Next Article in Journal
First Case of Typhoid Fever due to Extensively Drug-resistant Salmonella enterica serovar Typhi in Italy
Next Article in Special Issue
Upscaling the Surveillance of Tick-Borne Pathogens in the French Caribbean Islands
Previous Article in Journal
Methylobacterium spp. as Emerging Opportunistic Premise Plumbing Pathogens
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Evaluation of Disease Causality of Rare Ixodes ricinus-Borne Infections in Europe

1
Centre for Infectious Diseases Research, National Institute for Public Health and the Environment, P.O. Box 1, Bilthoven 3720 BA, The Netherlands
2
Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers Location Academic Medical Center, Amsterdam 1105 AZ, The Netherlands
*
Author to whom correspondence should be addressed.
These two authors contributed equally.
Pathogens 2020, 9(2), 150; https://doi.org/10.3390/pathogens9020150
Submission received: 13 February 2020 / Revised: 21 February 2020 / Accepted: 21 February 2020 / Published: 24 February 2020
(This article belongs to the Collection Advances in Tick Research)

Abstract

:
In Europe, Ixodes ricinus ticks transmit pathogens such as Borrelia burgdorferi sensu lato and tick-borne encephalitis virus (TBEV). In addition, there is evidence for transmission to humans from I. ricinus of Anaplasma phagocytophilum, Babesia divergens, Babesia microti, Babesia venatorum, Borrelia miyamotoi, Neoehrlichia mikurensis, Rickettsia helvetica and Rickettsia monacensis. However, whether infection with these potential tick-borne pathogens results in human disease has not been fully demonstrated for all of these tick-borne microorganisms. To evaluate the available evidence for a causative relation between infection and disease, the current study analyses European case reports published from 2008 to 2018, supplemented with information derived from epidemiological and experimental studies. The evidence for human disease causality in Europe found in this review appeared to be strongest for A. phagocytophilum and B. divergens. Nonetheless, some knowledge gaps still exist. Importantly, comprehensive evidence for pathogenicity is lacking for the remaining tick-borne microorganisms. Such evidence could be gathered best through prospective studies, for example, studies enrolling patients with a fever after a tick bite, the development of specific new serological tools, isolation of these microorganisms from ticks and patients and propagation in vitro, and through experimental studies.

1. Introduction

Lyme borreliosis and tick-borne encephalitis (TBE) are caused by infections with Borrelia burgdorferi sensu lato and tick-borne encephalitis virus (TBEV), respectively. These well-established tick-borne diseases are frequently reported in Europe, and several studies have indicated a rise in the incidence of these diseases over the last decades [1,2,3]. In Europe, both pathogens are transmitted to humans by the bite of Ixodes ricinus. During such a bite, humans might also be exposed and sometimes infected with other microorganisms transmitted by I. ricinus [4]. These potential tick-borne pathogens (TBPs) include Anaplasma phagocytophilum, Babesia divergens, Babesia microti, Babesia venatorum, Borrelia miyamotoi, Neoehrlichia mikurensis, Rickettsia helvetica, and Rickettsia monacensis, which have all been associated with human disease in Europe in case reports [5]. Although the number of case reports describing European human infections and disease associated with these agents is accumulating, the evidence for causality of infection with these microorganisms and human disease has not been fully demonstrated. Whenever a tick-borne pathogen is transmitted to a human host, the disease will not always ensue [6], because detrimental health effects are but one outcome of the interaction between a host and a microorganism [7]. Thus, the term pathogen points to a microorganism having the potential to cause disease following transmission [8].
Some of the I. ricinus-borne microorganisms are closely or more distantly related to species that are established disease agents in other tick species or continents. For example, R. helvetica, which is frequently encountered in I. ricinus throughout Europe, is related to other Spotted Fever Group Rickettsia, whose pathogenicity has been undisputedly shown. These include Rickettsia rickettsii, the causative agent of Rocky Mountain spotted fever (RMSF), a disease with a typical clinical picture, including a characteristic macular rash [9,10]. Epidemiological studies associate RMSF to Dermacentor ticks. Indeed, R. rickettsii could be successfully cultured from ticks, but also from patients [11,12]. Furthermore, infected ticks were able to transmit R. rickettsii when experimentally fed on animals [13,14], and R. rickettsii was found in endothelial cells of vascular lesions in acute cases [12,15]. In addition, Mediterranean spotted fever (MSF) is caused by Rickettsia conorii and also presents itself with a typical clinical picture, namely an eschar at the site of the tick-bite [12,16]. Rhipicephalus sanguineus ticks were identified as the main vector of R. conorii, which could be isolated and cultured both from patients and ticks [17,18,19]. Later, the ability of R. conorii to cause endothelial damage was proven in vivo [17,20,21]. In contrast, although R. helvetica has been isolated and cultivated from I. ricinus and a human patient [22], a typical clinical picture and consistent pathology of patients with a R. helvetica infection are lacking, as well as epidemiological evidence of association with disease [23,24,25].
A. phagocytophilum was established as the etiologic agent of Human Granulocytic Anaplasmosis (HGA) in the United States of America (USA) [26,27,28]. Patients lack characteristic symptoms, but develop acute fever, malaise, headache, myalgia, and/or arthralgia, often with a recent history of a tick-bite. HGA was initially misdiagnosed as Human Monocytotropic Ehrlichiosis (HME), a disease caused by Ehrlichia chaffeensis and transmitted by Amblyomma americanum [29,30]. A. phagocytophilum has successfully been cultivated from patient blood and Ixodes scapularis ticks [29,31,32] and transmission has been demonstrated in experimental settings [27,33]. Furthermore, A. phagocytophilum infects granulocytes, inducing leukocytopenia, often accompanied, in humans as well as in experimental animal models, by thrombocytopenia and elevated liver enzymes, e.g., Alanine aminotransferase (ALT) or Aspartate transaminase (AST) [27]. Epidemiological studies in the USA, where HGA is notifiable, show a steadily rising incidence [34]. In contrast to the USA, HGA is not notifiable in European countries, and the number of described cases is relatively low. The differences in the number of identified cases of HGA in the USA and Europe could be caused by limited awareness, but also by differences in vector, host susceptibility, and importantly, the pathogenicity of circulating A. phagocytophilum strains.
As long as the ability to cause disease by the European genospecies of potential TBPs has not been firmly established, it is difficult to assess to what extent public health professionals should increase awareness amongst the public and medical professionals and implement routine diagnostic modalities for these microorganisms. However, these public health actions are important prerequisites to be able to adequately assess the severity and incidence of I. ricinus-borne infectious diseases other than Lyme Borreliosis and TBE in Europe [35].
The nature of these I. ricinus-borne agents makes it difficult, if not impossible, to meet Koch’s postulates, as has previously been discussed [35,36,37]. In order to evaluate their involvement in human disease, several pitfalls must be overcome. Firstly, the clinical symptoms, as well as observed abnormalities in blood, attributed to infections with tick-borne microorganisms other than B. burgdorferi s.l. and TBEV, are non-characteristic and relatively mild in immunocompetent individuals. Without knowledge of a tick-bite, such a clinical picture could be attributed to any common infectious disease agent, e.g., influenza virus. Secondly, the isolation and propagation of microorganisms from patients is an important prerequisite for the establishment of their pathogenicity. Unfortunately, most of these tick-borne microorganisms are difficult to cultivate, even in dedicated laboratories [38]. For example, the cultivation and in vitro propagation of B. miyamotoi [39] and the cultivation of N. mikurensis has only been described in the literature recently [40,41]. Although serological or PCR-based methods have been shown to be useful as diagnostic tools for infectious diseases, they provide only limited evidence for (active) infection [42,43,44]. Thirdly, I. ricinus is frequently infected with more than one microorganism, enabling human exposure and possibly infection with multiple microorganisms, referred to as co-infections, after a singular or consecutive tick-bite. This could confuse the identification of the causative agent of the disease. For example, erythema migrans (EM), a typical manifestation of Lyme borreliosis, has been reported in patients in which infection with N. mikurensis or B. miyamotoi was detected by molecular methods [45,46]. This does not mean that these microorganisms are able to cause EM; it is much more likely that the EM is caused by an infection with B. burgdorferi s.l. and that the other microorganisms were present in the blood, either giving rise to other symptoms or indolent. Furthermore, the detection of R. helvetica and R. monacensis DNA and antibodies in patients with or suspected to have Lyme neuroborreliosis is a puzzling phenomenon and it remains to be elucidated whether coinfection with these spotted fever Rickettsiae has any clinical implications at all [24,25]. Fourthly, a final level of complexity is that some of these other I. ricinus-borne microorganisms might be able to cause disease only under specific conditions, i.e., when a patient is immunocompromised or has a co-infection.
Therefore, the aim of this literature study was to evaluate the published European case reports on tick-borne diseases—other than Lyme Borreliosis or TBE—in order to identify which clinical symptoms are associated with probable infections. This information, as well as data from epidemiological and experimental studies, was used to assess whether there is evidence for a causative relation between infection with these tick-borne microorganisms and human disease in Europe. The literature search was limited to the decade between 01-01-2008 and 01-03-2018, in order to assess the current situation. Our study focused only on microorganisms found in the European I. ricinus ticks and molecularly proven to be transmitted to their human host after a tick-bite, and not on microorganisms found in I. scapularis or I. persulcatus [6].

2. Results

2.1. Literature Search

Our literature search resulted in 3366 unique titles, out of which 3303 did not meet our search criteria based on the abstracts (Table S1), thus yielding 63 papers. In addition to the literature search, we manually included four additional case studies as part of a structural search. These, in total, 67 remaining papers were screened by reading the full text and 60 published case studies were found relevant for this study (Figure 1). These 60 European case studies, from between January 2008 and March 2018, described 77 individual cases. A. phagocytophilum case reports were most common, accounting for 23 of the papers and 32 cases. This was followed by 20 Babesia case reports describing 21 cases, of which 11 were associated with B. divergens, while seven and three were suspected of B. microti or B. venatorum infection, respectively. A total of 8 publications described 14 N. mikurensis cases, while 6 publications described 3 and 4 cases suspected of R. helvetica or R. monacensis infection, respectively. Finally, three B. miyamotoi cases were described in three publications. Infection with the various I. ricinus borne microorganisms was confirmed by molecular biology methods such as PCR, serology, and microscopy (Table 1).

2.2. Extracted Data

2.2.1. Anaplasma Phagocytophilum

Clinical Presentation

The most common symptoms reported in the 32 European cases of anaplasmosis analyzed [47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69] were fever, headache, malaise, and myalgia (41%–91%). Arthralgia and erythematous skin manifestations such as a maculopapular rash were less common (25% and 22%, respectively), and fewer cases presented with lymphadenopathy, chills, rigors, abdominal complaints, anemia, splenomegaly, and darkened urine (3%–19%) (Table 2). General laboratory findings included elevated C-reactive protein (CRP) values, leukocytopenia, thrombocytopenia, elevated liver enzymes and lactate dehydrogenase (LDH) values (Suppl. data). One patient was immunocompromised, and hospitalization was required in 88% of cases. Most patients (94%) received antibiotic treatment mainly in the form of doxycycline and subsequently recovered. In two cases, the outcome was fatal.

Exposure to Ticks

Eleven (34%) patients recalled a tick-bite, and 12 (38%) of the cases reported exposure to ticks.

Culture from Ticks or Patient Materials

Although A. phagocytophilum was not cultured in any of the case reports analyzed in this study, culture in a HL-60 promyelocytic cell line from a European patient has been successfully performed in the past in the Czech Republic [70].

Pathology

Both the European and American variants of A. phagocytophilum have been shown to infect human and animal host reservoir neutrophils, hence the name Human Granulocytic Anaplasmosis (HGA) [42,71,72]. The disruptive effect on infected neutrophils may lead to detrimental effects on the host’s immune response and poor regulation of inflammatory processes. This is believed to give rise to secondary pathological findings such as tissue damage in the spleen, liver, and kidneys [71]. In farm animals, such as sheep and cattle, the severity of the symptoms was affected by factors such as the host immune status or pre-existing infections [73]. In the cases evaluated in this study, the primary damage to neutrophils and secondary organ damage can be deducted from the reported laboratory test results such as leukocytopenia and elevated liver enzymes, respectively (Table 2). Moreover, bacteria were visualized in 25% of cases in Giemsa stained blood or bone marrow smears.

