Next Article in Journal
Stable Enzymatic Nanoparticles from Nucleases, Proteases, Lipase and Antioxidant Proteins with Substrate-Binding and Catalytic Properties
Next Article in Special Issue
A Comprehensive Review of Risk Factors for Venous Thromboembolism: From Epidemiology to Pathophysiology
Previous Article in Journal
Transcriptome-Based Traits of Radioresistant Sublines of Non-Small Cell Lung Cancer Cells
Previous Article in Special Issue
Thrombosis Models: An Overview of Common In Vivo and In Vitro Models of Thrombosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Finding the Needle in the Haystack: Serological and Urinary Biomarkers in Behçet’s Disease: A Systematic Review

1
Department of Clinical and Biological Sciences, School of Specialization of Clinical Pathology, University of Turin, 10124 Turin, Italy
2
Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-Net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit, San Giovanni Bosco Hub Hospital, University of Turin, 10124 Turin, Italy
*
Author to whom correspondence should be addressed.
These authors have contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(3), 3041; https://doi.org/10.3390/ijms24033041
Submission received: 15 November 2022 / Revised: 16 January 2023 / Accepted: 17 January 2023 / Published: 3 February 2023
(This article belongs to the Special Issue New Advances in Thrombosis)

Abstract

:
Urinary and serological markers play an essential role in the diagnostic process of autoimmune diseases. However, to date, specific and reliable biomarkers for diagnosing Behçet’s disease (BD) are still lacking, negatively affecting the management of these patients. To analyze the currently available literature on serological and urinary BD biomarkers investigated in the last 25 years, we performed a systematic literature review using the Population, Intervention, Comparison, and Outcomes (PICO) strategy. One hundred eleven studies met the eligibility criteria (6301 BD patients, 5163 controls). Most of them were retrospective, while five (5%) were prospective. One hundred ten studies (99%) investigated serological biomarkers and only two (2%) focused on urinary biomarkers. One hundred three studies (93%) explored the diagnostic potential of the biomolecules, whereas sixty-two (56%) tested their effect on disease activity monitoring. Most articles reported an increase in inflammatory markers and pro-oxidant molecules, with a decrease in antioxidants. Promising results have been shown by the omics sciences, offering a more holistic approach. Despite the vast number of investigated markers, existing evidence indicates a persistent gap in BD diagnostic/prognostic indices. While new steps have been taken in the direction of pathogenesis and disease monitoring, international efforts for the search of a diagnostic marker for BD are still needed.

1. Introduction

Behçet’s disease (BD) is a multisystemic inflammatory condition often described as a part of the vasculitic spectrum, whose etiology, although not fully characterized, is attributed to a complex inter-relationship between the genetic background and the dysregulation of both the innate and the adaptive immune system [1]. Females and males are equally affected, with a worse disease progression in males due to ocular, vascular, and neurological involvement [2]. Diagnosis onset is collocated between 25 and 30 years old, although countries with a low disease prevalence may show a delayed time of diagnosis [3].
The distribution of BD is widespread; however, it is more prevalent in countries along the ancient “Silk Road”, from the Mediterranean area to the far east, where it is associated with the distribution of the major histocompatibility complex antigen HLA-B51 [4].Even though the first description of BD dates back to 1937, its diagnosis still relies entirely on clinical criteria [5], and a laboratory test to help identify patients with BD is still lacking. Unfortunately, the most common symptoms of BD, including oro-genital aphthae, skin lesions, arthritis, and uveitis, overlap with other autoimmune diseases, such as inflammatory bowel conditions or connective tissue diseases, and the differential diagnosis may become a real challenge [6,7]. Moreover, the disease course tends to be considerably prolonged, and it may take months or years before all the typical signs and symptoms appear. Unfortunately, for most patients, an early diagnosis of BD can be an unrealistic goal and having one or more biomarkers of BD could drastically change how BD is diagnosed and ultimately help clinical evaluation. A biomarker is a measurable characteristic of the body that may indicate a particular biological state or condition [8]. Biomarkers are employed in many fields of medicine, such as disease diagnosis, disease activity evaluation, prognosis, and therapy monitoring. Since the 1950s, new biomarkers for BD have been studied to be applicable to all populations in which the disease is prevalent. Still, there is no consensus on a shared biomarker for BD to be evaluated by testing in the diagnostic routine. Recently, omics sciences have helped solve this diagnostic gap in multiple diseases using a promising holistic approach, but at the moment, they are not yet integrated into standard clinical care [9,10]. A system based on omics sciences is also needed in BD—on the one hand, for diagnosing BD patients early; and, on the other hand, for identifying different profiles of BD patients based on their disease activity, prognosis, and response to therapy. In order to contribute to this growing field of research, this study aimed to systematically review the currently available literature on the identification and characterization of the clinical utility of serological and urinary BD biomarkers investigated in the last 25 years.

2. Methods

2.1. Literature Search Strategy

A detailed literature search screening Ovid MEDLINE, In-Process and Other Non-Indexed Citation, the National Library of Medicine’s (NLM), and the in-process database for Ovid MEDLINE, from inception to November 2021, was performed a priori to identify original articles analyzing the diagnostic role of urinary and serological biomarkers in BD. The Population, Intervention, Comparison, and Outcomes strategy (PICO) was adopted to identify the best keywords to use in database queries. The following keywords and medical subject heading (MESH) terms were used in all possible combinations using Boolean operators: Behçet’s syndrome; retinal vasculitis; biomarkers; inflammation mediators; immune checkpoint proteins; pathogen-associated molecular pattern molecules.

2.2. Selection of the Studies

We screened and selected full-text articles, analyzing the titles and abstracts. After the first screening phase, we evaluated the selected abstracts and the full texts to determine eligibility. Papers retrieved by the literature search but reporting insufficient data according to the chosen PICO strategy were excluded. The online search was limited to case-control, cohort, and case-series studies. Studies with a small sample size (n < 20), conference abstracts, reviews, and animal studies were excluded. Articles written in languages other than English were excluded. The selection and inclusion criteria were determined a priori.
We considered studies eligible if they met the following inclusion criteria:
Studies that included at least 20 patients diagnosed with BD following the current International Study Group Criteria [5];
Studies that analyzed urinary biomarkers, serological biomarkers, or both;
Ex vivo studies (in vitro studies were excluded).
Four independent reviewers (MA, DM, PM, and LR) systematically analyzed the abstracts and full texts of the articles meeting the inclusion criteria; any disagreements were resolved by consensus. If consensus could not be achieved, a third party (MR) provided an assessment of eligibility. As the data on eligibility were dichotomous (eligible: yes/no), agreement at both the title and abstract review and the full article review stages was determined by the calculation of Cohen’s kappa coefficient (k > 8). We performed the present study according to the PRISMA guidelines [11].

2.3. Data Extraction and Data Synthesis

Data were extracted in an electronic database, summarized, analyzed, and discussed. For each study, the following data were identified: study design, country of origin, type of biomarker, methods used for detection, sample size, pathergy and HLAB51 positivity, type of involvement (systemic/organ-specific), different marker concentrations in BD, and controls and measures of association. The homogeneity of studies was assessed per each diagnostic maker. Quantitative synthesis was considered inappropriate due to the heterogeneity among studies in the population set, the type of biomarker analyzed, and the methods used for the identifications used in different studies. Therefore, a qualitative narrative synthesis was performed.

3. Results and Discussion

3.1. Systematic Literature Search

We retrieved 637 articles from the initial search (Figure 1).
Three hundred forty-four studies were excluded after the title and abstract screening because they did not fit the selection criteria described above. We further assessed twohundred ninety-three studies for eligibility. We excluded one hundred eighty-two studies because they did not meet the inclusion criteria, were not focused on biomarkers, did not reach statistically significant results, or were not in English. Finally, one hundred eleven articles were eligible for the qualitative synthesis.
Figure 2 shows the number of studies per year included in this systematic review. Furthermore, Table 1 displays the main characteristics of the analyzed studies, including the number of patients, study design, biomarkers tested, and accuracy.
A total of 6301 patients with BD (1813 with active, 1543 with inactive BD, and 2945 cases in which the activity of BD was not addressed in the study) met the inclusion criteria and were further analyzed. There were 5163 included controls, consisting of 4171 healthy controls (HC) and 992 patients with autoimmune diseases (such as SLE, AR, SS, multiple sclerosis, and vasculitis). Most studies were retrospective, whereas six had a prospective design.
Considering the extensive geographical diffusion of BD, we analyzed the countries of origin in which all included studies was performed. The global map of Figure 3 shows the publication rate of the analyzed studies per country: Turkey and South Korea were the most represented countries. Interestingly, it is possible to identify the characteristic spread of BD studies along the ancient Silk Road.

3.2. Biomarkers and Their Roles in Diagnosis and Disease Activity

A total of 110 studies (99%) investigated serological biomarkers, while only two (2%) tested their population with urinary biomarkers. Most of the included studies (103; 93%) were designed to investigate the diagnostic potential of the biomolecules, while 62 (56%) tested their ability to differentiate between different stages of disease activity. A comprehensive view of all the examined markers is given in Table 2. The most important are cited in the following paragraphs.

3.2.1. Conventional Inflammation Markers and Soluble Proteins

The erythrocyte sedimentation rate (ESR) and C reactive protein (CRP), two inflammation indices, have been assessed by 14 (13%) and 19 (17%) studies on BD, respectively. An increase in their values has been reported with a total agreement rate among the articles.
The neutrophil-to-lymphocyte ratio (NLR) is a parameter analyzed through a hemocytometer. It has been investigated as a biomarker in seven (6%) articles; all the studies reported a significant increase in the NLR in BD patients, especially in patients with active disease.
Tumor necrosis factor-alpha (TNF-α) is a cytokine that regulates the immune system, inflammatory response, and apoptosis. Serum TNF-α has been analyzed in eight studies (7%). Its levels were remarkably increased in BD patients compared to healthy controls, whereas there have been inconclusive results on the correlation between high TNF-α levels and BD activity [29,49,58,61,69,81,103,117].
In the sub-group of interleukins (ILs), fifteen different molecules have been studied as serological markers in 22 studies (20%). Among them, IL-8 had increased levels in BD sera compared to controls in three different studies (3%) [50,51,75], with high rates of sensibility and specificity in differentiating active and inactive patients, as reported in four articles (4%) [19,36,50,51].Moreover, IL-6 has been investigated as a serological marker in six articles (5%), highlighting a potential in BD diagnosing but not in the activity disease classification.
Adenosine deaminase (ADA) has been the main focus of three studies (3%). ADA is a marker of T-lymphocyte activation, whose serological levels were found to be markedly elevated in BD patients compared to controls [28,34,40].
Anti-alpha enolase antibodies (AAEA) have been evaluated in three studies (3%). They consist of a heterogeneous group of antibodies directed toward surface proteins in endothelial cells, which have been found to increase in many inflammatory diseases, including SLE, AR, and vasculitis. Additionally, in this case, the serological levels of both IgG and IgM AAEA seemed to be significantly elevated in BD patients, particularly during the active phase [60,92,107].

3.2.2. Oxidant and Anti-Oxidant Molecules

Reactive oxygen species (ROS), including nitric oxide (NO), are products of oxidative stress and are usually released in inflammatory sites by the innate immune response and endothelial cells. In seven articles (6%), the authors described the NO levels to be significantly enhanced in the serum and urine of BD patients compared to HC [24,26,27,29,32,102]. Significant differences were noticed in patients with active disease in comparison to inactive patients [24,26,27,102,122].
Malondialdehyde (MDA), one of the final products of lipid peroxidation triggered by the free radicals of oxidative stress, was reported to be elevated in BD sera in comparison to HC, even if it was not a promising biomarker of BD activity of disease [38,56,102].
Super oxide dismutase (SOD) and catalase are anti-inflammatory enzymes involved in oxidative stress. The dosages of SOD and RBC catalase levels [28,102] showed significant reductions in BD patients, especially in samples collected during the phase of disease activity.

3.2.3. microRNAs

Several miRNAs (including miR-93, miR-106b, miR-25, miR-146a, miR-326, and miR-181b) were assessed by three studies (3%) included in this systematic review. In particular, in a case-control study of 47 BD patients [103], the authors observed that the miR-155 levels increased in BD patients compared to HC. However, these results were not corroborated by the authors of two other studies [104,106], where a conspicuous decrease in miR-155 levels was conversely observed when testing their BD patients.

