Lyme disease: A rigorous review of diagnostic criteria and treatment

34
Review Lyme disease: A rigorous review of diagnostic criteria and treatment Andrea T. Borchers a , Carl L. Keen b , Arthur C. Huntley c , M. Eric Gershwin a, * a Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616, USA b Department of Nutrition, University of California at Davis, Davis, CA 95616, USA c Department of Dermatology, University of California at Davis, Davis, CA 95616, USA article info Article history: Received 8 September 2014 Received in revised form 15 September 2014 Accepted 16 September 2014 Available online xxx Keywords: Lyme disease Spirochetes Autoimmunity Diagnostic criteria abstract Lyme disease was originally identied in Lyme, Connecticut, based upon an unusual cluster of what appeared to be patients with juvenile rheumatoid arthritis. It was subsequently identied as a new clinical entity originally called Lyme arthritis based on the observation that arthritis was a major clinical feature. However, Lyme arthritis is now called Lyme disease based upon the understanding that the clinical features include not only arthritis, but also potential cardiac, dermatologic and neurologic ndings. Lyme disease typically begins with an erythematous rash called erythema migrans (EM). Approximately 4e8% of patients develop cardiac, 11% develop neurologic and 45e60% of patients manifest arthritis. The disease is transmitted following exposure to a tick bite containing a spirochete in a genetically susceptible host. There is considerable data on spirochetes, including Borrelia burgdorferi (Bb), the original bacteria identied in this disease. Lyme disease, if an organism had not been identied, would be considered as a classic autoimmune disease and indeed the effector mechanisms are similar to many human diseases manifest as loss of tolerance. The clinical diagnosis is highly likely based upon appropriate serology and clinical manifestations. However, the serologic features are often mis- interpreted and may have false positives if conrmatory laboratory testing is not performed. Antibiotics are routinely and typically used to treat patients with Lyme disease, but there is no evidence that pro- longed or recurrent treatment with antibiotics change the natural history of Lyme disease. Although there are animal models of Lyme disease, there is no system that faithfully recapitulates the human disease. Further research on the effector mechanisms that lead to pathology in some individuals should be further explored to develop more specic therapy. © 2014 Elsevier Ltd. All rights reserved. 1. Introduction Lyme disease takes its name from the town of Lyme in Con- necticut, U.S.A., where an unusual cluster of cases with an initial diagnosis of juvenile rheumatoid arthritis occurred in the mid 1970s [1]. Closer examination of this community revealed that the recurrent attacks of arthritis in these patients appeared to be a new clinical entity, originally called Lyme arthritis (LA). After it became clear that arthritis constituted only one of the late manifestations of a multisystem disease that included dermatological, neurological and cardiac manifestations [2], the name was changed to Lyme disease. Early prospective studies in the US revealed the natural history of this disease in untreated patients. Lyme often begins with an erythematous rash, consistent with an entity called erythema mi- grans (EM) or erythema chronicum migrans that had rst been described in 1909 in Europe [3]. Some patients (20%) experience no further signs and symptoms, the remainder may develop neuro- logical (11%), cardiac (4e8%) or rarely ocular abnormalities, and 45e60% eventually develop arthritis [4,5]. Not only EM, but also a meningoradiculoneuritis had long been known to occur after tick bites in Europe and to be associated with each other [6]. In 1982 Burgdorfer et al. [7] isolated a previously unidentied spirochete from deer ticks (Ixodes dammini, now Ixodes scapularis)(Fig. 1). Their and other data demonstrated that this was the etiological agent of Lyme disease [7e9], which was named Borrelia burgdorferi (Bb) in honor of one of its original discoverers [10]. These bacteria Abbreviations: ACA, acrodermatitis chronica atrophicans; AI, antibody index; AV, atrioventricular; Bb, Borrelia burgdorferi; CDC, Centers for Disease Control and Prevention; CNS, central nervous system; CSF, cerebral spinal uid; EIA, enzyme immunoassay; DbpA, decorin-binding protein A; EM, erythema migrans; LA, Lyme arthritis; LNB, Lyme neuroborreliosis; PCR, polymerase chain reaction. * Corresponding author. Division of Rheumatology, Allergy and Clinical Immu- nology, University of California at Davis, School of Medicine, 451 Health Sciences Drive, Suite 6510, Davis, CA 95616, USA. Tel.: þ1 530 752 2884; fax: þ1 530 752 4669. E-mail address: [email protected] (M.E. Gershwin). Contents lists available at ScienceDirect Journal of Autoimmunity journal homepage: www.elsevier.com/locate/jautimm http://dx.doi.org/10.1016/j.jaut.2014.09.004 0896-8411/© 2014 Elsevier Ltd. All rights reserved. Journal of Autoimmunity xxx (2014) 1e34 Please cite this article in press as: Borchers AT, et al., Lyme disease: A rigorous review of diagnostic criteria and treatment, Journal of Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

Transcript of Lyme disease: A rigorous review of diagnostic criteria and treatment

lable at ScienceDirect

Journal of Autoimmunity xxx (2014) 1e34

Contents lists avai

Journal of Autoimmunity

journal homepage: www.elsevier .com/locate/ jaut imm

Review

Lyme disease: A rigorous review of diagnostic criteria and treatment

Andrea T. Borchers a, Carl L. Keen b, Arthur C. Huntley c, M. Eric Gershwin a, *

a Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA 95616, USAb Department of Nutrition, University of California at Davis, Davis, CA 95616, USAc Department of Dermatology, University of California at Davis, Davis, CA 95616, USA

a r t i c l e i n f o

Article history:Received 8 September 2014Received in revised form15 September 2014Accepted 16 September 2014Available online xxx

Keywords:Lyme diseaseSpirochetesAutoimmunityDiagnostic criteria

Abbreviations: ACA, acrodermatitis chronica atroAV, atrioventricular; Bb, Borrelia burgdorferi; CDC, CenPrevention; CNS, central nervous system; CSF, cerebimmunoassay; DbpA, decorin-binding protein A; EM,arthritis; LNB, Lyme neuroborreliosis; PCR, polymeras* Corresponding author. Division of Rheumatology

nology, University of California at Davis, School of MDrive, Suite 6510, Davis, CA 95616, USA. Tel.: þ1 5304669.

E-mail address: [email protected] (M.E. G

http://dx.doi.org/10.1016/j.jaut.2014.09.0040896-8411/© 2014 Elsevier Ltd. All rights reserved.

Please cite this article in press as: BorcheAutoimmunity (2014), http://dx.doi.org/10.1

a b s t r a c t

Lyme disease was originally identified in Lyme, Connecticut, based upon an unusual cluster of whatappeared to be patients with juvenile rheumatoid arthritis. It was subsequently identified as a newclinical entity originally called Lyme arthritis based on the observation that arthritis was a major clinicalfeature. However, Lyme arthritis is now called Lyme disease based upon the understanding that theclinical features include not only arthritis, but also potential cardiac, dermatologic and neurologicfindings. Lyme disease typically begins with an erythematous rash called erythema migrans (EM).Approximately 4e8% of patients develop cardiac, 11% develop neurologic and 45e60% of patientsmanifest arthritis. The disease is transmitted following exposure to a tick bite containing a spirochete in agenetically susceptible host. There is considerable data on spirochetes, including Borrelia burgdorferi (Bb),the original bacteria identified in this disease. Lyme disease, if an organism had not been identified,would be considered as a classic autoimmune disease and indeed the effector mechanisms are similar tomany human diseases manifest as loss of tolerance. The clinical diagnosis is highly likely based uponappropriate serology and clinical manifestations. However, the serologic features are often mis-interpreted and may have false positives if confirmatory laboratory testing is not performed. Antibioticsare routinely and typically used to treat patients with Lyme disease, but there is no evidence that pro-longed or recurrent treatment with antibiotics change the natural history of Lyme disease. Althoughthere are animal models of Lyme disease, there is no system that faithfully recapitulates the humandisease. Further research on the effector mechanisms that lead to pathology in some individuals shouldbe further explored to develop more specific therapy.

© 2014 Elsevier Ltd. All rights reserved.

1. Introduction

Lyme disease takes its name from the town of Lyme in Con-necticut, U.S.A., where an unusual cluster of cases with an initialdiagnosis of juvenile rheumatoid arthritis occurred in the mid1970s [1]. Closer examination of this community revealed that therecurrent attacks of arthritis in these patients appeared to be a newclinical entity, originally called Lyme arthritis (LA). After it became

phicans; AI, antibody index;ters for Disease Control andral spinal fluid; EIA, enzymeerythema migrans; LA, Lymee chain reaction., Allergy and Clinical Immu-edicine, 451 Health Sciences752 2884; fax: þ1 530 752

ershwin).

rs AT, et al., Lyme disease:016/j.jaut.2014.09.004

clear that arthritis constituted only one of the late manifestations ofa multisystem disease that included dermatological, neurologicaland cardiac manifestations [2], the name was changed to Lymedisease.

Early prospective studies in the US revealed the natural historyof this disease in untreated patients. Lyme often begins with anerythematous rash, consistent with an entity called erythema mi-grans (EM) or erythema chronicum migrans that had first beendescribed in 1909 in Europe [3]. Some patients (20%) experience nofurther signs and symptoms, the remainder may develop neuro-logical (11%), cardiac (4e8%) or rarely ocular abnormalities, and45e60% eventually develop arthritis [4,5]. Not only EM, but also ameningoradiculoneuritis had long been known to occur after tickbites in Europe and to be associated with each other [6]. In 1982Burgdorfer et al. [7] isolated a previously unidentified spirochetefrom deer ticks (Ixodes dammini, now Ixodes scapularis) (Fig. 1).Their and other data demonstrated that this was the etiologicalagent of Lyme disease [7e9], which was named Borrelia burgdorferi(Bb) in honor of one of its original discoverers [10]. These bacteria

A rigorous review of diagnostic criteria and treatment, Journal of

Fig. 1. Ixodes scapularis, the primary vector for Lyme disease in eastern North America.Photo credit: A new view on lyme disease: rodents hold the key to annual risk. Gross L,PLoS Biology Vol. 4/6/2006, e182. http://dx.doi.org/10.1371/journal.pbio.0040182(Creative Commons Attribution 2.5 Generic license).

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e342

belong to the phylum Spirochetes, have an outer membrane sur-rounding a protoplasmic cylinder, are long (10e30 mm), but thin(0.18e0.25 mm in diameter), irregularly coiled, and highly motiledue to a 7e11 flagella inserted at each end of the cell and containedin the periplasmic space. They have a small linear chromosome anda variable number of linear and circular plasmids (up to 21 plas-mids, 12 of which are linear, while 9 are circular) [11]. The genomeis highly complex, with almost 8% of all open reading framesencoding lipoproteins and another 6% devoted to motility andchemotaxis. Since open reading frames in the Borrelia genomeoften lack significant homology with previously annotated genes,the function of a considerable portion of their products remains tobe elucidated. Interestingly, if an organism had not been identified,Lyme would be considered as a classic autoimmune disease withsimilarities to other human diseases with loss of tolerance andseemingly common effector mechanisms [12e15].

Spirochetes with similar morphology, protein profile and anti-genic determinants were detected in Ixodes ricinus ticks fromSwitzerland and Ixodes pacificus ticks from Oregon [7,16] and sub-sequently in Ixodes persulcatus ticks in Russia [17]. Genotypingsubsequently established that Lyme disease in Europe was causednot only by Bb sensu stricto, but also by two genotypically distinctspecies, which were named Borrelia afzelii and Borrelia garinii andsubsumed under the B. burgdorferi sensu lato complex (here Bor-relia species abbreviated as Borrelia spp.), which now contains atleast 20 confirmed or proposed genomic species (see Table 1). Ofthese, Borrelia spielmanii, Borrelia bavariensis, Borrelia bissettii,Borrelia lusitaniae, Borrelia americana, Borrelia andersonii and Bor-relia valaisiana also have pathogenic potential (see Table 1 for theavailable evidence). Of particular note, it had long been held that Bbis the only species causing Lyme disease in the US, but PCR-baseddetection of both B. americana and B. andersonii has been re-ported in blood or skin of a number of US patients, some of themwith EM and/or other manifestations of Lyme disease [18,19]. Inaddition, B. bissettii, which has been isolated from the cerebralspinal fluid (CSF) of a European patient with Lyme disease [20], hasalso been detected by PCR in serum of three Californian subjects,but details on whether this was associated with any disease man-ifestations are not available [21]. It should be kept in mind, how-ever, that detection by PCR or even isolation by culture of a

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

particular species of Borrelia does not prove its pathogenicity sinceit is not uncommon for patients to simultaneously harbor two ormore species [22e25]. While direct PCR of tissue samples appearsto be more sensitive than culture-based typing, it does not reliablyamplify all strains identified by culture [26].

Borrelia spp. are maintained in a complex enzootic cycleinvolving one or more vertebrate host reservoir species and one ormore tick species. The major vectors transmitting pathogenic Bor-relia spp. all belong to the hard-bodied ticks of the genus Ixodes,with I. scapularis transmitting Bb in the East, mid-Atlantic andupper Midwest of the US and I. pacificus transmitting Bb andB. bissettii in the Western parts of the US, whereas I. ricinus is themajor European vector for Bb, B. afzelii, and B. garinii along withother potentially pathogenic and non-pathogenic Borrelia spp., andI. persulcatus, whose geographical distribution partially overlapswith I. ricinus, is the main vector of B. afzelii and B. garinii in easternregions of Europe and in Asia. Note, however, that there have beenisolated reports implicating other tick genera in transmitting Bband other Borrelia spp., including Amblyomma americanum inFlorida and Georgia [18] and ticks of the genus Dermacentor inSpain [27]. The biology of Borrelia spp. is closely tied to the lifecycleof the tick vector. Ticks take one blood meal during each of thethree stages of their life cycle, i.e., as larvae, nymphs, and adults.There is little to no transovarial transmission of Borrelia spp.infection, therefore ticks become infected when larvae or nymphsfeed on an infected host, infection is thenmaintained transstadiallyand can be passed on to the host providing the next blood meal,including humans.

2. Epidemiology of Lyme disease

Almost all confirmed cases of Lyme disease have occurred in theNorthern hemisphere, the majority coming from the United Statesand Europe (including the European part of Russia), far fewer fromAsia, and some from Northern Africa [28]. In the US, Lyme diseasehas been a nationally notifiable disease since 1991 and the numberof reported cases rose steadily from 9908 in 1992 to a peak ofalmost 30,000 confirmed cases in 2009 [29]. Since then, the inci-dence appears to have stabilized below 25,000 confirmed casesannually. There are numerous indications that underreportingrepresents a considerable problem [30e32], and scientists from theUS Centers for Disease Control and Prevention (CDC) actually esti-mate that the true number of cases may be closer to 300,000 eachyear [33]. On the other hand, it is equally clear that there is aconsiderable over-diagnosis of Lyme disease based upon misinter-pretation of the serology and/or over-zealous interpretation ofinternet information [34].

The Northeast, mid-Atlantic region and upper Midwest are theprimary areas of endemicity, and just 10 states (Connecticut,Delaware, Massachusetts, Maryland, Minnesota, New Jersey, NewYork, Pennsylvania, Rhode Island and Wisconsin) account for �93%of annual cases. But evenwithin these endemic areas, the incidencevaries considerably, exceeding 500/105 in certain counties [35].Among the cases reported to the CDC, AfricaneAmericans aresignificantly underrepresented [36,37]. This may be due to a lowerrisk of exposure since African Americans more rarely live in highlyendemic rural areas. However, the results of a detailed analysis ofcases in an endemic area of Maryland revealed that area of resi-dence alone could not fully account for the different incidence ratesof African Americans compared to European Americans [36].Instead, a significantly lower rate of EM and an excess risk ofarthritis among African Americans suggested that under-recognition of EM e or unawareness of its clinical significance e

resulted in a disproportionate number of late manifestations in thispopulation. Unlike many other immune-mediated diseases, there is

A rigorous review of diagnostic criteria and treatment, Journal of

Table 1Borrelia spp., their vectors, and evidence for their pathogenicity (culture from human tissue, demonstration of their DNA in human tissue, pathogenicity in mice). (List of species and vectors based on [600,601]).

Borrelia species Vector Geographicaldistribution

Culturedfrom blood

Cultured from EM Isolated from other tissue

Positive PCR Pathogenicity in mice

B. afzelii I. ricinus, I. persulcatus,I. hexagonus

Asia, Europe Rarely [104,105] [20,111,167,168,391] Synovial fluid or tissue,CSF [20,24,110,113,385]ACA [395,396]BL [184]

CSF, blood [104,388];synovial fluid [602,603];heart valve [366]

[604]

B. americana I. pacificus, I. minor USA Blood or skin [18,19]B. andersonii I. dentatus USA Blood or skin [18,19]B. bavariensis

(formerly OspAserotype 4 ofB. garinii)

I. ricinus Europe [391] EM [412]

B. bissettiia I. ricinus, I. scapularis,I. pacificus, I. minor,I. affinis

Europe, USA [605] CSF [20,605];BL [184]

Serum [21]; blood [606];cardiactissue [607]

[608]

B. burgdorferi sensustricto

I. ricinus, I. scapularis,I. pacificus,I. spinipalpis, I. hexagonis,I. affinis, I. minor,I. muris

Europe, USA [101,103,405] [20,80,164,166] Synovial fluid, CSF[20,24,112,385]Heart [20]ACA [395,396]

CSF, blood [104,220,388];synovialfluid or tissue[118,268,384,578,602,603]

[379,380,609,610]

B. californiensis I. pacificus, I. jellisonii,I. spinipalpis

USA

B. carolinensis I. minor USAB. garinii I. ricinus, I. persulcatus,

I. nipponensis,I. uriae

Asia, Europe Rarely [105,110] [20,111,166,168] Synovial fluid or tissue, CSF[20,24,111,385]ACA [110,395,396]BL [394]

CSF, blood [104,388];synovial fluid [602,603]

[369,611,612]

B. finlandensis I. ricinusB. japonica I. ovatus Japan In inbred mice [613], but

not in outbred mice [614]B. kurtenbachiia I. scapularis, (I. ricinus) Europe, USA CSF or skin [605] Weak [615], absent [616];B. lusitaniae I. ricinus Europe, North Africa [617] Yes, but not arthritogenic

[611,618]B. sinica I. ovatus ChinaB. spielmanii I. ricinus Europe [20,145,382,619,620]B. tanukii I. tanuki JapanB. turdi I. turdus JapanB. valaisiana

(VS116)I. ricinus, I. granulatus,I. nipponensis,I. columnae

Asia, Europe Skin [23]; blood [621]; CSF[622]; B. valaisiana-relatedspeciesin blood [234]

[611]

B. yangtze I. granulatus, I. nipponensis,Haemaphysalis longicornis

China

Genomospecies 2 I. pacificus, I. spinipalpis USA

a B. bissettii and B. kurtenbachii (initially called Bb strain 25015) were originally both included in the B. bissettii genospecies. Therefore, it is not always possible in older studies to determine which patient isolates belong towhich of these novel species.

A.T.Borchers

etal./

Journalof

Autoim

munity

xxx(2014)

1e34

3

Pleasecite

thisarticle

inpress

as:Borchers

AT,

etal.,

Lymedisease:

Arigorous

reviewof

diagnosticcriteria

andtreatm

ent,Journal

ofAutoim

munity

(2014),http://dx.doi.org/10.1016/j.jaut.2014.09.004

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e344

no obvious sex bias (see below) nor is there any significant data onconcordance in twins or within large family pedigrees [38e40].

In most of Europe, Lyme disease is not a reportable disease, anduntil recently there were no standardized case definitions [41].Therefore, the available data are less reliable and difficult tocompare because a variety of active and passive surveillancemethods were used to arrive at the available estimates. Nonethe-less, it is clear that Lyme borreliosis is highly endemic in much ofEurope, with the highest incidence reported from southern Swe-den, Lithuania, Germany, Austria, and Slovenia. The total number ofannual cases in Europe is estimated to be ~3-fold higher than thenumber of the cases reported to the CDC [28]. A rise in the inci-dence of Lyme borreliosis similar to that observed in the USA hasbeen reported from some European regions [42e44]. Increasedawareness of the disease almost certainly accounts for some of thisrise, but climate change with a subsequent expansion of the terri-tory of vector ticks, along with changes in land use, resulting inincreases in reservoir hosts for ticks, and changes in residentialdistribution and human recreational behavior also contribute;there is also the potential error of a false positive diagnosis[42,45e49].

While the CDC data in the USA suggest that males constituteslightly more than half of the reported cases (54% overall and 61% ofpediatric cases) [37], data from Europe often suggest a slight tomarked female preponderance [28,42,50], but an even distributionhas been reported from some areas [51e53]. The age distributionamong cases with Lyme disease is generally bimodal, with thehighest incidence rates seen in children aged 5e9 years and inadults >50 years of age in both the US and Europe[28,35,37,42,51,54].

3. Risk of infection after a tick bite and factors influencing it

3.1. Risk of infection

In areas that are endemic for Lyme disease, the risk of Borreliainfection after the bite of an infected tick is only 1% and 3% in the US[55e57], and 3e12% in Europe [58e62]. However, at least one-fourth, and sometimes more than half, of European subjects sero-convert without developing clinical manifestations of Lyme disease[58e61]. In a cohort of highly exposed individuals, almost all of theseroconverters remained asymptomatic [63]. This is the moreremarkable since a substantial proportion (�30%) of European pa-tients with EM never develop seropositivity [64e66]. In contrast,asymptomatic seroconversion is quite rare in the US [67,68]; themechanism for this is unclear and further studies are suggested.

3.2. Environmental and behavioral factors

Risk factors for Borrelia spp. infection are time spent in endemicareas, time spent outdoors, and factors associated with tick density[69e71]. Therefore, the most effective way to minimize the risk ofBorrelia infection is to avoid tick habitats, such as wooded areas,shrubs, tall grass and particularly the edges where these differenttypes of vegetation meet. Wearing protective clothing, using DEETinsect repellents, conducting a visual inspection every night tocheck for attached ticks, and prompt removal of attached ticks areother important preventive measures. Of note, a previous diagnosisof Lyme disease was identified, along with age, as an independentpredictor of Bb infection in a study that investigated both envi-ronmental and behavioral risk factors [70]. This probably reflectsthat the same population subgroups stays at risk of repeated in-fections because of the same behavioral or environmental factorsthat increased their original risk. The role of environment versusheritability is well established in human immune diseases [72]. The

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

high rates of reinfection observed in several cohort studies confirmthat a previous history of Lyme disease constitutes a risk factor forreinfection [73e75]. This also means that previous infection doesnot protect from reinfection. Mice do develop immunity, but it isstrain-specific [76], and there are recent empirical data suggestingthat humans also develop strain-specific immunity that lasts for�6years [77]. This could explain the extraordinarily low rate ofsymptomatic infection (~2%) in a Swiss cohort with frequentexposure to ticks most likely containing a wide variety of strains[63]. It is important to note that Lyme disease follows the pattern ofa classic infection, much like that of post-streptococcal glomeru-lonephritis or rheumatic heart disease. It is not related to trauma,exercise, automobile accidents, or weather. It is strictly a hostpathogen relationship. Indeed, both in the case of Lyme as well asany other human immunologically mediated disease, the role forenvironmental associated factors has reasonably defined criteria[78].

3.3. Season and tick developmental stage

In both the Eastern US and most parts of Europe, peak onset ofLyme disease occurs during the summer months, when nymphalabundance is highest and nymphs are actively questing, whereasfew cases occur during the late fall and the first few months of theyear when adult ticks actively seek a host [51,54,64,79,80]. How-ever, in some European regions, a secondary peak in the EM inci-dence is seen late in the fall (October, November), suggesting thatactively questing adults also contribute to human Borrelia in-fections [64,81], and in Russia, Borrelia spp. are transmitted almostexclusively by adult I. persulcatus ticks [62]. Infection rates arehigher in adult than in nymphal ticks [20,79,82], but nymphs aremore abundant and are much smaller and more difficult to detectand, consequently, are more likely to stay attached longer [59].Duration of tick attachment has been identified as an importantrisk factor for infection, at least in the US.

3.4. Duration of tick attachment

Spirochetes reside in the midgut of unfed ticks. Once the bloodmeal reaches the midgut, the spirochetes begin to replicate andthen disseminate via the hemocoel to the salivary glands [83].There are experimental data demonstrating that Bb-infectedI. scapularis nymphs transfer considerable numbers of spirochetesas early as 24 h after attachment [84]. Nonetheless, in experimentalanimals, infection rarely occurs within 24 h of tick attachment, andpeak infection rates are not reached until 48e72 h of attachment[85e90]. The time required for infection to occur appears to dependon the target host species [85,86] and on the infecting strain of Bb[88]. In accordance with these findings, the risk of infection forresidents of endemic areas of the US was found to be minimal ifticks stayed attached for <72 h and to increase significantly withlonger attachment duration [55,67].

Data on the transmission of Borrelia spp. by I. ricinus ticks arelimited and obtained in gerbils [91,92]. These data suggest thatconsiderable rates of host infectionmay be observed as early as 17 hpost-attachment [91] and that I. ricinus transmits B. afzelii soonerand more efficiently compared to Bb [92]. In accordance with theexperimental animal data, attachment time does not appear to be amajor factor in determining the risk of infection in Europe [59,61],and there are a number of reports of European subjects becominginfected after a tick attachment duration of �24 h [59,60,64],particularly in Russia [62]. It is often hypothesized that the earliertransmission of European Borrelia spp. may be attributable to ahigher rate of systemic infection in unfed I. ricinus nymphs,including infection of the salivary glands, but data in support of this

A rigorous review of diagnostic criteria and treatment, Journal of

Fig. 2. Erythema migrans e erythematous target-like plaque of Lyme disease. Theprimary skin lesion of Borrelia infection is noted for centrifugal expansion, sometimesleaving central clearing. Photo Credit: James Gathany Content Providers(s): CDC/JamesGathany e This media comes from the Centers for Disease Control and Prevention'sPublic Health Image Library (PHIL), with identification number #9875.

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 5

hypothesis are limited [93e95] and the rates of systemic infectionof unfed I. scapularis nymphs in the US are quite similar to thosereported from Europe [96].

3.5. Inoculum size

The minimal infective dose in humans is unknown, but only lownumbers of spirochetes (20e60/gland) can be detected in salivaryglands of feeding ticks [83,89,97], suggesting that a small inoculumis sufficient to cause infection and disease. In mice (mostly C3H),the ID50 (infectious dose at which 50% of animals became infected)for subcutaneous needle inoculation of salivary gland extracts ob-tained from partially fed nymphal ticks was 18 spirochetes [98]. TheID50 values in syringe inoculation experiments are somewhathigher and greatly depend on the Bb strain under investigation[83,99,100].

4. Manifestations and diagnosis

4.1. Stages of Lyme disease

It is customary to divide Lyme disease into three stages. The“primary lesion” or early localized stage occurs at the site of Borreliainoculation. The infection is manifest as erythema migrans(migrating redness), an erythematous papule that expandscentrifugally over days to become a plaque, with or without centralclearing (Fig. 2). With the development of EM, the patient may alsohave constitutional symptoms such as fever, malaise, and headache.The skin lesion may have localized pain or pruritus, and regionallymphadenopathy may develop. In ~20% of untreated subjects, thisearly stage with EM remains the only manifestation of Lyme dis-ease. In the first few weeks after infection, spirochetes can becultured from blood of up to 45% of American patients, indicatingthat they disseminate via the bloodstream to other tissues[101e103]. Note, however, that culture isolation from blood ismuchless frequently achieved in European patients (0.7e7.7%)[104e106], possibly suggesting other routes of dissemination. The“secondary lesions” of Borrelia infection, the early disseminatedstage, comprises multiple EM lesions, widespread erythematousplaques that are smaller but morphologically similar to the initiallesion. The early disseminated stage may also be associated withLyme neuroborreliosis (LNB), and carditis. European patients mayalso develop a skin lesion called borrelial lymphocytoma duringthis stage (Fig. 3). The “tertiary skin lesion” of this infection, iden-tified for decades before the associationwith Borreliawas known, isacrodermatitis chronic atrophicans, a focal area of atrophic skinusually involving an extremity. Acordermatitis chronica atrophi-cans (ACA) is primarily found in Europe, and is most commonlyassociated with the late disseminated stage, which can also includelate neurological manifestations. Although LA can occur during theearly disseminated stage, it is more commonly observed during thelate disseminated stage.

4.2. Case definitions

There are no standardized diagnostic criteria for Lyme borre-liosis. Unfortunately, this has led to both over and under diagnosisof Lyme disease. Clearly the disease requires the tick, the spirocheteand the host. In other words, the presence of ticks by themselvesdoes not in any fashion lead to the diagnosis of Lyme disease. TheCDC has published case definitions for surveillance purposes (seeTable 2), but emphasizes that these are not intended as diagnosticcriteria [107]. The European Concerted Action on Lyme Borreliosis(EUCALB) has proposed clinical case definitions for use in clinicalsettings and epidemiological investigations (see Table 3) (EUCALB,

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

www.eucalb.com) [41]. Except for EM, the manifestations of Lymedisease are not specific, but can have a variety of causes. It istherefore vital to obtain a detailed history in order to establishprobable exposure to Ixodes ticks in an endemic area at an appro-priate time of year and to obtain appropriate and definitive labo-ratory confirmation. This is a point that should be stronglyemphasized to avoid misdiagnosis.

4.3. Laboratory confirmation

4.3.1. CultureWith the exception of EM, all manifestations of Lyme borreliosis

require laboratory confirmation. The diagnostic gold standard is theisolation of Borrelia spp. by culture with subsequent PCR-based orother confirmation of its identify. Since culture is expensive andrequires special media and laboratory expertise, it is not routinelyperformed particularly since results are not available for 2e6weeksand, therefore, are not useful for clinical decision making. In addi-tion, the sensitivity of the procedure is 40e70% for EM[64,80,108,109], but only ~3e17% for CSF samples [104,110,111], andvery low for synovial fluid or tissue samples [112,113]. Therefore,negative results do not exclude the diagnosis of Lyme borreliosis.

A rigorous review of diagnostic criteria and treatment, Journal of

Fig. 3. Borrelial lymphocytoma. Although these lesions more commonly occur oncooler areas such as ear lobes and scrotum, the may appear elsewhere including at theoriginal site of inoculation and present as erythematous to purplish discrete nodules.Photo credit: “Borrelial lymphocytoma” by Spectrum11 e Own work. Licensed underCreative Commons Zero, Public Domain Dedication via Wikimedia Commons e http://commons.wikimedia.org/wiki/File:Borrelial_lymphocytoma.jpg#mediaviewer/File:Borrelial_lymphocytoma.jpg.

Table 2CDC case definitions for surveillance purposes [107].

Case definitions CDC

Erythemamigrans

A skin lesion that typically begins as a red macule orpapule and expands over a period of days to weeks toform a large round lesion, often with partial centralclearing.the largest diameter must reach a size �5 cm;the diagnosis must be made by a physician;laboratory confirmation is recommended for personswith no known exposure.

Neuroborreliosis Any of the following, alone or in combination:lymphocytic meningitis;cranial neuritis, particularly facial palsy (may bebilateral);radiculoneuropathy;encephalomyelitis.Encephalomyelitis must be confirmed by Bb-specificantibody production in CSF

Musculoskeletalsystem

Recurrent brief attacks of objective joint swelling in oneor a few joints, sometimes followed by chronic arthritisin one or a few joints

Cardiovascularsystem

Acute onset of high-grade (2nd or 3rd degree)atrioventricular conduction defects that resolve in daysto weeks and are sometimes associated withmyocarditis

Suspected A case of EM without known exposure (defined ashaving been �30 days before the onset of EM inwooded, brushy, or grassy areas in a county in whichLyme disease is endemic)A casewith laboratory evidence of infection but withoutavailable clinical information

Probable Any other case of physician-diagnosed Lyme diseasethat has laboratory evidence of infection

Confirmed A case of EM with a known exposureA case of EM with laboratory evidence of infection andwithout a known exposureA case with at least one late manifestation that haslaboratory evidence of infection

Laboratoryevidence

Positive culture for Bb ORTwo-tier testing (for specific antibodies) interpretedusing established criteria, wherePositive IgM is sufficient only when �30 d fromsymptom onsetPositive IgG is sufficient at any point during illnessSingle-tier IgG immunoblot seropositivity usingestablished criteriaCSF antibody positive for Bb by EIA or IFA, when the titeris higher than it was in serum

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e346

4.3.2. PCRPCR on DNA extracted from tissue or fluid specimens is useful

for the confirmation of Borrelia infection, particularly in the syno-vium of patients with LA and also in cases of diagnostic uncertainty,but is generally performed only for research purposes. A large va-riety of methods have been used, but none of these is standardizedand, accordingly, distinct methods yield rather divergent results[104]. Its sensitivity is 75e80% in EM [23,108,114], 15e30% in CSF[104,115,116], and 60e85% in synovial fluid samples [117,118].Again, negative findings do not exclude the diagnosis of Lymedisease.

4.3.3. SerologySingle serological tests yield false-positive results in subjects

with other spirochetal infections; spirochete infection of gums isvery common. Hence, both the CDC and European guidelinesstrongly recommend a two-tier approach. The two steps consist of asensitive enzyme immunoassay (EIA), or rarely an indirect immu-nofluorescence assay, followed by immunoblotting of samples thatare positive or indeterminate in the first step. The CDC has pub-lished recommendations on the number and types of IgM and IgGbands that have to be present in order to consider immunoblotresults positive [119]. These rules should not be applied to patientswho were infected in Europe since the existence of at least 3pathogenic species requires species-specific interpretations rules[120]. In addition, with the currently available serological tests, theBorrelia-specific antibody repertoire seems more restricted [121],fewer patients with EM alone ever develop seropositivity (�70%compared to 80e86%) [64e66,122,123], and subclinical infectionappears to be considerablymore common in European compared toUS patients [56e61,63,67,68]. All of these factors are likely to lowerthe sensitivity and specificity of serological tests in the diagnosis ofLyme disease acquired in Europe. Known positive and negativesamples must be included in all assays; this is critical.

Obtaining serological confirmation of infection is hampered by avariety of problems. Antibodies against Borrelia spp. are slow todevelop, with IgM generally not being detectable for the first 1e2

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

weeks after infection and IgG often not emerging for 4e6 weeks[51,122e127]. In patients who are seronegative at presentation butin whom there is strong suspicion of Borrelia infection, it is advis-able to obtain evidence of seroconversion, preferably within 8e14days after presentation [123,126]. However, some patients withsolitary EM as their only manifestation may never seroconvert[122,123], particularly in Europe [64e66]. The rate of seropositivitycorrelates with the duration of symptoms before diagnosis andtreatment not only in samples taken at presentation[51,126,128,129], but also in samples obtained during follow-up,indicating that early antibiotic treatment may abrogate the devel-opment of seropositivity [66,130]. There is great variability in thespecific antigens recognized by individual sera and the extent towhich serum antibodies recognize homologous proteins from otherpathogenic Borrelia spp. or even from heterologous strains of thesame species [99,121,131e133].

