Next Article in Journal
Functional Characterization and Structural Analysis of NADH Oxidase Mutants from Thermus thermophilus HB27: Role of Residues 166, 174, and 194 in the Catalytic Properties and Thermostability
Next Article in Special Issue
Multiple Lineages of Usutu Virus (Flaviviridae, Flavivirus) in Blackbirds (Turdus merula) and Mosquitoes (Culex pipiens, Cx. modestus) in the Czech Republic (2016–2019)
Previous Article in Journal
Cryptosporidium Prevalence in Calves and Geese Co-Grazing on Four Livestock Farms Surrounding Two Reservoirs Supplying Public Water to Mainland Orkney, Scotland
Previous Article in Special Issue
A Dengue Vaccine: Will It be Accepted and Is It Feasible? Lessons from Barranquilla, Colombia, and Merida, Venezuela
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Inflammatory Immune Responses in Patients with Tick-Borne Encephalitis: Dynamics and Association with the Outcome of the Disease

1
Department of Infectious Diseases, University Medical Center Ljubljana, Japljeva 2, 1525 Ljubljana, Slovenia
2
Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
3
Institute for Biostatistics and Medical Informatics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
4
Department of Mathematics, Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, 6000 Koper, Slovenia
5
Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
6
Division of Rheumatology, Allergy and Immunology, Center for Immunology and Inflammatory Diseases, Masachusetts General Hospital/Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
*
Author to whom correspondence should be addressed.
Microorganisms 2019, 7(11), 514; https://doi.org/10.3390/microorganisms7110514
Submission received: 26 September 2019 / Revised: 17 October 2019 / Accepted: 29 October 2019 / Published: 31 October 2019
(This article belongs to the Special Issue Emerging Vector Borne Infections: A Novel Threat for Global Health)

Abstract

:
Information on the association of inflammatory immune responses and disease outcome after tick-borne encephalitis (TBE) is limited. In the present study, we assessed the levels of 24 cytokines/chemokines associated with innate and adaptive immune responses in matched serum and cerebrospinal fluid (CSF) samples of 81 patients at first visit, and in serum at follow-up time points. Serum levels of several cytokines/chemokines obtained during the meningoencephalitic phase of TBE differed compared to the levels at a follow-up visit 2 months later; several significant differences were also found in cytokine/chemokine levels in serum at 2 months compared to the last time point, 2–7 years after acute illness. Cytokines/chemokines levels in CSF or serum obtained at the time of acute illness or serum levels obtained 2 months after the onset of TBE did not have predictive value for an unfavorable outcome 2–7 years later. In contrast, serum levels of mediators associated with Th17 responses were lower in patients with unfavorable outcome whereas those associated with other adaptive or innate immune responses were higher at the last visit in those with an unfavorable outcome. These findings provide new insights into the immunopathogenesis of TBE and implicate inflammatory immune responses with post-encephalitic syndrome years after the initial infection.

1. Introduction

Tick-borne encephalitis (TBE) is a flaviviral infection of the central nervous system, which is endemic in many parts of Europe and Asia. It is transmitted to humans predominantly through Ixodes spp. tick bites. There is no specific antiviral treatment for TBE, but the disease is preventable by vaccination [1,2,3,4]. TBE is caused by three subtypes of the TBE virus (TBEV); European, Siberian, and Far-Eastern. Most patients infected with the European virus subtype who develop symptoms have a biphasic disease course. The initial phase presents as mild febrile illness associated with general malaise, fatigue, headache, myalgia and arthralgia, and others. The second phase of TBE presents as meningitis in about 50% of adult patients, meningoencephalitis in 40%, or meningoencephalomyelitis in 5–10%. In children, meningitis is the predominant clinical manifestation. The case fatality rate is between 0.5 and 2%. In addition, at least 30% of patients develop a post-encephalitic syndrome (PES), and approximately 5% are affected by permanent pareses [3,4,5,6].
The pathogenesis of TBE, including the interaction of TBEV with innate and adaptive immunity is not well understood. Although the clinical presentation of TBE infection is believed to be driven predominantly by the host immune responses, knowledge of the type of immune responses involved and their impact on symptomology, severity, and outcome of TBE is limited.
In addition to TBEV, several other members of the flavivirus genus, such as the Japanese encephalitis virus, dengue virus, and West Nile virus are pathogenic to humans and may cause encephalitis. The role played by the immune response caused by these viruses is poorly understood; more information is available on the acute illness than on the outcome. Dengue virus and Japanese encephalitis virus infection of the central nervous system induce expression of multiple cytokines, which may augment inflammation and other pathological changes in the infected brain [7]. Similar cerebrospinal fluid (CSF) levels of cytokines in dengue encephalitis and Japanese encephalitis patients were reported [7] and differences in the immunological response of the Japanese encephalitis and non-Japanese encephalitis cases were exposed [8]. It is of interest that in patients with Japanese encephalitis, dengue encephalitis, and nonspecific encephalitis, cytokines and chemokines did not correlate with severity of illness [9]. The reports on the association of the levels of inflammatory immune mediators in patients with flavivirus central nervous system infection are rare. It seems that during Japanese encephalitis virus infection, elevated levels of proinflammatory cytokines and chemokines are associated with a poor outcome, but whether they are simply a correlate of severe disease or contribute to pathogenesis remains to be determined [10].
In the current study, in an effort to delineate immune responses in the clinical course and outcome of TBE, we assessed 24 cytokines and chemokines associated with innate and adaptive (T and B cell) immune responses in patients with TBE who were followed systematically from the initial visit during acute illness for at least 2 years to evaluate their post-treatment status. All patients were from Central Europe (Slovenia) where the European TBEV subtype prevails. The levels of these inflammatory mediators were determined in matched CSF and serum samples at first visit, and in serum at follow-up time points, and correlated with unfavorable outcomes in TBE.

2. Materials and Methods

The study was approved by the Medical Ethics Committee of the Ministry of Health of the Republic of Slovenia (No 0120-467/2017/3). Each participant provided written informed consent.