3.2.4. New “Omics” Sciences

Two studies (2%) have addressed the serum metabolomics state of BD patients, starting with an untargeted approach and subsequently validating a specific panel of biomarkers on an independent cohort.
Through the gas chromatography/time-of-flight mass spectrometry GC/TOF-MS, Ahn and colleagues isolated a panel of five metabolites (decanoic acid, fructose, tagatose, linoleic acid, and oleic acid) able to differentiate BD patients from HC with high sensitivity and specificity, at 100% and 97.1%, respectively [125]. Concurrently, Zheng et al. observed that high serum levels of two polyunsaturated fatty acids (PUFAs), linoleic acid (LA) and arachidonic acid (AA), discriminated BD patients and HC efficiently with high sensitivity (95% for PUFAs and 95% for LA) and specificity (65 for PUFAs and 88% for LA) [110].
In their previous work, Ahn and colleagues also assessed the urinary metabolomic profiles of BD patients. The authors identified a combination of metabolites (guanine, pyrrole-2-carboxylate, 3-hydroxypyridine, mannose, L-citrulline, galactonate, isothreonate, sedoheptuloses, hypoxanthine, and gluconic acid lactone) able to identify BD patients with high sensitivity (96.7%) and specificity (93.3%) [93].
Two studies (2%) have widely investigated the serum proteomic asset using matrix-assisted laser desorption ionization–time-of-flight mass spectrometry (MALDI-TOF-MS). A first model based on 39 proteins could distinguish BD and HC with a sensitivity of 83.67% and a specificity of 89.87% [65]. The second study detected significant upregulation of fibrin, apolipoprotein A-IV, and serum amyloid A (SAA) in the sera of BD patients with active disease at the intestinal level compared to controls [94].

4. Discussion

BD is a rare multisystemic vasculitis whose symptoms and signs often overlap with other autoimmune diseases, leading to delayed diagnosis and occasionally inappropriate therapy. The pathogenesis of BD has not been fully elucidated. However, the dysregulation of the innate and acquired immune systems in a facilitative environment plays a crucial role in disease development [2] (Figure 4).
Further, unlike other autoimmune diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis (AR), or other vasculitis, specific biomarkers for BD have not yet been identified, negatively affecting the early diagnosis and management of BD patients.
In this systematic review, we carefully reviewed all the relevant articles published in the current literature to identify the international efforts made in identifying specific serological and urinary BD biomarkers.
Considering the well-known inflammatory nature of BD, most studies have shown an increase in inflammatory biomarkers in BD patients, such as CRP, ESR, and numerous cytokines, including TNFα, IL-1β, IL-6, IL-8, IL-17, and IL-23 (Table 2).
Unfortunately, despite a high agreement rate among the articles, their lack of specificity makes them a nonoptimal diagnostic tool, whereas they can be helpful in disease monitoring.
To date, there is consensus on the involvement of lymphocytes in the BD pathogenesis, in particular, T helper cells that produce IL-17 (Th17) and T regulatory (Tregs) cells [106,126,127]. In the presence of IL-23, Th naive cells differentiate in the Th17 phenotype and migrate at mucosal surfaces, where, through the secretion of IL-17, they induce the recruitment of neutrophils and activate epithelial cells, mediating the inflammatory process [128]. Conversely, Treg cells play a role in inhibiting the immune response triggered by the resident microflora in the mucosa by the secretion of TGF-β and IL-10 [129]. Interestingly, Th17 and Tregs share common pathways during differentiation, and their cell count is fundamental for maintaining the balance between pro-inflammatory and anti-inflammatory conditions in mucosal tissues [128]. Multiple studies on BD have reported a decrease in Treg cells, alongside an upregulation of Th17 cells and neutrophils (both as an absolute value and NLR) [76,80,84,87,106,116,122,123]. Activated neutrophils may reach the inflammatory sites, triggering a substantial oxidative stress response by releasing ROS, which can contribute to the disease progression over time. It is worth mentioning that increased levels of numerous pro-oxidants were observed in BD sera and urines; among them, ADA NO, advanced oxidation protein products (AOPPs), and some of the final products of lipid peroxidation, such as MDA and thiobarbituric acid-reactive substances (TBARS) [28,31,34,40,102] (Table 2). On the contrary, many studies have described lower anti-oxidant levels in the sera of BD patients, such as catalase and SOD, confirming the dysregulation of the production of pro-oxidants and anti-oxidant substances in BD [28,102]. However, similarly to the inflammation biomarkers, pro- and anti-oxidants remain aspecific and could be used for monitoring BD patients but, due to their low specificity, do not have a pivotal role in BD diagnosis.
MiRNAs are small non-coding RNA (19-23-nucleotide length) that inhibit translation by binding mRNAs. Recently, some miRNAs have been investigated as putative BD markers. In particular, low serum levels of miR-155 were detected in active BD patients, and higher levels were observed during disease remission [104,106]. It is known that miR-155 is involved in switching off the inflammatory response by downregulating IL-6 and IL-1β and upregulating IL-10, an inhibitory interleukin [130]. In fact, high serum levels of IL-6 and IL-1β and low levels IL-10 were observed in active BD [14,75,102,106,115,122].
Moreover, the role of miR-155 in blocking BD progression was confirmed by the increase in Th17 and the release of IL-17. These mechanisms are mediated by the inhibition of E26 transformation-specific-1 (ETS-1), a gene upregulated in BD [106,115,131]. One could hypothesize that lower levels of miR-155 might lead to low CD4+ T cells, Th17, and IL17 and increased ETS-1 during active BD [131]. However, Kolhai et al. reported an increase in the miR-155 levels in BD patients, in concomitance with a reduction of Ets-1 and an elevation of Th17 cells, suggesting a pro-inflammatory role and a potential therapeutic target [103]. While the current scientific interest is focused on miRNAs for improving our understanding of BD pathogenesis, their potential in diagnostic testing for BD remains to be elucidated.
Considering that BD disease is often described as an ensemble of phenotypes with different clinical characteristics, a future challenge could be to test if these phenotypes exhibit different miRNA patterns [132]. This could not only improve our knowledge about pathogenic processes underlying the various phenotypes but could also represent a step toward a more tailored therapeutic approach.
To date, new “omics“ science, such as proteomics and metabolomics, has provided a comprehensive analysis of endogenous proteins and metabolites. With the use of metabolomics, one can potentially detect the alterations of physiological and pathological metabolites at the early stages of the disease due to its excellent sensitivity. In BD, two metabolomic tests have been developed and subsequently validated with reported high specificity and sensitivity [110,125]. Unfortunately, although this approach seems to be very promising, these tests are extremely expensive and complex, and therefore, are far from being routinely available for diagnostic or follow-up testing.
In addition to metabolome investigations from blood samples, many studies have recently focused on analyzing the fecal metabolome alterations, resulting from changes in the gut microbial communities in BD patients [133,134]
Since the intercorrelation between diet and gut microbiota is well known, studying intestinal altered metabolic profiles and the microbial community imbalance of BD patients is paving the way to new therapeutic approaches based on nutritional interventions [135].
We acknowledge that this study suffers from some limitations, mainly due to the vast heterogeneity of the included studies regarding the number of patients, control groups, and the types of biomarkers and assays used. Moreover, although it was possible to include only BD diagnosed following the ISGBD criteria, there was no standard disease activity score in the past. Only recently has a consensus on a common definition of Behçet’s disease activity been reached by developing and validating the Behçet’s Disease Current Activity Form (BDCAF) score [124]; however, it is not objectionable because it is a subjective score based on referred symptoms. For these reasons, a meta-analysis of the studies could not be performed.

5. Conclusions

In conclusion, despite the enormous efforts from the scientific community to identify potential biomarkers in BD, much more work must be done. While identifying novel aspecific biomarkers might help us better understand BD’s pathogenesis and might also find a place for monitoring disease activity during follow-up, we are still far from identifying potential diagnostic biomarkers for this complex and rare disease. Proteomics, metabolomics, and microbiome analysis might help in the near future to identify potential candidates to help researchers and ultimately clinicians to better identify patients suffering from BD.