Both IgM and IgG Borrelia-specific antibodies may persist foryears in some patients [123,126,134,135], which make it impossibleto distinguish between past and newly acquired infections based onseropositivity alone. The background positivity rate in the generalpopulation may be high in endemic areas, ranging from 5 to 8.4% inUS studies with two-tier testing [136e138], and from 3 to 15% in

A rigorous review of diagnostic criteria and treatment, Journal of

Table 3Case definitions from the European Union Concerted Action on Lyme Borreliosis (EUCALB, www.eucalb.com) [41]

Manifestation Clinical Essential laboratory evidence Supporting laboratory/clinical evidence

EM Expanding red or bluish-red patch(�5 cm in diameter),a with or withoutcentral clearing. Advancing edgetypically distinct, often intenselycolored, not markedly elevated

None Detection of Borrelia spp. by cultureand/or PCR from skin biopsy

Borrelial lymphocytoma(rare)

Painless bluish-red nodule or plaque,usually on ear lobe, ear helix, nipple orscrotum

Seroconversion or positive serologyb

Histology in unclear casesHistologyDetection of Borrelia spp. by cultureand/or PCR from skin biopsyRecent or concomitant EM

ACA Long-standing red or bluish-red lesions,usually on the extensor surfaces ofextremities. Initial doughy swelling.Lesions eventually become atrophic.Possible skin induration and fibroidnodules over bony prominences

High level of specific serum IgG HistologyDetection of Borrelia spp. by cultureand/or PCR from skin biopsy

LNB In adults mainly meningo-radiculitis,meningitis;rarely encephalitis, myelitis;very rarely cerebral vasculitis.In children mainly meningitis and facialpalsy

Pleocytosis and demonstration ofintrathecal specific antibody synthesisc

HistologyDetection of Borrelia spp. by cultureand/or PCR from CSFIntrathecal synthesis of total IgM and/orIgG and/or IgASpecific serum antibodiesRecent or concomitant EM

Lyme arthritis Recurrent attacks or persistingobjective joint swelling in one or a fewlarge joints. Alternative explanationsmust be excluded

Specific serum IgG antibodies; usuallyin high concentrations

Synovial fluid analysisDetection of Borrelia spp. by cultureand/or PCR from synovial fluid and/ortissue

Lyme carditis Acute onset of atrio-ventricular (IeIII)conduction disturbances, rhythmdisturbances, sometimes myocarditis orpancarditis.Alternative explanations must beexcluded

Specific serum antibodies Detection of Borrelia spp. by cultureand/or PCR from endomyocardialbiopsyRecent or concomitant EM and/orneurologic disorders

Ocular manifestations Conjunctivitis, uveitis, papillitis,episcleritis, keratitis

Specific serum antibodies Recent or concomitant Lyme borreliosismanifestationsDetection of Borrelia spp. by cultureand/or PCR from ocular fluid

a If <5 cm, a history of tick bite, a delay (after the tick bite) of �2 days and an expanding rash at the site of the tick bite is required.b Initial and follow-up samples should be tested in parallel in order to avoid changes by inter-assay variation.c In early cases intrathecally produced specific antibodies may still be absent.

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 7

various European regions and some Chinese provinces [139e142].But seroprevalence rates �20% and even as high as 37% have beenreported in high-risk groups, such as forestry workers or orienteers,by single-tier testing [28,63,143]. In addition, even commerciallyavailable diagnostic systems are not standardized and their resultscan differ substantially [129,144,145]. Further (see below) there arefalse positive results and data must be critically evaluated.

First generation assays are based on Borrelia spp. sonicates, butthese are derived from cultured spirochetes, which do not expressmany of the immunogenic proteins that are elaborated in vivo.Second and third generation EIA and immunoblot assays use avariety of recombinant proteins that are expressed in the vertebratehost, such as decorin-binding protein A (DbpA), variable majorprotein-like sequence, expressed (VlsE), or BBK32, and alsorecombinant forms of homologous proteins from differentgenospecies, truncated proteins or synthetic peptides[120,133,144,146e151]. In a protein microarray study, sera frompatients with Lyme borreliosis react with a variety of antigens thathad not been identified by conventional methods, most likelybecause they are expressed only in vivo [132]. While these datawere obtained in patients with late Lyme disease, they offer thepotential of improving the sensitivity of serological assays in earlyLyme borreliosis, which may remain suboptimal even with thirdgeneration assays [129,150,152].

Immunoblots are subject not only to intra and interlaboratoryvariation but also to subjective interpretation [153]. Again, knownpositive and negative samples and appropriate use of dilutions is

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

necessary. Since no single EIA has sufficient specificity to be usedalone, efforts are being made to find a combination of EIAs thatyields the same or higher sensitivity and specificity compared toconventional two-tier testing [150,152,154]. A promising candidatefor one of these assays is a 26-mer peptide from the sixth invariantregion (C6) of the VlsE lipoprotein [124,139,152,154]. However, thisassay still does not eliminate the problem of cross-reactivity withother spirochetal and viral antigens and autoantigens, at least inEuropean patients [139], but this contrasts with results from the US[155]. In addition, the sixth invariant region of VlsE may not be ashighly conserved among strains of the three major pathogenicBorrelia genospecies as the original studies suggested [156], leadingto false negative results in a subset of European patients [157].Nonetheless, there are data suggesting that a USwhole cell sonicateELISA followed by a C6 ELISA can be used to identify patients withLyme regardless of their travel history, i.e., even if they acquiredtheir infection in Europe [158]. In contrast, conventional US two-tiered testing has very poor sensitivity in infections acquired inEurope.

4.4. Erythema migrans

The earliest sign of Borrelia infection is EM in ~70% of the casesreported to the CDC [159],�90% in cohorts of pediatric and adult USpatients [160e162], and 70e95% in European epidemiologicalstudies [51,53,54,163]. Adults are most commonly bitten on thelower limbs, buttocks or groin, less frequently on the trunk or arms,

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e348

and rarely on the head, whereas the bite is on the head or neck in20e40% of children [28,64,80,160,164]. Onset is most commonly inthe warm summer months, and is rare from December to March.EM begins as a red macule or papule and expands over a period ofdays to weeks into a large round or oval red or bluish-red lesionthat can reach a size of up to 73 cm [165,166]. Central clearing mayeventually leave an annular lesion or a rash with the bull's eyeappearance that is considered to be so typical of EM. Actually,however, central clearing is seen in only 19% of US patients ascompared to almost 80% of European patients according to a meta-analysis [165], confirming the results of direct comparisons be-tween US and European patients [166e169]. Occasionally, ulcera-tion or vesicles may occur at the center of the lesion [80,164].Pruritus is reported by 33 and 38% of US and European patients,respectively [165]. In about 21% of US patients, but only 12% ofEuropean patients, secondary EM lesions develop [165]. Lymph-adenopathy is observed in 13e22% of patients.

EM is frequently accompanied, sometimes preceded, by sys-temic symptoms, in particular fatigue, headache, stiff neck, myal-gias, arthralgias, fever, nausea and dysesthesia [165]. The frequencyof systemic symptoms overall and of most of the individualsymptoms is lower in European patients with EM caused mostly byB. afzelii and B. garinii compared to EM caused by Bb in the US. Inaddition, lesions caused by B. garinii grow faster and reach a largersize than those caused by B. afzelii [109,168]. They were also foundto cause local symptoms, such as itching, burning, or pain moreoften, be accompanied by myalgia and chills more frequently andbe associated with a higher seropositivity rate, even though pa-tients with B. garinii infection presented earlier after symptomonset [168].

EM was once considered to be virtually pathognomonic of Lymedisease. Actually, this no longer holds true, at least in certain areas ofthe United States where a Lyme-like illness referred to as southerntick-associated rash illness (STARI) causes skin lesions that areindistinguishable from EM [165]. It is transmitted by the lone startick (A. americanum), but the agent causing STARI remains un-identified. The situation is further complicated by the fact that therehave been reports of the isolation of Borrelia spp. from A. ameri-canum [18], and the range of this tick is expanding into areas whereLyme disease is endemic. Otherwise, EM is considered to be suffi-ciently distinct in appearance to allow its clinical diagnosis withoutany laboratory confirmation in a patient with the appropriateexposure history. Serological testing is not advised since many pa-tients have not yet developed anti-Borrelia antibodies by the timethey presentwith EM [122e126].While the CDC case definitions forsurveillance purposes state that a solitary lesion must reach at least5 cm in size [107], lesions smaller than 5 cm can be diagnosed as EMby experienced physicians according to European case definitions[41] (see also Table 3). When erythematous lesions develop duringtick attachment or within 24 h after tick removal they most likelyrepresent hypersensitivity reactions. A variety of rashes can mimicEM (see the excellent review by Tibbles and Edlow [165] for thefeatures that help distinguish them from EM).

4.5. Other cutaneous manifestations of LB

4.5.1. Acrodermatitis chronica atrophicans (ACA)ACA is a late cutaneous manifestation of Lyme borreliosis that

primarily affects middle-aged and elderly women [170,171]. It oc-curs in 1e7% of European patients with Lyme disease [51,52,54,172],but was not seen in any Russian [53] or Chinese patients [173], andonly rare cases have been described in the US [174,175]. Its onset isvery insidious, with the appearance of a bluish-red discoloration ofa skin area with doughy swelling usually on the sun-exposedextensor surfaces of the hands and feet. In the beginning, it is

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

usually unilateral, often on the same extremity that had beenaffected by EM months to years previously, but only ~20% of pa-tients give a history of EM [171]. ACA may later become symmet-rical and eventually affect all extremities in some patients [176].The initial inflammation can persist for years, but eventually isreplaced by atrophy of the skin [177]. Unlike EM and borreliallymphocytoma, it does not resolve spontaneously. Almost 20% ofpatients with ACA exhibit fibrous nodules particularly around theelbow or, less frequently, on other bony prominences such as theknee; others (~7%) have band-like fibrous lesions over the ulna ortibia [178]. At least 30e40% of patients develop a sensory orsensorimotor polyneuropathy that is most pronounced in the ex-tremities with cutaneous involvement [176,179].

Patients with ACA are usually strongly IgG-positive to a largevariety of borrelial antigens [121,178]. The differential diagnosis ofACA depends on the stage of the lesion and includes venousinsufficiency, acrocyanosis, livedo reticularis, lymphedema, andchilblains [175]. Histopathology may be helpful in supporting thediagnosis. A prominent histopathological feature is telangiectasesin association with a mostly perivascular infiltrate consistingmainly of lymphocytes with a variable number of plasma cells andmacrophages [178,180e182]. This infiltrate can be band-like, patchyor diffuse.

4.5.2. Borrelial lymphocytomaBorrelial lymphocytoma, also called lymphocytoma cutis or

lymphadenosis benigna cutis, is a presenting manifestation in0.3e4.5% of European cases of Lyme borreliosis[51,52,54,163,172,183], but is rarely reported from the eastern partsof Europe [53], while some cases from China have been described[173]. Children are affected more often than adults [51,54]. This B-cell pseudolymphoma manifests as a solitary red to bluish-redplaque or nodule a few centimeters in diameter, which may bepainful to touch [177,184e186]. It usually develops within weeks tomonths after infection, sometimes in association with EM or othermanifestations of early disseminated disease, but occasionally hasalso been observed late in the course of Lyme borreliosis togetherwith ACA [184]. It shows a strong predilection for earlobes inchildren, and nipples or genital areas in adults, but can develop inother regions of the body. A majority of patients either recall a tickbite or develop EM in the vicinity of the affected ear lobe or nipple,but in others the borrelial lymphocytomamay develop at a distancefrom the causative tick bite [177,184,185].

Solitary lymphocytoma cutis can have a variety of causes,although Borrelia infection is the most frequent one in endemicareas. Therefore, in the absence of EM, serological confirmation isrequired. The vast majority of patients with borrelial lymphocy-toma has Borrelia-specific IgM and/or IgG antibodies at presenta-tion or seroconverts over the course of the ensuing weeks[184e186], but some remain seronegative throughout the acutephase and follow-up [185,186]. If patients are seronegative, histo-pathology may be required, which shows a dense, diffuse lym-phocytic infiltrate with plasma cells throughout the dermis, rarelyextending into the epidermis [186]. Frequently, though not invari-ably, there are prominent lymphoid follicles with germinal centersthat e unlike other B-cell pseudolymphomas e typically lack amantle zone [186]. If untreated, borrelial lymphocytoma can persistfor months. With appropriate antibiotic treatment resolutiongenerally occurs within 3e4 weeks, but may take considerablylonger when diagnosis and treatment are delayed [184,185].

4.6. Lyme neuroborreliosis (LNB)

LNB most commonly develops within a few days to 3 monthsafter infection and can be the presenting and the only

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 9

manifestation of Lyme disease [28,187e189]. Therefore, the onset ofearly LNB occurs most frequently in June/July to October/November, depending on the latitude [187,188,190e192]. Approxi-mately 10e15% of patients with untreated EM will developneurological sequelae [5,193]. Note, however, that when patientsare closely monitored and their earliest manifestations of Lymedisease are rigorously treated, as in two large US vaccine trials, theproportion of cases presenting with LNB can be as low as 2% or less[162,194]. Among the cases of Lyme disease reported to the CDC,12e14% have LNB [35]. Generally, LNB is the presenting manifes-tation in 16e23% of European patients with Lyme disease[51,53,163,195], although considerably lower rates have been re-ported in some regions [52,54]. All of the available epidemiologicaldata show the highest rates of LNB in children, followed by adults�50 years of age [51,54,187,196]. A very consistent finding is amale:female ratio of 1.5:1 among European LNB patients[187,188,191,196,197].

The classical triad of neurological manifestations in Lyme bor-reliosis, as originally described in Europe before knowledge of theetiological agent [198,199], consists of lymphocytic meningitis,cranial neuritis and painful radiculoneuritis alone or in variouscombinations. Although any cranial nerve can be affected, facialpalsy is the most common cranial neuropathy. This triad has beenobserved in North American patients, where lymphocytic menin-gitis with headache and stiff neck is most prominent, oftenaccompanied by mild encephalitis, whereas radiculoneuritis is lesspronounced [200,201]. Cranial neuritis is seen in about one half ofcases. Of note, among the cases reported to the CDC between 2001and 2010, Bell's palsy represented 9%, radiculoneuropathy 4%, andmeningitis/encephalitis 1%, respectively [159,202]. In contrast, inEuropean LNB, severe radiculitic pain that frequently worsens atnight is a common presenting symptom and is eventually reportedby 70e90% of adult patients [172,187,188]. CSF lymphocytic pleo-cytosis is present in the vast majority of European patientsdescribed to date [172,187,188,196], and now is a diagnostic crite-rion for definite LNB [203]. However clinical signs of meningitis arerare in adult patients [187,188]. Cranial neuritis affects 40e50% ofadult patients [51,172,187,188,196], with more than 80% of thesepatients having facial palsy, which is bilateral in over one third ofadults [187,188,200,201]. Paresis of other peripheral nerves canaccompany cranial neuropathies or occur in isolation. In a Europeanstudy, most patients with EM and moderate to severe symptomssuggestive of neurologic involvement (such as headache, stiff neck,vertigo, memory or concentration problems, sleep disturbances, orparesthesias) did not have laboratory evidence of infection and/orinflammation of the central nervous system (CNS) [145]. On theother hand, radicular pain, meningeal signs, and peripheral facialpalsy along with larger EM size and seasonal occurrence (onsetfrom May till August) were significantly associated with the pres-ence of lymphocytic pleocytosis, which was in turn predictive ofmicrobiologically proven LNB. Symptoms of CNS involvement suchas myelitis or encephalitis are rare in early LNB [187,188].

The characteristics of LNB in children differ from those seen inadults, with less frequent and less severe radicular pain, morefrequent headache and clinical signs of meningitis, while the fre-quency of facial palsy is often higher, but bilateral facial palsy is rare[172,187,189,190,196,204,205]. As a result, facial palsy and menin-gitis are the most frequent clinical manifestations of childhood LNB[189,190,192,206,207]. Lyme disease can be the most commonidentifiable etiology of facial nerve palsy in children in someendemic areas of the US [208] as well as Europe [189,191,209],particularly duringMayeOctoberwhen the vastmajority of cases ofLNB present [190]. Importantly, LNB in children may be quiteinsidious with slight meningeal signs, but otherwise mostly non-specific signs and symptoms such as headache, intermittent low-

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

grade fever, loss of appetite and weight loss [192,205]. Rarely,children with meningitis may develop raised intracranial pressureand present with the typical clinical picture of pseudotumor cerebri[189,210].

There are also late manifestations of LNB. An encephalopathythat arises months or evenmany years after the initial infection hasbeen described mainly in US patients many of whom had not beentreated or had received delayed or inadequate treatment [211,212].These patients had objective evidence of slight cognitive dysfunc-tion and memory impairment along with sleep disturbances andmood changes. Some had evidence of CNS infection, includingintrathecal Bb-specific antibody production, i.e., a positive anti-Bbantibody index (AI), increased CSF protein levels or, more rarely,CSF pleocytosis [211e213], and their disease may have representedmild forms of encephalomyelitis. However, most of the patientsdiagnosed with Lyme encephalopathy did not have evidence of CNSinflammation and it has been suggested that they had a “toxicmetabolic” encephalopathy similar to that seen in other viral,bacterial or autoimmune inflammatory states [214]. In addition,chronic distal axonal polyneuropathies, manifesting mainly asintermittent distal paresthesias, have been described in US patients[211,215]. A similar distal neuropathy is seen almost exclusively inconjunction with ACA in Europe [176,179,216]. Another late disor-der seen primarily in Europe is progressive encephalomyelitis,which manifests with cerebellar ataxia, cognitive deficits, spasticbladder paresis, and para and tetraparesis, quite frequently inconjunction with cranial nerve palsies [187,188,217]. In addition,rare cases of chronic meningoencephalitis, meningomyelitis, andradiculomyelitis have been described [187,188].

The clinical diagnosis of LNB is highly likely when a patientpresents with classical triad of lymphocytic meningitis, cranialneuritis and painful radiculoneuritis, or with one or more of thesemanifestations in combination with a well-documented EM. Bilat-eral facial palsy, as seen in a considerable portion of adult patientswith LNB, is rarely seen in facial palsy of other etiologies[187,188,200,201]. Nonetheless, the definite diagnosis requireslaboratory confirmation, particularly when the symptoms ofpossible LNB are less characteristic. In the US, serum anti-Bb anti-bodies are considered to be adequate for this purpose inmost cases.Patients with LNB are usually seropositive at presentation or se-roconvert during the ensuing weeks [187,218]. When the CNS isinvolved, CSF analysis should show lymphocytic pleocytosis,elevated protein levels, and a positive intrathecal AI. In the case ofLyme encephalomyelitis, demonstration of a positive intrathecalanti-Bb AI is necessary to meet the CDC case definitions for sur-veillance. There have been no recent cohort studies of LNB in theUS, much less any recent systematic analyses of the sensitivity andspecificity of intrathecal AI and other CSF results. Therefore, the roleof CSF analysis in the diagnosis and management of otherneurological manifestations of Lyme disease remains uncertain[214,219e221].

Some European investigators have expressed considerableskepticism regarding the broad spectrum of neurological manifes-tations that have been attributed to Bb in the US without stringentdocumentation of specific intrathecal antibody production or evenpleocytosis [187,222]. Evidence of a positive Borrelia-specific anti-body index has long been the “gold standard” for the diagnosis ofLNC in Europe. It may be detectable in some patients with signs andsymptoms of LNB who still lack detectable specific antibodies inserum [129,144,188,223]. Unlike Borrelia-specific serum antibodies,which are present in a sizable subset of the general population[136,138,139], a specific AI in CSF is less likely to represent anincidental finding, at least in patients without a prior history of LBN.However, it may still be absent very early in the disease course[187,188,223,224] and may continue for months or years after

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3410

successful antibiotic therapy [225,226], and even for decades insome untreated patients [222]. Unfortunately, patients often seekalternative therapies, including herbals and a variety of over-the-counter medications; there is no basis for the use of these in pa-tients with Lyme disease [227].

The guidelines published by the European Federation ofNeurological Societies (EFNS) distinguish between definite andpossible LNB [203]. Cases of definite LNB have to fulfill thefollowing three criteria: 1) neurological symptoms suggestive ofLNB, 2) CSF pleocytosis, and 3) intrathecal production of Bb-specificantibodies in CSF. For possible LNB, two of these three criteria haveto be fulfilled; and if criterion 3) is lacking, Borrelia-specific anti-bodies have to be detectable in serum after a duration of 6 weeks.Patients with i) peripheral neuropathy, ii) a clinical diagnosis of ACAand iii) Borrelia-specific serum antibodies also fulfill the criteria fordefinite diagnosis.

Some cases of facial palsy or other neurological manifestationsof Lyme disease that occur in conjunctionwith, or following shortlyafter, physician-documented EM may lack evidence of specificintrathecal antibody production, pleocytosis or both [188]. Someclassify such cases as LNB [188,191,197], and this might help avoidlumbar punctures in children [172]. It can even be argued thatlumbar puncture is unnecessary in European patients because itsresults do not influence treatment decisions [221] since it has beenshown that European patients with LNB can be treated with thesame regimen of oral doxycycline as is used for EM alone [228].

4.7. Cardiac manifestations

Carditis is a manifestation of early disseminated disease, pre-senting within days and up to threemonths after the onset of EM orother signs and symptoms of early Lyme disease, but it can be thesole presenting feature [229e231]. While early prospective studiesfrom an endemic area in the US revealed cardiac abnormalities in4e8% of patients with Lyme disease some of whom were system-atically evaluated for cardiac involvement [4,5,174], the incidencerates are much lower in more recent adult and pediatric cohorts[160,164,232,233]. According to the CDC case definitions only high-grade (second or third degree) atrioventricular (AV) conductiondefects represent notifiable cases of Lyme carditis, and theserepresent �1% of cases of Lyme disease reported to the CDC from2001 to 2010 [159]. Even though the European case definitions ofLyme cardioborreliosis are more comprehensive (see Table 3), �1%of patients presented with carditis in epidemiological studies invarious endemic regions in Europe [51,53,54,163]. Similarly lowincidence rates of cardiac abnormalities have been reported fromNortheastern China [173,234]. Lyme carditis affects primarily sub-jects in their 20se40s, with men outnumbering women by at least3:1 [230], while only a moderate male preponderance wasobserved among pediatric patients [231]. Details of what has beencoined the autoimmune side of heart disease has been reviewedelsewhere [235,236].

The most commonmanifestation of Lyme carditis is acute onset,rapidly fluctuating AV conduction disturbances [229e231,237,238].Electrophysiological studies show that different levels of the AVconduction system can be affected, but most commonly the block isabove the bundle of His, frequently within the AV node [229,230].Among the cases reported to date, almost half progressed to com-plete heart block, at least 20% had second grade AV block, and atemporary pacemaker was required in about one-third of thesepatients, whereas implantation of a permanent pacemakerwas rare[230,231,237,239]. In addition, myocarditis, pericarditis, endo-carditis, pericardial effusion, prolongation of the corrected QT-interval, ST-T wave changes and, in rare cases, congestive heartfailure have been reported. While patients with Lyme carditis can

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

be completely asymptomatic, patients with high-grade AV blockusually show symptoms such as light-headedness, syncope or nearsyncope, shortness of breath, palpitations, and/or chest pain.Among pediatric patients with early disseminated Lyme disease,the lack of cardiopulmonary symptoms was found to be highlyspecific (99%) for the absence of carditis [231]. AV conduction dis-turbances can fluctuate and worsen rapidly, and patients with first-degree block and a PR interval �300 ms are at increased risk ofprogression to complete heart block [229,231]. Because of thepotentially fatal course of Lyme carditis [240e243], such patientsneed to be hospitalized and closely monitored.

Lyme carditis should be suspected in younger subjects withsymptoms of cardiac involvement in the absence of other apparentrisk factors and with a plausible history of exposure to Ixodes ticks.Patients with carditis are usually seropositive at presentation orseroconvert soon thereafter [231,239,240].

The potential role of Borrelia spp. in dilated cardiomyopathyremains uncertain. In 1990, Stanek et al. [244] reported the isola-tion of Borrelia spirochetes from, and their visualization in, endo-myocardial biopsy tissue of a patient with dilated cardiomyopathywho was strongly seropositive for antibodies against Bb. Morerecently, 24% of endomyocardial biopsy specimens from 41 Czechpatients with recent-onset dilated cardiomyopathy yielded positivePCR results for Borrelia spp. compared to none of the controls withend-stage coronary artery disease [245]. However, this was notreplicated in Dutch patients, and conflicting results also have beenobtained regarding an increased seroprevalence of Borrelia infec-tion among patients with dilated cardiomyopathy [246].

4.8. Lyme arthritis

If left untreated, ~60% of patients with EM develop briefrecurrent episodes of arthritis, often preceded and accompanied byarthralgias and myalgias [4]. Patients with EM would be treatedwith antibiotics today and would rarely if ever develop arthritis.Therefore, patients diagnosed with LA today must come mainlyfrom those subsets of patients whose EM goes unnoticed or un-diagnosed or who do not develop EM or any other typical earlymanifestations of Lyme disease. Approximately one-fourth ofadults and between 16 and 37% of childrenwith LA recall a tick bite[172,247e250]. More importantly, almost 30% of adults give ahistory of EM [172], even in case series recruited within the lastdecade [247], while the proportion in children has ranged between4 and 18% in recent cohorts [247,251,252]. According to data from alarge vaccine trial in which participants were very closely moni-tored, 16% of patients with Bb infection have only systemic symp-toms (primarily headaches and arthralgias) [253] that would notlikely be recognized as Lyme disease by the patient or the treatingphysician. An additional 7e11% of the participants show asymp-tomatic seroconversion [68,254]. Even under the worst-case sce-nario that 60% of these two patient subgroups develop LA, onewould expect arthritis to be the presenting manifestation in only~16% of patients with Lyme disease in the US. Rather surprisingly,patients with LA continue to represent ~30% of the cases reportedto the CDC [159]. In contrast, the observed rate in large case seriesof adults and children with Lyme disease has been �6% [160,161].In other words, the majority of cases of LA in the US would seem tobe preventable. LA constitutes 4e8% of cases of Lyme borreliosis inEuropean epidemiological studies [51,53,54,163], although the re-sults of a Swedish study suggest that LA may be underdiagnosed[255]. While Lyme disease overall affects men and women withsimilar frequency, up to 75% of adults and 70% of children with LAare male not only in the US [4,247,248,250,251,256e258], but alsoin Europe [170,172,249,259,260].

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 11

LA can develop within days or weeks after EM or other mani-festations of early Lyme disease in both adults and children, butusually is a late manifestation that appears months or even yearsafter infection in untreated patients [2,4,53,256,261]. Therefore,onset of LA can be observed throughout the year [247e249,259]. LAis characterized by brief, but recurrent, attacks of monoarticular orasymmetric oligoarticular swelling and pain that generally affectlarge joints, most commonly the knee, or episodes of migratorypolyarthritis involving large and small joints [4,249,256,258e265].In early series, the temporomandibular joints were also quitefrequently involved [1,4], but this has rarely been described sincethen. While the attacks of arthritis eventually subside in manyuntreated patients, some develop chronic synovitis and, in rarecases, have evidence of erosions and permanent joint destructionand dysfunction [4,265]. This erosive arthritis histopathologicallyresembles rheumatoid arthritis [266,267]. With the notableexception of a European pediatric cohort [249], the affected jointsare usually painful, but the pain is often surprisingly mild consid-ering that the effusions are frequently large [1,252,256,258]. Rangeof motion is limited in the majority of patients and a considerableportion of patients is unable to bear weight on the affectedjoint [248,249,252,264]. Approximately one third of patientspresent with fever or give a history of recent fever[247,248,251,252,259,262]. Fatigue and malaise are fairly commonsymptoms, but other systemic signs and symptoms are rare in pa-tients with LA [1,4].

In accordance with arthritis mostly being a late manifestation ofLyme borreliosis, patients with LA generally are strongly seroposi-tive for Borrelia-specific IgG [121,131,247,252,258]. Since the anti-body repertoire expands over time, such patients usually displayIgG reactivity with numerous Western blot bands [121,131,258].Occasionally, however, LA presents shortly after the onset of EM [4],and serology may still be negative [268]. In such cases, amplifica-tion of spirochetal DNA from joint fluid or tissue is up to 85% sen-sitive and invaluable in supporting the diagnosis [117,118,268],whereas culture of Borrelia spp. from synovial fluid or tissue hasvery rarely been successful [112,113].

Markers of inflammation are elevated in peripheral blood andsynovial fluid [248,249,252,263,269], but may be in the normalrange in up to ~20% of patients [160,258,262]. None of these find-ings are specific for LA. Radiographs of the affected joint(s) mayshow effusions and soft tissue swelling, but are otherwise normal[4,249,269]. The differential diagnosis of LA includes primarily ju-venile idiopathic arthritis and septic arthritis; in all cases there arecommon cytokine, chemokine and cognate receptor alterations[270]. The ability to differentiate between LA and septic arthritiswould spare patients with LA unnecessary surgical irrigation anddebridement. Unfortunately, attempts to find predictors haveyielded inconsistent results [248,252,264].

5. Treatment

5.1. Treatment guidelines

The manifestations of Lyme disease are routinely treated withantibiotics; therapy hastens the resolution and largely prevents thedevelopment of other disease manifestations. The classes of anti-biotics that have shown the greatest effectiveness against Borreliaspirochetes is b-lactams (in particular cephalosporins) tetracyclinesand, to a lesser extent, macrolides. The best treatment approach, inparticular the duration of therapy, is a matter of ongoing debate. Itis quite evident that not all patients, and most certainly not allspecies or strains of Borrelia [113] respond equally to the antibioticsmost commonly used in the treatment of Lyme disease. Based onthe available evidence from randomized controlled trials (RCTs),

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

treatment recommendations have been published by the InfectiousDiseases Society of America (IDSA) [221], the American Academy ofPediatrics [271], and by a variety of national and supranationalassociations in Europe [203,272] (www.eucalb.com). As the com-parison between the guidelines published by the IDSA and theEUCALB in Table 4 illustrates, the approaches to therapy are largelysimilar on both sides of the Atlantic, but there are some minordifferences in the recommended dosage and treatment duration.Note that the International Lyme and Associated Diseases Societyhas also published guidelines for the management of Lyme disease[273]. These advocate more aggressive and longer treatmentcourses for patients with persistent symptoms or refractory dis-ease, but much of the terminology is poorly defined and the cited“evidence” is largely restricted to older studies that support such anapproach and ignores much of the other available evidence. Theresults of several RCTs and a large retrospective study do not showany benefit in extending the duration of treatment with doxycy-cline from 10 days to 15 or 20 days in patients with EM [274e276].Retrospectively obtained data suggest a similar conclusion foramoxicillin [276]. In another double-blind RCT, treatment of pa-tients with disseminated Lyme borreliosis with a 3-week course ofi.v. ceftriaxone followed by 100 days of amoxicillin did not improvethe outcome compared to a 3-week course of ceftriaxone alone[277]. These findings do not exclude that some patients may benefitfrom longer or more aggressive therapy, but the risk-benefit ratiohas to be carefully weighed for each individual patient.

5.2. Antimicrobial prophylaxis after a tick bite

Due to the low rate of infection after a tick bite, even if the tick isshown to be positive for Borrelia spp. [55e61], treatment of a tickbite before any symptoms occur is not generally recommended inEurope [272]. However, a randomized placebo-controlled trial inthe US demonstrated the effectiveness of single dose of 200 mgdoxycycline within 72 h after removal of an I. scapularis tick inpreventing the development of EM [67]. Other manifestations ofLyme disease were not observed in this study subjects. In Russia,treatment with 200 mg doxycycline for 3e5 days after the tick bitereduced the infection rate by a factor of 11 [62]. The IDSA does notrecommend routine use of prophylaxis but states that a single doseof doxycycline may be offered provided that 1) the tick can bereliably identified as I. scapularis and has been attached for �36 h,2) prophylaxis can be givenwithin 72 h of removal of the tick, 3) thelocal infection rate of ticks is �20% and 4) doxycycline is not con-traindicated [221].

5.3. Antibiotic-refractory LA

LA that persists for >3 months after initiation of recommendedtreatment regimens with either i.v. antibiotics for >4 weeks or oralantibiotics for >8 weeks has been referred to as treatment-resistantor antibiotic-refractory LA [278,279]. According to the IDSA, symp-tomatic treatment with non-steroidal anti-inflammatory drugs(NSAIDs), disease-modifying anti-rheumatic drugs (DMARDs), orintra-articular corticosteroids is recommended for such patients ifPCR of synovial fluid or tissue does not show any borrelial DNA[221]. Note, however, that PCR is not 100% sensitive, hence negativePCR results do not necessarily mean the absence of spirochetes.Patients who still do not improve may benefit from arthroscopicsynovectomy, but this is not consistently successful [249].

5.4. Monitoring of Lyme carditis

Because of the rarity of Lyme carditis, the role of antibiotics in itstreatment is not fully established, particularly since Lyme carditis

A rigorous review of diagnostic criteria and treatment, Journal of

Table 4Treatment recommendations by the IDSA and EUCALB (In the Daily dose recommendations by EUCALB, “same” indicates that the same dose as recommended by the IDSA).

Condition Compound Route Adults (A),children (C)

IDSA EUCALB

Daily dose Duration(in days)

Daily dose Duration(in days)

EM, BL Doxycycline Oral A 2 � 100 mg 14 (10e21) Same 14 (10e21)Ca 2 � 2 mg/kg Same

Amoxicillin Oral A 3 � 500 mg 14 (14e21) 3 � 500e1000 mg 14 (10e21)C 50 mg/kg in 3 divided doses 25e50 mg/kg

Cefuroxime axetil Oral A 2 � 500 mg 14 (14e21) Same 14 (10e21)C 30 mg/kg in 2 divided doses 30e40 mg/kg

Penicillin V Oral A 3 � 1.0e1.5 Mio 14 (10e21)C 0.1e0.15 Mio/kg

Azithromycin e not recommendedas first line therapyb

Oral A 2 � 500 mg 7e10 2 � 500 mg 1st day1 � 500 mg Next 4 days

C 20 mg/kg 1st day10 mg/kg Next 4 days

Multiple EM As for EM above As for neuroborreliosisLNB Ceftriaxone i.v. A 2 g 14 (10e28) Same 14 (10e30)

C 50e75 mg/kg 50e100 mg/kgPenicillin G i.v. A 18e24 Mio in 4 divided doses 20 Mio 14 (10e30)

C 0.2e0.4 Mio/kg in 4 divideddoses

0.25e0.5 Mio/kg

Cefotaxime i.v. A 2 g every 8 h “Also effective”C 150e200 mg/kg

(Doxycycline may be adequate)d oral 2 � 100e200 mg (10e28) 2 � 100 mg or 1 � 200 mg 21 (14e30)Arthritis Doxycycline Oral 2 � 100 mg 28 Same 21 (14e30)

Amoxicillin Oral A 3 � 500 mg 28 3 � 500e1000 mg 21 (14e30)C 50 mg/kg in 3 divided doses 25e50 mg/kg

Cefuroxime axetil Oral A 2 � 500 mg 28C 30 mg/kg in 2 divided doses

Ceftriaxone i.v. A 2 g 21 (14e30)C 50e100 mg/kg

ACA Ceftriaxone i.v. A 2 g 21 (14e30)C 50e100 mg/kg

Doxycycline Oral 2 � 100 mg 21 (14e30)Amoxicillin Oral A 3 � 500e1000 mg 21 (14e30)

C 25e50 mg/kgCardio-

borreliosisParenteral antibiotic such as ceftriaxonefor hospitalized patients (as in LNB);followed by an oral antibiotic forcompletion of therapy (as in EM)

As for arthritis

c In Europe, azithromycin is primarily considered as an alternative treatment for patients who should not take doxycycline and are allergic to penicillin, such as children andpregnant or breast-feeding women.

a Doxycycline is not recommended for children <8 years of age or pregnant or breastfeeding women.b Macrolide antibiotics are less effective than other antimicrobials and should be reserved for patients for whom amoxicillin, doxycycline and cefuroxime axetil are

contraindicated.d In a non-inferiority RCT, oral doxycycline was shown as effective as i.v. ceftriaxone for the treatment of European LNB [228], confirming the results of earlier open-label

studies [221], but it is not yet clear whether this extends to North American patients with neurological manifestations.

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3412

regresses within one to six weeks without antimicrobial therapy[229]. Nonetheless, antibiotic treatment is recommended becausespirochetes have been visualized in endomyocardial biopsy speci-mens with various staining methods [231,280] or detected by PCR[240,241] and because carditis usually occurs at an early stage ofLyme disease when antibiotic treatment can prevent other diseasemanifestation. Carditis is a potentially life-threatening manifesta-tion of Lyme disease, and several fatal cases have been reported[240e242], one in a patient coinfected with Bb and Babesia microti[243]. Therefore, and since AV conduction disturbances can fluc-tuate and worsen rapidly it is highly advisable to hospitalize andclosely monitor patients with second or third degree block or firstdegree block and a PR interval �300 ms.

