Abstract
Tumor necrosis factor alpha (TNF-α) and interleukin-17 (IL-17) are two pro-inflammatory cytokines involved in the pathophysiology of spondyloarthritis (SpA). Therapies targeting TNF-α or IL-17 are used as a second line among SpA patients failing non-steroidal anti-inflammatory drugs. The choice of such treatment has to take into account the patient’s comorbidities. Neurologic diseases are common and their association with SpA deserves to be studied. Therefore, the role of TNF-α and IL-17 cytokines is worth investigating in these neuropsychiatric diseases. This review aimed to explore the role of TNF-α and IL-17 in the pathogenesis of uveitis, multiple sclerosis, neuromyelitis optica, Alzheimer’s disease, Parkinson’s disease and depression. This update is critical to guide the therapeutic management of these co-morbidities in SpA patients.
Keywords: Alzheimer’s disease, central nervous system, cytokines, depression, IL-17, multiple sclerosis, neuromyelitis optica, Parkinson’s disease, spondyloarthritis, tumor necrosis factor alpha, uveitis
Introduction
Spondyloarthritis (SpA) is a systemic inflammatory disorder affecting the axial skeleton, peripheral joints and entheses. Extra-articular manifestation such as inflammatory bowel disease, psoriasis and uveitis are often associated. SpA affects 0.1–1% of the general population. Comorbidities are frequent and the most common are represented by osteoporosis, cardiovascular disease, cancer and infections. 1 However, other comorbidities deserve to be investigated, such as neurodegenerative and neuropsychiatric diseases. Indeed, SpA is often diagnosed in young adults, but as life expectancy increases, we are faced with aging patients who may also have neurodegenerative diseases such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). Moreover, recent evidence indicates that SpA patients face an increased risk of depression (odds ratio 1.8) compared with the general population. 2 The authors also reported a prevalence of depression in nearly 11% of SpA patients and that up to 1.5% of all SpA patients suffered from dementia, PD, or multiple sclerosis (MS). Psychological distress symptoms contribute to impaired quality of life both directly and indirectly by influencing disease activity.3–5 These observations led us to take a general interest in pathologies of the central nervous system (CNS), including eye/optic nerve, and more particularly their coexistence with SpA. We therefore decided to focus on certain neurological pathologies due to their frequency in the general population (AD, PD, depression) and/or their potential severity (multiple sclerosis, neuromyelitis optica and uveitis).
Tumor necrosis factor alpha (TNF-α) and interleukin-17 (IL-17) are two pro-inflammatory cytokines involved in the pathophysiology of SpA. Therefore, it is worth investigating their role in neuropsychiatric diseases. It was hypothesized that TNF-α produced by macrophages plays an important role in neurodevelopment and in the pathophysiology of various neuropsychiatric conditions.6,7 Actually, TNF-α may activate the hypothalamo–pituitary–adrenocortical axis and serotonergic neuronal transporters, resulting in neuronal destruction or neurotoxic release of glutamate. 8 However, the role of IL-17 remains unclear in these neurologic disorders. In order to optimize the management of SpA patients, it seems necessary to study the role of pro-inflammatory cytokines in the CNS and the impact of treatments used in SpA on these neurological diseases.
In this review, we aimed to explore the role of TNF-α and IL-17 cytokines in the pathogenesis of uveitis, MS, AD, PD and depression and the effect of treatment targeting TNF-α and IL-17 on these neuropsychiatric diseases (Table 1). First, we will focus on the epidemiology of each neurological comorbidity in SpA, then in the pathophysiology mechanisms and targeted treatments. We conducted a selected review in PubMed database prior to December 2020 using the following MeSH terms: TNF-α, IL-17, uveitis, Alzheimer’s disease, Parkinson’s disease, depression, multiple sclerosis, neuromyelitis optica.
Table 1.
Comparative efficacy of TNF-α and IL-17 targeted treatments in neuropsychiatric diseases.
Anti-IL-17 | Anti-TNF-α | |
---|---|---|
Uveitis | ± | + |
Multiple sclerosis | + | − |
Neuromyelitis optica | Undefined | − |
Alzheimer’s disease | Undefined | ± |
Parkinson’s disease | Undefined | Undefined |
Depression | − | − |
, positive action; −, negative action; ±, inconclusive action.
Uveitis
Epidemiology of uveitis in SpA
The estimated prevalence of uveitis in all its types – anterior, intermediate, posterior, or global (panuveitis) – is about 1 per 1000 individuals. The incidence of uveitis is reported between 19 and 88 per 100,000 patients/year. 9 Its impact can be major since its occurrence is complicated by visual morbidity. 10 SpA is the most common systemic disease associated with uveitis. Studies have concluded that approximately 80% of patients with a history of acute anterior uveitis associated with HLA-B27 gene suffered from SpA family disorders.11,12 It is worth mentioning that this frequency is about three times lower for HLA-B27 negative patients. 13 Conversely, SpA without HLA-B27 genetic background, particularly in women, has a poorer visual prognosis, probably due to diagnostic delay. 14 Uveitis occurs in 30–50% of ankylosing SpA and predominates in males. Among 902 patients with uveitis recruited by rheumatologists, nearly 50% were recurrent. 15 They reported two independent factors associated with uveitis: HLA-B27 gene and disease duration greater than 10 years. Furthermore, it should be noted that occurrence of uveitis is not related to the activity or severity of the underlying SpA. 16
The role of the IL-17/TNF-α pathway in the pathophysiology of uveitis
Uveitis is an intraocular inflammatory process, mainly mediated by T-cells. 17 T-cell activation increases the production of pro-inflammatory cytokines such as IL-17 and/or TNF-α. In humans, the presence of IL-17 and TNF-α is increased in the aqueous humor of patients with SpA. 18 Low levels of TNF-α and high levels of IL-17 are found in the serum of patients with uveitis. IL-17 expression was particularly correlated with disease activity. 19 Similarly, the IL-17 serum levels in contrast to TNF-α expression can differentiate non-infectious uveitis from toxoplasmosis or post-viral uveitis. 20 A recent study shows a clear increase in Th17-cells and a significant decrease in regulatory T-cells (Treg-cells) in patients with HLA-B27 positive uveitis. 21
Animal models of experimental autoimmune uveitis (EAU) provide a better understanding of the pathophysiological mechanisms of uveitis, despite some limitations. Depending on the model of EAU used (genetically modified or requiring the administration of antigenic peptide), Th17 or Th1 immune pathways can be differentially modulated. 22 Some authors have shown an overexpression of TNF-α in EAU. 23 Others reported a predominance of Th17-cells, whose number was correlated with the severity of uveitis. 24 In this outstanding study, it was suggested that Th17-cells may also contribute to uveitis pathogenesis via TNF-α induction. 24 In addition, animal studies have demonstrated the efficacy of antibodies blocking IL-17 or TNF-α to reduce the intensity of uveitis, highlighting their therapeutic value in the disease. 25 However, the strategy of targeting IL-17 may be harmful since it has resulted in retinal photoreceptor toxicity 25 and a “paradoxical” anti-inflammatory effect of the administration of IL-17 in the EAU model was observed in another study. 26
Uveitis occurrence under anti-IL-17/anti-TNF-α therapy in SpA patients
SpA patients frequently have recurrent episodes of uveitis. 15 Most of these episodes resolve within 3 months and are usually treated with topical corticosteroids. Monoclonal antibodies targeting TNF-α, in particular adalimumab (ADA) or infliximab, prevent new relapses of uveitis, although some may occur while receiving these treatments. 27 In contrast, the soluble TNF-α trapping receptor (etanercept) is more frequently associated with the emergence of new uveitis than monoclonal antibodies. 28 The incidence rate of uveitis, adjusted for exposure to TNF-α inhibitors (TNFis), in patients with SpA ranges from 2.6 to 3.5 per 100 patients/year.29–31 A recent analysis of patients included in the Phase III clinical trials of secukinumab (SCK; anti-IL-17 antibodies) revealed a lower incidence of 1.4 per 100 patients/year. 32 However, these studies are not similar, and we cannot conclude that uveitis occurs less frequently with anti-IL-17 therapy versus anti-TNF-α therapy.