Author Contributions

Conceptualization, M.A., M.R., P.M. and S.S.; methodology, M.A., M.R., D.M., P.M. and L.R.; formal analysis, M.A., M.R., S.S., A.B., S.G.F. and I.C.; data curation, M.A., M.R., D.M., P.M. and L.R.; writing—original draft preparation, M.A., M.R. and D.M.; writing—review and editing, A.B., S.G.F., I.C., E.M., D.R. and S.S.; supervision, E.M., D.R. and S.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Leccese, P.; Alpsoy, E. Behçet’s Disease: An Overview of Etiopathogenesis. Front. Immunol. 2019, 10, 1067. [Google Scholar] [CrossRef] [PubMed]
  2. Bettiol, A.; Prisco, D.; Emmi, G. Behçet: The syndrome. Rheumatology 2020, 59, 101–107. [Google Scholar] [CrossRef] [PubMed]
  3. Oguz, I.D.; Hizli, P.; Gonul, M. The Epidemiology of Behçet’s Disease. In Behcet’s Disease; Gonul, M., Kartal, S.P., Eds.; BoD—Books on Demand: Norderstedt, Germany, 2017. [Google Scholar] [CrossRef]
  4. Alpsoy, E. Behçet’s Disease: A Comprehensive Review with a Focus on Epidemiology, Etiology and Clinical Features, and Management of Mucocutaneous Lesions. J. Dermatol. 2016, 43, 620–632. [Google Scholar] [CrossRef] [PubMed]
  5. International Study Group for Behçet’s Disease. Criteria Fordiagnosis of Behçet’s Disease. Lancet 1990, 335, 1078–1080. [Google Scholar]
  6. Kim, D.H.; Cheon, J.H. Intestinal Behçet’s Disease: A True Inflammatory Bowel Disease or Merely an Intestinal Complication of Systemic Vasculitis? Yonsei Med. J. 2016, 57, 22–32. [Google Scholar] [CrossRef]
  7. Valenti, S.; Gallizzi, R.; De Vivo, D.; Romano, C. Intestinal Behçet and Crohn’s Disease: Two Sides of the Same Coin. Pediatr. Rheumatol. Online J. 2017, 15, 1–8. [Google Scholar] [CrossRef]
  8. Shi, G.; Zhang, Z.; Li, Q. New Biomarkers in Autoimmune Disease. J. Immunol. Res. 2017, 2017, 1–4. [Google Scholar] [CrossRef]
  9. Guma, M.; Tiziani, S.; Firestein, G.S. Metabolomics in Rheumatic Diseases: Desperately Seeking Biomarkers. Nat. Rev. Rheumatol. 2016, 12, 269–281. [Google Scholar] [CrossRef]
  10. Giacomelli, R.; Afeltra, A.; Alunno, A.; Bartoloni-Bocci, E.; Berardicurti, O.; Bombardieri, M.; Bortoluzzi, A.; Caporali, R.; Caso, F.; Cervera, R.; et al. Guidelines for Biomarkers in Autoimmune Rheumatic Diseases—Evidence Based Analysis. Autoimmun. Rev. 2019, 18, 93–106. [Google Scholar] [CrossRef]
  11. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
  12. Wickham, H. 2016. ggplot2: Elegant Graphics for Data Analysis, Springer-Verlag New York. Available online: https://ggplot2.tidyverse.org/ (accessed on 16 January 2023).
  13. R Core Team, version 0.98.1091. 2014. Available online: https://support.posit.co/hc/en-us/articles/200716783-RStudio-IDE-Release-History (accessed on 16 January 2023).
  14. Yosipovitch, G.; Shohat, B.; Bshara, J.; Wysenbeek, A.; Weinberger, A. Elevated Serum Interleukin 1 Receptors and Interleukin 1B in Patients with Behçet’s Disease: Correlations with Disease Activity and Severity. Isr. J. Med. Sci. 1995, 31, 345–348. [Google Scholar] [CrossRef]
  15. Deǧer, O.; Örem, A.; Akyol, N.; Bahadir, S.; Yildirmiş, S. Polymorphonuclear leukocyte elastase levels in patients with Behçet’s disease. Clin. Chim. Acta 1995, 236, 129–134. [Google Scholar] [CrossRef]
  16. Direskeneli, H.; Keser, G.; D’Cruz, D.; Khamashta, M.A.; Akoglu, T.; Yazici, H.; Yurdakul, S.; Hamuryudan, V.; Özgün, S.; Goral, A.J.; et al. Anti-endothelial cell antibodies, endothelial proliferation and von Willebrand factor antigen in Behçet’s disease. Clin. Rheumatol. 1995, 14, 55–61. [Google Scholar] [CrossRef]
  17. Uslu, T.; Erem, C.; Tosun, M.; Deger, O. Plasma endothelin-1 levels in Behçet’s disease. Clin. Rheumatol. 1997, 16, 59–61. [Google Scholar] [CrossRef]
  18. Çayirli, E.A.; Er, H.; Yilmaz, E. The Levels of Plasma Interleukin-2 and Soluble Interleukin-2R in Behçet’s Disease: A Marker of Disease Activity. J. Dermatol. 1998, 25, 513–516. [Google Scholar] [CrossRef]
  19. Katsantonis, J.; Adler, Y.; Orfanos, C.E.; Zouboulis, C.C. Adamantiades-Behçet’s Disease: Serum IL-8 Is a More Reliable Marker for Disease Activity than C-Reactive Protein and Erythrocyte Sedimentation Rate. Dermatology 2000, 201, 37–39. [Google Scholar] [CrossRef]
  20. Eksioglu-Demiralp, E.; Direskeneli, H.; Ergun, T.; Fresko, I.; Akoglu, T. Increased CD4 + CD16 + and CD4 + CD56 + T cell subsets in Behçet’s disease. Rheumatol. Int. 1999, 19, 23–26. [Google Scholar] [CrossRef]
  21. Freysdottir, J.; Lau, S.-H.; Fortune, F. γδ T cells in Behçet’s disease (BD) and recurrent aphthous stomatitis (RAS). Clin. Exp. Immunol. 1999, 118, 451–457. [Google Scholar] [CrossRef]
  22. Krause, I.; Monselise, Y.; Milo, G.; Weinberger, A. Anti-Saccharomyces cerevisiae Antibodies. Adamantiades-Behçet’s Dis. 2003, 528, 201–204. [Google Scholar] [CrossRef]
  23. Evereklioglu, C.; Inaloz, H.; Kirtak, N.; Doganay, S.; Bulbul, M.; Otlu, B.; Ozerol, E.; Er, H.; Ozbek, E. Serum leptin concentration is increased in patients with Behcet’s syndrome and is correlated with disease activity. Br. J. Dermatol. 2002, 147, 331–336. [Google Scholar] [CrossRef]
  24. Er, H.; Evereklioglu, C.; Cumurcu, T.; Türköz, Y.; Özerol, E.; Şahin, K.; Daganay, S. Serum Homocysteine Level Is Increased and Correlated with Endothelin-1 and Nitric Oxide in Behçet’s Disease. Br. J. Ophthalmol. 2002, 86, 653–657. [Google Scholar] [CrossRef] [PubMed]
  25. Saglam, K.; Yilmaz, I.M.; Saglam, A.; Ulgey, M.; Bulucu, F.; Baykal, Y. Levels of circulating intercellular adhesion molecule-1 in patients with Behçet’s disease. Rheumatol. Int. 2001, 21, 146–148. [Google Scholar] [CrossRef] [PubMed]
  26. Evereklioglu, C.; Turkoz, Y.; Er, H.; Inaloz, H.S.; Ozbek, E.; Cekmen, M. Increased Nitric Oxide Production in Patients with Behçet’s Disease: Is It a New Activity Marker? J. Am. Acad. Dermatol. 2002, 46, 50–54. [Google Scholar] [CrossRef] [PubMed]
  27. Evereklioglu, C.; Özbek, E.; Çekmen, M.; Mehmet, N.; Duygulu, F.; Ozkiris, A.; Çalip, M.; Er, H.; Türköz, Y. Urinary Nitric Oxide Levels Are Increased and Correlated with Plasma Concentrations in Patients with Behçet’s Disease: Is It a New Urinary Activity Marker? Nephrology. 2003, 8, 231–238. [Google Scholar] [CrossRef]
  28. Erkiliç, K.; Evereklioglu, C.; Çekmen, M.; Özkiris, A.; Duygulu, F.; Dogan, H. Adenosine Deaminase Enzyme Activity Is Increased and Negatively Correlates with Catalase, Superoxide Dismutase and Glutathione Peroxidase in Patients with Behçet’s Disease: Original Contributions/Clinical and Laboratory Investigations. Mediat. Inflamm. 2003, 12, 107–116. [Google Scholar] [CrossRef]
  29. Akdeniz, N.; Esrefoglu, M.; Keles, M.; Karakuzu, A.; Atasoy, M. Serum Interleukin-2, Interleukin-6, Tumour Necrosis Factor-Alpha and Nitric Oxide Levels in Patients With Behçet’s Disease. Ann. Acad. Med. 2004, 33, 596–599. [Google Scholar]
  30. Sari, R.A.; Kiziltunc, A.; Tays’Y, S.; Akdem’Yr, S.; Gündogdu, M. Levels of soluble E-selectin in patients with active Behçet’s disease. Clin. Rheumatol. 2004, 24, 55–59. [Google Scholar] [CrossRef]
  31. Yazici, C.; Köse, K.; Çaliş, M.; Demir, M.; Kirnap, M.; Ateş, F. Increased Advanced Oxidation Protein Products in Behçet’s Disease: A New Activity Marker? Br. J. Dermatol. 2004, 151, 105–111. [Google Scholar] [CrossRef]
  32. Duygulu, F.; Evereklioglu, C.; Calis, M.; Borlu, M.; Çekmen, M.; Ascioglu, O. Synovial Nitric Oxide Concentrations Are Increased and Correlated with Serum Levels in Patients with Active Behçet’s Disease: A Pilot Study. Clin. Rheumatol. 2005, 24, 324–330. [Google Scholar] [CrossRef]
  33. Ureten, K.; Ertenli, I.; Oztürk, M.A.; Kiraz, S.; Onat, A.M.; Tuncer, M.; Okur, H.; Akdogan, A.; Apras, S.; Calgüneri, M. Neutrophil CD64 expression in Behçet’s disease. J. Rheumatol. 2005, 32(5), 849–852. [Google Scholar] [PubMed]
  34. Calis, M.; Ates, F.; Yazici, C.; Kose, K.; Kirnap, M.; Demir, M.; Borlu, M.; Evereklioglu, C. Adenosine Deaminase Enzyme Levels, Their Relation with Disease Activity, and the Effect of Colchicine on Adenosine Deaminase Levels in Patients with Behçet’s Disease. Rheumatol. Int. 2005, 25, 452–456. [Google Scholar] [CrossRef]
  35. Qiao, H.; Sonoda, K.-H.; Ariyama, A.; Kuratomi, Y.; Kawano, Y.-I.; Ishibashi, T. CXCR2 Expression on Neutrophils is Upregulated During the Relapsing Phase of Ocular Behçet Disease. Curr. Eye Res. 2005, 30, 195–203. [Google Scholar] [CrossRef]
  36. Gür-Toy, G.; Lenk, N.; Yalcin, B.; Aksaray, S.; Alli, N. Serum Interleukin-8 as a Serologic Marker of Activity in Behçet’s Disease. Int. J. Dermatol. 2005, 44, 657–660. [Google Scholar] [CrossRef]
  37. Basak, C.; Yunus, S.; Ahmet, G.; Ilker, E.; Naci, C. Activation Markers in Behçet’s Disease. SKINmed. Dermatol. Clin. 2005, 4, 282–286. [Google Scholar] [CrossRef]
  38. Yardim-Akaydin, S.; Sepici, A.; Özkan, Y.; Şimşek, B.; Sepici, V. Evaluation of Allantoin Levels as a New Marker of Oxidative Stress in Behçet’s Disease. Scand. J. Rheumatol. 2006, 35, 61–64. [Google Scholar] [CrossRef]
  39. Köse, O.; Arca, E.; Akgül, Ö.; Erbil, K. The levels of serum neopterin in Behçet’s disease—Objective marker of disease activity. J. Dermatol. Sci. 2006, 42, 128–130. [Google Scholar] [CrossRef]
  40. Canpolat, F.; Ünver, M.; Eskioǧlu, F.; Kösebalaban, Ş.; Durmazlar, S.P.K. Serum and Erythrocyte Adenosine Deaminase Activities in Patients with Behçet’s Disease. Int. J. Dermatol. 2006, 45, 1053–1056. [Google Scholar] [CrossRef]