2.1. Evaluation of Patients

This study is based on 420 adult patients who were diagnosed with TBE in the years 2007–2012 at the Department of Infectious Diseases, UMC Ljubljana, Slovenia, and assessed for long-term outcome in 2014. Detailed clinical information and evaluation criteria on these patients, and the inflammatory profile in the acute meningoencephalitic phase of illness in a subset of them, were reported previously [11,12]. Briefly, clinical and laboratory information on the course and severity of acute illness was obtained and the outcome of TBE was assessed at follow-up visits. During hospitalization (initial visit) and at each follow-up visit, each patient was evaluated for the presence of clinical signs and subjective symptoms. Determination of TBE-associated symptoms was made if patients reported newly developed symptoms or worsening of existing symptoms since the onset of TBE without another medical explanation.
We defined TBE sequelae as objective neurological signs (ataxia, tremor, cranial and spinal nerve pareses, etc.) and subjective symptoms (headache, dizziness, concentration and memory disorders, emotional lability, sleep disorders, fatigue, myalgias and/or arthralgias, etc.) that match the criteria for TBE-associated symptoms at examination 2–7 years after the acute illness.
An unfavorable long-term clinical outcome was defined as the existence of PES defined with the presence of two or more subjective symptoms fulfilling the criteria for TBE-associated symptoms or at least one objective neurological sign at the last follow-up visit 2–7 years after the acute illness.

2.2. Selection of Patients for the Analysis of Cytokine and Chemokine Levels

The present study comprised 81 patients selected from the larger cohort of 420 patients in a previous study on the clinical assessment of long-term outcome of TBE [11,12]. Patients were selected to have comparable numbers of patients with favorable versus unfavorable outcome and based on the availability of matched CSF specimens and serum samples obtained at the time of acute illness, and serum samples at follow-up examinations at 2 months and 2–7 years after study entry. Samples were aliquoted and frozen at −80 °C for an average of 76 (48–80) months for serum and 80 (78–90) months for CSF prior to testing. Samples did not undergo freeze-thaw cycles except for this cytokine analysis. No significant differences were observed in the demographic or clinical characteristics of the 81 patients who were selected for the present study and the rest of the patients in the larger cohort, with the exception that the proportion of patients with unfavorable outcome at 2–7 years after TBE was higher in the present study due to the selection criteria.

2.3. Chemokine and Cytokine Determinations

The concentration of 24 cytokines was measured in serum and CSF samples with four different Human Cytokine/Chemokine Panels (Milliplex Map, Merck, Germany) on the MagPix instrument (Luminex, Austin, TX, USA), according to the manufacturer’s instructions. Investigated cytokines/chemokines are associated with innate (GMCSF, IFNα, IL-1β, IL-6, IL-8, TNFα, MCP1 (CCL2), MIP1α (CCL3), IL-10), adaptive Th1 (IFNγ, IL-12P40, IL-12P70, CXCL10, CXCL9, CCL19), Th17 (IL-17F, IL-17A, IL-22, IL-21, IL-23, IL-25 (IL-17E), IL-27), and B cell immune response (CXCL12, CXCL13). All samples were previously aliquoted and diluted to a final concentration of 1:5. Samples in a single panel were performed on the same day and all plates were analyzed simultaneously with the Milliplex Analyst 5.1 software in order to minimize inter-assay variation. Inflammatory profiles were assessed at first visit in matched CSF and serum samples, and in serum at follow-up time points (at 2 months and 2–7 years). Samples from CSF were not available from follow-up visits.

2.4. Statistical Analyses

The categorical variables were summarized as frequencies and percentages (with their 95% confidence intervals (CI)), and the numerical variables as medians (interquartile ranges, IQR).
The Wilcoxon signed rank test was used to compare the levels of the 24 cytokines/chemokines in serum at subsequent follow-up times (at first visit during acute illness, 2 months later and at the last follow-up visit 2–7 years after TBE). Differences between groups were summarized as the ratio of the medians (medians at a follow-up visit to median at previous visit; values above 1 indicate greater median concentrations at the later visit). p values from the univariate tests and those adjusted for multiple comparisons using a multivariate permutation procedure were reported [13].
A similar analysis, based on the Mann-Whitney test was performed to compare the levels of the 24 cytokines/chemokines measured in the acute phase (CSF and serum), and at the two follow-up times to compare patients with PES and those with a favorable outcome of TBE at the last visit. The distributions of the mediators for patients with and without PES were displayed graphically using box and whisker plots.
Adjusted p values < 0.05 were considered statistically significant. Statistical analysis was performed using R statistical language (R), version 3.5.0. [14].

3. Results

As we reported previously, the analysis of the levels of cytokines and chemokines in matched serum and CSF samples obtained during meningoencephalitic phase of TBE revealed that cytokines and chemokines associated with innate immune responses or Th1 adaptive immune responses were significantly higher in CSF, whereas mediators associated with Th17 and B cell immune responses were generally higher in serum [12]. In the present study, we compared the levels of inflammatory mediators at first visit during acute illness and at follow-up time points to evaluate whether immune responses at each time point were associated with the outcome of the disease. Since there is no etiologic treatment for TBE, the natural course of the disease had been disrupted only by symptomatic therapy (most often analgetics, and rarely, nonsteroid antirheumatic drugs, none of our patients received steroids) in some of our patients.
The basic clinical, laboratory, and demographic findings during the acute illness in patients included in the present study is shown in Table 1. Of 81 patients, 43 (53%) fulfilled the criteria for PES: 37 (46%) reported ≥ 2 TBE-associated symptoms, and 11 (13%) had ≥ 1 objective neurological sign at the follow-up visit 2–7 years after acute illness (five patients had objective sequellae of TBE as well as pronounced TBE-associated symptoms).

3.1. Comparison of Inflammatory Responses in Serum during Acute Illness and Follow-Up

Serum samples for these analyses were available at the time of acute illness (n = 81, time 0), and at 2 months (n = 74) and 2–7 years thereafter (n = 81).
The analysis of the levels of serum cytokines/chemokines at different time points identified several statistically significant differences (Table 2).
A comparison of the findings at the first visit and follow-up at 2 months revealed a statistically significant decrease for five mediators associated with innate (IFNα (p = 0.003), TNFα (p = 0.002)), adaptive Th17 (IL-21 (p < 0.001), IL-27 (p < 0.001)), and B cell (CXCL12 (p = 0.038)) responses. In contrast, three mediators associated with innate or Th1 adaptive responses (MCP1 (p < 0.001), CXCL9 (p = 0.023), and CCL19 (p < 0.001)) were significantly higher at the 2-month follow-up. It is possible that these responses may decrease during acute illness and are normalized 2 months later.
Even greater statistically significant differences were found when comparing the cytokine and chemokine levels in serum at 2 months, and at the last time point 2–7 years later. The levels of most innate immune mediators were generally higher at the last visit, suggesting that these levels may initially be reduced 2 months after acute illness. In contrast, mediators associated with Th1 and Th17 responses generally decreased over time, whereas the levels of B cell mediators remained steady after the 2-month visit (Table 2, Figure S1).
In three patients with TBE and concomitant Lyme neuroborreliosis, the levels of cytokines/chemokines in serum and their dynamics seemed comparable to those found in patients without coinfection (data not shown).