6. Outcome

6.1. Erythema migrans (EM)

In RCTs, EM usually resolves within a few days or weeks afterinitiation of treatment, although the process may take up to 8weeks [275,281e285]. Non-specific signs and symptoms, such as

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

arthralgias, myalgias, fatigue, headache, irritability, stiff neck andparesthesias, often last longer, but their number and severitysteadily decreases over the following months [66,275,281,286]. Byone year after initiation of treatment, between 70 and >90% of USpatients return to their pre-Lyme disease health status[80,282,283,287e289], while rates of 84e98% have been reportedfrom Europe [66,275,286,290,291]. The prognosis in pediatric pa-tients also seems to be very good [160,292]. Treatment failures dooccur, and while major extracutaneous manifestations of Lymedisease often develop during or shortly after treatment, some occurmonths after therapy [160,274,281,283e285,289,290].

6.2. Lyme neuroborreliosis (LNB)

In many cases, the signs and symptoms of LNB resolve sponta-neously [189,211,222,293]. However, antimicrobial therapy not onlyhastens the resolution of symptoms [294], but also preventsobjective and subjective sequelae of LNB and the development ofother major manifestations of Lyme disease [293]. Symptoms ofearly LNB start to improve within a few days after initiation ofantibiotic therapy, and some patients recover completely by the

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 13

end of treatment [189,294e297]. However, in most cases diseaseresolution is gradual. In particular motor and sensory deficits maybe slow to improve [228,294,295,298]. Some patients may requireretreatment because of insufficient improvement or even wors-ening. Generally, between 70 and 85% of patients with LNB make acomplete recovery, usually within 6e12 months after initiation oftherapy [196,296e302], and this rate may reach 90% in patientswith facial palsy [303]. The results of long-term follow-up studiessuggest that there is little further improvement after 1 year[197,304].

Residual neurologic findings can be documented in 5e28% ofpatients �1 year after therapy, including remaining facial nervedysfunction, other pareses, radiculopathies, vision or hearing loss,ataxia and various other findings [197,298,300,304e306]. Whilethey are generally considered to be mild to moderate, more thanhalf of the LNB patients identified in a population-based study inSweden felt that the neurologic sequelae persisting 5 years after theacute illness restricted their daily activities and almost one fifth hadnot been able to resume their pre-Lyme disease occupation [197]. In26 Swedish patients with CNS involvement with a median follow-up of 6 months (1e18 months), four patients (15%) had permanentdisabilities that seriously affected their daily lives, namelysphincter dysfunction in 2, unilateral hearing loss in 1, and nearcomplete unilateral peripheral facial palsy in 1 [307]. Patients whohad had LNB 30months previously also reported significantly lowerhealth-related quality of life (HrQoL) as assessed by the physicaland mental component of the Short-Form 36 (SF-36) and scoredhigher on the Fatigue Severity Scale (FSS) compared to matchedcontrols [305]. Their depression and apathy scores were also higher,but both patients and controls had low scores overall. Predictors ofthe physical health (the physical component summary score ofthe SF-36) were pretreatment symptom duration >6 weeks andincomplete recovery at 4 months, while the mental componentsummary score was predicted by incomplete recovery at 4 months.Almost half of the variation in fatigue (assessed by the fatigueseverity scale) was explained by pretreatment symptom duration>6 weeks, higher pretreatment clinical scores and non-recovery at4 months [304]. Others similarly found an association betweenpretreatment symptom duration and persistence of symptomspost-treatment [196], but particularly of remaining objectiveneurologic sequelae [197,298].

The complete resolution rate in children with facial nerve palsyranges between 80 and 96%, depending to some extent on thethoroughness of the assessment and the length of follow-up[308e312]. Of note, among pediatric patients who had beentreated for Borrelia-associated facial palsy 3e5 years earlier and hadall declared themselves completely cured, approximately one-halfstill had subjective feelings of facial dysfunction and a similarproportion had objective evidence of mild to moderate facial nervedysfunction upon neurophysiological testing, but there was littlecorrelation between the two [311]. Such patients may have diffi-culties with closing their eyes or pronouncing words, cosmeticcomplaints and social problems [310,313]. More limited data areavailable on the long-term outcome of pediatric LNB overall andthese are highly variable, indicating between 8 and 27% of patientsdo not recover completely [197,204,313,314]. In addition to residualfacial nerve dysfunction, such children can have objective evidenceof other neurologic sequelae that can be sensory, motor or both incharacter [313]. Unlike adult LNB, pre-treatment symptom durationwas not associated with outcome in pediatric patients [204,313].

6.3. Lyme arthritis

LA eventually resolves spontaneously, but this may take years,with one child having been reported to have attacks of arthritis in

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

the 7th and 8th year after onset [4,261]. Resolution can be hastenedconsiderably with antibiotic therapy [315]. The majority of patientswith LA require only a single course of antimicrobial therapy, butthis proportion varies between 48 and 90% for US adults [247,278]and between 52 and 95% for US children [247,250,251,256,258,316].Similar ranges have been reported from Europe [259,260,317].Resolution of LA usually occurs gradually over the course of severalmonths after a 4-week course of oral antibiotics [278]. Therefore, itis advisable towait for threemonths before retreatment [221]. Evenif not all physicians heed this recommendation, it is obvious thatsubstantial numbers of patients need to be re-treated because ofinsufficient response to a single course of antibiotics, and some ofthose patients fail to respond even to a second or third course ofantibiotics [249,278,316,318]. Such antibiotic-refractory LA (per-sisting for >3 months after initiation of recommended treatmentregimens) has been estimated to occur in approximately 10% ofadult US patients [278,319], but this proportion may actually beconsiderably higher [247]. Some data, including the results of arecent direct comparison between adults and children [247], sug-gest a more benign prognosis for pediatric LA in the US[250,258,262,263]. However, this is not confirmed in other in-vestigations [251,316], one of which found that 39% of pediatricpatients took >6 months and 13% took >12 months for diseaseresolution, and the last patient was free of arthritis after ~19months [316]. Unfortunately, there was very limited information onthe number of courses of antibiotics and other treatment modal-ities in these children. Similarly, LA is refractory to antibiotics in aconsiderable portion (up to 23%) of children and adolescent fromsome European countries, and 16% of patients still had ongoingarthritis 10 months to 1 year after initiation of therapy [249,320].Lyme arthritis eventually resolves in all patients, though it may takemonths or years and may require NSAIDs, DMARDs, intraarticularcorticosteroid injections, or synovectomy.

No predictor of antibiotic responsiveness could be identified inpediatric cohorts [251,316]. It remains uncertain whether intra-articular steroid therapy prior to antibiotic treatment is associatedwith a poorer response [259,278,321]. Cumulative duration of ep-isodes of arthritis, but not the overall pre-treatment symptomduration, was associated with non-responder status [278]. Inaddition, the HLA-DR4 specificity [278], particularly in conjunctionwith serological reactivity to the outer surface protein A (OspA)antigen [279], and more specifically HLA-DRB1*0401, DRB1*0101and related alleles have been associated with treatment-resistantLA [319]. There is also no available data on the role, if any, of epi-genetics in Lyme disease [322].

There are few data on residual symptoms after antibiotic ther-apy in patients with LA. In a prospective population-based study ofpediatric LA, none of the 90 patients had active arthritis a median of7 years after diagnosis, but four reported mild to moderateimpairment in school and sports activities due to continuingmusculoskeletal complaints [256] and such complaints have alsobeen reported to persist in ~8% of European patients [260,320].

6.4. Post-Lyme disease syndrome

In RCTs and prospective cohort studies, residual subjectivesymptoms after antibiotic therapy are reported by 3e27% of pa-tients with EM [75,80,160,274,281,283,289] and 10e40% of patientswith LNB [197,298,300,304e306,323]. Such information is largelylacking for patients with LA. The most common symptoms after EMalone are arthralgias, myalgias, fatigue, headache, irritability, stiffneck and paresthesias [197,298,300,304e306,323]. Problems withmemory and concentration may be other prominent symptoms insome patients with a history of Lyme disease [324e326], and thesealong with fatigue, pain and paresthesias also are the most

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3414

common symptoms reported by patients with a history of LNB[197,298,304,327]. It should be kept in mind, however, that most ofthese symptoms are quite common in the general population.Conflicting results have been obtained in studies addressing thequestion of whether these symptoms are more frequent or moresevere in patients with a history of Lyme disease compared tohealthy controls. Compared to matched controls, no significantdifferences were seen in European patients with EM [275,286],Swedish children with a history of LNB [204,313], or in cohorts ofUS patients with a history of any type of Lyme disease [293,328]. Incontrast, others found a significantly higher frequency of subjectivesymptoms in pediatric and adult patients with a history of LNB[305,323,329] or of any manifestation of Lyme disease [324,330]and significantly poorer global health and functional status[305,324,330]. Thus, it currently remains unclear towhat extent thesymptoms persisting after antibiotic treatment are a direct or in-direct consequence of Lyme disease.When specifically asked, only aminority of patients consider Lyme disease to be the cause of theirsymptoms and difficulties with activities of daily living[81,328,329].

Patients with a history of LNB not only have subjective com-plaints of memory problems, but assessment of neuropsychologicalfunction �30 months after treatment revealed deficits particularlyin verbal memory, but also in visual memory, attention/executivefunction, and processing speed in adult patients compared tohealthy controls [302,306], though not in pediatric patients with ahistory of Lyme facial palsy [329]. In other patient cohorts, there isoften little correlation between subjective perception and objectiveneuropsychological evidence of memory deficits [325,330]. Yet,numerous studies have shown that a subset of these patients hasobjective evidence of memory impairment, particularly on verbaltasks, compared to healthy controls or patients who recoveredcompletely [324,325,331e336]. However, this is not an entirelyconsistent finding [293,330,337,338] and comparison to diseasecontrols has also yielded conflicting results [324,325,332e334,336].A variety of other domains are affected less consistently. In contrastto adults, adolescents exhibited greater impairment of visualmemory than of verbal memory [339].

The subjective symptoms and objective cognitive deficits per-sisting in some patients with Lyme borreliosis resemble what hasbeen described in untreated or inadequately treated US patientswho developed a subtle encephalopathy years after the onset oftheir disease [211,212,333,340,341]. Only some of these patientshad evidence of CNS infection or inflammation such as intrathecalantibody production or elevated CSF protein concentrations[211,213,340,342]. Kaplan et al. [343] considered such patients tohave encephalopathy if they had abnormal CSF results or a positivePCR reaction for Bb DNA, and to have post-Lyme disease syndrome(PLDS) if CSF analysis yielded normal results. Since then, the defi-nition of PLDS (or post-treatment Lyme disease syndrome or“chronic Lyme disease”) has been expanded considerably and theseterms are now used to describe patients who have any kind ofpersistent symptoms after what is considered adequate antibiotictherapy for their manifestation(s) of Lyme disease. Here again arigorous evaluation is indicated to be certain there are not othercauses, i.e., to avoid mislabeling a patient as having Lyme disease.

Unfortunately, there is no standardized case definition for PLDS,and this may account for some of the discrepant results obtained instudies of cognitive function after Lyme disease. The IDSA hasproposed such a definition, which requires 1) a documentedepisode of early or late Lyme disease fulfilling the CDC case defi-nitions for surveillance purposes, 2) resolution or stabilization ofthe objective manifestations of Lyme disease following a generallyaccepted treatment regimen, and 3) onset of symptoms within 6months of the diagnosis of Lyme disease and persistence of

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

continuous or recurrent subjective symptoms of fatigue, wide-spread musculoskeletal pain, or complaints of cognitive difficultiesfor �6 months after completion of antibiotic therapy [221]. Afurther requirement is that these symptoms are of sufficientseverity to result in a decline of physical and/or social functioning.There are also a variety of exclusion criteria intended to insure thatthe patient does not have coinfections, objective manifestations ofLyme disease or other undiagnosed or previously diagnosed un-derlying diseases or conditions that would explain the patient'scomplaints. The challenge will be to find, and agree upon, the toolsfor assessing symptoms and their functional impact before thesecase definitions can be considered useful in unraveling the inci-dence, nature, and pathogenesis of PLDS [344].

6.4.1. Treatment of PLDSThere have been several randomized, double-masked, placebo-

controlled trials of antibiotic retreatment in patients with persis-tent symptoms after what is considered appropriate therapy fortheir manifestations of Lyme disease [326,336,345,346]. The in-clusion criteria and other basic characteristics of the three trialsthat used i.v. ceftriaxone for the retreatment and included anintention-to-treat analysis are summarized in Table 5. In the studyby Krupp et al. [345], the improvement in fatigue was similar in thetwo groups at 1 month, but sustained at 6 months only in thosepatients who received i.v. ceftriaxone, resulting in a significantdifference at 6 months. Cognitive function was mildly impairedcompared to normative data in both groups andwas not affected bytreatment. Fallon et al. [336] found that the ceftriaxone group hadsignificantly greater generalized improvement in cognitive func-tion at 12 weeks, but this was not sustained at 24 week. In patientswith greater symptom severity at baseline, ceftriaxone was asso-ciated with significantly greater improvement in physical func-tioning and current pain at 12 and 24 weeks, and in fatigue at 12weeks only. The effectiveness of antibiotic retreatment for fatigueseen in the Krupp study [345] was confirmed in a post-hoc analysisof the Fallon data [336,347]. There was no significant treatmenteffect on any of the outcome measures in the trials conducted byKlempner et al. [326,338]. In all three studies, treatment wasassociated with several serious or even life-threatening adverseevents.

Based on these results, several guidelines contain the conclusionthat further antibiotic therapy of patients with subjective symp-toms that persist for �6 months after treatment with recom-mended antibiotic regimens is “not useful” [221] “ineffective”[203], or has “not shown sustained benefit” [272]. These state-ments are false as written since two of these trials did show asignificant and sustained benefit in at least a subset of patients[336,345]. They also bring up the question of what the precise ex-pectations were from repeated antimicrobial treatment in patientswho had already been treated with multiple and prolonged coursesof antibiotics. Did anybody truly expect yet another course of cef-triaxone would cure patients who had failed to respond toextended antibiotic therapy? Be that as it may, the interpretation ofthese results and the question of whether or not patients withdisabling subjective symptoms should be retreated with (yetanother course of) antibiotics remains the subject of intense con-troversy [347e350]. What seems to be lacking from this discussionis the well-known fact that antibiotic therapy can induce consid-erable disturbances of the intestinal microflora. Indeed, there hasbeen no study of the microbiome in patients with Lyme diseasedespite increased evidence of the importance of the microbiome inimmune mediated pathology [351,352]. An intact microflora hasimportant functions in preventing the colonization of the gastro-intestinal tract by pathogenic bacteria and in providing continuedstimulation to the gut-associated immune system and thereby

A rigorous review of diagnostic criteria and treatment, Journal of

Table 5Characteristics of three placebo-controlled randomized controlled trials of antibiotic retreatment in PLDS.

Reference Krupp et al. [345] Fallon et al. [336] Klempner et al. [326,338] Klempner et al. [326]

n 55 37 Patients, 20 healthycontrols

78 (Seropositive) 51 (Seronegative)

Inclusion criteria 1) Age 18e70 years2) History of physician-

documented EM- or CDC-defined latemanifestations of Lymedisease confirmed bypositive ELISA andWestern blot serology

3) Completion (6 monthsbefore study entry) ofstandard antibiotic ther-apy for Lyme disease

4) Current severe fatigue(score �4.0 on a modifiedversion of FFS-11)

1) Age 18e65 years2) History of physician-

documented EM- or CDC-defined manifestations ofLyme disease and apositive or equivocalELISA confirmed byWestern blot serology

3) Current positive IgGWestern blot

4) Treatment for Lyme dis-ease with �3 weeks of i.v.ceftriaxone completed �4months before study entry

5) Subjective memoryimpairment that startedafter the onset of Lymedisease

6) Objective evidence ofmemory impairment (asassessed with the Wechs-ler Memory Scale-III)

1) Age �18 years;2) History of acute Lyme

disease acquired in the USand;a) History of single or

multiple EM, earlyneurologic or cardiacsymptoms attributedto Lyme disease, radi-culoneuropathy, or LA

3) Current positive IgGWestern blot;

4) Physician-documentedtreatment of acute Lymedisease with a recom-mended antibioticregimen;

5) Onset of chronic symp-tomsa within 6 months ofthe initial infection withBb and their persistencefor �6 months but �12years

As in seropositive patientsbut with a history ofphysician-documented EMand current negative IgGWestern blot

Treatment drug i.v. ceftriaxone 2 g/d orplacebo

i.v. ceftriaxone 2 g/d orplacebo

i.v. ceftriaxone 2 g/d for 30days followed by oraldoxycycline 200 mg/d for 60days or placebo

Same as seronegative

Treatment duration 28 days 10 weeks 30 þ 60 ¼ 90 days Same as seronegativeOutcome measures 1) Change in fatigue;

2) Change in mental speed;3) Clearance of Borrelia (as

assessed by OspA antigenin CSF)

1) Neurocognitiveperformance;

2) Physical outcome

1) Health-related quality oflife;

2) Cognitive function

Same as seronegative

Days of prior antibiotictherapy (mean except inKlempner trials)

A 44 ± 22 daysP 57 ± 64 days

An i.v. 76 ± 61A oral 240 ± 310P i.v. 58 ± 40P oral 179 ± 231

A median 71 (IQR 42e113)P median 60 (IQR 42e98)

A median 57 (IQR 33e120)P median 49 (IQR 31e83)

A ¼ antibiotic group, An i.v. ¼ previous i.v. antibiotic treatment received by the antibiotic group, A oral ¼ previous oral antibiotic treatment received by the antibiotic group.P ¼ placebo group.

a These self-reported symptoms included widespread musculoskeletal pain, cognitive impairment, radicular pain, paresthesias, dysesthesias, or profound fatigue andinterfered with the patient's functioning.

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 15

shaping the responses of the overall immune system [353].Therefore, it may be time to finally consider the role of repeated andprolonged antibiotic treatment itself in the symptoms of patientswith PLDS.

7. Pathogenesis

7.1. Animal models

Elucidation of the pathogenesis of Lyme disease is hampered bythe lack of an animal model that reproduces all the facets of humanLyme disease. The relative absence of appropriate models likelyreflects the disparity that exists between genetic susceptibility andenvironmental factors [354]. The rhesus macaque develops most ofthe manifestations seen in humans, including EM and peripheralneuropathy [355], but is not widely available and difficult andcostly to handle. Specific-pathogen-free beagle dogs with tick-transmitted Bb infection represent the most faithful model of hu-man LA because they develop recurrent episodes of mono or oli-goarticular arthritis (lameness) after an incubation period of �2months [356e358]. Rabbits are one of the few species that developEM after Bb infection but they eventually clear the infection, whileuntreated patients appear to remain persistently infected

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

[315,359,360]. A variety of smaller animals, including rats, hamster,and gerbils, have also been used. The most widely employed modelis various strains of inbred laboratory mice, but while susceptiblestrains develop arthritis and carditis, neurological manifestationsand EM are absent. Arthritis develops early after infection, resolvesspontaneously, and does not show the typical recurrent attacksseen in human patients, with the exception of mice deficient inCD28 [361]. In addition, the infiltrate in murine carditis is domi-nated by macrophages with only scattered lymphocytes[362e364], whereas lymphocytes are the major constituents of thehuman cardiac infiltrate [241,280,365], even if a case of B. afzelii-associated endocarditis with an infiltrate consisting mostly ofmacrophages, neutrophils and eosinophils has been reported [366].

Mice are most commonly infected by syringe inoculation of oneof a few select cultured strains of Bb, but cultured spirochetes ex-press a different protein profile than spirochetes transmitted byticks and individual strains of Bb differ in their pathogenicity[367e369]. In addition, syringe inoculation of cultured spirochetesneglects the suppressive effects of tick saliva on the host immuneresponse. Yet, this effect is substantial, as evidenced by the pro-found alterations in the cytokine profile of mice infested by unin-fected ticks compared to normal mice [370], the decreasedleukocyte recruitment to the skin and the increased spirochete

A rigorous review of diagnostic criteria and treatment, Journal of

Table 6Common methods for typing Borrelia spp. strains.

Loci Method Resulting groups

Intergenic spacer 16Se23S (rrserrlA)

PCR-based RFLPanalysis

Ribosomal spacer type(RST) 1e3

Intergenic spacer 5Se23S (rrferrlB)

PCR þ sequencing orPCR-based RFLPanalysis

Intergenic spacer (IGS)types

OspA (plasmid-borne) Monoclonal antibodies Serotypes 1e8ospC (plasmid-borne) PCR þ (Cold) single-

strand conformationalpolymorphism (SSCP)analysis;PCR þ Reverse line blot

Based on <2% or >8%sequence divergence,�70 groups designatedby combinations ofletters for the Borreliaspp. and numbers

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3416

burden in various tissues of mice co-inoculated with tick salivarygland extract and Borrelia spp. compared to Borrelia spp. infectionalone [371e373], and by major differences in the immune responseof mice with tick-transmitted infection compared to mice infectedvia needle inoculation [374e376]. In vitro, tick saliva or salivarygland extract can inhibit the functions of DCs, macrophages, neu-trophils and NK cells, and suppress T cell responses and humoralimmunity [377]. NK and other regulatory cells have been shown toplay a key role in autoimmunity [352]. Mice, dogs andmonkeys canbe inoculated with Borrelia spp. via nymphs that were infectedeither by capillary feeding or by placement on mice that had beeninfected by syringe inoculation [357,378e381]. Arthritis andmyocarditis develop later after infection and persist longer in tick-inoculated mice compared to needle-inoculated mice [378e380].Cystitis is seen in mice with tick-borne infection, but is rarely re-ported from needle-inoculated mice, even though their bladderalso frequently harbors high spirochete numbers [369,382]. Adrawback of this model is that the size of the inoculum cannot bestandardized.

7.2. Factors that influence pathogenesis

7.2.1. Organotropism of the different Borrelia genospeciesAs the data summarized in Table 1 show, each of the three major

pathogenic species of Borrelia can cause essentially all of the typicalmanifestations of Lyme disease. Nonetheless, there are numerousdata suggesting that each of them is associatedwith distinct clinicalmanifestations. Thus Bb shows tropism for the joint [383,384], asalso suggested by the apparently higher incidence of LA in the US,where Bb is essentially the only genospecies causing Lyme borre-liosis, compared to Europe, where Bb is relatively uncommon.B. garinii is most strongly associated with LNB [111,145,385e390],and B. afzelii appears to be overrepresented among skin isolates[20,23,24,111,390e392], in particular ACA [111,393e396].

However, there are some major shortcomings in the currentlyavailable data. Determination of the infecting species was based oncultured isolates in many of these studies, but culture of Borreliaspp. is not very sensitive, particularly in CSF samples[104,110,111,397], and even less so in synovial fluid or tissue sam-ples [112,113]. PCR-based amplification of DNA isolated directlyfrom tissue specimens generally shows greater sensitivity [114], butlargely depends on the specific PCR target [23,104], still lackssensitivity, particularly in CSF [104,115,116], and does not neces-sarily detect Borrelia spp. in all samples that are culture positive[104,108]. Furthermore, since patients with EM are now routinelytreated with antibiotics, determination of the infecting species ofBorrelia in samples from extracutaneous lesions is likely to yieldresults that are skewed towards those strains that do not also causeeasily recognizable EM lesions. Few of these studies examinedwhatis likely to be a major determinant of the probability of acquiring aninfectionwith a particular species of Borrelia, namely its prevalencein regional ticks [20,24,111,386,391,398]. The results are somewhatconflicting, most likely because of the small number of ticks and/orhuman samples examined in some of these investigations. Inaddition, tick and human samples were collected during differenttime periods and in different locations, thereby not taking intoconsideration that there may be considerable temporal andregional variation in the number of infected ticks and the distri-bution of Borrelia spp. in ticks [20,71,399,400]. Overall, the resultssuggest that B. garinii may be underrepresented in skin rather thanoverrepresented in CSF [20,24,386,391]. Note, however, thatB. afzelii infection of the nervous system may simply be under-recognized because it does not cause the typical clinical picture ofEuropean LNB associated with B. garinii infection, namely radi-culoneuritis and/or peripheral facial palsy [110]. Instead, detection

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

of B. afzelii in CSF was associated with mostly nonspecific symp-toms, and most of the patients from whose CSF B. afzelii could becultured did not fulfill the European diagnostic criteria for LNB.

7.2.2. Virulence of the infecting Borrelia strainA variety of typing tools have been developed for the study of

spirochete evolution and phylogenetics. The ones most frequentlyused for classifying strains of Borrelia species into subtypes arelisted in Table 6. The biodiversity of Borrelia spp. subtypes decreasesfrom ticks to EM lesions and further to sites of disseminated disease[391,401]. In other words, ospC, RST, or other subtypes of Bb,B. afzelii and B. garinii differ in their infectivity and ability to causedisseminated disease [26,402e408]. This may be partly mediatedby differential abilities to induce pro-inflammatory cytokines[409]; the mechanisms remain enigmatic and recent technologies,including use of microRNAs, have not yet been studied [236].Nonetheless, the available data remain somewhat controversial[410e412]. One of the reasons for these discrepancies may be theuse of spirochetemia as a marker of dissemination. Yet, the rela-tionship between hematogenous spread and disseminated diseasemay be more complex than generally appreciated. This is high-lighted by the findings that even non-disseminating strains cancause transient spirochetemia in mice [413], and that hematoge-nous dissemination is not necessarily a sign of greater pathoge-nicity, at least in European patients with Lyme disease [65,105].

This may also explain why experimental models have failed toconsistently reproduce an association between specific RST typesand disseminated disease, even though individual RST types differin their pathogenicity [367e369,380]. It is also possible that thefactors determining dissemination differ between humans andlaboratory mice or that the RST classification does not have suffi-cient discriminatory power to predict the ability to disseminate andto cause severe disease manifestations. Typing of ospC alone mayalso be insufficient to capture the capacity for causing disseminatedinfection, since multilocus sequencing allows grouping of Bb iso-lates into clonal complexes (many of which contain more than oneospC type) that better predict the propensity for dissemination[414]. Thus, the association between ospC major groups and inva-sive disease may be due to strong linkage disequilibrium ratherthan a direct effect of OspC [414]. This is, however, in contrast todata suggesting that ospC type is related to disseminated diseasebecause invasive and noninvasive ospC types differ in their affinityfor plasminogen, which is activated to plasmin, a serine proteasethat degrades extracellular matrix and, thereby, enables bacterialdissemination [413].

It also should be kept in mind that detection by PCR or evenisolation by culture of a particular Borrelia genotype does not proveits pathogenicity since patients may harbor more than one strain[26,404,405,415]. In addition, almost all of the typing studies were

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 17

performed on culture isolates, but culture-based genotype analysisunderestimates the genetic diversity of the infecting spirochetes.This was most clearly demonstrated by the detection of mixtures ofgenotypes in 43% of samples that were typed directly in tissuesamples compared to barely 6% of cultured samples [26]. In otherwords, isolates may be selected for their ability to grow in culturerather than for their pathogenic potential. On the other hand, dif-ferences in the results from culture-based and direct typing ofpaired patient specimens indicate that the direct method also doesnot reflect the full diversity of the infectious strains [26].

7.2.3. Host genetic factorsNone of the above studies take host factors into account. Yet,

individual strains of mice clearly differ in their susceptibility to thedevelopment of arthritis and carditis [416e419]. These and otherdata underscore that pathology is at least partly determined by hostgenetic factors in mice [420e423]. While mice with most H-2haplotypes develop moderate to severe arthritis, the H-2d haplo-type was associated with resistance [417]. However, there is evi-dence to suggest that murine susceptibility or resistance to arthritisis not attributable to the MHC itself, but is determined by genesoutside the MHC [421e425]. Rhesus macaques from separate col-onies differ in the frequency with which they develop certainmanifestations of Lyme disease, such as myositis or peripheralneuritis [426]. This suggests that in this outbred animal model,genetic host factors also play a role.

There are indications that the differential ability for rapidspirochete clearance, thereby limiting the pathogen burden in thejoint, is an important and probably genetically determined factor indecreasing the susceptibility for Bb-induced arthritis in certainmouse strains after infection via syringe [378,420,421,427] or tickbite [428]. Yet, other protective mechanisms must be in play inC57BL/6 mice (B6), which develop only mild arthritis despitespirochete numbers in the joint similar to those seen in arthritis-susceptible C3H mice [421]. Conversely, IL-10-deficient mice havea significantly lower tissue spirochete burden, yet develop moresevere arthritis than their wild-type counterparts [429]. In US pa-tients, no association was detected between the number of Bbspirochetes in EM biopsy samples and the number or severity ofsystemic symptoms [108]. Actually, the spirochete burden wasinversely associated with the size and the duration of the EM le-sion(s) [108], which have been shown to be directly correlated[164]. Therefore, these results suggest that there is a reduction ofthe number of spirochetes in the skin near the tick bite over time[108]. This could be a simple dilution effect as spirochetes spreadover a larger area of skin, but could also reflect immune-mediatedelimination of spirochetes or their dissemination into the bloodstream. In contrast, higher spirochete load (mostly of B. afzelii) inskin biopsy specimens was associated with the presence of sys-temic symptoms and of central clearing in the EM lesion of Euro-pean patients [114].

7.2.4. CoinfectionsIxodes ticks are vectors for a variety of infectious organisms

other than Borrelia spp. In North America, these include Anaplasmaphagocytophilum, the cause of human granulocytic anaplasmosis(HGA, formerly human granulocytic ehrlichiosis), B. microti, themain cause of babesiosis, and the relapsing fever spirochete Borreliamiyamotoi. These can also be transmitted by I. ricinus andI. persulcatus ticks in Europe and Asia, which additionally can carrythe flavivirus that causes tick-borne encephalitis. The existence ofsimultaneous infectionwith Bb and A. phagocytophilum or B. microtihas been firmly established by their cultivation from patient tissueswith subsequent PCR-based confirmation of their identity[430,431]. Based on these and serological methods, the reported

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

rates of human coinfection with Bb and A. phagocytophilum or B.microti vary between 2e11% and 2e40%, respectively, depending onthe case definitions, the region and possibly the year(s) underinvestigation [138,431e434]. Coinfections with all three agentshave been reported [433].

In experimental animals, simultaneous infection with Bb andA. phagocytophilum can increase the pathogen load in various tis-sues, markedly alter the cytokine pattern, result in a redistributionof splenic T cell subsets, and enhance the severity of LA, with theprecise nature and combination of effects depending on the mousestrain and the route of infection (needle inoculation versus tick-mediated infection) [435,436]. When tick-transmittedA. phagocytophilum infection preceded tick-mediated Bb infectionby 8 days, the burden of spirochetes, but not of A. phagocytophilum,was enhanced in the heart and certain other tissues, but carditisprevalence and severity were not altered [437]. Together, these datasuggest that A. phagocytophilum can have immunosuppressive ef-fects and thereby enhance disease severity. Such an immunosup-pressive effect may also be operative in humans as indicated by thefinding that A. phagocytophilum coinfection in Lyme disease isassociated with a significantly lower number of Borrelia-specific IL-12-secreting, though not IFNg-secreting cells [438].

In human patients, simultaneous infection with Bb and A.phagocytophilum can increase the number of symptoms comparedto Lyme borreliosis alone [431,433], although this is not a consistentfinding [432]. Whether coinfected patients have leucopenia andthrombocytopenia as frequently as patients with HGA alone is alsocontroversial [431,432]. There are data suggesting that these dis-crepancies are at least partly attributable to different case defini-tions [431], but they also may have been a result of identifying mildsubclinical cases when actively searching for coinfections. Similarly,patients coinfected with Bb and B. microtiwere found to experiencemore frequent and diverse symptoms and a more severe and pro-longed disease compared to patients infected with one or the otherpathogen alone [138,433]. While coinfections with Bb andB. miyamotoi have been described in US patients [439], it currentlyremains unknown to what extent this influences the course orseverity of either disease.

7.3. The host immune response in the pathogenesis of Lyme disease

The Borrelia genome does not encode any known toxins;therefore, it seems likely that much of the tissue damage in patientswith Lyme disease is due to the strong host immune response thatBorrelia spp. induces. In order to establish the contribution of theimmune response to the pathogenesis of the various manifesta-tions of Lyme disease it would first be necessary to elucidate whatconstitutes the most appropriate human immune response toBorrelia infection. However, this is difficult because it remains un-clear whether infected patients who remain asymptomatic or onlydevelop EM do so 1) because of a highly efficient immune response,2) because the size of the inoculum was sufficient to induce sero-conversion, but not large enough to cause clinical symptoms, 3) theinfecting strain was of limited pathogenic potential, 4) because ofother as yet unknown host (genetic) characteristics, or 5) somecombination of several or all of these factors. As long as the answersto these questions are lacking, it remains difficult to distinguishbetween beneficial and pathogenic immune responses.

7.3.1. Immune evasionThe Borrelia spirochete has developed a variety of mechanisms

that allow it to adapt to the remarkably different environments itencounters as it cycles through its host reservoirs and tick vectors.One of these is the ability to rapidly alter the expression of surfaceproteins that allows it to transition from the tick midgut into its

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3418

new host while evading the host immune system. Among the mostthoroughly investigated changes are the downregulation of OspAand the upregulation of OspC when the blood meal starts to arrivein the tickmidgut. OspAmediates spirochete attachment to the tickmidgut, while OspC is required in the initial establishment ofinfection in the vertebrate host [440]. This protein can bind theIxodes salivary gland protein Salp15 and thereby prevents recog-nition by antibodies and antibody-mediated killing. In addition,Borrelia spirochetes express a variety of proteins that can bind andneutralize complement, although their importance in vivo largelyremains to be elucidated [440e442]. Inside the new host, expres-sion of OspC is downregulated under the pressure of the host im-mune response [443,444]. Simultaneously, upregulation of otherproteins can be observed, in particular the VlsE protein [443e445].Recombinational switching between an expression locus and 15silent cassettes results in continuous variation in the sequence ofthis protein, and this antigenic variation is another importantmechanism of immune evasion. In addition, Borrelia spirochetesexpress a variety of surface proteins that allow them to bindextracellular matrix proteins such as fibronectin, laminin, decorin,integrins, and glycosaminoglycans [446]. This combined with theability to co-opt host matrix metalloproteinases (MMPs) is likely tobe important for their dissemination through tissue, but binding toextracellular matrix proteins may also provide a means to evadehumoral immunity by hiding in an immune-privileged site.

7.3.2. EM and the early immune responseAfter a tick deposits Borrelia spirochetes in the skin, an influx of

neutrophils is observed at the bite site in mice [447], as is alsoobserved in needle-inoculated mice and rabbits [448,449]. By thetime the EM lesion develops in rabbits, the initial neutrophil infil-trate is replaced by lymphocyteswith scatteredmacrophages [449].This is similar to what is seen in human biopsy samples[181,412,450,451] and may explain why neutrophils are rarelydetected in human EM lesions [450,451]. At least in vitro, neutro-phils and monocytes are not efficient at phagocytosing unopson-ized Bb [452,453], but are able to kill spirochetes through a varietyof mechanisms [454], including neutrophil extracellular traps(NETs), which are web-like structures with antibacterial propertiesthat arise from a specific type of cell death called NETosis [447]. Yet,this process does not appear to be entirely effective. Isolatedmacrophages are capable of phagocytosing and degrading Borreliaspirochetes in the presence or absence of opsonization [452,455].Therefore, they would be expected to play an important role in theearly defense against infection not only by eliminating spirochetesbut also by secreting cytokines that recruit other immune cells.However, in mice infected via tick bite, there appears to be littleclearance of tick-transmitted spirochetes from the site of tickattachment in the first few days, suggesting that in vivo, macro-phages and neutrophils are not effective in eliminating Borrelia[84]. This may be due to the ability of tick salivary components toreduce phagocytosis of human neutrophils and mouse macro-phages [377].