Transmission Experiments

Transmission of a Dutch strain of A. phagocytophilum from infected I. ricinus to naïve dogs has been shown, as well as the acquisition of the bacteria by I. ricinus in vitro [74].Moreover, a longitudinal study on sheep-to-tick transmission efficiency under natural conditions found molecular evidence of I. ricinus infected with A. phagocytophilum after feeding on their sheep host [75]. Moreover, the bacterium has been detected in I. ricinus ticks as well as animal reservoirs in multiple European countries [76,77,78,79]. Moreover, human tick-bite studies have shown an association between a tick-bite by I. ricinus ticks and the presence of A. phagocytophilum in patient blood. For example, a study in which 626 blood samples of Dutch patients who reported a tick-bite, were analyzed for the presence of tick-borne microorganisms found five samples positive for A. phagocytophilum by molecular screening [6]. Another study in Poland, in which whole blood, serum, and cerebrospinal fluid (CSF) samples from 118 symptomatic patients who reported a tick-bite were screened for tick-borne microorganisms, reported molecular detection of A. phagocytophilum in 14 of the samples [80]. These data support the assumption that this microorganism can be transmitted from I. ricinus to both animals and humans.

2.2.2. Babesia Species

Clinical Presentation

In the seven cases where B. microti was reported as the probable cause of disease [81,82,83,84,85,86,87], fever was reported for all patients, anemia in all except one, and also malaise, chills, headache, and abdominal complaints were relatively common (ranging from 57% to 43%). Skin manifestations were reported only in one case (Table 2). General laboratory findings included elevated CRP, thrombocytopenia, elevated liver enzymes, bilirubin, and LDH (Suppl. Data). Hospitalization was required in 86% of the cases. No fatal cases were described between 2008 and 2018. All patients received treatment with a combination of antibiotics and antiparasitic medication such as azithromycin-atovaquone or proguanil and quinine. It is important to note that infection with B. microti was acquired outside of Europe, namely North America and Uruguay, and solely diagnosed and/or treated in Europe.
In ten additional publications [88,89,90,91,92,93,94,95,96,97], 11 babesiosis cases were reported where infection with B. divergens was the probable cause of disease, and fever was the most common symptom (82%). Anemia, malaise, jaundice, abdominal complaints and darkened urine were all relatively common (ranging from 45% to 27%), while headache, myalgia and arthralgia were reported for only two patients (Table 2). General laboratory findings included elevated CRP values, leukocytopenia, thrombocytopenia, and elevated liver enzymes, bilirubin, LDH and kidney function (Table 1 and Suppl. data). Hospitalization was required in 91% of the 11 cases, 64% were immunocompromised, out of which 54% were splenectomized. Treatment was administered to 91% of patients. In one case, doxycycline was provided while most followed the guideline advised dual therapy with an antibiotic and antiparasitic medication. Of the nine cases that received combination-therapy, three were fatal, two of which were immunocompromised. Additionally, an immunocompromised patient without treatment died.
Finally, three cases were associated with B. venatorum infection [98,99,100]. Symptoms included anemia (100%), fever, darkened urine (67%), rigors and myalgia (33%). General laboratory findings included elevated CRP values, to a lesser extent thrombocytopenia, and elevated bilirubin, LDH, and kidney function. (Suppl. data). One case required hospitalization, and in all cases, treatment with quinine and clindamycin or azithromycin and atovaquone was followed by recovery. All three patients were immunocompromised and had undergone splenectomy.

Exposure to Ticks

In B. microti suspected cases, patients recalled a tick-bite in 14% of the cases, and exposure to ticks was reported in the remaining 86% of the cases. For cases where B. divergens infection was detected, patients recalled a tick-bite in 45% of cases and exposure to ticks was reported in the other 55% of cases. Exposure to ticks was reported in 33% of the B. venatorum suspected cases.

Culture from Ticks or Patient Materials

None of the Babesia species were cultured in the case reports analyzed. However, B. divergens has been successfully cultured, propagated, and sequenced from samples deriving from a French patient and it has also been cultivated from European cattle [101,102,103]. B. microti has been isolated from field voles in Germany, and B. venatorum has been cultured from roe deer erythrocytes in France [104,105].

Pathology

Babesia spp. are intraerythrocytic parasites, and symptoms such as anemia, jaundice, and hemoglobinuria (presenting as darkened urine) are a result of hemolysis [106].
In the cases evaluated for B. microti, B. divergens, and B. venatorum, intra-erythrocytic parasites could be visualized in Giemsa stained blood smears of 91%, 86%, and 33% of patients, respectively. Parameters linked to hemolysis, such as anemia and elevated bilirubin, were also reported. Thrombocytopenia was common in B. microti (86%) and B. divergens (73%) cases and less frequent in B. venatorum (33%) cases.

Transmission Experiments

A European strain of B. microti was transmitted in vivo from I. ricinus to gerbils and vice versa [107]. B. divergens was transmitted to I. ricinus ticks through feeding on inoculated gerbils and cattle [108]. Moreover, B. venatorum was transmitted to I. ricinus ticks fed on infected sheep blood and vice versa [109]. These findings demonstrate that I. ricinus is a competent vector for all three Babesia species.

2.2.3. Borrelia miyamotoi

Clinical Presentation

Three European cases with suspected B. miyamotoi disease (BMD) were included in this study [110,111,112], of which two patients were immunocompromised. Symptoms reported included neurological complaints in the two immunocompromised cases due to meningoencephalitis and headache (67%). Fever, abdominal complaints, myalgia, arthralgia, weight loss, and lymphadenopathy were observed in one case (33%). The two immunocompromised patients received antibiotic treatment (ceftriaxone) and recovered. The remaining patient recovered without treatment and showed elevated CRP values, leukocytopenia, and thrombocytopenia.

Exposure to Ticks

Patients recalled tick-bites in all three cases.

Culture from Ticks or Patient Materials

B. miyamotoi was isolated and cultivated in modified Kelly–Pettenkofer medium supplemented with 10% fetal bovine serum from I. ricinus egg masses resulting in two Dutch isolates, of which one was propagated and both were sequenced [113].

Pathology

As opposed to the hematogenous disease commonly seen in immunocompetent cases, in immunocompromised patients, BMD has been reported to manifest neurologically (e.g., meningoencephalitis) [114]. However, recently (after the study period of this review) two new neurological BMD cases have been diagnosed in Sweden, of which one case was immunocompetent [115]. Neurological involvement is demonstrated in the CSF by pleocytosis, visible spirochetes by dark-field microscopy and by molecular analysis specific for relapsing fever Borrelia and Borrelia miyamotoi [115]. Pleocytosis and visible spirochetes in the CSF by dark-field microscopy were also reported in the Dutch immunodeficient case included in this study [110,114].

Transmission Experiments

A Dutch tick isolate of B. miyamotoi has been shown to be infective to I. ricinus fed on infected CD1 mice and vice versa (unpublished) [116]. Moreover, field-collected B. miyamotoi infected I. ricinus larvae have been shown to transmit the spirochete to naive rodents [117]. This suggests I. ricinus is a competent vector for B. miyamotoi transmission.

2.2.4. Neoehrlichia mikurensis

Clinical Presentation

The most prevalent symptom in the 14 cases of neoehrlichiosis included [46,118,119,120,121,122,123] fever (86%), less common were skin manifestations (erythematous rash and EM) and anemia (36% and 29%, respectively), and in fewer cases, symptoms such as malaise, chills, arthralgia, abdominal complaints, edema, myalgia, and weight loss were reported (ranging from 14% to 7%). General laboratory findings included elevated CRP values, leukocytopenia, thrombocytopenia, and elevated liver enzymes (Suppl. data). Hospitalization was required for 79% of the patients. All patients received treatment mainly in the form of doxycycline. Two cases received phenoxymethylpenicillin, and one case received piperacillin-tazobactam, ciprofloxacin, and fosfomycin. All cases but the latter recovered. The majority of cases (n = 10; 71%) were immunocompromised, and five of these (50%) had undergone splenectomy.

Exposure to Ticks

Five (36%) patients recalled a tick-bite and two (14%) of the cases reported exposure to ticks.

Culture from Ticks or Patient Materials

Only recently, N. mikurensis was reported to be cultivable in human primary endothelial cells derived from skin microvasculature as well as pulmonary arteries and in the I. ricinus IRE/CTVM20 cell line. Cultivation was performed using PCR-positive blood from Swedish patients [41].

Pathology

The pathology of neoehrlichiosis is not yet fully understood. As N. mikurensis was isolated and propagated in human primary endothelial cells and was visualized in the cytoplasm of circulating endothelial cells of infected patients, it has been suggested that endothelial cells are the infectious target for the bacteria [41]. There is evidence in reported cases for vascular events, such as deep vein thrombosis, thromboembolic events, and transitory ischemic accidents [124]. Moreover, two of the cases in this study presented with arterial aneurysms [119,122].

Transmission Experiments

N. mikurensis has been widely molecularly detected in I. ricinus in multiple European countries, such as Belgium, France, Germany, The Czech Republic, Portugal, Poland, The Netherlands, and Sweden [125,126,127,128]. Moreover, the bacterium has been isolated from xenodiagnostic I. ricinus larvae fed on infected rodents, further implicating this tick species serves as a vector for the bacteria [129]. As far as we are aware, no studies have been published on tick transmission in animal models.

2.2.5. Spotted Fever Rickettsiae

Clinical Presentation

Three R. helvetica case reports were included [22,130,131]. All these cases developed fever and headache. Accompanying photophobia was common (67%), while skin manifestations (such as a macular rash), myalgia and arthralgia, were presented in only one case. Laboratory findings included elevated CRP values, elevated erythrocyte sedimentation rate (ESR), leukocytopenia, thrombocytopenia, and elevated liver enzymes and kidney function (Suppl. data). The three cases required hospitalization and recovered following antibiotic treatment with doxycycline.
In the four cases where infection with R. monacensis was reported as the probable cause of disease [132,133,134], skin manifestations (such as erythematous rash and EM) were reported. One of the cases, in which the EM was reported, was found to be co-infected with B. afzelii. Fever and headaches were common symptoms (75%) and anemia was reported in one patient. Laboratory findings included leukocytopenia, as well as elevated ESR and bilirubin (Suppl. data). One patient required hospitalization, but all patients recovered following antibiotic treatment with doxycycline and in one case with azithromycin.

Exposure to Ticks

No tick-bites were reported in the R. helvetica cases, however, all patients had been exposed to ticks. Of the R. monacensis cases 50% reported a tick-bite and 25% were exposed to ticks.

Culture from Ticks or Patient Materials

R. helvetica has been cultivated in African green monkey kidney fibroblast cells (VERO) from CSF of two Swedish cases included in this study [22,131], of which one was also infected by Herpes simplex virus. The bacterium has also been successfully cultured and sequenced from questing I. ricinus ticks in VERO cells [135,136]. R. monacensis was isolated from a patient’s blood in Spain and cultivated in VERO cells [132]. Moreover, isolation of R. monacensis from I. ricinus ticks in VERO, mouse fibroblast (L-929), and tick (IRE11, DAE100, ISE6) cell lines have been reported [137].

Pathology

R. helvetica DNA has been detected by using a semi-nested PCR of the 16S rRNA and 17-kDa outer-membrane protein genes, in the pericardium and in a lymph node from the pulmonary hilum in one patient and in a coronary artery and the heart muscle of another, in two Swedish cases of sudden cardiac death postulated to be due to a R. helvetica infection. The bacteria were also visualized in macrophages and endothelial cells using Gimenez and May-Grünwald-Giemsa (MGG) staining, wherein their structure was confirmed by transmission electron microscopy [138]. R. helvetica has also been visualized in granulomatous endothelial tissue and macrophages from Swedish patients with sarcoidosis, even though an association with the disease was not established [139]. Additionally, R. helvetica DNA has been detected in CSF of patients in The Netherlands and Denmark [24,140]. In two Danish cases, R. helvetica was cultured from CSF, leukocytopenia was observed, and a macular rash was reported for one of the patients, supporting the findings above. The pathology of R. monacensis has not been studied, but the rashes described in the case reports analyzed, suggest endothelial tissue tropism.

