Skip to main content

REVIEW article

Front. Immunol., 10 May 2019
Sec. Autoimmune and Autoinflammatory Disorders
This article is part of the Research Topic Neutrophil-mediated skin diseases: immunology and genetics View all 11 articles

Behçet’s Disease: An Overview of Etiopathogenesis

\nPietro LeccesePietro Leccese1Erkan Alpsoy*Erkan Alpsoy2*
  • 1Rheumatology Institute of Lucania (IRel) and the Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
  • 2Department of Dermatology and Venereology, School of Medicine, Akdeniz University, Antalya, Turkey

Behçet’s disease (BD) is a systemic inflammatory disease with a chronic, relapsing-remitting course of unknown etiology hallmarked predominantly by mucocutaneous lesions and ocular involvement. BD shares some common features with autoimmune and autoinflammatory diseases and spondyloarthropathies (MHC-I-opathies). It is related to more than one pathogenic pathway triggered by environmental factors such as infectious agents in genetically predisposed subjects. The interplay between genetic background and immune system is linked to the BD presentation. Genetic factors have been investigated extensively, and several recent genome-wide association studies have confirmed HLA-B*51 to be the strongest genetic susceptibility factor. However, new non-HLA susceptibility genes have been identified. Genetic variations in the genes encoding the cytokines could affect their function and be associated with disease susceptibility. Infectious agents such as Streptococcus sanguinis or the differences in salivary or gut microbiome composition can be considered to trigger the innate-derived inflammation, which is, subsequently, sustained by adaptive immune responses. Altered trimming of microbial and/or endogenous peptides by endoplasmic reticulum aminopeptidase 1 (ERAP1), presented by HLA-B*51, may play a key role in BD pathogenesis causing an alteration in T cell balance with downregulation of Tregs and expansion of Th1 and Th17. The activity of neutrophils is increased and there is an intense neutrophil infiltration in the early stage of inflammation in organs affected by the disease. Association with HLA-B*51 and increased IL-17 response seems to have an important role in neutrophil activity. In this paper, we provide an overview of the most recent advances on BD etiopathogenesis.

Introduction

Behçet’s disease (BD) is a systemic inflammatory disease with a chronic, relapsing-remitting course, and its etiology is still unknown. The disease is characterized by a range of clinical manifestations including oral aphthae, genital ulcers, skin lesions, ocular, vascular, articular, gastrointestinal, urogenital, pulmonary, and neurologic involvement. BD is prevalent in regions along the “Silk Road,” extending from Japan to Mediterranean countries. BD often begins between the ages of 20–40. The disease is equally distributed between men and women and the diagnosis can be made only on the basis of clinical symptoms and signs. The course of the disease is more severe in male patients with younger age at onset and an increased number of organs affected at diagnosis (1). Disease can be recognized by clinical findings because of the absence of a universally accepted diagnostic laboratory test. BD diagnosis is largely based on mucocutaneous symptoms which are a common characteristic of various diagnostic criteria used in the diagnosis of the disease so far (2). The International Study Group for Behçet’s disease criteria (requires the presence of oral ulcer plus any two of recurrent genital ulcer, typical eye lesions, typical cutaneous lesions, or a positive skin pathergy test) is the most commonly used and internationally recognized diagnostic criteria by the authors of this field (2, 3).

Besides considerable morbidity, BD has increased mortality because of the pulmonary artery and large vessel, neurological, and gastrointestinal involvements. Therefore, knowing the etiopathogenesis of BD is extremely important to better understand the disease and, more importantly, to develop targeted therapies. BD has been listed among autoimmune diseases by some authors because of positive response to classical immunosuppressive agents and involvement of autoantigens and antigen-specific T cells. Others claim the disease should be included in the group of autoinflammatory diseases because of unprovoked episodes of inflammation without evidence of antigen-specific T cells or autoantibodies, increased activity of neutrophils, elevated levels of interleukin (IL)-1β (4). Most authors evaluate the disease as a spondyloarthropathy (MHC-I-opathy) based on Human Leukocyte Antigen (HLA) class I association and epistatic endoplasmic reticulum aminopeptidase 1 (ERAP-1) interactions, increased T helper (Th) 17 type immune response, neutrophilic inflammation and barrier dysfunction in environmentally exposed organs (5). According to the current literature, BD cannot be definitely classified under any of these three groups and defining it as autoimmune, autoinflammatory or spondyloarthropathy appears to be a simplified approach (6). BD shares some common features with all the above-mentioned entities and involves more than one pathogenic pathway triggered by environmental factors such as infectious agents in genetically predisposed subjects. We will discuss the most recent evidences on the etiology of BD under the subtitles of infectious, genetic and immunological etiology sections of this review (1, 7, 8).

Infections

Infectious agents have long been proposed as triggering factors in BD development. Antigens from viruses such as herpes simplex virus (HSV)-1 or bacteria belonging to Streptococcus species such as Streptococcus sanguinis have been suspected to have high homology with human proteins such as heat-shock proteins (HSP) and the cross-reaction leads to an immune response in genetically predisposed individuals (1, 9). Professor Hulusi Behçet was indeed one of the first authors who regarded the disease as possibly related to an infectious agent (10). Several studies have investigated the association between HSV-1 and BD. Studd et al. in an situ DNA-RNA hybridization method, detected a higher frequency of hybridization between HSV-1 DNA and complementary RNA in mononuclear cells of BD patients compared with healthy controls. The results show the presence of at least a portion of the HSV-1 genome in mononuclear cells of BD patients (11).

Several Streptococcus strains have become increasingly important in infectious etiology. The development of some clinical manifestations of the disease in hypersensitivity tests against streptococcal antigens is one of the most relevant evidences (12). In addition, antibodies against S. sanguinis and S. pyogenes were obtained more frequent in BD patients than in controls (13). Streptococcal 65-kDa HSP from an uncommon serotype (KTH-1, strain BD113-20) of oral S. sanguinis has been reported to be an important trigger in the pathogenesis (14). Neurofilament medium (Nf-M) was recently suggested as possible antigen able to trigger an immune response via molecular mimicry with bacterial HSP-65 (15). Immunoglobulin M in BD patients has been reported able to react with some streptococcal proteins such as streptococcal α-enolase and glyceraldehyde 3-phosphate dehydrogenase (16).