Efficacy of targeting IL-17 or TNF-α on uveitis
The availability of different treatment options in the management of uveitis will depend on the severity and its impact on quality of life. In the event of topical steroid and/or local corticosteroid infiltration failures (ineffectiveness or corticosteroid dependence), treatment with monoclonal antibodies targeting TNF-α may be appropriate. Two randomized, double-blind, placebo-controlled trials using ADA versus placebo have been conducted in the treatment of non-infectious uveitis (intermediate, posterior, or panuveitis). The VISUAL I study included patients with active uveitis despite 2 weeks of oral corticosteroids. 33 Relapse occurred less frequently in the ADA arm (40 mg every 2 weeks after a loading dose of 80 mg). The VISUAL II study with the same methodology was conducted in patients with a controlled uveitis under oral corticosteroid therapy between 10 and 35 mg per day. 34 In this study, relapses were significantly less frequent in the ADA group. Therefore, ADA is recommended for patients who have an inadequate response to prior oral corticosteroid therapy, as well as for patients requiring steroid sparing despite immunosuppressive drugs. For treatments targeting IL-17 or IL-23, only SCK has been evaluated in clinical trials. Some authors have reported a successful experience with ustekinumab (blocking the p40 subunit of the IL-12/IL-23 axis) in some patients.35,36 Currently, several Phase II trials are in progress with this molecule (NCT02648581, NCT03847272, NCT02911116). For SCK, a first study was conducted on individuals with anterior (n = 4) or posterior (n = 12) uveitis with encouraging results. 37 Subsequently, three randomized controlled trials (SHIELD, INSURE, ENDURE) evaluating subcutaneous SCK (300 mg every 2 weeks) against placebo failed to demonstrate any advantage regardless of the patient’s phenotype (refractory Behcet’s uveitis, active non-Behcet’s uveitis despite corticosteroids or requiring immunosuppression). 38 More recently, a new study with a small sample of 30 patients suggested that intravenous SCK (10 mg/kg per 2 weeks or 30 mg/kg per 4 weeks) may be better than the subcutaneous formulation. 39 Thus, it is possible that subcutaneous bioavailability may be partly the cause of SCK treatment failure in the randomized studies.
A specific well-conducted study addressing the anterior uveitis typically found in SpA is missing. Several different causes of uveitis are generally grouped together in clinical trials. The heterogeneity in diseases inducing uveitis and anatomical localizations hinders the transfer of results for SpA. Finally, it is worth remembering that MS can rarely be associated with intermediate uveitis. 40 As TNFis may promote demyelinating lesions 41 another therapeutic target will have to be considered.
MS
Epidemiology of MS in SpA
MS is a chronic demyelinating disorder of the CNS. It is an heterogeneous disease, which may be categorized into clinically isolated syndrome, relapsing and/or remitting MS (RRMS), secondary progressive MS and primary progressive MS. The prevalence of MS in SpA patients can be estimated AS from 0.7% to 1.75%. 42 In most cases reported, SpA preceded the first signs of MS. The interval between the two conditions ranged from 3 to 21 years. 43 One study described four cases of patients who did not previously receive TNFi at the time of MS diagnosis. 44 Most studies conclude that the association of SpA and MS seemed to be a coincidence.42,45
The role of the IL-17/TNF-α pathway in the pathophysiology of MS
MS is a T-cell mediated disease of the CNS (mainly CD8+ cells). In the initial process of RRMS, activated auto aggressive T-cells cross the blood–brain barrier. The activation of T-cells induces recruitment of other inflammatory cells, including macrophages, by secretion of numerous cytokines. So there is an accumulation of macrophages and lymphocytes in the CNS, resulting in demyelination and destruction of axons. 46
IL-17A is an important player in several autoimmune and inflammatory diseases such as MS and SpA. Several cell types have been described as potential producers of IL-17A, such as astrocytes and oligodendrocytes. Many studies have described the role of IL-17 in MS and all have concluded that there is a clear link between IL-17 expression and disease development.46–49
In vitro studies and preclinical animal models have shown that IL-17A plays a major role in MS. Numerous studies using experimental models of MS in mice have found a clear association of Th17 and IL-17 with disease severity and progression.47,48 In humans, IL-17A-producing memory T lymphocytes have been found in brain lesions of MS patients but not in normal samples. 48 In addition, elevated mononuclear cells expressing IL-17A were reported in the cerebrospinal fluid and blood of patients with MS. Two additional studies found increased serum TNF-α and IL-17 levels in MS patients compared with healthy subjects.50,51
TNF-α is secreted by innate immune cells such as macrophages, monocytes and differentiated T-cells. In MS, TNF-α levels were found to be increased in active MS lesions and in an animal model of MS. However, it was determined that TNFα plays a dual role in MS. TNF-α and its receptors can either promote neuroinflammation and secondary neuronal damage or exert protective functions under pathological conditions.46,52
Efficacy of targeting IL-17 or TNF-α on MS
TNF-α has two biologically active forms (soluble and transmembrane), as well as two different receptors. TNF-α-receptor 1 (TNFR1) has pro-inflammatory and cytotoxic activity, and the other, TNF-α-receptor 2 (TNFR2), has a rather cytoprotective action. Thus, activation of TNFR2 could improve the differentiation of oligodendrocytes and promote remyelination by inhibiting the pro-inflammatory activity of microglia.52–55
This can explain why TNFis, due to their non-selectivity, could increase disease activity in MS or induce CNS demyelination in SpA patients treated with TNFis. Moreover, it is believed that in MS an imbalance in favor of pro-inflammatory TNFR1-mediated signaling pathways outweighs beneficial TNFR2-mediated effects. Despite a beneficial effect on monoclonal antibody targeting TNFR1 in experimental autoimmune encephalomyelitis, 56 a TNFR1-Fc, lenercept, induced an increased number of magnetic resonance imaging (MRI) lesions without clinical effect in MS patients. 57 Others theories have also been proposed, such as the impermeability of blood–brain barrier to TNFis, the dysregulation of TNF-α in patients with RRMS, the alteration of cytokine responses by downregulating interleukin-10 and upregulating interleukin-12 and interferon-γ or the fact that TNFis may unmask an underlying latent infection, which can lead to autoimmune demyelination 54 or aseptic meningitis reflecting CNS inflammation induced by infliximab. 58 Indeed, several case reports have indicated that demyelinating diseases could be a serious adverse event following TNFi treatment54,59 such as etanercept 60 or ADA. 55 Literature reviews of previously published cases42,55 identified 20 cases of MS onset upon TNFi therapies.
A randomized controlled study reported interesting results on MS disease activity by IL-17A inhibition. 61 Compared with placebo, SCK significantly reduced the number of cumulative new-gadolinium-enhanced T1 lesions by 67% but non-significantly reduced the number of combined unique active lesions, which was the primary endpoint. No additional adverse events were reported in the SCK arm compared with placebo. 61 Some cases reported the efficacy of SCK in patients with concomitant MS and SpA62,63 with stability of brain MRI lesions, absence of new relapse and remission of neurological symptoms. Considering these data, IL-17A blockade may become an alternative option for SpA patients with MS.
Focus on neuromyelitis optica
Neuromyelitis optica (NMO) is a severe and relapsing demyelinating disease of the CNS that preferentially affects the spinal cord (myelitis) and the optic nerves (optic neuritis). It was initially recognized as a subtype of MS but it is mainly characterized by the presence of specific serum autoantibodies directed against aquaporin 4. 64
Several studies indicated that Th17-cells and Th17-cell-cytokines could play an essential role in the pathophysiology of NMO by degrading the blood–brain barrier and promoting the release of autoantibodies. Also suggested is a collaboration between Th17-cells and B-cells inducing CNS damage. 65 A meta-analysis showed that the number of Th17-cells and serum IL-17 levels were higher in patients with NMO compared with healthy subjects. 66
According to some studies, TNF-α seems to be neuroprotective from inflammatory damage with practically undetectable TNF-α levels in NMO patients. 67 One case of NMO was reported with positive myelin oligodendrocyte glycoprotein antibodies following TNFi for a pustular psoriasis. 68 There were no data concerning IL-17 blockade in NMO.
AD
Epidemiology of AD in SpA
AD is the most common form of dementia in the elderly, resulting in a progressive decline in a number of cognitive functions. At histology level, AD is characterized by age-related aggregation of the amyloid β-protein affecting microglia, astrocytes and neurons. Indirect evidence suggests that initiation is due to strong local inflammatory responses. The prevalence of AD in SpA is unknown but a Korean study found that SpA patients showed a significantly higher prevalence of overall dementia and Alzheimer’s dementia. The prevalence was 1.37% for overall dementia and 0.99% for AD in the SpA group 69 while it was 0.87% and 0.63% respectively in the control group.