Transmission Experiments

Both R. helvetica and R. monacensis have been repeatedly detected by molecular means both from I. ricinus ticks and, to a lesser extent, from wildlife in various European countries [137,141,142,143,144,145,146,147,148,149]. As far as we are aware, there are no published transmission experiments in laboratory animals for either species.

3. Discussion, Conclusions and Knowledge Gaps

Since Koch’s postulates are not applicable to all potential disease agents, alternatively, the sum of various alternative approaches could provide sufficient evidence to support causality between a microorganism and human disease, in this case, a tick-borne disease. Such has been the succession of events for several established tick-borne pathogens, for example, for B. burgdorferi s.l. and TBEV. Case reports can provide extensive qualitative, but not quantitative data, thus showing circumstantial, but insufficient scientific evidence for disease causality. The cases presented are usually exceptional ones, such as immunocompromised patients, and because the studies are retrospective in nature, the reports suffer from reporter bias. As part of the current study, by supplementing the data from case reports on A. phagocytophilum, B. venatorum, B. divergens, B. microti, B. miyamotoi, N. mikurensis, R. helvetica, and R. monacensis with epidemiological and experimental studies, we have assessed their causal relationship to human disease. Thus, we found that to varying degrees, the existing evidence is incomplete for all the microorganisms mentioned above, but seemed strongest for A. phagocytophilum and B. divergens. This approach also provided a comprehensive outlook on the current knowledge gaps that need to be filled (Table 3).
A. phagocytophilum causes illness in immunocompetent patients in Europe, and the case reports amounted to half of all papers included in this review. The clinical picture, laboratory test results, and low fatality rate were reminiscent of what has been described for HGA in the USA [26,43]. A rash was reported in a minority of patients, which is not considered to be a specific sign of HGA and may point to co-infections with other tick-borne bacteria, such as Rickettsia or Borrelia spp. [134,153,154,155], as was confirmed for one of these cases [56]. In Europe A. phagocytophilum has been cultivated from a patient’s blood and its tissue tropism, namely neutrophils, has been demonstrated [42,70]. A. phagocytophilum has not been isolated from I. ricinus. However, there is ample evidence of its association with I. ricinus ticks [76,77] and evidence that this tick species can be infected by natural reservoirs [75].
In accordance with published data [156,157], most cases reporting Babesia infection in Europe were attributed to B. divergens, and, while disease has been associated with immunocompromised and splenectomized patients [157], almost half of the cases were immunocompetent. Four cases were fatal, and three of these were immunocompromised and one was immunocompetent and had a co-existing Lyme borreliosis infection [90]. Co-infections with Babesia are a known possibility in Lyme borreliosis patients and should be considered in cases from endemic regions presenting with high fever, anemia, thrombocytopenia, leukocytopenia, or other hemolysis signs, or in cases who do not respond to treatment [158]. Co-infection of B. microti and B. burgdorferi s.l. has been found to increase both the severity and duration of illness in North America [153,159,160], such correlations have not been shown for co-infection with B. divergens and B. burgdorferi s.l. Our analyses also suggested that B. microti is able to cause disease in Europe, but mainly as an imported agent presenting as a non-fatal illness in immunocompetent patients. Although it is present in I. ricinus and European wildlife and has been detected in human samples from Germany, Poland, and Switzerland [157], the number and severity of B. microti cases, be it from lack of awareness or genetic difference, is low when compared to North America [157]. In the USA, 2358 cases were reported in 2017 and fatality rates of up to 6%–9% have been reported in immunocompetent patients [156,161]. The first European originated case, probably acquired by a blood transfusion, was published in 2007 [162], however, the protozoa have not been cultivated from European patients or I. ricinus. Cultivation of European strains would facilitate genomic analyses in the search for differences in pathogenicity when compared to North American strains. The few B. venatorum cases reported were relatively mild and affected immunocompromised patients. Babesia venatorum can be transmitted by I. ricinus in laboratory settings [109]. However, cultivation from ticks and patients remains to be performed and would also assist the development of specific serological tests for diagnostic purposes and future population studies.
Borrelia miyamotoi is an emerging tick-borne microorganism whose potential to cause disease has only recently been disclosed [114]. Since the first case reports in 2011 in Russia, over 450 cases have been published, mainly from Asia and North America, with a total of 5 cases from Europe. B. miyamotoi is able to cause an acute febrile disease with flu-like symptoms. Skin lesions, such as EM, have been reported in B. miyamotoi suspected cases, but are presumed to be related to a co-infection with the etiologic agent of Lyme disease, B. burgdorferi s.l. In immunocompromised cases, of which most originate from Europe, the disease seems to develop neurologically, however, a meningoencephalitis in an immunocompetent case has been reported. Nevertheless, more European cases need to be evaluated to assess the full extent of the clinical symptoms and effectiveness of the available diagnostic modalities. B. miyamotoi has not been cultivated from an individual infected by an I. ricinus tick-bite. However, it has been cultivated from 12 Russian patients, some from European Russia, of which 6 were sequenced and published [163], with I. persulcatus being the most likely vector. Cultivation of more B. miyamotoi isolates from I. ricinus and European human cases would enable comparative genomic analyses between isolates from Europe, North America, and Asia [164]. This could be an enormous impetus for basic, molecular, and epidemiological research and could lead to the development of specific diagnostic tools for this emerging potential TBP.
Another emerging potential TBP is N. mikurensis [124]. The findings of this study support the growing evidence of N. mikurensis as a disease agent affecting the immunocompromised population, and show it is also found in immunocompetent patients. EM was reported in two cases where co-infection with A. phagocytophilum in one and B. burgdorferi s.l. in the other was present [46]. A Norwegian study, in which EM patients were screened by PCR for the presence of N. mikurensis, reported seven positive samples, of which only three were positive for B. burgdorferi s.l. by serology [165]. However, this could be explained by the low diagnostic accuracy of serology in early Lyme borreliosis [166]. Three other cases analyzed in this study, of immunocompromised patients presenting with a rash, were diagnosed by a positive N. mikurensis PCR, and in two cases by transmission electron micrographs. However, no serology was performed in order to rule out a co-infection that could have caused the rash [120,121]. Although Ehrlichia species are known to infect granulocytes or mononuclear phagocytes [167], the bacterium has been cultivated from patient’s blood in a tick cell line and in human primary endothelial cells [41]. Routine cultivation, as well as cultivation from ticks would enable development of the serological assays, which are currently lacking from the routine practice.
Consistent with it being a spotted fever group Rickettsia, three R. monacensis cases presented with a rash, and in one case in which an EM was present, a co-infection with B. afzelii was detected [134]. However, no studies have been performed to elucidate the specific pathology of this microorganism. The clinical picture reported for R. helvetica in this study included headaches and photophobia, which is suggestive of neurological involvement. One of the cases presented with subacute meningitis [22] and although the diagnosis of neuroborreliosis was rejected based on serology, the test was performed in an early disease stage when test sensitivity infamously falls short. Another case presented with meningoencephalitis [131] and was positive for Herpes Simplex Virus 2, which could account for the clinical picture. In the three case reports included in the study R. helvetica was either isolated and cultivated or isolated, accompanied by seroconversion indicating that this microorganism can cause infection in humans. However, proof found of a Rickettsia infection does not imply or translate to symptomatic disease. Therefore the evidence should not be mistaken for disease causation, which has not been demonstrated [22,131]. Interestingly, R. helvetica has been detected as a co-infection with Lyme neuroborreliosis both in patient blood and CSF in a Dutch study [24]. R. helvetica has also been found as a co-infection in Danish Lyme borreliosis and neuroborreliosis patients [140], however the involvement of this bacterium with neurological symptoms has not been studied. Although both R. helvetica and R. monacensis have been isolated from patients and I. ricinus ticks, only few case reports have been published on cases caused by these species and no transmission experiments have been performed to date, in contrast to, for example, R. conorii, an established human pathogen in Europe.
In all case reports analyzed in this study, diagnostic tools such as microscopy, serology, and molecular analysis were required in order to reach a diagnosis. In some cases, these tools were employed retrospectively, as they were not accessible in routine practice. Moreover, the cultivation of microorganisms such as A. phagocytophilum, N. mikurensis, and Babesia spp. was not attempted as their growth requires dedicated settings, is technically demanding, and time-consuming. Even in experimental settings, cultivation of these microorganisms can be arduous. To date, cultivation from I. ricinus has been reported only for B. miyamotoi, R. helvetica, and R. monacensis [113,135,137] and cultivation from I. ricinus acquired cases has not been performed for B. venatorum, B. microti, and B. miyamotoi.
The additional evidence needed in order to conclusively appraise disease causality and elucidate the impact of I. ricinus transmitted infections in Europe for these tick-borne microorganisms can be achieved by the fulfillment of several steps. Firstly, to enhance our understanding of the tissue tropism and specific pathology of microorganisms, such as R. helvetica, R. monacensis, and N. mikurensis, experimental transmission experiments using laboratory animals should be performed. Secondly, new and specific serological tools should be developed for the diagnosis of I. ricinus-borne microorganisms. This could be facilitated by cultivation of the tick-borne microorganisms from patients or ticks, followed by next-generation sequencing and bioinformatic analyses, for the discovery of new potential antigens. Thirdly, prospective studies need to be performed including patients with febrile illness after a tick-bite, the common denominator in the clinical manifestation of the different tick-borne microorganisms.

4. Materials and Methods

4.1. Literature Search

The literature search was performed using the Pubmed and Embase databases (Table S1). In addition, a structural search was performed in order to find additional cases and information.

4.2. Selection Criteria

The search focused on suspected disease cases with Anaplasma phagocytophilum, Babesia divergens, Babesia microti, Babesia venatorum, Borrelia miyamotoi, Neoehrlichia mikurensis, Rickettsia helvetica, and Rickettsia monacensis. Case reports and series of patients diagnosed in Europe, published in the period between 01-01-2008 and 01-03-2018, were included. The process was performed independently by TA and DH, and when disagreements occurred, the study was re-evaluated by both TA and DH under the supervision of HS, KK, and JH. Publications written in English, Dutch, and Spanish were included.

4.3. Data Extraction

Parameters extracted for all case reports/series were: Sex, age, immune status, underlying health conditions, pre-existing treatments, history of a tick-bite or exposure to ticks, clinical symptoms, general laboratory findings, hospitalization, treatment, disease outcome, diagnostic evaluation, excluded causes of disease and co-infections (Table S2–S9). Within clinical symptoms, “fever” was attributed to a case whenever the measured temperature exceeded ≥ 37.5 ℃ (99,5 oF) [168] or when the authors reported fever as a symptom. History of a tick-bite was recorded whenever a patient recalled a tick bite on presentation. Exposure to ticks was recorded whenever a patient worked in a high-risk occupation, e.g., forest ranger, had engaged in activities considered to be high risk for a tick bite such as hiking, or had been exposed to animals.

4.4. Data Analysis

For each tick-borne microorganism, cases were evaluated for evidence of a causal relationship between the microorganism and the disease described by establishing the nature and frequency of five criteria: The clinical presentation, association to tick-bite(s), culture from ticks or patient materials, transmission experiments, and the ability to induce specific pathology. In addition, for the analyses on cultivation from ticks or patients, transmission experiments and pathology, information was added by selectively searching the literature.

Supplementary Materials

The following are available online at https://www.mdpi.com/2076-0817/9/2/150/s1, Table S1: S1_search criteria, Table S2: S2_A. phagocytophilum, Table S3: S3_B. divergens, Table S4: S4_B. microti, Table S5: S5_B. venatorum, Table S6: S6_B. miyamotoi, Table S7: S7_N. mikurensis, Table S8: S8_R. helvetica, Table S9: S9_R. monacensis.

Author Contributions

Conceptualization, T.A. and H.S.; data curation, T.A. and D.H.; data analysis, T.A.; writing—original draft preparation, TA; writing—review and editing, T.A., D.H., KK, J.W.R.H, and H.S.; supervision, H.S. and J.W.R.H.; project administration, H.S. and J.W.R.H.. All authors have read and agreed to the published version of the manuscript.