Cho et al. demonstrated that the S. sanguinis GroEL protein is a target of the serum anti-S. sanguinis IgA antibody. In addition, serum IgA reactivity against recombinant S. sanguinis GroEL has been correlated to reactivity against recombinant human hnRNP A2/B1 suggesting how autoreactive lymphocytes may be activated by infectious triggering (17).

As BD usually starts from the oral mucosa, it has been speculated that oral microbial flora may be implicated in the pathogenesis of the disease (18). BD patients can develop new-onset oral ulceration or experience both cutaneous and systemic flare-ups following dental procedures or surgical treatments for chronic tonsillitis (19, 20). Antimicrobial agents have been used successfully for treating various disease symptoms (21). Several previous studies and our experience showed oral health impairment in BD patients compared with healthy subjects (18, 22, 23). Oral health improvement in BD patients may positively modify their disease course. Dental treatments in BD patients could be associated with a relapse of oral aphthae in the short time but could decrease their number in longer follow-up (~6 months) (24), also leading to better oral health in the long-term follow-up. Higher levels of various Streptococci were found in the oral mucosa of BD patients. In addition, S. sanguinis strain resulted able to induce the secretion of inflammatory cytokines by the KTH-1 cells. It is plausible that an inflammation process induced by infectious agents in subjects with predisposing genetic background leads to the development of BD (25, 26).

No association between BD and other bacterial species such as Borrelia burgdorferi and or Helicobacter pylori have been found (27, 28). Cytomegalovirus, Epstein-Barr virus, Parvovirus B19, Varicella zoster virus, Hepatitis virus have also been investigated as possible triggering factors but these studies were characterized by low-level evidences (29, 30).

Recent studies have shown that the differences in salivary or gut microbiome composition may have a role in the pathogenesis. In a study of the salivary microbiome using high- throughput sequencing of the 16S rRNA V4 region, Coit et al. reported that BD patients have a significantly less diverse microbial community structure than healthy controls (31). In another study, Consolandi et al. compared the fecal microbiota of BD patients to healthy controls. They reported both a peculiar dysbiosis of the gut microbiota and a significant decrease of butyrate production in BD patients. Authors speculated that a defect of butyrate production might lead to both reduced T regulatory cells (Tregs) responses and activation of immunopathological T-effector responses (32).

In summary, until now, no infectious agent has been isolated as the specific etiologic agent. Additionally, results of antibacterial and antiviral treatments are controversial. However, there is a general agreement that infectious agents or microbiome is not directly responsible for the emergence of BD, but they play a triggering role in the development of the disease by causing dysfunction of the immune system.

Genetics

Increased prevalence of the disease along the ancient “Silk Road,” familial aggregation, association with the genes inside the major histocompatibility complex (MHC) region and outside the MHC region are the main evidence of genetic influence and a complex inheritance model of disease (33, 34). The strongest genetic susceptibility factor for BD is located inside the MHC class I region including the Human Leukocyte Antigen-B51 (HLA-B*51). The odds ratio for individuals carrying HLA-B*51/B5 allele to develop BD compared with no-carriers was found to be 5.78 (33).

Genome-wide association studies (GWAS) have clearly shown the role of several single nucleotide polymorphisms (SNPs) in the etiopathogenesis of various diseases, including BD (3541). In a multicenter study, Hughes et al. studied the association between HLA-B*51 and BD as well as other risk loci within the HLA region: 8572 variants were screened, and imputation and meta-analysis of 24834 variants were performed in two independent groups of BD patients. The most significant association was with rs116799036, which is located between HLA-B and MHC Class I Polypeptide-Related Sequence A (MICA) (42). Recently, Takeuchi et al. genotyped 1900 Turkish BD and 1779 controls with the Immunochip and demonstrated that the major BD-related polymorphism was known as rs1050502, an HLA-B*51 gene variant (43). However, the presence of HLA-B*51 alone only partially explains the genetic disease risk and all clinical manifestations of BD. Several recent GWAS have confirmed the association between BD and HLA-B*51, except for Fei et al's investigation. These studies also revealed new susceptibility loci both on other HLA Class I regions and on non-HLA genes (3541). These genes provide a significant role in understanding disease pathogenesis and offer novel treatment strategies.

In general, BD-associated gene polymorphisms were localized in molecules responding to microorganisms, as well as in genes encoding cytokines and adhesion molecules. Polymorphisms within genes encoding the cytokines may affect their function and may be associated with disease predisposition (44). Researchers identified several non-HLA genetic associations by GWAS including ERAP1, IL23 receptor (IL23R), IL-23R/IL-12RB2, IL-10, and STAT genes (38, 45).

ERAP1 variations have been identified as significant predictive loci of BD susceptibility. The gene encodes an amino-peptidase having the critical role to trim N-terminal of peptides. This mechanism was affected by the amino acids sequence of the corresponding protein (4651). ERAP1 is characterized by several common polymorphisms encoding variant amino acids related not only to BD, but also to ankylosing spondylitis (AS) and psoriasis (4751). The same SNPs associated to BD risk resulted protective against AS and psoriasis: this effect depends on the different HLA interacting with ERAP1 (46, 49). ERAP1 polymorphisms was a risk factor preferentially in BD patients with HLA-B*51-positivity; ERAP1 rs17482078 (p.Arg725Gln) might influence the peptide repertoire binding to HLA-B*51 (47). A recent paper suggested the critical role of the altered peptide presentation by HLA-B*51 in influencing disease pathogenesis (52). Based on these alterations, T-cell and natural killer (NK) cell recognition were probably affected, providing the basis for the association of ERAP1 and HLA-B*51 with BD (53). A very recent alternative pathogenic hypothesis linking HLA-B*51 with BD involves the gut microbiome and the HLA-B*51 misfolding. Both ER stress and unfolded proteins were consequences of the misfolding and also the inflammation trigger. Some combination of the misfolded proteins probably influences BD pathogenesis, but this point has not yet been addressed in BD patients and several small studies reported a role in AS pathogenesis with HLA-B-27 (52).