3.2. Association of Cytokine and Chemokine Levels and Outcome of TBE

Since several inflammatory mediators were associated with more severe acute illness [12], we next assessed the potential association between cytokine and chemokine levels in CSF or serum at the initial visit and in serum at follow-up time points (2 months and 2–7 years thereafter) with the clinical outcome of TBE, i.e., with the presence of PES 2–7 years after TBE.
Patients with PES at last visit and those who had a favorable outcome had very similar levels of the individual cytokines/chemokines at the time of acute illness in CSF and serum, and at the 2-month follow-up time point in serum (Figure 1, Figure 2 and Figure 3 and Table 3). None of the mediators was statistically significantly associated with the outcome after correcting the analyses for multiple testing. Thus, based on our data, the cytokines/chemokines levels in CSF or serum within the first 2 months after the onset of TBE did not have predictive value for identifying patients with unfavorable outcome 2–7 years later.
However, associations between the levels of inflammatory mediators and unfavorable outcomes were observed at the last visit (Table 3 and Figure 4). At the last visit, 2–7 years after TBE, the levels of Th17 mediators in serum were generally lower in patients with PES. In contrast, the levels of most mediators associated with innate, Th1 or B cell responses were higher for patients with PES at the 2–7 year time point.

4. Discussion

Although most patients recover within weeks following TBE, in some cases objective neurological signs endure and approximately one third of adult patients report persistent symptoms such as fatigue, arthralgias and myalgias, headache, dizziness, sleep disorders, emotional lability, memory and concentration disorders, etc., termed PES. The underlying causes of this syndrome remain unclear. A possible explanation for PES is that these symptoms are due to inappropriate activation of host immune responses following TBEV infection.
A literature search revealed several reports that have characterized the levels of inflammatory mediators during the meningoencephalitic phase of TBE [12,15,16,17,18,19,20,21,22,23,24,25,26]. Several of these studies demonstrated an association between heightened inflammatory markers and greater disease severity during acute illness [12,19,23,24]. However, only two previous studies evaluated the association of inflammation and disease outcome after TBE. In a study on 44 adult patients with TBE, no correlation between the IFN-γ, TNF-α, IL-10 or IL-6 concentration and the (long-term) outcome was found [19]. In contrast, a study of children (n = 22) with TBE, which correlated the levels of 16 cytokines and chemokines and 5 markers of neuronal damage in CSF with long-term outcomes, suggested that the mechanism underlying the central nervous system pathology and TBE sequelae is related to the degree of inflammation in CNS and that high levels of IFN-γ, IL-4, IL-6 and IL-8 in CSF during acute illness might indicate a risk for incomplete recovery in childhood TBE [27].
In our current study we comprehensively characterized the inflammatory immune responses in serum and CSF in TBE patients who were followed systematically from acute infection to PES, to better understand the role of the immune responses in the clinical course and outcome of TBE. For this purpose, a cohort of 81 patients who presented with TBE were followed systematically for up to 7 years. Matched serum and CSF samples were collected at the first visit and serum samples were collected at each subsequent visit and correlated with clinical information to identify the type of immune responses related to persistent symptoms associated with PES. Although many cytokines and chemokines may have overlapping functions, we classified them according to the particular immune response they are most commonly associated with to help interpret the results.
We expected that 2 months after TBE, when all patients were afebrile and the vast majority had improved substantially, their inflammatory immune responses would return to baseline. Indeed, we identified statistically significant differences in the levels of mediators associated with innate, Th1, Th17, and B cell responses between the acute phase and 2 months later. Of interest, statistically significant differences were also observed for the levels of many of these mediators between 2-month time point and 2–7 years after TBE. These results suggest that inflammatory imbalance after TBE is rather long-lasting and would need further assessment. One of the possible explanations for these surprising findings is selection bias due to the larger proportion of patients with PES 2–7 years after TBE (53%) in our group of 81 patients in comparison to the cohort of 420 patients assessed for clinical outcome (33%) [11]. However, differences in serum values of cytokines/chemokines in patients who fulfilled the criteria for PES and those who did not were small and not statistically significant.
In the present study, the cytokines/chemokines levels in CSF or serum obtained at the time of acute illness as well as serum levels of the inflammatory mediators obtained 2 months after the onset of TBE did not have predictive value for early identification of patients who may have an unfavorable outcome 2–7 years later. However, associations between immune mediators and PES were observed at the last time point (2–7 years after TBE). Of interest, the levels of Th17 mediators in serum tended to be lower for patients with PES at the last time point compared to those whose symptoms resolved, suggesting that more robust Th17 responses may be helpful in disease resolution. In contrast, cytokines/chemokines representing the other immune response groups were higher in patients with PES, supporting the notion that inappropriate immune responses may contribute to adverse clinical outcomes.
Dysregulated Th17 immunity plays an important role in autoimmune disease, presumably by leading to inappropriate inflammatory immune responses that are associated with tissue pathology, which is implicated in the pathogenesis of several inflammatory and autoimmune diseases including multiple sclerosis and rheumatoid arthritis [28,29,30,31,32,33]. However, Th17 immunity is also important in the control of extracellular pathogens, and may also shape responses to viral infections, both directly and indirectly. The type of Th17 immunity that develops, either pathogenic or protective, is thought to be shaped by the type of stimulus and also by the composition and duration of exposure to the local inflammatory milieu. In patients with Lyme borreliosis, a tick-borne spirochetal infection, Th17 responses appear to be protective during early infection. However, prolonged exposure to high levels of Th17 inflammatory mediators appears to lead to post-treatment complications, including post-Lyme borreliosis symptoms after erythema migrans, and autoimmune phenomena in patients with antibiotic-refractory Lyme arthritis [34]. In viral infections, Th17 responses could cause immunopathology directly by secreting potent pro-inflammatory cytokines and activating local myeloid cells, and indirectly by inhibition of cytotoxic T lymphocyte responses, thereby enhancing viral replication. It is of interest that in the present study the levels of Th17 mediators were not higher but lower or equal in patients having PES in comparison to those with a favorable outcome after TBE, while the levels of several mediators representing the innate or Th1 adaptive responses were typically higher in patients with PES. These findings would suggest that in TBE, robust Th17 responses may play a protective role, whereas prolonged and/or excessive innate or Th1 adaptive responses may be disadvantageous.
Our study has several limitations. Although the design of the present study enabled insights into the innate or acquired (T and B cells) immune responses and the correlation of these responses with the clinical course and outcome of TBE, CSF was available only at initial visit but not at follow-up visits. Thus, if the inflammatory changes associated with the unfavorable outcome of TBE were occurring predominantly in the central nervous system (CSF) and not in serum, we might miss essential information for the elucidation of the pathogenesis of unfavorable outcome of TBE. For practical and ethical reasons, the access to CSF specimens is limited. In addition, follow-up serum samples in the present study were limited to the time points at 2 months and 2–7 years after TBE. Although we anticipated that inflammatory immune responses would return to baseline 2 months after TBE (these differences were indeed established), we also observed substantial differences in cytokine/chemokine levels between 2 months and 2–7 years after TBE. Assessment of inflammatory mediator levels at additional times, for example at 6 months and/or 12 months after TBE would be needed to ascertain more precisely when the inflammatory responses subsided. Moreover, comparison of serum inflammatory levels in patients with TBE and in healthy control subjects would facilitate the interpretation of findings. Although several statistically significant differences were found comparing serum levels of cytokines/chemokines at different time points, the levels were relatively modest and the interpretation of the biological relevance of the differences remains questionable. Finally, although the number of patients included in the present study (n = 81) was substantially higher than in previous reports, this number is still relatively lower than required to overcome the large number of variables in human disease, which could limit the ability to detect significant differences.
In conclusion, our findings demonstrate that infection with TBEV triggers a broad spectrum local (CSF) and systemic (serum) immune response that may take months to years to return to homeostasis. Moreover, dysregulation of these responses years after TBE, as observed with lower Th17 responses but higher innate and Th1 responses, is associated with unfavorable outcomes in patients with PES. These findings provide new insights into the immunopathogenesis of TBE and implicate certain inflammatory immune responses with PES years after the initial infection.