Funding

T.A., D.H., J.W.R.H., and H.S. were supported by a grant from the ZonMw (project number 50-52200-98-313, Ticking on Pandora’s box) and a grant from the European Interreg North Sea Region program, as part of the NorthTick project. KK is supported by the Dutch Ministry of Health, Welfare, and Sports. The funders had no role in the study design and interpretation, or the decision to submit the work for publication.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hofhuis, A.; Harms, M.; van den Wijngaard, C.; Sprong, H.; van Pelt, W. Continuing increase of tick bites and Lyme disease between 1994 and 2009. Ticks Tick Borne Dis. 2015, 6, 69–74. [Google Scholar] [CrossRef]
  2. Jaenson, T.G.; Hjertqvist, M.; Bergstrom, T.; Lundkvist, A. Why is tick-borne encephalitis increasing? A review of the key factors causing the increasing incidence of human TBE in Sweden. Parasit. Vectors 2012, 5, 184. [Google Scholar] [CrossRef] [Green Version]
  3. Kunze, U. The International Scientific Working Group on Tick-Borne Encephalitis (ISW TBE): Review of 17 years of activity and commitment. Ticks Tick Borne Dis. 2016, 7, 399–404. [Google Scholar] [CrossRef]
  4. Sprong, H.; Azagi, T.; Hoornstra, D.; Nijhof, A.M.; Knorr, S.; Baarsma, M.E.; Hovius, J.W. Control of Lyme borreliosis and other Ixodes ricinus-borne diseases. Parasit. Vectors 2018, 11, 145. [Google Scholar] [CrossRef] [Green Version]
  5. Rizzoli, A.; Silaghi, C.; Obiegala, A.; Rudolf, I.; Hubalek, Z.; Foldvari, G.; Plantard, O.; Vayssier-Taussat, M.; Bonnet, S.; Spitalska, E.; et al. Ixodes ricinus and Its Transmitted Pathogens in Urban and Peri-Urban Areas in Europe: New Hazards and Relevance for Public Health. Front. Public Health 2014, 2, 251. [Google Scholar] [CrossRef]
  6. Jahfari, S.; Hofhuis, A.; Fonville, M.; van der Giessen, J.; van Pelt, W.; Sprong, H. Molecular Detection of Tick-Borne Pathogens in Humans with Tick Bites and Erythema Migrans, in the Netherlands. PLoS Negl. Trop. Dis. 2016, 10, e0005042. [Google Scholar] [CrossRef] [Green Version]
  7. Casadevall, A.; Pirofski, L.A. Microbiology: Ditch the term pathogen. Nature 2014, 516, 165–166. [Google Scholar] [CrossRef]
  8. Pirofski, L.A.; Casadevall, A. Q and A: What is a pathogen? A question that begs the point. BMC Biol. 2012, 10, 6. [Google Scholar] [CrossRef] [Green Version]
  9. Portillo, A.; de Sousa, R.; Santibanez, S.; Duarte, A.; Edouard, S.; Fonseca, I.P.; Marques, C.; Novakova, M.; Palomar, A.M.; Santos, M.; et al. Guidelines for the Detection of Rickettsia spp. Vector Borne Zoonotic Dis. 2017, 17, 23–32. [Google Scholar] [CrossRef] [Green Version]
  10. Abdad, M.Y.; Abou Abdallah, R.; Fournier, P.E.; Stenos, J.; Vasoo, S. A Concise Review of the Epidemiology and Diagnostics of Rickettsioses: Rickettsia and Orientia spp. J. Clin. Microbiol. 2018, 56. [Google Scholar] [CrossRef] [Green Version]
  11. Wolbach, S.B.; Schlesinger, M.J. The cultivation of the micro-organisms of Rocky Mountain spotted fever (Dermocentroxenus rickettsi) and of typhus (Rickettsia prowazeki) in tissue plasma cultures. J. Med. 1923, 44, 231–256. [Google Scholar]
  12. Parola, P.; Paddock, C.D.; Raoult, D. Tick-borne rickettsioses around the world: Emerging diseases challenging old concepts. Clin. Microbiol. Rev. 2005, 18, 719–756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Wilson, L.B.; Chowning, W.M. Studies in Pyroplasmosis hominis (“spotted fever” or “tick fever” of the Rocky Mountains). J. Infect. Dis. 1904, 1, 31–57. [Google Scholar] [CrossRef] [Green Version]
  14. Wolbach, S.B. Studies on Rocky Mountain spotted fever. J. Med. Res. 1919, 41, 2–197. [Google Scholar]
  15. Silverman, D.J. Rickettsia rickettsii-induced cellular injury of human vascular endothelium in vitro. Infect. Immun. 1984, 44, 545–553. [Google Scholar] [CrossRef] [Green Version]
  16. Olmer, D. Sur une infection épidémique, avec exanthème de nature indéterminée. Mars. Med. 1925, 22, 1291–1293. [Google Scholar]
  17. Rovery, C.; Brouqui, P.; Raoult, D. Questions on Mediterranean spotted fever a century after its discovery. Emerg. Infect. Dis. 2008, 14, 1360–1367. [Google Scholar] [CrossRef]
  18. Marrero, M.; Raoult, D. Centrifugation-shell vial technique for rapid detection of Mediterranean spotted fever rickettsia in blood culture. Am. J. Trop. Med. Hyg. 1989, 40, 197–199. [Google Scholar] [CrossRef]
  19. Durand, P.; Conseil, E. Transmission expérimentale de la fièvre boutonneuse par Rhipicephalus sanguineus. C. R. Acad. Sci. 1930, 190, 1244. [Google Scholar]
  20. George, F.; Brouqui, P.; Boffa, M.C.; Mutin, M.; Drancourt, M.; Brisson, C.; Sampol, J. Demonstration of Rickettsia conorii-induced endothelial injury in vivo by measuring circulating endothelial cells, thrombomodulin, and von Willebrand factor in patients with Mediterranean spotted fever. Blood 1993, 82, 2109–2116. [Google Scholar] [CrossRef] [Green Version]
  21. Walker, D.H.; Occhino, C.; Tringali, G.R.; Di Rosa, S.; Mansueto, S. Pathogenesis of rickettsial eschars: The tache noire of boutonneuse fever. Hum. Pathol. 1988, 19, 1449–1454. [Google Scholar] [CrossRef]
  22. Nilsson, K.; Elfving, K.; Pahlson, C. Rickettsia helvetica in patient with meningitis, Sweden, 2006. Emerg. Infect. Dis. 2010, 16, 490–492. [Google Scholar] [CrossRef] [PubMed]
  23. Svendsen, C.B.; Milman, N.; Nielsen, H.W.; Krogfelt, K.A.; Larsen, K.R. A prospective study evaluating the presence of Rickettsia in Danish patients with sarcoidosis. Scand. J. Infect. Dis. 2009, 41, 745–752. [Google Scholar] [CrossRef] [PubMed]
  24. Koetsveld, J.; Tijsse-Klasen, E.; Herremans, T.; Hovius, J.W.; Sprong, H. Serological and molecular evidence for spotted fever group Rickettsia and Borrelia burgdorferi sensu lato co-infections in The Netherlands. Ticks Tick Borne Dis. 2016, 7, 371–377. [Google Scholar] [CrossRef] [PubMed]
  25. Ocias, L.F.; Dessau, R.B.; Lebech, A.M.; Jorgensen, C.S.; Petersen, R.F.; Krogfelt, K.A. Evidence of rickettsiae in Danish patients tested for Lyme neuroborreliosis: A retrospective study of archival samples. BMC Infect. Dis. 2018, 18, 325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Bakken, J.S.; Dumler, J.S. Clinical diagnosis and treatment of human granulocytotropic anaplasmosis. Ann. N. Y. Acad. Sci. 2006, 1078, 236–247. [Google Scholar] [CrossRef]
  27. Bakken, J.S.; Dumler, J.S. Human granulocytic anaplasmosis. Infect. Dis. Clin. N. Am. 2015, 29, 341–355. [Google Scholar] [CrossRef] [Green Version]
  28. Dumic, I.; Patel, J.; Hart, M.; Niendorf, E.R.; Martin, S.; Ramanan, P. Splenic Rupture as the First Manifestation of Babesia Microti Infection: Report of a Case and Review of Literature. Am. J. Case Rep. 2018, 19, 335–341. [Google Scholar] [CrossRef] [Green Version]
  29. Bakken, J.S. The discovery of human granulocytotropic ehrlichiosis. J. Lab. Clin. Med. 1998, 132, 175–180. [Google Scholar] [CrossRef]
  30. Chen, S.M.; Dumler, J.S.; Bakken, J.S.; Walker, D.H. Identification of a granulocytotropic Ehrlichia species as the etiologic agent of human disease. J. Clin. Microbiol. 1994, 32, 589–595. [Google Scholar] [CrossRef] [Green Version]
  31. Massung, R.F.; Levin, M.L.; Munderloh, U.G.; Silverman, D.J.; Lynch, M.J.; Kurtti, T.J. Isolation of Anaplasma phagocytophilum strain Ap-variant 1 in a tick-derived cell line. Ann. N. Y. Acad. Sci. 2006, 1078, 541–544. [Google Scholar] [CrossRef] [PubMed]
  32. Goodman, J.L.; Nelson, C.; Vitale, B.; Madigan, J.E.; Dumler, J.S.; Kurtti, T.J.; Munderloh, U.G. Direct cultivation of the causative agent of human granulocytic ehrlichiosis. N. Engl. J. Med. 1996, 334, 209–215. [Google Scholar] [CrossRef] [PubMed]
  33. Telford, S.R., 3rd; Dawson, J.E.; Katavolos, P.; Warner, C.K.; Kolbert, C.P.; Persing, D.H. Perpetuation of the agent of human granulocytic ehrlichiosis in a deer tick-rodent cycle. Proc. Natl. Acad. Sci. USA 1996, 93, 6209–6214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Epidemiology and Statistics. Available online: https://www.cdc.gov/anaplasmosis/stats/index.html (accessed on 1 January 2019).
  35. Tijsse-Klasen, E.; Koopmans, M.P.; Sprong, H. Tick-borne pathogen—Reversed and conventional discovery of disease. Front. Public Health 2014, 2, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Inglis, T.J. Principia aetiologica: Taking causality beyond Koch’s postulates. J. Med. Microbiol. 2007, 56, 1419–1422. [Google Scholar] [CrossRef]
  37. Falkow, S. Molecular Koch’s postulates applied to microbial pathogenicity. Rev. Infect. Dis. 1988, 10, S274–S276. [Google Scholar] [CrossRef] [Green Version]
  38. Raoult, D. Uncultured candidatus neoehrlichia mikurensis. Clin. Infect. Dis. 2014, 59, 1042. [Google Scholar] [CrossRef]
  39. Wagemakers, A.; Oei, A.; Fikrig, M.M.; Miellet, W.R.; Hovius, J.W. The relapsing fever spirochete Borrelia miyamotoi is cultivable in a modified Kelly-Pettenkofer medium, and is resistant to human complement. Parasit. Vectors 2014, 7, 418. [Google Scholar] [CrossRef] [Green Version]
  40. Wenneras, C.; Bloemberg, G.; Bogdan, C. Reply to Raoult. Clin. Infect. Dis. 2014, 59, 1042–1043. [Google Scholar] [CrossRef]
  41. Wass, L.; Grankvist, A.; Bell-Sakyi, L.; Bergstrom, M.; Ulfhammer, E.; Lingblom, C.; Wenneras, C. Cultivation of the causative agent of human neoehrlichiosis from clinical isolates identifies vascular endothelium as a target of infection. Emerg. Microbes Infect. 2019, 8, 413–425. [Google Scholar] [CrossRef] [Green Version]
  42. Brouqui, P.; Bacellar, F.; Baranton, G.; Birtles, R.J.; Bjoersdorff, A.; Blanco, J.R.; Caruso, G.; Cinco, M.; Fournier, P.E.; Francavilla, E.; et al. Guidelines for the diagnosis of tick-borne bacterial diseases in Europe. Clin. Microbiol. Infect. 2004, 10, 1108–1132. [Google Scholar] [CrossRef]
  43. Socolovschi, C.; Mediannikov, O.; Raoult, D.; Parola, P. Update on tick-borne bacterial diseases in Europe. Parasite 2009, 16, 259–273. [Google Scholar] [CrossRef] [Green Version]