The association between SNPs of IL-10 and IL-23R/IL-12RB2 genes and BD was demonstrated in Turkish (35, 40) and Japanese population (35, 39). A reduced mRNA expression in BD patients monocytes was recognized in the presence of the A-allele of rs1518111 IL-10 compared with wild-type G-allele. PBMCs or monocytes produced significantly less IL-10 following stimulation with Toll-like receptor (TLR) ligands in individuals homozygous for A-allele of rs1518111 (35). Afkari et al. showed that IL-10 rs1800872 A allele contributes to BD genetic risk by modulating IL-10 expression: BD patient group showed lower gene expression levels compared to the controls (54). Most disease-associated GWAS variants were found to be localized on the IL-23R side of the hotspot. These results indicate the association of BD with IL-23R rather than IL-12RB2. The association of IL23R rs17375018 and a haplotype of four gene variations and BD was reported, but no functional data were available for this variation. Targeted resequencing of IL23R in BD Japanese and Turkish patients showed novel association pieces of evidence including the reduced frequency of those rare missense variations with a protective role by reduced IL-23-dependent IL-17 production, as demonstrated in Crohn's disease (35). IL23 induces T cell activation for IL17 production and is one of the most significant activators of Th17 pathway (1). The association between BD susceptibility and IL23R-IL12RB2 locus was confirmed in a Korean population: the intergenic rs1495965 SNP was significantly related with BD risk both in discovery and replication phases (55).

Association between STAT4 rs7574070 and BD was underlined in different studies (35, 37, 38). In addition, the disease-associated A allele was related to increased gene expression, greater severity of disease course and higher IL-17 production (35). IL1A-IL1B, IRF8, and CEBPB-PTPN1 were three novel disease markers recently identified by direct genotyping in GWAS besides ADO-EGR2 discovered by imputation (43).

The variation of the promoter region of TNF has also been reported as a risk marker for BD. Alterations of TNF expression related to gene polymorphisms may be responsible for the higher cytokine activity (56, 57). Polymorphic alleles were more frequent in BD patients and were related to higher TNF production by monocytes or mononuclear cells (45, 57). Mutations in the Mediterranean fever gene were also considered additional BD susceptibility factors (45).

The role of other genes located outside the HLA region, encoding chemokines (e.g., CCR1-CCR3, CCR5), cytokines (such as for IL-1β, IL-6, IL-8, IL-12, IL-17, IL-18, IL-23), oxidative stress-related proteins (glutathione transferase and myeloperoxidase), cell membrane receptors (TNFRSF1A, TLR2, 4, 7, 9), immunoregulatory proteins (e.g., IRF1, IRF5, CTLA-4, NF-jB), extracellular proteins (like ICAM- 1, MMP-9), and others including those for KLRC4, TNFAIP3, DEFA1, NEMO, NOD2, TLR4, and FUT2 were analyzed in several investigations with conflicting findings (45, 5862).

Besides genetic contribution, also epigenetic processes, such as DNA methylation, histone modification, and non-coding RNAs, microRNA (miRNA) in particular, have been suggested as involved in BD pathogenesis (45, 63). The epigenetic aspects were also investigated by analyzing miRNA signatures associated with BD patients with active disease and showed that miRNAs target pathways relevant in BD, such as TNF, IFN-γ, and vascular endothelial growth factor receptor signaling cascades (64, 65). Alipour et al. reported that disease pathogenesis could be affected by altered methylation levels of interspersed repetitive sequences (IRs) elements, as well as by histone modifications and miRNA regulation, in particular, higher levels of miR-182 and miR-3591-3p and lower levels of miR-155, miR-638 and miR-4488 (63).

Recently, Zhou et al. screened a Caucasian family formed by an affected mother and two affected daughters presenting with oral and genital ulcers, uveitis, and arthralgia/arthritis clinical signs. Exome sequencing revealed two strong candidate variants, p.C78W of TNFRSF9 and p.L227X of TNFAIP3 genes. These mutations affect immune cell survival and proinflammatory cytokine production. Therefore, one or two of this mutation may contribute to this dominantly-inherited condition and can help us to understand how BD symptoms develop (66).

Immunity

Activated innate immunity plays an important role in the pathogenesis of BD. Microbial triggers are sensed and processed by the innate immune system via pathogen-related and/or danger-associated molecular patterns. Overproduction of inflammatory cytokines by innate immune cells such as macrophages and dendritic cells may cause a higher production of adaptive Th1- and Th17-related cytokines. BD lesions in their early stages are predominated by neutrophils which are major immunoregulatory cell group of the innate immune system. Another member of innate immunity, natural killer (NK) cells are also found in BD lesions (67).

BD is considered as a neutrophilic vasculitis and the role of neutrophils in BD pathogenesis has long been known (7). Surface molecules, indicating neutrophil activation status (CD10, CD14, and CD16), oxidative burst and phagocytic function of neutrophils have been explored and the presence of proactive neutrophils in BD patients was reported (68). Tissue injury in BD can be modulated by neutrophils in several manners: neutrophils were hyperactivated, probably HLA B*51-associated, and usually were involved in perivascular infiltration (68, 69). No significant differences were observed in the oxidative burst, phagocytic microbicide activities or cytokine pattern when BD patients and controls were compared in Perazzio and colleagues' study. However, significant differences in phagocytic dysfunction were found in patients with severe active disease compared with subjects with mild disease (45, 70). In addition, the structural and functional modification of fibrinogen resulted related to reactive oxygen species and neutrophil activation via neutrophil NADPH oxidase (69). Therefore, neutrophil activation was considered as the main source of oxidative stress through the oxidation of proteins. Hyper-activated neutrophils secrete some cytokines that are both autocrine and also stimulate Th1 cells (45). Recently, Yavuz et al. reported that testosterone causes a significant neutrophil activation together with Th-1 type immune alterations which may explain a more aggressive disease with a higher mortality rate in male BD patients (71).