The role of the IL-17/TNF-α pathway in the pathophysiology of AD
Pro-inflammatory cytokines can be considered as important factors for induction/stimulation of AD and particularly the IL-23/IL-17A pathway. 70 First of all, high serum levels of IL-17A and IL-23 were observed in the AD Chinese patients compared with controls, with higher IL-23 serum level in the female AD patients. 71 Since a high level was observed in human, suppression of IL-23 signaling pathway (via inhibition of p40 subunit by local or systemic delivery) reduces AD-like pathology in animal models.72,73 Thus, it appears that suppression of Th17 development leads to reduced AD-like pathogenesis. Numbers and functions of IL-23-producing monocytes/macrophages were increased in AD patients. 74 IL-23 was able to induce senescence-accelerated mouse prone 8 strain, as an AD animal model. 75 In addition to IL-23, Th17-cells and IL-17A itself were involved in AD pathogenesis. In AD patients, IL-17A was elevated in the cerebrospinal fluid, 76 with an increased activity and differentiation of Th17-cells through RORγ expression. 77 So, the IL-23/17 pathway is involved in AD induction and the development of the first symptoms. 78 However, the late symptoms were not mediated by the IL-23/17 pathway as suggested by the decrease in IL-17 and IL-23 levels reported in established AD patients compared with age- and sex-matched healthy controls.79,80
It therefore appears that IL-17A plays an essential role in the initiation and maintenance of AD and its complications during ageing. However, once the disease is established, the influence of IL-17A declines in parallel with the reduction of immune responses.
Factors explaining high IL-17A expression in AD
In vitro, it was shown that amyloid-β protein was able to produce IL-17A after tissular hypoxia. 81 In an AD animal model, gram-negative respiratory infection induced T-cell infiltration and consequently amyloid-β deposition, leading to increased glial cell activation and neurodegenerative functions. 82 To explain this association with bacteria, research focused on innate immune response and in particular TLR4. IL-17A was upregulated in an AD transgenic mouse model in a TLR4-dependent manner. 83 TLR4 was involved in recognition of amyloid-β and induction of IL-23/17 pathway through NF-κB. 84 Two genetic factors also regulated IL-17A expression in AD. The human leukocyte antigen-DRB1*1501 alleles play crucial roles in induction of T helper cells (including Th17-cells) against amyloid-β protein in human models. 85 In Han Chinese population, the G allele of rs1884444 polymorphism within IL-23 receptor gene was associated with AD. 86
Mechanisms involved by IL-17A in AD
IL-17A is a strong stimulator for neutrophils in the blood.87,88 In AD animal models, increasing number, functions and recruitment of neutrophils to the inflamed CNS can be a strong mechanism explaining involvement of the IL-23/17 pathway in AD. Expression of FasL on the Th17 and interaction of the molecule with Fas was also reported to induce apoptosis in the neurons in an AD animal model. 89 So, Th17 mediates AD symptoms through induction of neuroinflammation, leading to recruitment and activation of immune cells, such as neutrophils and microglia, which results in damage to neurons.
TNF-α in AD
Both microglia and neurons expressed TNF-α at physiological levels, and it appears to be a key pro-inflammatory cytokine upregulated in AD patients with interaction with AD genes.90,91 Among these genes, a significant association has been found between a polymorphism of exon 6 of TNFR2 and late AD. 91 Furthermore, several TNF-α promoter polymorphisms associated with its increased expression were also associated with inflammatory and infectious diseases. A meta-analysis suggests that TNF-α polymorphism G308A could be associated with an increased risk of AD in China and a decreased risk of AD in Northern European populations. 92 TNF-α is elevated in AD and stimulates the synthesis of amyloid plaques in animal models of AD and in the human brain.93–95 The high level of TNF-α measured in serum appears to be predictive of the severity of the disease. 96
Efficacy of targeting IL-17 or TNF-α on AD
Many indirect effects of various approaches improved AD signs and could be due to IL-17. All-trans-retinoic acid downregulates IL-17A within peripheral blood mononuclear cells of AD patients and was associated with improvement of AD symptoms. 91 Downregulation of IL-17A by injection of transforming growth factor-β reduced neuron damage in an animal model of AD. 97 It was highlighted that flavonoid resveratrol downregulated IL-17A and was beneficial on AD symptoms. 98 A vaccine approach with Aβ 42 DNA improved AD symptoms through suppression of Th17-cell proliferation and IL-17A secretion. 77 However, no direct systemic evidence of targeting IL-23/17 axis in human is available, mainly due to lack of ability of these antibodies to penetrate the blood–brain barrier.
One review suggested a beneficial effect of TNFi on cognition and behavior based on evidence from animal studies of AD models. 99 There were only three studies in humans and two of them concluded that TNFis are beneficial in AD patients, but studies had a poor level of evidence. 99 Some other results from animal models with TNF receptor knockout suggest an adverse use of TNFi in AD, with worsening extracellular Aβ deposition. 100 Butchart et al. 101 found some interesting trends that favored etanercept, but there were no statistically significant changes in cognition, behavior or global function. Overall, TNFi seemed to have a poor efficacy because most antibodies show poor penetration of the blood–brain barrier via passive diffusion. Furthermore, it would necessary to obtain a mild to moderate inhibition level in order not to induce adverse events resulting from blocking the physiological roles of TNF-α in the CNS. 94
PD
Epidemiology of PD in SpA
PD is a neurodegenerative disease characterized by progressive loss of dopaminergic neurons and presence of neuroinflammation. Approximately 2–3% of elderly people are affected by PD. No treatments currently exist to prevent PD and dopaminergic substitution treatments just relieve the consequences of dopaminergic neuron loss. To date, no association between psoriatic arthritis or axial SpA and PD has been identified. However, patients with psoriasis may have on average 38% increased risk to develop PD, possibly as a result of chronic inflammation. 102
The role of the IL-17/TNF-α pathway in the pathophysiology of PD
PD is characterized by the presence of Lewy bodies that result from an abnormal accumulation of vesicles and aggregated peptides. Alpha-synuclein (α-syn), a pre-synaptic protein, has been recognized as its main constituent. 103 The presence of self-reactive T-cells directed against α-syn has recently been identified in Parkinson’s patients, suggesting a potential autoimmune origin of the disease. 104 Neuronal death results from T-cells’ infiltration into microglia and is promoted by the presence of cytokines and pro-inflammatory chemokines produced by neuronal aggregates. 105
IL-17 and PD
The involvement of Th17 in PD has been confirmed in two studies using mouse models of PD. In the group of mice treated with intraperitoneal injections of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, an increased proportion of Th17-cells in the substantia nigra and ventral midbrain was observed. Immunization of mice with nitrated α-syn partially resulted in Th17 polarization of CD4+ T-cells and impaired Treg-cell function.106,107 Another animal study based on transgenic human rats showed that the presence of the LRRK2 G2019S gene, one of the major mutations of PD-related gene, was not associated with changes in brain Th17-cells but, rather, with decreases in peripheral Th17-cells. 108
Limited data are available based on clinical studies, and results are heterogeneous according to the technique used. Indeed, patients with PD appear to have a similar level of Th17-cells compared with controls but may contain a higher proportion of cells ready to express IL-17 upon activation. 109 The proportion of IL17-producing cells that can induce an increased Th17/IL-17 activity is unknown. According to Kustrimovic et al., 110 the amount of IL-17 actually secreted by lymphocytes in PD patients is unchanged, whereas it is increased in very few subjects according to Sommer et al. 111 Moreover, Kustrimovic et al. 110 reported a reduction in the absolute number of Th17-cells per volume of blood in patients with PD, which means that increased presence of IL-17-producing cells is not necessarily correlated with increasing Th17/IL-17-dependent systemic pro-inflammatory effects. Thus, evidence of the involvement of Th17 in PD is limited and controversial.
TNF-α and PD
TNF-α expression may contribute to faster dopaminergic neuron degeneration and it was found that a higher genetic pro-inflammatory profile confers a higher risk to develop PD earlier. 112 Some studies have demonstrated abnormally elevated levels of TNF-α in PD patients 113 whereas others found significantly low levels. 114 Kouchaki et al. 115 showed that the serum levels of TNF-α and IL-27 may be important prognostic biomarkers of PD with correlation between the serum level of those factors and severity of PD. Moreover, it was shown that TNF-α levels measured in the striatum and cerebrospinal fluid were significantly higher in PD patients than those in controls. 116 One study aimed to determine TNF-α levels in tear samples obtained from patients with PD and to analyze the relationship between TNF-α values and PD characteristics. They found that tear TNF-α values were significantly higher in patients with PD than in control subjects but without relationship to the duration or severity of PD. 117
Efficacy of targeting IL-17 or TNF-α on PD
No case report has been published of Parkinson’s syndrome in patients treated with biotherapies (IL-17 or IL-23 inhibitors; TNF-α blockers) in the following indications: psoriasis, psoriatic arthritis and axial SpA. However, in inflammatory bowel disease, Peter et al. 118 showed that early exposure to TNFi was associated with substantially reduced PD incidence but further studies are required to determine whether TNFi administered to high-risk PD individuals may mitigate this risk.
