Skip to main content
  • Review Article
  • Open access
  • Published:

Behçet’s disease physiopathology: a contemporary review

Abstract

Behçet’s disease, also known as the Silk Road Disease, is a rare systemic vasculitis disorder of unknown etiology. Recurrent attacks of acute inflammation characterize Behçet’s disease. Frequent oral aphthous ulcers, genital ulcers, skin lesions and ocular lesions are the most common manifestations. Inflammation is typically self-limiting in time and relapsing episodes of clinical manifestations represent a hallmark of Behçet’s disease. Other less frequent yet severe manifestations that have a major prognostic impact involve the eyes, the central nervous system, the main large vessels and the gastrointestinal tract. Behçet’s disease has a heterogeneous onset and is associated with significant morbidity and premature mortality. This study presents a current immunological review of the disease and provides a synopsis of clinical aspects and treatment options.

Description

Behçet’s Disease (BD) is a rare systemic vasculitis disorder of unknown etiology characterized by recurrent attacks of oral aphthous ulcers, genital sores, and ocular lesions (triple-symptom complex). Clinical manifestations of acute inflammation are typically self-limiting in time with relapsing episodes of varied intensity leading to sequelae. Frequency and duration of relapses are unpredictable and follow no discernible pattern of onset. The disease occasionally generates severe manifestations involving the cardiovascular system, the central nervous system, and the gastrointestinal tract. Behçet’s Disease is classified as a systemic vasculitis associated with significant morbidity and mortality, particularly in males with early age onset [13]. Currently, there exists no specific diagnostic tool or serum biomarker to identify and quantify the severity of BD; prognosis is difficult to ascertain. Behçet’s disease typically appears between the third and the fourth decades of age, infrequently developing before puberty or after the fifth decade of life. Earlier age onset is correlated with more severe clinical manifestations and mortality [2, 4, 5]. The disease appears to occur more frequently and is associated with more severe ocular, neurological, and cardiovascular clinical manifestations in males [1, 3]. Treatment options are unspecific and aim to relieve symptoms and control disease progression and severity. Disease management usually includes systemic anti-inflammatory and/or immune-modulating drugs [1, 3, 6].

Prevalence of BD worldwide ranges between 0.1/1000 and 1/10,000 with a significant presence in Asian countries 30°–40° north of the Equator from the Mediterranean to Japan, hence the term Silk Road Disease [1, 2]. Turkey has reported the highest prevalence ranging from 80 to 420/100,000 [7]. Disease occurrence in other Asian countries such as Saudi Arabia, Iran, Korea, China, and Japan, has been found to oscillate between 13.5 and 85/100,000 with latter possessing the highest reported incidence in East Asia [8]. Occurrence is far lower in Western countries ranging from 0.12 to 0.64/100,000 [1, 9]. Immigrants from countries with high prevalence tend to have inferior risks of developing the disease pursuant to emigration to low prevalence countries [2, 7]. Disease incidence amongst Turkish individuals in Germany was found to be 21/100,000, which, although significantly higher than that of the German population at large (0.42–0.55/100,000), remains inferior to that of their country of origin [5, 9]. Whereas BD primarily affects males in western Asia, it is more commonplace amongst females in Japan and Korea [5]. Familial aggregation of BD has been reported to occur in 1–18 % of the population, with a higher incidence of familial association in juvenile patients [10]. The pattern of disease inheritance does not follow Mendelian rules as narrated by multi-case family studies [10, 11]. A correlation with HLA class I antigens, notably HLA-B51, has been observed [2, 7].

Clinical features

As a systemic vasculitis disease, BD may affect almost all vascularized systems (Table 1) and is characterized by episodes of relapses and remissions with sequelae [1, 3, 6]. Although several clinical manifestations are associated with BD, the triple-system complex of oral and genital aphthae, and uveitis first described by Behçet in 1937 generally illustrates disease pattern. Children exhibit more frequent perianal aphthosis and arthralgia, less frequent genital ulcers and vascular involvement and a more severe course of uveitis [1, 1012]. Over the past few years, modern treatment strategies, involving immunosuppressant therapy, and the use of aggressive approaches have led to improvements in the prognosis of severe forms of BD [13, 14]. Prognosis for the disease is usually reserved, especially when ocular, cardiovascular, neurological, and gastrointestinal manifestations appear [6, 15, 16].

Table 1 Main manifestations of Behçet’s disease

Oral ulcers

Recurrent oral ulcers represent the earliest disease manifestation in 47–86 % of patients [16]. It may take years for the other symptoms to appear afterwards, and oral ulcers are observed in all patients during their clinical course. Lesions have disciform appearance with round and sharp erythematous border, covered with a grayish-white pseudomembrane or a central yellowish fibrinous base and grow rapidly from a flat ulcer to a deep sore [1]. They may occur as single ulcers or in crops and heal with little scarring; prognosis is usually favorable [1, 5]. Oral ulcers most commonly affect the gingival and buccal mucosa, tongue and lips yet may also appear in the soft and hard palates, pharynx, and tonsils [1]. Minor ulcers (<1 cm in diameter) heal without scarring in 4–14 days whereas major ulcers (>1 cm in diameter) are more painful and heal within 2–6 weeks. Herpetiform ulcers occur in recurrent crops of small ulcers that are 0.2–0.3 cm in diameter are painful and may coalesce. Treatment is usually symptomatic and prognosis of oral ulcerations is favorable [5, 16].

Genital ulcers

Genital ulcers develop in 57–93 % of patients [17]. They are painful and morphologically resemble oral ulcers, but are larger, deeper, have more irregular margins and heal with white or pigmented scars [16]. Male genital lesions most commonly involve the scrotum and usually leave a scar that will help with the diagnosis retrospectively. They may also affect the epididymis; penile lesions are less frequent [1]. In females, vulvar, vaginal and cervical lesions are especially common [5]. Rarely, deep vaginal lesions may perforate the bladder resulting in fistulae [16, 17]. Both males and females may develop perineal, perianal and groin lesions [16]. In cases of fistulae and internal lesions, prognosis is unfavorable when the infectious risk is inadequately measured [1, 5].

Ocular manifestations

Ocular disease, involving the retina and the uvea, occurs in 30–70 % of BD patients and is associated with high morbidity [15]. It is the primary cause of blindness in approximately 25 % of patients despite aggressive corticosteroid treatment [15]. Ocular symptoms occur more frequently amongst males and are associated with disease severity, even though prognosis is improving with the use of aggressive immunosuppressant therapy [1, 5]. They usually occur 2–3 years after the onset of oral or genital ulcerations but remain the first disease manifestation in 10–20 % of patients [1]. Ocular disease is a chronic relapsing bilateral non-granulomatous uveitis that may involve the anterior segment, the posterior segment or both (panuveitis) [6, 13]. The latter is associated with worse prognosis and is more common among males [1]. Ocular disease is characterized by the formation of hypopyon—a visible layer of pus in the anterior chamber—observed in one-third of patients [5]. Episodes of anterior uveitis subside spontaneously; repeated attacks result in irreversible structural deformities [13, 17]. Ocular manifestations also include iridocyclitis, keratitis, episcleritis, scleritis, vitritis, vitreous hemorrhage, retinal vasculitis, retinal vein occlusion, retinal neovascularization and optic neuritis [15].