Supplementary Materials

The following are available online at https://www.mdpi.com/2076-2607/7/11/514/s1. Figure S1: Concentrations of inflammatory immune markers in serum of patients with tick-borne encephalitis at the time of acute illness (up to 14 days from the beginning of meningoencephalitic phase of the disease), 2 months later, and 2–7 years after the onset of the disease. Concentrations are in ng/mL for IL-17F, IL-23, IL-25 and IL-27, and in pg/mL for all other inflammatory immune mediators. Last FU = last follow-up visit (2–7 years after the onset of tick-borne encephalitis).

Author Contributions

Conceptualization, P.B., K.S. and F.S.; methodology, P.B., M.K., S.L.-F., K.R.-R., M.P. and T.A.-Ž.; formal analysis, L.L., P.B., K.S. and F.S.; data curation, L.L., P.B., K.S. and F.S.; writing – original draft preparation, P.B., F.S. and K.S.; writing – review and editing, P.B., F.S., L.L., M.K., S.L.-F, K.R.-R., M.P., T.A.-Ž. and K.S.; funding acquisition, F.S. and K.S.

Funding

This research was funded by the Slovenian Research Agency, grant number P3-0296 and by grants from Global Lyme Alliance and MGH-ECOR.

Conflicts of Interest

The authors Petra Bogovič, Lara Lusa, Miša Korva, Katarina Resman-Rus, Miša Pavletič, Stanka Lotrič-Furlan, and Tatjana Avšič-Županc declare no conflict of interest. Klemen Strle served on the scientific advisory board for Roche for development of Lyme disease serological diagnostics and received support by grants from Global Lyme Alliance and MGH-ECOR. Franc Strle served on the scientific advisory board for Roche on Lyme disease serological diagnostics, received research support from the Slovenian Research Agency [grant number P3-0296], and is an unpaid member of the steering committee of the ESCMID Study Group on Lyme Borreliosis/ESGBOR. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Dobler, G.; Erber, W.; Bröker, M.; Schmitt, H.J. The TBE Book, 2nd ed.; Global Health Press: Singapore, 2019; ISBN 978-981-14-0914-1. [Google Scholar]
  2. Dumpis, U.; Crook, D.; Oksi, J. Tick-borne encephalitis. Clin. Infect. Dis. 1999, 28, 882–890. [Google Scholar] [CrossRef] [PubMed]
  3. Bogovic, P.; Strle, F. Tick-borne encephalitis: A review of epidemiology, clinical characteristics, and management. World J. Clin. Cases 2015, 3, 430–441. [Google Scholar] [CrossRef] [PubMed]
  4. Taba, P.; Schmutzhard, E.; Forsberg, P.; Lutsar, I.; Ljøstad, U.; Mygland, Å.; Levchenko, I.; Strle, F.; Steiner, I. EAN consensus review on prevention, diagnosis and management of tick-borne encephalitis. Eur. J. Neurol. 2017, 24, 1214–1261. [Google Scholar] [CrossRef] [PubMed]
  5. Bogovič, P.; Lotrič-Furlan, S.; Avšič-Županc, T.; Lusa, L.; Strle, F. Factors associated with severity of tick-borne encephalitis: A prospective observational study. Travel Med. Infect. Dis. 2018, 26, 25–31. [Google Scholar] [CrossRef] [PubMed]
  6. 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]
  7. Li, H.; Li, Y.; Wen, B.; Zhang, J.; Wang, C.; Song, Z.; Li, S.; Qu, X.; Huang, R.; Liu, W. Dengue virus and Japanese encephalitis virus infection of the central nervous system share similar profiles of cytokine accumulation in cerebrospinal fluid. Cent. Eur. J. Immunol. 2017, 42, 218–222. [Google Scholar] [CrossRef]
  8. Mittal, M.; Ansari, M.J.; Abrar, S.; Pandey, A.K. Cytokine Profile in JE and non JE Encephalitis. Int. J. Sci. Res. (IJSR) 2016, 5, 1112–1117. [Google Scholar]
  9. Kalita, J.; Srivastava, R.; Mishra, M.K.; Basu, A.; Misra, U.K. Cytokines and chemokines in viral encephalitis: a clinicoradiological correlation. Neurosci. Lett. 2010, 473, 48–51. [Google Scholar] [CrossRef]
  10. Winter, P.M.; Dung, N.M.; Loan, H.T.; Kneen, R.; Wills, B.; Thu, L.T.; House, D.; White, N.J.; Farrar, J.J.; Hart, C.A.; et al. Proinflammatory cytokines and chemokines in humans with Japanese encephalitis. J. Infect. Dis. 2004, 190, 1618–1626. [Google Scholar] [CrossRef]
  11. Bogovič, P.; Stupica, D.; Rojko, T.; Lotrič-Furlan, S.; Avšič-Županc, T.; Kastrin, A.; Lusa, L.; Strle, F. The long-term outcome of tick-borne encephalitis in Central Europe. Ticks Tick-Borne Dis. 2018, 9, 369–378. [Google Scholar] [CrossRef]
  12. Bogovič, P.; Lusa, L.; Korva, M.; Pavletič, M.; Rus, K.R.; Lotrič-Furlan, S.; Avšič-Županc, T.; Strle, K.; Strle, F. Inflammatory Immune Responses in the Pathogenesis of Tick-Borne Encephalitis. J. Clin. Med. 2019, 8, 731. [Google Scholar] [CrossRef]
  13. Westfall, P.H.; Young, S.S. Resampling-Based Multiple Testing; Wiley: New York, NY, USA, 1993; ISBN 978-0-471-55761-6. [Google Scholar]
  14. R Core Team. A Language and Environment for Statistical Computing; R Foundation for Statistical Computing: Vienna, Austria, 2019; Available online: http://www.R-project.org (accessed on 20 June 2019).
  15. Kondrusik, M.; Pancewicz, S.; Zajkowska, J.; Hermanowska-Szpakowicz, T. Tumor necrosis factor alpha and interleukin 1-beta in serum of patients with tick-borne encephalitis. Pol. Merkur. Lekarski. 2001, 11, 26–28. [Google Scholar] [PubMed]
  16. Atrasheuskaya, A.V.; Fredeking, T.M.; Ignatyev, G.M. Changes in immune parameters and their correction in human cases of tick-borne encephalitis. Clin. Exp. Immunol. 2003, 131, 148–154. [Google Scholar] [CrossRef] [PubMed]
  17. Lepej, S.Z.; Misić-Majerus, L.; Jeren, T.; Rode, O.D.; Remenar, A.; Sporec, V.; Vince, A. Chemokines CXCL10 and CXCL11 in the cerebrospinal fluid of patients with tick-borne encephalitis. Acta Neurol. Scand. 2007, 115, 109–114. [Google Scholar] [CrossRef] [Green Version]