Depression
Epidemiology of depression in SpA
Depression has frequently been reported to be associated with other physical diseases and changes in the cytokine system. In primary care, the average reported prevalence of depression was 12%. 119 In SpA, pooled prevalence of at least moderate depression was 15% using the Hospital Anxiety and Depression Scale threshold of ⩾11. 120 The prevalence of depression was similar between axial SpA, radiographic SpA and non-radiographic-axial SpA. Patients with depression had significantly higher disease activity. 120 Mood, sleep disorders and pain seem to contribute independently to the poor quality of life in patients with SpA.121–123
The role of the IL-17/TNF-α pathway in the pathophysiology of depression
IL-17 and depression
During depression, the role of IL-17 is unclear. In a mouse model, IL-17A has been shown to be involved in depression associated with psoriatic inflammation via the NF-κB and p38MAPK pathways, which play a significant role in upregulation of inflammatory mediators in the brain.124–126 In humans, the presence of higher baseline IL-17 levels appears to be associated with greater symptom reduction in depressed patients treated with association of selective serotonin reuptake inhibitors and bupropion. 127
TNF-α and depression
The association between TNF-α and depression is controversial. Several mechanisms were highlighted to explain how TNF-α may lead to depression or depressive symptoms : the activation of the hypothalamus-pituitary-adrenal axis, the activation of neuronal serotonin transporters and the stimulation of the indoleamine 2,3-dioxygenase, which leads to tryptophan depletion. 128 In a meta-analysis, peripheral TNF-α levels were not significantly increased in the elderly with depression as compared with controls. 129 Another meta-analysis revealed that TNF-α G-308A polymorphism is not associated with susceptibility to depression. 130 Conversely, Himmerich et al. 131 found that acutely depressed inpatients had higher levels of sTNF-R p55, sTNF-R p75 and TNF-α than healthy subjects. This is consistent with another meta-analysis which reported significantly higher concentrations of proinflammatory cytokines (TNF-α and IL-6) in depressed subjects compared with control subjects. 132
Efficacy of targeting IL-17 or TNF-α on depression
There is no evidence of elevated risk for depression, anxiety or suicide with SCK in a pooled analysis of data from 10 clinical studies in moderate-to-severe plaque psoriasis. 121 However, a case was described of depression exacerbation in a psoriatic arthritis patient possibly induced by SCK. 122 Suicidal ideation was mainly reported with another IL-17 blocker. 123
On the other hand, one study found that TNFi (infliximab) may be effective in the treatment of depression, anxiety and quality of life in patients with active SpA. 134 It also described some rare cases of suicide in patients treated with TNFi. In one case, a patient treated with adalimumab developed anti-TNF-α-induced lupus and suicidal ideations. 135 However, infliximab was able to reduce signs of depression in the case of systemic inflammation. 136
The potential missing link between cytokine-induced chronic inflammatory diseases such as SpA and depression could be sleep disorders. 137 Targeting TNF-α and IL-17 decreases pain in SpA; the positive effect of biological disease-modifying anti-rheumatic drugs (bDMARDs) on sleep architecture is barely discriminable from the efficacy of bDMARDs on disease activity. The role of cytokine imbalance on chronic pain and sleep disorders has been neglected in the field of rheumatology. Imeri and Opp 138 described the regulation of sleep by the immune system in the early 20th. TNF inhibition by etanercept improved sleep patterns as well as sleepiness in two randomized placebo-controlled, double-blind trials involving abstinent alcohol-dependent male adults and patients with obstructive sleep apnea.139,140 Data studying the impact of targeting IL-17 in sleep disorders in non-rheumatic population are not available.
Fibromyalgia
Fibromyalgia is a syndrome of unknown etiology characterized by widespread chronic pain very often associated with fatigue, anxiety, depression and sleep disorders. Its prevalence in SpA ranges from 12.5% to 15%141–143 and 2–7% in the general population. 144 In axial SpA patients, fibromyalgia reaches a prevalence of 25%. 145 Fibromyalgia is likely associated with a CNS sensitization impairment due to stress such as sleep deprivation and chronic pain rather than with cytokine imbalance. As chronic painful stimulations modulate gray matter,129,130 function, morphology of small nerve fibers 131 and structural plasticity of the brain, fibromyalgia might be considered as a common comorbidity of SpA. 132
IL-17 and fibromyalgia
Plasma levels of IL-17A were compared in plasma from 58 fibromyalgia patients and 39 healthy women matched for age and body mass index using the technique of cytometric bead array. 146 IL-17 levels were significantly higher in fibromyalgia patients. The origin of IL-17 amongst IL-17-producing cells was not addressed by this study. A positive but mild correlation between IL-17 and TNF-α plasma concentration (r = 0.396, p = 0.003) was found. Otherwise, non-pharmacological intervention such as progressive resistance exercise or relaxation therapy, known to improve fibromyalgia outcome measures,147,148 do not impact IL-17 levels. 149
Conclusion
At the time of personalized medicine, the prescriber should consider patients’ comorbidities. According to neuropsychiatric diseases observed in SpA patients, we reviewed arguments to choose the best treatment. In SpA with MS, the best strategy would be to block the IL-17 pathway. In SpA with severe uveitis, the best strategy would be a monoclonal antibody targeting TNF-α. Even if in some neurological diseases, the exact role of cytokines and the effect of their blockage remains uncertain, it is important to keep in mind that the use of a treatment targeting TNF-α or IL-17 in SpA is likely to interfere with diseases of the CNS (Table 1).
Footnotes
Conflict of interest statement: E. How Shing Koy has received honoraria from Novartis; P. Labauge has no disclosures; A. Baillet has received consultancy fees, and/or honoraria from Abbvie, Novartis, Medac, Pfizer, Galapagos, UCB; C. Prati has received consultancy fees, and/or honoraria, and/or grants from Novartis, Lilly, Abbvie, UCB, Fresenius; H. Marotte has received consultancy fees, and/or honoraria from Abbvie, Biogen, BMS, Lilly, MSD, Nordic Pharma, Novartis, Pfizer, Sanofi, UCB; Y-M. Pers has received consultancy fees, and/or honoraria from Novartis, Pfizer.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This review has been written with logistical and financial support from Novartis.
ORCID iD: Yves-Marie Pers
https://orcid.org/0000-0001-5927-3773
Contributor Information
Elsa How Shing Koy, Department of Rheumatology, Hôpital Nord, CHU Saint-Etienne, Saint-Etienne, France; Department Internal Medicine, Le Corbusier Hospital, Firminy, France.
Pierre Labauge, MS Unit, Department of Neurology, Montpellier University Hospital, Montpellier Cedex 5, France.
Athan Baillet, Université Grenoble-Alpes, GREPI TIMC, UMR 5525, Grenoble France.
Clément Prati, Department of Rheumatology, CHRU de BESANCON, University Teaching Hospital, Besançon, France.
Hubert Marotte, Department of Rheumatology, Hôpital Nord, CHU Saint-Etienne, Saint-Etienne, France; INSERM U1059, University of Lyon, Saint-Etienne, France.
Yves-Marie Pers, IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France; Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Lapeyronie University Hospital, 371 Avenue du Doyen Gaston Giraud, Montpellier Cedex 5, 34295, France.