Recognition of Borrelia Osps and other lipoproteins by toll-likereceptors, in particular (TLR)-2 heterodimers with TLR-1 and TLR-6 in conjunction with the CD14 co-receptor, plays a central role inthe activation of the innate immune system [456,457]. However,full activation of the innate inflammatory response seems torequire phagocytosis of Bb followed by activation of the phag-osomal TLRs, TLR-7, -8 and -9, by their respective bacterial nucleicacid ligands [458e460]. There are indications that other patternrecognition receptors, in particular nucleotide-binding oligomeri-zation domain (NOD)2, also play a role [457,461e463]. TLRs areimportant not only in activating the innate immune response, butTLR-dependent recognition of pathogens by dendritic cells (DCs)

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

also is a central element in shaping the adaptive immune response[456]. Large numbers of Langerhans cells have been detected notonly at the periphery of the EM lesion, but also in the adjacent non-erythematous skin [451]. DCs were also enriched in blister fluidraised above EM lesions and displayed surfacemarkers of activationand maturation [464]. Both Langerhans cells and dermal DCs havebeen shown to be capable of phagocytosing, processing and pre-senting Borrelia spirochetes in vitro [465,466]. When pulsed withBb, Langerhans cells and splenic DCs can induce helper T cells tostimulate B cell production of Bb-specific antibodies in vitro [467].When adoptively transferred into syngeneic mice, Bb-pulsedsplenic DCs can also protect mice from infection after challengewith Bb, most likely because of their ability to induce earlier helperT cell responses and Bb-specific antibody production.

In addition, DCs have been shown to play a role in the activationof CD8þ T cells. Borrelia spp. are thought to be mainly extracellularpathogens, and electron microscopically, spirochete-like structuresare often observed to be aligned with collagen fibrils in skin[465,468], synovium [469], ligament [470], and heart [243]. This islikely to be due to their expression of Dbps, which bind to decorinexpressed on collagen fibrils. The role of CD8 cells in a variety ofautoimmune diseases has been shown to be critical not only for thepromotion, but also the prevention of disease [471]. Consequently,the role of CD8þ T cells has long been neglected even thoughseveral lines of evidence suggest that they are an importantcomponent of the immune response to Borrelia infection. Trans-mission electronmicroscopy studies revealed that, upon incubationof normal human skin fragments with Bb, dermal DCs containedspirochetes both in phagolysosomes and free in the cytoplasm, andthe same was observed when isolated dermal DCs were incubatedwith Bb [465]. Such cytosplasmic spirochetes could then be pro-cessed and presented by MHC I to CD8 T cells. Bb lacks the ability torecycle membrane lipids and lipoproteins and, therefore, shedsthem in the form of liposome-like blebs. Liposomes prepared fromBb lipids and lipoproteins as a model for native blebs quicklyreached the cytoplasm of a variety of cells via membrane fusion,whereas uptake via phagocytosis was never observed [472].Importantly, human DCs loaded with Bb-liposomes generated Bb-specific CD8þ T cells capable of killing Bb-liposome-treated autol-ogous T cell blasts. This raises the possibility (to date untested) thatsuch Bb-specific CD8þ T cells could kill cells that have taken up Bbblebs and present Bb antigens. Such cells include DCs, and theirdeath could contribute to the ineffectiveness of the early immuneresponse in preventing spirochete dissemination. In vitro, phago-cytosis of Bb induces macrophages to undergo apoptosis [455].Uptake of antigen-containing apoptotic blebs from such macro-phages by DCs could lead to cross-presentation of these antigens.

EM is thought to arise from the multiplication and spreading ofspirochetes in the skin and their induction of both innate andadaptive inflammatory immune responses. Blister fluid raisedabove EM lesions was enriched for monocytes/macrophages andDCs and these showed an activated phenotype [464]. Both CD4þand CD8þ T cells, particularly of the antigen-experienced memoryphenotype, were also enriched, and this agrees with the observa-tion that CD8þ T cells constitute a substantial portion of the infil-trate in EM and ACA lesions [451,473]. There are surprisingly fewinvestigations of the cytokine production in skin, and these showconsiderable interindividual variation in the protein or mRNAexpression of a large variety of cytokines and chemokines, withIFNg, TNFa, IL-1, IL-6 and IL-10 being most consistently detected[181,464,474e476]. There is, however, no basis for treatment withthe newer biologics and indeed there has been very little dissection,unlike in rheumatoid arthritis, to understand the complexity ofcytokine production and interactions [270,477]. Strong expressionof IFNg in EM lesions and high Bb-specific IFNg recall responses

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 19

during convalescencewere found to be associated with the absenceof residual non-specific symptoms after treatment [475,478]. Thissuggests that IFNg plays a role in early spirochete elimination,which would be consistent with its well-known role in macrophageactivation.

7.3.3. Pathogenesis of LNBThere is evidence that Bb can invade the nervous system early

after infection, but even the detection of Bb DNA in CSF is notnecessarily associated with neurological symptoms [220,479].Thus, it appears that the mere presence of Bb spirochetes is notsufficient to cause clinical disease. On the other hand, the rapidresponse to antibiotic therapy suggests that the presence of viablespirochetes is required for symptomatic LNB. The frequency (up to45%) with which Bb can be isolated from the blood of patients withearly Lyme disease in the US suggests that hematogenous spread isa frequent route of dissemination to tissues like the nervous system[101e103]. In Europe, however, spirochetemia is far less frequentlydetected [104e106]. In addition, in European patients with LNB,there often is a topographical association between the site of thetick bite or EM and the neurological manifestations, such as the firstparesis in adults [187] or facial palsy in children (who are morefrequently bitten in the head and neck region) [189]. Such a topo-graphical association has not been observed in American patients[200]. This suggests that European Borrelia spp. might spread viamigration along peripheral nerves.

In vitro, Bb can adhere to, and be internalized by, neural and glialcells, which may interfere with their function and induce cell death[480,481]. However, cytotoxicity is only seen at high multiplicitiesof infection (MOIs) [480], which are unlikely to be reached in thenervous system given how rarely spirochetes can be cultured ordetected via PCR in CSF, or visualized in biopsy or autopsy material.There is indirect evidence that Schwann cells in peripheral nervelesions of Bb-infected rhesus monkeys produce nitric oxide andTNFa [381]. This is consistent with evidence that Bb can also inducea variety of neurotoxic substances in vitro, including nitric oxideand pro-inflammatory cytokines such as IL-6 or TNFa from glialcells [482] and IL-6, IL-8 and CCL2 from human Schwann cell cul-tures and dorsal root ganglia tissue explants obtained from rhesusmonkeys [483]. Dexamethasone not only reduced the production ofthese mediators but also the Bb-induced apoptosis observed insensory neurons in this model, indicating that apoptosis was due tothe Bb-induced inflammatory response.

Histopathology of peripheral nerve biopsy specimens from pa-tients with neurological manifestations of Lyme disease shows aperivascular infiltrate of lymphocytes, plasma cells and histiocytesin association with patchy multifocal axonal degeneration[179,484e486]. A lymphocytic infiltrate has also been observed inthe meninges [365], extending into the white and gray matter in anadvanced case, along with infiltration of dorsal root ganglia andnerve roots [484,487]. It is likely that these infiltrating immunecells also contribute to the production of inflammatory andneurotoxic substances. T-cell lines and clones isolated from the CSFof patients with LNB show a strong polarization towards a type 1cytokine pattern (high production of IFNg and mostly undetectablelevels of IL-4) [488,489]. When CSF mononuclear cells (MNC) fromadult patients with LNB are stimulated with borrelial antigensin vitro, there is a marked upregulation of the number of IFNg-secreting cells [490e494], and this upregulation is significantlygreater compared to peripheral blood, suggesting a strongcompartmentalization of the Borrelia-specific IFNg response[490,494]. However, the number of responder cells shows pro-nounced variation between patients [490e494], as do the con-centrations of IFNg and essentially all other cytokines andchemokines that have been measured in CSF of patients with LNB

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

[495e497]. The actual concentrations of IFNg in CSF are remarkablylow [495,497]. Not only have CD8þ T cells been detected in nervebiopsy samples from patients with ACA-associated polyneuropathy[216], but clonal expansion of CD8 T cells has been demonstrated inthe CSF of patients with acute LNB [498]. The expanded clonesproduced TNFa and some IFNg in response to stimulation withBorrelia antigens. While the role of these Borrelia-specific CD8þ Tcells in LNB remains to be established, their detection in tissues thatare targeted soon after Borrelia infection suggests a role in earlyhost defense. On the other hand, peripheral CD8þ and gd T cellswere identified as the predominant sources of Borrelia-specificIFNg production in patients with chronic Lyme borreliosis (mostlyLNB) [499]. In view of the strong CSF compartmentalization of theimmune response, the relevance of these findings is unclear andthe precise role of this T cell subset remains to be elucidated.

A Swedish group of investigators presented data suggesting thatfailure to upregulate IL-4 production after an initial IFNg responsewas associated with a chronic course of LNB [490,492], as were lowCSF concentrations of TNFa and TGFb [491]. However, this is diffi-cult to reconcile with the observation that highly elevated numbersof Borrelia-specific IFNg-secreting T cells can still be detected insome patients after complete recovery [490,494]. The reportedassociations should be interpreted with great caution since theywere based on small numbers of patients andmultiple comparisonswithout statistical correction, and the statistically significant dif-ferences often seemed to be driven by a small number of patientswhose cytokine responses were markedly higher than the mean[490e492].

A highly consistent finding is that CSF of patients with LNBcontains high levels of B-cell chemoattractants and among these,CXCL13 plays a key in the recruitment of B cells [500,501]. B cellsconstituted an average of 16.5% of all CSF cells in patients with LNB,this proportion being significantly higher compared to patientswith viral meningitis or multiple sclerosis [502]. There are datasuggesting that B cells could contribute to the pathogenesis of LNBby producing antibodies that cross-react with neuronal antigens viamolecular mimicry; molecular mimicry has been a classic mecha-nism of autoimmune disease [503]. Both OspA and the flagellarprotein of Bb share antigenic determinants with neuronal tissuecomponents [504,505], and Bb-specific antibodies cross-reactivewith neuronal antigens have been demonstrated in serum of pa-tients with LNB [506,507]. Humoral immune responses to self an-tigens may also develop independently of molecular mimicryduring Borrelia-induced CNS inflammation [508,509]. These aretransient and most likely arise as a result of tissue damage thatunmasks novel epitopes. The rapid response of the manifestationsof LNB to antibiotics in the vast majority of patients suggests thatsuch autoimmune responses do not contribute to the developmentof chronic disease [510]. However, unlike more classical autoim-mune diseases, there is again very little evidence to suggest using Bcell therapy to treat patients with Lyme disease [511,512]. Whetherand to what extent any of these mechanisms play a role in thepathogenesis of LNB remains to be elucidated.

7.3.4. Pathogenesis of Lyme arthritis7.3.4.1. Murine Bb-induced arthritis. Bb spirochetes can readily becultured from the joints of mice needle-inoculated with as few as102 or 103 spirochetes [448,513,514] and quantitative PCR showsthe ankle to be one of the most heavily infected tissues [368,420].Mice without detectable Bb DNA in the joint essentially show nojoint inflammation, suggesting that the presence of spirochetes isrequired for the development of arthritis [368]. The murine jointinfiltrate consists primarily of neutrophils and other leukocytes,while lymphocytes are sparse [515,516]. In mice infected with Bbvia syringe inoculation, T cells are not necessary for the

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3420

development of arthritis [418,517,518], nor does adoptive transfer ofT cells from Bb-infected animals protect recipient SCID mice fromthe development of arthritis [519]. T cells also do not play a role inthe resolution of arthritis [518,520]. They may actually enhancepathology, as evidenced by the observation thatmice lacking both Band T cells have less severe arthritis and carditis than mice lackingonly B cells [521]. The results of T-cell reconstitution experiments inRag1 knockout mice indicated that T cells, more specifically CD4 Tcells, were primarily responsible for increasing the severity ofarthritis [521]. However, this contrast with other data showing thatdepletion of CD4þ T cells resulted in greater arthritis severity,whereas depletion of CD8þ T cells improved it [513]. In murineLyme disease, T cell help is also not absolutely required for pro-duction of protective and disease-resolving antibodies [522e525].Instead, T cell-independent B cell responses are of major impor-tance in host defense [518,526].

Nonetheless, there are data suggesting that Bb-specific T cellcytokine responses may play a role in the pathogenesis of arthritis.Arthritis-susceptible C3H and the more arthritis-resistant BALB/cmice differ in the production of IL-4 and IFNg during antigen-specific recall responses, suggesting that these prototypical type 1and type 2 cytokines may play a role in disease severity or duration,possibly by influencing tissue spirochete burden [514,520,527,528].However, the findings are inconsistent, hence the contributions ofIL-4 or IFNg to spirochete elimination or disease resolution aredifficult to determine. The results of studies with a variety of knock-outmice show that neither IL-4 nor IFNg are absolutely required forthe development of arthritis in mice infected via needle inoculation[529e532].

Due to the dearth of direct comparisons, it is currently unclear towhat extent the cytokine profile of mice with tick-transmittedinfection differs from that seen in syringe-inoculated mice.Antigen-specific recall responses in mice with tick-borne infectionwere assessed earlier postinfection than in most needle inoculationstudies [374,533] and showed a weak Th1 pattern in BALB/c mice[374]. In contrast, restimulation of lymph node cells from needle-inoculated BALB/c mice with Bb lysate resulted in a weak mixedTh1/Th2 response [374]. Since these cells were harvested 9 dayspostinfection, these data are consistent with the observation thatBALB/c mice infected via needle inoculation started to develop Bb-specific IL-4 production in addition to IFNg secretion at some pointbetween d4 and d14 postinfection [528]. In contrast, stimulation oflymph node cells with ConA yielded a strong Th2 pattern in BALB/cmice infected via tick bite [374], as has been observed by others[533]. This Th2 skewing was shown to be due to tick-transmittedfactors [533]. Arthritis-susceptible C3H mice showed even stron-ger Th2 polarization, and treatment with IFNg, IL-2, and TNFa aloneor in combination during the first 10 days after tick placementprotected these mice from Bb infection, the protection being almostcomplete with the combination of all three cytokines [370].Conversely, neutralization of IL-4 or IL-5 before the placement ofinfected ticks resulted in significant increases in spirochete burdenin various tissues [534].

After infection with Bb via syringe inoculation, exogenous IL-10did not attenuate arthritis inwild-typemice [535], but deficiency ofIL-10 in both C3H and B6 mice was associated with more severearthritis despite a marked decrease in the tissue spirochete burden,particularly in the ankle joint [429,535]. This suggests that IL-10 isvital for establishing the right balance between spirochete clear-ance and protection of host tissue from an overly exuberant in-flammatory response. Indeed, there are data suggesting that theeffects of IL-10 deficiency are due to enhanced macrophage pro-duction of pro-inflammatory cytokines [100], and are partlymediated by deficient downregulation not only of IFNg productionin the joint [536], but also of systemic IL-17 synthesis [537]. IL-17

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

and several of the cytokines involved in the differentiation andmaintenance of Th17 cells, such as IL-15 and particularly IL-23, alsoplay a central role in the development of arthritis in the Borreliaspp.-vaccination and -infection model [538e543]. In this model, B6mice are vaccinated with B. bissettii and challenged with Bb 21 dlater, with the vaccination step priming Borrelia-specific T cellswhile infection with a heterologous strain circumvents the pro-tective antibody response. Note that in this model, the infiltrate inthe joint contains lymphocytes in addition to neutrophils andmacrophages [544]. In contrast, IL-17 seems to play at best a minorrole in the standardmurine model, the C3Hmouse infected with Bbvia needle inoculation, which is largely T-cell independent [545].

Numerous studies have also documented that B cells, while notrequired for the development of arthritis, are necessary for remis-sion of arthritis [418,517,518,520,521,546]. Whether they are alsosufficient remains somewhat controversial. For example, in theabsence of natural killer T (NKT) cells, arthritis did not resolve inmice with tick-transmitted Bb infection despite increased anti-Bbantibody production [363].

If B cells and T cells are not required for the development ofarthritis in the commonly used murine models, this suggests thatinnate inflammatory responses make a major contribution to Bb-induced arthritis. While the available data do not suggest a sub-stantial role for NK cells, there is evidence that neutrophilscontribute to the pathogenesis of murine LA via their ability toproduce inflammatory cytokines and chemokines, and data fromthe hamster model also implicate macrophages [544].

7.3.4.2. Pathogenesis of human LA. Perivascular accumulation oflymphocytes and macrophages and scattered mast cells in theinterstitium are typical histopathological findings in patients withsevere, mostly chronic LA [266]. Some samples may also showlymphoid follicles with the cellular composition, though not thearchitecture, of germinal centers. Neutrophils are not generallyseen in the synovium in LA, but it has been hypothesized that theyare present in the acute stages, but are soon replaced by lympho-cytes and plasma cells [365]. Support for this theory comes fromthe observation that histopathology in a patient with LA whoresponded to antibiotic therapy showed granulation-like tissue andan infiltrate consisting mainly of neutrophils, whereas a treatment-resistant patient showed the typical histopathology of chronicarthritis [547]. Neutrophils are abundant in synovial fluid, wherethey often represent ~80e90% of cells [248,548], suggesting thatthey contribute to the pathogenesis of LA. They possess the abilityto produce a variety of inflammatory cytokines, such as IL-1b, TNFaand IL-15. Both TNFa and IL-1b are principal mediators of jointdestruction in other forms of inflammatory arthritis due to theireffects on bone resorption, resorption of proteoglycans in cartilage,and their ability to stimulate the release of additional pro-inflammatory cytokines and other inflammatory mediators. Timeto resolution of an arthritis episode was found to be longest inpatients with the highest levels of IL-1b and the lowest levels of itsnatural antagonist, IL-1Ra [549]. In addition, neutrophils can pro-duce chemokines that attract Th1 cells, Th17 cells or both, such asCXCL10, CCL2, and CCL20 [550]. The concentrations of all of thesechemoattractants were found to be high in synovial fluid of patientswith LA [550].

The initial focus in the assessment of the roles of cytokines in thepathogenesis of LA was on Th1 cells. Immunohistochemistryshowed that IFNg was the predominant cytokine in the synovialmembrane, while lesser numbers of cells expressed IL-4, IL-10 andIL-12 [551]. The Borrelia-specific recall response in synovial fluidMNC from patients with LA is also polarized towards a type 1cytokine pattern [551,552], and such a pattern is also seen in T celllines or clones obtained from PBMC or synovial fluid [553,554],

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 21

which sometimes co-produce IFNg and IL-10, but only rarely IL-4[555]. There are data suggesting that the ratio of Th1:Th2secreting cells in synovial fluid correlates with the severity of Lymearthritis [552] and that high synovial fluid IFNg concentrations areassociated with antibiotic-refractory arthritis [556]. However, noassociation between IFNg mRNA expression in synovial fluid MNCor synovial membrane and responder status was detected [551].

As has also been reported from CSF of patients with LNB [498],Borrelia-specific CD8þ T cell lines could be generated from pe-ripheral blood and synovial fluid of five patients with LA [557].Specific HLA class I-restricted cytotoxic activity could only bedemonstrated with samples obtained after the resolution ofarthritis, not during active arthritis. Unlike what is seen in themurinemodel of LA [513], this seems to imply that CD8 Tcells play arole in the resolution of LA. The detection of cytotoxic activity onlyafter disease resolution may explain why others failed to establishBb-specific cytotoxic T cell clones from PBMC or synovial fluid cellsof patients with chronic LA [553], although this failure may havebeen due to methodological issues rather than to the true absenceof such cells [557]. Unlike rheumatoid arthritis, there has not been acomprehensive genetic analysis [558].

There is increasing evidence that not only Th1 cells, but alsoTh17 cells contribute to the pathogenesis of LA not only in thevaccination-infection model of murine Bb-induced arthritis, butalso in human LA. Synovial fluid of patients with LA was shown tocontain IL-17-producing T cells [559] and significantly elevatedlevels of IL-17 compared to controls [550]. In addition, theneutrophil-activating protein A (NapA) of Bb was detectable in sy-novial fluid of patients with LA, this protein stimulated the pro-duction of high levels of IL-17 as well as IFNg from synovial fluid Tcells, and NapA-specific Th17 cells accumulated in synovial fluid,but could not be detected in peripheral blood [550,560]. NapA alsotime and dose-dependently induced the production of IL-23 byneutrophils and monocytes and of other cytokines involved in thedifferentiation and maintenance of Th17 cells in monocytes. On theother hand, a polymorphism in the receptor of IL-23 was associatedwith significantly lower in vitro production of IL-17 by Bb-stimu-lated PBMC, but did not influence the frequency of arthritis or otherchronic manifestations of Lyme disease [561]. Together, these re-sults suggest that synovial rather than peripheral production of IL-17 contributes to Bb-induced joint inflammation.

It has long been held that tissue damage results from the stronginflammatory response to Bb since the spirochete does not expresstoxins. However, it was recently discovered that Bb expresses aprotease with aggrecanase activity [562], and this may enable it tocause direct damage to joint tissue. In addition, Bb is capable ofinducing the expression of variety of host matrix metal-loproteinases (MMPs) as well as the aggrecanase ADAMST-4, butnot ADAMST-5, by human chondrocytes in vitro [563e566].Furthermore, several MMPs as well as ADAMST-4 are elevated inthe synovial fluid of patients with LA. Additional MMPs weredetected mainly in the synovial fluid of patients with persistentsymptoms after antibiotic therapy [564]. There are data suggestingthat thesewere not directly induced by Bb, but that their expressionresulted from the activation of innate inflammatory responses.Altogether, these data suggest that a variety of enzymes capable ofdegrading the matrix component aggrecan play a role in thedegradation of cartilage and subsequent joint damage in human LA.

7.3.4.3. The pathogenesis of antibiotic-refractory LA7.3.4.3.1. The autoimmune mechanism hypothesis. Compared to

patients with antibiotic-responsive LA, patients with antibiotic-refractory arthritis more frequently possess HLA-DRB1*0401 andrelated alleles [279,319,567], which are also associated with theseverity of rheumatoid arthritis. US patients with LA frequently

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

develop seroreactivity to OspA near the onset of prolonged episodesof arthritis [279]. The highest risk of non-response to treatment wasobserved in the group of patients who had both the HLA-DR4specificity and elaborated an anti-OspA antibody response [278].The association of antibiotic-refractory arthritis with certain HLAclass II alleles implicates CD4þ T cells in the pathogenesis ofantibiotic-refractory arthritis. The association of these same alleleswith severity of rheumatoid arthritis suggests that an autoimmunemechanism might play a role in perpetuating the inflammatoryresponse, even after the apparent elimination of infection withantibiotics [117,568,569]. Indeed, PBMC or synovial fluid T cells ofpatients with treatment-resistant LA exhibited a significantlyhigher frequency of proliferative and IFNg responses to theimmunodominant epitope of OpsA (OspA164e173) compared to pa-tients with antibiotic-responsive arthritis [570]. Use of an algorithmto predict epitopes presented by DRB1*0401 resulted in the iden-tification of LFA-1 as a candidate autoantigen, and OspA164e173-specific DRB1*0401 þ CD4þ T cells isolated by tetramer technologyrecognized the relevant epitope of LFA-1 [571]. However, its lack ofbinding to DRB1*0101, the other HLA-DR molecule associated withantibiotic-refractory arthritis, makes its relevance doubtful [571].Other candidates include cytokeratin-10 [572] and endothelial cellgrowth factor [573].

This autoimmune hypothesis ignores that LA eventually resolvesin essentially all patients. It is somewhat difficult to comprehendhow an infection-induced autoimmune process spontaneouslydisappears after it was capable of maintaining joint inflammationfor months or years after the apparent eradication of the pathogen.On the other hand, post-streptococcal rheumatic heart disease alsofades and would be considered as a classic autoimmune disease.Additionally, it is well known that patients with Behcet's syndromeundergo spontaneous remission over time and this is likely alsotrue of a number of auto-inflammatory diseases [574,575]. Inaddition, a humoral response to OspA is not commonly seen inEuropean patients with LA [121,576], and the immunodominantepitope identified in Bb OspA does not exist in B. afzelii or B. garinii[577]. This has been proposed as an explanation for the putativeabsence of antibiotic-refractory arthritis in Europe. However, theavailable data clearly show the existence of treatment-resistant LAin European children [249,259,320], and some adult cases have alsobeen reported [550,551,578,579]. Therefore a different pathoge-netic mechanism must be responsible for antibiotic-refractory LA,at least in Europe.

7.3.4.3.2. The spirochete persistence hypothesis. Borrelia spp.establishes persistent infection in a variety of vertebrate hosts, suchas laboratory mice, rats, hamsters, gerbils, guinea pigs, dogs, andnon-human primates, even though these animals mount a strongimmune response that considerably reduces the spirochete burden.Persistent infection has also been observed in untreated humans[244,580], as most clearly demonstrated by the isolation of spiro-chetes from an ACA lesion of more than 10 years' duration [359].The question then arises of whether Borrelia spirochetes can evadenot only the host immune response but also treatment with anti-biotics and thereby account for the persistent symptoms of patientswith treatment-resistant LA. It has been postulated that they coulddo so by hiding in sites inaccessible to both, such as the extracel-lular matrix. Bb expresses various surface proteins that can bind avariety of extracellular matrix proteins such as fibronectin, laminin,decorin, integrins, and glycosaminoglycans. Electron microscopyshows spirochete-like structures to be aligned with collagen fibrilsin various tissues, including synovium [469] and ligament [470].Another possibility is the intracellular seclusion of spirochetes. Ithas been shown that a several different cell types are capable ofinternalizing Bb in vitro and that spirochetes remain viable forseveral weeks of co-culture [481]. However, it currently remains

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3422

unknownwhether and towhat extent suchmechanisms contributeto borrelial immune evasion and potential persistence after anti-biotic treatment.

In patients with antibiotic-refractory arthritis, borrelial DNA canquite often be detected by PCR in synovial fluid or tissue for up to9 months after the beginning of the first course of antibiotic ther-apy [117,268,569,578,581,582]. However, positive PCR results arenot associated with recurrence or duration of arthritis after therapy[569]. In samples obtained �12 months after the start of therapy,PCR results are generally negative [568,569], although longerpersistence has occasionally been reported in synovial fluid [583]and muscle [584]. Since PCR is not 100% sensitive, a negative PCRresult does not prove the absence of spirochetes. On the other hand,PCR cannot distinguish between viable and non-viable organisms.Attempts to detect borrelial mRNA transcripts as a marker ofmetabolic activity failed in three synovial fluid samples that yieldedpositive PCR results for borrelial DNA [569]. This suggests that thisspirochetal DNA originated from non-viable organisms. In contrast,a few investigators have been able to culture Borrelia spp. fromsynovial samples or tendon for up to ~10 months after antibiotictreatment [112,113,470]. In addition, analysis of plasma cells ob-tained via laser-capture microdissection from joint tissue of pa-tients with antibiotic-refractory LA provided evidence of ongoing insitu diversification and affinity maturation of the antibody reper-toire, even though PCR yielded negative results for Bb DNA [585];the authors suggested that this was due to the continued presenceof an autoantigen; yet, persistence of Bb antigens cannot beexcluded.

Similar to what had previously been described in a treated pa-tient [586], Wormser et al. [587] reported the visualization of a fewmostly non-motile spirochetes in culture medium inoculated withsynovial fluid from an untreated patient. These spirochetes couldnot be subcultured and appeared to be enmeshed in some form ofmatrix of unknown composition. This led to the hypothesis thatthey represent non-viable, but morphologically intact, organismssome component of which might be capable of triggering an in-flammatory response upon release into the joint space [587]. Someconfirmation for this hypothesis comes from experimental animals[588]. Through the use of real-time intravital microscopy and Bbstably transformed to express the green fluorescent protein (GFP),large deposits of nonmotile fluorescentmaterial could be visualizedat the interface of ear dermis and cartilage and in patellar enthesesof both sham-treated and doxycycline-treated Myd88-deficientmice (chosen because of their higher pathogen burden), whilemotile spirochetes were detectable only in sham-treated animals[588]. Immunization of naïve mice with patellar homogenatesinduced seroconversion in the recipients and enhanced macro-phage TNFa production in vitro regardless of the antibiotic treat-ment status of the donor. In a separate experiment, the results ofxenodiagnosis and culture of blood, ear skin and urinary bladderssuggested that no viable spirochetes persisted inwild-type B6 micewith tick-transmitted Bb infection, and only 1/21 Myd88-deficientmice was culture-positive after treatment with doxycycline in thedrinking water. However, borrelial DNA could still be detected inear skin, but not tibiotarsal joints, after antibiotic treatment.Nonetheless, confirmation that the patellar homogenate fromantibiotic-treated animals was unable to induce infection wouldhave been desirable. These results strongly suggest that immuno-genic spirochete material persists near cartilage and in enthesesafter antibiotic treatment and could provide a mechanism for thedevelopment of antibiotic-resistant LA if it could be shown thatthese spirochetal remnants persisted for at least several months.

In contrast to these findings, other studies in mice infected withBb via syringe inoculation or tick bite show the persistence not onlyof borrelial DNA in various tissues, but also of a small number of

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

non-cultivatable spirochetes [589e591]. Conflicting results havebeen obtained regarding their infectivity, but they do not causepathology. While tissue DNA levels continuously decrease until8 months after completion of antibiotic treatment, they resurge tolevels similar to those in saline-treated animals at 12 months aftertreatment [592]. Despite declining antibody levels, this is notaccompanied by tissue inflammation, suggesting that viable spi-rochetes survive, but remain non-pathologic. Other animal studieshave yielded inconsistent results regarding the persistence ofviable spirochetes after antibiotic treatment [593]. This may partlybe due to the use of different inoculum sizes, failure to consider thepharmacodynamics of the specific model, and varying outcomemeasures. Of note, in syringe-inoculated rhesus macaques treatedwith antibiotics during the disseminated stage of disease, Bb DNAand RNA (as a marker of viability) could be detected in some of thetreated animals and a few spirochetes could be recovered fromxenodiagnostic ticks [594]. A limited number of slow-growing, butmetabolically active, spirochetes could be cultured from some ofthe animals. Similarly, tissues remained not only PCR-positive, butspirochetes could be cultured from some antibiotic-treated dogswith tick-transmitted Bb infection [593,595].

Correspondingly, Borrelia spp. can occasionally be cultured fromsynovial samples or tendon for up to ~10 months after antibiotictreatment [112,113,470], the site of previous EM lesions up to 3months after therapy [285,596,597], and from CSF of patients withLNB up to at least 7 months after treatment [297,597], even if this isnot always associated with recurrent or persistent symptoms [597].While such cases are rare, they nonetheless suggest that the sur-vival of some viable spirochetes after antibiotic treatment couldoccasionally contribute to some of the long-lasting manifestationsof Lyme disease, including antibiotic-refractory LA.

7.3.5. Potential pathogenesis of PLDSEvidence is emerging that the persistence of symptoms after

antibiotic treatment may be associated with immunological ab-normalities in a subset of patients. In particular, compared to pa-tients with a history of Lyme disease without persisting symptomsafter treatment, a significantly higher frequency of anti-neuralantibody reactivity was seen in convenience samples of patientswith PLDS who had participated in two of the antibiotic retreat-ment trials [326,598]. These anti-neural antibodies reacted with alarge variety of neural tissue components, suggesting that they arenot directly involved in the pathogenesis of PLDS. In Slovenianpatients with EM, there was a strong correlation between theserum concentrations of the Th17-associated cytokine IL-23 atpresentation and the subsequent persistence or development ofsymptoms 2 months after antibiotic treatment [599]. Those withpersistent symptoms more frequently had antibodies againstendothelial cell growth factor, a putative autoantigen implicated inthe persistence of arthritis after antibiotic therapy [573], and levelsof IL-23 also correlated with the concentrations of these autoanti-bodies in this subset of patients [599].

8. Conclusions

Lyme disease is an infectious cause of recurrent bouts of arthritisoften part of a multi-system disease that involves cardiac, skin andneurologic manifestations. It typically begins with an erythematousrash but can also include a number of other dermatologic abnor-malities. Approximately 20% of patients experience only dermato-logic features and do not progress further. However, 40e60% ofpatients eventually develop an inflammatory arthropathy. Thedisease is initiated following exposure of a genetically at risk host toa tick that carries a spirochete. There is now extensive research onthe molecular characteristics of these spirochetal infections,

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 23

including their genomic organization. Virtually all confirmed casesof Lyme disease occur in the Northern hemisphere and there isinformation that there is both under as well as over reporting ofLyme disease. The under reporting likely comes from failure toconsider the diagnosis as well as patients who undergo sponta-neous remission. The over reporting comes from an over zealousdiagnosis based on misinterpretation of the serologic findings andinaccuracies of internet information. Further research to identifythe mechanism of action and effector mechanisms will lead toimproved therapies. Finally, physician education is cristical to anaccurate diagnosis.

References

[1] Steere AC, Malawista SE, Snydman DR, Shope RE, Andiman WA, Ross MR,et al. Lyme arthritis: an epidemic of oligoarticular arthritis in children andadults in three Connecticut communities. Arthritis Rheum 1977;20:7e17.

[2] Steere AC, Bartenhagen NH, Craft JE, Hutchinson GJ, Newman JH, Rahn DW,et al. The early clinical manifestations of Lyme disease. Ann Intern Med1983;99:76e82.

[3] Afzelius A. Verhandlungen der Dermatologischen Gesellschaft zu Stockholm,28. Oktober 1909. Arch Dermatol Syph 1910;101:404.

[4] Steere AC, Schoen RT, Taylor E. The clinical evolution of Lyme arthritis. AnnIntern Med 1987;107:725e31.

[5] Steere AC, Broderick TF, Malawista SE. Erythema chronicum migrans andLyme arthritis: epidemiologic evidence for a tick vector. Am J Epidemiol1978;108:312e21.

[6] Stanek G, Strle F. Lyme disease: European perspective. Infect Dis Clin NorthAm 2008;22:327e39 [vii].

[7] Burgdorfer W, Barbour AG, Hayes SF, Benach JL, Grunwaldt E, Davis JP. Lymedisease e a tick-borne spirochetosis? Science 1982;216:1317e9.

[8] Steere AC, Grodzicki RL, Kornblatt AN, Craft JE, Barbour AG, BurgdorferW, et al.The spirochetal etiology of Lyme disease. N Engl J Med 1983;308:733e40.

[9] Benach JL, Bosler EM, Hanrahan JP, Coleman JL, Habicht GS, Bast TF, et al.Spirochetes isolated from the blood of two patients with Lyme disease.N Engl J Med 1983;308:740e2.

[10] Johnson RC, Schmid GP, Hyde FW, Steigerwalt AG, Brenner DJ. Borreliaburgdorferi sp. nov.: etiologic agent of Lyme disease. Int J Syst Bacteriol1984;34:496e7.

[11] Brisson D, Drecktrah D, Eggers CH, Samuels DS. Genetics of Borrelia burg-dorferi. Annu Rev Genet 2012;46:515e36.

[12] Horai R, Silver PB, Chen J, Agarwal RK, Chong WP, Jittayasothorn Y, et al.Breakdown of immune privilege and spontaneous autoimmunity in miceexpressing a transgenic T cell receptor specific for a retinal autoantigen.J Autoimmun 2013;44:21e33.

[13] Pillai S. Rethinking mechanisms of autoimmune pathogenesis. J Autoimmun2013;45:97e103.

[14] Anaya JM. Common mechanisms of autoimmune diseases (the autoimmunetautology). Autoimmun Rev 2012;11:781e4.

[15] Hayter SM, Cook MC. Updated assessment of the prevalence, spectrum andcase definition of autoimmune disease. Autoimmun Rev 2012;11:754e65.