  18. Zajkowska, J.; Moniuszko-Malinowska, A.; Pancewicz, S.A.; Muszyńska-Mazur, A.; Kondrusik, M.; Grygorczuk, S.; Swierzbińska-Pijanowska, R.; Dunaj, J.; Czupryna, P. Evaluation of CXCL10, CXCL11, CXCL12 and CXCL13 chemokines in serum and cerebrospinal fluid in patients with tick borne encephalitis (TBE). Adv. Med. Sci. 2011, 56, 311–317. [Google Scholar] [CrossRef]
  19. Günther, G.; Haglund, M.; Lindquist, L.; Forsgren, M.; Andersson, J.; Andersson, B.; Sköldenberg, B. Tick-borne encephalitis is associated with low levels of interleukin-10 in cerebrospinal fluid. Infect. Ecol. Epidemiol. 2011, 1. [Google Scholar] [CrossRef]
  20. Cerar, T.; Ogrinc, K.; Lotric-Furlan, S.; Kobal, J.; Levicnik-Stezinar, S.; Strle, F.; Ruzić-Sabljic, E. Diagnostic value of cytokines and chemokines in Lyme neuroborreliosis. Clin. Vaccine Immunol. 2013, 20, 1578–1584. [Google Scholar] [CrossRef]
  21. Palus, M.; Formanová, P.; Salát, J.; Žampachová, E.; Elsterová, J.; Růžek, D. Analysis of serum levels of cytokines, chemokines, growth factors, and monoamine neurotransmitters in patients with tick-borne encephalitis: identification of novel inflammatory markers with implications for pathogenesis. J. Med. Virol. 2015, 87, 885–892. [Google Scholar] [CrossRef]
  22. Pietikäinen, A.; Maksimow, M.; Kauko, T.; Hurme, S.; Salmi, M.; Hytönen, J. Cerebrospinal fluid cytokines in Lyme neuroborreliosis. J. Neuroinflamm. 2016, 13, 273. [Google Scholar] [CrossRef]
  23. Grygorczuk, S.; Parczewski, M.; Świerzbińska, R.; Czupryna, P.; Moniuszko, A.; Dunaj, J.; Kondrusik, M.; Pancewicz, S. The increased concentration of macrophage migration inhibitory factor in serum and cerebrospinal fluid of patients with tick-borne encephalitis. J. Neuroinflamm. 2017, 14, 126. [Google Scholar] [CrossRef] [Green Version]
  24. Grygorczuk, S.; Świerzbińska, R.; Kondrusik, M.; Dunaj, J.; Czupryna, P.; Moniuszko, A.; Siemieniako, A.; Pancewicz, S. The intrathecal expression and pathogenetic role of Th17 cytokines and CXCR2-binding chemokines in tick-borne encephalitis. J. Neuroinflamm. 2018, 15, 115. [Google Scholar] [CrossRef] [PubMed]
  25. Markowicz, M.; Schötta, A.M.; Kundi, M.; Bogovič, P.; Ogrinc, K.; Strle, F.; Stanek, G. CXCL13 concentrations in cerebrospinal fluid of patients with Lyme neuroborreliosis and other neurological disorders determined by Luminex and ELISA. Ticks Tick-Borne Dis. 2018, 9, 1137–1142. [Google Scholar] [CrossRef] [PubMed]
  26. Koper, O.M.; Kamińska, J.; Grygorczuk, S.; Zajkowska, J.; Kemona, H. CXCL9 concentrations in cerebrospinal fluid and serum of patients with tick-borne encephalitis. Arch. Med. Sci. 2018, 14, 313–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Fowler, Å.; Ygberg, S.; Bogdanovic, G.; Wickström, R. Biomarkers in Cerebrospinal Fluid of Children With Tick-borne Encephalitis: Association With Long-term Outcome. Pediatr. Infect. Dis. J. 2016, 35, 961–966. [Google Scholar] [CrossRef] [PubMed]
  28. Bettelli, E.; Korn, T.; Oukka, M.; Kuchroo, V.K. Induction and effector functions of T(H)17 cells. Nature 2008, 453, 1051–1057. [Google Scholar] [CrossRef] [PubMed]
  29. Gaffen, S.L.; Jain, R.; Garg, A.V.; Cua, D.J. The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat. Rev. Immunol. 2014, 14, 585–600. [Google Scholar] [CrossRef] [PubMed]
  30. Duerr, R.H.; Taylor, K.D.; Brant, S.R.; Rioux, J.D.; Silverberg, M.S.; Daly, M.J.; Steinhart, A.H.; Abraham, C.; Regueiro, M.; Griffiths, A.; et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 2006, 314, 1461–1463. [Google Scholar] [CrossRef]
  31. Australo-Anglo-American Spondyloarthritis Consortium (TASC); Reveille, J.D.; Sims, A.M.; Danoy, P.; Evans, D.M.; Leo, P.; Pointon, J.J.; Jin, R.; Zhou, X.; Bradbury, L.A.; et al. Genome-wide association study of ankylosing spondylitis identifies non-MHC susceptibility loci. Nat. Genet. 2010, 42, 123–127. [Google Scholar] [CrossRef]
  32. Liu, Y.; Helms, C.; Liao, W.; Zaba, L.C.; Duan, S.; Gardner, J.; Wise, C.; Miner, A.; Malloy, M.J.; Pullinger, C.R.; et al. A genome-wide association study of psoriasis and psoriatic arthritis identifies new disease loci. PLoS Genet. 2008, 4, e1000041. [Google Scholar] [CrossRef]
  33. Wellcome Trust Case Control Consortium; Australo-Anglo-American Spondylitis Consortium (TASC); Burton, P.R.; Clayton, D.G.; Cardon, L.R.; Craddock, N.; Deloukas, P.; Duncanson, A.; Kwiatkowski, D.P.; McCarthy, M.I.; et al. Association scan of 14,500 nonsynonymous SNPs in four diseases identifies autoimmunity variants. Nat. Genet. 2007, 39, 1329–1337. [Google Scholar] [CrossRef]
  34. Strle, K.; Stupica, D.; Drouin, E.E.; Steere, A.C.; Strle, F. Elevated levels of IL-23 in a subset of patients with post-lyme disease symptoms following erythema migrans. Clin. Infect. Dis. 2014, 58, 372–380. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Levels of cytokines/chemokines in cerebrospinal fluid obtained during the meningoencephalitic phase of tick-borne encephalitis according to the presence or absence of post-encephalytic syndrome at the last follow-up visit (2–7 years after acute illness). Concentrations are in ng/mL for IL-17F, IL-23, IL-25 and IL-27, and in pg/mL for all other inflammatory immune mediators. PES last FU = post-encephalitic syndrome present at last follow-up visit.
Figure 1. Levels of cytokines/chemokines in cerebrospinal fluid obtained during the meningoencephalitic phase of tick-borne encephalitis according to the presence or absence of post-encephalytic syndrome at the last follow-up visit (2–7 years after acute illness). Concentrations are in ng/mL for IL-17F, IL-23, IL-25 and IL-27, and in pg/mL for all other inflammatory immune mediators. PES last FU = post-encephalitic syndrome present at last follow-up visit.