References
- 1. Moltó A, Etcheto A, van der Heijde D, et al. Prevalence of comorbidities and evaluation of their screening in spondyloarthritis: results of the international cross-sectional ASAS-COMOSPA study. Ann Rheum Dis 2016; 75: 1016–1023. [DOI] [PubMed] [Google Scholar]
- 2. Zhao SS, Robertson S, Reich T, et al. Prevalence and impact of comorbidities in axial spondyloarthritis: systematic review and meta-analysis. Rheumatology (Oxford) 2020; 59(Suppl. 4): iv47–iv57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Kotsis K, Voulgari PV, Drosos AA, et al. Health-related quality of life in patients with ankylosing spondylitis: a comprehensive review. Expert Rev Pharmacoecon Outcomes Res 2014; 14: 857–872. [DOI] [PubMed] [Google Scholar]
- 4. Baysal O, Durmuş B, Ersoy Y, et al. Relationship between psychological status and disease activity and quality of life in ankylosing spondylitis. Rheumatol Int 2011; 31: 795–800. [DOI] [PubMed] [Google Scholar]
- 5. Batmaz İ, Sarıyıldız MA, Dilek B, et al. Sleep quality and associated factors in ankylosing spondylitis: relationship with disease parameters, psychological status and quality of life. Rheumatol Int 2013; 33: 1039–1045. [DOI] [PubMed] [Google Scholar]
- 6. Fricchione G, Daly R, Rogers MP, et al. Neuroimmunologic influences in neuropsychiatric and psychophysiologic disorders. Acta Pharmacol Sin 2001; 22: 577–587. [PubMed] [Google Scholar]
- 7. Montgomery SL, Bowers WJ. Tumor necrosis factor-alpha and the roles it plays in homeostatic and degenerative processes within the central nervous system. J Neuroimmune Pharmacol 2012; 7: 42–59. [DOI] [PubMed] [Google Scholar]
- 8. Himmerich H, Berthold-Losleben M, Pollmächer T. The relevance of the TNF-alpha system in psychiatric disorders. Fortschr Neurol Psychiatr 2009; 77: 334–345. [DOI] [PubMed] [Google Scholar]
- 9. Martin TM, Rosenbaum JT. An update on the genetics of HLA B27-associated acute anterior uveitis. Ocul Immunol Inflamm 2011; 19: 108–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Nussenblatt RB. The natural history of uveitis. Int Ophthalmol 1990; 14: 303–308. [DOI] [PubMed] [Google Scholar]
- 11. Monnet D, Breban M, Hudry C, et al. Ophthalmic findings and frequency of extraocular manifestations in patients with HLA-B27 uveitis: a study of 175 cases. Ophthalmology 2004; 111: 802–809. [DOI] [PubMed] [Google Scholar]
- 12. Juanola X, Loza Santamaría E, Cordero-Coma M; SENTINEL Working Group. Description and prevalence of spondyloarthritis in patients with anterior uveitis: the SENTINEL interdisciplinary collaborative project. Ophthalmology 2016; 123: 1632–1636. [DOI] [PubMed] [Google Scholar]
- 13. Khan MA, Haroon M, Rosenbaum JT. Acute anterior uveitis and spondyloarthritis: more than meets the eye. Curr Rheumatol Rep 2015; 17: 59. [DOI] [PubMed] [Google Scholar]
- 14. Smith WM. Gender and spondyloarthropathy-associated uveitis. J Ophthalmol 2013; 2013: 928264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Canouï-Poitrine F, Lekpa FK, Farrenq V, et al. Prevalence and factors associated with uveitis in spondylarthritis patients in France: results from an observational survey. Arthritis Care Res (Hoboken) 2012; 64: 919–924. [DOI] [PubMed] [Google Scholar]
- 16. El Maghraoui A. Extra-articular manifestations of ankylosing spondylitis: prevalence, characteristics and therapeutic implications. Eur J Intern Med 2011; 22: 554–560. [DOI] [PubMed] [Google Scholar]
- 17. Dick AD. Immune mechanisms of uveitis: insights into disease pathogenesis and treatment. Int Ophthalmol Clin 2000; 40: 1–18. [DOI] [PubMed] [Google Scholar]
- 18. Mei Y, Pan F, Gao J, et al. Increased serum IL-17 and IL-23 in the patient with ankylosing spondylitis. Clin Rheumatol 2011; 30: 269–273. [DOI] [PubMed] [Google Scholar]
- 19. El-Asrar AMA, Struyf S, Kangave D, et al. Cytokine profiles in aqueous humor of patients with different clinical entities of endogenous uveitis. Clin Immunol 2011; 139: 177–184. [DOI] [PubMed] [Google Scholar]
- 20. Sauer A, Villard O, Creuzot-Garcher C, et al. Intraocular levels of interleukin 17A (IL-17A) and IL-10 as respective determinant markers of toxoplasmosis and viral uveitis. Clin Vaccine Immunol 2015; 22: 72–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Zhuang Z, Wang Y, Zhu G, et al. Imbalance of Th17/Treg cells in pathogenesis of patients with human leukocyte antigen B27 associated acute anterior uveitis. Sci Rep 2017; 7: 40414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Luger D, Silver PB, Tang J, et al. Either a Th17 or a Th1 effector response can drive autoimmunity: conditions of disease induction affect dominant effector category. J Exp Med 2008; 205: 799–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Curnow SJ, Falciani F, Durrani OM, et al. Multiplex bead immunoassay analysis of aqueous humor reveals distinct cytokine profiles in uveitis. Invest Ophthalmol Vis Sci 2005; 46: 4251–4259. [DOI] [PubMed] [Google Scholar]
- 24. Amadi-Obi A, Yu C-R, Liu X, Mahdi RM, et al. TH17 cells contribute to uveitis and scleritis and are expanded by IL-2 and inhibited by IL-27/STAT1. Nat Med 2007; 13: 711–718. [DOI] [PubMed] [Google Scholar]
- 25. Kezic JM, Glant TT, Rosenbaum JT, et al. Neutralization of IL-17 ameliorates uveitis but damages photoreceptors in a murine model of spondyloarthritis. Arthritis Res Ther 2012; 14: R18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Ke Y, Liu K, Huang G-Q, et al. Anti-inflammatory role of IL-17 in experimental autoimmune uveitis. J Immunol 2009; 182: 3183–3190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Lie E, Lindström U, Zverkova-Sandström T, et al. Tumour necrosis factor inhibitor treatment and occurrence of anterior uveitis in ankylosing spondylitis: results from the Swedish biologics register. Ann Rheum Dis 2017; 76: 1515–1521. [DOI] [PubMed] [Google Scholar]
- 28. Gao X, Wendling D, Botteman MF, et al. Clinical and economic burden of extra-articular manifestations in ankylosing spondylitis patients treated with anti-tumor necrosis factor agents. J Med Econ 2012; 15: 1054–1063. [DOI] [PubMed] [Google Scholar]
- 29. Wendling D, Joshi A, Reilly P, et al. Comparing the risk of developing uveitis in patients initiating anti-tumor necrosis factor therapy for ankylosing spondylitis: an analysis of a large US claims database. Curr Med Res Opin 2014; 30: 2515–2521. [DOI] [PubMed] [Google Scholar]
- 30. van der Heijde D, Dougados M, Landewé R, et al. Sustained efficacy, safety and patient-reported outcomes of certolizumab pegol in axial spondyloarthritis: 4-year outcomes from RAPID-axSpA. Rheumatology (Oxford) 2017; 56: 1498–1509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Heldmann F, Brandt J, van der Horst-Bruinsma IE, et al. The European Ankylosing Spondylitis Infliximab Cohort (EASIC): a European multicentre study of long term outcomes in patients with ankylosing spondylitis treated with infliximab. Clin Exp Rheumatol 2011; 29: 672–680. [PubMed] [Google Scholar]
- 32. Deodhar A, Miceli-Richard C, Baraliakos X, et al. SAT0270 Low incidence of both new-onset and flares of uveitis in secukinumab-treated patients with ankylosing spondylitis: clinical trial and post-marketing safety analysis. Ann Rheum Dis 2018; 77(Suppl. 2): 999. [Google Scholar]
- 33. Jaffe GJ, Dick AD, Brézin AP, et al. Adalimumab in patients with active noninfectious uveitis. N Engl J Med 2016; 375: 932–943. [DOI] [PubMed] [Google Scholar]
- 34. Nguyen QD, Merrill PT, Jaffe GJ, et al. Adalimumab for prevention of uveitic flare in patients with inactive non-infectious uveitis controlled by corticosteroids (VISUAL II): a multicentre, double-masked, randomised, placebo-controlled phase 3 trial. Lancet 2016; 388: 1183–1192. [DOI] [PubMed] [Google Scholar]
- 35. Chateau T, Angioi K, Peyrin-Biroulet L. Two cases of successful ustekinumab treatment for non-infectious uveitis associated with Crohn’s disease. J Crohns Colitis 2020; 14: 571. [DOI] [PubMed] [Google Scholar]