Ocular symptoms include blurred vision, photophobia, lacrimation, floaters, hyperemia and periorbital or global pain [1, 5]. Recurrent inflammatory attacks are associated with secondary complications such as posterior and peripheral anterior synechia, iris atrophy, cataract resulting from inflammation or treatment, secondary glaucoma (occasionally neovascular), atrophic retina, optic atrophy, macular edema, macular degeneration, retinal veins occlusion, sheathed vessels, chorioretinal scars, and proliferative vitreoretinopathy and phthisis bulbi [15]. Prognosis is correlated with frequency, severity of ocular inflammation, and extent of lesions, and remains in many cases unfavorable.

Cutaneous manifestations

Skin involvement affects 38–99 % of BD patients [1, 3]. Cutaneous manifestations commonly include papulopustular (28–96 %) and acne-like lesions [10, 17]. Wounds exhibit a wide distribution affecting the face, limbs, trunk, and buttocks [1]. Skin lesions are characterized by thrombosis and vasculitis [3]. Early lesions exhibit leukocytoclastic vasculitis or neutrophilic vascular reactions whereas mature lesions are characterized by lymphocytic vasculitis [17].

Erythema nodosum lesions occur in 15–78 % of patients, mainly in females, in the lower limbs [1, 5]. Erythema nodosum lesions do not ulcer and may heal spontaneously leaving residual pigmentation. Cutaneous ulcers are rare and only affect 3 % of BD patients [1]. They resemble aphthous ulcers, are recurrent and usually heal with scarring, in the neck, breast, axillae, inguinal region, legs and interdigital skin of the feet [16]. Prognosis for most cutaneous lesions in BD is usually favorable.

Neurological manifestations

Neurological involvement in BD (neuro-BD) occurs in 5–10 % of patients and is more frequent in males [18, 19]. It usually occurs around 5 years after the onset of the disease and is associated with long-term morbidity and mortality [1, 4, 20]. Neurological disease affects the central nervous system (CNS) more frequently than the peripheral nervous system [21, 22].

Neuro-BD may be parenchymal (80 % of patients), non-parenchymal or mixed brain disease [1, 18]. Parenchymal brain disease affects the brainstem and/or basal ganglia and is correlated with a poor prognosis [18, 23]. Non-parenchymal brain disease is characterized by subsets of cerebral venous thromboses, notably dural sinus thrombosis, arterial vasculitis and aseptic meningitis and comprises the most devastating symptom category of BD resulting in a range of neurological deficits that may lead to coma and death [18, 19].

Headache syndromes represent the most common neurological symptom and occur in 70 % of patients [18]. Most parenchymal neuro-BD cases present as subacute meningoencephalitis that exhibit subacute onset and is associated with exacerbation of systemic manifestations [21, 22]. Flare-ups reach a peak within few days and may last for periods of weeks. Brainstem involvement, including ophthalmoparesis, cranial neuropathy and cerebellar or pyramidal dysfunction, has additionally been reported [1, 18, 23, 24]. Cerebral or spinal cord involvement has been observed in association with subcortical dementia and accompanied by ataxia [21, 24]. Cerebral hemispheric involvement, including encephalopathy, hemiparesis, hemisensory loss, seizures and dysphasia, and mental changes, including cognitive dysfunction and psychosis, have been observed as well [18, 20, 24]. Spinal cord involvement, manifested by pyramidal signs in the limbs, sensory level dysfunction and sphincter dysfunction, has also been reported [18, 25]. Other less common clinical symptoms involving the CNS have been reported such as stroke, epilepsy, brain tumor-like neuro-BD, movement disorders, acute meningeal syndrome, optic neuropathy, spinal cord involvement and asymptomatic and subclinical neurological involvement [21, 24, 25].

Non-parenchymal neuro-BD, also referred to as vasculo-BD or angio-BD, involves the main vascular structures of the CNS [23]. Clinical syndromes include vascular disorders, intracranial hypertension and intracranial aneurysms [26]. More rarely, mixed parenchymal and non-parenchymal disease has also been reported [18].

Classically, meningitis or meningoencephalitis, neurological deficits including motor disturbances and brainstem symptoms and psychiatric symptoms including personality changes develop in patients with neuro-BD [5, 18]. These symptoms are associated with disease exacerbations and gradually cause irreversible disability [5]. At late stages, dementia develops in approximately one-third of patients. Cognitive impairment in neuro-BD patients includes poor memory, attention and motivation, in addition to personality changes [27]. Prognosis for neuro-BD, in all its forms, is unfavorable [17, 18].

Cardiovascular manifestations

Behçet’s disease may affect blood vessels of different sizes and types, including arteries and veins, as well as the heart organ [28]. Cardiovascular features have been reported to affect 7–49 % of patients, more frequently in males [1, 29]. They occur 3–16 years after the onset of BD [30].

Vascular BD commonly affects veins causing recurrent superficial thrombophlebitis and deep venous thrombosis in 30–40 % of patients [18, 31, 32]. Thromboses of the superior and inferior vena cava (0.2–9 % of patients), dural sinuses and supra-hepatic veins (2–3.2 % of patients) and pulmonary arterial aneurysms (1 % of patients) are associated with poor prognoses [1, 29, 34]. Occlusion and aneurysms of major arteries commonly lead to bleeding, infarction and organ failure, particularly in instances of pulmonary aneurysm [5, 33]. Rupture of aneurysms may be fatal. At the level of lungs, thrombosis, aneurysm and arteriobronchial fistula cause recurrent episodes of dyspnea, cough, chest pain, and hemoptysis [5].

Cardiac involvement includes pericarditis, myocarditis, endocarditis, mitral valve prolapse, valve lesions, intracardiac thrombosis, endomyocardial fibrosis, myocardiopathy, and coronary artery lesions [30, 34, 35]. The prognosis in these cases is unfavorable with frequent recurrences [1]. The highest direct mortality rate in cardiovascular involvement was attributed to large vessel vasculitis as a result of sudden death by aneurysm rupture or thrombosis (up to 9.8 % of patients in one Turkish study) [4, 12, 30].

Articular manifestations

Articular involvement occurs in 45–60 % of patients and includes arthralgia, monoarthritis or polyarthritis [36, 37]. Non-erosive, non-deforming oligoarthralgia commonly involving the knees, ankles, elbows, and wrists is the most frequent manifestation [1, 5]. Neutrophilic and mononuclear cell infiltrates in the synovium and small vessel lesions with thrombosis typically characterize articular disease [5]. Destructive changes rarely occur in patients with articular involvement [37]. As anti-inflammatory treatment is generally effective, prognosis is usually favorable [38].

Gastrointestinal manifestations

Gastrointestinal involvement occurs in 3–26 % of patients and varies among different populations [39, 40]. It is much more frequent in Japan than in the Middle East and the Mediterranean region [13]. Mucosal inflammation and ulceration occur throughout the gastrointestinal tract and are mostly common in the ileocecal region [40, 41]. The esophagus, ascending colon, and transverse colon are less frequently involved [5]. Clinical symptoms include anorexia, vomiting, dyspepsia, diarrhea, melena, abdominal pain and, less frequently, perforation requiring surgical intervention [39, 41]. Prognosis is unfavorable as gastrointestinal involvement is typically acute and chronic.