  41. Kwon, S.R.; Lim, M.J.; Park, S.G.; Moon, Y.S.; Park, W. Decreased protein S activity is related to the disease activity of Behcet’s disease. Rheumatol. Int. 2006, 27, 39–43. [Google Scholar] [CrossRef]
  42. Briani, C.; Doria, A.; Marcolongo, R.; Tognon, S.; Ruggero, S.; Toffanin, E.; Ermani, M.; Ghirardello, A.; Zampieri, S.; Semenzato, G. Increased titres of IgM anti-heparan sulfate antibody in Behçet’s disease. Ann. Rheum. Dis. 2006, 24. [Google Scholar]
  43. Sarican, T.; Ayabakan, H.; Turkmen, S.; Kalaslioglu, V.; Baran, F.; Yenice, N. Homocysteine: An activity marker in Behçet’s disease? J. Dermatol. Sci. 2007, 45, 121–126. [Google Scholar] [CrossRef]
  44. Lee, Y.J.; Kang, S.W.; Song, J.K.; Park, J.J.; Bae, Y.D.; Lee, E.Y.; Song, Y.W. Serum galectin-3 and galectin-3 binding protein levels in Behçet’s disease and their association with disease activity. Ann. Rheum. Dis. 2007, 25. [Google Scholar]
  45. Pay, S.; Abbasov, T.; Erdem, H.; Musabak, U.; Simsek, I.; Pekel, A.; Akdogan, A.; Sengul, A.; Dinc, A. Serum MMP-2 and MMP-9 in Patients with Behçet’s Disease: Do Their Higher Levels Correlate to Vasculo-Behçet’s Disease Associated with Aneurysm Formation? Clin. Exp. Rheumatol. 2007, 25 Suppl. 45, S70–S75. [Google Scholar] [PubMed]
  46. Öztürk, M.A.; Ünverdi, S.; Oktar, S.O.; Bukan, N.; Gülbahar, Ö.; Üreten, K.; Göker, B.; Haznedaroglu, S.; Sungur, G.; Çiftçi, T.U.; et al. Vascular endothelial growth factor and carotid intima-media thickness in patients with Behçet’s disease. Clin. Rheumatol. 2008, 27, 961–966. [Google Scholar] [CrossRef] [PubMed]
  47. Turan, B.; Pfister, K.; Diener, P.; Hell, M.; Möller, B.; Boyvat, A.; Ergin, S.; Villiger, P.M. Soluble tumour necrosis factor receptors sTNFR1 and sTNFR2 are produced at sites of inflammation and are markers of arthritis activity in Behçet’s disease. Scand. J. Rheumatol. 2008, 37, 135–141. [Google Scholar] [CrossRef]
  48. Kutlay, S.; Calayoglu, R.; Boyvat, A.; Turkcapar, N.; Sengul, S.; Keven, K.; Nergizoglu, G. Circulating endothelial cells: A disease activity marker in Behçet’s vasculitis? Rheumatol. Int. 2008, 29, 159–162. [Google Scholar] [CrossRef]
  49. Curnow, S.J.; Pryce, K.; Modi, N.; Knight, B.; Graham, E.M.; Stewart, J.E.; Fortune, F.; Stanford, M.R.; Murray, P.I.; Wallace, G.R. Serum Cytokine Profiles in Behçet’s Disease: Is There a Role for IL-15 in Pathogenesis? Immunol. Lett. 2008, 121, 7–12. [Google Scholar] [CrossRef]
  50. Polat, M.; Vahaboglu, G.; Önde, U.; Eksioglu, M. Classifying Patients with Behçet’s Disease for Disease Severity, Using a Discriminating Analysis Method. Clin. Exp. Dermatol. 2009, 34, 151–155. [Google Scholar] [CrossRef]
  51. Durmazlar, S.P.K.; Ulkar, G.B.; Eskioglu, F.; Tatlican, S.; Mert, A.; Akgul, A. Significance of Serum Interleukin-8 Levels in Patients with Behcet’s Disease: High Levels May Indicate Vascular Involvement. Int. J. Dermatol. 2009, 48, 259–264. [Google Scholar] [CrossRef]
  52. Habibagahi, Z.; Habibagahi, M.; Heidari, M. Raised concentration of soluble form of vascular endothelial cadherin and IL-23 in sera of patients with Behçet’s disease. Mod. Rheumatol. 2009, 20, 154–159. [Google Scholar] [CrossRef]
  53. Fadini, G.P.; Tognon, S.; Rodriguez, L.; Boscaro, E.; Baesso, I.; Avogaro, A.; Semenzato, G.; Agostini, C.; Marcolongo, R. Low levels of endothelial progenitor cells correlate with disease duration and activity in patients with Behçet’s disease. Ann. Rheum. Dis. 2009, 27. [Google Scholar]
  54. Choe, J.-Y.; Park, S.-H.; Kim, S.-K. Serum angiopoietin-1 level is increased in patients with Behçet’s disease. Jt. Bone Spine 2010, 77, 340–344. [Google Scholar] [CrossRef]
  55. Donmez, A.; Aksu, K.; Aydın, H.; Keser, G.; Cagirgan, S.; Doganavsargil, E.; Tombuloglu, M. The plasma levels of activated thrombin activatable fibrinolysis inhibitor and thrombomodulin in Behçet Disease and their association with thrombosis. Thromb. Res. 2010, 126, 207–210. [Google Scholar] [CrossRef]
  56. Sezer, E.D.; Aksu, K.; Caglayan, O.; Keser, G.; Karabulut, G.; Ercan, G. DNA Damage and Its Relationship with Other Oxidative Stress Parameters in Behcet’s Disease. Rheumatol. Int. 2012, 32, 217–222. [Google Scholar] [CrossRef]
  57. Özden, M.; Çaycı, Y.; Tekin, H.; Çoban, A.; Aydın, F.; Şentürk, N.; Bek, Y.; Cantürk, T.; Turanlı, A. Serum galectin-3 levels in patients with Behçet’s disease: Association with disease activity over a long-term follow-up. J. Eur. Acad. Dermatol. Venereol. 2011, 25, 1168–1173. [Google Scholar] [CrossRef]
  58. Pehlivan, Y.; Onat, A.M.; Okumuş, S.; Kisacik, B.; Comez, G.; Comez, A.; Goktepe, F.; Ceylan, N.O. The Role of Resistin in Behçet’s Disease: A Turkish Experiment. Mod. Rheumatol. 2012, 22, 571–575. [Google Scholar] [CrossRef]
  59. Ahn, J.K.; Hwang, J.-W.; Oh, J.-M.; Bae, E.-K.; Lee, J.; Lee, Y.S.; Koh, E.-M.; Cha, H.-S. Increased α-defensin-1 expression in Korean patients with Behcet’s disease. Jt. Bone Spine 2011, 78, 593–597. [Google Scholar] [CrossRef]
  60. Shin, S.J.; Kim, B.C.; Il Kim, T.; Lee, S.K.; Lee, K.H.; Kim, W.H. Anti-Alpha-Enolase Antibody as a Serologic Marker and Its Correlation with Disease Severity in Intestinal Behçet’s Disease. Dig. Dis. Sci. 2011, 56, 812–818. [Google Scholar] [CrossRef]
  61. Jung, Y.S.; Kim, S.W.; Yoon, J.Y.; Lee, J.H.; Jeon, S.M.; Hong, S.P.; Il Kim, T.; Kim, W.H.; Cheon, J.H. Expression of a Soluble Triggering Receptor Expressed on Myeloid Cells-1 (STREM-1) Correlates with Clinical Disease Activity in Intestinal Behcet’s Disease. Inflamm. Bowel Dis. 2011, 17, 2130–2137. [Google Scholar] [CrossRef]
  62. Vural, B.; Uğurel, E.; Tüzün, E.; Kürtüncü, M.; Zuliani, L.; Çavuş, F.; Içöz, S.; Erdağ, E.; Gül, A.; Güre, A.O.; et al. Anti-neuronal and stress-induced-phosphoprotein 1 antibodies in neuro-Behçet’s disease. J. Neuroimmunol. 2011, 239, 91–97. [Google Scholar] [CrossRef]
  63. López-Longo, F.J.; Arias-Salgado, E.G.; Martín, M.; Jiménez-Yuste, V.; de la Rúa, A.R.; Bello, I.F.; Álvarez, M.T.; Butta, N.V. Platelet soluble CD40L and matrix metalloproteinase 9 activity are proinflammatory mediators in Behçet disease patients. Thromb. Haemost. 2012, 107, 88–98. [Google Scholar] [CrossRef]
  64. Gündüz, E.; Teke, H.; Bilge, N..Y.; Cansu, D..; Bal, C.; Korkmaz, C.; Gülbaş, Z. Regulatory T cells in Behçet’s disease: Is there a correlation with disease activity? Does regulatory T cell type matter? Rheumatol. Int. 2013, 33, 3049–3054. [Google Scholar] [CrossRef]
  65. Wang, X.; Zhang, M.F.; Xie, J.; Li, Z.L.; Wang, P. Behcet’s Disease with Active Uveitis: Detection of Serum Protein Biomarkers Using MALDI-TOF-MS. Anat. Rec. 2012, 295, 1168–1173. [Google Scholar] [CrossRef]
  66. Örem, A.; Yayli, S.; Arıca, D.; Akcan, B.; Yücesan, F.; Bahadir, S. Lipoprotein-associated phospholipase A2level in patients with Behçet’s disease. J. Eur. Acad. Dermatol. Venereol. 2012, 27, e289–e293. [Google Scholar] [CrossRef]
  67. Hamzaoui, K.; Kaabachi, W.; Fazaa, B.; Zakraoui, L.; Mili-Boussen, I.; Haj-Sassi, F. Serum IL-33 levels and skin mRNA expression in Behçet’s disease. Ann. Rheum. Dis. 2013, 31, S6–S14. [Google Scholar]
  68. Vural, B.; Şehitoğlu, E.; Çavuş, F.; Yalçınkaya, N.; Haytural, H.; Küçükerden, M.; Ulusoy, C.; Uğurel, E.; Turan, S.; Bulut, L.; et al. Mitochondrial carrier homolog 1 (Mtch1) antibodies in neuro-Behçet’s disease. J. Neuroimmunol. 2013, 263, 139–144. [Google Scholar] [CrossRef]
  69. Shaker, O.G.; Tawfic, S.O.; El-Tawdy, A.M.; El-Komy, M.H.M.; El Menyawi, M.; Heikal, A.A. Expression of TNF-α, APRIL and BCMA in Behcet’s Disease. J. Immunol. Res. 2014, 2014, 380–405. [Google Scholar] [CrossRef]
  70. Xun, Y.; Chen, P.; Yan, H.; Yang, W.; Shi, L.; Chen, G.; Du, H. Identification of prohibitin as an antigen in Behcet’s disease. Biochem. Biophys. Res. Commun. 2014, 451, 389–393. [Google Scholar] [CrossRef]
  71. Vayá, A.; Rivera, L.; Todolí, J.; Hernandez, J.L.; Laiz, B.; Ricart, J.M. Haematological, biochemical and inflammatory parameters in inactive Behçet’s disease. Its association with red blood cell distribution width. Clin. Hemorheol. Microcirc. 2014, 56, 319–324. [Google Scholar] [CrossRef]
  72. Balta, I.; Balta, S.; Koryurek, O.M.; Demirkol, S.; Mikhailidis, D.P.; Celik, T.; Cakar, M.; Kucuk, U.; Eksioglu, M.; Kurt, Y.G. Serum endocan levels as a marker of disease activity in patients with Behçet disease. J. Am. Acad. Dermatol. 2013, 70, 291–296. [Google Scholar] [CrossRef]
  73. Ozuguz, P.; Karabulut, A.A.; Tulmac, M.; Kisa, U.; Kocak, M.; Gunduz, O. Markers of Endothelial Dysfunction and Evaluation of Vascular Reactivity Tests in Behçet Disease. Angiology 2013, 65, 937–943. [Google Scholar] [CrossRef]
  74. Mejía, J.-C.; Espinosa, G.; Tàssies, D.; Reverter, J.-C.; Cervera, R. Endogenous thrombin potential in Behçet’s disease: Relationship with thrombosis and anticoagulant therapy. Ann. Rheum. Dis. 2014, 32. [Google Scholar]
  75. Lopalco, G.; Lucherini, O.M.; Vitale, A.; Talarico, R.; Lopalco, A.; Galeazzi, M.; Lapadula, G.; Cantarini, L.; Iannone, F. Putative Role of Serum Amyloid-A and Proinflammatory Cytokines as Biomarkers for Behcet’s Disease. Medicine. 2015, 94, e1858. [Google Scholar] [CrossRef]
  76. Yuksel, M.; Yildiz, A.; Oylumlu, M.; Turkcu, F.M.; Bilik, M.Z.; Ekinci, A.; Elbey, B.; Tekbas, E.; Alan, S. Novel Markers of Endothelial Dysfunction and Inflammation in Behçet’s Disease Patients with Ocular Involvement: Epicardial Fat Thickness, Carotid Intima Media Thickness, Serum ADMA Level, and Neutrophil-to-Lymphocyte Ratio. Clin. Rheumatol. 2016, 35, 701–708. [Google Scholar] [CrossRef]