- 36. Mugheddu C, Atzori L, Del Piano M, et al. Successful ustekinumab treatment of noninfectious uveitis and concomitant severe psoriatic arthritis and plaque psoriasis. Dermatol Ther. Epub ahead of print 18 August 2017. DOI: 10.1111/dth.12527. [DOI] [PubMed] [Google Scholar]
- 37. Hueber W, Patel DD, Dryja T, et al. Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis. Sci Transl Med 2010; 2: 52ra72. [DOI] [PubMed] [Google Scholar]
- 38. Dick AD, Tugal-Tutkun I, Foster S, et al. Secukinumab in the treatment of noninfectious uveitis: results of three randomized, controlled clinical trials. Ophthalmology 2013; 120: 777–787. [DOI] [PubMed] [Google Scholar]
- 39. Letko E, Yeh S, Foster CS, et al. Efficacy and safety of intravenous secukinumab in noninfectious uveitis requiring steroid-sparing immunosuppressive therapy. Ophthalmology 2015; 122: 939–948. [DOI] [PubMed] [Google Scholar]
- 40. Messenger W, Hildebrandt L, Mackensen F, et al. Characterisation of uveitis in association with multiple sclerosis. Br J Ophthalmol 2015; 99: 205–209. [DOI] [PubMed] [Google Scholar]
- 41. Seror R, Richez C, Sordet C, et al. Pattern of demyelination occurring during anti-TNF-α therapy: a French national survey. Rheumatology (Oxford) 2013; 52: 868–874. [DOI] [PubMed] [Google Scholar]
- 42. Wendling D, Flipo R-M, Breban M, et al. Coexistence of spondyloarthropathy and multiple sclerosis: a series of 21 cases. Ann Rheum Dis 2008; 67: 901–903. [DOI] [PubMed] [Google Scholar]
- 43. Borman P, Tuncay F, Köybaşi M, et al. Coexistence of ankylosing spondylitis and multiple sclerosis. Acta Neurol Belg 2011; 111: 340–343. [PubMed] [Google Scholar]
- 44. Fominykh V, Shevtsova T, Arzumanian N, et al. Coexistence of multiple sclerosis and ankylosing spondylitis: report of four cases from Russia and review of the literature. J Clin Neurosci 2017; 44: 230–233. [DOI] [PubMed] [Google Scholar]
- 45. Calin A. Is there an association between ankylosing spondylitis and multiple sclerosis? Ann Rheum Dis 1989; 48: 971–972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Wang K, Song F, Fernandez-Escobar A, et al. The properties of cytokines in multiple sclerosis: pros and cons. Am J Med Sci 2018; 356: 552–560. [DOI] [PubMed] [Google Scholar]
- 47. Waisman A, Hauptmann J, Regen T. The role of IL-17 in CNS diseases. Acta Neuropathol 2015; 129: 625–637. [DOI] [PubMed] [Google Scholar]
- 48. Kolbinger F, Huppertz C, Mir A, et al. IL-17A and multiple sclerosis: signaling pathways, producing cells and target cells in the central nervous system. Curr Drug Targets 2016; 17: 1882–1893. [DOI] [PubMed] [Google Scholar]
- 49. Kuwabara T, Ishikawa F, Kondo M, et al. The role of IL-17 and related cytokines in inflammatory autoimmune diseases. Mediators Inflamm 2017; 2017: 908061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Ashtari F, Madanian R, Shaygannejad V, et al. Serum levels of IL-6 and IL-17 in multiple sclerosis, neuromyelitis optica patients and healthy subjects. Int J Physiol Pathophysiol Pharmacol 2019; 11: 267–273. [PMC free article] [PubMed] [Google Scholar]
- 51. Trenova AG, Slavov GS, Draganova-Filipova MN, et al. Circulating levels of interleukin-17A, tumor necrosis factor-alpha, interleukin-18, interleukin-10, and cognitive performance of patients with relapsing-remitting multiple sclerosis. Neurol Res 2018; 40: 153–159. [DOI] [PubMed] [Google Scholar]
- 52. Pegoretti V, Baron W, Laman JD, et al. Selective modulation of TNF–TNFRs signaling: insights for multiple sclerosis treatment. Front Immunol 2018; 9: 925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Medler J, Wajant H. Tumor necrosis factor receptor-2 (TNFR2): an overview of an emerging drug target. Expert Opin Ther Targets 2019; 23: 295–307. [DOI] [PubMed] [Google Scholar]
- 54. Kemanetzoglou E, Andreadou E. CNS demyelination with TNF-α blockers. Curr Neurol Neurosci Rep 2017; 17: 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Engel S, Luessi F, Mueller A, et al. PPMS onset upon adalimumab treatment extends the spectrum of anti-TNF-α therapy-associated demyelinating disorders. Ther Adv Neurol Disord 2020; 13: 1756286419895155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Williams SK, Maier O, Fischer R, et al. Antibody-mediated inhibition of TNFR1 attenuates disease in a mouse model of multiple sclerosis. PLoS One 2014; 9: e90117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. The Lenercept Multiple Sclerosis Study Group and the University of British Columbia MS/MRI Analysis Group. TNF neutralization in MS: results of a randomized, placebo-controlled multicenter study. Neurology 1999; 53: 457–465. [PubMed] [Google Scholar]
- 58. Marotte H, Charrin JE, Miossec P. Infliximab-induced aseptic meningitis. Lancet 2001; 358: 1784. [DOI] [PubMed] [Google Scholar]
- 59. Dreyer L, Magyari M, Laursen B, et al. Risk of multiple sclerosis during tumour necrosis factor inhibitor treatment for arthritis: a population-based study from DANBIO and the Danish Multiple Sclerosis Registry. Ann Rheum Dis 2016; 75: 785–786. [DOI] [PubMed] [Google Scholar]
- 60. Gomez-Gallego M, Meca-Lallana J, Fernandez-Barreiro A. Multiple sclerosis onset during etanercept treatment. Eur Neurol 2008; 59: 91–93. [DOI] [PubMed] [Google Scholar]
- 61. Havrdová E, Belova A, Goloborodko A, et al. Activity of secukinumab, an anti-IL-17A antibody, on brain lesions in RRMS: results from a randomized, proof-of-concept study. J Neurol 2016; 263: 1287–1295. [DOI] [PubMed] [Google Scholar]
- 62. Macaluso F, Guggino G, Mauro D, et al. Safety and efficacy of secukinumab treatment in a patient with ankylosing spondylitis and concomitant multiple sclerosis. Clin Exp Rheumatol 2019; 37: 1096. [PubMed] [Google Scholar]
- 63. Cortese A, Lucchetti R, Altobelli A, et al. Secukinumab may be a valid treatment option in patients with CNS demyelination and concurrent ankylosing spondylitis: Report of two clinical cases. Mult Scler Relat Disord 2019; 35: 193–195. [DOI] [PubMed] [Google Scholar]
- 64. Flanagan EP, Cabre P, Weinshenker BG, et al. Epidemiology of aquaporin-4 autoimmunity and neuromyelitis optica spectrum. Ann Neurol 2016; 79: 775–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Lin J, Li X, Xia J. Th17 cells in neuromyelitis optica spectrum disorder: a review. Int J Neurosci 2016; 126: 1051–1060. [DOI] [PubMed] [Google Scholar]
- 66. Hou M-M, Li Y-F, He L-L, et al. Proportions of Th17 cells and Th17-related cytokines in neuromyelitis optica spectrum disorders patients: a meta-analysis. Int Immunopharmacol 2019; 75: 105793. [DOI] [PubMed] [Google Scholar]
- 67. Pentón-Rol G, Cervantes-Llanos M, Martínez-Sánchez G, et al. TNF-alpha and IL-10 downregulation and marked oxidative stress in neuromyelitis optica. J Inflamm (Lond) 2009; 6: 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Lommers E, Depierreux F, Hansen I, et al. NMOSD with anti-MOG antibodies following anti-TNFα therapy: a case report. Mult Scler Relat Disord 2018; 26: 37–39. [DOI] [PubMed] [Google Scholar]
- 69. Jang H-D, Park J-S, Kim DW, et al. Relationship between dementia and ankylosing spondylitis: A nationwide, population-based, retrospective longitudinal cohort study. PLoS One 2019; 14: e0210335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Griffin WST. Neuroinflammatory cytokine signaling and Alzheimer’s disease. N Engl J Med 2013; 368: 770–771. [DOI] [PubMed] [Google Scholar]
- 71. Chen J-M, Jiang G-X, Li Q-W, et al. Increased serum levels of interleukin-18, -23 and -17 in Chinese patients with Alzheimer’s disease. Dement Geriatr Cogn Disord 2014; 38: 321–329. [DOI] [PubMed] [Google Scholar]
- 72. Tan M-S, Yu J-T, Jiang T, et al. IL12/23 p40 inhibition ameliorates Alzheimer’s disease-associated neuropathology and spatial memory in SAMP8 mice. J Alzheimers Dis 2014; 38: 633–646. [DOI] [PubMed] [Google Scholar]
- 73. vom Berg J, Prokop S, Miller KR, et al. Inhibition of IL-12/IL-23 signaling reduces Alzheimer’s disease–like pathology and cognitive decline. Nat Med 2012; 18: 1812–1819. [DOI] [PubMed] [Google Scholar]
- 74. Saresella M, Marventano I, Calabrese E, et al. A complex proinflammatory role for peripheral monocytes in Alzheimer’s disease. J Alzheimers Dis 2014; 38: 403–413. [DOI] [PubMed] [Google Scholar]