Diagnosis

A Behçet’s disease diagnosis is typically confirmed by elimination of other disease scenarios, even when a triple-symptom complex is evident [10]. As there exists no targeted diagnostic test for BD, diagnosis of clinical symptoms is challenging, especially when symptoms are non-concomitant. The International Chapel Hill guidelines of 1990 (revised in 2012) propose the presence of recurrent oral ulcerations (at least three times in 1 year) plus any two of the following: recurrent genital ulceration (aphthous ulceration with scarring); eye lesions (anterior uveitis, posterior uveitis, cells in vitreous on slit lamp examination, retinal vasculitis); skin lesions (erythema nodosum, pseudofolliculitis or papulopustular lesions, acneiform nodules in post-adolescent patients not on corticosteroid therapy); and/or a positive pathergy test (read by a physician at 24–48 h) (Supplementary Table 1) [42]. Nevertheless, given the difficulty of diagnosis, it is vital for clinicians to thoroughly investigate the clinical history of BD patients to exclude diseases with similar manifestations before providing confirmation (Table 2) [1].

Table 2 Common erroneous diagnoses presented to Behçet’s disease patients

Behçet’s disease is characterized by episodes of relapses and remissions, and is associated with periods in which patients may exhibit few overt clinical symptoms [1].

Elevated levels of various inflammatory markers are sensitive (but not specific) to BD including C-reactive protein, erythrocyte sedimentation rate, peripheral leukocyte and platelet counts and serum cytokines (TNF-α, IFN-γ, IL-1β, IL-6, and IL-8) [35, 43]. Moderate anemia of chronic disease may also be present; however, autoantibodies, such as antinuclear, rheumatoid factor, cryoglobulinemia and antineutrophil cytoplasmic antibodies (ANCA) have not been detected in BD, hence the classification of the disease as a non-ANCA vasculitis [43]. Pathergy is a characteristic feature of BD and is characterized by a non-specific hyper-reactivity of the skin to minor trauma [1, 10]. The positivity of the test, reduced upon surgical pre-cleansing of the skin, also varies with geographical location (more than 60 % of Middle Eastern patients, 15 % of Korean patients and 5 % of Caucasian patients) [1].

Over the years, five major independent sets of criteria have been used for diagnosing BD, each characterized by its own clinical features and frequencies, as well as nature of criteria that should be met in order for the diagnosis to be positive [44, 45]. All required a confirmation of the three major symptoms initially described by Behçet as a separate clinical entity (oral ulceration, genital ulceration, and eye lesions). The International Study Group (ISG) for BD proposed the main international standards for diagnosing BD [1]. The study group compared the performance of the previously defined sets of criteria and established a new set of symptom manifestations for BD. These criteria, published in 1990 and last updated in 2013, provided simpler means for diagnosis of BD, excluded rare and subjective features, and showed more specificity with little or no loss of sensitivity to previously used tools [5, 48].

In 2004, Lawton et al. conducted a study in an attempt to define a set of clinical features that may be used as a standard index for measurement of BD activity [8]. Five countries participated in this study, namely China, Korea, Iraq, Turkey and UK. Using the Rasch method, the study analyzed 14 items that formed an index of disease activity (Supplementary Fig. 1) [8]. This BD Current Activity Form is used to develop an overall score for a disease activity index in clinical trials on BD involving therapeutic interventions [46].

Pathogenesis

Various hypotheses attempt to shed light on the etiology of BD including innate and adaptive immunity actors as well as environmental factors [47]. Epidemiological data suggest the involvement of both genetic and environmental factors in the development of BD [5, 10, 45]. The disease is assumed to stem from an autoimmune response, which, in turn may be triggered by infectious or other environmental agents in genetically susceptible individuals [1, 5] (Fig. 1).

Fig. 1
figure 1

Summary of pathogenesis of Behçet disease. It is presumed that environmental factors trigger CD4+ T cells activation in genetically susceptible individuals, leading to the secretion of cytokines that stimulate other inflammation-inducing immune cells resulting in an uncontrolled autoimmune cascade in vascular tissue. SNP single nucleotide polymorphism, IL interleukin, HLA human Leukocyte Antigen, HSP heat shock proteins, INFγ interferon gamma, TNF-α tumor necrosis factor alpha, CD mature Th cells expressing a surface protein identified by a number, NKT natural killer T cells, Th T helper cells, ϒσ T cells T lymphocytes

Four criteria support evidence of genetic influence on susceptibility to BD: peculiar geographical distribution, occasions of familial aggregation, correlation with class I HLA antigens (HLA-B51) and polymorphisms in genes that control immune responses [2, 7, 48, 49]. The fact that Turkish immigrants in Germany had significantly higher disease prevalence than Turkish nationals who did not immigrate also corroborates the environment influence on susceptibility [7, 54]. Yazici et al. and Ohno et al. reported a close association between HLA-B5 and BD development [7, 54]. They demonstrate that HLA-B5 is heterogeneous in composition and includes at least HLA-B51 and HLA-B52. Within the MHC locus, HLA-B51 and HLA-B5701 were associated with disease pathogenesis, most notably in populations along the Old Silk Route. In Iranian Azari patients, HLA-B35 frequency was higher in the patient than control group, and the frequency of HLA-B51, HLA-B52, and HLA-BW4 was significantly elevated [50].

However, the reported risk increase varied between studies from 1.3 to 16 [7, 54]. Similarly, various independent associations of HLA-A and HLA-C regions have been described, with unspecific evidence that remains to be confirmed [51, 52]. Other genes from the MHC locus have additionally been investigated, including several TNF genes and the MHC class I-related gene (MICA), yet their exact involvement remains undetermined as they may conceivably be activated a posteriori by an HLA-B51-induced inflammatory cascade [55, 58]. Ombrello et al. depicted an association of the MHC-I loci and BD [53]. They propose that the peptide-binding specificity of MHC-I molecules by killer immunoglobulin-like receptors on natural killer cells and T lymphocytes plays a primary role in autoimmunity. Non-MHC genes, namely interleukin 1 (IL-1), coagulation factor V, intercellular adhesion molecule 1 (ICAM-1), and endothelial nitric oxide synthase (eNOS) have been also investigated in BD pathogenesis [56, 59]. Although several associations were determined, definitive conclusions have not been established as to their precise role. Similarly, association findings of genome-wide association studies have identified also established associations with IL10, IL23R, CCR1, STAT4, KLRC4, GIMAP2/GIMAP4, and UBAC2 genes in Behçet’s disease patients of different ethnicities. Deep resequencing of targeted genes identified additional associations with rare variants in TLR4, MEFV, and NOD2 genes as well as shared inflammatory pathways with spondyloarthropathies [51, 52].

Infectious agents such as bacteria and/or viruses have been implicated as possible instigators of a dysfunction in inflammatory response in BD patients [45, 54]. However, most viral, fungal, bacterial and parasitic causes have been eliminated as disease instigators; scans for microorganisms such as Streptococcus sanguis, Herpes simplex virus, Epstein–Barr virus and cytomegalovirus lead to negative results in BD patients during periods of both flare-up and remission [45, 55, 58]. Recent results indicate that both a peculiar dysbiosis of the gut microbiota and a significant decrease of butyrate production are present in patients with BD patients. As butyrate is able to promote differentiation of T regulatory cells, a defect of butyrate production might lead to both reduced Treg responses and activation of immuno-pathological T-effector responses [56, 59].