NK cells were also identified in BD lesions where seems they have a role in driving the CD4+ Th1 response which is the main feature of BD lesions (72, 73). However, several studies underlined increased NK cells in the peripheral blood, in particular during the active phases of the disease (72, 74, 75).

Dysregulation of the immune system contributes to BD etiopathogenesis, with increased systemic levels of inflammatory cytokines (45, 72). It's well known that CD4+T cells can differentiate into two types: Th1 cells subset, which secretes IFN-γ, IL-2, and TNF and promotes cell-mediated immunity, and Th2 cells, which produce IL-4, IL-5, IL-10, and IL-13 and promote antibody-mediated immunity (76).

The alteration of T cell balance, especially Th1/Th17 expansion and decreased regulation by Tregs, are supposed to have a significant role in BD pathogenesis (7, 43). In particular, increased frequencies of Th17 cells were reported in the BD cutaneous lesions (77). Th17 and IL-17 pathways might have a part in the development and/or activity of BD (1). Increased production of IL-17, IL-23, and IFN-γ by PBMCs besides increased frequencies of IL-17 and IFN-γ producing T cells in BD patients with active uveitis was reported (78). IL-17 levels of BD patients with active stages of uveitis, oral and genital ulcers and articular symptoms were significantly higher compared with patients with inactive stages of the same symptoms. Hamzaoui et al. demonstrated that the percentage of circulating Th17 cells and plasma interleukin IL-17 levels were increased in active BD (52, 79). Increased neutrophil activity and neutrophil infiltration in the affected organs of BD might be caused by the increased IL-17 response (80). A recent study reported that, under Th17-stimulating conditions, T cells express both IL-17 and IFN-γ. Production of large amounts of IL-17 and IFN-γ by all lymphocyte subsets in BD patients were associated with increased innate responses, early tissue neutrophil infiltrations and late adaptive immunity (67). Moreover, in experimental autoimmune uveitis (EAU) the role of Herpesvirus entry mediator (HVEM), a member of the Tumor Necrosis Factor Receptor family, has been evaluated. The HVEM seemed to be involved as a co-signaling molecule inducing both Th1 and Th17 responses in EAU. In addition, in the same mouse model, the use of anti-HVEM antibodies blocking HVEM co-signal ameliorated EAU (81).

Takeuchi et al. compared the proinflammatory and Th1-, Th2-, and Th17-related cytokines frequency in a group of BD patients with recurrent uveitis and a group of remitted uveitis before and after infliximab treatment. They found higher levels of IL-1β, IL-4, IL-17A, IL-17F, IL-21, IL-22, IL-31, IFN-γ, sCD40L, and TNF-α, with a significant difference for IL-17F, in BD-recurrent uveitis patients respect to the BD-remitted uveitis group, before drug infusion. In addition, only IL-10 levels were found higher in the remission group than in the other group (82). Emmi et al. showed that cytotoxic Th1 and Th 17 cells can play a role in inducing mucosal damage during the early stages in BD patient with active intestinal involvement (83). These results confirm that Th17 and IL-17 pathway are active and play an important role, particularly in acute attacks of the disease. Conversely, a reduction in Tregs and cytokine IL-10 were notified in the disease (72, 84).

Due to recent progress in molecular methods and basic scientific researches, our knowledge about the disease has considerably increased. GWAS have become a very important step in understanding BD pathogenesis. New genes such as ERAP1 have been introduced which help to understand the possible pathogenic mechanism of HLA-B*51. In the future, similar studies in different populations with a higher number of patients will provide significant advances in the etiopathology of BD. Despite all these advances, clinical expression of the disease is quite heterogeneous and show regional differences. The underlying environmental and genetic factors of this situation are not fully elucidated. Being a complex disease, BD is related more than one pathogenic pathway. Although, management of the disease has evolved noticeably because of more effective and targeted therapies we still need new treatment options for severe and non-responsive cases such as biological treatments developed for the underlying etiopathological mechanism (85).

In conclusion, environmental factors (S. sanguinis etc.) or the differences in salivary or gut microbiome composition can trigger the innate-derived inflammation, which may be subsequently sustained by adaptive immune responses. Epistatic interactions between HLA-B*51 and ERAP1 variants seems to cause T cell homestasis perturbation, especially Th1 and Th 17 activation and Tregs response suppression. The activity of neutrophils is increased and there is an intense neutrophil infiltration in the early stage of inflammation in organs affected by the disease. Association with HLA-B*51 and increased IL-17 response have a key role in the neutrophil activity (Figure 1).

FIGURE 1
www.frontiersin.org

Figure 1. Possible regulation mechanisms in the etiopathogenesis of Behçet’s disease.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. 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) 43:620–32. doi: 10.1111/1346-8138.13381

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Alpsoy E, Donmez L, Onder M, Gunasti S, Usta A, Karincaoglu Y, et al. Clinical features and natural course of Behcet's disease in 661 cases: a multicentre study. Br J Dermatol. (2007) 157:901–6. doi: 10.1111/j.1365-2133.2007.08116.x

PubMed Abstract | CrossRef Full Text | Google Scholar

3. International Study Group for Behçet’s disease. Criteria for diagnosis of Behçet’s disease. Lancet. (1990) 335:1078–80.

4. Gul A. Pathogenesis of Behçet’s disease: autoinflammatory features and beyond. Semin Immunopathol. (2015) 37:413–8. doi: 10.1007/s00281-015-0502-8

PubMed Abstract | CrossRef Full Text | Google Scholar

5. McGonagle D, Aydin SZ, Gül A, Mahr A, Direskeneli H. ‘MHC-I-opathy’-unified concept for spondyloarthritis and Behçet disease. Nat Rev Rheumatol. (2015) 11:731–40. doi: 10.1038/nrrheum.2015.147

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Hatemi G, Karatemiz G, Yazici H. Behçet’s disease: an MHC-I-opathy? Clin Exp Rheumatol. (2017) 35(Suppl 104):5.