Microorganisms 07 00514 g001
Figure 2. Levels of cytokines/chemokines in serum obtained during the meningoencephalitic phase of tick-borne encephalitis according to the presence or absence of post-encephalytic syndrome at the last follow-up visit (2–7 years after acute illness). Concentrations are in ng/mL for IL-17F, IL-23, IL-25 and IL-27, and in pg/mL for all other inflammatory immune mediators. PES last FU = post-encephalitic syndrome present at last follow-up visit.
Figure 2. Levels of cytokines/chemokines in serum obtained during the meningoencephalitic phase of tick-borne encephalitis according to the presence or absence of post-encephalytic syndrome at the last follow-up visit (2–7 years after acute illness). Concentrations are in ng/mL for IL-17F, IL-23, IL-25 and IL-27, and in pg/mL for all other inflammatory immune mediators. PES last FU = post-encephalitic syndrome present at last follow-up visit.
Microorganisms 07 00514 g002
Figure 3. Levels of cytokines/chemokines in serum obtained 2 months after first sampling according to the presence or absence of post-encephalytic syndrome at the last follow-up visit (2–7 years after tick-borne encephalitis). Concentrations are in ng/mL for IL-17F, IL-23, IL-25 and IL-27, and in pg/mL for all other inflammatory immune mediators. PES last FU = post-encephalitic syndrome present at last follow-up visit.
Figure 3. Levels of cytokines/chemokines in serum obtained 2 months after first sampling according to the presence or absence of post-encephalytic syndrome at the last follow-up visit (2–7 years after tick-borne encephalitis). Concentrations are in ng/mL for IL-17F, IL-23, IL-25 and IL-27, and in pg/mL for all other inflammatory immune mediators. PES last FU = post-encephalitic syndrome present at last follow-up visit.
Microorganisms 07 00514 g003
Figure 4. Levels of cytokines/chemokines in serum obtained from patients 2–7 years after tick-borne encephalitis according to the presence or absence of post-encephalytic syndrome at the same time point. Concentrations are in ng/mL for IL-17F, IL-23, IL-25 and IL-27, and in pg/mL for all other inflammatory immune mediators. PES last FU = post-encephalitic syndrome present at last follow-up visit (at the time point when levels of cytokines/chemokines were determined).
Figure 4. Levels of cytokines/chemokines in serum obtained from patients 2–7 years after tick-borne encephalitis according to the presence or absence of post-encephalytic syndrome at the same time point. Concentrations are in ng/mL for IL-17F, IL-23, IL-25 and IL-27, and in pg/mL for all other inflammatory immune mediators. PES last FU = post-encephalitic syndrome present at last follow-up visit (at the time point when levels of cytokines/chemokines were determined).
Microorganisms 07 00514 g004
Table 1. Basic demographic, clinical, and laboratory data on the acute illness for 81 adult patients with tick-borne encephalitis in whom levels of cytokines and chemokines in serum and cerebrospinal fluid were determined.
Table 1. Basic demographic, clinical, and laboratory data on the acute illness for 81 adult patients with tick-borne encephalitis in whom levels of cytokines and chemokines in serum and cerebrospinal fluid were determined.
CharacteristicNumber (%, 95% CI) or Median (IQR)
Male sex40 (49.4; 38.1–60.7)
Age (years)56 (43–63)
Males58 (43–62.5)
Females55 (45–63)
Underlying illnesses39 (48.1; 36.9–59.5)
Monophasic course of illness 35 (43.2; 32.2–54.7)
Clinical presentation
Meningitis33 (40.7; 30.0–52.2)
Meningoencephalitis40 (49.4; 38.1–60.7)
Meningoencephalomyelitis8 (9.9; 4.4–18.5)
Treatment in intensive care unit7 (8.6; 3.6–17.0)
Duration (days)5 (3.5–9)
Artificial ventilation: number; duration (days)2 (28.6; 3.7–71.0); 6 (5–7)
Duration of illness before CSF and first blood sample obtained (days) a5 (3–6) b
CSF leukocyte count (× 106 cells/L)76 (37–134)
CSF protein concentration (g/L)0.70 (0.53–0.91)
Elevated (> 0.45 g/L) 67 (82.7; 72.7–90.2)
Concomitant Lyme neuroborreliosis c3/78 (3.8; 0.8–10.8)
Positive B. burgdorferi sensu lato IgG antibodies d7/78 (9.0; 3.7–17.6)
a In patients with a biphasic course of illness, this figure was based on the time period from the beginning of the second (meningoencephalitic) phase until hospitalization. b Data available for 77 patients. c Diagnosis of concomitant Lyme neuroborreliosis was based on intrathecal synthesis of Borrelia burgdorferi sensu lato specific IgG or IgM antibodies or isolation of B. burgdorferi sensu lato from CSF. d The sole indication of borrelial infection. CI, confidence interval; IQR, interquartile range; CSF, cerebrospinal fluid.
Table 2. Serum concentrations of cytokines/chemokines obtained in patients with tick-borne encephalitis early in the meningoencephalic phase of the illness, and at follow-up visits 2 months and 2–7 years after tick-borne encephalitis.
Table 2. Serum concentrations of cytokines/chemokines obtained in patients with tick-borne encephalitis early in the meningoencephalic phase of the illness, and at follow-up visits 2 months and 2–7 years after tick-borne encephalitis.
Cytokine/ChemokineConcentrations (pg/mL) Median (IQR)Ratio of Medians
ME Phase of TBE2 m Later2–7 y Later2 m/0 mpa ValueAdjusted p a Value 2–7 y/2 mpb ValueAdjusted p b Value
Innate
GMCSF0.86
(0.47–1.68)