Matzinger proposed that immune responses in situations similar to BD develop from a continuous autoimmune cascade as a result of signals emitted by injured host cells [55, 58]. This ‘danger model’ reaction proposes that the immune reaction overreacts to external stimuli. T cells and other antigen-presenting cells (APC) take hold of a process that feeds on itself in cases of a favorable genetic terrain. The ‘pattern-recognition theory’ consigns the injury by a ‘non-self’ entity to a permanent aggression, activating an uncontrolled adaptive response. Heat-shock proteins (HSP60) given their resemblance with pathogenic proteins have been proposed in this context [61, 64]. Medzhitov proposes that autoimmune disease such as BD may be an autoantigen-derived autoinflammatory disease [56]. This adaptive reaction to external triggers persists in situations of permanent pathogenic presence via autoantigen-activated dendritic T or B cells. This explanation might help to explain why both anti-microbial agents and immunosuppressant therapies may come across as effective in autoimmune diseases in general and BD in particular [57].

The depiction of Toll-like receptors (TLRs) as innate structures that detect and react to permanent proteins in infectious agents additionally suggests mechanisms by which the body’s reaction to ‘non-self’ develops into a state of autoinflammation [50, 62]. This may play a role in BD for several reasons: either the loss of built-in regulation of TLRs, or the erroneous recognition of self-proteins by TLRs [62]. Cohen proposed that TLRs and the receptors IL-1, IL-18, and IL-33 play a crucial role in these circumstances: a state of hyperactivation becomes permanent, which brings about inflammatory tissue damage as observed in BD [58].

Histopathological observations in BD indicate diffuse necrosis, along with extensive intraluminal transudative fluid [59]. Extravasation of red blood cells has also been noted, suggesting occlusion with recanalization with rare areas of perforation [39, 42]. Diffuse venous thrombosis and focal myonecrosis are likewise frequent [18, 32]. Examined tissue exhibits necrotic and cystic parenchyma, along with high infiltration by mononuclear and polymorphonuclear leukocytes [60, 65]. Dense polymorphonuclear infiltration is commonly observed around arterioles, and is comprised mainly of eosinophils [19]. Macrophages are abundantly observed around venules, which, unlike arterioles, exhibit sparse acute inflammatory cells in infiltrates comprised mainly of lymphocytes [27, 32]. Areas surrounding lesions commonly exhibit edema, astrocytosis, and macrophage infiltrates with scattered microglial nodules [48]. Lymphocytic infiltrates are predominantly constituted of CD3+ T cells, of which CD4+ or CD8+ T cells can be (rarely) identified; CD20+ B cells are also present [6164]. Some CD68+ macrophages and monocytes have been observed [65]. Consequently, a role for pro-inflammatory mediators secreted by infiltrating T cells and monocytes in inducing apoptosis of neurons has been suggested.

Several studies reported the involvement of specific cytokines as mediators in BD [6670]. Peripheral blood of BD patients contains IFN-γ and IL-12, suggesting the involvement of Th1 cells [7173]. Elevated IL-1, IL-6, IL-18, TNF-α and chemokine levels are thought to account for activation of innate and adaptive immune responses [73, 75]. Neutrophils of BD patients are hyperactivated, as shown by their increased phagocytosis and superoxide production, enhanced chemotaxis and elevated production of lysosomal enzymes [57, 73]. Accordingly, lymphocytes of BD patients exhibit abnormal functions as clonal expansion of autoreactive T cells specific for HSP60 peptides [59, 74]. Moreover, γδ T cells are more prevalent in blood and mucosal lesions of BD patients [42, 58]. It has consequently been suggested that interactions between T cells, neutrophils and APCs may contribute to the pathogenesis of BD (Fig. 1) [19, 57, 60, 77].

Saadoun et al. and Geri et al. additionally reported evidence for the involvement of IL-21 in BD [35, 75]. Both studies observed that Th1 cells and IL-21 were significantly increased in serum of active BD patients compared to patients in remission and healthy controls [41, 76, 81]. The addition of serum from active BD patients to that of healthy controls increased the frequency of Th17 and Th1 cells and decreased FoxP3 expression in CD4+ T cells. IL-21-producing CD4+ T cells were notably expanded in peripheral blood of patients with active disease compared to those in remission and healthy controls, and were positively correlated with Th17 cells, and negatively correlated with Tregs [7779]. Levels of the other IL-17A-promoting cytokines (IL-1β, IL-6, TGF-β, and IL-23) did not show significant difference between the active, remission, and healthy groups [73, 78, 84]. The frequency of Tregs was significantly decreased in BD patients with active disease and remission compared to healthy controls; a similar pattern was noted for activated Tregs, notably in cases of CNS involvement [78]. These results suggest that IL-21 induces the differentiation of Th17 cells and the production of Th17 and polymorphonuclear leukocyte chemoattractants in CNS lesions of BD patients [80, 81]. IL-21 also demonstrated a role in T cell homeostasis of BD [81, 82]. IL-21 may contribute to the pathogenesis of BD by disrupting the equilibrium between Th17 cells and Tregs [77, 83]. This is consistent with the findings of Nurieva et al. who demonstrated the expression of IL-21 by mouse Th17 cells and its autocrine induction of Th17 cell differentiation while inhibiting the expression of FoxP3 [74]. The expression of IL-17 and IL-21 mRNA was significantly increased in Th17 cells compared to other effector cells [45, 84, 85]. IL-21 expression appeared to be induced by IL-6 alone, rather than by an adjoined effect of IL-6 and TGF-β [35, 82, 84]. IL-21 also upregulated its own expression, indicating an autocrine regulation [35, 50, 86]. IL-21 expression was also shown to be regulated by STAT3, unlike IL-17 expression, which was reported to be regulated by RORγ [74]. STAT3-deficient Th cells failed to produce IL-21 mRNA and protein, which were normally expressed in RORγ-deficient Th cells [90]. IL-21 thus appeared to play an important role in Th17 cell differentiation; activated naïve Th cells differentiated into Th17 cells in the presence of IL-21 [89]. This effect of IL-21 was synergized by the addition of TGF-β [82]. Combined treatment enhanced the generation of Th17 cells and inhibited FoxP3 expression [76, 84].

IL-23 was also found to potentiate the effect of IL-21/TGF-β co-treatment as IL-21 upregulated the expression of IL-23R, RORγ and Th17 cytokines and inhibited the expression of TGF-β and FoxP3 [75]. These results indicate that IL-21 selectively regulates Th17 differentiation [70, 79, 89].

Vollmer et al. reported differential effects for IL-21 during the initiation and progression of Experimental Allergic Encephalomyelitis (EAE) [83]. Treatment of mice with IL-21 prior to EAE induction significantly increased disease severity [67]. In contrast, administration of IL-21 during EAE progression had less effect [83]. IL-21 did not affect antigen-specific T cell proliferation, but rather increased autoreactive Th1 cells upon pretreatment [82]. IL-21 also showed differential effects on IFN-γ production by natural killer cells that was enhanced upon pretreatment [67, 70]. IL-21 pretreatment significantly increased the levels of circulating IgG and IgG2b antibodies, thus demonstrating an effect for IL-21 on natural killer cells and on autoantibody production [70, 74]. IL-21 seems to be induced in T cells by IL-6 and is necessary and sufficient for inducing Th17 cell differentiation via STAT3-dependent upregulation of RORγ [72, 74]. Thus, IL-21 acts in an autocrine manner, similar to the action of IFN-γ in Th1 cells and IL-4 in Th2 cells [46, 87]. IL-21 also appears to modulate the balance between Th17 cells and Tregs in an experimental model of colitis [86]. In this latter model, IL-21 treatment reduced the TGF-β-mediated expression of FoxP3 in CD4+ T cells and the suppressive function of Tregs, and preferentially induced the expansion of FoxP3 T cells [89]. Autocrine IL-21 expression suppresses the induction of Tregs and induces the generation of Th17 cells [70, 76, 8486].