- 75. Wang J, Cheng X, Zhang X, et al. Neuroendocrine immunomodulation network dysfunction in SAMP8 mice and PrP-hAβPPswe/PS1ΔE9 mice: potential mechanism underlying cognitive impairment. Oncotarget 2016; 7: 22988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Hu WT, Chen-Plotkin A, Grossman M, et al. Novel CSF biomarkers for frontotemporal lobar degenerations. Neurology 2010; 75: 2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Saresella M, Calabrese E, Marventano I, et al. Increased activity of Th-17 and Th-9 lymphocytes and a skewing of the post-thymic differentiation pathway are seen in Alzheimer’s disease. Brain Behav Immun 2011; 25: 539–547. [DOI] [PubMed] [Google Scholar]
- 78. Browne TC, McQuillan K, McManus RM, et al. IFN-γ production by amyloid β–specific Th1 cells promotes microglial activation and increases plaque burden in a mouse model of Alzheimer’s disease. J Immunol 2013; 190: 2241–2251. [DOI] [PubMed] [Google Scholar]
- 79. Doecke JD, Laws SM, Faux NG, et al. Blood-based protein biomarkers for diagnosis of Alzheimer disease. Arch Neurol 2012; 69: 1318–1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Hu WT, Watts K, Grossman M, et al. Reduced CSF p-Tau181 to Tau ratio is a biomarker for FTLD-TDP. Neurology 2013; 81: 1945–1952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Yin Y, Wen S, Wang D. Hypoxia enhances stimulating effect of amyloid beta peptide (25-35) for Interleukin 17 and T helper lymphocyte subtype 17 upregulation in cultured peripheral blood mononuclear cells. Microbiol Immunol 2009; 53: 281–286. [DOI] [PubMed] [Google Scholar]
- 82. McManus R, Higgins S, Mills K, et al. Respiratory infection promotes T cell infiltration and amyloid-β deposition in APP/PS1 mice. Neurobiol Aging 2014; 35: 109–121. [DOI] [PubMed] [Google Scholar]
- 83. Jin J-J, Kim H-D, Maxwell JA, et al. Toll-like receptor 4-dependent upregulation of cytokines in a transgenic mouse model of Alzheimer’s disease. J Neuroinflammation 2008; 5: 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Ravari A, Mirzaei T, Kennedy D, et al. Chronoinflammaging in Alzheimer; a systematic review on the roles of toll like receptor 2. Life Sci 2017; 171: 16–20. [DOI] [PubMed] [Google Scholar]
- 85. Zota V, Nemirovsky A, Baron R, et al. HLA-DR alleles in amyloid β-peptide autoimmunity: a highly immunogenic role for the DRB1*1501 allele. J Immunol 2009; 183: 3522–3530. [DOI] [PubMed] [Google Scholar]
- 86. Liu Y, Yu J, Zhang W, et al. Interleukin-23 receptor polymorphisms are associated with Alzheimer’s disease in Han Chinese. J Neuroimmunol 2014; 271: 43–48. [DOI] [PubMed] [Google Scholar]
- 87. Ley K, Smith E, Stark MA. IL-17A-producing neutrophil-regulatory Tn lymphocytes. Immunol Res 2006; 34: 229–242. [DOI] [PubMed] [Google Scholar]
- 88. von Vietinghoff S, Ley K. Interleukin 17A controls interleukin 17F production and maintains blood neutrophil counts in mice. J Immunol 2009; 183: 865–873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Zhang J, Ke K-F, Liu Z, et al. Th17 cell-mediated neuroinflammation is involved in neurodegeneration of Aβ1-42-induced Alzheimer’s disease model rats. PLoS One 2013; 8:e75786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Alvarez A, Cacabelos R, Sanpedro C, et al. Serum TNF-alpha levels are increased and correlate negatively with free IGF-I in Alzheimer disease. Neurobiol Aging 2007; 28: 533–536. [DOI] [PubMed] [Google Scholar]
- 91. Perry RT, Collins JS, Wiener H, et al. The role of TNF and its receptors in Alzheimer’s disease. Neurobiol Aging 2001; 22: 873–883. [DOI] [PubMed] [Google Scholar]
- 92. Wang T. TNF-alpha G308A polymorphism and the susceptibility to Alzheimer’s disease: an updated meta-analysis. Arch Med Res 2015; 46: 24–30.e1. [DOI] [PubMed] [Google Scholar]
- 93. Brosseron F, Krauthausen M, Kummer M, et al. Body fluid cytokine levels in mild cognitive impairment and Alzheimer’s disease: a comparative overview. Mol Neurobiol 2014; 50: 534–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Decourt B, Lahiri DK, Sabbagh MN. Targeting tumor necrosis factor alpha for Alzheimer’s disease. Curr Alzheimer Res 2017; 14: 412–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Fillit H, Ding WH, Buee L, et al. Elevated circulating tumor necrosis factor levels in Alzheimer’s disease. Neurosci Lett 1991; 129: 318–320. [DOI] [PubMed] [Google Scholar]
- 96. Paganelli R, Di Iorio A, Patricelli L, et al. Proinflammatory cytokines in sera of elderly patients with dementia: levels in vascular injury are higher than those of mild-moderate Alzheimer’s disease patients. Exp Gerontol 2002; 37: 257–263. [DOI] [PubMed] [Google Scholar]
- 97. Chen J-H, Ke K-F, Lu J-H, et al. Protection of TGF-β1 against neuroinflammation and neurodegeneration in Aβ1–42-induced Alzheimer’s disease model rats. PLoS One 2015; 10:e0116549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Diaz-Gerevini G, Repossi G, Dain A, et al. Beneficial action of resveratrol: how and why? Nutrition 2016; 32: 174–178. [DOI] [PubMed] [Google Scholar]
- 99. Ekert JO, Gould RL, Reynolds G, et al. TNF alpha inhibitors in Alzheimer’s disease: a systematic review. Int J Geriatr Psychiatry 2018; 33: 688–694. [DOI] [PubMed] [Google Scholar]
- 100. Montgomery SL, Mastrangelo MA, Habib D, et al. Ablation of TNF-RI/RII expression in Alzheimer’s disease mice leads to an unexpected enhancement of pathology: implications for chronic pan-TNF-α suppressive therapeutic strategies in the brain. Am J Pathol 2011; 179: 2053–2070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Butchart J, Brook L, Hopkins V, et al. Etanercept in Alzheimer disease: a randomized, placebo-controlled, double-blind, phase 2 trial. Neurology 2015; 84: 2161–2168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Ungprasert P, Srivali N, Kittanamongkolchai W. Risk of Parkinson’s disease among patients with psoriasis: a systematic review and meta-analysis. Indian J Dermatol 2016; 61: 152–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Power JHT, Barnes OL, Chegini F. Lewy bodies and the mechanisms of neuronal cell death in Parkinson’s disease and dementia with lewy bodies. Brain Pathol 2017; 27: 3–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Sulzer D, Alcalay RN, Garretti F, et al. T cells from patients with Parkinson’s disease recognize α-synuclein peptides. Nature 2017. 29; 546: 656–661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Harms AS, Cao S, Rowse AL, et al. MHCII is required for α-synuclein-induced activation of microglia, CD4 T cell proliferation, and dopaminergic neurodegeneration. J Neurosci 2013; 33: 9592–9600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Reynolds AD, Stone DK, Hutter JAL, et al. Regulatory T cells attenuate Th17 cell-mediated nigrostriatal dopaminergic neurodegeneration in a model of Parkinson’s disease. J Immunol 2010; 184: 2261–2271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Liu Z, Huang Y, Cao B-B, et al. Th17 cells induce dopaminergic neuronal death via LFA-1/ICAM-1 interaction in a mouse model of Parkinson’s disease. Mol Neurobiol 2017; 54: 7762–7776. [DOI] [PubMed] [Google Scholar]
- 108. Park J, Lee J-W, Cooper SC, et al. Parkinson disease-associated LRRK2 G2019S transgene disrupts marrow myelopoiesis and peripheral Th17 response. J Leukoc Biol 2017; 102: 1093–1102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Storelli E, Cassina N, Rasini E, et al. Do Th17 lymphocytes and IL-17 contribute to Parkinson’s disease? A systematic review of available evidence. Front Neurol 2019; 10: 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Kustrimovic N, Comi C, Magistrelli L, et al. Parkinson’s disease patients have a complex phenotypic and functional Th1 bias: cross-sectional studies of CD4+ Th1/Th2/T17 and Treg in drug-naïve and drug-treated patients. J Neuroinflammation 2018; 15: 205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Sommer A, Marxreiter F, Krach F, et al. Th17 lymphocytes induce neuronal cell death in a human iPSC-based model of Parkinson’s disease. Cell Stem Cell 2018; 23: 123–131.e6. [DOI] [PubMed] [Google Scholar]