PubMed Abstract | Google Scholar

7. Greco A, De Virgilio A, Ralli M, Ciofalo A, Mancini P, Attanasio G, et al. Behçet’s disease: New insights into pathophysiology, clinical features and treatment options. Autoimmun Rev. (2018) 17:567–75. doi: 10.1016/j.autrev.2017.12.006

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Yazici H, Seyahi E, Hatemi G, Yazici Y. Behçet syndrome: a contemporary view. Nat Rev Rheumatol. (2018) 14:119. doi: 10.1038/nrrheum.2018.3

CrossRef Full Text | Google Scholar

9. Lehner T. The role of heat shock protein, microbial and autoimmune agents in the aetiology of Behçet’s disease. Int Rev Immunol. (1997) 14:21–32. doi: 10.3109/08830189709116842

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Behçet H. Uber rezidivierende aphthose, durch ein Virus verursachte Geschwure, am Mund, am Auge, und an den Genitalien. Dermatol Wochenschr. (1937) 105:1152–7.

Google Scholar

11. Studd M, McCance DJ, Lehner T. Detection of HSV-1 DNA in patients with Behçet’s syndrome and in patients with recurrent oral ulcers by the polymerase chain reaction. J Med Microbiol. (1991) 34:39–43. doi: 10.1099/00222615-34-1-39

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Togashi A, Saito S, Kaneko F, Nakamura K, Oyama N. Skin prick test with self-saliva in patients with oral aphthoses: a diagnostic pathergy for Behcet's disease and recurrent aphthosis. Inflamm Allergy Drug Targets. (2011) 10:164–70. doi: 10.2174/187152811795564109

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Yokota K, Hayashi S, Fujii N, Yoshikawa K, Kotake S, Isogai E, et al. Antibody response to oral streptococci in Behcet's disease. Microbiol Immunol. (1992) 36:815–22. doi: 10.1111/j.1348-0421.1992.tb02083.x

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Cho SB, Cho S, Bang D. New insights in the clinical understanding of Behçet’s disease. Yonsei Med J. (2012) 53:35–42. doi: 10.3349/ymj.2012.53.1.35

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Lule S, Colpak AI, Balci-Peynircioglu B, Gursoy-Ozdemir Y, Peker S, Kalyoncu U, et al. Behçet Disease serum is immunoreactive to neurofilament medium which share common epitopes to bacterial HSP-65, a putative trigger. J Autoimmun. (2017) 84:87–96. doi: 10.1016/j.jaut.2017.08.002

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Cho SB, Lee JH, Ahn KJ, Cho S, Park YB, Lee SK, et al. Identification of streptococcal proteins reacting with sera from Behçet’s disease and rheumatic disorders. Clin Exp Rheumatol. (2010) 28:S31–8.

PubMed Abstract | Google Scholar

17. Cho SB, Zheng Z, Ahn KJ, Choi MJ, Cho S, Kim DY, et al. Serum IgA reactivity against GroEL of Streptococcus sanguinis and human heterogeneous nuclear ribonucleoprotein A2/B1 in patients with Behçet disease. Br J Dermatol. (2013) 168:977–83. doi: 10.1111/bjd.12128

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Mumcu G, Ergun T, Inanc N, Fresko I, Atalay T, Hayran O, et al. Oral health is impaired in Behçet’s disease and is associated with disease severity. Rheumatology. (2004) 43:1028–33. doi: 10.1093/rheumatology/keh236

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Kaneko F, Oyama N, Nishibu A. Streptococcal infection in the pathogenesis of Behçet’s disease and clinical effects of minocycline on the disease symptoms. Yonsei Med J. (1997) 38:444–54. doi: 10.3349/ymj.1997.38.6.444

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Mizushima Y, Matsuda T, Hoshl K, Ohno S. Induction of Behçet’s disease symptoms after dental treatment and streptococcal antigen skin test. J Rheumatol. (1988) 15:1029–30.

PubMed Abstract | Google Scholar

21. Alpsoy E. Behçet’s disease: treatment of mucocutaneous lesions. Clin Exp Rheumatol. (2005) 23:532–9.

PubMed Abstract | Google Scholar

22. Akman A, Kacaroglu H, Donmez L, Bacanli A, Alpsoy E. Relationship between periodontal findings and Behçet’s disease: a controlled study. J Clin Periodontol. (2007) 34:485–91. doi: 10.1111/j.1600-051X.2007.01085.x

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Celenligil-Nazliel H, Kansu E, Ebersole J. Periodontal findings and systemic antibody responses to oral microorganisms in Behçet’s disease. J Periodontol. (1999) 70:1449–56. doi: 10.1902/jop.1999.70.12.1449

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Karacayli U, Mumcu G, Simsek I, Pay S, Kose O, Erdem H, et al. The close association between dental and periodontal treatments and oral ulcer course in Behçet’s disease: a prospective clinical study. J Oral Pathol Med. (2009) 38:410–5. doi: 10.1111/j.1600-0714.2009.00765.x

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Kaneko F, Togashi A, Nomura E, Nakamura K. A new diagnostic way for Behçet’s disease: skin prick with self-saliva. Genet Res Int. (2014) 581468. doi: 10.1155/2014/581468

CrossRef Full Text | Google Scholar

26. Akman A, Sallakci N, Kacaroglu H, Tosun O, Yavuzer U, Alpsoy E, et al. Relationship between periodontal findings and the TNF-alpha Gene 1031T/C polymorphism in Turkish patients with Behçet’s disease. J Eur Acad Dermatol Venereol. (2008) 22:950–7. doi: 10.1111/j.1468-3083.2008.02678.x

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Onen F, Tuncer D, Akar S, Birlik M, Akkoc N. Seroprevalence of Borrelia burgdorferi in patients with Behçet’s disease. Rheumatol Int. (2003) 23:289–93. doi: 10.1007/s00296-003-0313-4