Furthermore, the effects of IL-21 on the differentiation of B cells and generation of plasma cells were reported by Ozaki et al. [67, 88]. IL-21 was found to enhance Ig production, isotype switching and production of plasma cells. Habib et al. reported the requirement of γc chain for IL-21R signaling and for IL-21-induced activation of the JAK/STAT pathway [96]. This study also demonstrated that γc-associated JAK3 tyrosine kinase is an important mediator of γc-dependent IL-21 signaling [89].

Reduced Tregs in BD patients have been shown to suppress antigen-specific T cell responses [71]. Immune tolerance towards self-antigens is initiated in the thymus and involves clonal deletion of autoreactive T lymphocytes. Peripheral exposure to extrathymic self-antigens as well as nonpathogenic foreign substances also plays a role in immune tolerance [60]. One proposed mechanism for peripheral tolerance involves active immune suppression mediated by T cells [71]. This study reported the generation of a unique CD4+ T cell subset that displayed immunoregulatory properties [71]. Mouse ovalbumin-specific naïve CD4+ T cells stimulated with splenic APCs and ovalbumin peptide in the presence of IL-10 displayed a cytokine profile (high levels of IL-10 and IL-5 and low levels of IL-2 and IL-4) that was distinct from that of Th0, Th1 or Th2 cells, and exhibited low proliferation in response to antigenic stimulation [46, 90, 94]. These IL-10-induced cells, now known as Tregs, were designated T regulatory cells 1 (Tr1) [11, 71]. It is worth mentioning that antigen-specific Tr1 cells could be generated in vitro from naïve CD4+ T cells of humans as well [80, 94, 97]. It was further demonstrated that antigen-specific activation of Tr1 inhibits antigen-specific proliferation of other T cells, and thus suppresses antigen-specific T cell responses, in both mice and humans [85, 86]. This may be explained by the cytokine profile of Tr1 cells that includes high levels of the immunosuppressive cytokine IL-10 and low levels of the T cell growth-promoting cytokines IL-2 and IL-4 [82, 93, 97]. Tr1 cells in essence prevented the development of T cell-mediated disease in mice.

Treatment

As no curative solution is currently available, treatment of BD attempts to relieve symptoms, resolve inflammation, limit tissue damage, reduce recurrence frequency and severity, and avoid life-threatening complications. Choice of treatment depends on the combinations of clinical symptoms and the severity of organ involvement, with priority given to treatment of ocular, gastrointestinal, CNS, and cardiovascular manifestations [6, 44, 45].

A variety of approaches have been proposed for BD including anti-inflammatory and immunosuppressive therapies [3, 14, 91]. Long-term treatment has been associated with significant adverse effects and comorbidity and no treatment protocol has demonstrated enduring healing outcomes [83, 92, 93]. Treatment in refractory disease usually involves a combination of therapies including corticosteroids in addition to chemical immunosuppressants and/or biologic monoclonal antibodies (Table 3) [17, 49, 50]. The European League Against Rheumatism (EULAR) proposed comprehensive treatment guidelines for the management of BD [5]. The multidisciplinary recommendations combined current evidence from clinical trials with input from an expert multinational committee (Table 4) [1, 44, 94].

Table 3 Characteristic therapies for Behçet’s disease
Table 4 EULAR general treatment recommendations for the treatment of Behçet’s disease

As mentioned above, prior evidence suggests the involvement of IL-17, IL-21, and TGF-β in the pathogenesis of BD as the level of these markers was correlated with disease activity [73, 78, 82, 84]. Treatments that aim to target these cytokines may constitute a promising target for novel therapy [94, 95, 97, 98]. In this context, IL-21 blocking agents (neutralizing antibodies or receptors) have been suggested for patients with severe or refractory BD, given that IL-21 blockade restored the altered balance between Th17 cells and Tregs [35, 67, 70, 89]. Further studies are also warranted to elucidate signaling pathways associated with IL-17 in BD patients with the goal of developing targeted therapies [73, 75, 90]. Similarly, the regulation of anti-inflammatory cytokines such as TGF-β by current treatments such as Aprelimast show significant promise [95].

Conclusion

Irrespective of recent therapeutic advancements, the clinical prognosis of BD patients remains unfavorable. The primary goal is to focus on identifying the pathogenesis of the disease and providing therapies that inhibit the underlying autoimmune inflammatory cascade. Current research points towards targeted cytokines that play a role in the physiopathology of Behçet’s disease.

Highlights

  • Recurrent oral and genital ulcers and ocular lesions characterize Behçet’s disease (BD).

  • Severe manifestations involve the eyes, the CNS, the main large vessels and the gastrointestinal tract.

  • Innate and adaptive immunity responses result in an uncontrolled autoimmune cascade in vascular tissue.

  • Environmental factors trigger T cells activation in genetically susceptible individuals, resulting in secretion of cytokines that stimulate other inflammation-inducing immune cells.

  • New treatments point towards targeted cytokines such as TNF-alpha, IL-21 and IL-17.

Abbreviations

SNP:

Single nucleotide polymorphism

IL-:

Interleukin

HLA:

Human leukocyte antigen

HSP:

Heat-shock proteins

INFγ:

Interferon gamma

TNF-α:

Tumor necrosis factor alpha

CD:

Mature Th cells expressing a surface protein identified by a number

NKT:

Natural killer T cells

Th:

T helper cells

ϒσ T cells:

T lymphocytes

References

  1. Mendes D, Correia M, Barbedo M, Vaio T, Mota M, Gonçalves O, Valente J (2009) Behçet’s disease—a contemporary review. J Autoimmun 32(3–4):178–188

    Article  CAS  PubMed  Google Scholar 

  2. Verity DH, Marr JE, Ohno S, Wallace GR, Stanford MR (1999) Behçet’s disease, the silk road and HLA-B51: historical and geographical perspectives. Tissue Antigens 54(3):213–220

    Article  CAS  PubMed  Google Scholar 

  3. Sfikakis PP, Markomichelakis N, Alpsoy E, Assaad-Khalil S, Bodaghi B, Gul A et al (2007) Anti-TNF therapy in the management of behçet’s disease-review and basis for recommendations. Rheumatology (Oxford) 46(5):736–741

    Article  CAS  Google Scholar 

  4. Saadoun D, Wechsler B, Desseaux K, Le Thi Huong D, Amoura Z, Resche-Rigon M, Cacoub P (2010) Mortality in Behçet’s disease. Arthritis Rheum 62(9):2806–2812

    Article  CAS  PubMed  Google Scholar 

  5. Sakane T, Takeno M, Suzuki N (1999) Behçet’s disease. N Engl J Med 341(17):1284–1291

    Article  CAS  PubMed  Google Scholar 

  6. Sfikakis PP (2002) Behçet’s disease: a new target for anti-tumor necrosis factor treatment. Ann Rheum Dis 61(Suppl 2):51–53

    Article  Google Scholar 

  7. Yazici H, Akokan G, Yalçin B, Müftüoğlu A (1977) The high prevalence of HLA-B5 in Behçet’s disease. Clin Exp Immunol 30(2):259–261

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Lawton G, Bhakta BB, Chamberlain MA, Tennant A (2004) The Behçet’s disease activity index. Rheumatology (Oxford) 43(1):73–78