- 112. Lindenau JD, Altmann V, Schumacher-Schuh AF, et al. Tumor necrosis factor alpha polymorphisms are associated with Parkinson’s disease age at onset. Neurosci Lett 2017; 658: 133–136. [DOI] [PubMed] [Google Scholar]
- 113. Alam Q, Alam MZ, Mushtaq G, et al. Inflammatory process in Alzheimer’s and Parkinson’s diseases: central role of cytokines. Curr Pharm Des 2016; 22: 541–548. [DOI] [PubMed] [Google Scholar]
- 114. Gupta V, Garg RK, Khattri S. Levels of IL-8 and TNF-α decrease in Parkinson’s disease. Neurol Res 2016; 38: 98–102. [DOI] [PubMed] [Google Scholar]
- 115. Kouchaki E, Kakhaki RD, Tamtaji OR, et al. Increased serum levels of TNF-α and decreased serum levels of IL-27 in patients with Parkinson disease and their correlation with disease severity. Clin Neurol Neurosurg 2018; 166: 76–79. [DOI] [PubMed] [Google Scholar]
- 116. Mogi M, Harada M, Riederer P, et al. Tumor necrosis factor-alpha (TNF-alpha) increases both in the brain and in the cerebrospinal fluid from parkinsonian patients. Neurosci Lett 1994; 165: 208–210. [DOI] [PubMed] [Google Scholar]
- 117. Çomoğlu SS, Güven H, Acar M, et al. Tear levels of tumor necrosis factor-alpha in patients with Parkinson’s disease. Neurosci Lett 2013; 553: 63–67. [DOI] [PubMed] [Google Scholar]
- 118. Peter I, Dubinsky M, Bressman S, et al. Anti-tumor necrosis factor therapy and incidence of Parkinson disease among patients with inflammatory bowel disease. JAMA Neurol 2018; 75: 939–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Stubbs B, Aluko Y, Myint PK, et al. Prevalence of depressive symptoms and anxiety in osteoarthritis: a systematic review and meta-analysis. Age Ageing 2016; 45: 228–235. [DOI] [PubMed] [Google Scholar]
- 120. Zhao S, Thong D, Miller N, et al. The prevalence of depression in axial spondyloarthritis and its association with disease activity: a systematic review and meta-analysis. Arthritis Res Ther 2018; 20: 140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Strober BE, Langley RGB, Menter A, et al. No elevated risk for depression, anxiety or suicidality with secukinumab in a pooled analysis of data from 10 clinical studies in moderate-to-severe plaque psoriasis. Br J Dermatol 2018; 178: e105–e107. [DOI] [PubMed] [Google Scholar]
- 122. Komori T, Otsuka A, Honda Y, et al. Exacerbation of depression in a psoriatic arthritis patient possibly induced by secukinumab. Eur J Dermatol 2016; 26: 506–507. [DOI] [PubMed] [Google Scholar]
- 123. Macfarlane GJ, Rotariu O, Jones GT, et al. Determining factors related to poor quality of life in patients with axial spondyloarthritis: results from the British Society for Rheumatology Biologics Register (BSRBR-AS). Ann Rheum Dis 2020; 79: 202–208. [DOI] [PubMed] [Google Scholar]
- 124. Nadeem A, Ahmad SF, Al-Harbi NO, et al. IL-17A causes depression-like symptoms via NFκB and p38MAPK signaling pathways in mice: implications for psoriasis associated depression. Cytokine 2017; 97: 14–24. [DOI] [PubMed] [Google Scholar]
- 125. Beurel E, Lowell JA. Th17 cells in depression. Brain Behav Immun 2018; 69: 28–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Lu Y, Ho CS, Liu X, et al. Chronic administration of fluoxetine and pro-inflammatory cytokine change in a rat model of depression. PLoS One 2017; 12: e0186700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Jha MK, Minhajuddin A, Gadad BS, et al. Interleukin 17 selectively predicts better outcomes with bupropion-SSRI combination: novel T cell biomarker for antidepressant medication selection. Brain Behav Immun 2017; 66: 103–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Berthold-Losleben M, Himmerich H. The TNF-alpha system: functional aspects in depression, narcolepsy and psychopharmacology. Curr Neuropharmacol 2008; 6: 193–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Ng A, Tam WW, Zhang MW, et al. IL-1β, IL-6, TNF- α and CRP in elderly patients with depression or Alzheimer’s disease: systematic review and meta-analysis. Sci Rep 2018; 8: 12050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Shin K-H, Jeong HC, Choi D-H, et al. Association of TNF-alpha G-308A gene polymorphism with depression: a meta-analysis. Neuropsychiatr Dis Treat 2017; 13: 2661–2668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Himmerich H, Fulda S, Linseisen J, et al. Depression, comorbidities and the TNF-alpha system. Eur Psychiatry 2008; 23: 421–429. [DOI] [PubMed] [Google Scholar]
- 132. Dowlati Y, Herrmann N, Swardfager W, et al. A meta-analysis of cytokines in major depression. Biol Psychiatry 2010; 67: 446–457. [DOI] [PubMed] [Google Scholar]
- 133. Koo J, Ho RS, Thibodeaux Q. Depression and suicidality in psoriasis and clinical studies of brodalumab: a narrative review. Cutis 2019; 104: 361–365. [PubMed] [Google Scholar]
- 134. Ertenli I, Ozer S, Kiraz S, et al. Infliximab, a TNF-α antagonist treatment in patients with ankylosing spondylitis: the impact on depression, anxiety and quality of life level. Rheumatol Int 2012; 32: 323–330. [DOI] [PubMed] [Google Scholar]
- 135. Jafri F, Sammut A. A rare case of suicidal ideation related to Adalimumab use. Open Access Rheumatol 2018; 10: 113–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Raison CL, Rutherford RE, Woolwine BJ, et al. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA Psychiatry 2013; 70: 31–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Irwin MR. Sleep and inflammation: partners in sickness and in health. Nat Rev Immunol 2019; 19: 702–715. [DOI] [PubMed] [Google Scholar]
- 138. Imeri L, Opp MR. How (and why) the immune system makes us sleep. Nat Rev Neurosci 2009; 10: 199–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Irwin MR, Olmstead R, Valladares EM, et al. Tumor necrosis factor antagonism normalizes rapid eye movement sleep in alcohol dependence. Biol Psychiatry 2009; 66: 191–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Vgontzas AN, Zoumakis E, Lin H-M, et al. Marked decrease in sleepiness in patients with sleep apnea by etanercept, a tumor necrosis factor-alpha antagonist. J Clin Endocrinol Metab 2004; 89: 4409–4413. [DOI] [PubMed] [Google Scholar]
- 141. Haliloglu S, Carlioglu A, Akdeniz D, et al. Fibromyalgia in patients with other rheumatic diseases: prevalence and relationship with disease activity. Rheumatol Int 2014; 34: 1275–1280. [DOI] [PubMed] [Google Scholar]
- 142. Salaffi F, De Angelis R, Carotti M, et al. Fibromyalgia in patients with axial spondyloarthritis: epidemiological profile and effect on measures of disease activity. Rheumatol Int 2014; 34: 1103–1110. [DOI] [PubMed] [Google Scholar]
- 143. Azevedo VF, Paiva E, dos S, et al. Occurrence of fibromyalgia in patients with ankylosing spondylitis. Rev Bras Reumatol 2010; 50: 646–650. [PubMed] [Google Scholar]
- 144. Yunus MB. The prevalence of fibromyalgia in other chronic pain conditions. Pain Res Treat 2012; 2012: 584573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Baraliakos X, Regel A, Kiltz U, et al. Patients with fibromyalgia rarely fulfil classification criteria for axial spondyloarthritis. Rheumatology (Oxford) 2018; 57: 1541–1547. [DOI] [PubMed] [Google Scholar]
- 146. Pernambuco AP, Schetino LPL, Alvim CC, et al. Increased levels of IL-17A in patients with fibromyalgia. Clin Exp Rheumatol 2013; 31(6 Suppl. 79): S60–S63. [PubMed] [Google Scholar]
- 147. Larun L, Brurberg KG, Odgaard-Jensen J, et al. Exercise therapy for chronic fatigue syndrome. Cochrane Database Syst Rev 2017; 4: CD003200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Häuser W, Klose P, Langhorst J, et al. Efficacy of different types of aerobic exercise in fibromyalgia syndrome: a systematic review and meta-analysis of randomised controlled trials. Arthritis Res Ther 2010; 12: R79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Ernberg M, Christidis N, Ghafouri B, et al. Plasma cytokine levels in fibromyalgia and their response to 15 weeks of progressive resistance exercise or relaxation therapy. Mediators Inflamm 2018; 2018: 3985154. [DOI] [PMC free article] [PubMed] [Google Scholar]