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Ersoy O, Ersoy R, Yayar O, Demirci H, Tatlican S. H pylori infection in patients with Behçet’s disease. World J Gastroenterol. (2007) 13:2983–5. doi: 10.3748/wjg.v13.i21.2983

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Pineton de Chambrun M, Wechsler B, Geri G, Cacoub P, Saadoun D. New insights into the pathogenesis of Behçet’s disease. Autoimmun Rev. (2012) 11:687–98. doi: 10.1016/j.autrev.2011.11.026

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Saleh Z, Arayssi T. Update on the therapy of Behçet disease. Ther Adv Chronic Dis. (2014) 5:112–34. doi: 10.1177/2040622314523062

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Coit P, Mumcu G, Ture-Ozdemir F, Unal AU, Alpar U, Bostanci N, et al. Sequencing of 16S rRNA reveals a distinct salivary microbiome signature in Behcet's disease. Clin Immunol. (2016) 169:28–35. doi: 10.1016/j.clim.2016.06.002

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Consolandi C, Turroni S, Emmi G, Severgnini M, Fiori J, Peano C, et al. Behcet's syndrome patients exhibit specific microbiome signature. Autoimmun Rev. (2015) 14:269–76. doi: 10.1016/j.autrev.2014.11.009

PubMed Abstract | CrossRef Full Text | Google Scholar

33. de Menthon M, Lavalley MP, Maldini C, Guillevin L, Mahr A. HLA-B51/B5 and the risk of Behçet’s disease: a systematic review and meta-analysis of case-control genetic association studies. Arthritis Rheum. (2009) 61:1287–96. doi: 10.1002/art.24642

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Gul A, Inanc M, Ocal L, Aral O, Konice M. Familial aggregation of Behçet’s disease in Turkey. Ann Rheum Dis. (2010) 59:622–5. doi: 10.1136/ard.59.8.622

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Gul A. Genetics of Behçet’s disease: lessons learned from genomewide association studies. Curr Opin Rheumatol. (2014) 26:56–63. doi: 10.1097/BOR.0000000000000003

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Lee YJ, Horie Y, Wallace GR, Choi YS, Park JA, Choi JY, et al. Genome-wide association study identifies GIMAP as a novel susceptibility locus for Behçet’s disease. Ann Rheum Dis. (2013) 72:1510–6. doi: 10.1136/annrheumdis-2011-200288

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Kirino Y, Bertsias G, Ishigatsubo Y, Mizuki N, Tugal-Tutkun I, Seyahi E, et al. Genome-wide association analysis identifies new susceptibility loci for Behçet’s disease and epistasis between HLA-B-51 and ERAP1. Nat Genet. (2013) 45:202–7. doi: 10.1038/ng.2520

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Hou S, Yang Z, Du L, Jiang Z, Shu Q, Chen Y, et al. Identification of a susceptibility locus in STAT4 for Behçet‘s disease in Han Chinese in a genome-wide association study. Arthritis Rheum. (2012) 64:4104–13. doi: 10.1002/art.37708

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Mizuki N, Meguro A, Ota M, Ohno S, Shiota T, Kawagoe T, et al. Genome-wide association studies identify IL23R-IL12RB2 and IL10 as Behçet’s disease susceptibility loci. Nat Genet. (2010) 42:703–6. doi: 10.1038/ng.624

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Remmers EF, Cosan F, Kirino Y, Ombrello MJ, Abaci N, Satorius C, et al. Genome-wide association study identifies variants in the MHC class I, IL10, and IL23R-IL12RB2 regions associated with Behçet’s disease. Nat Genet. (2010) 42:698–702. doi: 10.1038/ng.625

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Fei Y, Webb R, Cobb BL, Direskeneli H, Saruhan-Direskeneli G, Sawalha AH. Identification of novel genetic susceptibility loci for Behçet’s disease using a genome-wide association study. Arthritis Res Ther. (2009) 11:R66. doi: 10.1186/ar2695

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Hughes T, Coit P, Adler A, Yilmaz V, Aksu K, Düzgün N, et al. Identification of multiple independent susceptibility loci in the HLA region in Behçet’s disease. Nat Genet. (2013) 45:319–24. doi: 10.1038/ng.2551

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Takeuchi M, Mizuki N, Meguro A, Ombrello MJ, Kirino Y, Satorius C, et al. Dense genotyping of immune-related loci implicates host responses to microbial exposure in Behçet’s disease susceptibility. Nat Genet. (2017) 49:438–43. doi: 10.1038/ng.3786

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Huang Y, Yu H, Cao Q, Deng J, Huang X, Kijlstra A, et al. The association of chemokine gene polymorphisms with VKH and Behçet’s disease in a chinese han population. Biomed Res Int. (2017) 2017:1274960. doi: 10.1155/2017/1274960

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Salmaninejad A, Gowhari A, Hosseini S, Aslani S, Yousefi M, Bahrami T, et al. Genetics and immunodysfunction underlying Behçet’s disease and immunomodulant treatment approaches. J Immunotoxicol. (2017) 14:137–51. doi: 10.1080/1547691X.2017.1346008

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Padula MC, Leccese P, Padula AA, D'Angelo S, Martelli G. ERAP1 molecular characterization: identification of a de novo allelic variant. HLA. (2018) 2018:13262. doi: 10.1111/tan.13262

CrossRef Full Text | Google Scholar

47. Takeuchi M, Ombrello MJ, Kirino Y, Erer B, Tugal-Tutkun I, Seyahi E, et al. A single endoplasmic reticulum aminopeptidase-1 protein allotype is a strong risk factor for Behçet’s disease in HLA-B*51 carriers. Ann Rheum Dis. (2016) 75:2208–11. doi: 10.1136/annrheumdis-2015-209059

PubMed Abstract | CrossRef Full Text | Google Scholar

48. López de Castro JA, Alvarez-Navarro C, Brito A, Guasp P, Martín-Esteban A, Sanz-Bravo A. Molecular and pathogenic effects of endoplasmic reticulum aminopeptidases ERAP1 and ERAP2 in MHC-I-associated inflammatory disorders: towards a unifying view. Mol Immunol. (2016) 77:193–204. doi: 10.1016/j.molimm.2016.08.005