  77. Bassyouni, I.H.; Sharaf, M.; Wali, I.E.; Mansour, H.M. Clinical significance of Angiopoietin-1 in Behcet’s disease patients with vascular involvement. Hear. Vessel. 2015, 31, 918–924. [Google Scholar] [CrossRef]
  78. Tulunay, A.; Dozmorov, M.G.; Ture-Ozdemir, F.; Yilmaz, V.; Eksioglu-Demiralp, E.; Alibaz-Oner, F.; Ozen, G.; Wren, J.D.; Saruhan-Direskeneli, G.; Sawalha, A.H.; et al. Activation of the JAK/STAT pathway in Behcet’s disease. Genes Immun. 2014, 16, 170–175. [Google Scholar] [CrossRef]
  79. Belguendouz, H.; Messaoudene, D.; Lahmar-Belguendouz, K.; Djeraba, Z.; Otmani, F.; Terahi, M.; Tiar, M.; Hartani, D.; Lahlou-Boukoffa, O.; Touil-Boukoffa, C. In vivo and in vitro IL-18 production during uveitis associated with Behçet disease: Effect of glucocorticoid therapy. J. Fr. Ophtalmol. 2015, 38, 206–212. [Google Scholar] [CrossRef]
  80. Ozturk, C.; Balta, S.; Balta, I.; Demirkol, S.; Celik, T.; Turker, T.; Iyisoy, A.; Eksioglu, M. Neutrophil-Lymphocyte Ratio and Carotid-Intima Media Thickness in Patients with Behçet Disease without Cardiovascular Involvement. Angiology 2015, 66, 291–296. [Google Scholar] [CrossRef]
  81. Türkcü, F.M.; Şahin, A.; Cingü, A.K.; Kaya, S.; Yüksel, H.; Cinar, Y.; Batmaz, İ. Serum Omentin, Resistin and Tumour Necrosis Factor-α Levels in Behcet Patients with and without Ocular Involvement. Graefes Arch. Clin. Exp. Ophthalmol. 2015, 253, 1565–1568. [Google Scholar] [CrossRef]
  82. de Souza, A.W.S.; Perazzio, S.F.; de França, N.R.; Andrade, L.E.C.; Bijl, M.; Westra, J.; Kallenberg, C.G.M. High mobility group box 1 serum levels are increased in Behçet’s disease, but not associated with disease activity or disease manifestations. Rheumatology 2015, 54, 2151–2155. [Google Scholar] [CrossRef]
  83. Seo, J.; Ahn, Y.; Zheng, Z.; Kim, B.; Choi, M.; Bang, D.; Kim, D. Clinical significance of serum YKL -40 in Behçet disease. Br. J. Dermatol. 2015, 174, 1337–1344. [Google Scholar] [CrossRef]
  84. Yolbas, S.; Yildirim, A.; Gozel, N.; Uz, B.; Koca, S.S. Hematological Indices May Be Useful in the Diagnosis of Systemic Lupus Erythematosus and in Determining Disease Activity in Behçet’s Disease. Med. Princ. Pract. 2016, 25, 510–516. [Google Scholar] [CrossRef] [PubMed]
  85. Hu, C.-J.; Pan, J.-B.; Song, G.; Wen, X.-T.; Wu, Z.-Y.; Chen, S.; Mo, W.-X.; Zhang, F.-C.; Qian, J.; Zhu, H.; et al. Identification of Novel Biomarkers for Behcet Disease Diagnosis Using Human Proteome Microarray Approach. Mol. Cell. Proteom. 2017, 16, 147–156. [Google Scholar] [CrossRef] [PubMed]
  86. Mejía, J.C.; Ortiz, T.; Tàssies, D.; Solanich, X.; Vidaller, A.; Cervera, R.; Reverter, J.C.; Espinosa, G. Procoagulant microparticles are increased in patients with Behçet’s disease but do not define a specific subset of clinical manifestations. Clin. Rheumatol. 2015, 35, 695–699. [Google Scholar] [CrossRef] [PubMed]
  87. Balkarli, A.; Kucuk, A.; Babur, H.; Erbasan, F. Neutrophil/Lymphocyte Ratio and Mean Platelet Volume in Behçet’ s Disease. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 3045–3050. [Google Scholar]
  88. Park, J.-S.; Kang, M.-I.; Ha, Y.-J.; Song, J.J.; Park, Y.-B.; Lee, S.-K.; Lee, S.-W. Serum anti-lysozyme is associated with disease activity of Behçet’s disease. Int. J. Rheum. Dis. 2016, 20, 261–268. [Google Scholar] [CrossRef]
  89. Cantarini, L.; Pucino, V.; Vitale, A.; Talarico, R.; Lucherini, O.M.; Magnotti, F.; De Rosa, V.; Galgani, M.; Alviggi, C.; Marone, G.; et al. Immunometabolic biomarkers of inflammation in Behçet’s disease: Relationship with epidemiological profile, disease activity and therapeutic regimens. Clin. Exp. Immunol. 2016, 184, 197–207. [Google Scholar] [CrossRef]
  90. Cure, E.; Icli, A.; Uslu, A.U.; Baykara, R.A.; Sakiz, D.; Ozucan, M.; Yavuz, F.; Arslan, S.; Cure, M.C.; Kucuk, A. Atherogenic index of plasma may be strong predictor of subclinical atherosclerosis in patients with Behçet disease. Z. Rheumatol. 2016, 76, 259–266. [Google Scholar] [CrossRef]
  91. Jiang, Y.; Zang, M.; Li, S. Serum PLR and LMR in Behçet’s disease. Medicine 2017, 96, e6981. [Google Scholar] [CrossRef]
  92. Kang, S.E.; Lee, S.J.; Lee, J.Y.; Yoo, H.J.; Park, J.K.; Lee, E.Y.; Lee, E.B.; Song, Y.W. Serum Levels of IgG Antibodies against Alpha-Enolase Are Increased in Patients with Behçet’s Disease and Are Associated with the Severity of Oral Ulcer, Erythrocyte Sedimentation Rates, and C-Reactive Protein. Clin. Exp. Rheumatol. 2017, 35, S67–S74. [Google Scholar]
  93. Ahn, J.K.; Kim, J.; Hwang, J.; Song, J.; Kim, K.H.; Cha, H.S. Urinary Metabolomic Profiling to Identify Potential Biomarkers for the Diagnosis of Behcet’s Disease by Gas Chromatography/Time-of-Flight-Mass Spectrometry. Int. J. Mol. Sci. 2017, 18, 2309. [Google Scholar] [CrossRef]
  94. Lee, H.J.; Kim, J.H.; Kim, S.W.; Joo, H.A.; Lee, H.W.; Kim, Y.S.; Park, S.J.; Hong, S.P.; Il Kim, T.; Kim, W.H.; et al. Proteomic Analysis of Serum Amyloid A as a Potential Marker in Intestinal Behçet’s Disease. Dig. Dis. Sci. 2017, 62, 1953–1962. [Google Scholar] [CrossRef]
  95. Ha, Y.-J.; Park, J.-S.; Kang, M.-I.; Lee, S.-K.; Park, Y.-B.; Lee, S.-W. Increased serum interleukin-32 levels in patients with Behçet’s disease. Int. J. Rheum. Dis. 2017, 21, 2167–2174. [Google Scholar] [CrossRef]
  96. Lopalco, G.; Lucherini, O.M.; Lopalco, A.; Venerito, V.; Fabiani, C.; Frediani, B.; Galeazzi, M.; Lapadula, G.; Cantarini, L.; Iannone, F. Cytokine Signatures in Mucocutaneous and Ocular Behçet’s Disease. Front. Immunol. 2017, 8, 200. [Google Scholar] [CrossRef]
  97. Omma, A.; Sandikci, S.C.; Colak, S.; Tecer, D.; Yücel, C.; Ozbalkan, Z. Serum calprotectin and ischemia modified albumin levels as markers of disease activity in Behçet’s disease. Adv. Dermatol. Allergol. 2018, 35, 609–613. [Google Scholar] [CrossRef]
  98. Koca, T.T. Clinical Significance of Serum Bilirubin in Behçet’s Disease. J. Transl. Intern. Med. 2018, 6, 185–188. [Google Scholar] [CrossRef]
  99. Enecik, M.; Mavi, B.; Yücel, C.; Keskin, G.; Yıldız, M. The importance of Serum Interleukin-20 Levels in Patients with Behçet’s Disease. Adv. Clin. Exp. Med. 2018, 27, 1391–1395. [Google Scholar] [CrossRef]
  100. Harmanci, K.; Akan, O.Y.; Pirildar, T.; Ortan, P.; Ulman, C. The Evaluation of the Relationship between sTREM-1, VEGF-B, and VEGF Gene Expression Levels with Disease Activity of Behçet’s Patients. Dis. Markers 2018, 2018, 1–5. [Google Scholar] [CrossRef]
  101. Lucherini, O.M.; Vitale, A.; Orlando, I.; Sota, J.; Fabiani, C.; Franceschini, R.; Simpatico, A.; Frediani, B.; Galeazzi, M.; Tosi, G.M.; et al. Serum immunoglobulin D levels in patients with Behçet’s disease according to different clinical manifestations. Ann. Rheum. Dis. 2018, 36, 110–115. [Google Scholar]
  102. Chekaoui, A.; Lahmar, K.; Belguendouz, H.; Mazari, F.; Terahi, M.; Hakem, D.; Youinou, P.; Touil-Boukoffa, C. Increased IL-1β Levels Are Associated with an Imbalance of “Oxidant/Antioxidant” Status during Behçet’s Disease. Eur. Cytokine Netw. 2018, 29, 95–102. [Google Scholar] [CrossRef]
  103. Kolahi, S.; Farajzadeh, M.J.; Alipour, S.; Abhari, A.; Farhadi, J.; Bahavarnia, N.; Malek Mahdavi, A.; Khabbazi, A.; Sakhinia, E. Determination of Mir-155 and Mir-146a Expression Rates and Its Association with Expression Level of TNF-α and CTLA4 Genes in Patients with Behcet’s Disease. Immunol. Lett. 2018, 204, 55–59. [Google Scholar] [CrossRef]
  104. Hassouna, S.S.; Tayel, M.Y.; ElKaffash, D.M.; Abdelhady, A.M.; Elsayed, E.H. MicroRNA155 Expression in Relation to BDCAF Scored Behçet’s Disease in an Egyptian Patients’ Sample. Open Rheumatol. J. 2018, 12, 115–122. [Google Scholar] [CrossRef] [PubMed]
  105. Kurtipek, G.S.; Kesli, R.; Akyurek, F.T.; Akyurek, F.; Ataseven, A.; Terzi, Y. Plasma-soluble urokinase plasminogen activator receptor (suPAR) levels in Behçet’s disease and correlation with disease activity. Int. J. Rheum. Dis. 2016, 21, 866–870. [Google Scholar] [CrossRef] [PubMed]
  106. Ahmadi, M.; Yousefi, M.; Abbaspour-Aghdam, S.; Dolati, S.; Aghebati-Maleki, L.; Eghbal-Fard, S.; Khabbazi, A.; Rostamzadeh, D.; Alipour, S.; Shabani, M.; et al. Disturbed Th17/Treg Balance, Cytokines, and MiRNAs in Peripheral Blood of Patients with Behcet’s Disease. J. Cell. Physiol. 2019, 234, 3985–3994. [Google Scholar] [CrossRef] [PubMed]
  107. Prado, L.L.; Goncalves, C.R.; Viana, V.T.; Saad, C.G.S.; Bonfa, E. Anti-Alpha-Enolase Antibodies in Behçet’s Disease: A Marker of Mucocutaneous and Articular Disease Activity? Clin. Exp. Rheumatol. 2018, 36, 28–32. [Google Scholar] [PubMed]
  108. Acikgoz, N.; Kurtoğlu, E.; Yagmur, J.; Kapicioglu, Y.; Cansel, M.; Ermis, N. Elevated Monocyte to High-Density Lipoprotein Cholesterol Ratio and Endothelial Dysfunction in Behçet Disease. Angiology 2017, 69, 65–70. [Google Scholar] [CrossRef]
  109. Hasan, M.S.; Ryan, P.L.; A Bergmeier, L.; Fortune, F. Circulating NK cells and their subsets in Behçet’s disease. Clin. Exp. Immunol. 2017, 188, 311–322. [Google Scholar] [CrossRef]
  110. Zheng, W.; Wu, X.; Goudarzi, M.; Shi, J.; Song, W.; Li, C.; Liu, J.; Chen, H.; Zhang, X.; Zeng, X.; et al. Metabolomic Alterations Associated with Behçet’s Disease. Arthritis Res. Ther. 2018, 20, 1–10. [Google Scholar] [CrossRef]
  111. Şahin, A.; Ilhan, A.; Derin, M.E.; Doğan, H.O.; Şahin, M. Low levels of pannexin-1 in Behçet’s syndrome. Int. J. Rheum. Dis. 2019, 22, 1474–1478. [Google Scholar] [CrossRef]