  44. Sanchez, E.; Vannier, E.; Wormser, G.P.; Hu, L.T. Diagnosis, Treatment, and Prevention of Lyme Disease, Human Granulocytic Anaplasmosis, and Babesiosis: A Review. JAMA 2016, 315, 1767–1777. [Google Scholar] [CrossRef]
  45. Platonov, A.E.; Karan, L.S.; Kolyasnikova, N.M.; Makhneva, N.A.; Toporkova, M.G.; Maleev, V.V.; Fish, D.; Krause, P.J. Humans infected with relapsing fever spirochete Borrelia miyamotoi, Russia. Emerg. Infect. Dis. 2011, 17, 1816–1823. [Google Scholar] [CrossRef]
  46. Grankvist, A.; Sandelin, L.L.; Andersson, J.; Fryland, L.; Wilhelmsson, P.; Lindgren, P.E.; Forsberg, P.; Wenneras, C. Infections with Candidatus Neoehrlichia mikurensis and Cytokine Responses in 2 Persons Bitten by Ticks, Sweden. Emerg. Infect. Dis. 2015, 21, 1462–1465. [Google Scholar] [CrossRef] [PubMed]
  47. Psaroulaki, A.; Koliou, M.; Chochlakis, D.; Ioannou, I.; Mazeri, S.; Tselentis, Y. Anaplasma phagocytophilum infection in a child. Pediatr. Infect. Dis. J. 2008, 27, 664–666. [Google Scholar] [CrossRef] [PubMed]
  48. Chochlakis, D.; Koliou, M.; Ioannou, I.; Tselentis, Y.; Psaroulaki, A. Kawasaki disease and Anaplasma sp. infection of an infant in Cyprus. Int. J. Infect. Dis. 2009, 13, e71–e73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Hindryckx, P.; D’Heygere, F. A 42-year-old man with persistent fever after holiday. Tijdschr. Geneeskd. 2009, 65, 495–496. [Google Scholar] [CrossRef]
  50. Galloo, X.; Wiels, W.; Du Four, S.; Surmont, M.; Mertens, R. Beyond Lyme: Tick-borne illness in Europe. Acta Clin. Belg. 2016, 71, 40. [Google Scholar]
  51. Chochlakis, D.; Psaroulaki, A.; Kokkini, S.; Kostanatis, S.; Arkalati, E.; Karagrannaki, E.; Tsiatis, K.; Tselentis, Y.; Gikas, A. First evidence of Anaplasma infection in Crete, Greece. Report of six human cases. Clin. Microbiol. Infect. 2009, 15, 8–9. [Google Scholar] [CrossRef] [Green Version]
  52. Lotric-Furlan, S.; Ruzic-Sabljic, E.; Strle, F. Concomitant human granulocytic anaplasmosis and Lyme neuroborreliosis. Clin. Microbiol. Infect. 2009, 15, 28–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Psaroulaki, A.; Chochlakis, D.; Ioannou, I.; Florentia, A.; Gikas, A.; Tselentis, Y. Acute anaplasmosis in humans in Cyprus. Clin. Microbiol. Infect. 2009, 15, 10–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Haschke-Becher, E.; Bernauer, R.; Walleczek, A.M.; Apfalter, P.; Afazel-Saeedi, S.; Kraus, J.; Ladurner, G.; Strasser, P. First detection of the Anaplasma phagocytophilum groEL-A genotype in man. J. Infect. 2010, 60, 300–305. [Google Scholar] [CrossRef] [PubMed]
  55. Novakova, M.; Vichova, B.; Majlathova, V.; Lesnakova, A.; Pochybova, M.; Petko, B. First case of human granulocytic anaplasmosis from Slovakia. Ann. Agric. Environ. Med. 2010, 17, 173–175. [Google Scholar]
  56. Vogl, U.M.; Presterl, E.; Stanek, G.; Ramharter, M.; Gattringer, K.B.; Graninger, W. First described case of human granulocytic anaplasmosis in a patient in Eastern Austria. Wien. Med. Wochenschr. 2010, 160, 91–93. [Google Scholar] [CrossRef]
  57. Jereb, M.; Pecaver, B.; Tomazic, J.; Muzlovic, I.; Avsic-Zupanc, T.; Premru-Srsen, T.; Levicnik-Stezinar, S.; Karner, P.; Strle, F. Severe human granulocytic anaplasmosis transmitted by blood transfusion. Emerg. Infect. Dis. 2012, 18, 1354–1357. [Google Scholar] [CrossRef] [PubMed]
  58. Koebel, C.; Kern, A.; Edouard, S.; Hoang, A.T.; Celestin, N.; Hansmann, Y.; Jaulhac, B.; Brouqui, P.; De Martino, S.J. Human granulocytic anaplasmosis in eastern France: Clinical presentation and laboratory diagnosis. Diagn. Microbiol. Infect. Dis. 2012, 72, 214–218. [Google Scholar] [CrossRef]
  59. Vanicek, J.; Stastnik, M.; Kianicka, B.; Bares, M.; Bulik, M. Rare neurological presentation of human granulocytic anaplasmosis. Eur. J. Neurol. 2013, 20, e70–e72. [Google Scholar] [CrossRef]
  60. Hagedorn, P.; Imhoff, M.; Fischer, C.; Domingo, C.; Niedrig, M. Human granulocytic anaplasmosis acquired in Scotland, 2013. Emerg. Infect. Dis. 2014, 20, 1079–1081. [Google Scholar] [CrossRef]
  61. Hing, M.; Woestyn, S.; Van Bosterhaut, B.; Desbonnet, Y.; Heyman, P.; Cochez, C.; Silaghi, C.; Sprong, H.; Fournier, P.E.; Raoult, D.; et al. Diagnosis of human granulocytic anaplasmosis in Belgium by combining molecular and serological methods. New Microbes New Infect. 2014, 2, 177–178. [Google Scholar] [CrossRef] [Green Version]
  62. Picco, P.; Naselli, A.; Pala, G.; Rizzo, F.; Damasio, B.; Buoncompagni, A.; Martini, A. Whole-body MRI as an unconventional diagnostic tool in a pediatric patient with systemic infection. Acta Radiol. Short Rep. 2014, 3, 2047981614549571. [Google Scholar] [CrossRef] [PubMed]
  63. Welc-Faleciak, R.; Kowalec, M.; Zajkowska, J.; Pancewicz, S.A.; Sinski, E. Clinical and molecular features of one case of human infection with Anaplasma phagocytophilum from Podlaskie Province in eastern Poland. Ann. Agric. Environ. Med. 2015, 22, 414–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Garcia, J.C.; Nunez, M.J.; Portillo, A.; Oteo, J.A. Human anaplasmosis: Two case-reports. Enferm. Infecc. Microbiol. Clin. 2015, 33, 68–69. [Google Scholar] [CrossRef]
  65. Markowicz, M.; Schotta, A.M.; Wijnveld, M.; Stanek, G. Human granulocytic anaplasmosis acquired in Connecticut, USA, diagnosed in Vienna, Austria, 2015. Diagn. Microbiol. Infect. Dis. 2016, 84, 347–349. [Google Scholar] [CrossRef] [PubMed]
  66. Pokorn, M.; Zupanc, T.A.; Strle, F. Pediatric Human Granulocytic Anaplasmosis is Rare in Europe. Pediatr. Infect. Dis. J. 2016, 35, 358–359. [Google Scholar] [CrossRef]
  67. Tsiodras, S.; Spanakis, N.; Spanakos, G.; Pervanidou, D.; Georgakopoulou, T.; Campos, E.; Petra, T.; Kanellopoulos, P.; Georgiadis, G.; Antalis, E.; et al. Fatal human anaplasmosis associated with macrophage activation syndrome in Greece and the Public Health response. J. Infect. Public Health 2017, 10, 819–823. [Google Scholar] [CrossRef]
  68. Lagler, H.; Harrison, N.; Kussmann, M.; Obermuller, M.; Burgmann, H.; Makristathis, A.; Ramharter, M. Direct detection of Anaplasma phagocytophilum by polymerase chain reaction followed by electrospray ionization mass spectrometry from human blood. Int. J. Infect. Dis. 2017, 60, 61–63. [Google Scholar] [CrossRef] [Green Version]
  69. Vikse, J.; Klos, J.; Berg, A. A travelling camper with a spiking fever, headache, myalgia, hepatitis, and intracellular inclusions. Lancet Infect. Dis. 2017, 17, 1318. [Google Scholar] [CrossRef]
  70. Hulinska, D.; Votypka, J.; Plch, J.; Vlcek, E.; Valesova, M.; Bojar, M.; Hulinsky, V.; Smetana, K. Molecular and microscopical evidence of Ehrlichia spp. and Borrelia burgdorferi sensu lato in patients, animals and ticks in the Czech Republic. New Microbiol. 2002, 25, 437–448. [Google Scholar]
  71. Dumler, J.S.; Choi, K.S.; Garcia-Garcia, J.C.; Barat, N.S.; Scorpio, D.G.; Garyu, J.W.; Grab, D.J.; Bakken, J.S. Human granulocytic anaplasmosis and Anaplasma phagocytophilum. Emerg. Infect. Dis. 2005, 11, 1828–1834. [Google Scholar] [CrossRef]
  72. Granquist, E.G.; Aleksandersen, M.; Bergstrom, K.; Dumler, S.J.; Torsteinbo, W.O.; Stuen, S. A morphological and molecular study of Anaplasma phagocytophilum transmission events at the time of Ixodes ricinus tick bite. Acta Vet. Scand. 2010, 52, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Stuen, S. Anaplasma phagocytophilum—The most widespread tick-borne infection in animals in Europe. Vet. Res. Commun. 2007, 31, 79–84. [Google Scholar] [CrossRef] [PubMed]
  74. Fourie, J.J.; Evans, A.; Labuschagne, M.; Crafford, D.; Madder, M.; Pollmeier, M.; Schunack, B. Transmission of Anaplasma phagocytophilum (Foggie, 1949) by Ixodes ricinus (Linnaeus, 1758) ticks feeding on dogs and artificial membranes. Parasit. Vectors 2019, 12, 136. [Google Scholar] [CrossRef] [PubMed]
  75. Ogden, N.H.; Casey, A.N.; French, N.P.; Woldehiwet, Z. A review of studies on the transmission of Anaplasma phagocytophilum from sheep: Implications for the force of infection in endemic cycles. Exp. Appl. Acarol. 2002, 28, 195–202. [Google Scholar] [CrossRef]
  76. Tomassone, L.; Berriatua, E.; De Sousa, R.; Duscher, G.G.; Mihalca, A.D.; Silaghi, C.; Sprong, H.; Zintl, A. Neglected vector-borne zoonoses in Europe: Into the wild. Vet. Parasitol. 2018, 251, 17–26. [Google Scholar] [CrossRef] [Green Version]
  77. Jahfari, S.; Coipan, E.C.; Fonville, M.; van Leeuwen, A.D.; Hengeveld, P.; Heylen, D.; Heyman, P.; van Maanen, C.; Butler, C.M.; Foldvari, G.; et al. Circulation of four Anaplasma phagocytophilum ecotypes in Europe. Parasit. Vectors 2014, 7, 365. [Google Scholar] [CrossRef] [Green Version]
  78. Stuen, S.; Granquist, E.G.; Silaghi, C. Anaplasma phagocytophilum—A widespread multi-host pathogen with highly adaptive strategies. Front. Cell. Infect. Microbiol. 2013, 3, 31. [Google Scholar] [CrossRef] [Green Version]
  79. Foldvari, G.; Jahfari, S.; Rigo, K.; Jablonszky, M.; Szekeres, S.; Majoros, G.; Toth, M.; Molnar, V.; Coipan, E.C.; Sprong, H. Candidatus Neoehrlichia mikurensis and Anaplasma phagocytophilum in urban hedgehogs. Emerg. Infect. Dis. 2014, 20, 496–498. [Google Scholar] [CrossRef]
  80. Dunaj, J.; Moniuszko-Malinowska, A.; Swiecicka, I.; Andersson, M.; Czupryna, P.; Rutkowski, K.; Zambrowski, G.; Zajkowska, J.; Grygorczuk, S.; Kondrusik, M.; et al. Tick-borne infections and co-infections in patients with non-specific symptoms in Poland. Adv. Med. Sci. 2018, 63, 167–172. [Google Scholar] [CrossRef]
  81. Ramharter, M.; Walochnik, J.; Lagler, H.; Winkler, S.; Wernsdorfer, W.H.; Stoiser, B.; Graninger, W. Clinical and molecular characterization of a near fatal case of human babesiosis in Austria. J. Travel Med. 2010, 17, 416–418. [Google Scholar] [CrossRef] [Green Version]
  82. Holler, J.G.; Roser, D.; Nielsen, H.V.; Eickhardt, S.; Chen, M.; Lester, A.; Bang, D.; Frandsen, C.; David, K.P. A case of human babesiosis in Denmark. Travel Med. Infect. Dis. 2013, 11, 324–328. [Google Scholar] [CrossRef] [PubMed]