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Ombrello MJ, Kastner DL, Remmers EF. Endoplasmic reticulum-associated amino-peptidase 1 and rheumatic disease: genetics. Curr Opin Rheumatol. (2015) 27:349–56. doi: 10.1097/BOR.0000000000000189

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Reeves E, Edwards CJ, Elliott T, James E. Naturally occurring ERAP1 haplotypes encode functionally distinct alleles with fine substrate specificity. J Immunol. (2013) 191:35–43. doi: 10.4049/jimmunol.1300598

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Kochan G, Krojer T, Harvey D, Fischer R, Chen L, Vollmar M, et al. Crystal structures of the endoplasmic reticulum aminopeptidase-1 (ERAP1) reveal the molecular basis for N-terminal peptide trimming. Proc Natl Acad Sci USA. (2011) 108:7745–50. doi: 10.1073/pnas.1101262108

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Giza M, Koftori D, Chen L, Bowness P. Is Behçet’s disease a ‘class 1-opathy’? The role of HLA-B*51 in the pathogenesis of Behçet’s disease. Clin Exp Immunol. (2018) 191:11–8. doi: 10.1111/cei.13049

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Guasp P, Barnea E, González-Escribano MF, Jiménez-Reinoso A, Regueiro JR, Admon A, et al. The Behçet’s disease-associated variant of the aminopeptidase ERAP1 shapes a low-affinity HLA-B*51 peptidome by differential subpeptidome processing. J Biol Chem. (2017) 292:9680–9. doi: 10.1074/jbc.M117.789180

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Afkari B, Babaloo Z, Dolati S, Khabazi A, Jadidi-Niaragh F, Talei M, et al. Molecular analysis of interleukin-10 gene polymorphisms in patients with Behçet’s disease. Immunol Lett. (2018) 194:56–61. doi: 10.1016/j.imlet.2017.12.008

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Kang EH, Kim S, Park MY, Choi JY, Choi IA, Kim MJ, et al. Behçet’s disease risk association fine-mapped on the IL23R-IL12RB2 intergenic region in Koreans. Arthritis Res Ther. (2017) 19:227. doi: 10.1186/s13075-017-1435-5

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Abdolmohammadi R, Bonyadi M. Polymorphisms of promoter region of TNF-α gene in iranian azeri turkish patients with Behçet’s disease. J Korean Med Sci. (2017) 32:33–7. doi: 10.3346/jkms.2017.32.1.33

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Akman A, Sallakci N, Coskun M, Bacanli A, Yavuzer U, Alpsoy E, et al. TNF-alpha gene 1031 T/C polymorphism in Turkish patients with Behçet’s disease. Br J Dermatol. (2006) 155:350–6. doi: 10.1111/j.1365-2133.2006.07348.x

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Sousa I, Shahram F, Francisco D, Davatchi F, Abdollahi BS, Ghaderibarmi F, et al. Brief report: association of CCR1, KLRC4, IL12A-AS1, STAT4, and ERAP1 With Behçet’s disease in Iranians. Arthritis Rheumatol. (2015) 67:2742–8. doi: 10.1002/art.39240

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Takeuchi M, Kastner DL, Remmers EF. The immunogenetics of Behçet’s disease: a comprehensive review. J Autoimmun. (2015) 64:137–48. doi: 10.1016/j.jaut.2015.08.013

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Hisamatsu T, Naganuma M, Matsuoka K, Kanai T. Diagnosis and management of intestinal Behçet’s disease. Clin J Gastroenterol. (2014) 7:205–12. doi: 10.1007/s12328-014-0488-0

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Mat M, Sevim A, Fresko I, Tuzun Y. Behçet’s disease as a systemic disease. Clin Dermatol. (2014) 32:435–42. doi: 10.1016/j.clindermatol.2013.11.012

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Burillo-Sanz S, Montes-Cano MA, García-Lozano JR, Ortiz-Fernández L, Ortego-Centeno N, García-Hernández, et al. Mutational profile of rare variants in inflammasome-related genes in Behçet disease: a next generation sequencing approach. Sci Rep. (2017) 7:8453. doi: 10.1038/s41598-017-09164-7

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Alipour S, Nouri M, Sakhinia E, Samadi N, Roshanravan N, Ghavami A, et al. Epigenetic alterations in chronic disease focusing on Behçet’s disease: review. Biomed Pharmacother. (2017) 91:526–33. doi: 10.1016/j.biopha.2017.04.106

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Puccetti A, Fiore PF, Pelosi A, Tinazzi E, Patuzzo G, Argentino G, et al. Gene expression profiling in Behçet’s disease indicates an autoimmune component in the pathogenesis of the disease and opens new avenues for targeted therapy. J Immunol Res. (2018) 2018:4246965. doi: 10.1155/2018/4246965

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Puccetti A, Pelosi A, Fiore PF, Patuzzo G, Lunardi C, Dolcino M. MicroRNA expression profiling in Behçet’s disease. J Immunol Res. (2018) 2018:2405150. doi: 10.1155/2018/2405150

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Zhou Q, Laxer R, Pelletier M, Ramaswamy M, Wang H-Y, Chin D, et al. OR9-001 - Exome sequencing in monogenic Behçet-like disease. Pediatr Rheumatol Online J. (2013) 11(supl 1):A184. doi: 10.1186/1546-0096-11-S1-A184

CrossRef Full Text | Google Scholar

67. Deniz R, Tulunay-Virlan A, Ture Ozdemir F, Unal AU, Ozenm G, Alibaz-Oner, et al. Th17-Inducing conditions lead to in vitro activation of both Th17 and Th1 responses in Behçet’s disease. Immunol Invest. (2017) 46:518–25. doi: 10.1080/08820139.2017.1306865

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Eksioglu-Demiralp E, Direskeneli H, Kibaroglu A, Yavuz S, Ergun T, Akoglu T. Neutrophil activation in Behçet’s disease. Clin Exp Rheumatol. (2001) 19(5 Suppl. 24):S19–S24.