  112. Bassyouni, I.H.; Mohammed, W.H.S.; Taha, F.M.; El Refai, R.M. Clinical significance of CCN2/connective tissue growth factor in Behçet’s disease patients. Int. J. Rheum. Dis. 2019, 22, 1459–1465. [Google Scholar] [CrossRef]
  113. Arica, D.A.; Akşan, B.; Örem, A.; Altinkaynak, B.A.; Yayli, S.; Sönmez, M. High levels of endothelial progenitor cells and circulating endothelial cells in patients with Behçet’s disease and their relationship to disease activity. An. Bras. de Dermatol. 2019, 94, 320–326. [Google Scholar] [CrossRef]
  114. Sandikci, S.C.; Colak, S.; Omma, A.; Enecik, M.E.; Ozbalkan, Z.; Neselioglu, S.; Erel, O. An investigation of thiol/disulfide homeostasis in patients with Behçet’s disease. Arch. Med Sci. 2020, 16, 1353–1359. [Google Scholar] [CrossRef]
  115. Talaat, R.M.; Sibaii, H.; Bassyouni, I.H.; El-Wakkad, A. IL-17, IL-10, IL-6, and IFN-γ in Egyptian Behçet’s Disease: Correlation with Clinical Manifestations. Eur. Cytokine Netw. 2019, 30, 15–22. [Google Scholar] [CrossRef]
  116. Gheita, T.A.; Sakr, B.R.; Rabea, R.E.; Abd ElHamid, S.M. Value of Hematological Indices versus VEGF as Biomarkers of Activity in Behçet’s Disease. Clin. Rheumatol. 2019, 38, 2201–2210. [Google Scholar] [CrossRef]
  117. El Boghdady, N.A.; Shaker, O.G. Role of Serum MiR-181b, Proinflammatory Cytokine, and Adhesion Molecules in Behçet’s Disease. J. Interferon Cytokine Res. 2019, 39, 347–354. [Google Scholar] [CrossRef]
  118. Balbaba, M.; Ulaş, F.; Postacı, S.A.; Öz, B.; Aydın, S. Serum Cortistatin Levels in Patients with Ocular Active and Ocular Inactive Behçet Disease. Ocul. Immunol. Inflamm. 2019, 28, 601–605. [Google Scholar] [CrossRef]
  119. A Hassan, W.; Behiry, E.G.; Abdelshafy, S.; Salem, T.; A Baraka, E. Assessment of Endocan Serum Level in Patients with Behçet Disease: Relation to Disease Activity and Carotid Intima Media Thickness. Egypt J. Immunol. 2020, 27, 129–139. [Google Scholar]
  120. Hussain, M.; Chen, P.; Zhang, Y.; Tian, Y.; Du, H. Moesin expression is correlated with its involvement in patients with Behcet’s disease. Arch. Med Sci. 2020, 16, 924–930. [Google Scholar] [CrossRef]
  121. Hussain, M.; Ma, F.; Chen, P.; Tian, Y.; Du, H. Circulation autoantibodies against C-terminus of NuMA in patients with Behçet’s disease. Central Eur. J. Immunol. 2020, 45, 86–92. [Google Scholar] [CrossRef]
  122. Djaballah-Ider, F.; Touil-Boukoffa, C. Effect of Combined Colchicine-Corticosteroid Treatment on Neutrophil/Lymphocyte Ratio: A Predictive Marker in Behçet Disease Activity. Inflammopharmacology 2020, 28, 819–829. [Google Scholar] [CrossRef]
  123. Cheng, L.; Li, L.; Liu, C.; Yan, S.; Chen, H.; Li, H.; Zhang, F.; Chen, H.; Li, Y. Variation of Red Blood Cell Parameters in Behcet’s Disease: Association with Disease Severity and Vascular Involvement. Clin. Rheumatol. 2021, 40, 1457–1464. [Google Scholar] [CrossRef]
  124. Choi, H.J.; Seo, M.R.; Ryu, H.J.; Baek, H.J. Cross-Cultural Adaptation and Validation of the Behcet’s Disease Current Activity Form in Korea. Korean J. Intern. Med. 2015, 30, 714. [Google Scholar] [CrossRef] [PubMed]
  125. Ahn, J.K.; Kim, J.; Hwang, J.; Song, J.; Kim, K.H.; Cha, H.S. Potential Metabolomic Biomarkers for Reliable Diagnosis of Behcet’s Disease Using Gas Chromatography/ Time-of-Flight-Mass Spectrometry. Jt. Bone Spine 2018, 85, 337–343. [Google Scholar] [CrossRef]
  126. Filleron, A.; Tran, T.A.; Hubert, A.; Letierce, A.; Churlaud, G.; KonCrossed, D.; Sign-Paut, I.; Saadoun, D.; Cezar, R.; Corbeau, P.; et al. Regulatory T Cell/Th17 Balance in the Pathogenesis of Paediatric Behçet Disease. Rheumatology. 2021, 61, 422–429. [Google Scholar] [CrossRef] [PubMed]
  127. Na, S.Y.; Park, M.J.; Park, S.; Lee, E.S. Up-Regulation of Th17 and Related Cytokines in Behçet’s Disease Corresponding to Disease Activity. Clin. Exp. Rheumatol. 2013, 31, 32–40. [Google Scholar] [PubMed]
  128. Zhou, L.; Lopes, J.E.; Chong, M.M.W.; Ivanov, I.I.; Min, R.; Victora, G.D.; Shen, Y.; Du, J.; Rubtsov, Y.P.; Rudensky, A.Y.; et al. TGF-β-Induced Foxp3 Inhibits Th17 Cell Differentiation by Antagonizing RORγt Function. Nature 2008, 453, 236. [Google Scholar] [CrossRef] [PubMed]
  129. Ivanov, I.I.; Tuganbaev, T.; Skelly, A.N.; Honda, K. T Cell Responses to the Microbiota. Annu. Rev. Immunol. 2022, 40, 559–587. [Google Scholar] [CrossRef]
  130. Zhou, Q.; Xiao, X.; Wang, C.; Zhang, X.; Li, F.; Zhou, Y.; Kijlstra, A.; Yang, P. Decreased MicroRNA-155 Expression in Ocular Behcet’s Disease but Not in Vogt Koyanagi Harada Syndrome. Invest. Ophthalmol. Vis. Sci. 2012, 53, 5665–5674. [Google Scholar] [CrossRef]
  131. Na, S.Y.; Park, M.J.; Park, S.; Lee, E.S. MicroRNA-155 Regulates the Th17 Immune Response by Targeting Ets-1 in Behçet’s Disease. Clin. Exp. Rheumatol. 2016, 34, 56–63. [Google Scholar]
  132. Seyahi, E. Phenotypes in Behçet’s syndrome. Intern. Emerg. Med. 2019, 14, 67. [Google Scholar] [CrossRef]
  133. Consolandi, C.; Turroni, S.; Emmi, G.; Severgnini, M.; Fiori, J.; Peano, C.; Biagi, E.; Grassi, A.; Rampelli, S.; Silvestri, E.; et al. Behçet’s syndrome patients exhibit specific microbiome signature. Autoimmun. Rev. 2015, 14, 269–276. [Google Scholar] [CrossRef]
  134. Wang, Q.; Yi, S.; Su, G.; Du, Z.; Pan, S.; Huang, X.; Cao, Q.; Yuan, G.; Kijlstra, A.; Yang, P. Changes in the Gut Microbiome Contribute to the Development of Behcet’s Disease via Adjuvant Effects. Front Cell Dev Biol. 2021, 9, 716760. [Google Scholar] [CrossRef] [PubMed]
  135. Pagliai, G.; Dinu, M.; Fiorillo, C.; Becatti, M.; Turroni, S.; Emmi, G.; Sofi, F. Modulation of gut microbiota through nutritional interventions in Behçet’s syndrome patients (the MAMBA study): Study protocol for a randomized controlled trial. Trials 2020, 21, 511. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flowchart of the literature search strategy.
Figure 1. Flowchart of the literature search strategy.
Ijms 24 03041 g001
Figure 2. Graphical representation of the number of studies per year included in this systematic review. The scatter plot was established using the package Ggplot2 [12] of R studio [13].
Figure 2. Graphical representation of the number of studies per year included in this systematic review. The scatter plot was established using the package Ggplot2 [12] of R studio [13].
Ijms 24 03041 g002
Figure 3. Graphical representation of the global origin of the publication rate of the analyzed studies per country, colored by BD study rate. The graphical representation was computed by log-transforming the number of research papers published by each country. It is possible to recognize the Silk Road pattern. The map was created using the package Ggplot2 [124] of R studio [12].
Figure 3. Graphical representation of the global origin of the publication rate of the analyzed studies per country, colored by BD study rate. The graphical representation was computed by log-transforming the number of research papers published by each country. It is possible to recognize the Silk Road pattern. The map was created using the package Ggplot2 [124] of R studio [12].
Ijms 24 03041 g003
Figure 4. Mechanisms underlying Behçet’s disease’s etiopathogenesis.
Figure 4. Mechanisms underlying Behçet’s disease’s etiopathogenesis.
Ijms 24 03041 g004
Table 1. Main characteristics of the studies included in the analysis.
Table 1. Main characteristics of the studies included in the analysis.
REFYearFirst AuthorCountryDesignBD Patients, nControls, nBiomarker TestedUrinary/
Serologic
Diagnostic/Activity
[14]1995Yosipovitch et al.IsraelRetrospective2520IL-1BSDiagnostic
SIL-2RSDiagnostic
[15]1995Deǧer et al.TurkeyRetrospective42 (20 active)40PMN elastaseSDiagnostic/Activity
[16]1995Direskeneli et al.UKRetrospective70 (56 active)52AECASDiagnostic/Activity
vVFSDiagnostic
[17]1997Uslu et al.TurkeyRetrospective2718ET-1SDiagnostic
[18]1998Alpsoy et al.TurkeyRetrospective32 (14 active)20IL-2SDiagnostic
SIL-2RSActivity
[19]2000Katsantonis et al.GermanyRetrospective34 (25 active)N/AIL-8SActivity
[20]2000Eksioglu-Demiralp et al.TurkeyRetrospective3755CD4+CD16+SDiagnostic
CD4+CD56+SDiagnostic
[21]2000Freysdottir et al.UKRetrospective2026T-γδSDiagnostic
CD56SDiagnostic
[22]2002Krause et al.IsraelRetrospective2720IgG ASCASDiagnostic
IgA ASCASDiagnostic
[23]2002Evereklioglu et al.TurkeyRetrospective35 (18 active)20LeptinSDiagnostic/Activity
[24]2002Er et al.TurkeyRetrospective43 (20 active)52ET-1SDiagnostic/Activity
HomocysteineSDiagnostic/Activity
NOSDiagnostic/Activity
[25]2002Saglam et al.TurkeyRetrospective44 (23 active)30cICAM-1SDiagnostic/Activity
[26]2002Evereklioglu et al.TurkeyRetrospective52 (27 active)32NOSDiagnostic/Activity
[27]2003Evereklioglu et al.TurkeyRetrospective36 (16 active)20NO (urinary)UDiagnostic/Activity
NO (serum)SDiagnostic/Activity
[28]2003Erkiliç et al.TurkeyRetrospective35 (17 active)20ADASDiagnostic/Activity
TBARSSDiagnostic
Plasmatic SODSDiagnostic/Activity
RBC SODSDiagnostic/Activity
Plasmatic GSHPxSDiagnostic/Activity
RBC GSHPxSDiagnostic/Activity
RBC CatalaseSDiagnostic
[29]2004Akdeniz et al.TurkeyRetrospective2716IL-6SDiagnostic
Il-2SDiagnostic
TNF-αSDiagnostic
NOSDiagnostic
[30]2004Sari et al.TurkeyRetrospective2320E-selectineSDiagnostic
ESRSDiagnostic
PCRSDiagnostic
[31]2004Yazici et al.TurkeyRetrospective49 (31 active)40MPOSDiagnostic/Activity
AOPPSDiagnostic/Activity
ThiolSDiagnostic/Activity