  83. Poisnel, E.; Ebbo, M.; Berda-Haddad, Y.; Faucher, B.; Bernit, E.; Carcy, B.; Piarroux, R.; Harle, J.R.; Schleinitz, N. Babesia microti: An unusual travel-related disease. BMC Infect. Dis. 2013, 13, 99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Surgers, L.; Belkadi, G.; Foucard, A.; Lalande, V.; Girard, P.M.; Hennequin, C. Babesiosis and Lyme disease co-infection in a female patient returning from the United States. Med. Mal. Infect. 2015, 45, 490–492. [Google Scholar] [CrossRef] [PubMed]
  85. Jablonska, J.; Zarnowska-Prymek, H.; Stanczak, J.; Kozlowska, J.; Wiercinska-Drapalo, A. Symptomatic co-infection with Babesia microti and Borrelia burgdorferi in patient after international exposure; a challenging case in Poland. Ann. Agric. Environ. Med. 2016, 23, 387–389. [Google Scholar] [CrossRef]
  86. Arsuaga, M.; Gonzalez, L.M.; Lobo, C.A.; de la Calle, F.; Bautista, J.M.; Azcarate, I.G.; Puente, S.; Montero, E. First Report of Babesia microti-Caused Babesiosis in Spain. Vector Borne Zoonotic Dis. 2016, 16, 677–679. [Google Scholar] [CrossRef] [Green Version]
  87. De Ramon, C.; Cid, J.; Rodriguez-Tajes, S.; Alvarez-Martinez, M.J.; Valls, M.E.; Fernandez, J.; Lozano, M. Severe Babesia microti infection in an American immunocompetent patient diagnosed in Spain. Transfus. Apher. Sci. 2016, 55, 243–244. [Google Scholar] [CrossRef]
  88. Mcnamara, R.; van der Velde, J.; Meike, R. Babesiosis-an unusual cause of acute confusion in an elderly man. Eur. Geriatr. Med. 2010, 1, S158. [Google Scholar]
  89. Browne, S.; Ryan, Y.; Goodyer, M.; Gilligan, O. Fatal babesiosis in an asplenic patient. Br. J. Haematol. 2010, 148, 494. [Google Scholar] [CrossRef]
  90. Haapasalo, K.; Suomalainen, P.; Sukura, A.; Siikamaki, H.; Jokiranta, T.S. Fatal babesiosis in man, Finland, 2004. Emerg. Infect. Dis. 2010, 16, 1116–1118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Martinot, M.; Zadeh, M.M.; Hansmann, Y.; Grawey, I.; Christmann, D.; Aguillon, S.; Jouglin, M.; Chauvin, A.; De Briel, D. Babesiosis in immunocompetent patients, Europe. Emerg. Infect. Dis. 2011, 17, 114–116. [Google Scholar] [CrossRef]
  92. Gonzalez, L.M.; Rojo, S.; Gonzalez-Camacho, F.; Luque, D.; Lobo, C.A.; Montero, E. Severe babesiosis in immunocompetent man, Spain, 2011. Emerg. Infect. Dis. 2014, 20, 724–726. [Google Scholar] [CrossRef] [PubMed]
  93. Gonzalez, L.M.; Castro, E.; Lobo, C.A.; Richart, A.; Ramiro, R.; Gonzalez-Camacho, F.; Luque, D.; Velasco, A.C.; Montero, E. First report of Babesia divergens infection in an HIV patient. Int. J. Infect. Dis. 2015, 33, 202–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Morch, K.; Holmaas, G.; Frolander, P.S.; Kristoffersen, E.K. Severe human Babesia divergens infection in Norway. Int. J. Infect. Dis. 2015, 33, 37–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Tanyel, E.; Guler, N.; Hokelek, M.; Ulger, F.; Sunbul, M. A case of severe babesiosis treated successfully with exchange transfusion. Int. J. Infect. Dis. 2015, 38, 83–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. O’Connell, S.; Lyons, C.; Abdou, M.; Patowary, R.; Aslam, S.; Kinsella, N.; Zintl, A.; Hunfeld, K.P.; Wormser, G.P.; Gray, J.; et al. Splenic dysfunction from celiac disease resulting in severe babesiosis. Ticks Tick Borne Dis. 2017, 8, 537–539. [Google Scholar] [CrossRef] [PubMed]
  97. Asensi, V.; Gonzalez, L.M.; Fernandez-Suarez, J.; Sevilla, E.; Navascues, R.A.; Suarez, M.L.; Lauret, M.E.; Bernardo, A.; Carton, J.A.; Montero, E. A fatal case of Babesia divergens infection in Northwestern Spain. Ticks Tick Borne Dis. 2018, 9, 730–734. [Google Scholar] [CrossRef]
  98. Haselbarth, K.; Kurz, M.; Hunfeld, K.P.; Krieger, G. Babesiosis in an immunocompromised German patient. Med. Klin. 2008, 103, 104–107. [Google Scholar] [CrossRef]
  99. Blum, S.; Gattringer, R.; Haschke, E.; Walochnik, J.; Tschurtschenthaler, G.; Lang, F.; Oberbauer, R. The case: Hemolysis and acute renal failure. Babesiosis. Kidney Int. 2011, 80, 681–683. [Google Scholar] [CrossRef] [Green Version]
  100. Blackberg, J.; Lazarevic, V.L.; Hunfeld, K.P.; Persson, K.E.M. Low-virulent Babesia venatorum infection masquerading as hemophagocytic syndrome. Ann. Hematol. 2018, 97, 731–733. [Google Scholar] [CrossRef]
  101. Malandrin, L.; L’Hostis, M.; Chauvin, A. Isolation of Babesia divergens from carrier cattle blood using in vitro culture. Vet. Res. 2004, 35, 131–139. [Google Scholar] [CrossRef] [Green Version]
  102. Gorenflot, A.; Brasseur, P.; Precigout, E.; L’Hostis, M.; Marchand, A.; Schrevel, J. Cytological and immunological responses to Babesia divergens in different hosts: Ox, gerbil, man. Parasitol. Res. 1991, 77, 3–12. [Google Scholar] [CrossRef] [PubMed]
  103. Cuesta, I.; Gonzalez, L.M.; Estrada, K.; Grande, R.; Zaballos, A.; Lobo, C.A.; Barrera, J.; Sanchez-Flores, A.; Montero, E. High-Quality Draft Genome Sequence of Babesia divergens, the Etiological Agent of Cattle and Human Babesiosis. Genome Announc. 2014, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Krampitz, H.E. Babesia microti: Morphology, distribution and host relationship in Germany. Zentralbl. Bakteriol. Orig. A 1979, 244, 411–415. [Google Scholar] [PubMed]
  105. Bonnet, S.; Jouglin, M.; L’Hostis, M.; Chauvin, A. Babesia sp. EU1 from roe deer and transmission within Ixodes ricinus. Emerg. Infect. Dis. 2007, 13, 1208–1210. [Google Scholar] [CrossRef] [PubMed]
  106. Gray, J.; Zintl, A.; Hildebrandt, A.; Hunfeld, K.P.; Weiss, L. Zoonotic babesiosis: Overview of the disease and novel aspects of pathogen identity. Ticks Tick Borne Dis. 2010, 1, 3–10. [Google Scholar] [CrossRef] [PubMed]
  107. Gray, J.; von Stedingk, L.V.; Gurtelschmid, M.; Granstrom, M. Transmission studies of Babesia microti in Ixodes ricinus ticks and gerbils. J. Clin. Microbiol. 2002, 40, 1259–1263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Lewis, D.; Young, E.R. The transmission of a human strain of Babesia divergens by Ixodes ricinus ticks. J. Parasitol. 1980, 66, 359–360. [Google Scholar] [CrossRef]
  109. Bonnet, S.; Brisseau, N.; Hermouet, A.; Jouglin, M.; Chauvin, A. Experimental in vitro transmission of Babesia sp. (EU1) by Ixodes ricinus. Vet. Res. 2009, 40, 21. [Google Scholar] [CrossRef] [Green Version]
  110. Hovius, J.W.; de Wever, B.; Sohne, M.; Brouwer, M.C.; Coumou, J.; Wagemakers, A.; Oei, A.; Knol, H.; Narasimhan, S.; Hodiamont, C.J.; et al. A case of meningoencephalitis by the relapsing fever spirochaete Borrelia miyamotoi in Europe. Lancet 2013, 382, 658. [Google Scholar] [CrossRef] [Green Version]
  111. Boden, K.; Lobenstein, S.; Hermann, B.; Margos, G.; Fingerle, V. Borrelia miyamotoi-Associated Neuroborreliosis in Immunocompromised Person. Emerg. Infect. Dis. 2016, 22, 1617–1620. [Google Scholar] [CrossRef]
  112. Hoornstra, D.; Koetsveld, J.; Sprong, H.; Platonov, A.E.; Hovius, J.W. Borrelia miyamotoi Disease in an Immunocompetent Patient, Western Europe. Emerg. Infect. Dis. 2018, 24, 1770–1772. [Google Scholar] [CrossRef] [Green Version]
  113. Kuleshov, K.V.; Hoornstra, D.; Sprong, H.; Platonov, A.E.; Hovius, J.W. Draft Whole-Genome Sequences of Two Western European Borrelia miyamotoi Isolates. Microbiol. Resour. Announc. 2019, 8. [Google Scholar] [CrossRef] [Green Version]
  114. Wagemakers, A.; Staarink, P.J.; Sprong, H.; Hovius, J.W. Borrelia miyamotoi: A widespread tick-borne relapsing fever spirochete. Trends Parasitol. 2015, 31, 260–269. [Google Scholar] [CrossRef]
  115. Henningsson, A.J.; Asgeirsson, H.; Hammas, B.; Karlsson, E.; Parke, A.; Hoornstra, D.; Wilhelmsson, P.; Hovius, J.W. Two Cases of Borrelia miyamotoi Meningitis, Sweden, 2018. Emerg. Infect. Dis. 2019, 25, 1965–1968. [Google Scholar] [CrossRef] [Green Version]
  116. Narasimhan, S.; Hoornstra, D. Ixodes scapularis and Ixodes ricinus murine transmission of an American, European and Japanese Borrelia miyamotoi tick strain. Unpublished.
  117. Van Duijvendijk, G.; Coipan, C.; Wagemakers, A.; Fonville, M.; Ersoz, J.; Oei, A.; Foldvari, G.; Hovius, J.; Takken, W.; Sprong, H. Larvae of Ixodes ricinus transmit Borrelia afzelii and B. miyamotoi to vertebrate hosts. Parasit. Vectors 2016, 9, 97. [Google Scholar] [CrossRef] [Green Version]
  118. Fehr, J.S.; Bloemberg, G.V.; Ritter, C.; Hombach, M.; Luscher, T.F.; Weber, R.; Keller, P.M. Septicemia caused by tick-borne bacterial pathogen Candidatus Neoehrlichia mikurensis. Emerg. Infect. Dis. 2010, 16, 1127–1129. [Google Scholar] [CrossRef] [Green Version]
  119. Von Loewenich, F.D.; Geissdorfer, W.; Disque, C.; Matten, J.; Schett, G.; Sakka, S.G.; Bogdan, C. Detection of “Candidatus Neoehrlichia mikurensis” in two patients with severe febrile illnesses: Evidence for a European sequence variant. J. Clin. Microbiol. 2010, 48, 2630–2635. [Google Scholar] [CrossRef] [Green Version]
  120. Welinder-Olsson, C.; Kjellin, E.; Vaht, K.; Jacobsson, S.; Wenneras, C. First case of human "Candidatus Neoehrlichia mikurensis" infection in a febrile patient with chronic lymphocytic leukemia. J. Clin. Microbiol. 2010, 48, 1956–1959. [Google Scholar] [CrossRef] [Green Version]
  121. Pekova, S.; Vydra, J.; Kabickova, H.; Frankova, S.; Haugvicova, R.; Mazal, O.; Cmejla, R.; Hardekopf, D.W.; Jancuskova, T.; Kozak, T. Candidatus Neoehrlichia mikurensis infection identified in 2 hematooncologic patients: Benefit of molecular techniques for rare pathogen detection. Diagn. Microbiol. Infect. Dis. 2011, 69, 266–270. [Google Scholar] [CrossRef]
  122. Grankvist, A.; Andersson, P.O.; Mattsson, M.; Sender, M.; Vaht, K.; Hoper, L.; Sakiniene, E.; Trysberg, E.; Stenson, M.; Fehr, J.; et al. Infections with the tick-borne bacterium "Candidatus Neoehrlichia mikurensis" mimic noninfectious conditions in patients with B cell malignancies or autoimmune diseases. Clin. Infect. Dis. 2014, 58, 1716–1722. [Google Scholar] [CrossRef] [PubMed]