[16] Barbour AG, Burgdorfer W, Hayes SF, P�eter O, Aeschlimann A. Isolation of acultivable spirochete from Ixodes ricinus ticks of Switzerland. Curr Microbiol1983;8:123e6.

[17] Korenberg EI, Kryuchechnikov VN, Kovalevsky YV. Advances in in-vestigations of Lyme borreliosis in the territory of the former USSR. Eur JEpidemiol 1993;9:86e91.

[18] Clark KL, Leydet B, Hartman S. Lyme borreliosis in human patients in Floridaand Georgia, USA. Int J Med Sci 2013;10:915e31.

[19] Clark KL, Leydet BF, Threlkeld C. Geographical and genospecies distributionof Borrelia burgdorferi sensu lato DNA detected in humans in the USA. J MedMicrobiol 2014;63:674e84.

[20] Fingerle V, Schulte-Spechtel UC, Ruzic-Sabljic E, Leonhard S, Hofmann H,Weber K, et al. Epidemiological aspects and molecular characterization ofBorrelia burgdorferi s.l. from southern Germany with special respect to thenew species Borrelia spielmanii sp. nov. Int J Med Microbiol 2008;298:279e90.

[21] Girard YA, Fedorova N, Lane RS. Genetic diversity of Borrelia burgdorferi anddetection of B. bissettii-like DNA in serum of north-coastal California resi-dents. J Clin Microbiol 2011;49:945e54.

[22] Demaerschalck I, Ben Messaoud A, De Kesel M, Hoyois B, Lobet Y, Hoet P,et al. Simultaneous presence of different Borrelia burgdorferi genospecies inbiological fluids of Lyme disease patients. J Clin Microbiol 1995;33:602e8.

[23] Rijpkema SG, Tazelaar DJ, Molkenboer MJ, Noordhoek GT, Plantinga G,Schouls LM, et al. Detection of Borrelia afzelii, Borrelia burgdorferi sensustricto, Borrelia garinii and group VS116 by PCR in skin biopsies of patientswith erythema migrans and acrodermatitis chronica atrophicans. ClinMicrobiol Infect 1997;3:109e16.

[24] Schaarschmidt D, Oehme R, Kimmig P, Hesch RD, Englisch S. Detection andmolecular typing of Borrelia burgdorferi sensu lato in Ixodes ricinus ticks and

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

in different patient samples from southwest Germany. Eur J Epidemiol2001;17:1067e74.

[25] Ruzic-Sabljic E, Arnez M, Logar M, Maraspin V, Lotric-Furlan S, Cimperman J,et al. Comparison of Borrelia burgdorferi sensu lato strains isolated fromspecimens obtained simultaneously from two different sites of infection inindividual patients. J Clin Microbiol 2005;43:2194e200.

[26] Liveris D, Varde S, Iyer R, Koenig S, Bittker S, Cooper D, et al. Genetic diversityof Borrelia burgdorferi in Lyme disease patients as determined by cultureversus direct PCR with clinical specimens. J Clin Microbiol 1999;37:565e9.

[27] Lled�o L, Gegúndez MI, Gim�enez-Pardo C, �Alamo R, Fern�andez-Soto P,Nuncio MS, et al. A seventeen-year epidemiological surveillance study ofBorrelia burgdorferi infections in two provinces of northern Spain. Int J En-viron Res Public Health 2014;11:1661e72.

[28] Hub�alek Z. Epidemiology of Lyme borreliosis. Curr Probl Dermatol 2009;37:31e50.

[29] Centers for Disease Control and Prevention. Reported cases of Lyme diseaseby Year, United States, 2003-2012. 2013. www.cdc.gov/lyme/stats/chartstables/casesbyyear.html [accessed 15.03.04].

[30] Naleway AL, Belongia EA, Kazmierczak JJ, Greenlee RT, Davis JP. Lyme diseaseincidence in Wisconsin: a comparison of state-reported rates and rates froma population-based cohort. Am J Epidemiol 2002;155:1120e7.

[31] Meek JI, Roberts CL, Smith Jr EV, Cartter ML. Underreporting of Lyme diseaseby Connecticut physicians. J Public Health Manag Pract 1992;1996(2):61e5.

[32] Coyle BS, Strickland GT, Liang YY, Pena C, McCarter R, Israel E. The publichealth impact of Lyme disease in Maryland. J Infect Dis 1996;173:1260e2.

[33] Kuehn BM. CDC estimates 300,000 US cases of Lyme disease annually. JAMA2013;310:1110.

[34] Anderson G, Berk M, Maes M. Biological phenotypes underpin the physio-somatic symptoms of somatization, depression, and chronic fatigue syn-drome. Acta psychiatr Scand 2014;129:83e97.

[35] Bacon RM, Kugeler KJ, Mead PS. Surveillance for Lyme diseaseeUnited States,1992-2006. MMWR Surveill Summ 2008;57:1e9.

[36] Fix AD, Pena CA, Strickland GT. Racial differences in reported Lyme diseaseincidence. Am J Epidemiol 2000;152:756e9.

[37] Lyme diseaseeUnited States, 2003-2005. MMWR Morb Mortal Wkly Rep2007;56:573e6.

[38] Pollard KM. Gender differences in autoimmunity associated with exposure toenvironmental factors. J Autoimmun 2012;38:J177e86.

[39] Pennell LM, Galligan CL, Fish EN. Sex affects immunity. J Autoimmun2012;38:J282e91.

[40] Shoenfeld Y, Tincani A, Gershwin ME. Sex gender and autoimmunity.J Autoimmun 2012;38:J71e3.

[41] Stanek G, Fingerle V, Hunfeld KP, Jaulhac B, Kaiser R, Krause A, et al. Lymeborreliosis: clinical case definitions for diagnosis and management inEurope. Clin Microbiol Infect 2011;17:69e79.

[42] Zeman P, Benes C. Spatial distribution of a population at risk: an importantfactor for understanding the recent rise in tick-borne diseases (Lyme bor-reliosis and tick-borne encephalitis in the Czech Republic). Ticks Tick BorneDis 2013;4:522e30.

[43] Svihrova V, Hudeckova H, Jesenak M, Schwarzova K, Kostanova Z, Ciznar I.Lyme borreliosis e analysis of the trends in Slovakia, 1999-2008. FoliaMicrobiol (Praha) 2011;56:270e5.

[44] Fül€op B, Poggensee G. Epidemiological situation of Lyme borreliosis in Ger-many: surveillance data from six Eastern German States, 2002 to 2006.Parasitol Res 2008;103(Suppl. 1):S117e20.

[45] Trajer A, Bobvos J, Paldy A, Krisztalovics K. Association between incidence ofLyme disease and spring-early summer season temperature changes inHungary e 1998-2010. Ann Agric Environ Med 2013;20:245e51.

[46] Levy S. The Lyme disease debate: host biodiversity and human disease risk.Environ Health Perspect 2013;121:A120e5.

[47] Brownstein JS, Holford TR, Fish D. Effect of climate change on Lyme diseaserisk in North America. Ecohealth 2005;2:38e46.

[48] Randolph SE. Evidence that climate change has caused ‘emergence’ of tick-borne diseases in Europe? Int J Med Microbiol 2004;293(Suppl. 37):5e15.

[49] Lindgren E, Talleklint L, Polfeldt T. Impact of climatic change on the northernlatitude limit and population density of the disease-transmitting Europeantick Ixodes ricinus. Environ Health Perspect 2000;108:119e23.

[50] Bennet L, Halling A, Berglund J. Increased incidence of Lyme borreliosis insouthern Sweden following mild winters and during warm, humid summers.Eur J Clin Microbiol Infect Dis 2006;25:426e32.

[51] Berglund J, Eitrem R, Ornstein K, Lindberg A, Ringer A, Elmrud H, et al. Anepidemiologic study of Lyme disease in southern Sweden. N Engl J Med1995;333:1319e27.

[52] Altpeter E, Zimmermann H, Oberreich J, Peter O, Dvorak C. Tick relateddiseases in Switzerland, 2008 to 2011. Swiss Med Wkly 2013;143:w13725.

[53] Lesnyak O, Laikovskaya E, Kufko I, Bruinink H, Baranova N, Rijpkema S.Clinical features of Lyme borreliosis in the middle Urals and distribution ofBorrelia burgdorferi sensu lato species in local Ixodes persulcatus ticks. Zen-tralbl Bakteriol 1998;288:111e9.

[54] Huppertz HI, B€ohme M, Standaert SM, Karch H, Plotkin SA. Incidence of Lymeborreliosis in the Würzburg region of Germany. Eur J Clin Microbiol InfectDis 1999;18:697e703.

[55] Sood SK, Salzman MB, Johnson BJ, Happ CM, Feig K, Carmody L, et al.Duration of tick attachment as a predictor of the risk of Lyme disease in anarea in which Lyme disease is endemic. J Infect Dis 1997;175:996e9.

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3424

[56] Shapiro ED, Gerber MA, Holabird NB, Berg AT, Feder Jr HM, Bell GL, et al.A controlled trial of antimicrobial prophylaxis for Lyme disease after deer-tick bites. N Engl J Med 1992;327:1769e73.

[57] Costello CM, Steere AC, Pinkerton RE, Feder Jr HM. A prospective study of tickbites in an endemic area for Lyme disease. J Infect Dis 1989;159:136e9.

[58] Maiwald M, Oehme R, March O, Petney TN, Kimmig P, Naser K, et al.Transmission risk of Borrelia burgdorferi sensu lato from Ixodes ricinus ticks tohumans in southwest Germany. Epidemiol Infect 1998;121:103e8.

[59] Huegli D, Moret J, Rais O, Moosmann Y, Erard P, Malinverni R, et al. Pro-spective study on the incidence of infection by Borrelia burgdorferi sensu latoafter a tick bite in a highly endemic area of Switzerland. Ticks Tick Borne Dis2011;2:129e36.

[60] Fryland L, Wilhelmsson P, Lindgren PE, Nyman D, Ekerfelt C, Forsberg P. Lowrisk of developing Borrelia burgdorferi infection in the south-east of Swedenafter being bitten by a Borrelia burgdorferi-infected tick. Int J Infect Dis2011;15:e174e81.

[61] Nahimana I, Gern L, Blanc DS, Praz G, Francioli P, Peter O. Risk of Borreliaburgdorferi infection in western Switzerland following a tick bite. Eur J ClinMicrobiol Infect Dis 2004;23:603e8.

[62] Korenberg EI, Vorobyeva NN, Moskvitina HG, Gorban L. Prevention of bor-reliosis in persons bitten by infected ticks. Infection 1996;24:187e9.

[63] Fahrer H, van der Linden SM, Sauvain MJ, Gern L, Zhioua E, Aeschlimann A.The prevalence and incidence of clinical and asymptomatic Lyme borreliosisin a population at risk. J Infect Dis 1991;163:305e10.

[64] Strle F, Nelson JA, Ruzic-Sabljic E, Cimperman J, Maraspin V, Lotric-Furlan S,et al. European Lyme borreliosis: 231 culture-confirmed cases involvingpatients with erythema migrans. Clin Infect Dis 1996;23:61e5.

[65] Oksi J, Marttila H, Soini H, Aho H, Uksila J, Viljanen MK. Early disseminationof Borrelia burgdorferi without generalized symptoms in patients with ery-thema migrans. APMIS 2001;109:581e8.

[66] Glatz M, Golestani M, Kerl H, Müllegger RR. Clinical relevance of different IgGand IgM serum antibody responses to Borrelia burgdorferi after antibiotictherapy for erythema migrans: long-term follow-up study of 113 patients.Arch Dermatol 2006;142:862e8.

[67] Nadelman RB, Nowakowski J, Fish D, Falco RC, Freeman K, McKenna D, et al.Prophylaxis with single-dose doxycycline for the prevention of Lyme diseaseafter an Ixodes scapularis tick bite. N Engl J Med 2001;345:79e84.

[68] Steere AC, Sikand VK, Schoen RT, Nowakowski J. Asymptomatic infectionwith Borrelia burgdorferi. Clin Infect Dis 2003;37:528e32.

[69] Berglund J, Eitrem R. Tick-borne borreliosis in the archipelago of southernSweden. Scand J Infect Dis 1993;25:67e72.

[70] Finch C, Al-Damluji MS, Krause PJ, Niccolai L, Steeves T, O'Keefe CF, et al.Integrated assessment of behavioral and environmental risk factors for Lymedisease infection on Block Island, Rhode Island. PLoS One 2014;9:e84758.

[71] Beytout J, George JC, Malaval J, Garnier M, Beytout M, Baranton G, et al. Lymeborreliosis incidence in two French departments: correlation with infectionof Ixodes ricinus ticks by Borrelia burgdorferi sensu lato. Vector Borne Zoo-notic Dis 2007;7:507e17.

[72] Selmi C, Lu Q, Humble MC. Heritability versus the role of the environment inautoimmunity. J Autoimmun 2012;39:249e52.

[73] Bennet L, Berglund J. Reinfection with Lyme borreliosis: a retrospectivefollow-up study in southern Sweden. Scand J Infect Dis 2002;34:183e6.

[74] Salazar JC, Gerber MA, Goff CW. Long-term outcome of Lyme disease inchildren given early treatment. J Pediatr 1993;122:591e3.

[75] Nowakowski J, Nadelman RB, Sell R, McKenna D, Cavaliere LF, Holmgren D,et al. Long-term follow-up of patients with culture-confirmed Lyme disease.Am J Med 2003;115:91e6.

[76] Barthold SW. Specificity of infection-induced immunity among Borreliaburgdorferi sensu lato species. Infect Immun 1999;67:36e42.

[77] Khatchikian CE, Nadelman RB, Nowakowski J, Schwartz I, Wormser GP,Brisson D. Evidence for strain-specific immunity in patients treated for earlyLyme disease. Infect Immun 2014;82:1408e13.

[78] Miller FW, Pollard KM, Parks CG, Germolec DR, Leung PS, Selmi C, et al.Criteria for environmentally associated autoimmune diseases. J Autoimmun2012;39:253e8.

[79] Falco RC, McKenna DF, Daniels TJ, Nadelman RB, Nowakowski J, Fish D, et al.Temporal relation between Ixodes scapularis abundance and risk for Lymedisease associated with erythema migrans. Am J Epidemiol 1999;149:771e6.

[80] Smith RP, Schoen RT, Rahn DW, Sikand VK, Nowakowski J, Parenti DL, et al.Clinical characteristics and treatment outcome of early Lyme disease in pa-tients with microbiologically confirmed erythema migrans. Ann Intern Med2002;136:421e8.

[81] Lipsker D, Antoni-Bach N, Hansmann Y, Jaulhac B. Long-term prognosis of pa-tients treated for erythemamigrans in France. Br J Dermatol 2002;146:872e6.

[82] Rauter C, Hartung T. Prevalence of Borrelia burgdorferi sensu lato genospeciesin Ixodes ricinus ticks in Europe: a metaanalysis. Appl Environ Microbiol2005;71:7203e16.

[83] Dunham-Ems SM, Caimano MJ, Pal U, Wolgemuth CW, Eggers CH, Balic A,et al. Live imaging reveals a biphasic mode of dissemination of Borreliaburgdorferi within ticks. J Clin Invest 2009;119:3652e65.

[84] Hodzic E, Feng S, Freet KJ, Borjesson DL, Barthold SW. Borrelia burgdorferipopulation kinetics and selected gene expression at the host-vector inter-face. Infect Immun 2002;70:3382e8.

[85] Piesman J, Mather TN, Sinsky RJ, Spielman A. Duration of tick attachment andBorrelia burgdorferi transmission. J Clin Microbiol 1987;25:557e8.

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

[86] Piesman J, Maupin GO, Campos EG, Happ CM. Duration of adult femaleIxodes dammini attachment and transmission of Borrelia burgdorferi, withdescription of a needle aspiration isolation method. J Infect Dis 1991;163:895e7.

[87] Piesman J. Dynamics of Borrelia burgdorferi transmission by nymphal Ixodesdammini ticks. J Infect Dis 1993;167:1082e5.

[88] des Vignes F, Piesman J, Heffernan R, Schulze TL, Stafford 3rd KC, Fish D.Effect of tick removal on transmission of Borrelia burgdorferi and Ehrlichiaphagocytophila by Ixodes scapularis nymphs. J Infect Dis 2001;183:773e8.

[89] Ohnishi J, Piesman J, de Silva AM. Antigenic and genetic heterogeneity ofBorrelia burgdorferi populations transmitted by ticks. Proc Natl Acad Sci U S A2001;98:670e5.

[90] Hojgaard A, Eisen RJ, Piesman J. Transmission dynamics of Borrelia burg-dorferi s.s. during the key third day of feeding by nymphal Ixodes scapularis(Acari: Ixodidae). J Med Entomol 2008;45:732e6.

[91] Kahl O, Janetzki-Mittmann C, Gray JS, Jonas R, Stein J, de Boer R. Risk ofinfection with Borrelia burgdorferi sensu lato for a host in relation to theduration of nymphal Ixodes ricinus feeding and the method of tick removal.Zentralbl Bakteriol 1998;287:41e52.

[92] Crippa M, Rais O, Gern L. Investigations on the mode and dynamics oftransmission and infectivity of Borrelia burgdorferi sensu stricto and Borreliaafzelii in Ixodes ricinus ticks. Vector Borne Zoonotic Dis 2002;2:3e9.

[93] Korenberg EI, Moskvitina GG. Interrelationships between different Borreliagenospecies and their principal vectors. J Vector Ecol 1996;21:178e85.

[94] Lebet N, Gern L. Histological examination of Borrelia burgdorferi infections inunfed Ixodes ricinus nymphs. Exp Appl Acarol 1994;18:177e83.

[95] Leuba-Garcia S, Kramer MD, Wallich R, Gern L. Characterization of Borreliaburgdorferi isolated from different organs of Ixodes ricinus ticks collected innature. Zentralbl Bakteriol 1994;280:468e75.

[96] Piesman J. Dispersal of the Lyme disease spirochete Borrelia burgdorferi tosalivary glands of feeding nymphal Ixodes scapularis (Acari: Ixodidae). J MedEntomol 1995;32:519e21.

[97] Piesman J, Schneider BS, Zeidner NS. Use of quantitative PCR to measuredensity of Borrelia burgdorferi in the midgut and salivary glands of feedingtick vectors. J Clin Microbiol 2001;39:4145e8.

[98] Lima CM, Zeidner NS, Beard CB, Soares CA, Dolan MC, Dietrich G, et al. Dif-ferential infectivity of the Lyme disease spirochete Borrelia burgdorferiderived from Ixodes scapularis salivary glands and midgut. J Med Entomol2005;42:506e10.

[99] Hanson MS, Cassatt DR, Guo BP, Patel NK, McCarthy MP, Dorward DW, et al.Active and passive immunity against Borrelia burgdorferi decorin bindingprotein A (DbpA) protects against infection. Infect Immun 1998;66:2143e53.

[100] Lazarus JJ, Meadows MJ, Lintner RE, Wooten RM. IL-10 deficiency promotesincreased Borrelia burgdorferi clearance predominantly through enhancedinnate immune responses. J Immunol 2006;177:7076e85.

[101] Nowakowski J, McKenna D, Nadelman RB, Bittker S, Cooper D, Pavia C, et al.Blood cultures for patients with extracutaneous manifestations of Lymedisease in the United States. Clin Infect Dis 2009;49:1733e5.

[102] Nowakowski J, Schwartz I, Liveris D, Wang G, Aguero-Rosenfeld ME, Girao G,et al. Laboratory diagnostic techniques for patients with early Lyme diseaseassociated with erythema migrans: a comparison of different techniques.Clin Infect Dis 2001;33:2023e7.

[103] Wormser GP, McKenna D, Carlin J, Nadelman RB, Cavaliere LF, Holmgren D,et al. Brief communication: hematogenous dissemination in early Lymedisease. Ann Intern Med 2005;142:751e5.

[104] Cerar T, Ogrinc K, Cimperman J, Lotric-Furlan S, Strle F, Ruzic-Sabljic E.Validation of cultivation and PCR methods for diagnosis of Lyme neuro-borreliosis. J Clin Microbiol 2008;46:3375e9.

[105] Maraspin V, Ruzic-Sabljic E, Cimperman J, Lotric-Furlan S, Jurca T, Picken RN,et al. Isolation of Borrelia burgdorferi sensu lato from blood of patients witherythema migrans. Infection 2001;29:65e70.

[106] Maraspin V, Ogrinc K, Ruzic-Sabljic E, Lotric-Furlan S, Strle F. Isolation ofBorrelia burgdorferi sensu lato from blood of adult patients with borreliallymphocytoma, Lyme neuroborreliosis, Lyme arthritis and acrodermatitischronica atrophicans. Infection 2011;39:35e40.

[107] Centers for Disease Control and Prevention, National Notifiable DiseasesSurveillance System (NNDSS). Lyme disease (Borrelia burgdorferi) 2011case definition. 2011. http://wwwncdcgov/NNDSS/script/casedefaspx?CondYrID¼752&DatePub¼1/1/2011 [accessed 14.02.14].

[108] Liveris D, Wang G, Girao G, Byrne DW, Nowakowski J, McKenna D, et al.Quantitative detection of Borrelia burgdorferi in 2-millimeter skin samples oferythema migrans lesions: correlation of results with clinical and laboratoryfindings. J Clin Microbiol 2002;40:1249e53.

[109] Carlsson SA, Granlund H, Jansson C, Nyman D, Wahlberg P. Characteristics oferythema migrans in Borrelia afzelii and Borrelia garinii infections. Scand JInfect Dis 2003;35:31e3.

[110] Strle F, Ruzic-Sabljic E, Cimperman J, Lotric-Furlan S, Maraspin V. Compari-son of findings for patients with Borrelia garinii and Borrelia afzelii isolatedfrom cerebrospinal fluid. Clin Infect Dis 2006;43:704e10.

[111] van Dam AP, Kuiper H, Vos K, Widjojokusumo A, de Jongh BM, Spanjaard L,et al. Different genospecies of Borrelia burgdorferi are associated with distinctclinical manifestations of Lyme borreliosis. Clin Infect Dis 1993;17:708e17.

[112] Schmidli J, Hunziker T, Moesli P, Schaad UB. Cultivation of Borrelia burg-dorferi from joint fluid three months after treatment of facial palsy due toLyme borreliosis. J Infect Dis 1988;158:905e6.

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 25

[113] Preac Mursic V, Marget W, Busch U, Pleterski Rigler D, Hagl S. Kill kinetics ofBorrelia burgdorferi and bacterial findings in relation to the treatment ofLyme borreliosis. Infection 1996;24:9e16.

[114] O'Rourke M, Traweger A, Lusa L, Stupica D, Maraspin V, Barrett PN, et al.Quantitative detection of Borrelia burgdorferi sensu lato in erythema migransskin lesions using internally controlled duplex real time PCR. PLoS One2013;8:e63968.

[115] Lebech AM. Polymerase chain reaction in diagnosis of Borrelia burgdorferiinfections and studies on taxonomic classification. APMIS Suppl 2002:1e40.

[116] Nocton JJ, Bloom BJ, Rutledge BJ, Persing DH, Logigian EL, Schmid CH, et al.Detection of Borrelia burgdorferi DNA by polymerase chain reaction in ce-rebrospinal fluid in Lyme neuroborreliosis. J Infect Dis 1996;174:623e7.

[117] Nocton JJ, Dressler F, Rutledge BJ, Rys PN, Persing DH, Steere AC. Detection ofBorrelia burgdorferi DNA by polymerase chain reaction in synovial fluid frompatients with Lyme arthritis. N Engl J Med 1994;330:229e34.

[118] Jones KL, McHugh GA, Glickstein LJ, Steere AC. Analysis of Borrelia burgdorferigenotypes in patients with Lyme arthritis: high frequency of ribosomal RNAintergenic spacer type 1 strains in antibiotic-refractory arthritis. ArthritisRheum 2009;60:2174e82.

[119] Recommendations for test performance and interpretation from the SecondNational Conference on Serologic Diagnosis of Lyme Disease. MMWR MorbMortal Wkly Rep 1995;44:590e1.

[120] Wilske B, Fingerle V, Schulte-Spechtel U. Microbiological and serologicaldiagnosis of Lyme borreliosis. FEMS Immunol MedMicrobiol 2007;49:13e21.

[121] Dressler F, Ackermann R, Steere AC. Antibody responses to the threegenomic groups of Borrelia burgdorferi in European Lyme borreliosis. J InfectDis 1994;169:313e8.

[122] Aguero-Rosenfeld ME, Nowakowski J, McKenna DF, Carbonaro CA,Wormser GP. Serodiagnosis in early Lyme disease. J Clin Microbiol 1993;31:3090e5.

[123] Engstrom SM, Shoop E, Johnson RC. Immunoblot interpretation criteria forserodiagnosis of early Lyme disease. J Clin Microbiol 1995;33:419e27.

[124] Steere AC, McHugh G, Damle N, Sikand VK. Prospective study of serologictests for Lyme disease. Clin Infect Dis 2008;47:188e95.

[125] Glatz M, Fingerle V, Wilske B, Ambros-Rudolph C, Kerl H, Müllegger RR.Immunoblot analysis of the seroreactivity to recombinant Borrelia burgdor-feri sensu lato antigens, including VlsE, in the long-term course of treatedpatients with erythema migrans. Dermatology 2008;216:93e103.

[126] Aguero-Rosenfeld ME, Nowakowski J, Bittker S, Cooper D, Nadelman RB,Wormser GP. Evolution of the serologic response to Borrelia burgdorferi intreated patients with culture-confirmed erythema migrans. J Clin Microbiol1996;34:1e9.

[127] Craft JE, Grodzicki RL, Shrestha M, Fischer DK, Garcia-Blanco M, Steere AC.The antibody response in Lyme disease. Yale J Biol Med 1984;57:561e5.

[128] Tveitnes D, Oymar K, Natas O. Laboratory data in children with Lyme neu-roborreliosis, relation to clinical presentation and duration of symptoms.Scand J Infect Dis 2009;41:355e62.

[129] Cerar T, Ogrinc K, Strle F, Ruzic-Sabljic E. Humoral immune responses inpatients with Lyme neuroborreliosis. Clin Vaccine Immunol 2010;17:645e50.

[130] Stanek G, Breier F, Menzinger G, Schaar B, Hafner M, Partsch H. Erythemamigrans and serodiagnosis by enzyme immunoassay and immunoblot withthree Borrelia species. Wien Klin Wochenschr 1999;111:951e6.

[131] Dressler F, Whalen JA, Reinhardt BN, Steere AC. Western blotting in theserodiagnosis of Lyme disease. J Infect Dis 1993;167:392e400.

[132] Xu Y, Bruno JF, Luft BJ. Profiling the humoral immune response to Borreliaburgdorferi infection with protein microarrays. Microb Pathog 2008;45:403e7.

[133] Goettner G, Schulte-Spechtel U, Hillermann R, Liegl G, Wilske B, Fingerle V.Improvement of Lyme borreliosis serodiagnosis by a newly developed re-combinant immunoglobulin G (IgG) and IgM line immunoblot assay andaddition of VlsE and DbpA homologues. J Clin Microbiol 2005;43:3602e9.

[134] Kalish RA, Leong JM, Steere AC. Early and late antibody responses to full-length and truncated constructs of outer surface protein A of Borrelia burg-dorferi in Lyme disease. Infect Immun 1995;63:2228e35.

[135] Hammers-Berggren S, Lebech AM, Karlsson M, Svenungsson B, Hansen K,Stiernstedt G. Serological follow-up after treatment of patients with ery-thema migrans and neuroborreliosis. J Clin Microbiol 1994;32:1519e25.

[136] Hilton E, DeVoti J, Benach JL, Halluska ML, White DJ, Paxton H, et al. Sero-prevalence and seroconversion for tick-borne diseases in a high-risk popu-lation in the northeast United States. Am J Med 1999;106:404e9.

[137] Smith PF, Benach JL, White DJ, Stroup DF, Morse DL. Occupational risk ofLyme disease in endemic areas of New York State. Ann N Y Acad Sci1988;539:289e301.

[138] Krause PJ, Telford 3rd SR, Spielman A, Sikand V, Ryan R, Christianson D, et al.Concurrent Lyme disease and babesiosis. Evidence for increased severity andduration of illness. JAMA 1996;275:1657e60.

[139] Tjernberg I, Kruger G, Eliasson I. C6 peptide ELISA test in the serodiagnosis ofLyme borreliosis in Sweden. Eur J Clin Microbiol Infect Dis 2007;26:37e42.

[140] Dehnert M, Fingerle V, Klier C, Talaska T, Schlaud M, Krause G, et al. Sero-positivity of Lyme borreliosis and associated risk factors: a population-basedstudy in children and adolescents in Germany (KiGGS). PLoS One 2012;7:e41321.

[141] Skogman BH, Ekerfelt C, Ludvigsson J, Forsberg P. Seroprevalence of BorreliaIgG antibodies among young Swedish children in relation to reported tick

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

bites, symptoms and previous treatment for Lyme borreliosis: a population-based survey. Arch Dis Child 2010;95:1013e6.

[142] Hao Q, Geng Z, Hou XX, Tian Z, Yang XJ, Jiang WJ, et al. Seroepidemiologicalinvestigation of Lyme disease and human granulocytic anaplasmosis amongpeople living in forest areas of eight provinces in China. Biomed Environ Sci2013;26:185e9.

[143] Lakos A, Igari Z, Solymosi N. Recent lesson from a clinical and seroepide-miological survey: low positive predictive value of Borrelia burgdorferiantibody testing in a high risk population. Adv Med Sci 2012;57:356e63.

[144] Henningsson AJ, Christiansson M, Tjernberg I, Lofgren S, Matussek A. Labo-ratory diagnosis of Lyme neuroborreliosis: a comparison of three CSF anti-Borrelia antibody assays. Eur J Clin Microbiol Infect Dis 2013;33:797e803.

[145] Ogrinc K, Lotric-Furlan S, Maraspin V, Lusa L, Cerar T, Ruzic-Sabljic E, et al.Suspected early Lyme neuroborreliosis in patients with erythema migrans.Clin Infect Dis 2013;57:501e9.

[146] Stanek G, Lusa L, Ogrinc K, Markowicz M, Strle F. Intrathecally produced IgGand IgM antibodies to recombinant VlsE, VlsE peptide, recombinant OspCand whole cell extracts in the diagnosis of Lyme neuroborreliosis. MedMicrobiol Immunol 2013;203:125e32.

[147] Burbelo PD, Issa AT, Ching KH, Cohen JI, Iadarola MJ, Marques A. Rapid,simple, quantitative, and highly sensitive antibody detection for Lyme dis-ease. Clin Vaccine Immunol 2010;17:904e9.

[148] Skogman BH, Croner S, Forsberg P, Ernerudh J, Lahdenne P, Sillanpaa H, et al.Improved laboratory diagnostics of Lyme neuroborreliosis in children bydetection of antibodies to new antigens in cerebrospinal fluid. Pediatr InfectDis J 2008;27:605e12.

[149] Schulte-Spechtel U, Fingerle V, Goettner G, Rogge S, Wilske B. Molecularanalysis of decorin-binding protein A (DbpA) reveals five major groupsamong European Borrelia burgdorferi sensu lato strains with impact for thedevelopment of serological assays and indicates lateral gene transfer of thedbpA gene. Int J Med Microbiol 2006;296(Suppl. 40):250e66.

[150] Bacon RM, Biggerstaff BJ, Schriefer ME, Gilmore Jr RD, Philipp MT, Steere AC,et al. Serodiagnosis of Lyme disease by kinetic enzyme-linked immunosor-bent assay using recombinant VlsE1 or peptide antigens of Borrelia burg-dorferi compared with 2-tiered testing using whole-cell lysates. J Infect Dis2003;187:1187e99.

[151] Heikkil€a T, Sepp€al€a I, Saxen H, Panelius J, Yrj€an€ainen H, Lahdenne P. Species-specific serodiagnosis of Lyme arthritis and neuroborreliosis due to Borreliaburgdorferi sensu stricto, B. afzelii, and B. garinii by using decorin bindingprotein A. J Clin Microbiol 2002;40:453e60.

[152] Branda JA, Linskey K, Kim YA, Steere AC, Ferraro MJ. Two-tiered antibodytesting for Lyme disease with use of 2 enzyme immunoassays, a whole-cellsonicate enzyme immunoassay followed by a VlsE C6 peptide enzymeimmunoassay. Clin Infect Dis 2011;53:541e7.

[153] Robertson J, Guy E, Andrews N, Wilske B, Anda P, Granstrom M, et al.A European multicenter study of immunoblotting in serodiagnosis of Lymeborreliosis. J Clin Microbiol 2000;38:2097e102.

[154] Jansson C, Carlsson SA, Granlund H, Wahlberg P, Nyman D. Analysis ofBorrelia burgdorferi IgG antibodies with a combination of IgG ELISA and VlsEC6 peptide ELISA. Clin Microbiol Infect 2005;11:147e50.

[155] Liang FT, Steere AC, Marques AR, Johnson BJ, Miller JN, Philipp MT. Sensitiveand specific serodiagnosis of Lyme disease by enzyme-linked immunosor-bent assay with a peptide based on an immunodominant conserved regionof Borrelia burgdorferi VlsE. J Clin Microbiol 1999;37:3990e6.

[156] Liang FT, Alvarez AL, Gu Y, Nowling JM, Ramamoorthy R, Philipp MT. Animmunodominant conserved region within the variable domain of VlsE, thevariable surface antigen ofBorrelia burgdorferi. J Immunol 1999;163:5566e73.

[157] Tjernberg I, Sillanp€a€a H, Sepp€al€a I, Eliasson I, Forsberg P, Lahdenne P. Antibodyresponses to borrelia IR(6) peptide variants and the C6 peptide in Swedishpatients with erythema migrans. Int J Med Microbiol 2009;299:439e46.

[158] Branda JA, Strle F, Strle K, Sikand N, Ferraro MJ, Steere AC. Performance ofUnited States serologic assays in the diagnosis of Lyme borreliosis acquiredin Europe. Clin Infect Dis 2013;57:333e40.

[159] Centers for Disease Control and Prevention. Clinical manifestations ofconfirmed Lyme disease cases e United States, 2001-2010. 2013. www.cdc.gov/lyme/stats/chartstables/casesbysymptom.html [accessed 15.03.04].

[160] Gerber MA, Shapiro ED, Burke GS, Parcells VJ, Bell GL. Lyme disease inchildren in southeastern Connecticut. Pediatric Lyme Disease Study Group.N Engl J Med 1996;335:1270e4.

[161] Wormser GP, McKenna D, Nadelman RB, Nowakowski J, Weinstein A. Lymedisease in children. N Engl J Med 1997;336:1107. author reply e 8.

[162] Sigal LH, Zahradnik JM, Lavin P, Patella SJ, Bryant G, Haselby R, et al.A vaccine consisting of recombinant Borrelia burgdorferi outer-surface pro-tein A to prevent Lyme disease. Recombinant Outer-Surface Protein A LymeDisease Vaccine Study Consortium. N Engl J Med 1998;339:216e22.

[163] Christova I, Komitova R. Clinical and epidemiological features of Lyme bor-reliosis in Bulgaria. Wien Klin Wochenschr 2004;116:42e6.

[164] Nadelman RB, Nowakowski J, Forseter G, Goldberg NS, Bittker S, Cooper D,et al. The clinical spectrum of early Lyme borreliosis in patients with culture-confirmed erythema migrans. Am J Med 1996;100:502e8.

[165] Tibbles CD, Edlow JA. Does this patient have erythema migrans? JAMA2007;297:2617e27.

[166] Strle F, Ruzic-Sabljic E, Logar M, Maraspin V, Lotric-Furlan S, Cimperman J,et al. Comparison of erythema migrans caused by Borrelia burgdorferi andBorrelia garinii. Vector Borne Zoonotic Dis 2011;11:1253e8.