    Article  CAS  Google Scholar 

  9. Savey L, Resche-Rigon M, Wechsler B, Comarmond C, Piette JC, Cacoub P, Saadoun D (2014) Ethnicity and association with disease manifestations and mortality in Behçet’s disease. Orphanet J Rare Dis 27(9):42

    Article  Google Scholar 

  10. Gül A, Inanç M, Ocal L, Aral O, Koniçe M (2000) Familial aggregation of Behçet’s disease in turkey. Ann Rheum Dis 59(8):622–625

    Article  PubMed  PubMed Central  Google Scholar 

  11. Remmers EF, Cosan F, Kirino Y, Ombrello MJ, Abaci N, Satorius C et al (2010) Genome-wide association study identifies variants in the MHC class I, IL10, and IL23R-IL12RB2 regions associated with Behçet’s disease. Nat Genet 42(8):698–702

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Saadoun D, Wechsler B, Terrada C, Hajage D, Le Thi Huong D, Resche-Rigon M et al (2010) Azathioprine in severe uveitis of Behçet’s disease. Arthritis Care Res (Hoboken). 62(12):1733–1738

    Article  CAS  PubMed  Google Scholar 

  13. Desbois AC, Wechsler B, Resche-Rigon M, Piette JC, Huong D Le T, Amoura Z et al (2012) Immunosuppressants reduce venous thrombosis relapse in Behçet’s disease. Arthritis Rheum 64(8):2753–2760

    Article  CAS  PubMed  Google Scholar 

  14. Kump LI, Moeller KL, Reed GF, Kurup SK, Nussenblatt RB, Levy-Clarke GA (2008) Behçet’s disease: comparing 3 decades of treatment response at the national eye institute. Can J Ophthalmol 43(4):468–472

    Article  PubMed  PubMed Central  Google Scholar 

  15. Alpsoy E, Zouboulis CC, Ehrlich GE (2007) Mucocutaneous lesions of Behçet’s disease. Yonsei Med J 48(4):573–585

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Alpsoy E, Donmez L, Bacanli A, Apaydin C, Butun B (2003) Review of the chronology of clinical manifestations in 60 patients with Behçet’s disease. Dermatology 207(4):354–356

    Article  PubMed  Google Scholar 

  17. Al-Araji A (2009) Neuro-Behçet’s disease: epidemiology, clinical characteristics, and management. Lancet Neurol 8(2):192

    Article  PubMed  Google Scholar 

  18. Arai Y, Kohno S, Takahashi Y, Miyajima Y, Tsutusi Y (2006) Autopsy case of neuro-Behçet’s disease with multifocal neutrophilic perivascular inflammation. Neuropathology 26(6):579–585

    Article  PubMed  Google Scholar 

  19. Hadfield MG, Aydin A, Lippman HR, Sanders KM (1997) Neuro-Behçet’s disease. Clin Neuropathol 16(2):55–60

    CAS  PubMed  Google Scholar 

  20. Noel N, Bernard R, Wechsler B, Resche-Rigon M, Depaz R, Le Thi Huong Boutin D et al (2014) Long-term outcome of neuro-Behçet’s disease. Arthritis Rheum 66(5):1306–1314

    Article  Google Scholar 

  21. Kidd D (2012) Neurological complications of Behçet’s syndrome. Curr Neurol Neurosc Rep 12(6):675–679

    Article  CAS  Google Scholar 

  22. Siva A, Kantarci OH, Saip S, Altintas A, Hamuryudan V, Islak C et al (2001) Behçet’s disease: diagnostic and prognostic aspects of neurological involvement. J Neurol 248(2):95–103

    Article  CAS  PubMed  Google Scholar 

  23. Kikuchi H, Aramaki K, Hirohata S (2008) Effect of infliximab in progressive neuro-Behçet’s syndrome. J Neurol Sci 272(1):99–105

    Article  CAS  PubMed  Google Scholar 

  24. Saadoun D, Wechsler B, Resche-Rigon M, Trad S, Le Thi Huong D, Sbai A et al (2009) Cerebral venous thrombosis in Behçet’s disease. Arthritis Rheum 61(4):518–526

    Article  CAS  PubMed  Google Scholar 

  25. Akman-Demir G (1999) Clinical patterns of neurological involvement in Behçet’s disease: evaluation of 200 patients. Brain 122(11):2171–2182

    Article  PubMed  Google Scholar 

  26. Kizilkilic O, Albayram S, Adaletli I, Ak H, Islak C, Kocer N (2003) Endovascular treatment of Behçet’s disease-associated intracranial aneurysms: report of two cases and review of the literature. Neuroradiology 45(5):328–334

    Article  PubMed  Google Scholar 

  27. Öktem-Tanör Ö, Baykan-Kurt B, Gürvit IH, Akman-Demir G, Serdaroğlu P (1999) Neuropsychological follow-up of 12 patients with neuro-Behçet’s disease. J Neurol 246(2):113–119

    Article  PubMed  Google Scholar 

  28. Saadoun D, Asli B, Wechsler B, Houman H, Geri G, Desseaux K et al (2012) Long-term outcome of arterial lesions in Behçet’s disease: a series of 101 patients. Medicine (Baltimore) 91(1):18–24

    Article  Google Scholar 

  29. Desbois A, Wechsler B, Cluzel P, Helft G, Boutin D, Piette JC et al (2014) Cardiovascular involvement in Behçet’s disease. Rev Med Int 35(2):103–111

    Article  Google Scholar 

  30. Tursen U, Gurler A, Boyvat A (2003) Evaluation of clinical findings according to sex in 2313 turkish patients with Behçet’s disease. Int J Dermatol 42(5):346–351

    Article  PubMed  Google Scholar 

  31. Kural-Seyahi E, Fresko I, Seyahi N, Ozyazgan Y, Mat C, Hamuryudan V et al (2003) The long-term mortality and morbidity of Behçet syndrome: a 2-decade outcome survey of 387 patients followed at a dedicated center. Medicine 82(1):60–76

    Article  PubMed  Google Scholar 

  32. Desbois A, Rautou P, Biard L, Belmatoug N, Wechsler B, Resche-Rigon M et al (2014) Behçet’s disease in Budd-Chiari syndrome. Orphanet J Rare Dis 9(1):104

    Article  PubMed  PubMed Central  Google Scholar 

  33. Hamuryudan V, Yurdakul S, Moral F et al (1994) Pulmonary arterial aneurysms in behçet’s syndrome: a report of 24 cases. Br J Rheumatol 33(1):48–51

    Article  CAS  PubMed  Google Scholar 

  34. Geri G, Terrier B, Rosenzwajg M, Wechsler B, Touzot M, Seilhean D et al (2011) Critical role of IL-21 in modulating Th17 and regulatory T cells in Behçet’s disease. J Allergy Clin Immunol 128(3):655

    Article  CAS  PubMed  Google Scholar 

  35. Atzeni F, Sarzi-Puttini P, Doria A, Boiardi L, Pipitone N, Salvarani C (2005) Behçet’s disease and cardiovascular involvement. Lupus 14(9):723–726

    Article  CAS  PubMed  Google Scholar 

  36. Ait Badi MA, Zyani M, Kaddouri S, Niamane R, Hda A, Algayres J (2008) Les manifestations articulaires de la maladie de Behçet. À propos de 79 cas. Rev Med Int 29(4):277–282