0.865
(0.31–1.68)
0.72
(0.26–1.68)
1.010.9561.0000.830.4230.999
IFNα3.58
(0.30–10.69)
0.71
(0.30–6.16)
0.71
(0.30–10.69)
0.20<0.0010.0031.000.0460.511
IL-1β0.48
(0.24–0.92)
0.32
(0.23–0.69)
0.84
(0.29–3.37)
0.670.0060.0802.63<0.001<0.001
IL-60.30
(0.30–1.86)
0.30
(0.30–0.76)
0.86
(0.30–14.32)
1.000.0280.3322.87<0.001<0.001
IL-85.52
(1.05–25.07)
1.91
(0.62–15.06)
108.22
(23.67–314.44)
0.350.0470.50256.66<0.001<0.001
TNFα12.01
(5.38–26.84)
6.43
(4.37–16.39)
39.32
(14.98–60.02)
0.54<0.0010.0026.12<0.001<0.001
MCP1117.30
(74.27–185.77)
206.25
(158.30–259.33)
223.39
(180.29–269.72)
1.76<0.001<0.0011.080.0690.646
MIP1α1.01
(1.01–30.24)
1.01
(1.01–8.80)
59.21
(17.18–129.04)
1.000.0140.18058.62<0.001<0.001
IL-100.23
(0.23–0.23)
0.23
(0.23–0.23)
0.23
(0.23–0.23)
1.000.7891.0001.000.7891.000
Th1
IFNγ1.1
9(0.45–2.83)
1.05
(0.45–2.43)
0.45
(0.17–1.48)
0.880.3500.9990.43<0.001<0.001
IL-12P401.18
(1.18–1.18)
1.18
(1.18–1.18)
1.18
(1.18–1.18)
1.001.0001.0001.000.3710.999
IL-12P700.38
(0.33–0.92)
0.38
(0.33–0.92)
0.33
(0.33–0.72)
1.000.6471.0000.870.0110.153
CXCL917.42
(10.22–28.51)
22.21
(11.67–40.55)
24.01
(12.11–40.78)
1.280.0020.0231.080.7711.000
CXCL1088.34
(56.41–171.02)
91.1
9(59.58–147.12)
101.97
(69.51–147.12)
1.0320.6241.0001.120.5861.000
CCL1911.82
(6.21–21.16)
24.08
(11.42–31.48)
20.07
(11.98–28.47)
2.04<0.001<0.0010.830.0510.543
Th17
IL-17F c0.32
(0.32–0.32)
0.32
(0.32–0.32)
0.32
(0.32–0.47)
1.000.2890.9971.00<0.001d0.002
IL-17A0.31
(0.31–0.31)
0.31
(0.31–0.31)
0.31
(0.31–0.31)
1.000.0180.2311.000.3640.999
IL-220.21
(0.21–0.45)
0.22
(0.21–0.47)
0.21
(0.21–0.34)
1.020.3190.9990.980.0580.588
IL-215.70
(2.71–14.11)
2.71
(2.71–4.75)
20.31
(11.35–27.41)
0.48<0.001<0.0017.49<0.001<0.001
IL-23 c0.25
(0.25–0.25)
0.25
(0.25–0.25)
0.25
(0.25–0.25)
1.000.2240.9881.000.1820.961
IL-25 c0.32
(0.32–0.32)
0.32
(0.32–0.32)
0.32
(0.32–0.32)
1.000.2810.9971.001.0001.000
IL-27 c2.63
(1.70–3.86)
1.52
(0.90–2.12)
0.94
(0.59–1.54)
0.58<0.001<0.0010.62<0.001<0.001
B cell
CXCL1222.40
(16.30–30.84)
26.24
(19.40–36.08)
34.36
(25.30–40.26)
0.780.0030.0401.310.1690.935
CXCL1339.26
(24.44–61.32)
30.74
(18.98–52.88)
33.59
(23.29–41.51)
1.170.0750.6831.09<0.0010.005
The Wilcoxon signed rank test with continuity correction was used for comparisons of individual cytokines/chemokines; univariate p values were adjusted for multiple comparisons (adjusted p) with a multivariate permutation procedure. p values for significantly different concentrations are shown in bold. a Comparison of findings during acute illness and 2 months later. b Comparison of findings obtained at visit 2 months and 2–7 years after TBE. c Concentrations are given in ng/mL. d Medians and IQRs are the same but the other data (in the 1st and 4th quartiles) are not. IQR, interquartile range; ME, meningoencephalitic; TBE, tick-borne encephalitis; m, month, y, year.
Table 3. Correlation of cytokine and chemokine levels obtained at the time of acute illness, 2 months later, and 2–7 years later with the outcome of tick-borne encephalitis 2–7 years after acute illness.
Table 3. Correlation of cytokine and chemokine levels obtained at the time of acute illness, 2 months later, and 2–7 years later with the outcome of tick-borne encephalitis 2–7 years after acute illness.
Cytokine/ChemokineUnfavorable Outcome * of Tick-Borne Encephalitis 2–7 Years after Acute Illness
CSF, Time 0Serum, Time 0Serum, Time 2 MonthsSerum, Time 2–7 Years
Median Ratio a
(PES/Others)
p Value bAdjusted p Value cMedian Ratio a
(PES/Others)
p Value bAdjusted p Value cMedian Ratio a
(PES/Others)
p Value bAdjusted p Value cMedian Ratio a
(PES/Others)
p Value bAdjusted p Value c
Innate
GMCSF0.840.72810.990.74211.400.378110.9431
IFNα10.573110.627110.598110.9141
IL-1β0.930.99610.570.0040.2150.830.92111.540.5301
IL-61.480.206110.598110.41113.100.3321
IL-81.080.89110.400.538110.94411.520.1911
TNFα1.110.97410.780.0850.9980.830.39611.310.5901
MCP11.010.91411.070.34211.120.23411.100.3481
MIP1α0.560.0210.7690.080.0690.99510.84012.020.2351
IL-1010.910110.623110.969110.9581
Th1
IFNγ1.370.274110.84210.610.425110.5981
IL-12P400.820.247110.958110.149110.3601
IL-12P701.070.636110.93811.520.11711.150.3851
CXCL91.370.34911.340.30211.150.58111.550.0830.998
CXCL101.010.57310.890.91310.900.31211.200.4241
CCL191.650.0610.9901.480.13011.180.63811.300.0260.837
Th17
IL-17F1NANA10.316110.70910.910.0790.998
IL-17A1NANA10.298110.0210.76710.4051
IL-2210.13510.860.19310.820.790110.0540.982
IL-2110.93110.470.0340.91510.57310.740.0310.879
IL-231NANA10.222110.734110.1591
IL-251NANA10.120110.344110.1351
IL-271.140.76911.190.59611.090.65710.830.5251
B cell
CXCL120.440.51310.900.36411.000.86311.170.3391
CXCL131.120.69111.020.74410.910.34111.070.2301
* Unfavorable outcome was defined by the presence of post-encephalitic syndrome. a Mann-Whitney tests were used to compare the levels of the examined cytokines/chemokines at first visit and follow-up times in the group of patients with and without post-encephalitic syndrome at the last follow-up visit. We reported the fold change of the medians (defined as the ratio of medians observed in the group of patients with post-encephalitic syndrome and in the group without post-encephalitic syndrome at the last follow-up visit; values > 1 indicate greater median concentrations in the group with post-encephalitic syndrome. b p values from the Mann-Whitney tests. c p values from the Mann-Whitney tests adjusted for multiple comparisons. CSF, cerebrospinal fluid; NA, not available (because no variability in concentrations was present).