  123. Andreasson, K.; Jonsson, G.; Lindell, P.; Gulfe, A.; Ingvarsson, R.; Lindqvist, E.; Saxne, T.; Grankvist, A.; Wenneras, C.; Marsal, J. Recurrent fever caused by Candidatus Neoehrlichia mikurensis in a rheumatoid arthritis patient treated with rituximab. Rheumatology 2015, 54, 369–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Portillo, A.; Santibanez, P.; Palomar, A.M.; Santibanez, S.; Oteo, J.A. ‘Candidatus Neoehrlichia mikurensis’ in Europe. New Microbes New Infect. 2018, 22, 30–36. [Google Scholar] [CrossRef] [PubMed]
  125. Heylen, D.; Fonville, M.; van Leeuwen, A.D.; Sprong, H. Co-infections and transmission dynamics in a tick-borne bacterium community exposed to songbirds. Environ. Microbiol. 2016, 18, 988–996. [Google Scholar] [CrossRef]
  126. Richter, D.; Matuschka, F.R. “Candidatus Neoehrlichia mikurensis,” Anaplasma phagocytophilum, and lyme disease spirochetes in questing european vector ticks and in feeding ticks removed from people. J. Clin. Microbiol. 2012, 50, 943–947. [Google Scholar] [CrossRef] [Green Version]
  127. Jahfari, S.; Fonville, M.; Hengeveld, P.; Reusken, C.; Scholte, E.J.; Takken, W.; Heyman, P.; Medlock, J.M.; Heylen, D.; Kleve, J.; et al. Prevalence of Neoehrlichia mikurensis in ticks and rodents from North-west Europe. Parasit. Vectors 2012, 5, 74. [Google Scholar] [CrossRef] [Green Version]
  128. Andersson, M.; Bartkova, S.; Lindestad, O.; Raberg, L. Co-infection with ‘Candidatus Neoehrlichia Mikurensis’ and Borrelia afzelii in Ixodes ricinus ticks in southern Sweden. Vector Borne Zoonotic Dis. 2013, 13, 438–442. [Google Scholar] [CrossRef]
  129. Burri, C.; Schumann, O.; Schumann, C.; Gern, L. Are Apodemus spp. mice and Myodes glareolus reservoirs for Borrelia miyamotoi, Candidatus Neoehrlichia mikurensis, Rickettsia helvetica, R. monacensis and Anaplasma phagocytophilum? Ticks Tick Borne Dis. 2014, 5, 245–251. [Google Scholar] [CrossRef]
  130. Nilsson, K. Septicaemia with Rickettsia helvetica in a patient with acute febrile illness, rash and myasthenia. J. Infect. 2009, 58, 79–82. [Google Scholar] [CrossRef]
  131. Nilsson, K.; Wallmenius, K.; Pahlson, C. Coinfection with Rickettsia helvetica and Herpes Simplex Virus 2 in a Young Woman with Meningoencephalitis. Case Rep. Infect. Dis. 2011, 2011, 469194. [Google Scholar] [CrossRef] [Green Version]
  132. Jado, I.; Oteo, J.A.; Aldamiz, M.; Gil, H.; Escudero, R.; Ibarra, V.; Portu, J.; Portillo, A.; Lezaun, M.J.; Garcia-Amil, C.; et al. Rickettsia monacensis and human disease, Spain. Emerg. Infect. Dis. 2007, 13, 1405–1407. [Google Scholar] [CrossRef]
  133. Madeddu, G.; Mancini, F.; Caddeo, A.; Ciervo, A.; Babudieri, S.; Maida, I.; Fiori, M.L.; Rezza, G.; Mura, M.S. Rickettsia monacensis as cause of Mediterranean spotted fever-like illness, Italy. Emerg. Infect. Dis. 2012, 18, 702–704. [Google Scholar] [CrossRef]
  134. Tijsse-Klasen, E.; Sprong, H.; Pandak, N. Co-infection of Borrelia burgdorferi sensu lato and Rickettsia species in ticks and in an erythema migrans patient. Parasit. Vectors 2013, 6, 347. [Google Scholar] [CrossRef] [Green Version]
  135. Elfving, K.; Lukinius, A.; Nilsson, K. Life cycle, growth characteristics and host cell response of Rickettsia helvetica in a Vero cell line. Exp. Appl. Acarol. 2012, 56, 179–187. [Google Scholar] [CrossRef] [Green Version]
  136. Dong, X.; El Karkouri, K.; Robert, C.; Gavory, F.; Raoult, D.; Fournier, P.E. Genomic comparison of Rickettsia helvetica and other Rickettsia species. J. Bacteriol. 2012, 194, 2751. [Google Scholar] [CrossRef]
  137. Simser, J.A.; Palmer, A.T.; Fingerle, V.; Wilske, B.; Kurtti, T.J.; Munderloh, U.G. Rickettsia monacensis sp. nov., a spotted fever group Rickettsia, from ticks (Ixodes ricinus) collected in a European city park. Appl. Environ. Microbiol. 2002, 68, 4559–4566. [Google Scholar] [CrossRef] [Green Version]
  138. Nilsson, K.; Lindquist, O.; Pahlson, C. Association of Rickettsia helvetica with chronic perimyocarditis in sudden cardiac death. Lancet 1999, 354, 1169–1173. [Google Scholar] [CrossRef]
  139. Nilsson, K.; Pahlson, C.; Lukinius, A.; Eriksson, L.; Nilsson, L.; Lindquist, O. Presence of Rickettsia helvetica in granulomatous tissue from patients with sarcoidosis. J. Infect. Dis. 2002, 185, 1128–1138. [Google Scholar] [CrossRef] [Green Version]
  140. Nielsen, H.; Fournier, P.E.; Pedersen, I.S.; Krarup, H.; Ejlertsen, T.; Raoult, D. Serological and molecular evidence of Rickettsia helvetica in Denmark. Scand. J. Infect. Dis. 2004, 36, 559–563. [Google Scholar] [CrossRef]
  141. Heylen, D.; Fonville, M.; Docters van Leeuwen, A.; Stroo, A.; Duisterwinkel, M.; van Wieren, S.; Diuk-Wasser, M.; de Bruin, A.; Sprong, H. Pathogen communities of songbird-derived ticks in Europe’s low countries. Parasit. Vectors 2017, 10, 497. [Google Scholar] [CrossRef]
  142. Akl, T.; Bourgoin, G.; Souq, M.L.; Appolinaire, J.; Poirel, M.T.; Gibert, P.; Abi Rizk, G.; Garel, M.; Zenner, L. Detection of tick-borne pathogens in questing Ixodes ricinus in the French Pyrenees and first identification of Rickettsia monacensis in France. Parasite 2019, 26, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Gajda, E.; Hildebrand, J.; Sprong, H.; Bunkowska-Gawlik, K.; Perec-Matysiak, A.; Coipan, E.C. Spotted fever rickettsiae in wild-living rodents from south-western Poland. Parasit. Vectors 2017, 10, 413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Elfving, K.; Malmsten, J.; Dalin, A.M.; Nilsson, K. Serologic and Molecular Prevalence of Rickettsia helvetica and Anaplasma phagocytophilum in Wild Cervids and Domestic Mammals in the Central Parts of Sweden. Vector Borne Zoonotic Dis. 2015, 15, 529–534. [Google Scholar] [CrossRef]
  145. Laaksonen, M.; Klemola, T.; Feuth, E.; Sormunen, J.J.; Puisto, A.; Makela, S.; Penttinen, R.; Ruohomaki, K.; Hanninen, J.; Saaksjarvi, I.E.; et al. Tick-borne pathogens in Finland: Comparison of Ixodes ricinus and I. persulcatus in sympatric and parapatric areas. Parasit. Vectors 2018, 11, 556. [Google Scholar] [CrossRef] [Green Version]
  146. Minichova, L.; Hamsikova, Z.; Mahrikova, L.; Slovak, M.; Kocianova, E.; Kazimirova, M.; Skultety, L.; Stefanidesova, K.; Spitalska, E. Molecular evidence of Rickettsia spp. in ixodid ticks and rodents in suburban, natural and rural habitats in Slovakia. Parasit. Vectors 2017, 10, 158. [Google Scholar] [CrossRef] [Green Version]
  147. Biernat, B.; Stanczak, J.; Michalik, J.; Sikora, B.; Cieniuch, S. Rickettsia helvetica and R. monacensis infections in immature Ixodes ricinus ticks derived from sylvatic passerine birds in west-central Poland. Parasitol. Res. 2016, 115, 3469–3477. [Google Scholar] [CrossRef] [Green Version]
  148. Honig, V.; Carolan, H.E.; Vavruskova, Z.; Massire, C.; Mosel, M.R.; Crowder, C.D.; Rounds, M.A.; Ecker, D.J.; Ruzek, D.; Grubhoffer, L.; et al. Broad-range survey of vector-borne pathogens and tick host identification of Ixodes ricinus from Southern Czech Republic. FEMS Microbiol. Ecol. 2017, 93. [Google Scholar] [CrossRef]
  149. Parola, P.; Paddock, C.D.; Socolovschi, C.; Labruna, M.B.; Mediannikov, O.; Kernif, T.; Abdad, M.Y.; Stenos, J.; Bitam, I.; Fournier, P.E.; et al. Update on tick-borne rickettsioses around the world: A geographic approach. Clin. Microbiol. Rev. 2013, 26, 657–702. [Google Scholar] [CrossRef] [Green Version]
  150. Piesman, J.; Gern, L. Lyme borreliosis in Europe and North America. Parasitology 2004, 129, S191–S220. [Google Scholar] [CrossRef]
  151. Valarcher, J.F.; Hagglund, S.; Juremalm, M.; Blomqvist, G.; Renstrom, L.; Zohari, S.; Leijon, M.; Chirico, J. Tick-borne encephalitis. Rev. Sci. Tech. 2015, 34, 453–466. [Google Scholar] [CrossRef] [Green Version]
  152. Mansfield, K.L.; Johnson, N.; Phipps, L.P.; Stephenson, J.R.; Fooks, A.R.; Solomon, T. Tick-borne encephalitis virus—A review of an emerging zoonosis. J. Gen. Virol. 2009, 90, 1781–1794. [Google Scholar] [CrossRef] [PubMed]
  153. Wormser, G.P.; Dattwyler, R.J.; Shapiro, E.D.; Halperin, J.J.; Steere, A.C.; Klempner, M.S.; Krause, P.J.; Bakken, J.S.; Strle, F.; Stanek, G.; et al. The clinical assessment, treatment, and prevention of lyme disease, human granulocytic anaplasmosis, and babesiosis: Clinical practice guidelines by the Infectious Diseases Society of America. Clin. Infect. Dis. 2006, 43, 1089–1134. [Google Scholar] [CrossRef] [PubMed]
  154. Hulinska, D.; Votypka, J.; Vanousova, D.; Hercogova, J.; Hulinsky, V.; Drevova, H.; Kurzova, Z.; Uherkova, L. Identification of Anaplasma phagocytophilum and Borrelia burgdorferi sensu lato in patients with erythema migrans. Folia Microbiol. 2009, 54, 246–256. [Google Scholar] [CrossRef] [PubMed]
  155. Dvorakova Heroldova, M.; Dvorackova, M. Seroprevalence of Anaplasma phagocytophilum in patients with suspected Lyme borreliosis. Epidemiol. Mikrobiol. Imunol. 2014, 63, 297–302. [Google Scholar] [PubMed]
  156. Vannier, E.; Krause, P.J. Human babesiosis. N. Engl. J. Med. 2012, 366, 2397–2407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Hildebrandt, A.; Gray, J.S.; Hunfeld, K.P. Human babesiosis in Europe: What clinicians need to know. Infection 2013, 41, 1057–1072. [Google Scholar] [CrossRef]
  158. Swanson, S.J.; Neitzel, D.; Reed, K.D.; Belongia, E.A. Coinfections acquired from ixodes ticks. Clin. Microbiol. Rev. 2006, 19, 708–727. [Google Scholar] [CrossRef] [Green Version]
  159. Krause, P.J.; Telford, S.R., 3rd; Spielman, A.; Sikand, V.; Ryan, R.; Christianson, D.; Burke, G.; Brassard, P.; Pollack, R.; Peck, J.; et al. Concurrent Lyme disease and babesiosis. Evidence for increased severity and duration of illness. JAMA 1996, 275, 1657–1660. [Google Scholar] [CrossRef]
  160. Belongia, E.A. Epidemiology and impact of coinfections acquired from Ixodes ticks. Vector Borne Zoonotic Dis. 2002, 2, 265–273. [Google Scholar] [CrossRef]
  161. Parasites—Babesiosis. Available online: https://www.cdc.gov/parasites/babesiosis/data-statistics/index.html (accessed on 16 December 2019).