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3426

[167] Strle F, Nadelman RB, Cimperman J, Nowakowski J, Picken RN, Schwartz I,et al. Comparison of culture-confirmed erythema migrans caused by Borreliaburgdorferi sensu stricto in New York State and by Borrelia afzelii in Slovenia.Ann Intern Med 1999;130:32e6.

[168] Logar M, Ruzic-Sabljic E, Maraspin V, Lotric-Furlan S, Cimperman J, Jurca T,et al. Comparison of erythema migrans caused by Borrelia afzelii and Borreliagarinii. Infection 2004;32:15e9.

[169] Zajkowska J, Hermanowska-Szpakowicz T, Coyle P, Ostrowska J, Pancewicz S,Kondrusik M. Comparative study of early Lyme disease: erythema migrans inNew York state and northeastern Poland. Med Sci Monit 2002;8:CR37e43.

[170] Strle F, Wormser GP, Mead P, Dhaduvai K, Longo MV, Adenikinju O, et al.Gender disparity between cutaneous and non-cutaneous manifestations ofLyme borreliosis. PLoS One 2013;8:e64110.

[171] Asbrink E, Hovmark A, Olsson I. Clinical manifestations of acrodermatitischronica atrophicans in 50 Swedish patients. Zentralbl Bakteriol MikrobiolHyg A 1986;263:253e61.

[172] Dhote R, Basse-Guerineau AL, Beaumesnil V, Christoforov B, Assous MV. Fullspectrum of clinical, serological, and epidemiological features of complicatedforms of Lyme borreliosis in the Paris, France, area. Eur J Clin Microbiol InfectDis 2000;19:809e15.

[173] Ai CX, Wen YX, Zhang YG, Wang SS, Qiu QC, Shi ZX, et al. Clinical manifes-tations and epidemiological characteristics of Lyme disease in Hailin county,Heilongjiang Province, China. Ann N Y Acad Sci 1988;539:302e13.

[174] Steere AC, Taylor E, Wilson ML, Levine JF, Spielman A. Longitudinal assess-ment of the clinical and epidemiological features of Lyme disease in adefined population. J Infect Dis 1986;154:295e300.

[175] Müllegger RR, Glatz M. Skin manifestations of Lyme borreliosis: diagnosisand management. Am J Clin Dermatol 2008;9:355e68.

[176] Kindstrand E, Nilsson BY, Hovmark A, Pirskanen R, Asbrink E. Peripheralneuropathy in acrodermatitis chronica atrophicans e a late Borrelia mani-festation. Acta Neurol Scand 1997;95:338e45.

[177] Asbrink E, Hovmark A. Early and late cutaneous manifestations in Ixodes-borne borreliosis (erythema migrans borreliosis, Lyme borreliosis). Ann N YAcad Sci 1988;539:4e15.

[178] Brehmer-Andersson E, Hovmark A, Asbrink E. Acrodermatitis chronicaatrophicans: histopathologic findings and clinical correlations in 111 cases.Acta Derm Venereol 1998;78:207e13.

[179] Kristoferitsch W, Sluga E, Graf M, Partsch H, Neumann R, Stanek G, et al.Neuropathy associated with acrodermatitis chronica atrophicans. Clinicaland morphological features. Ann N Y Acad Sci 1988;539:35e45.

[180] Buechner SA, Rufli T, Erb P. Acrodermatitis chronic atrophicans: a chronic T-cell-mediated immune reaction against Borrelia burgdorferi? Clinical, histo-logic, and immunohistochemical study of five cases. J Am Acad Dermatol1993;28:399e405.

[181] Müllegger RR, McHugh G, Ruthazer R, Binder B, Kerl H, Steere AC. Differentialexpression of cytokine mRNA in skin specimens from patients with ery-thema migrans or acrodermatitis chronica atrophicans. J Invest Dermatol2000;115:1115e23.

[182] de Koning J, Tazelaar DJ, Hoogkamp-Korstanje JA, Elema JD. Acrodermatitischronica atrophicans: a light and electron microscopic study. J Cutan Pathol1995;22:23e32.

[183] Lipsker D, Hansmann Y, Limbach F, Clerc C, Tranchant C, Grunenberger F,et al. Disease expression of Lyme borreliosis in northeastern France. Eur JClin Microbiol Infect Dis 2001;20:225e30.

[184] Maraspin V, Cimperman J, Lotric-Furlan S, Ruzic-Sabljic E, Jurca T, Picken RN,et al. Solitary borrelial lymphocytoma in adult patients. Wien KlinWochenschr 2002;114:515e23.

[185] Strle F, Pleterski-Rigler D, Stanek G, Pejovnik-Pustinek A, Ruzic E,Cimperman J. Solitary borrelial lymphocytoma: report of 36 cases. Infection1992;20:201e6.

[186] Colli C, Leinweber B, Mullegger R, Chott A, Kerl H, Cerroni L. Borrelia burg-dorferi-associated lymphocytoma cutis: clinicopathologic, immunopheno-typic, and molecular study of 106 cases. J Cutan Pathol 2004;31:232e40.

[187] Hansen K, Lebech AM. The clinical and epidemiological profile of Lymeneuroborreliosis in Denmark 1985-1990. A prospective study of 187 patientswith Borrelia burgdorferi specific intrathecal antibody production. Brain1992;115(Pt 2):399e423.

[188] Oschmann P, Dorndorf W, Hornig C, Schafer C, Wellensiek HJ,Pflughaupt KW. Stages and syndromes of neuroborreliosis. J Neurol1998;245:262e72.

[189] Christen HJ, Hanefeld F, Eiffert H, Thomssen R. Epidemiology and clinicalmanifestations of Lyme borreliosis in childhood. A prospective multicentrestudy with special regard to neuroborreliosis. Acta Paediatr Suppl 1993;386:1e75.

[190] Øymar K, Tveitnes D. Clinical characteristics of childhood Lyme neuro-borreliosis in an endemic area of northern Europe. Scand J Infect Dis2009;41:88e94.

[191] Bremell D, Hagberg L. Clinical characteristics and cerebrospinal fluid pa-rameters in patients with peripheral facial palsy caused by Lyme neuro-borreliosis compared with facial palsy of unknown origin (Bell's palsy). BMCInfect Dis 2011;11:215.

[192] Sundin M, Hansson ME, Engman ML, Orvell C, Lindquist L, Wide K, et al.Pediatric tick-borne infections of the central nervous system in an endemicregion of Sweden: a prospective evaluation of clinical manifestations. Eur JPediatr 2012;171:347e52.

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

[193] Asbrink E, Olsson I, Hovmark A. Erythema chronicum migrans Afzelius inSweden. A study on 231 patients. Zentralbl Bakteriol Mikrobiol Hyg A1986;263:229e36.

[194] Steere AC, Sikand VK. The presenting manifestations of Lyme disease and theoutcomes of treatment. N Engl J Med 2003;348:2472e4.

[195] Stanek G, Flamm H, Groh V, Hirschl A, Kristoferitsch W, Neumann R, et al.Epidemiology of borrelia infections in Austria. Zentralbl Bakteriol MikrobiolHyg A 1987;263:442e9.

[196] Henningsson AJ, Malmvall BE, Ernerudh J, Matussek A, Forsberg P. Neuro-borreliosis e an epidemiological, clinical and healthcare cost study from anendemic area in the south-east of Sweden. Clin Microbiol Infect 2010;16:1245e51.

[197] Berglund J, Stjernberg L, Ornstein K, Tykesson-Joelsson K, Walter H. 5-yFollow-up study of patients with neuroborreliosis. Scand J Infect Dis2002;34:421e5.

[198] Garin C, Bujadoux A. Paralysie par les tiques [Paralysis by ticks]. J M�ed Lyon1922;71:765e7;a translation is published in Clin Infect Dis 1993;16(1):168e9.

[199] Bannwarth A. Chronische lymphocyt€are Meningitis, endzündliche Poly-neuritis und “Rheumatismus”. Ein Beitrag zum Problem “Allergie und Ner-vensystem” in zwei Teilen [Chronic lymphocytic meningitis, inflammatorypolyneuritis and “rheumatism”. A contribution on the problem of “allergyand the nervous system” in two parts]. Arch für Psychiatr Nervenkrankh1941;113:284e376.

[200] Reik L, Steere AC, Bartenhagen NH, Shope RE, Malawista SE. Neurologicabnormalities of Lyme disease. Med Baltim 1979;58:281e94.

[201] Pachner AR, Steere AC. The triad of neurologic manifestations of Lyme dis-ease: meningitis, cranial neuritis, and radiculoneuritis. Neurology 1985;35:47e53.

[202] Greco A, Gallo A, Fusconi M, Marinelli C, Macri GF, de Vincentiis M. Bell'spalsy and autoimmunity. Autoimmun Rev 2012;12:323e8.

[203] Mygland A, Ljøstad U, Fingerle V, Rupprecht T, Schmutzhard E, Steiner I.EFNS guidelines on the diagnosis and management of European Lyme neu-roborreliosis. Eur J Neurol 2010;17:8e16. e1e4.

[204] Skogman BH, Croner S, Nordwall M, Eknefelt M, Ernerudh J, Forsberg P. Lymeneuroborreliosis in children: a prospective study of clinical features, prog-nosis, and outcome. Pediatr Infect Dis J 2008;27:1089e94.

[205] Broekhuijsen-van Henten DM, Braun KP, Wolfs TF. Clinical presentation ofchildhood neuroborreliosis; neurological examination may be normal. ArchDis Child 2010;95:910e4.

[206] Belman AL, Iyer M, Coyle PK, Dattwyler R. Neurologic manifestations inchildren with North American Lyme disease. Neurology 1993;43:2609e14.

[207] Bingham PM, Galetta SL, Athreya B, Sladky J. Neurologic manifestations inchildren with Lyme disease. Pediatrics 1995;96:1053e6.

[208] Cook SP, Macartney KK, Rose CD, Hunt PG, Eppes SC, Reilly JS. Lymedisease and seventh nerve paralysis in children. Am J Otolaryngol1997;18:320e3.

[209] Tveitnes D, Øymar K, Natas O. Acute facial nerve palsy in children: how oftenis it Lyme borreliosis? Scand J Infect Dis 2007;39:425e31.

[210] Kan L, Sood SK, Maytal J. Pseudotumor cerebri in Lyme disease: a case reportand literature review. Pediatr Neurol 1998;18:439e41.

[211] Logigian EL, Kaplan RF, Steere AC. Chronic neurologic manifestations of Lymedisease. N Engl J Med 1990;323:1438e44.

[212] Halperin JJ, Krupp LB, Golightly MG, Volkman DJ. Lyme borreliosis-associatedencephalopathy. Neurology 1990;40:1340e3.

[213] Halperin JJ, Luft BJ, Anand AK, Roque CT, Alvarez O, Volkman DJ, et al. Lymeneuroborreliosis: central nervous system manifestations. Neurology1989;39:753e9.

[214] Halperin JJ. Nervous system Lyme disease. Handb Clin Neurol 2014;121:1473e83.

[215] Halperin J, Luft BJ, Volkman DJ, Dattwyler RJ. Lyme neuroborreliosis. Pe-ripheral nervous system manifestations. Brain 1990;113(Pt 4):1207e21.

[216] Kindstrand E, Nilsson BY, Hovmark A, Nennesmo I, Pirskanen R, Solders G,et al. Polyneuropathy in late Lyme borreliosis e a clinical, neurophysiologicaland morphological description. Acta Neurol Scand 2000;101:47e52.

[217] Ackermann R, Rehse-Kupper B, Gollmer E, Schmidt R. Chronic neurologicmanifestations of erythema migrans borreliosis. Ann N Y Acad Sci 1988;539:16e23.

[218] Kaiser R. False-negative serology in patients with neuroborreliosis and thevalue of employing of different borrelial strains in serological assays. J MedMicrobiol 2000;49:911e5.

[219] Steere AC, Berardi VP, Weeks KE, Logigian EL, Ackermann R. Evaluation of theintrathecal antibody response to Borrelia burgdorferi as a diagnostic test forLyme neuroborreliosis. J Infect Dis 1990;161:1203e9.

[220] Luft BJ, Steinman CR, Neimark HC, Muralidhar B, Rush T, Finkel MF, et al.Invasion of the central nervous system by Borrelia burgdorferi in acutedisseminated infection. JAMA 1992;267:1364e7.

[221] Wormser GP, Dattwyler RJ, Shapiro ED, Halperin JJ, Steere AC, Klempner MS,et al. The clinical assessment, treatment, and prevention of Lyme disease,human granulocytic anaplasmosis, and babesiosis: clinical practice guide-lines by the Infectious Diseases Society of America. Clin Infect Dis 2006;43:1089e134.

[222] Krüger H, Reuss K, Pulz M, Rohrbach E, Pflughaupt KW, Martin R, et al.Meningoradiculitis and encephalomyelitis due to Borrelia burgdorferi: afollow-up study of 72 patients over 27 years. J Neurol 1989;236:322e8.

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 27

[223] Blanc F, Jaulhac B, Fleury M, de Seze J, de Martino SJ, Remy V, et al. Relevanceof the antibody index to diagnose Lyme neuroborreliosis among seropositivepatients. Neurology 2007;69:953e8.

[224] Ljøstad U, Skarpaas T, Mygland A. Clinical usefulness of intrathecal antibodytesting in acute Lyme neuroborreliosis. Eur J Neurol 2007;14:873e6.

[225] Kaiser R. Variable CSF findings in early and late Lyme neuroborreliosis: afollow-up study in 47 patients. J Neurol 1994;242:26e36.

[226] Hammers-Berggren S, Hansen K, Lebech AM, Karlsson M. Borrelia burgdor-feri-specific intrathecal antibody production in neuroborreliosis: a follow-upstudy. Neurology 1993;43:169e75.

[227] Chang C, Gershwin ME. Integrative medicine in allergy and immunology.Clin Rev Allergy Immunol 2013;44:208e28.

[228] Ljøstad U, Skogvoll E, Eikeland R, Midgard R, Skarpaas T, Berg A, et al. Oraldoxycycline versus intravenous ceftriaxone for European Lyme neuro-borreliosis: a multicentre, non-inferiority, double-blind, randomised trial.Lancet Neurol 2008;7:690e5.

[229] Steere AC, Batsford WP, Weinberg M, Alexander J, Berger HJ, Wolfson S, et al.Lyme carditis: cardiac abnormalities of Lyme disease. Ann Intern Med1980;93:8e16.

[230] van der Linde MR. Lyme carditis: clinical characteristics of 105 cases. Scand JInfect Dis Suppl 1991;77:81e4.

[231] Costello JM, Alexander ME, Greco KM, Perez-Atayde AR, Laussen PC. Lymecarditis in children: presentation, predictive factors, and clinical course.Pediatrics 2009;123:e835e41.

[232] Rubin DA, Sorbera C, Nikitin P, McAllister A, Wormser GP, Nadelman RB.Prospective evaluation of heart block complicating early Lyme disease.Pacing Clin Electrophysiol 1992;15:252e5.

[233] Williams CL, Strobino B, Lee A, Curran AS, Benach JL, Inamdar S, et al. Lymedisease in childhood: clinical and epidemiologic features of ninety cases.Pediatr Infect Dis J 1990;9:10e4.

[234] Ni XB, Jia N, Jiang BG, Sun T, Zheng YC, Huo QB, et al. Lyme borreliosis causedby diverse genospecies of Borrelia burgdorferi sensu lato in northeasternChina. Clin Microbiol Infect 2014;20:808e14.

[235] Agmon-Levin N, Selmi C. The autoimmune side of heart and lung diseases.Clin Rev Allergy Immunol 2013;44:1e5.

[236] Ando Y, Yang GX, Kenny TP, Kawata K, Zhang W, Huang W, et al. Over-expression of microRNA-21 is associated with elevated pro-inflammatorycytokines in dominant-negative TGF-beta receptor type II mouse.J Autoimmun 2013;41:111e9.

[237] McAlister HF, Klementowicz PT, Andrews C, Fisher JD, Feld M, Furman S.Lyme carditis: an important cause of reversible heart block. Ann Intern Med1989;110:339e45.

[238] Sigal LH. Early disseminated Lyme disease: cardiac manifestations. Am J Med1995;98:25Se8S. discussion 8Se9S.

[239] Midttun M, Lebech AM, Hansen K, Videbaek J. Lyme carditis: a clinical pre-sentation and long time follow-up. Scand J Infect Dis 1997;29:153e7.

[240] Three sudden cardiac deaths associated with Lyme carditis e United States,November 2012-July 2013. MMWR Morb Mortal Wkly Rep 2013;62:993e6.

[241] Tavora F, Burke A, Li L, Franks TJ, Virmani R. Postmortem confirmation of Lymecarditis with polymerase chain reaction. Cardiovasc Pathol 2008;17:103e7.

[242] Cary NR, Fox B, Wright DJ, Cutler SJ, Shapiro LM, Grace AA. Fatal Lyme car-ditis and endodermal heterotopia of the atrioventricular node. Postgrad MedJ 1990;66:134e6.

[243] Marcus LC, Steere AC, Duray PH, Anderson AE, Mahoney EB. Fatal pancarditisin a patient with coexistent Lyme disease and babesiosis. Demonstration ofspirochetes in the myocardium. Ann Intern Med 1985;103:374e6.

[244] Stanek G, Klein J, Bittner R, Glogar D. Isolation of Borrelia burgdorferi from themyocardium of a patient with longstanding cardiomyopathy. N Engl J Med1990;322:249e52.

[245] Kubanek M, Sramko M, Berenova D, Hulinska D, Hrbackova H, Maluskova J,et al. Detection of Borrelia burgdorferi sensu lato in endomyocardial biopsyspecimens in individuals with recent-onset dilated cardiomyopathy. Eur JHeart Fail 2012;14:588e96.

[246] Dubrey SW, Bhatia A, Woodham S, Rakowicz W. Lyme disease in the UnitedKingdom. Postgrad Med J 2014;90:33e42.

[247] Daikh BE, Emerson FE, Smith RP, Lucas FL, McCarthy CA. Lyme arthritis: acomparison of presentation, synovial fluid analysis, and treatment course inchildren and adults. Arthritis Care Res Hob 2013;65:1986e90.

[248] Thompson A, Mannix R, Bachur R. Acute pediatric monoarticular arthritis:distinguishing Lyme arthritis from other etiologies. Pediatrics 2009;123:959e65.

[249] Huppertz HI, Karch H, Suschke HJ, Doring E, Ganser G, Thon A, et al. Lymearthritis in European children and adolescents. The Pediatric RheumatologyCollaborative Group. Arthritis Rheum 1995;38:361e8.

[250] Rose CD, Fawcett PT, Eppes SC, Klein JD, Gibney K, Doughty RA. PediatricLyme arthritis: clinical spectrum and outcome. J Pediatr Orthop 1994;14:238e41.

[251] Tory HO, Zurakowski D, Sundel RP. Outcomes of children treated for Lymearthritis: results of a large pediatric cohort. J Rheumatol 2010;37:1049e55.

[252] Deanehan JK, Kimia AA, Tan Tanny SP, Milewski MD, Talusan PG, Smith BG,et al. Distinguishing Lyme from septic knee monoarthritis in Lyme disease-endemic areas. Pediatrics 2013;131:e695e701.

[253] Steere AC, Dhar A, Hernandez J, Fischer PA, Sikand VK, Schoen RT, et al.Systemic symptoms without erythema migrans as the presenting picture ofearly Lyme disease. Am J Med 2003;114:58e62.

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

[254] Steere AC, Sikand VK, Meurice F, Parenti DL, Fikrig E, Schoen RT, et al.Vaccination against Lyme disease with recombinant Borrelia burgdorferiouter-surface lipoprotein A with adjuvant. Lyme Disease Vaccine StudyGroup. N Engl J Med 1998;339:209e15.

[255] Berglund J, Hansen BU, Eitrem R. Lyme arthritis e a common manifestationin a highly endemic area in Sweden. J Rheumatol 1995;22:695e701.

[256] Gerber MA, Zemel LS, Shapiro ED. Lyme arthritis in children: clinicalepidemiology and long-term outcomes. Pediatrics 1998;102:905e8.

[257] Schoen RT. Acute inflammatory arthritis: monoarthritis risk stratification inLyme disease. Nat Rev Rheumatol 2013;9:261e2.

[258] Saulsbury FT. Lyme arthritis in 20 children residing in a non-endemic area.Clin Pediatr (Phila) 2005;44:419e21.

[259] Hammers-Berggren S, Andersson U, Stiernstedt G. Borrelia arthritis inSwedish children: clinical manifestations in 10 children. Acta Paediatr1992;81:921e4.

[260] Renaud I, Cachin C, Gerster JC. Good outcomes of Lyme arthritis in 24 pa-tients in an endemic area of Switzerland. Jt Bone Spine 2004;71:39e43.

[261] Szer IS, Taylor E, Steere AC. The long-term course of Lyme arthritis in chil-dren. N Engl J Med 1991;325:159e63.

[262] Eichenfield AH, Goldsmith DP, Benach JL, Ross AH, Loeb FX, Doughty RA,et al. Childhood Lyme arthritis: experience in an endemic area. J Pediatr1986;109:753e8.

[263] Culp RW, Eichenfield AH, Davidson RS, Drummond DS, Christofersen MR,Goldsmith DP. Lyme arthritis in children. An orthopaedic perspective. J BoneJt Surg Am 1987;69:96e9.

[264] Milewski MD, Cruz Jr AI, Miller CP, Peterson AT, Smith BG. Lyme arthritis inchildren presenting with joint effusions. J Bone Jt Surg Am 2011;93:252e60.

[265] Bianchi G, Rovetta G, Monteforte P, Fumarola D, Trevisan G, Crovato F, et al.Articular involvement in European patients with Lyme disease. A report of32 Italian patients. Br J Rheumatol 1990;29:178e80.

[266] Johnston YE, Duray PH, Steere AC, Kashgarian M, Buza J, Malawista SE, et al.Lyme arthritis. Spirochetes found in synovial microangiopathic lesions. Am JPathol 1985;118:26e34.

[267] Steere AC, Duray PH, Butcher EC. Spirochetal antigens and lymphoid cellsurface markers in Lyme synovitis. Comparison with rheumatoid synoviumand tonsillar lymphoid tissue. Arthritis Rheum 1988;31:487e95.

[268] Jaulhac B, Chary-Valckenaere I, Sibilia J, Javier RM, Piemont Y, Kuntz JL, et al.Detection of Borrelia burgdorferi by DNA amplification in synovial tissuesamples from patients with Lyme arthritis. Arthritis Rheum 1996;39:736e45.

[269] Willis AA, Widmann RF, Flynn JM, Green DW, Onel KB. Lyme arthritis pre-senting as acute septic arthritis in children. J Pediatr Orthop 2003;23:114e8.

[270] Boissier MC, Semerano L, Challal S, Saidenberg-Kermanac'h N, Falgarone G.Rheumatoid arthritis: from autoimmunity to synovitis and joint destruction.J Autoimmun 2012;39:222e8.

[271] American Academy of Pediatrics. Lyme disease. Red book 2012. Report of theCommittee of Infectious Diseases. Elk Grove, IL: American Academy of Pe-diatrics; 2012. p. 474e9.

[272] British Infection Association. The epidemiology, prevention, investigationand treatment of Lyme borreliosis in United Kingdom patients: a positionstatement by the British Infection Association. J Infect 2011;62:329e38.

[273] Cameron D, Gaito A, Harris N, Bach G, Bellovin S, Bock K, et al. Evidence-based guidelines for the management of Lyme disease. Expert Rev Anti InfectTher 2004;2:S1e13.

[274] Wormser GP, Ramanathan R, Nowakowski J, McKenna D, Holmgren D,Visintainer P, et al. Duration of antibiotic therapy for early Lyme disease. Arandomized, double-blind, placebo-controlled trial. Ann Intern Med2003;138:697e704.

[275] Stupica D, Lusa L, Ruzic-Sabljic E, Cerar T, Strle F. Treatment of erythemamigrans with doxycycline for 10 days versus 15 days. Clin Infect Dis2012;55:343e50.

[276] Kowalski TJ, Tata S, Berth W, Mathiason MA, Agger WA. Antibiotic treatmentduration and long-term outcomes of patients with early Lyme disease from aLyme disease-hyperendemic area. Clin Infect Dis 2010;50:512e20.

[277] Oksi J, Nikoskelainen J, Hiekkanen H, Lauhio A, Peltomaa M, Pitkaranta A,et al. Duration of antibiotic treatment in disseminated Lyme borreliosis: adouble-blind, randomized, placebo-controlled, multicenter clinical study.Eur J Clin Microbiol Infect Dis 2007;26:571e81.

[278] Steere AC, Levin RE, Molloy PJ, Kalish RA, Abraham 3rd JH, Liu NY, et al.Treatment of Lyme arthritis. Arthritis Rheum 1994;37:878e88.

[279] Kalish RA, Leong JM, Steere AC. Association of treatment-resistant chronicLyme arthritis with HLA-DR4 and antibody reactivity to OspA and OspB ofBorrelia burgdorferi. Infect Immun 1993;61:2774e9.

[280] Reznick JW, Braunstein DB, Walsh RL, Smith CR, Wolfson PM, Gierke LW,et al. Lyme carditis. Electrophysiologic and histopathologic study. Am J Med1986;81:923e7.

[281] Luger SW, Paparone P, Wormser GP, Nadelman RB, Grunwaldt E, Gomez G,et al. Comparison of cefuroxime axetil and doxycycline in treatment of pa-tients with early Lyme disease associated with erythema migrans. Antimi-crobial Agents Chemother 1995;39:661e7.

[282] Massarotti EM, Luger SW, Rahn DW, Messner RP, Wong JB, Johnson RC, et al.Treatment of early Lyme disease. Am J Med 1992;92:396e403.

[283] Nadelman RB, Luger SW, Frank E, Wisniewski M, Collins JJ, Wormser GP.Comparison of cefuroxime axetil and doxycycline in the treatment of earlyLyme disease. Ann Intern Med 1992;117:273e80.

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3428

[284] Strle F, Ruzic E, Cimperman J. Erythema migrans: comparison of treatmentwith azithromycin, doxycycline and phenoxymethylpenicillin. J AntimicrobChemother 1992;30:543e50.

[285] Strle F, Preac-Mursic V, Cimperman J, Ruzic E, Maraspin V, Jereb M. Azi-thromycin versus doxycycline for treatment of erythema migrans: clinicaland microbiological findings. Infection 1993;21:83e8.

[286] Cerar D, Cerar T, Ruzic-Sabljic E, Wormser GP, Strle F. Subjective symptomsafter treatment of early Lyme disease. Am J Med 2010;123:79e86.

[287] Luft BJ, Dattwyler RJ, Johnson RC, Luger SW, Bosler EM, Rahn DW, et al.Azithromycin compared with amoxicillin in the treatment of erythema mi-grans. A double-blind, randomized, controlled trial. Ann Intern Med1996;124:785e91.

[288] Dattwyler RJ, Volkman DJ, Conaty SM, Platkin SP, Luft BJ. Amoxycillin plusprobenecid versus doxycycline for treatment of erythema migrans borre-liosis. Lancet 1990;336:1404e6.

[289] Dattwyler RJ, Luft BJ, Kunkel MJ, Finkel MF, Wormser GP, Rush TJ, et al.Ceftriaxone compared with doxycycline for the treatment of acute dissem-inated Lyme disease. N Engl J Med 1997;337:289e94.

[290] Barsic B, Maretic T, Majerus L, Strugar J. Comparison of azithromycin anddoxycycline in the treatment of erythema migrans. Infection 2000;28:153e6.

[291] Weber K, Wilske B, Preac-Mursic V, Thurmayr R. Azithromycin versuspenicillin V for the treatment of early Lyme borreliosis. Infection 1993;21:367e72.

[292] Eppes SC, Childs JA. Comparative study of cefuroxime axetil versus amoxi-cillin in children with early Lyme disease. Pediatrics 2002;109:1173e7.

[293] Kalish RA, Kaplan RF, Taylor E, Jones-Woodward L, Workman K, Steere AC.Evaluation of study patients with Lyme disease, 10-20-year follow-up.J Infect Dis 2001;183:453e60.

[294] Steere AC, Pachner AR, Malawista SE. Neurologic abnormalities of Lymedisease: successful treatment with high-dose intravenous penicillin. AnnIntern Med 1983;99:767e72.

[295] Kohlhepp W, Oschmann P, Mertens HG. Treatment of Lyme borreliosis.Randomized comparison of doxycycline and penicillin G. J Neurol 1989;236:464e9.

[296] Pfister HW, Preac-Mursic V, Wilske B, Einhaupl KM. Cefotaxime vs penicillinG for acute neurologic manifestations in Lyme borreliosis. A prospectiverandomized study. Arch Neurol 1989;46:1190e4.

[297] Pfister HW, Preac-Mursic V, Wilske B, Schielke E, Sorgel F, Einhaupl KM.Randomized comparison of ceftriaxone and cefotaxime in Lyme neuro-borreliosis. J Infect Dis 1991;163:311e8.

[298] Ljøstad U, Mygland A. Remaining complaints 1 year after treatment for acuteLyme neuroborreliosis; frequency, pattern and risk factors. Eur J Neurol2010;17:118e23.

[299] Karkkonen K, Stiernstedt SH, Karlsson M. Follow-up of patients treated withoral doxycycline for Lyme neuroborreliosis. Scand J Infect Dis 2001;33:259e62.

[300] Borg R, Dotevall L, Hagberg L, Maraspin V, Lotric-Furlan S, Cimperman J, et al.Intravenous ceftriaxone compared with oral doxycycline for the treatment ofLyme neuroborreliosis. Scand J Infect Dis 2005;37:449e54.

[301] Rohacova H, Hancil J, Hulinska D, Mailer H, Havlik J. Ceftriaxone in thetreatment of Lyme neuroborreliosis. Infection 1996;24:88e90.

[302] Eikeland R, Ljostad U, Mygland A, Herlofson K, Lohaugen GC. Europeanneuroborreliosis: neuropsychological findings 30 months post-treatment.Eur J Neurol 2012;19:480e7.

[303] Dotevall L, Hagberg L. Successful oral doxycycline treatment of Lymedisease-associated facial palsy and meningitis. Clin Infect Dis 1999;28:569e74.

[304] Eikeland R, Mygland A, Herlofson K, Ljostad U. Risk factors for a non-favorable outcome after treated European neuroborreliosis. Acta NeurolScand 2013;127:154e60.

[305] Eikeland R, Mygland A, Herlofson K, Ljostad U. European neuroborreliosis:quality of life 30 months after treatment. Acta Neurol Scand 2011;124:349e54.

[306] Benke T, Gasse T, Hittmair-Delazer M, Schmutzhard E. Lyme encephalopa-thy: long-term neuropsychological deficits years after acute neuro-borreliosis. Acta Neurol Scand 1995;91:353e7.

[307] Bremell D, Dotevall L. Oral doxycycline for Lyme neuroborreliosis withsymptoms of encephalitis, myelitis, vasculitis or intracranial hypertension.Eur J Neurol 2014;21:1162e7 [accessed 29.06.14].

[308] Peltomaa M, Saxen H, Sepp€al€a I, Viljanen M, Pyykk€o I. Paediatric facial pa-ralysis caused by Lyme borreliosis: a prospective and retrospective analysis.Scand J Infect Dis 1998;30:269e75.

[309] Peltomaa M, Pyykk€o I, Sepp€al€a I, Viljanen M. Lyme borreliosis and facialparalysis e a prospective analysis of risk factors and outcome. Am J Oto-laryngol 2002;23:125e32.

[310] Skogman BH, Croner S, Odkvist L. Acute facial palsy in children e a 2-yearfollow-up study with focus on Lyme neuroborreliosis. Int J Pediatr Oto-rhinolaryngol 2003;67:597e602.

[311] Bagger-Sj€ob€ack D, Remahl S, Ericsson M. Long-term outcome of facial palsyin neuroborreliosis. Otol Neurotol 2005;26:790e5.

[312] Jenke AC, Stoek LM, Zilbauer M, Wirth S, Borusiak P. Facial palsy: etiology,outcome and management in children. Eur J Paediatr Neurol 2011;15:209e13.

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

[313] Skogman BH, Glimaker K, Nordwall M, Vrethem M, Odkvist L, Forsberg P.Long-term clinical outcome after Lyme neuroborreliosis in childhood. Pedi-atrics 2012;130:262e9.

[314] Thorstrand C, Belfrage E, Bennet R, Malmborg P, Eriksson M. Successfultreatment of neuroborreliosis with ten day regimens. Pediatr Infect Dis J2002;21:1142e5.

[315] Steere AC, Green J, Schoen RT, Taylor E, Hutchinson GJ, Rahn DW, et al.Successful parenteral penicillin therapy of established Lyme arthritis. N EnglJ Med 1985;312:869e74.

[316] Brescia AC, Rose CD, Fawcett PT. Prolonged synovitis in pediatric Lymearthritis cannot be predicted by clinical or laboratory parameters. ClinRheumatol 2009;28:591e3.

[317] Valesova H, Mailer J, Havlik J, Hulinska D, Hercogova J. Long-term results inpatients with Lyme arthritis following treatment with ceftriaxone. Infection1996;24:98e102.

[318] Haugeberg G, Hansen IJ, Skarpaas T, Noraas S, Kjelland V. Lyme arthritis inSouthern Norway e an endemic area for Lyme borreliosis. BMC Infect Dis2014;14:185.

[319] Steere AC, Klitz W, Drouin EE, Falk BA, Kwok WW, Nepom GT, et al. Anti-biotic-refractory Lyme arthritis is associated with HLA-DR molecules thatbind a Borrelia burgdorferi peptide. J Exp Med 2006;203:961e71.

[320] Bentas W, Karch H, Huppertz HI. Lyme arthritis in children and adolescents:outcome 12 months after initiation of antibiotic therapy. J Rheumatol2000;27:2025e30.

[321] Dattwyler RJ, Halperin JJ, Volkman DJ, Luft BJ. Treatment of late Lyme bor-reliosis e randomised comparison of ceftriaxone and penicillin. Lancet1988;1:1191e4.

[322] De Santis M, Selmi C. The therapeutic potential of epigenetics in autoim-mune diseases. Clin Rev Allergy Immunol 2012;42:92e101.

[323] Vrethem M, Hellblom L, Widlund M, Ahl M, Danielsson O, Ernerudh J, et al.Chronic symptoms are common in patients with neuroborreliosis e aquestionnaire follow-up study. Acta Neurol Scand 2002;106:205e8.

[324] Shadick NA, Phillips CB, Logigian EL, Steere AC, Kaplan RF, Berardi VP, et al.The long-term clinical outcomes of Lyme disease. A population-basedretrospective cohort study. Ann Intern Med 1994;121:560e7.

[325] Ravdin LD, Hilton E, Primeau M, Clements C, Barr WB. Memory functioningin Lyme borreliosis. J Clin Psychiatry 1996;57:282e6.

[326] Klempner MS, Hu LT, Evans J, Schmid CH, Johnson GM, Trevino RP, et al. Twocontrolled trials of antibiotic treatment in patients with persistent symp-toms and a history of Lyme disease. N Engl J Med 2001;345:85e92.

[327] Treib J, Fernandez A, Haass A, Grauer MT, Holzer G, Woessner R. Clinical andserologic follow-up in patients with neuroborreliosis. Neurology 1998;51:1489e91.

[328] Seltzer EG, Gerber MA, Cartter ML, Freudigman K, Shapiro ED. Long-termoutcomes of persons with Lyme disease. JAMA 2000;283:609e16.

[329] V�azquez M, Sparrow SS, Shapiro ED. Long-term neuropsychologic and healthoutcomes of children with facial nerve palsy attributable to Lyme disease.Pediatrics 2003;112:e93e7.

[330] Shadick NA, Phillips CB, Sangha O, Logigian EL, Kaplan RF, Wright EA, et al.Musculoskeletal and neurologic outcomes in patients with previouslytreated Lyme disease. Ann Intern Med 1999;131:919e26.