    Article  CAS  Google Scholar 

  37. Kaklamani VG, Vaiopoulos G, Kaklamanis PG (1998) Behçet’s disease. Sem Arthritis Rheum 27(4):197–217

    Article  CAS  Google Scholar 

  38. Kastner DL. Intermittent and periodic arthritic syndromes. In: Koopman WJ (ed.) Arthritis and allied conditions: a textbook of rheumatology. Vol. 1. 13th ed. Baltimore: Williams and Wilkins; 1997:1279–1306

  39. Beales IL (1998) Gastrointestinal involvement in Behçet’s syndrome. Am J Gastroenterol 93(12):2633

    Article  CAS  PubMed  Google Scholar 

  40. Marshall SE (2004) Behçet’s disease. Best Pract Res Clin Rheumatol 18(3):291–311

    Article  CAS  PubMed  Google Scholar 

  41. Lee KS, Kim SJ, Lee BC, Yoon DS, Lee WJ, Chi HS (1997) Surgical treatment of intestinal Behçet’s disease. Yonsei Med J 38(6):455–460

    Article  CAS  PubMed  Google Scholar 

  42. Jennette JC, Falk RJ, Bacon PA, Basu N, Cid MC, Ferrario F et al (2013) 2012 revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheum 65:1–11

    Article  CAS  PubMed  Google Scholar 

  43. Link J, Söderström M, Olsson T, Höjeberg B, Ljungdahl A, Link H (1994) Increased transforming growth factor-beta, interleukin-4, and interferon-gamma in multiple sclerosis. Ann Neurology 36(3):379–386

    Article  CAS  Google Scholar 

  44. Cho SB, Cho S, Bang D (2012) New insights in the clinical understanding of Behçet’s disease. Yonsei Med J 53(1):35–42

    Article  CAS  PubMed  Google Scholar 

  45. International study group for Behçet’s disease (1990) Criteria for diagnosis of Behçet’s disease. Lancet 335(8697):1078–1080

    Google Scholar 

  46. Türsen Ü (2009) Activation markers in Behçet disease. Turkderm 43:74–86

    Google Scholar 

  47. Kawai T, Akira S (2010) The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol 11:373–384

    Article  CAS  PubMed  Google Scholar 

  48. de Menthon M, Lavalley MP, Maldini C, Guillevin L, Mahr A (2009) HLA-B51/B5 and the risk of Behçet’s disease: a systematic review and meta-analysis of case-control genetic association studies. Arthritis Rheum 61(10):1287–1296

    Article  PubMed  Google Scholar 

  49. Ohno S, Ohguchi M, Hirose S, Matsuda H, Wakisaka A, Aizawa M (1982) Close association of HLA-Bw51 with Behçet’s disease. Arch Ophthalmol 100(9):1455–1458

    Article  CAS  PubMed  Google Scholar 

  50. Shahneh FZ, Hamzavi F, Bayazi B, Bandehagh A, Baradaran B (2013) New insights into HLA class I association to Behçet’s syndrome in Iranian Azari patients. Auto Immun Highlights 4(3):101–102

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Gül A (2014) Genetics of Behçetʼs disease: lessons learned from genomewide association studies. Curr Opin Rheumatol 26:56–63

    Article  PubMed  Google Scholar 

  52. Takeuchi M, Kastner DL, Remmers EF (2015) The immunogenetics of Behçet’s disease: a comprehensive review. J Autoimmun 64:137–148

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ombrello MJ, Kirino Y, de Paul Bakker IW, Gül A, Kastner DL, Remmers EF (2014) Behçet disease-associated MHC class I residues implicate antigen binding and regulation of cell-mediated cytotoxicity. Proc Natl Acad Sci USA 111:8867–8872

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kobayashi M, Ito M, Nakagawa A, Matsushita M, Nishikimi N, Sakurai T, Nimura Y (2000) Neutrophil and endothelial cell activation in the vasa vasorum in vasculo-Behçet disease. Histopathology 36(4):362–371

    Article  CAS  PubMed  Google Scholar 

  55. Lehner T (1997) The role of heat shock protein, microbial and autoimmune agents in the aetiology of Behçet’s disease. Int Rev Immunol 14(1):21–32

    Article  CAS  PubMed  Google Scholar 

  56. Consolandi C, Turroni S, Emmi G, Severgnini M, Fiori J, Peano C et al (2015) Behçet’s syndrome patients exhibit specific microbiome signature. Autoimmun Rev 14(4):269–276

    Article  PubMed  Google Scholar 

  57. Hirohata S, Oka H, Mizushima Y (1992) Streptococcal-related antigens stimulate production of IL6 and interferon-γ by T cells from patients with Behçet’s disease. Cell Immunol 140(2):410–419

    Article  CAS  PubMed  Google Scholar 

  58. The Matzinger P, Model Danger (2002) A Renewed Sense of Self. Science. 296:301–305

    Google Scholar 

  59. Medzhitov R, Janeway CA (2002) Decoding the patterns of self and non self by the innate immune system. Science 296:298–300

    Article  CAS  PubMed  Google Scholar 

  60. Direskeneli H (2006) Autoimmunity vs autoinflammation in Behcet’s disease: do we oversimplify a complex disorder? Rheumatology (Oxford) 45:1461–1465

    Article  CAS  Google Scholar 

  61. Cohen P (2014) The TLR and IL-1 signalling network at a glance. J Cell Sci 127:2383–2390

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Hirohata S (2008) Histopathology of central nervous system lesions in Behçet’s disease. J Neurol Sci 267(1):41–47

    Article  CAS  PubMed  Google Scholar 

  63. Kidd G, Kivisäkk P, Ransohoff RM (2003) Three or more routes for leukocyte migration into the central nervous system. Nat Rev Immunol 3(7):569–581

    Article  PubMed  Google Scholar 

  64. Ozaki K, Spolski R, Ettinger R, Kim HP, Wang G, Qi CF et al (2004) Regulation of B cell differentiation and plasma cell generation by IL-21, a novel inducer of blimp-1 and bcl-6. J Immunol 173(9):5361

    Article  CAS  PubMed  Google Scholar 

  65. Tzartos JS, Friese MA, Craner MJ, Palace J, Newcombe J, Esiri MM, Fugger L (2008) Interleukin-17 production in central nervous system-infiltrating T cells and glial cells is associated with active disease in multiple sclerosis. Am J Pathol 172(1):146–155

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Anderson MS, Bluestone JA (2005) The NOD mouse: a model of immune dysregulation. Annu Rev Immunol 23(1):447–485

    Article  CAS  PubMed  Google Scholar 

  67. Bucher C, Koch L, Vogtenhuber C, Goren E, Munger M, Panoskaltsis-Mortari A et al (2009) IL-21 blockade reduces graft-versus-host-disease mortality by supporting inducible T regulatory cell generation. Blood 114(26):5375–5384

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. O’Garra A, Rouleau M, Antonenko S, Rouleau M, Antonenko S, de Vries JE, Roncarolo MG (1997) A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature 389(6652):737–742

    Article  PubMed  Google Scholar 

  69. Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, Yao Z, Blank RB et al (2007) Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity 26(3):371–381

    Article  CAS  PubMed  Google Scholar 

  70. Weaver CT, Hatton RD, Mangan PR, Harrington LE (2007) IL-17 family cytokines and the expanding diversity of effector T cell lineages. Annu Rev Immunol 25(1):821–852

    Article  CAS  PubMed  Google Scholar 

  71. Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D, Watowich SS, Dong C (2007) STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. J Biol Chem 282(13):9358–9363