PubMed Abstract | Google Scholar

69. Becatti M, Emmi G, Silvestri E, Bruschi G, Ciucciarelli L, Squatrito D, et al. Neutrophil activation promotes fibrinogen oxidation and thrombus formation in Behçet disease. Circulation. (2016) 33:302–11. doi: 10.1161/CIRCULATIONAHA.115.017738

CrossRef Full Text | Google Scholar

70. Perazzio SF, Soeiro-Pereira PV, deSouza AW, Condino-Neto A, Andrade LE. Behçet’s disease heterogeneity: cytokine production and oxidative burst of phagocytes are altered in patients with severe manifestations. Clin Exp Rheumatol. (2015) 33:S85–95.

PubMed Abstract | Google Scholar

71. Yavuz S, Akdeniz T, Hancer V, Bicakcigil M, Can M, Yanikkaya-Demirel G. Dual effects of testosterone in Behçet’s disease: implications for a role in disease pathogenesis. Genes Immun. (2016) 17:335–41. doi: 10.1038/gene.2016.28

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Hasan MS, Ryan PL, Bergmeier LA, Fortune F. Circulating NK cells and their subsets in Behçet’s disease. Clin Exp Immunol. (2017) 188:311–22. doi: 10.1111/cei.12939

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Yamaguchi Y, Takahashi H, Satoh T, Okazaki Y, Mizuki N, Takahashi K, et al. Natural killer cells control a T-helper 1 response in patients with Behçet’s disease. Arthritis Res Ther. (2010) 12:R80. doi: 10.1186/ar3005

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Treusch M, Vonthein R, Baur M, Günaydin I, Koch S, Stübiger N, et al. Influence of human recombinant interferon-alpha2a (rhIFN-alpha2a) on altered lymphocyte subpopulations and monocytes in Behçet’s disease. Rheumatology. (2004) 43:1275–82. doi: 10.1093/rheumatology/keh311

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Suzuki Y, Hoshi K, Matsuda T, Mizushima Y. Increased peripheral blood gamma delta1 T cells and natural killer cells in Behçet’s disease. J Rheumatol. (1992) 19:588–92.

Google Scholar

76. Ben Ahmed M, Houman H, Miled M, Dellagi K, Louzir H. Involvement of chemokines and Th1 cytokines in the pathogenesis of mucocutaneous lesions of Behçet’s disease. Arthritis Rheum. (2004) 50:2291–5. doi: 10.1002/art.20334

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Shimizu J, Takai K, Fujiwara N, Arimitsu N, Ueda Y, Wakisaka S, et al. Excessive CD4+ T cells co-expressing interleukin-17 and interferon-γ in patients with Behçet’s disease. Clin Exp Immunol. (2012) 168:68–74. doi: 10.1111/j.1365-2249.2011.04543.x

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Chi W, Zhu X, Yang P, Liu X, Lin X, Zhou H, et al. Upregulated IL-23 and IL-17 in Behçet patients with active uveitis. Invest Ophthalmol Vis Sci. (2008) 49:3058–64. doi: 10.1167/iovs.07-1390

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Hamzaoui K, Bouali E, Ghorbel I, Khanfir M, Houman H, Hamzaoui A. Expression of Th-17 and RORγt mRNA in Behçet’s Disease. Med Sci Monit. (2011) 17:CR227–34. doi: 10.12659/MSM.881720

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Ekinci NS, Alpsoy E, Karakas AA, Yilmaz SB, Yegin O. IL-17A has an important role in the acute attacks of Behçet’s disease. J Invest Dermatol. (2010) 130:2136–8. doi: 10.1038/jid.2010.114

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Sakoda Y, Nagai T, Murata S, Mizuno Y, Kurosawa H, Shoda H, et al. Pathogenic function of herpesvirus entry mediator in experimental autoimmune uveitis by induction of Th1- and Th17-type T cell responses. J Immunol. (2016) 196:2947–54. doi: 10.4049/jimmunol.1501742

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Takeuchi M, Karasawa Y, Harimoto K, Tanaka A, Shibata M, Sato T, et al. Analysis of Th cell-related cytokine production in Behçet disease patients with uveitis before and after infliximab treatment. Ocul Immunol Inflamm. (2017) 25:52–61. doi: 10.3109/09273948.2016.1158276

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Emmi G, Silvestri E, Bella CD, Grassi A, Benagiano M, Cianchi F, et al. Cytotoxic Th1 and Th17 cells infiltrate the intestinal mucosa of Behcet patients and exhibit high levels of TNF-α in early phases of the disease. Medicine. (2016) 95:49. doi: 10.1097/MD.0000000000005516

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Touzot M, Cacoub P, Bodaghi B, Soumelis V, Saadoun D. IFNa induces IL-10 production and tilt the balance between Th1 and Th17 in Behçet disease. Autoimmun Rev. (2015) 14:370–5. doi: 10.1016/j.autrev.2014.12.009

CrossRef Full Text | Google Scholar

85. Esatoglu SN, Hatemi G, Leccese P, Olivieri I. Highlights of the 17th International Conference on Behçet’s syndrome. Clin Exp Rheumatol. (2016) 34:3–9.

PubMed Abstract | oogle Scholar

Keywords: Behçet’s disease, etiology, genetics, immunology, infectious agents

Citation: Leccese P and Alpsoy E (2019) Behçet’s Disease: An Overview of Etiopathogenesis. Front. Immunol. 10:1067. doi: 10.3389/fimmu.2019.01067

Received: 26 September 2018; Accepted: 25 April 2019;
Published: 10 May 2019.

Edited by:

Angelo Valerio Marzano, University of Milan, Italy

Reviewed by:

Giacomo Emmi, University of Florence, Italy
Fiona Moghaddas, Walter and Eliza Hall Institute of Medical Research, Australia

Copyright © 2019 Leccese and Alpsoy. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Erkan Alpsoy, ealpsoy@akdeniz.edu.tr

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.