[331] Krupp LB, Masur D, Schwartz J, Coyle PK, Langenbach LJ, Fernquist SK, et al.Cognitive functioning in late Lyme borreliosis. Arch Neurol 1991;48:1125e9.

[332] Bujak DI, Weinstein A, Dornbush RL. Clinical and neurocognitive features ofthe post Lyme syndrome. J Rheumatol 1996;23:1392e7.

[333] Kaplan RF, Meadows ME, Vincent LC, Logigian EL, Steere AC. Memoryimpairment and depression in patients with Lyme encephalopathy: com-parison with fibromyalgia and nonpsychotically depressed patients.Neurology 1992;42:1263e7.

[334] Gaudino EA, Coyle PK, Krupp LB. Post-Lyme syndrome and chronic fatiguesyndrome. Neuropsychiatric similarities and differences. Arch Neurol1997;54:1372e6.

[335] Keilp JG, Corbera K, Slavov I, Taylor MJ, Sackeim HA, Fallon BA. WAIS-III andWMS-III performance in chronic Lyme disease. J Int Neuropsychol Soc2006;12:119e29.

[336] Fallon BA, Keilp JG, Corbera KM, Petkova E, Britton CB, Dwyer E, et al.A randomized, placebo-controlled trial of repeated IV antibiotic therapy forLyme encephalopathy. Neurology 2008;70:992e1003.

[337] Elkins LE, Pollina DA, Scheffer SR, Krupp LB. Psychological states and neu-ropsychological performances in chronic Lyme disease. Appl Neuropsychol1999;6:19e26.

[338] Kaplan RF, Trevino RP, Johnson GM, Levy L, Dornbush R, Hu LT, et al.Cognitive function in post-treatment Lyme disease: do additional antibioticshelp? Neurology 2003;60:1916e22.

[339] McAuliffe P, Brassard MR, Fallon B. Memory and executive functions in ad-olescents with posttreatment Lyme disease. Appl Neuropsychol 2008;15:208e19.

[340] Halperin JJ, Pass HL, Anand AK, Luft BJ, Volkman DJ, Dattwyler RJ. Nervoussystem abnormalities in Lyme disease. Ann N Y Acad Sci 1988;539:24e34.

[341] Bloom BJ, Wyckoff PM, Meissner HC, Steere AC. Neurocognitive abnormal-ities in children after classic manifestations of Lyme disease. Pediatr InfectDis J 1998;17:189e96.

[342] Halperin JJ. Abnormalities of the nervous system in Lyme disease: responseto antimicrobial therapy. Rev Infect Dis 1989;11(Suppl. 6):S1499e504.

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 29

[343] Kaplan RF, Jones-Woodward L, Workman K, Steere AC, Logigian EL,Meadows ME. Neuropsychological deficits in Lyme disease patients with andwithout other evidence of central nervous system pathology. Appl Neuro-psychol 1999;6:3e11.

[344] Aucott JN, Crowder LA, Kortte KB. Development of a foundation for a casedefinition of post-treatment Lyme disease syndrome. Int J Infect Dis2013;17:e443e9.

[345] Krupp LB, Hyman LG, Grimson R, Coyle PK, Melville P, Ahnn S, et al. Studyand treatment of post Lyme disease (STOP-LD): a randomized doublemasked clinical trial. Neurology 2003;60:1923e30.

[346] Cameron D. Severity of Lyme disease with persistent symptoms. Insightsfrom a double-blind placebo-controlled clinical trial. Minerva Med 2008;99:489e96.

[347] Fallon BA, Petkova E, Keilp JG, Britton CB. A reappraisal of the U.S. clinical trialsof post-treatment Lyme disease syndrome. Open Neurol J 2012;6:79e87.

[348] Delong AK, Blossom B, Maloney EL, Phillips SE. Antibiotic retreatment ofLyme disease in patients with persistent symptoms: a biostatistical review ofrandomized, placebo-controlled, clinical trials. Contemp Clin Trials 2012;33:1132e42.

[349] Klempner MS, Baker PJ, Shapiro ED, Marques A, Dattwyler RJ, Halperin JJ,et al. Treatment trials for post-Lyme disease symptoms revisited. Am J Med2013;126:665e9.

[350] Feder Jr HM, Johnson BJ, O'Connell S, Shapiro ED, Steere AC, Wormser GP,et al. A critical appraisal of “chronic Lyme disease”. N Engl J Med 2007;357:1422e30.

[351] Fasano A. Leaky gut and autoimmune diseases. Clin Rev Allergy Immunol2012;42:71e8.

[352] Henao-Mejia J, Elinav E, Thaiss CA, Licona-Limon P, Flavell RA. Role of theintestinal microbiome in liver disease. J Autoimmun 2013;46:66e73.

[353] Matricardi PM, Bonini S. High microbial turnover rate preventing atopy: asolution to inconsistencies impinging on the hygiene hypothesis? Clin ExpAllergy 2000;30:1506e10.

[354] Bogdanos DP, Smyk DS, Rigopoulou EI, Mytilinaiou MG, Heneghan MA,Selmi C, et al. Twin studies in autoimmune disease: genetics, gender andenvironment. J Autoimmun 2012;38:J156e69.

[355] Philipp MT, Aydintug MK, Bohm Jr RP, Cogswell FB, Dennis VA, Lanners HN,et al. Early and early disseminated phases of Lyme disease in the rhesusmonkey: a model for infection in humans. Infect Immun 1993;61:3047e59.

[356] Straubinger RK, Straubinger AF, Summers BA, Jacobson RH, Erb HN. Clinicalmanifestations, pathogenesis, and effect of antibiotic treatment on Lymeborreliosis in dogs. Wien Klin Wochenschr 1998;110:874e81.

[357] Straubinger RK, Straubinger AF, Harter L, Jacobson RH, Chang YF,Summers BA, et al. Borrelia burgdorferi migrates into joint capsules andcauses an up-regulation of interleukin-8 in synovial membranes of dogsexperimentally infected with ticks. Infect Immun 1997;65:1273e85.

[358] Chang YF, Novosel V, Chang CF, Summers BA, Ma DP, Chiang YW, et al.Experimental induction of chronic borreliosis in adult dogs exposed toBorrelia burgdorferi-infected ticks and treated with dexamethasone. Am J VetRes 2001;62:1104e12.

[359] Asbrink E, Hovmark A. Successful cultivation of spirochetes from skin lesionsof patients with erythema chronicum migrans Afzelius and acrodermatitischronica atrophicans. Acta Pathol Microbiol Immunol Scand B 1985;93:161e3.

[360] Craft JE, Fischer DK, Shimamoto GT, Steere AC. Antigens of Borrelia burg-dorferi recognized during Lyme disease. Appearance of a new immuno-globulin M response and expansion of the immunoglobulin G response latein the illness. J Clin Invest 1986;78:934e9.

[361] Iliopoulou BP, Alroy J, Huber BT. CD28 deficiency exacerbates joint inflam-mation upon Borrelia burgdorferi infection, resulting in the development ofchronic Lyme arthritis. J Immunol 2007;179:8076e82.

[362] Ruderman EM, Kerr JS, Telford 3rd SR, Spielman A, Glimcher LH,Gravallese EM. Early murine Lyme carditis has a macrophage predominanceand is independent of major histocompatibility complex class II-CD4þ T cellinteractions. J Infect Dis 1995;171:362e70.

[363] Tupin E, Benhnia MR, Kinjo Y, Patsey R, Lena CJ, Haller MC, et al. NKT cellsprevent chronic joint inflammation after infection with Borrelia burgdorferi.Proc Natl Acad Sci U S A 2008;105:19863e8.

[364] Olson Jr CM, Bates TC, Izadi H, Radolf JD, Huber SA, Boyson JE, et al. Localproduction of IFN-g by invariant NKT cells modulates acute Lyme carditis.J Immunol 2009;182:3728e34.

[365] Duray PH, Steere AC. Clinical pathologic correlations of Lyme disease bystage. Ann N Y Acad Sci 1988;539:65e79.

[366] Hidri N, Barraud O, de Martino S, Garnier F, Paraf F, Martin C, et al. Lymeendocarditis. Clin Microbiol Infect 2012;18:E531e2.

[367] Wang G, Ojaimi C, Wu H, Saksenberg V, Iyer R, Liveris D, et al. Diseaseseverity in a murine model of Lyme borreliosis is associated with the ge-notype of the infecting Borrelia burgdorferi sensu stricto strain. J Infect Dis2002;186:782e91.

[368] Wang G, Ojaimi C, Iyer R, Saksenberg V, McClain SA, Wormser GP, et al.Impact of genotypic variation of Borrelia burgdorferi sensu stricto on kineticsof dissemination and severity of disease in C3H/HeJ mice. Infect Immun2001;69:4303e12.

[369] Craig-Mylius KA, Lee M, Jones KL, Glickstein LJ. Arthritogenicity of Borreliaburgdorferi and Borrelia garinii: comparison of infection in mice. Am J TropMed Hyg 2009;80:252e8.

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

[370] Zeidner N, Dreitz M, Belasco D, Fish D. Suppression of acute Ixodes scapularis-induced Borrelia burgdorferi infection using tumor necrosis factor-alpha,interleukin-2, and interferon-gamma. J Infect Dis 1996;173:187e95.

[371] Machackova M, Obornik M, Kopecky J. Effect of salivary gland extract fromIxodes ricinus ticks on the proliferation of Borrelia burgdorferi sensu strictoin vivo. Folia Parasitol (Praha) 2006;53:153e8.

[372] Pechova J, Stepanova G, Kovar L, Kopecky J. Tick salivary gland extract-activated transmission of Borrelia afzelii spirochaetes. Folia Parasitol(Praha) 2002;49:153e9.

[373] Severinova J, Salat J, Krocova Z, Reznickova J, Demova H, Horka H, et al. Co-inoculation of Borrelia afzelii with tick salivary gland extract influences dis-tribution of immunocompetent cells in the skin and lymph nodes of mice.Folia Microbiol (Praha) 2005;50:457e63.

[374] Christe M, Rutti B, Brossard M. Cytokines (IL-4 and IFN-gamma) and anti-bodies (IgE and IgG2a) produced in mice infected with Borrelia burgdorferisensu stricto via nymphs of Ixodes ricinus ticks or syringe inoculations.Parasitol Res 2000;86:491e6.

[375] de Silva AM, Fikrig E, Hodzic E, Kantor FS, Telford 3rd SR, Barthold SW.Immune evasion by tickborne and host-adapted Borrelia burgdorferi. J InfectDis 1998;177:395e400.

[376] Hagman KE, Yang X, Wikel SK, Schoeler GB, Caimano MJ, Radolf JD, et al.Decorin-binding protein A (DbpA) of Borrelia burgdorferi is not protectivewhen immunized mice are challenged via tick infestation and correlateswith the lack of DbpA expression by B. burgdorferi in ticks. Infect Immun2000;68:4759e64.

[377] Berndtson K. Review of evidence for immune evasion and persistent infec-tion in Lyme disease. Int J Gen Med 2013;6:291e306.

[378] Schaible UE, Gern L, Wallich R, Kramer MD, Prester M, Simon MM. Distinctpatterns of protective antibodies are generated against Borrelia burgdorferi inmice experimentally inoculated with high and low doses of antigen.Immunol Lett 1993;36:219e26.

[379] Zeidner NS, Schneider BS, Dolan MC, Piesman J. An analysis of spirocheteload, strain, and pathology in a model of tick-transmitted Lyme borreliosis.Vector Borne Zoonotic Dis 2001;1:35e44.

[380] Dolan MC, Piesman J, Schneider BS, Schriefer M, Brandt K, Zeidner NS.Comparison of disseminated and nondisseminated strains of Borrelia burg-dorferi sensu stricto in mice naturally infected by tick bite. Infect Immun2004;72:5262e6.

[381] Roberts ED, Bohm Jr RP, Lowrie Jr RC, Habicht G, Katona L, Piesman J, et al.Pathogenesis of Lyme neuroborreliosis in the rhesus monkey: the earlydisseminated and chronic phases of disease in the peripheral nervous sys-tem. J Infect Dis 1998;178:722e32.

[382] Wang G, van Dam AP, Dankert J. Phenotypic and genetic characterization of anovel Borrelia burgdorferi sensu lato isolate from a patient with Lyme bor-reliosis. J Clin Microbiol 1999;37:3025e8.

[383] Jaulhac B, Heller R, Limbach FX, Hansmann Y, Lipsker D, Monteil H, et al.Direct molecular typing of Borrelia burgdorferi sensu lato species in synovialsamples from patients with Lyme arthritis. J Clin Microbiol 2000;38:1895e900.

[384] van der Heijden IM, Wilbrink B, Rijpkema SG, Schouls LM, Heymans PH, vanEmbden JD, et al. Detection of Borrelia burgdorferi sensu stricto by reverseline blot in the joints of Dutch patients with Lyme arthritis. Arthritis Rheum1999;42:1473e80.

[385] Busch U, Hizo-Teufel C, Boehmer R, Fingerle V, Nitschko H, Wilske B, et al.Three species of Borrelia burgdorferi sensu lato (B. burgdorferi sensu stricto,B. afzelii, and B. garinii) identified from cerebrospinal fluid isolates by pulsed-field gel electrophoresis and PCR. J Clin Microbiol 1996;34:1072e8.

[386] Eiffert H, Ohlenbusch A, Christen HJ, Thomssen R, Spielman A, Matuschka FR.Nondifferentiation between Lyme disease spirochetes from vector ticks andhuman cerebrospinal fluid. J Infect Dis 1995;171:476e9.

[387] Ornstein K, Berglund J, Nilsson I, Norrby R, Bergstrom S. Characterization ofLyme borreliosis isolates from patients with erythema migrans and neuro-borreliosis in southern Sweden. J Clin Microbiol 2001;39:1294e8.

[388] Ornstein K, Berglund J, Bergstrom S, Norrby R, Barbour AG. Three major LymeBorrelia genospecies (Borrelia burgdorferi sensu stricto, B. afzelii andB. garinii) identified by PCR in cerebrospinal fluid from patients with neu-roborreliosis in Sweden. Scand J Infect Dis 2002;34:341e6.

[389] Ruzic-Sabljic E, Lotric-Furlan S, Maraspin V, Cimperman J, Pleterski-Rigler D,Strle F. Analysis of Borrelia burgdorferi sensu lato isolated from cerebrospinalfluid. APMIS 2001;109:707e13.

[390] Wilske B, Preac-Mursic V, Gobel UB, Graf B, Jauris S, Soutschek E, et al. AnOspA serotyping system for Borrelia burgdorferi based on reactivity withmonoclonal antibodies and OspA sequence analysis. J Clin Microbiol1993;31:340e50.

[391] Tijsse-Klasen E, Pandak N, Hengeveld P, Takumi K, Koopmans MP, Sprong H.Ability to cause erythema migrans differs between Borrelia burgdorferi sensulato isolates. Parasit Vectors 2013;6:23.

[392] Kuiper H, Cairo I, Van Dam A, De Jongh B, Ramselaar T, Spanjaard L, et al.Solitary erythema migrans: a clinical, laboratory and epidemiological studyof 77 Dutch patients. Br J Dermatol 1994;130:466e72.

[393] Wienecke R, Zochling N, Neubert U, Schlupen EM, Meurer M, Volkenandt M.Molecular subtyping of Borrelia burgdorferi in erythema migrans and acro-dermatitis chronica atrophicans. J Invest Dermatol 1994;103:19e22.

[394] Busch U, Hizo-Teufel C, Bohmer R, Fingerle V, Rossler D, Wilske B, et al.Borrelia burgdorferi sensu lato strains isolated from cutaneous Lyme

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3430

borreliosis biopsies differentiated by pulsed-field gel electrophoresis. Scand JInfect Dis 1996;28:583e9.

[395] Picken RN, Strle F, Picken MM, Ruzic-Sabljic E, Maraspin V, Lotric-Furlan S,et al. Identification of three species of Borrelia burgdorferi sensu lato(B. burgdorferi sensu stricto, B. garinii, and B. afzelii) among isolates fromacrodermatitis chronica atrophicans lesions. J Invest Dermatol 1998;110:211e4.

[396] Ruzic-Sabljic E, Maraspin V, Lotric-Furlan S, Jurca T, Logar M, Pikelj-Pecnik A,et al. Characterization of Borrelia burgdorferi sensu lato strains isolated fromhuman material in Slovenia. Wien Klin Wochenschr 2002;114:544e50.

[397] Roux F, Boyer E, Jaulhac B, Dernis E, Closs-Prophette F, Puechal X. Lymemeningoradiculitis: prospective evaluation of biological diagnosis methods.Eur J Clin Microbiol Infect Dis 2007;26:685e93.

[398] Picken RN, Cheng Y, Strle F, Cimperman J, Maraspin V, Lotric-Furlan S, et al.Molecular characterization of Borrelia burgdorferi sensu lato from Sloveniarevealing significant differences between tick and human isolates. Eur J ClinMicrobiol Infect Dis 1996;15:313e23.

[399] Bormane A, Lucenko I, Duks A, Mavtchoutko V, Ranka R, Salmina K, et al.Vectors of tick-borne diseases and epidemiological situation in Latvia in1993-2002. Int J Med Microbiol 2004;293(Suppl. 37):36e47.

[400] Jouda F, Perret JL, Gern L. Density of questing Ixodes ricinus nymphs andadults infected by Borrelia burgdorferi sensu lato in Switzerland: spatio-temporal pattern at a regional scale. Vector Borne Zoonotic Dis 2004;4:23e32.

[401] Brisson D, Baxamusa N, Schwartz I, Wormser GP. Biodiversity of Borreliaburgdorferi strains in tissues of Lyme disease patients. PLoS One 2011;6:e22926.

[402] Seinost G, Dykhuizen DE, Dattwyler RJ, Golde WT, Dunn JJ, Wang IN, et al.Four clones of Borrelia burgdorferi sensu stricto cause invasive infection inhumans. Infect Immun 1999;67:3518e24.

[403] Wormser GP, Liveris D, Nowakowski J, Nadelman RB, Cavaliere LF,McKenna D, et al. Association of specific subtypes of Borrelia burgdorferi withhematogenous dissemination in early Lyme disease. J Infect Dis 1999;180:720e5.

[404] Wormser GP, Brisson D, Liveris D, Hanincova K, Sandigursky S,Nowakowski J, et al. Borrelia burgdorferi genotype predicts the capacity forhematogenous dissemination during early Lyme disease. J Infect Dis2008;198:1358e64.

[405] Dykhuizen DE, Brisson D, Sandigursky S, Wormser GP, Nowakowski J,Nadelman RB, et al. The propensity of different Borrelia burgdorferi sensustricto genotypes to cause disseminated infections in humans. Am J TropMed Hyg 2008;78:806e10.

[406] Earnhart CG, Buckles EL, Dumler JS, Marconi RT. Demonstration of OspC typediversity in invasive human Lyme disease isolates and identification ofpreviously uncharacterized epitopes that define the specificity of the OspCmurine antibody response. Infect Immun 2005;73:7869e77.

[407] Baranton G, Seinost G, Theodore G, Postic D, Dykhuizen D. Distinct levels ofgenetic diversity of Borrelia burgdorferi are associated with different aspectsof pathogenicity. Res Microbiol 2001;152:149e56.

[408] Lagal V, Postic D, Ruzic-Sabljic E, Baranton G. Genetic diversity among Bor-relia strains determined by single-strand conformation polymorphismanalysis of the ospC gene and its association with invasiveness. J ClinMicrobiol 2003;41:5059e65.

[409] Strle K, Jones KL, Drouin EE, Li X, Steere AC. Borrelia burgdorferi RST1 (OspCtype A) genotype is associated with greater inflammation and more severeLyme disease. Am J Pathol 2011;178:2726e39.

[410] Alghaferi MY, Anderson JM, Park J, Auwaerter PG, Aucott JN, Norris DE, et al.Borrelia burgdorferi ospC heterogeneity among human and murine isolatesfrom a defined region of northern Maryland and southern Pennsylvania: lackof correlation with invasive and noninvasive genotypes. J Clin Microbiol2005;43:1879e84.

[411] Jones KL, Glickstein LJ, Damle N, Sikand VK, McHugh G, Steere AC. Borreliaburgdorferi genetic markers and disseminated disease in patients with earlyLyme disease. J Clin Microbiol 2006;44:4407e13.

[412] Brandt FC, Ertas B, Falk TM, Metze D, Boer-Auer A. Genotyping of borreliafrom formalin-fixed paraffin-embedded skin biopsies of cutaneous borre-liosis and tick bite reactions by assays targeting the IGS-region, ospA, andOspC genes. Br J Dermatol 2014;171:528e43.

[413] Lagal V, Portnoi D, Faure G, Postic D, Baranton G. Borrelia burgdorferi sensustricto invasiveness is correlated with OspC-plasminogen affinity. MicrobesInfect 2006;8:645e52.

[414] Hanincova K, Mukherjee P, Ogden NH, Margos G, Wormser GP, Reed KD,et al. Multilocus sequence typing of Borrelia burgdorferi suggests existence oflineages with differential pathogenic properties in humans. PLoS One2013;8:e73066.

[415] Liveris D, Wormser GP, Nowakowski J, Nadelman R, Bittker S, Cooper D, et al.Molecular typing of Borrelia burgdorferi from Lyme disease patients by PCR-restriction fragment length polymorphism analysis. J Clin Microbiol1996;34:1306e9.

[416] Barthold SW, Beck DS, Hansen GM, Terwilliger GA, Moody KD. Lyme bor-reliosis in selected strains and ages of laboratory mice. J Infect Dis 1990;162:133e8.

[417] Schaible UE, Kramer MD, Wallich R, Tran T, Simon MM. Experimental Bor-relia burgdorferi infection in inbred mouse strains: antibody response and

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

association of H-2 genes with resistance and susceptibility to developmentof arthritis. Eur J Immunol 1991;21:2397e405.

[418] Schaible UE, Gay S, Museteanu C, Kramer MD, Zimmer G, Eichmann K, et al.Lyme borreliosis in the severe combined immunodeficiency (SCID) mousemanifests predominantly in the joints, heart, and liver. Am J Pathol1990;137:811e20.

[419] Schaible UE, Kramer MD, Justus CW, Museteanu C, Simon MM. Demonstra-tion of antigen-specific T cells and histopathological alterations in miceexperimentally inoculated with Borrelia burgdorferi. Infect Immun 1989;57:41e7.

[420] Yang L, Weis JH, Eichwald E, Kolbert CP, Persing DH, Weis JJ. Heritablesusceptibility to severe Borrelia burgdorferi-induced arthritis is dominant andis associated with persistence of large numbers of spirochetes in tissues.Infect Immun 1994;62:492e500.

[421] Ma Y, Seiler KP, Eichwald EJ, Weis JH, Teuscher C, Weis JJ. Distinct charac-teristics of resistance to Borrelia burgdorferi-induced arthritis in C57BL/6Nmice. Infect Immun 1998;66:161e8.

[422] Roper RJ, Weis JJ, McCracken BA, Green CB, Ma Y, Weber KS, et al. Geneticcontrol of susceptibility to experimental Lyme arthritis is polygenic andexhibits consistent linkage to multiple loci on chromosome 5 in four inde-pendent mouse crosses. Genes Immun 2001;2:388e97.

[423] Ma Y, Miller JC, Crandall H, Larsen ET, Dunn DM, Weiss RB, et al. Interval-specific congenic lines reveal quantitative trait loci with penetrant lymearthritis phenotypes on chromosomes 5, 11, and 12. Infect Immun 2009;77:3302e11.

[424] Brown CR, Reiner SL. Genes outside the major histocompatibility complexcontrol resistance and susceptibility to experimental Lyme arthritis. MedMicrobiol Immunol 2000;189:85e90.

[425] Yang L, Ma Y, Schoenfeld R, Griffiths M, Eichwald E, Araneo B, et al. Evidencefor B-lymphocyte mitogen activity in Borrelia burgdorferi-infected mice.Infect Immun 1992;60:3033e41.

[426] Pachner AR, Gelderblom H, Cadavid D. The rhesus model of Lyme neuro-borreliosis. Immunol Rev 2001;183:186e204.

[427] Pahl A, Kuhlbrandt U, Brune K, Rollinghoff M, Gessner A. Quantitativedetection of Borrelia burgdorferi by real-time PCR. J Clin Microbiol 1999;37:1958e63.

[428] Lederer S, Brenner C, Stehle T, Gern L, Wallich R, Simon MM. Quantitativeanalysis of Borrelia burgdorferi gene expression in naturally (tick) infectedmouse strains. Med Microbiol Immunol 2005;194:81e90.

[429] Brown JP, Zachary JF, Teuscher C, Weis JJ, Wooten RM. Dual role ofinterleukin-10 in murine Lyme disease: regulation of arthritis severity andhost defense. Infect Immun 1999;67:5142e50.

[430] Nadelman RB, Horowitz HW, Hsieh TC, Wu JM, Aguero-Rosenfeld ME,Schwartz I, et al. Simultaneous human granulocytic ehrlichiosis and Lymeborreliosis. N Engl J Med 1997;337:27e30.

[431] Horowitz HW, Aguero-Rosenfeld ME, Holmgren D, McKenna D, Schwartz I,Cox ME, et al. Lyme disease and human granulocytic anaplasmosis coinfec-tion: impact of case definition on coinfection rates and illness severity. ClinInfect Dis 2013;56:93e9.

[432] Belongia EA, Reed KD, Mitchell PD, Chyou PH, Mueller-Rizner N, Finkel MF,et al. Clinical and epidemiological features of early Lyme disease and humangranulocytic ehrlichiosis in Wisconsin. Clin Infect Dis 1999;29:1472e7.

[433] Krause PJ, McKay K, Thompson CA, Sikand VK, Lentz R, Lepore T, et al. Dis-ease-specific diagnosis of coinfecting tickborne zoonoses: babesiosis, humangranulocytic ehrlichiosis, and Lyme disease. Clin Infect Dis 2002;34:1184e91.

[434] Steere AC, McHugh G, Suarez C, Hoitt J, Damle N, Sikand VK. Prospectivestudy of coinfection in patients with erythema migrans. Clin Infect Dis2003;36:1078e81.

[435] Thomas V, Anguita J, Barthold SW, Fikrig E. Coinfection with Borrelia burg-dorferi and the agent of human granulocytic ehrlichiosis alters murine im-mune responses, pathogen burden, and severity of Lyme arthritis. InfectImmun 2001;69:3359e71.

[436] Zeidner NS, Dolan MC, Massung R, Piesman J, Fish D. Coinfection with Bor-relia burgdorferi and the agent of human granulocytic ehrlichiosis suppressesIL-2 and IFNg production and promotes an IL-4 response in C3H/HeJ mice.Parasite Immunol 2000;22:581e8.

[437] Holden K, Hodzic E, Feng S, Freet KJ, Lefebvre RB, Barthold SW. Coinfectionwith Anaplasma phagocytophilum alters Borrelia burgdorferi population dis-tribution in C3H/HeN mice. Infect Immun 2005;73:3440e4.

[438] Jarefors S, Karlsson M, Forsberg P, Eliasson I, Ernerudh J, Ekerfelt C. Reducednumber of interleukin-12 secreting cells in patients with Lyme borreliosispreviously exposed to Anaplasma phagocytophilum. Clin Exp Immunol2006;143:322e8.

[439] Krause PJ, Narasimhan S, Wormser GP, Barbour AG, Platonov AE, Brancato J,et al. Borrelia miyamotoi sensu lato seroreactivity and seroprevalence in thenortheastern United States. Emerg Infect Dis 2014;20:1183e90.

[440] Tilly K, Rosa PA, Stewart PE. Biology of infection with Borrelia burgdorferi.Infect Dis Clin North Am 2008;22:217e34 [v].

[441] Ekdahl KN, Henningsson AJ, Sandholm K, Forsberg P, Ernerudh J, Ekerfelt C.Immunity in borreliosis with special emphasis on the role of complement.Adv Exp Med Biol 2007;598:198e213.

[442] Kung F, Anguita J, Pal U. Borrelia burgdorferi and tick proteins supportingpathogen persistence in the vector. Future Microbiol 2013;8:41e56.

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 31

[443] Liang FT, Yan J, Mbow ML, Sviat SL, Gilmore RD, Mamula M, et al. Borreliaburgdorferi changes its surface antigenic expression in response to hostimmune responses. Infect Immun 2004;72:5759e67.

[444] Crother TR, Champion CI, Whitelegge JP, Aguilera R, Wu XY, Blanco DR, et al.Temporal analysis of the antigenic composition of Borrelia burgdorferi duringinfection in rabbit skin. Infect Immun 2004;72:5063e72.

[445] Crother TR, Champion CI, Wu XY, Blanco DR, Miller JN, Lovett MA. Antigeniccomposition of Borrelia burgdorferi during infection of SCID mice. InfectImmun 2003;71:3419e28.

[446] Radolf JD, Caimano MJ, Stevenson B, Hu LT. Of ticks, mice and men: under-standing the dual-host lifestyle of Lyme disease spirochaetes. Nat RevMicrobiol 2012;10:87e99.

[447] Menten-Dedoyart C, Faccinetto C, Golovchenko M, Dupiereux I, VanLerberghe PB, Dubois S, et al. Neutrophil extracellular traps entrap and killBorrelia burgdorferi sensu stricto spirochetes and are not affected by Ixodesricinus tick saliva. J Immunol 2012;189:5393e401.

[448] Xu Q, Seemanapalli SV, Reif KE, Brown CR, Liang FT. Increasing the recruit-ment of neutrophils to the site of infection dramatically attenuates Borreliaburgdorferi infectivity. J Immunol 2007;178:5109e15.

[449] Chong-Cerrillo C, Shang ES, Blanco DR, Lovett MA, Miller JN. Immunohisto-chemical analysis of Lyme disease in the skin of naive and infection-immunerabbits following challenge. Infect Immun 2001;69:4094e102.

[450] Berger BW, Clemmensen OJ, Ackerman AB. Lyme disease is a spirochetosis: areview of the disease and evidence for its cause. Am J Dermatopathol1983;5:111e24.

[451] Büchner SA, Rufli T. Erythema chronicum migrans: evidence for cellularimmune reaction in the skin lesion. Dermatologica 1987;174:144e9.

[452] Montgomery RR, Lusitani D, de Boisfleury Chevance A, Malawista SE. Humanphagocytic cells in the early innate immune response to Borrelia burgdorferi.J Infect Dis 2002;185:1773e9.

[453] Benach JL, Fleit HB, Habicht GS, Coleman JL, Bosler EM, Lane BP. Interactionsof phagocytes with the Lyme disease spirochete: role of the Fc receptor.J Infect Dis 1984;150:497e507.

[454] Lusitani D, Malawista SE, Montgomery RR. Borrelia burgdorferi are suscep-tible to killing by a variety of human polymorphonuclear leukocyte com-ponents. J Infect Dis 2002;185:797e804.

[455] Cruz AR, Moore MW, La Vake CJ, Eggers CH, Salazar JC, Radolf JD. Phagocy-tosis of Borrelia burgdorferi, the Lyme disease spirochete, potentiates innateimmune activation and induces apoptosis in human monocytes. InfectImmun 2008;76:56e70.

[456] Mason LM, Veerman CC, Geijtenbeek TB, Hovius JW. M�enage �a trois: Borrelia,dendritic cells, and tick saliva interactions. Trends Parasitol 2014;30:95e103.

[457] Berende A, Oosting M, Kullberg BJ, Netea MG, Joosten LA. Activation of innatehost defense mechanisms by Borrelia. Eur Cytokine Netw 2010;21:7e18.

[458] Petzke MM, Brooks A, Krupna MA, Mordue D, Schwartz I. Recognition ofBorrelia burgdorferi, the Lyme disease spirochete, by TLR7 and TLR9 induces atype I IFN response by human immune cells. J Immunol 2009;183:5279e92.

[459] Cervantes JL, Dunham-Ems SM, La Vake CJ, Petzke MM, Sahay B, Sellati TJ,et al. Phagosomal signaling by Borrelia burgdorferi in human monocytes in-volves Toll-like receptor (TLR) 2 and TLR8 cooperativity and TLR8-mediatedinduction of IFN-b. Proc Natl Acad Sci U S A 2011;108:3683e8.

[460] Cervantes JL, La Vake CJ, Weinerman B, Luu S, O'Connell C, Verardi PH, et al.Human TLR8 is activated upon recognition of Borrelia burgdorferi RNA in thephagosome of human monocytes. J Leukoc Biol 2013;94:1231e41.

[461] Liu N, Montgomery RR, Barthold SW, Bockenstedt LK. Myeloid differentiationantigen 88 deficiency impairs pathogen clearance but does not alterinflammation in Borrelia burgdorferi-infected mice. Infect Immun 2004;72:3195e203.

[462] Oosting M, Berende A, Sturm P, Ter Hofstede HJ, de Jong DJ, Kanneganti TD,et al. Recognition of Borrelia burgdorferi by NOD2 is central for the inductionof an inflammatory reaction. J Infect Dis 2010;201:1849e58.

[463] Oosting M, Buffen K, Malireddi SR, Sturm P, Verschueren I, Koenders MI,et al. Murine Borrelia arthritis is highly dependent on ASC and caspase-1, butindependent of NLRP3. Arthritis Res Ther 2012;14:R247.

[464] Salazar JC, Pope CD, Sellati TJ, Feder Jr HM, Kiely TG, Dardick KR, et al.Coevolution of markers of innate and adaptive immunity in skin and pe-ripheral blood of patients with erythema migrans. J Immunol 2003;171:2660e70.

[465] Filgueira L, Nestl�e FO, Rittig M, Joller HI, Groscurth P. Human dendritic cellsphagocytose and process Borrelia burgdorferi. J Immunol 1996;157:2998e3005.

[466] Suhonen J, Komi J, Soukka J, Lassila O, Viljanen MK. Interaction betweenBorrelia burgdorferi and immature human dendritic cells. Scand J Immunol2003;58:67e75.

[467] Mbow ML, Zeidner N, Panella N, Titus RG, Piesman J. Borrelia burgdorferi-pulsed dendritic cells induce a protective immune response against tick-transmitted spirochetes. Infect Immun 1997;65:3386e90.

[468] de Koning J. Histopathologic patterns of erythema migrans and borreliallymphocytoma. Clin Dermatol 1993;11:377e83.

[469] Nanagara R, Duray PH, Schumacher Jr HR. Ultrastructural demonstration ofspirochetal antigens in synovial fluid and synovial membrane in chronicLyme disease: possible factors contributing to persistence of organisms. HumPathol 1996;27:1025e34.

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

[470] H€aupl T, Hahn G, Rittig M, Krause A, Schoerner C, Sch€onherr U, et al.Persistence of Borrelia burgdorferi in ligamentous tissue from a patient withchronic Lyme borreliosis. Arthritis Rheum 1993;36:1621e6.

[471] Gravano DM, Hoyer KK. Promotion and prevention of autoimmune diseaseby CD8þ T cells. J Autoimmun 2013;45:68e79.

[472] Beermann C, Wunderli-Allenspach H, Groscurth P, Filgueira L. Lipoproteinsfrom Borrelia burgdorferi applied in liposomes and presented by dendriticcells induce CD8(þ) T-lymphocytes in vitro. Cell Immunol 2000;201:124e31.

[473] Aberer E, Klade H, Hobisch G. A clinical, histological, and immunohisto-chemical comparison of acrodermatitis chronica atrophicans and morphea.Am J Dermatopathol 1991;13:334e41.

[474] Müllegger RR, Means TK, Shin JJ, Lee M, Jones KL, Glickstein LJ, et al. Che-mokine signatures in the skin disorders of Lyme borreliosis in Europe:predominance of CXCL9 and CXCL10 in erythema migrans and acroderma-titis and CXCL13 in lymphocytoma. Infect Immun 2007;75:4621e8.

[475] Sj€owall J, Fryland L, Nordberg M, Sj€ogren F, Garpmo U, Jansson C, et al.Decreased Th1-type inflammatory cytokine expression in the skin is asso-ciated with persisting symptoms after treatment of erythema migrans. PLoSOne 2011;6:e18220.