    Article  CAS  PubMed  Google Scholar 

  72. Lee JW, Wang P, Kattah MG, Youssef S, Steinman L, DeFea K, Straus DS (2008) Differential regulation of chemokines by IL-17 in colonic epithelial cells. J Immunol 181(9):6536

    Article  CAS  PubMed  Google Scholar 

  73. Eastaff-Leung N, Mabarrack N, Barbour A, Cummins A, Barry S. Foxp3+ regulatory T cells, Th17 effector cells, and cytokine environment in inflammatory bowel disease. J Clin Immunol. 2010; 2009;30(1):80–9

  74. de Pineton Chambrun M, Wechsler B, Geri G, Cacoub P, Saadoun D (2012) New insights into the pathogenesis of Behçet’s disease. Autoimmun Rev 11(10):687–698

    Article  Google Scholar 

  75. Deng J, Younge BR, Olshen RA, Goronzy JJ, Weyand CM (2010) Th17 and Th1 T-cell responses in giant cell arteritis. Circulation 121(7):906–915

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Sakane T (1997) New perspective on Behçet’s disease. Int Rev Immunol 14(1):89–96

    Article  CAS  PubMed  Google Scholar 

  77. Kebir H, Kreymborg K, Ifergan I, Dodelet-Devillers A, Cayrol R, Bernard M et al (2007) Human Th17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation. Nat Med 13(10):1173–1175

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Saadoun D, Rosenzwajg M, Joly F, Six A, Carrat F, Thibault V et al (2011) Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med 365:2067–2077

    Article  CAS  PubMed  Google Scholar 

  79. Nurieva R, Yang XO, Martinez G, Zhang Y, Panopoulos AD, Ma L, Schluns K et al (2007) Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature 448(7152):480–483

    Article  CAS  PubMed  Google Scholar 

  80. Vollmer TL, Liu R, Price M, Rhodes S, La Cava A, Shi F (2005) Differential effects of IL-21 during initiation and progression of autoimmunity against neuroantigen. J Immunol 174(5):2696–2701

    Article  CAS  PubMed  Google Scholar 

  81. Brustle A, Heink S, Huber M, Rosenplänter C, Stadelmann C, Yu P et al (2007) The development of inflammatory Th-17 cells requires interferon-regulatory factor 4. Nat Immunol 8(9):958–966

    Article  PubMed  Google Scholar 

  82. Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A et al (2009) Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 30(6):899–911

    Article  CAS  PubMed  Google Scholar 

  83. Bottinelli D, Uccelli A, Engelhardt B, Benvenuto F, Bottinelli D, Lira S et al (2009) C-C chemokine receptor 6-regulated entry of T H-17 cells into the CNS through the choroid plexus is required for the initiation of EAE. Nat Immunol 10(5):514–523

    Article  PubMed  Google Scholar 

  84. Engelhardt B, Wolburg-Buchholz K, Wolburg H (2001) Involvement of the choroid plexus in central nervous system inflammation. Micros Res Tech 52(1):112–129

    Article  CAS  Google Scholar 

  85. Chen Q, Yang W, Gupta S, Biswas P, Smith P, Bhagat G, Pernis AB (2008) IRF-4-binding protein inhibits interleukin-17 and interleukin-21 production by controlling the activity of IRF-4 transcription factor. Immunity 29(6):899–911

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Fantini MC, Rizzo A, Fina D, Caruso R, Becker C, Neurath MF et al (2007) IL-21 regulates experimental colitis by modulating the balance between Treg and Th17 cells. Eur J Immunol 37(11):3155–3163

    Article  CAS  PubMed  Google Scholar 

  87. Matusevicius D, Kivisäkk P, He B, Kostulas N, Ozenci V, Fredrikson S, Link H (1999) Interleukin-17 mRNA expression in blood and CSF mononuclear cells is augmented in multiple sclerosis. Mult Scler 5(2):101–104

    Article  CAS  PubMed  Google Scholar 

  88. Dagur PK, Biancotto A, Stansky E, Sen HN, Nussenblatt RB, McCoy JP (2014) Secretion of interleukin-17 by CD8+ T cells expressing CD146 (MCAM). Clin Immunol 152:36–47

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ et al (2006) The orphan nuclear receptor RORgammat directs the differentiation program of pro-inflammatory IL-17+ T helper cells. Cell 126(6):1121

    Article  CAS  PubMed  Google Scholar 

  90. Seder RA, Paul WE, Davis MM, de Fazekas St Groth B (1992) The presence of interleukin 4 during in vitro priming determines the lymphokine-producing potential of CD4+ T cells from T cell receptor transgenic mice. J Exp Med 176(4):1091–1098

    Article  CAS  PubMed  Google Scholar 

  91. Wu X, Tan Y, Xing Q, Wang S (2013) IL-21 accelerates xenogeneic graft-versus-host disease correlated with increased B-cell proliferation. Protein Cell 4(11):863–871

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Habib T, Senadheera S, Weinberg K, Kaushansky K (2002) The common gamma chain (gamma c) is a required signaling component of the IL-21 receptor and supports IL-21-induced cell proliferation via JAK3. Biochemistry 41(27):8725

    Article  CAS  PubMed  Google Scholar 

  93. Touzot M, Cacoub P, Bodaghi B, Soumelis V, Saadoun D (2015) IFN-α induces IL-10 production and tilt the balance between Th1 and Th17 in Behçet disease. Autoimmun Rev 14(5):370–375

    Article  CAS  PubMed  Google Scholar 

  94. Comarmond C, Wechsler B, Bodaghi B, Cacoub P, Saadoun D (2014) Biotherapies in Behçet’s disease. Autoimmun Rev 13(7):762–769

    Article  PubMed  Google Scholar 

  95. Takamoto M, Kaburaki T, Numaga J, Fujino Y, Kawashima H (2007) Long-term infliximab treatment for behçet’s disease. Jpn J Ophthalmol 51(3):239–240

    Article  PubMed  Google Scholar 

  96. Sbaï A, Wechsler B, Duhaut P, Du-Boutin LT, Amoura Z, Cacoub P et al (2003) Neuro-Behçet’s disease (isolated cerebral thrombophlebitis excluded). Clinical pattern, prognostic factors, treatment and long term follow-up. Adv Exp Med Biol 528:371–376

    Article  PubMed  Google Scholar 

  97. Wechsler B, Lê Thi Huong DB, Saadoun D (2009) EULAR recommendations for the management of Behçet’s disease: evidence-based or experience-based medicine. Rev Med Interne 30(11):939–941

    Article  CAS  PubMed  Google Scholar 

  98. Hatemi G, Melikoglu M, Tunc R et al (2015) Apremilast for Behçet’s syndrome–a phase 2, placebo-controlled study. N Engl J Med 372:1510

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

The authors wish to thank the precious cooperation of the i2 lab—Integrative Immunology at Université Pierre et Marie Curie—The Sorbonne Universities, as well as the Department of Internal Medicine and Clinical Immunology at the La Pitié-Salpêtrière Hospital.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Patrice Cacoub.

Ethics declarations

Funding

None.

Conflict of interest

All authors declare that they have no conflict of interest.

Ethical approval

This article does not contain any studies with human participants or animals performed by any of the authors.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 394 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0), which permits use, duplication, adaptation, distribution, and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zeidan, M.J., Saadoun, D., Garrido, M. et al. Behçet’s disease physiopathology: a contemporary review. Autoimmun Highlights 7, 4 (2016). https://doi.org/10.1007/s13317-016-0074-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s13317-016-0074-1

Keywords