[32]2004Duygulu et al.TurkeyRetrospective23 (11 active)15NOSDiagnostic/Activity
[33]2005Ureten et al.TurkeyRetrospective72 (37 active)73CD64SDiagnostic/Activity
[34]2005Calis et al.TurkeyRetrospective75 (50 active)25ADASDiagnostic/Activity
[35]2005Qiao et al.JapanRetrospective35 (15 active)16CXCR2SDiagnostic/Activity
[36]2005Gür-Toy et al.TurkeyRetrospective670IL-8SActivity
CRPSDiagnostic
ESRSDiagnostic
[37]2005Coskun et al.TurkeyRetrospective40 (25 active)30NeopterinSDiagnostic/Activity
ESRPDiagnostic/Activity
CRPSDiagnostic/Activity
[38]2006Yardim-Akaydin et al.TurkeyRetrospective2343AllantoinSDiagnostic
MDASDiagnostic
Ascorbic acidSDiagnostic
[39]2006Kose et al.TurkeyRetrospective68 (51 active)17NeopterinSDiagnostic/Activity
[40]2006Canpolat et al.TurkeyRetrospective23 (10 active)20ADASDiagnostic/Activity
Erythrocyte ADASDiagnostic/Activity
[41]2006Kwon et al.South KoreaProspective211 (92 active)N/AProtein SSActivity
[42]2006Briani et al.ItalyRetrospective32118Anti-HS igMSDiagnostic
Anti-HS igGSDiagnostic
[43]2006Sarican et al.TurkeyRetrospective64 (25 active)26HomocysteineSDiagnostic/Activity
[44]2007Lee et al.South KoreaRetrospective50 (26 active)UKGal-3SDiagnostic/Activity
G3BPSActivity
[45]2007Pay S et al.TurkeyRetrospective58 (23 active)20MMP-2SDiagnostic
MMP-9SDiagnostic/Activity
[46]2008Öztürk et al.TurkeyRetrospective2121VEGFSDiagnostic
ESRSDiagnostic
CRPSDiagnostic
[47]2008Turan et al.TurkeyProspective35N/AsTNFR1SActivity
sTNFR2SActivity
[48]2008Kutlay et al.TurkeyRetrospective45 (33 active)15CECSDiagnostic/Activity
[49]2008Curnow et al.UKRetrospective52 (24 active)35IL-15SDiagnostic/Activity
CXCL-8SDiagnostic/Activity
TNF-αSDiagnostic/Activity
[50]2008Polat et al.TurkeyRetrospective3216IL-8SDiagnostic/Activity
[51]2008Durmazlar et al.TurkeyRetrospective45 (33 active)29IL-8SDiagnostic/Activity
[52]2009Habibagah et al.IranRetrospective53 (15 active)44IL-23SDiagnostic/Activity
E–cadherinSDiagnostic
[53]2009Fadini et al.ItalyRetrospective3027CD34+KDR+ EPCsSDiagnostic
CD34+CD133+KDR+ EPCsSDiagnostic
[54]2010Choe et al.South KoreaRetrospective59 (21 active)65Angiopoietin-1SDiagnostic
Angiopoietin-2SDiagnostic
[55]2010Donmez et al.TurkeyRetrospective89 (17 active)86aTAFISDiagnostic
Thrombomodulin Diagnostic
[56]2010Sezer et al.TurkeyRetrospective60 (33 active)46MDASDiagnostic
8-OHdGSDiagnostic/Activity
T-SHSDiagnostic
[57]2011Özden et al.TurkeyRetrospective7061Gal-3SDiagnostic/Activity
[58]2011Pehlivan et al.TurkeyRetrospective45 (25 active)30ResistinSDiagnostic/Activity
TNF-αSDiagnostic/Activity
[59]2011Ahn et al.South KoreaRetrospective71 (21 active)34α defensin1SActivity
αdefensin1 mRNASDiagnostic/Activity
[60]2011Shin et al.South KoreaRetrospective8023AAEASDiagnostic
[61]2011Jung et al.South KoreaRetrospective88 (30 severe, 12 moderate)10sTREM1SDiagnostic/Activity
TNF-α Diagnostic
[62]2011Vural et al.TurkeyRetrospective2040STIP-1SDiagnostic
[63]2012Bello et al.SpainRetrospective3028sCD40LSDiagnostic
MMP-9SDiagnostic
[64]2012Gündüz et al.TurkeyRetrospective40 (11 active)20CD4+CD25+FOXP3+TregSDiagnostic/Activity
CD4+FOXP3+TregSDiagnostic/Activity
[65]2012Wang et al.ChinaRetrospective4979Proteomic analysisSDiagnostic
[66]2013Örem et al.TurkeyRetrospective72 (40 active)30Lipoprotein-associated phospholipase A2SDiagnostic/Activity
CRPSDiagnostic/Activity
ESRSDiagnostic/Activity
[67]2013Hamzaoui et al.TunisiaRetrospective46 (20 active)70IL-33SDiagnostic/Activity
IL6SDiagnostic
IL7SDiagnostic
[68]2013Vural et al.TurkeyRetrospective144168MTCH1 AbSDiagnostic
[69]2014Shaker et al.EgyptRetrospective30 (20 active)20TNF- αSDiagnostic/Activity
APRILSDiagnostic/Activity
BCMASDiagnostic/Activity
BAFFSDiagnostic/Activity
CRPSDiagnostic/Activity
ESRSDiagnostic/Activity
[70]2014Xun et al.ChinaRetrospective58106ProhibitinSDiagnostic
[71]2014Vayà et al.SpainRetrospective8994RDWSDiagnostic
CRPSDiagnostic
FibrinogenSDiagnostic
LeucocytesSDiagnostic
NeutrophilsSDiagnostic
[72]2014Balta et al.TurkeyRetrospective33 (16 active)35EndocanSDiagnostic/Activity
CRPSDiagnostic
ESRSDiagnostic
[73]2014Ozuguz et al.TurkeyProspective4020ADMASDiagnostic
CRPSDiagnostic/Activity
ESRSDiagnostic/Activity
HomocysteineSDiagnostic/Activity
[74]2014Mejia et al.SpainProspective56 (17 active)56Prothrombin fragm. 1.2SDiagnostic/Activity
Factor VIIISDiagnostic/Activity
vWFSDiagnostic
[75]2015Lopalco et al.ItalyProspective5832IL-6SDiagnostic
IL-8SDiagnostic
IL-18SDiagnostic
IFN-αSDiagnostic
CXCL11SDiagnostic
[76]2015Yuksel et al.TurkeyRetrospective36 (17 active)35ADMASDiagnostic/Activity
NLRSDiagnostic/Activity
[77]2015Bassyouni et al.EgyptRetrospective4730Angiopoietin-1SDiagnostic
[78]2015Tulunay et al.TurkeyRetrospective2626STAT3SDiagnostic
[79]2015Belguendouz et al.AlgeriaRetrospective26 (16 active)17IL-18SActivity
[80]2015Ozturk et al.TurkeyRetrospective65 (40 active)62NLRSDiagnostic/Activity
[81]2015Turkcu et al.TurkeyRetrospective51 (25 active)24TNF-αSDiagnostic
ResistinSDiagnostic
OmentinSDiagnostic
[82]2015De Souza et al.BrazilRetrospective26 (13 active)20HMGB1SDiagnostic
[83]2015Seo et al.South KoreaRetrospective112 (66 active)45YKL-40SDiagnostic/Activity
[84]2016Yolbas et al.TurkeyRetrospective53 (6 active)55NLRSActivity
91
51+39
[85]2016Hu et al.ChinaRetrospective
Phase I
40 (identification)35Protein microarray
Phase II130 (validation)223Anti-CTDP1 AbSDiagnostic
[86]2016Mejia et al.SpainRetrospective5573Procoagulant microparticlesSDiagnostic
[87]2016Balkarli et al.TurkeyRetrospective186 (120 active)79NLRSDiagnostic
ESRSDiagnostic/Activity
CRPSDiagnostic
[88]2016Park et al.South KoreaRetrospective51 (29 active)N/AAnti-lysozymeSActivity
[89]2016Cantarini et al.ItalyRetrospective27 (57 total samples: 21 from active, 36 inactive)36CD40LSDiagnostic
LeptinSDiagnostic
sTNFRSDiagnostic
IL-6SDiagnostic
ESRSActivity
[90]2017Cure et al.TurkeyRetrospective8484AIPSDiagnostic/Activity
CRPSDiagnostic
[91]2017Jiang et al.ChinaRetrospective140 (108 active)107PLRSDiagnostic/Activity
LMRSDiagnostic
ESRSActivity
CRPSActivity
[92]2017Kang et al.South KoreaRetrospective110110AAEA IgGSDiagnostic
[93]2017Ahn JK et al.South KoreaRetrospective4441Panel of 10 urinary biomarkers: guanine, pyrrole-2-carboxylate, 3-hydroxypyroline, mannose, L-citrulline, galactonate, isothreonate, sedoheptulose, hypoxanthine, and gluconic acidlactonateUDiagnostic
GuanineUDiagnostic
Pyrrole-2-carboxylateUDiagnostic
3-hydroxypyrolineUDiagnostic
MannoseUDiagnostic
L-citrullineUDiagnostic
GalactonateUDiagnostic
IsothreonateUDiagnostic
SedoheptuloseUDiagnostic
HypoxanthineUDiagnostic
Gluconic acidlactonateUDiagnostic
[94]2017Lee et al.South KoreaRetrospective Phase I15 (identification)15Fibrin, apoliprorotein A-IV and SAASDiagnostic
Phase II49 (validation)41SAASDiagnostic
IL-1βSDiagnostic
[95]2017Ha et al.South KoreaRetrospective50 (29 active)35IL-32SDiagnostic
[96]2017Lopalco et al.ItalyRetrospective4619sTNFR1SDiagnostic
sTNFR2SDiagnostic
Chitinase3-like1SDiagnostic
gp130/sIL-6RbSDiagnostic
IL-26SDiagnostic
[97]2018Omma et al.TurkeyRetrospective93 (57 active)62CalprotectinSDiagnostic
CRPSDiagnostic
IMASDiagnostic
[98]2018Koca et al.TurkeyRetrospective7175BilirubinSDiagnostic
[99]2018Enecik et al.TurkeyRetrospective45 (28 active)25IL-20SDiagnostic
[100]2018Harmanci et al.TurkeyRetrospective3030VEGF gene expression levelsSDiagnostic
[101]2018Lucherini et al.ItalyRetrospective7229IgDSDiagnostic
[102]2018Chekaoui et al.AlgeriaRetrospective48 (28 active)41IL-1βSDiagnostic/Activity
NOSDiagnostic/Activity
AOPPSDiagnostic/Activity
MDASDiagnostic
SODSDiagnostic/Activity
[103]2018Kolahi et al.IranRetrospective4761mir-155SDiagnostic
TNF-α expressionSDiagnostic
[104]2018Ahn et al.South KoreaRetrospective4545Panel of 5 biomarkers: DA, fructose, tagatose, LA, and OASDiagnostic
[105]2018Saylam et al.TurkeyRetrospective3041suPARSDiagnostic
CRPSDiagnostic
[106]2018Ahmadi et al.IranRetrospective4758Th17SDiagnostic
TregSDiagnostic
RORɣt mRNASDiagnostic
FoxP3 mRNASDiagnostic
IL-17mRNASDiagnostic
IL-23 mRNASDiagnostic
TGF mRNASDiagnostic
IL-10 mRNASDiagnostic
IL-17SDiagnostic
IL-23SDiagnostic
IL-10SDiagnostic
TFG-betaSDiagnostic
miR-93SDiagnostic
miR-106bSDiagnostic
miR-25SDiagnostic
miR-146°SDiagnostic
miR-155SDiagnostic
miR-326SDiagnostic
[104]2018Hassouna et al.EgyptRetrospective3015miR-155SDiagnostic
[107]2018Prado et al.BrazilRetrospective97 (43 active)123AAEA IgMSDiagnostic/Activity
[108]2018Acikgoz et al.TurkeyRetrospective6050MHRSDiagnostic
[109]2018Hasan et al.UKRetrospective60 (44 active)60NKSDiagnostic
CD56DimSDiagnostic
CD56BrighSDiagnostic
[110]2018Zheng et al.ChinaRetrospective Phase I24 (identification)26PC (34:3)SDiagnostic
PC (40:8)SDiagnostic
LASDiagnostic
AASDiagnostic
Phase II25 (validation)19LASDiagnostic
27AASDiagnostic
[111]2019Şahin et al.TurkeyRetrospective4644Pannexin-1SDiagnostic
[112]2019Bassyouni et al.EgyptRetrospective8760CCN2SDiagnostic
[113]2019Arica et al.TurkeyRetrospective45 (32 active)28Early EPCsSDiagnostic/Activity
Late EPCsSDiagnostic/Activity
MMP9SDiagnostic
VEGFSDiagnostic/Activity
CRPSDiagnostic/Activity
ESRSDiagnostic
[114]2019Sandikci et al.TurkeyRetrospective150100SerumnativethiolSDiagnostic
Total thiolSDiagnostic
T-SHSDiagnostic
[115]2019Talaat et al.EgyptRetrospective6420IL-6SDiagnostic/Activity
IL-10SDiagnostic
IL-17SDiagnostic
[116]2019Gheita et al.EgyptRetrospective9660NLRSDiagnostic
PLRSDiagnostic
RDWSDiagnostic
MPVSDiagnostic
VEGFSDiagnostic