[476] Jones KL, Muellegger RR, Means TK, Lee M, Glickstein LJ, Damle N, et al.Higher mRNA levels of chemokines and cytokines associated with macro-phage activation in erythema migrans skin lesions in patients from theUnited States than in patients from Austria with Lyme borreliosis. Clin InfectDis 2008;46:85e92.

[477] Lahiri A, Pochard P, Le Pottier L, Tobon GJ, Bendaoud B, Youinou P, et al. Thecomplexity of the BAFF TNF-family members: implications for autoimmu-nity. J Autoimmun 2012;39:189e98.

[478] Glickstein L, Moore B, Bledsoe T, Damle N, Sikand V, Steere AC. Inflammatorycytokine production predominates in early Lyme disease in patients witherythema migrans. Infect Immun 2003;71:6051e3.

[479] Garcia-Monco JC, Villar BF, Alen JC, Benach JL. Borrelia burgdorferi in thecentral nervous system: experimental and clinical evidence for early inva-sion. J Infect Dis 1990;161:1187e93.

[480] Peters DJ, Benach JL. Borrelia burgdorferi adherence and injury to undiffer-entiated and differentiated neural cells in vitro. J Infect Dis 1997;176:470e7.

[481] Gilmore RDJ. Immunoseclusion and chronic infection by Borrelia burgdorferi.In: Embers ME, editor. The pathogenic spirochetes: strategies for evasion ofhost immunity and persistence. New York, NY: Springer ScienceþBusinessMedia; 2012. p. 19e39.

[482] Rupprecht TA, Koedel U, Fingerle V, Pfister HW. The pathogenesis of Lymeneuroborreliosis: from infection to inflammation. Mol Med 2008;14:205e12.

[483] Ramesh G, Santana-Gould L, Inglis FM, England JD, Philipp MT. The Lymedisease spirochete Borrelia burgdorferi induces inflammation and apoptosisin cells from dorsal root ganglia. J Neuroinflamm 2013;10:88.

[484] Meurers B, Kohlhepp W, Gold R, Rohrbach E, Mertens HG. Histopathologicalfindings in the central and peripheral nervous systems in neuroborreliosis. Areport of three cases. J Neurol 1990;237:113e6.

[485] Camponovo F, Meier C. Neuropathy of vasculitic origin in a case of Garin-Boujadoux-Bannwarth syndrome with positive borrelia antibody response.J Neurol 1986;233:69e72.

[486] Vallat JM, Hugon J, Lubeau M, Leboutet MJ, Dumas M, Desproges-Gotteron R.Tick-bite meningoradiculoneuritis: clinical, electrophysiologic, and histo-logic findings in 10 cases. Neurology 1987;37:749e53.

[487] Bertrand E, Szpak GM, Pilkowska E, Habib N, Lipczynska-Lojkowska W,Rudnicka A, et al. Central nervous system infection caused by Borreliaburgdorferi. Clinico-pathological correlation of three post-mortem cases.Folia Neuropathol 1999;37:43e51.

[488] Lünemann JD, Gelderblom H, Sospedra M, Quandt JA, Pinilla C, Marques A,et al. Cerebrospinal fluid-infiltrating CD4þ T cells recognize Borrelia burg-dorferi lysine-enriched protein domains and central nervous system auto-antigens in early Lyme encephalitis. Infect Immun 2007;75:243e51.

[489] Hemmer B, Gran B, Zhao Y, Marques A, Pascal J, Tzou A, et al. Identification ofcandidate T-cell epitopes and molecular mimics in chronic Lyme disease. NatMed 1999;5:1375e82.

[490] Ekerfelt C, Ernerudh J, Bunikis J, Vrethem M, Aagesen J, Roberg M, et al.Compartmentalization of antigen specific cytokine responses to the centralnervous system in CNS borreliosis: secretion of IFN-gamma predominatesover IL-4 secretion in response to outer surface proteins of Lyme diseaseBorrelia spirochetes. J Neuroimmunol 1997;79:155e62.

[491] Widhe M, Grusell M, Ekerfelt C, Vrethem M, Forsberg P, Ernerudh J. Cyto-kines in Lyme borreliosis: lack of early tumour necrosis factor-a and trans-forming growth factor-b1 responses are associated with chronicneuroborreliosis. Immunology 2002;107:46e55.

[492] Widhe M, Jarefors S, Ekerfelt C, Vrethem M, Bergstrom S, Forsberg P, et al.Borrelia-specific interferon-gamma and interleukin-4 secretion in cerebro-spinal fluid and blood during Lyme borreliosis in humans: association withclinical outcome. J Infect Dis 2004;189:1881e91.

[493] Widhe M, Skogman BH, Jarefors S, Eknefelt M, Enestrom G, Nordwall M, et al.Up-regulation of Borrelia-specific IL-4- and IFN-g-secreting cells in cere-brospinal fluid from children with Lyme neuroborreliosis. Int Immunol2005;17:1283e91.

[494] Wang WZ, Fredrikson S, Sun JB, Link H. Lyme neuroborreliosis: evidence forpersistent up-regulation of Borrelia burgdorferi-reactive cells secretinginterferon-gamma. Scand J Immunol 1995;42:694e700.

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3432

[495] Cerar T, Ogrinc K, Lotric-Furlan S, Kobal J, Levicnik-Stezinar S, Strle F, et al.Diagnostic value of cytokines and chemokines in Lyme neuroborreliosis. ClinVaccine Immunol 2013;20:1578e84.

[496] Henningsson AJ, Tjernberg I, Malmvall BE, Forsberg P, Ernerudh J. Indicationsof Th1 and Th17 responses in cerebrospinal fluid from patients with Lymeneuroborreliosis: a large retrospective study. J Neuroinflamm 2011;8:36.

[497] Cepok S, Zhou D, Vogel F, Rosche B, Grummel V, Sommer N, et al. The im-mune response at onset and during recovery from Borrelia burgdorferimeningoradiculitis. Arch Neurol 2003;60:849e55.

[498] Jacobsen M, Zhou D, Cepok S, Nessler S, Happel M, Stei S, et al. Clonalaccumulation of activated CD8þ T cells in the central nervous system duringthe early phase of neuroborreliosis. J Infect Dis 2003;187:963e73.

[499] Ekerfelt C, Jarefors S, Tynngard N, Hedlund M, Sander B, Bergstrom S, et al.Phenotypes indicating cytolytic properties of Borrelia-specific interferon-gsecreting cells in chronic Lyme neuroborreliosis. J Neuroimmunol 2003;145:115e26.

[500] Kowarik MC, Cepok S, Sellner J, Grummel V, Weber MS, Korn T, et al. CXCL13is the major determinant for B cell recruitment to the CSF during neuro-inflammation. J Neuroinflamm 2012;9:93.

[501] Rupprecht TA, Plate A, Adam M, Wick M, Kastenbauer S, Schmidt C, et al. Thechemokine CXCL13 is a key regulator of B cell recruitment to the cerebro-spinal fluid in acute Lyme neuroborreliosis. J Neuroinflamm 2009;6:42.

[502] Cepok S, Rosche B, Grummel V, Vogel F, Zhou D, Sayn J, et al. Short-livedplasma blasts are the main B cell effector subset during the course of mul-tiple sclerosis. Brain 2005;128:1667e76.

[503] Cusick MF, Libbey JE, Fujinami RS. Molecular mimicry as a mechanism ofautoimmune disease. Clin Rev Allergy Immunol 2012;42:102e11.

[504] Alaedini A, Latov N. Antibodies against OspA epitopes of Borrelia burgdorfericross-react with neural tissue. J Neuroimmunol 2005;159:192e5.

[505] Dai Z, Lackland H, Stein S, Li Q, Radziewicz R, Williams S, et al. Molecularmimicry in Lyme disease: monoclonal antibody H9724 to B. burgdorferiflagellin specifically detects chaperonin-HSP60. Biochim Biophys Acta1993;1181:97e100.

[506] Sigal LH, Tatum AH. Lyme disease patients' serum contains IgM antibodies toBorrelia burgdorferi that cross-react with neuronal antigens. Neurology1988;38:1439e42.

[507] Sigal LH. Cross-reactivity between Borrelia burgdorferi flagellin and a humanaxonal 64,000 molecular weight protein. J Infect Dis 1993;167:1372e8.

[508] Kuenzle S, von Budingen HC, Meier M, Harrer MD, Urich E, Becher B, et al.Pathogen specificity and autoimmunity are distinct features of antigen-driven immune responses in neuroborreliosis. Infect Immun 2007;75:3842e7.

[509] Baig S, Olsson T, Hojeberg B, Link H. Cells secreting antibodies to myelinbasic protein in cerebrospinal fluid of patients with Lyme neuroborreliosis.Neurology 1991;41:581e7.

[510] Brooks WH. Mechanisms and pathophysiology of autoimmune disease. ClinRev Allergy Immunol 2012;42:1e4.

[511] Cornec D, Devauchelle-Pensec V, Tobon GJ, Pers JO, Jousse-Joulin S, Saraux A.B cells in Sjogren's syndrome: from pathophysiology to diagnosis andtreatment. J Autoimmun 2012;39:161e7.

[512] Borchers AT, Leibushor N, Cheema GS, Naguwa SM, Gershwin ME. Immune-mediated adverse effects of biologicals used in the treatment of rheumaticdiseases. J Autoimmun 2011;37:273e88.

[513] Keane-Myers A, Nickell SP. T cell subset-dependent modulation of immunityto Borrelia burgdorferi in mice. J Immunol 1995;154:1770e6.

[514] Keane-Myers A, Nickell SP. Role of IL-4 and IFN-gamma in modulation ofimmunity to Borrelia burgdorferi in mice. J Immunol 1995;155:2020e8.

[515] Barthold SW, Persing DH, Armstrong AL, Peeples RA. Kinetics of Borreliaburgdorferi dissemination and evolution of disease after intradermal inocu-lation of mice. Am J Pathol 1991;139:263e73.

[516] Barthold SW, Sidman CL, Smith AL. Lyme borreliosis in genetically resistantand susceptible mice with severe combined immunodeficiency. Am J TropMed Hyg 1992;47:605e13.

[517] Barthold SW, deSouza M, Feng S. Serum-mediated resolution of Lymearthritis in mice. Lab Invest 1996;74:57e67.

[518] McKisic MD, Barthold SW. T-cell-independent responses to Borrelia burg-dorferi are critical for protective immunity and resolution of Lyme disease.Infect Immun 2000;68:5190e7.

[519] Schaible UE, Wallich R, Kramer MD, Nerz G, Stehle T, Museteanu C, et al.Protection against Borrelia burgdorferi infection in SCID mice is conferred bypresensitized spleen cells and partially by B but not T cells alone. IntImmunol 1994;6:671e81.

[520] Bockenstedt LK, Kang I, Chang C, Persing D, Hayday A, Barthold SW. CD4þ Thelper 1 cells facilitate regression of murine Lyme carditis. Infect Immun2001;69:5264e9.

[521] McKisic MD, Redmond WL, Barthold SW. Cutting edge: T cell-mediated pa-thology in murine Lyme borreliosis. J Immunol 2000;164:6096e9.

[522] Fikrig E, Barthold SW, Chen M, Chang CH, Flavell RA. Protective antibodiesdevelop, and murine Lyme arthritis regresses, in the absence of MHC class IIand CD4þ T cells. J Immunol 1997;159:5682e6.

[523] Fikrig E, Barthold SW, Chen M, Grewal IS, Craft J, Flavell RA. Protective an-tibodies in murine Lyme disease arise independently of CD40 ligand.J Immunol 1996;157:1e3.

[524] Mbow ML, Zeidner N, Gilmore Jr RD, Dolan M, Piesman J, Titus RG. Majorhistocompatibility complex class II-independent generation of neutralizing

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

antibodies against T-cell-dependent Borrelia burgdorferi antigens presentedby dendritic cells: regulation by NK and gd T cells. Infect Immun 2001;69:2407e15.

[525] Barthold SW, Hodzic E, Tunev S, Feng S. Antibody-mediated disease remis-sion in the mouse model of Lyme borreliosis. Infect Immun 2006;74:4817e25.

[526] Belperron AA, Dailey CM, Booth CJ, Bockenstedt LK. Marginal zone B-celldepletion impairs murine host defense against Borrelia burgdorferi infection.Infect Immun 2007;75:3354e60.

[527] Matyniak JE, Reiner SL. T helper phenotype and genetic susceptibility inexperimental Lyme disease. J Exp Med 1995;181:1251e4.

[528] Kang I, Barthold SW,PersingDH,Bockenstedt LK. T-helper-cell cytokines in theearly evolution of murine Lyme arthritis. Infect Immun 1997;65:3107e11.

[529] Glickstein L, Edelstein M, Dong JZ. Gamma interferon is not required forarthritis resistance in the murine Lyme disease model. Infect Immun2001;69:3737e43.

[530] Potter MR, Noben-Trauth N, Weis JH, Teuscher C, Weis JJ. Interleukin-4 (IL-4)and IL-13 signaling pathways do not regulate Borrelia burgdorferi-inducedarthritis in mice: IgG1 is not required for host control of tissue spirochetes.Infect Immun 2000;68:5603e9.

[531] Brown CR, Reiner SL. Experimental Lyme arthritis in the absence ofinterleukin-4 or gamma interferon. Infect Immun 1999;67:3329e33.

[532] Brown CR, Blaho VA, Fritsche KL, Loiacono CM. Stat1 deficiency exacerbatescarditis but not arthritis during experimental Lyme borreliosis. J InterferonCytokine Res 2006;26:390e9.

[533] Zeidner N, Mbow ML, Dolan M, Massung R, Baca E, Piesman J. Effects ofIxodes scapularis and Borrelia burgdorferi on modulation of the host immuneresponse: induction of a TH2 cytokine response in Lyme disease-susceptible(C3H/HeJ) mice but not in disease-resistant (BALB/c) mice. Infect Immun1997;65:3100e6.

[534] Zeidner NS, Schneider BS, Rutherford JS, Dolan MC. Suppression of Th2 cy-tokines reduces tick-transmitted Borrelia burgdorferi load in mice. J Parasitol2008;94:767e9.

[535] Brown CR, Lai AY, Callen ST, Blaho VA, Hughes JM, Mitchell WJ. Adenoviraldelivery of interleukin-10 fails to attenuate experimental Lyme disease.Infect Immun 2008;76:5500e7.

[536] Sonderegger FL, Ma Y, Maylor-Hagan H, Brewster J, Huang X, Spangrude GJ,et al. Localized production of IL-10 suppresses early inflammatory cellinfiltration and subsequent development of IFN-gamma-mediated Lymearthritis. J Immunol 2012;188:1381e93.

[537] Hansen ES, Medic V, Kuo J, Warner TF, Schell RF, Nardelli DT. Interleukin-10(IL-10) inhibits Borrelia burgdorferi-induced IL-17 production and attenuatesIL-17-mediated Lyme arthritis. Infect Immun 2013;81:4421e30.

[538] Burchill MA, Nardelli DT, England DM, DeCoster DJ, Christopherson JA,Callister SM, et al. Inhibition of interleukin-17 prevents the development ofarthritis in vaccinated mice challenged with Borrelia burgdorferi. InfectImmun 2003;71:3437e42.

[539] Amlong CA, Nardelli DT, Peterson SH, Warner TF, Callister SM, Schell RF.Anti-interleukin-15 prevents arthritis in Borrelia-vaccinated and -infectedmice. Clin Vaccine Immunol 2006;13:289e96.

[540] Kotloski NJ, Nardelli DT, Peterson SH, Torrealba JR, Warner TF, Callister SM,et al. Interleukin-23 is required for development of arthritis in mice vacci-nated and challenged with Borrelia species. Clin Vaccine Immunol 2008;15:1199e207.

[541] Nardelli DT, Kevin Luk KH, Kotloski NJ, Warner TF, Torrealba JR, Callister SM,et al. Role of IL-17, transforming growth factor-beta, and IL-6 in the devel-opment of arthritis and production of anti-outer surface protein A borre-liacidal antibodies in Borrelia-vaccinated and -challenged mice. FEMSImmunol Med Microbiol 2008;53:265e74.

[542] Nardelli DT, Cloute JP, Luk KH, Torrealba J, Warner TF, Callister SM, et al.CD4þ CD25þ T cells prevent arthritis associated with Borrelia vaccination andinfection. Clin Diagn Lab Immunol 2005;12:786e92.

[543] Nardelli DT, Burchill MA, England DM, Torrealba J, Callister SM, Schell RF.Association of CD4þ CD25þ T cells with prevention of severe destructivearthritis in Borrelia burgdorferi-vaccinated and challenged gammainterferon-deficient mice treated with anti-interleukin-17 antibody. ClinDiagn Lab Immunol 2004;11:1075e84.

[544] Nardelli DT, Callister SM, Schell RF. Lyme arthritis: current concepts and achange in paradigm. Clin Vaccine Immunol 2008;15:21e34.

[545] Nardelli DT, Luedtke JO, Munson EL, Warner TF, Callister SM, Schell RF.Significant differences between the Borrelia-infection and Borrelia-vaccina-tion and -infection models of Lyme arthritis in C3H/HeN mice. FEMSImmunol Med Microbiol 2010;60:78e89.

[546] Barthold SW, Feng S, Bockenstedt LK, Fikrig E, Feen K. Protective andarthritis-resolving activity in sera of mice infected with Borrelia burgdorferi.Clin Infect Dis 1997;25(Suppl. 1):S9e17.

[547] Hendrickx G, De Boeck H, Goossens A, Demanet C, Vandenplas Y. Persistentsynovitis in children with Lyme arthritis: two unusual cases. An immuno-genetic approach. Eur J Pediatr 2004;163:646e50.

[548] Deanehan JK, Nigrovic PA, Milewski MD, Tan Tanny SP, Kimia AA, Smith BG,et al. Synovial fluid findings in children with knee monoarthritis in Lymedisease endemic areas. Pediatr Emerg Care 2014;30:16e9.

[549] Miller LC, Lynch EA, Isa S, Logan JW, Dinarello CA, Steere AC. Balance ofsynovial fluid IL-1b and IL-1 receptor antagonist and recovery from Lymearthritis. Lancet 1993;341:146e8.

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e34 33

[550] Codolo G, Bossi F, Durigutto P, Bella CD, Fischetti F, Amedei A, et al.Orchestration of inflammation and adaptive immunity in Borrelia burgdor-feri-induced arthritis by neutrophil-activating protein A. Arthritis Rheum2013;65:1232e42.

[551] Yin Z, Braun J, Neure L, Wu P, Eggens U, Krause A, et al. T cell cytokinepattern in the joints of patients with Lyme arthritis and its regulation bycytokines and anticytokines. Arthritis Rheum 1997;40:69e79.

[552] Gross DM, Steere AC, Huber BT. T helper 1 response is dominant and local-ized to the synovial fluid in patients with Lyme arthritis. J Immunol1998;160:1022e8.

[553] Yssel H, Shanafelt MC, Soderberg C, Schneider PV, Anzola J, Peltz G. Borreliaburgdorferi activates a T helper type 1-like T cell subset in Lyme arthritis.J Exp Med 1991;174:593e601.

[554] Yssel H, Nakamoto T, Schneider P, Freitas V, Collins C, Webb D, et al. Analysisof T lymphocytes cloned from the synovial fluid and blood of a patient withlyme arthritis. Int Immunol 1990;2:1081e9.

[555] Pohl-Koppe A, Balashov KE, Steere AC, Logigian EL, Hafler DA. Identificationof a T cell subset capable of both IFN-g and IL-10 secretion in patients withchronic Borrelia burgdorferi infection. J Immunol 1998;160:1804e10.

[556] Shin JJ, Glickstein LJ, Steere AC. High levels of inflammatory chemokines andcytokines in joint fluid and synovial tissue throughout the course ofantibiotic-refractory Lyme arthritis. Arthritis Rheum 2007;56:1325e35.

[557] Busch DH, Jassoy C, Brinckmann U, Girschick H, Huppertz HI. Detection ofBorrelia burgdorferi-specific CD8þ cytotoxic T cells in patients with Lymearthritis. J Immunol 1996;157:3534e41.

[558] Kurko J, Besenyei T, Laki J, Glant TT, Mikecz K, Szekanecz Z. Genetics ofrheumatoid arthritis - a comprehensive review. Clin Rev Allergy Immunol2013;45:170e9.

[559] Infante-Duarte C, Horton HF, Byrne MC, Kamradt T. Microbial lipopeptidesinduce the production of IL-17 in Th cells. J Immunol 2000;165:6107e15.

[560] Codolo G, Amedei A, Steere AC, Papinutto E, Cappon A, Polenghi A, et al.Borrelia burgdorferi NapA-driven Th17 cell inflammation in Lyme arthritis.Arthritis Rheum 2008;58:3609e17.

[561] Oosting M, ter Hofstede H, van de Veerdonk FL, Sturm P, Kullberg BJ, van derMeer JW, et al. Role of interleukin-23 (IL-23) receptor signaling for IL-17responses in human Lyme disease. Infect Immun 2011;79:4681e7.

[562] Russell TM, Johnson BJ. Lyme disease spirochaetes possess an aggrecan-binding protease with aggrecanase activity. Mol Microbiol 2013;90:228e40.

[563] Hu LT, Eskildsen MA, Masgala C, Steere AC, Arner EC, Pratta MA, et al. Hostmetalloproteinases in Lyme arthritis. Arthritis Rheum 2001;44:1401e10.

[564] Lin B, Kidder JM, Noring R, Steere AC, Klempner MS, Hu LT. Differences insynovial fluid levels of matrix metalloproteinases suggest separate mecha-nisms of pathogenesis in Lyme arthritis before and after antibiotic treatment.J Infect Dis 2001;184:174e80.

[565] Behera AK, Hildebrand E, Scagliotti J, Steere AC, Hu LT. Induction of hostmatrix metalloproteinases by Borrelia burgdorferi differs in human andmurine Lyme arthritis. Infect Immun 2005;73:126e34.

[566] Behera AK, Hildebrand E, Szafranski J, Hung HH, Grodzinsky AJ, Lafyatis R,et al. Role of aggrecanase 1 in Lyme arthritis. Arthritis Rheum 2006;54:3319e29.

[567] Steere AC, Dwyer E, Winchester R. Association of chronic Lyme arthritis withHLA-DR4 and HLA-DR2 alleles. N Engl J Med 1990;323:219e23.

[568] Carlson D, Hernandez J, Bloom BJ, Coburn J, Aversa JM, Steere AC. Lack ofBorrelia burgdorferi DNA in synovial samples from patients with antibiotictreatment-resistant Lyme arthritis. Arthritis Rheum 1999;42:2705e9.

[569] Li X, McHugh GA, Damle N, Sikand VK, Glickstein L, Steere AC. Burden andviability of Borrelia burgdorferi in skin and joints of patients with erythemamigrans or Lyme arthritis. Arthritis Rheum 2011;63:2238e47.

[570] Chen J, Field JA, Glickstein L, Molloy PJ, Huber BT, Steere AC. Association ofantibiotic treatment-resistant Lyme arthritis with T cell responses to domi-nant epitopes of outer surface protein A of Borrelia burgdorferi. ArthritisRheum 1999;42:1813e22.

[571] Meyer AL, Trollmo C, Crawford F, Marrack P, Steere AC, Huber BT, et al. Directenumeration of Borrelia-reactive CD4 T cells ex vivo by using MHC class IItetramers. Proc Natl Acad Sci U S A 2000;97:11433e8.

[572] Ghosh S, Seward R, Costello CE, Stollar BD, Huber BT. Autoantibodies fromsynovial lesions in chronic, antibiotic treatment-resistant Lyme arthritis bindcytokeratin-10. J Immunol 2006;177:2486e94.

[573] Drouin EE, Seward RJ, Strle K, McHugh G, Katchar K, Londono D, et al. A novelhuman autoantigen, endothelial cell growth factor, is a target of T and B cellresponses in patients with Lyme disease. Arthritis Rheum 2013;65:186e96.

[574] Yazici H, Ugurlu S, Seyahi E. Behcet syndrome: is it one condition? Clin RevAllergy Immunol 2012;43:275e80.

[575] Zen M, Gatto M, Domeneghetti M, Palma L, Borella E, Iaccarino L, et al.Clinical guidelines and definitions of autoinflammatory diseases: contrastsand comparisons with autoimmunity e a comprehensive review. Clin RevAllergy Immunol 2013;45:227e35.

[576] Krause A, Burmester GR, Rensing A, Schoerner C, Schaible UE, Simon MM,et al. Cellular immune reactivity to recombinant OspA and flagellin fromBorrelia burgdorferi in patients with Lyme borreliosis. Complexity of humoraland cellular immune responses. J Clin Invest 1992;90:1077e84.

[577] Drouin EE, Glickstein LJ, Steere AC. Molecular characterization of theOspA161-175 T cell epitope associated with treatment-resistant Lymearthritis: differences among the three pathogenic species of Borrelia burg-dorferi sensu lato. J Autoimmun 2004;23:281e92.

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

[578] Priem S, Burmester GR, Kamradt T, Wolbart K, Rittig MG, Krause A. Detectionof Borrelia burgdorferi by polymerase chain reaction in synovial membrane,but not in synovial fluid from patients with persisting Lyme arthritis afterantibiotic therapy. Ann Rheum Dis 1998;57:118e21.

[579] Limbach FX, Jaulhac B, Puechal X, Monteil H, Kuntz JL, Piemont Y, et al.Treatment resistant Lyme arthritis caused by Borrelia garinii. Ann Rheum Dis2001;60:284e6.

[580] Miklossy J, Khalili K, Gern L, Ericson RL, Darekar P, Bolle L, et al. Borreliaburgdorferi persists in the brain in chronic Lyme neuroborreliosis and may beassociated with Alzheimer disease. J Alzheimers Dis 2004;6:639e49. dis-cussion 73e81.

[581] Bradley JF, Johnson RC, Goodman JL. The persistence of spirochetal nucleicacids in active Lyme arthritis. Ann Intern Med 1994;120:487e9.

[582] Lipowsky C, Altwegg M, Michel BA, Bruhlmann P. Detection of Borreliaburgdorferi by species-specific and broad-range PCR of synovial fluid andsynovial tissue of Lyme arthritis patients before and after antibiotic treat-ment. Clin Exp Rheumatol 2003;21:271e2.

[583] Battafarano DF, Combs JA, Enzenauer RJ, Fitzpatrick JE. Chronic septicarthritis caused by Borrelia burgdorferi. Clin Orthop Relat Res 1993:238e41.

[584] Frey M, Jaulhac B, Piemont Y, Marcellin L, Boohs PM, Vautravers P, et al.Detection of Borrelia burgdorferi DNA in muscle of patients with chronicmyalgia related to Lyme disease. Am J Med 1998;104:591e4.

[585] Ghosh S, Steere AC, Stollar BD, Huber BT. In situ diversification of the anti-body repertoire in chronic Lyme arthritis synovium. J Immunol 2005;174:2860e9.

[586] Snydman DR, Schenkein DP, Berardi VP, Lastavica CC, Pariser KM. Borreliaburgdorferi in joint fluid in chronic Lyme arthritis. Ann Intern Med 1986;104:798e800.

[587] Wormser GP, Nadelman RB, Schwartz I. The amber theory of Lyme arthritis:initial description and clinical implications. Clin Rheumatol 2012;31:989e94.

[588] Bockenstedt LK, Gonzalez DG, Haberman AM, Belperron AA. Spirochete an-tigens persist near cartilage after murine Lyme borreliosis therapy. J ClinInvest 2012;122:2652e60.

[589] Bockenstedt LK, Mao J, Hodzic E, Barthold SW, Fish D. Detection of attenu-ated, noninfectious spirochetes in Borrelia burgdorferi-infected mice afterantibiotic treatment. J Infect Dis 2002;186:1430e7.

[590] Hodzic E, Feng S, Holden K, Freet KJ, Barthold SW. Persistence of Borreliaburgdorferi following antibiotic treatment in mice. Antimicrob Agents Che-mother 2008;52:1728e36.

[591] Barthold SW, Hodzic E, Imai DM, Feng S, Yang X, Luft BJ. Ineffectiveness oftigecycline against persistent Borrelia burgdorferi. Antimicrob Agents Che-mother 2010;54:643e51.

[592] Hodzic E, Imai D, Feng S, Barthold SW. Resurgence of persisting non-cultivable Borrelia burgdorferi following antibiotic treatment in mice. PLoSOne 2014;9:e86907.

[593] Wormser GP, Schwartz I. Antibiotic treatment of animals infected withBorrelia burgdorferi. Clin Microbiol Rev 2009;22:387e95.

[594] Embers ME, Barthold SW, Borda JT, Bowers L, Doyle L, Hodzic E, et al.Persistence of Borrelia burgdorferi in rhesus macaques following antibiotictreatment of disseminated infection. PLoS One 2012;7:e29914.

[595] Straubinger RK, Summers BA, Chang YF, Appel MJ. Persistence of Borreliaburgdorferi in experimentally infected dogs after antibiotic treatment. J ClinMicrobiol 1997;35:111e6.

[596] Hunfeld KP, Ruzic-Sabljic E, Norris DE, Kraiczy P, Strle F. In vitro suscepti-bility testing of Borrelia burgdorferi sensu lato isolates cultured from patientswith erythema migrans before and after antimicrobial chemotherapy.Antimicrob Agents Chemother 2005;49:1294e301.

[597] Preac-Mursic V, Weber K, Pfister HW, Wilske B, Gross B, Baumann A, et al.Survival of Borrelia burgdorferi in antibiotically treated patients with Lymeborreliosis. Infection 1989;17:355e9.

[598] Chandra A, Wormser GP, Klempner MS, Trevino RP, Crow MK, Latov N, et al.Anti-neural antibody reactivity in patients with a history of Lyme borreliosisand persistent symptoms. Brain Behav Immun 2010;24:1018e24.

[599] Strle K, Stupica D, Drouin EE, Steere AC, Strle F. Elevated levels of IL-23 in asubset of patients with post-Lyme disease symptoms following erythemamigrans. Clin Infect Dis 2014;58:372e80.

[600] Margos G, Vollmer SA, Ogden NH, Fish D. Population genetics, taxonomy,phylogeny and evolution of Borrelia burgdorferi sensu lato. Infect Genet Evol2011;11:1545e63.

[601] Rudenko N, Golovchenko M, Grubhoffer L, Oliver Jr JH. Updates on Borreliaburgdorferi sensu lato complex with respect to public health. Ticks TickBorne Dis 2011;2:123e8.

[602] Vasiliu V, Herzer P, Rossler D, Lehnert G, Wilske B. Heterogeneity of Borreliaburgdorferi sensu lato demonstrated by an ospA-type-specific PCR in synovialfluid from patients with Lyme arthritis. Med Microbiol Immunol 1998;187:97e102.

[603] Eiffert H, Karsten A, Thomssen R, Christen HJ. Characterization of Borreliaburgdorferi strains in Lyme arthritis. Scand J Infect Dis 1998;30:265e8.

[604] van Burgel ND, Balmus NC, Fikrig E, van Dam AP. Infectivity of Borreliaburgdorferi sensu lato is unaltered in C3-deficient mice. Ticks Tick Borne Dis2011;2:20e6.

[605] Strle F, Picken RN, Cheng Y, Cimperman J, Maraspin V, Lotric-Furlan S, et al.Clinical findings for patients with Lyme borreliosis caused by Borrelia burg-dorferi sensu lato with genotypic and phenotypic similarities to strain 25015.Clin Infect Dis 1997;25:273e80.

A rigorous review of diagnostic criteria and treatment, Journal of

A.T. Borchers et al. / Journal of Autoimmunity xxx (2014) 1e3434

[606] Rudenko N, Golovchenko M, Ruzek D, Piskunova N, Mallatova N,Grubhoffer L. Molecular detection of Borrelia bissettii DNA in serum samplesfrom patients in the Czech Republic with suspected borreliosis. FEMSMicrobiol Lett 2009;292:274e81.

[607] Rudenko N, Golovchenko M, Mokracek A, Piskunova N, Ruzek D,Mallatova N, et al. Detection of Borrelia bissettii in cardiac valve tissue of apatient with endocarditis and aortic valve stenosis in the Czech Republic.J Clin Microbiol 2008;46:3540e3.

[608] Schneider BS, Schriefer ME, Dietrich G, Dolan MC, Morshed MG, Zeidner NS.Borrelia bissettii isolates induce pathology in a murine model of disease.Vector Borne Zoonotic Dis 2008;8:623e33.

[609] Piesman J, Dolan MC, Happ CM, Luft BJ, Rooney SE, Mather TN, et al. Durationof immunity to reinfection with tick-transmitted Borrelia burgdorferi innaturally infected mice. Infect Immun 1997;65:4043e7.

[610] Munson E, Nardelli DT, Du Chateau BK, Callister SM, Schell RF. Hamster andmurine models of severe destructive Lyme arthritis. Clin Dev Immunol2012;2012:504215.

[611] Escudero R, Barral M, Perez A, Vitutia MM, Garcia-Perez AL, Jimenez S, et al.Molecular and pathogenic characterization of Borrelia burgdorferi sensu latoisolates from Spain. J Clin Microbiol 2000;38:4026e33.

[612] Pachner AR, Dail D, Bai Y, Sondey M, Pak L, Narayan K, et al. Genotype de-termines phenotype in experimental Lyme borreliosis. Ann Neurol 2004;56:361e70.

[613] Kaneda K, Masuzawa T, Simon MM, Isogai E, Isogai H, Yasugami K, et al.Infectivity and arthritis induction of Borrelia japonica on SCID mice and

Please cite this article in press as: Borchers AT, et al., Lyme disease:Autoimmunity (2014), http://dx.doi.org/10.1016/j.jaut.2014.09.004

immune competent mice: possible role of galactosylceramide binding ac-tivity on initiation of infection. Microbiol Immunol 1998;42:171e5.

[614] Masuzawa T, Kurita T, Kawabata H, Muramatsu K, Uchikawa K, Yanagihara Y.Infectivity and early antibody response to Borrelia burgdorferi sensu latoisolated in Japan in outbred mice. Microbiol Immunol 1994;38:621e7.

[615] Fikrig E, Barthold SW, Persing DH, Sun X, Kantor FS, Flavell RA. Borreliaburgdorferi strain 25015: characterization of outer surface protein A andvaccination against infection. J Immunol 1992;148:2256e60.

[616] Anderson JF, Barthold SW, Magnarelli LA. Infectious but nonpathogenicisolate of Borrelia burgdorferi. J Clin Microbiol 1990;28:2693e9.

[617] Collares-Pereira M, Couceiro S, Franca I, Kurtenbach K, Schafer SM,Vitorino L, et al. First isolation of Borrelia lusitaniae from a human patient.J Clin Microbiol 2004;42:1316e8.

[618] Zeidner NS, Nuncio MS, Schneider BS, Gern L, Piesman J, Brandao O, et al.A Portuguese isolate of Borrelia lusitaniae induces disease in C3H/HeN mice.J Med Microbiol 2001;50:1055e60.

[619] Maraspin V, Ruzic-Sabljic E, Strle F. Lyme borreliosis and Borrelia spielmanii.Emerg Infect Dis 2006;12:1177.

[620] F€oldv�ari G, Farkas R, Lakos A. Borrelia spielmanii erythema migrans. HungEmerg Infect Dis 2005;11:1794e5.

[621] Saito K, Ito T, Asashima N, Ohno M, Nagai R, Fujita H, et al. Case report:Borrelia valaisiana infection in a Japanese man associated with traveling toforeign countries. Am J Trop Med Hyg 2007;77:1124e7.

[622] Diza E, Papa A, Vezyri E, Tsounis S, Milonas I, Antoniadis A. Borrelia valaisianain cerebrospinal fluid. Emerg Infect Dis 2004;10:1692e3.

A rigorous review of diagnostic criteria and treatment, Journal of