Share and Cite

MDPI and ACS Style

Bogovič, P.; Lusa, L.; Korva, M.; Lotrič-Furlan, S.; Resman-Rus, K.; Pavletič, M.; Avšič-Županc, T.; Strle, K.; Strle, F. Inflammatory Immune Responses in Patients with Tick-Borne Encephalitis: Dynamics and Association with the Outcome of the Disease. Microorganisms 2019, 7, 514. https://doi.org/10.3390/microorganisms7110514

AMA Style

Bogovič P, Lusa L, Korva M, Lotrič-Furlan S, Resman-Rus K, Pavletič M, Avšič-Županc T, Strle K, Strle F. Inflammatory Immune Responses in Patients with Tick-Borne Encephalitis: Dynamics and Association with the Outcome of the Disease. Microorganisms. 2019; 7(11):514. https://doi.org/10.3390/microorganisms7110514

Chicago/Turabian Style

Bogovič, Petra, Lara Lusa, Miša Korva, Stanka Lotrič-Furlan, Katarina Resman-Rus, Miša Pavletič, Tatjana Avšič-Županc, Klemen Strle, and Franc Strle. 2019. "Inflammatory Immune Responses in Patients with Tick-Borne Encephalitis: Dynamics and Association with the Outcome of the Disease" Microorganisms 7, no. 11: 514. https://doi.org/10.3390/microorganisms7110514

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