[117]2019El Boghdady et al.EgyptRetrospective5145TNF-αSDiagnostic
IL-6SDiagnostic
E-selectineSDiagnostic
VCAMSDiagnostic
miR-181bSDiagnostic
[118]2019Balbaba et al.TurkeyRetrospective48 (24 active)24CortistatinSDiagnostic
[119]2020Hassan et al.EgyptRetrospective4242EndocanSDiagnostic/Activity
[120]2020Hussain et al.ChinaRetrospective50100MoesinSDiagnostic
[121]2020Hussain et al.ChinaRetrospective3264NuMA AbSDiagnostic
[122]2020Djaballah-Ider et al.AlgeriaRetrospective61 (47 active)25NLRSActivity
NOSActivity
IL-4SActivity
IFN-gammaSActivity
[123]2021Cheng et al.ChinaRetrospective48 (34 active)96Lymphocyte countSDiagnostic/Activity
White blood cell countSDiagnostic
Neutrophil countSDiagnostic
Basophil countSDiagnostic/Activity
RDWSDiagnostic/Activity
MCHSDiagnostic
MCHCSDiagnostic
Platelet countSDiagnostic/Activity
PlateletcountSDiagnostic/Activity
MPVSDiagnostic/Activity
CRSSDiagnostic
PLRSDiagnostic/Activity
NLRSDiagnostic
MonocyteSActivity
LMRcountSDiagnostic
8-OHdG—8-hydroxy-2′-deoxyguanosine; AA—arachidonic acid; AAEA—anti-alpha-enolase antibodies; ADA—adenosine deaminase; ADMA—asymmetric dimethyl arginine; AECA—anti-endothelial cell antibodies; AIP—atherogenic index plasma, anti-HS—anti-heparin–sulfate antibodies; anti-CTDP1—anti-carboxy-terminal domain phosphatase subunit 1; AOPP—advanced oxidation protein products; APRIL—a proliferation-inducing ligand; ASCA—anti-Saccharomyces cerevisiae; aTAFI—activated thrombin activatable fibrinolysis inhibitor; BAFF—B-cell-activating factor; BCMA—B-cell maturation antigen; CEC- circulating endothelial cells; cICAM—circulating intercellular adhesion molecule-1; cNuM—anuclear mitotic apparatus protein located at the carboxyl terminus; CPR—C-reactive protein; CTGF—connective tissue growth factor;CXCL11—C-X-C motif chemokine 11; CXCR2—C-X-C motif chemokine receptor 2; DA—decanoic acid; Endocan—human endothelial cell-specific molecule-1; EPC—endothelial progenitor cells; ESR—erythrocyte sedimentation rate; ET-1—endothelin-1; ETP—endogenous thrombin potential; GAL-3—galectin-3; G3BP—galectin-3 binding protein; HMGB1—high-mobility group box 1; IgD—D immunoglobulin;IMA—ischemia-modified albumin; INFa—interferon alpha; INFg—interferon gamma; LA—linoleic acid; LMR—lymphocytes-to-monocytes ratio; LpPLA2—lipoprotein-associated phospholipase A2; MDA—manoldialdehyde; MHR—monocyte-to-high-density lipoprotein–cholesterol ratio; MMP—matrix metalloproteinase; MPO—plasma myeloperoxidase; MPV—mean platelet volume; MTCH1—mitochondrial carrier homolog 1; NLR—neutrophil-to-lymphocyte ratio; NO—nitric oxide; OA—oleic acid; PC—phosphatidylcholines; PLR—platelet-to-lymphocyte ratio; PMN—polymorph nuclear; Procoagulant MP—procoagulant microparticles; RDW—red cell distribution width; SAA—serum amyloid A; SIL-1R—Soluble interleukin-1 receptor; SIL6-RB-Soluble interleukin-6 receptor B; SOD—Superoxide dismutase; STIP1—Stress induced phosphoprotein 1; sTNFR—soluble tumor necrosis factor receptor; sTREM1—soluble triggering receptor expressed on myeloid cells; suPAR—soluble urokinase plasminogen activator receptor; TBARS—thiobarbituric acid-reactive substances TGF-b—transforming growth factor beta; TNFa—tumor necrosis factor alpha; T-SH—total sulfhydryl levels; VCAM—vascular cell adhesion molecule 1;VEGF—vascular endothelial growth factor; vWF—von Willebrand factor.
Table 2. Serological and urinary biomarkers investigated in the studies included in the systematic review.
Table 2. Serological and urinary biomarkers investigated in the studies included in the systematic review.
ILsIL-1β
[14,94,102]
IL-2
[18,29]
IL-4
[122]
IL-6 [29,67,75,89,115,117]IL-7
[67]
IL-8
[19,36,49,50,51,75]
IL-10
[106,115]
IL-15
[49]
IL-17
[106,115]
IL-18
[75,79]
IL-20
[99]
IL-23
[52,106]
IL-26
[96]
IL-32
[95]
IL-33
[67]
CytokinesTNF-α
[29,49,58,61,69,81,103,117]
TGF-β
[106]
APRIL
[69]
BAFF
[69]
INF-α
[75]
IFN-γ
[122]
CTGF
[112]
STAT3
[78]
CXCL11
[75]
Surface proteinsCD64
[33]
CXCR2
[35]
BMCA
[69]
VCAM
[117]
Soluble proteinsSIL-2R
[14,18]
PMN leukocyte elastase
[15]
AECA
[16]
vWF
[16,74]
ET-1
[17,24]
Anti-ASCA Ab [22]Leptin
[23,89]
Homocysteine [24,43,73]CRP [30,31,36,37,64,66,69,71,72,73,87,90,91,93,99,105,113,123]cICAM-1
[23]
Catalase
[28]
ADA
[28,34,40]
SOD
[28,102]
TBARS
[28]
E-selectine
[30,117]
MPO
[31]
Neopterin
[37,39]
VEGF [46,100,113,116]Protein S
[41]
antiHS
[42]
Gal-3
[44,57]
G3BP
[44]
MMP2
[45]
α-defensin1 [59]
sTNFR1 e 2 [47,89,96]E-Caderin [52]Angiopoietin1 [54,77]Resistin
[58,81]
Thrombomodulin [55]aTAFI
[55]
AAEA
[60,92,107]
sTREM1
[61,100]
STIP
[62]
sCD40L
[63,89]
MMP9
[63,113]
Lp-PLA2
[66]
MTCH1 Ab
[68]
Prohibitin [70]Endocan
[72,119]
ADMA
[73,76]
Omentin
[81]
HMBG1
[82]
Anti-lysozyme
[88]
Fibrinogen
[71]
Factor VIII [74]cNuMA Ab
[121]
anti-CTDP1 Ab [85]SAA
[94]
sIL6-RB
[96]
Chitinase3-like1 [83,96]Bilirubin
[98]
Calprotectin [97]IMA
[97]
IgD
[101]
suPAR
[105]
Pannexin-1
[111]
Cortistatin
[118]
Moesin
[120]
CellsCD4+CD16+
[19,20]
CD4+CD56+ [19,20]T γδ [21]CEC
[48]
CD34+KDR+EPCs [53,113]CD34+CD133+KDR+ EPCs [53]CD4+CD25+FOXP3+Treg
[64]
CD4+FOXP3+Treg
[64]
Treg
[106]
Th17
[106]
CD56 +
[109]
miRNAα-defensin 1
[59]
miR-155
[103,104,106]
miR-181b
[117]
miR-93
[106]
miR-106b
[106]
miR-25
[106]
miR-146a
[106]
miR-326
[106]
Metabolomic/
proteomic markers
DA, OA
Fructose, tagatose
[125]
LA
[110,125]
PC
[110]
AA
[110]
Panel of six proteomic biomarkers
[65]
OthersESR [30,31,36,37,46,52,66,69,72,87,89,91,99,113]NO
[24,26,27,29,32,102,122]
Thiol
[31,114]
AOPP
[31,102]
Allantoin
[38]
MDA
[38,56,102]
Ascorbic acid
[38]
8-OhdG
[56]
T-SH
[56,114]
PLR
[31,91,116]
LMR
[91,123]
NLR
[31,76,80,84,87,116,122]
AIP
[90]
RDW
[71]
ETP
[74]
MPV
[123]
RDW
[71,123]
Procoagulant MP [86]MHR
[108]
Urinary markersMetabolomic panel:
Guanine
Pyrrole-2-carboxylate
3-hydroxypyroline
Mannose
L-citrulline
Galactonate
Isothreonate
Sedoheptulose
Hypoxanthine
Gluconic acidlactonate [93]
NO
[14]
8-OHdG—8-hydroxy-2′-deoxyguanosine; AA—arachidonic acid; AAEA—anti-alpha-enolase antibodies; ADA—adenosine deaminase; ADMA—asymmetric dimethyl arginine; AECA—anti-endothelial cell antibodies; AIP—atherogenic index plasma, anti-HS—anti-heparin–sulfate antibodies; anti-CTDP1—anti-carboxy-terminal domain phosphatase subunit 1; AOPP—advanced oxidation protein products; APRIL—a proliferation-inducing ligand; ASCA—anti-Saccharomyces cerevisiae; aTAFI—activated thrombin activatable fibrinolysis inhibitor; BAFF—B-cell-activating factor; BCMA—B-cell maturation antigen; CEC- circulating endothelial cells; cICAM—circulating intercellular adhesion molecule-1; cNuM—a nuclear mitotic apparatus protein located at the carboxyl terminus; CPR—C-reactive protein; CTGF—connective tissue growth factor; CXCL11—C-X-C motif chemokine 11; CXCR2—C-X-C motif chemokine receptor 2; DA—decanoic acid; Endocan—human endothelial cell-specific molecule-1; EPC—endothelial progenitor cells; ESR—erythrocyte sedimentation rate; ET-1—endothelin-1; ETP—endogenous thrombin potential; GAL-3—galectin-3; G3BP—galectin-3 binding protein; HMGB1—high-mobility group box 1; IgD—D immunoglobulin; IMA—ischemia-modified albumin; INFa—interferon alpha; INFg—interferon gamma; LA—linoleic acid; LMR—lymphocytes-to-monocytes ratio; LpPLA2—lipoprotein-associated phospholipase A2; MDA—manoldialdehyde; MHR—monocyte-to-high-density lipoprotein–cholesterol ratio; MMP—matrix metalloproteinase; MPO—plasma myeloperoxidase; MPV—mean platelet volume; MTCH1—mitochondrial carrier homolog 1; NLR—neutrophil-to-lymphocyte ratio; NO—nitric oxide; OA—oleic acid; PC—phosphatidylcholines; PLR—platelet-to-lymphocyte ratio; PMN—polymorph nuclear; Procoagulant MP—procoagulant microparticles; RDW—red cell distribution width; SAA—serum amyloid A; SIL-1R—Soluble interleukin-1 receptor; SIL6-RB-Soluble interleukin-6 receptor B; SOD—Superoxide dismutase; STIP1—Stress induced phosphoprotein 1; sTNFR—soluble tumor necrosis factor receptor; sTREM1—soluble triggering receptor expressed on myeloid cells; suPAR—soluble urokinase plasminogen activator receptor; TBARS—thiobarbituric acid-reactive substances TGF-b—transforming growth factor beta; TNFa—tumor necrosis factor alpha; T-SH—total sulfhydryl levels; VCAM—vascular cell adhesion molecule 1; VEGF—vascular endothelial growth factor; vWF—von Willebrand factor.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Arbrile, M.; Radin, M.; Medica, D.; Miraglia, P.; Rilat, L.; Cecchi, I.; Foddai, S.G.; Barinotti, A.; Menegatti, E.; Roccatello, D.; et al. Finding the Needle in the Haystack: Serological and Urinary Biomarkers in Behçet’s Disease: A Systematic Review. Int. J. Mol. Sci. 2023, 24, 3041. https://doi.org/10.3390/ijms24033041

AMA Style

Arbrile M, Radin M, Medica D, Miraglia P, Rilat L, Cecchi I, Foddai SG, Barinotti A, Menegatti E, Roccatello D, et al. Finding the Needle in the Haystack: Serological and Urinary Biomarkers in Behçet’s Disease: A Systematic Review. International Journal of Molecular Sciences. 2023; 24(3):3041. https://doi.org/10.3390/ijms24033041

Chicago/Turabian Style

Arbrile, Marta, Massimo Radin, Davide Medica, Paolo Miraglia, Letizia Rilat, Irene Cecchi, Silvia Grazietta Foddai, Alice Barinotti, Elisa Menegatti, Dario Roccatello, and et al. 2023. "Finding the Needle in the Haystack: Serological and Urinary Biomarkers in Behçet’s Disease: A Systematic Review" International Journal of Molecular Sciences 24, no. 3: 3041. https://doi.org/10.3390/ijms24033041

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