  162. Hildebrandt, A.; Hunfeld, K.P.; Baier, M.; Krumbholz, A.; Sachse, S.; Lorenzen, T.; Kiehntopf, M.; Fricke, H.J.; Straube, E. First confirmed autochthonous case of human Babesia microti infection in Europe. Eur. J. Clin. Microbiol. Infect. Dis. 2007, 26, 595–601. [Google Scholar] [CrossRef]
  163. Koetsveld, J.; Kolyasnikova, N.M.; Wagemakers, A.; Toporkova, M.G.; Sarksyan, D.S.; Oei, A.; Platonov, A.E.; Hovius, J.W. Development and optimization of an in vitro cultivation protocol allows for isolation of Borrelia miyamotoi from patients with hard tick-borne relapsing fever. Clin. Microbiol. Infect. 2017, 23, 480–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Kuleshov, K.V.; Margos, G.; Fingerle, V.; Koetsveld, J.; Goptar, I.A.; Markelov, M.L.; Kolyasnikova, N.M.; Sarksyan, D.S.; Kirdyashkina, N.P.; Shipulin, G.A.; et al. Whole genome sequencing of Borrelia miyamotoi isolate Izh-4: Reference for a complex bacterial genome. BMC Genom. 2020, 21, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Quarsten, H.; Grankvist, A.; Hoyvoll, L.; Myre, I.B.; Skarpaas, T.; Kjelland, V.; Wenneras, C.; Noraas, S. Candidatus Neoehrlichia mikurensis and Borrelia burgdorferi sensu lato detected in the blood of Norwegian patients with erythema migrans. Ticks Tick Borne Dis. 2017, 8, 715–720. [Google Scholar] [CrossRef] [PubMed]
  166. Steere, A.C.; McHugh, G.; Damle, N.; Sikand, V.K. Prospective study of serologic tests for lyme disease. Clin. Infect. Dis. 2008, 47, 188–195. [Google Scholar] [CrossRef] [Green Version]
  167. Ismail, N.; Bloch, K.C.; McBride, J.W. Human ehrlichiosis and anaplasmosis. Clin. Lab. Med. 2010, 30, 261–292. [Google Scholar] [CrossRef] [Green Version]
  168. Del Bene, V.E. Temperature. In Clinical Methods: The History, Physical, and Laboratory Examinations, 3rd ed.; Walker, H.K., Hall, W.D., Hurst, J.W., Eds.; Butterworths: Boston, MA, USA, 1990. [Google Scholar]
Figure 1. Results of the systematic and structural literature search for case reports of I. ricinus-borne microorganisms in the period of January 2008-March 2018. The literature search was performed by two independent authors and the figure shows the selection process involved.
Figure 1. Results of the systematic and structural literature search for case reports of I. ricinus-borne microorganisms in the period of January 2008-March 2018. The literature search was performed by two independent authors and the figure shows the selection process involved.
Pathogens 09 00150 g001
Table 1. Summary of the molecular biology methods performed to confirm infection with I. ricinus-borne microorganisms in the case reports included in this study.
Table 1. Summary of the molecular biology methods performed to confirm infection with I. ricinus-borne microorganisms in the case reports included in this study.
MicroorganismNumber of CasesPCR (*Sequenced)SerologyBlood SmearBone Marrow SmearOther MicroscopyCultivation
A. phagocytophilum3223 (*17)2672--
B. microti77 (*5)26---
B. divergens117 (*7)510---
B. venatorum/EU133 (*3)12---
B. miyamotoi32 (*2)1--2-
N. mikurensis1414 (*13)---2-
R. helvetica33 (*3)2---2
R. monacensis44 (*4)3---1
*:Sequenced PCR results.
Table 2. Overview of European cases of I. ricinus-borne diseases between 1 January 2008 and 1 March 2018, based on the data of case reports (*) and of other studies (**) retrieved in the systematic literature search of this study. The table describes the number of cases published, clinical manifestations from case reports, exposure to ticks in case reports, cultivation, and propagation from patients or tick materials, evidence from case reports reflecting the pathology in animal models or in vitro observations and transmission experiments in laboratory or wild animals.
Table 2. Overview of European cases of I. ricinus-borne diseases between 1 January 2008 and 1 March 2018, based on the data of case reports (*) and of other studies (**) retrieved in the systematic literature search of this study. The table describes the number of cases published, clinical manifestations from case reports, exposure to ticks in case reports, cultivation, and propagation from patients or tick materials, evidence from case reports reflecting the pathology in animal models or in vitro observations and transmission experiments in laboratory or wild animals.
MicroorganismNumber of Cases *Clinical Manifestations *Exposure to Ticks *Culture from Ticks or Patients **Pathology *Transmission Experiments **
A. phagocytophilum32 [46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68]Fever (88%), headache (56%), malaise (41%), myalgia (41%), arthralgia (25%), erythematous skin manifestations such as morbilliform and maculopapular rashes (22%), lymphadenopathy (19%), abdominal complaints (16%),anemia (13%), chills (13%), splenomegaly (9%), rigors (6%), dark urine (3%)History of tick-bite (34%), exposure to ticks (38%)[70] (Cultivated from patient blood)Leucocytopenia (50%), positive blood/bone marrow smear (25%).[74](infection of I. ricinus from sheep and to naïve dogs), [75] (infection of I. ricinus from sheep under natural conditions)
B. microti7 [79,80,81,82,83,84,85]Fever (100%), anemia (86%), malaise (57%), chills (57%), headache (43%), abdominal complaints (43%), skin manifestation (erythematous rash) (14%)History of tick-bite (14%), exposure to ticks outside Europe (86%)Noneanemia (86%), thrombocytopenia (86%), elevated liver enzymes (86%), elevated kidney function (14%),[107] (infection of I. ricinus from and to gerbils)
B. divergens11 [86,87,88,89,90,91,92,93,94,95]Fever (82%), anemia (45%), malaise (36%), abdominal complaints (36%), jaundice (36%), dark urine (27%), headache (18%), myalgia (18%), arthralgia (18%)History of tick-bite (45%), exposure/risk (55%)[102] (Cultivated from patient blood)Thrombocytopenia (73%), elevated kidney values (55%), elevated liver enzymes (45%), anemia (45%)[108] (infection of I. ricinus to cattle and gerbils)
B. venatorum/EU13 [96,97,98]Anemia (100%), fever (67%), dark urine (67%), myalgia (33%), rigors (33%)Exposure/risk (33%)NoneAnemia (100%), elevated kidney values (100%) and thrombocytopenia (33%)[109] (infection of I. ricinus from and to sheep blood)
B. miyamotoi3 [107,108,109]Neurological complaints (67%), headache (67%), abdominal complaints (33%), myalgia (33%), arthalgia (33%), skin manifestations (33%), lymphadenopathy (33%), weight loss (33%)History of tick-bite (100%) [113] (Cultivated from I. ricinus)Unknown[117] (infection from wild I. ricinus to naive mice), [116] (infection of I. ricinus from and to CD1 mice)
N. mikurensis14 [43,112,113,114,115,116,117]Fever (86%), skin manifestation (erythematous rash, erysipelas-like rash and EM; 36%), anemia (29%), malaise (14%), arthralgia (14%), chills (14%), abdominal complaints (14%), oedema (14%), weight loss (7%), myalgia (7%)History of tick-bite (36%), exposure/risk (14%)[41] ( Cultivated from patient blood)Unknown[129] (infection from wild rodents to I. ricinus)
R. helvetica3 [19,126,127]Fever (100%), headache (100%), photophobia (67%), skin manifestation (macular rash) (33%), myalgia (33%), arthralgia (33%)Exposure/risk (100%)[22,131] (Cultivated from patient CSF), [135](Cultivated from I. ricinus)UnknownNone
R. monacensis4 [128,129,130],Skin manifestation (100%) (erythematous rash, maculopapular rash and EM), fever (75%), headache (75%), anemia (25%)History of tick-bite (50%), exposure/risk (25%)[132] (Cultivated from patient blood), [137](Cultivated from I. ricinus)UnknownNone
Table 3. Summary of existing evidence for I. ricinus borne microorganisms disease causality in Europe and current knowledge gaps based on the systematic literature search of this study and case reports between 1 January 2008 and 1 March 2018. The table describes the number of European derived cases, cultivation, and propagation from patients or tick materials, evidence from case reports reflecting the pathology in animal models or in vitro observations and transmission experiments in laboratory or wild animals. In addition, shown for comparison B. burgdorferi s.l. and TBEV, both established TBPs [150,151,152].
Table 3. Summary of existing evidence for I. ricinus borne microorganisms disease causality in Europe and current knowledge gaps based on the systematic literature search of this study and case reports between 1 January 2008 and 1 March 2018. The table describes the number of European derived cases, cultivation, and propagation from patients or tick materials, evidence from case reports reflecting the pathology in animal models or in vitro observations and transmission experiments in laboratory or wild animals. In addition, shown for comparison B. burgdorferi s.l. and TBEV, both established TBPs [150,151,152].
Tick-Borne MicroorganismEuropean Derived CasesCultivated fromPathologyTransmission Experiments
TicksPatients
B. burgdorferi s.l. +++++
TBE virus+++++
A. phagocytophilum+-++ +/&
B. divergens+-+++
B. microti---++
B. venatorum+--+ +
B. miyamotoi++--+/#
N. mikurensis+-+--
R. helvetica+++--
R. monacensis+++--
+: Published evidence, -: No evidence, +/&: Transmission experiments with wild animals/ticks and laboratory animals, +/#: Published and unpublished evidence.

Share and Cite

MDPI and ACS Style

Azagi, T.; Hoornstra, D.; Kremer, K.; Hovius, J.W.R.; Sprong, H. Evaluation of Disease Causality of Rare Ixodes ricinus-Borne Infections in Europe. Pathogens 2020, 9, 150. https://doi.org/10.3390/pathogens9020150

AMA Style

Azagi T, Hoornstra D, Kremer K, Hovius JWR, Sprong H. Evaluation of Disease Causality of Rare Ixodes ricinus-Borne Infections in Europe. Pathogens. 2020; 9(2):150. https://doi.org/10.3390/pathogens9020150

Chicago/Turabian Style

Azagi, Tal, Dieuwertje Hoornstra, Kristin Kremer, Joppe W. R. Hovius, and Hein Sprong. 2020. "Evaluation of Disease Causality of Rare Ixodes ricinus-Borne Infections in Europe" Pathogens 9, no. 2: 150. https://doi.org/10.3390/pathogens9020150

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

Article Metrics

Back to TopTop