Next Article in Journal
Iron at the Interface of Hepatocellular Carcinoma
Next Article in Special Issue
Adiponectin and Leptin Exert Antagonizing Effects on HUVEC Tube Formation and Migration Modulating the Expression of CXCL1, VEGF, MMP-2 and MMP-9
Previous Article in Journal
Putrescine Depletion Affects Arabidopsis Root Meristem Size by Modulating Auxin and Cytokinin Signaling and ROS Accumulation
Previous Article in Special Issue
The Inflammatory Profile of Obesity and the Role on Pulmonary Bacterial and Viral Infections
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Adiponectin Deregulation in Systemic Autoimmune Rheumatic Diseases

1
Department of Rheumatology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
2
Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
3
Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
4
FAMNIT, University of Primorska, 6000 Koper, Slovenia
5
Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(8), 4095; https://doi.org/10.3390/ijms22084095
Submission received: 16 March 2021 / Revised: 8 April 2021 / Accepted: 13 April 2021 / Published: 15 April 2021
(This article belongs to the Special Issue Adipokines in Health and Diseases)

Abstract

:
Deregulation of adiponectin is found in systemic autoimmune rheumatic diseases (SARDs). Its expression is downregulated by various inflammatory mediators, but paradoxically, elevated serum levels are present in SARDs with high inflammatory components, such as rheumatoid arthritis and systemic lupus erythematosus. Circulating adiponectin is positively associated with radiographic progression in rheumatoid arthritis as well as with cardiovascular risks and lupus nephritis in systemic lupus erythematosus. However, in SARDs with less prominent inflammation, such as systemic sclerosis, adiponectin levels are low and correlate negatively with disease activity. Regulators of adiponectin gene expression (PPAR-γ, Id3, ATF3, and SIRT1) and inflammatory cytokines (interleukin 6 and tumor necrosis factor α) are differentially expressed in SARDs and could therefore influence total adiponectin levels. In addition, anti-inflammatory therapy could also have an impact, as tocilizumab treatment is associated with increased serum adiponectin. However, anti-tumor necrosis factor α treatment does not seem to affect its levels. Our review provides an overview of studies on adiponectin levels in the bloodstream and other biological samples from SARD patients and presents some possible explanations why adiponectin is deregulated in the context of therapy and gene regulation.

1. The Adiponectin Paradox

Adiponectin expression is downregulated by inflammatory mediators such as interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α). Accordingly, decreased serum levels have been described in diseases with low-grade chronic inflammation such as type 2 diabetes, obesity, and atherosclerosis, and adiponectin deficiency has been linked to the pathogenesis of these diseases [1,2]. Paradoxically, high adiponectin levels are associated with various inflammatory diseases such as rheumatoid arthritis (RA), despite persistent inflammation, and have been associated with general and cardiovascular mortality [1]. Thus, it appears that the optimal window of adiponectin concentration is tight and any deregulation leads to a pathology. However, the explanation why adiponectin expression and regulation is different in some inflammatory diseases is not known. Resolving this paradox could provide an important insight into the pathogenesis of these diseases and clarify how adiponectin may be involved, potentially making it a treatment target.
A majority of up-to-date reviews addresses the association between circulating adiponectin levels and specific immune-mediated, rheumatic, or connective tissue diseases [3,4,5,6,7], or focuses on the role of adiponectin in the pathogenesis of these diseases [1,8,9,10].
In this review, we will examine the deregulation of adiponectin in the setting of several systemic autoimmune rheumatic diseases (SARDs) to try to provide some possible explanations for the paradoxically elevated levels in highly inflammatory SARDs. We will present adiponectin levels in circulation and in other biological samples in addition to associations with clinical manifestations of the diseases. Our review is the first in the field to highlight the effects of various drugs used in SARDs on circulating adiponectin levels and to suggest how inflammatory cytokines, such as interleukin-6 and tumor necrosis factor α, and gene regulation might contribute to deregulated adiponectin expression in SARDs.

1.1. Adiponectin Expression, Structure, Isoforms and Signaling

Adiponectin is the most abundant circulating adipokine in a human. It is present in serum at 0.01% to 0.05% of total proteins and plasma concentrations of 4 to 37 µg/mL [11]. They are in a range of about a thousand times higher than that of other hormones (e.g., insulin, leptin) and closer to the concentrations of some carrier proteins (e.g., retinol-binding proteins) [12]. Most adiponectin is produced by healthy adipose tissue, which is why it is classified as adipokine. Other tissues (Figure 1) and cells, such as skeletal myocytes, cardiomyocytes, osteoblasts, liver parenchyma cells, pituitary and endothelial cells may also be a source [13,14].
The structure of adiponectin consists of four parts, the N-terminal signal sequence, the variable region, the collagenous domain and the C1q-like globular domain [11]. After synthesis, adiponectin undergoes further post-translational modifications to form low molecular weight (LMW) trimers, medium molecular weight (MMW) hexamers or multimeric high molecular weight (HMW) isoforms (Figure 2a). They are secreted and bind differently to adiponectin receptors [15]. AdipoR1 and AdipoR2 are the best characterized receptors, while binding to T-cadherin and calreticulin is less studied. After binding to AdipoR1/R2, adiponectin stimulates two main signaling pathways, through PPAR-α and AMPK. Their activation and the activation of other downstream molecules leads to metabolic effects (e.g., fatty acid oxidation, glucose uptake), vasodilatation, as well as reduced apoptosis, inflammation, and fibrosis (Figure 2b). In AdipoR, pockets with ceramidase activity capable of metabolizing sphingosine to sphingosine-1-phosphate and thereby reducing cellular ceramide content were discovered [11].

1.2. What Affects Adiponectin Levels?

Adiponectin levels are influenced by several physiological, pathological and external factors. They are dependent on ethnicity and correlate negatively with body mass index, but positively with age. High levels of adiponectin are present in centenarians, where they may act as a compensatory response in maintaining metabolic and redox homeostasis. Interestingly, minimal levels are observed early in the morning, suggesting that the circadian rhythm may also be involved in regulation. In addition, sexual dimorphism is observed, with higher total and higher HMW adiponectin concentrations in women. This could be due to the inhibitory effect of testosterone on HMW adiponectin production [11], and due to the fact that females (after puberty) generally have a higher percentage of body fat than males [16]. Adiponectin expression is affected by endoplasmic reticulum stress, oxidative stress [17] and β-adrenergic activation [15]. It has been shown that some therapeutic agents, e.g., antidiabetic agents thiazolidinediones (TZD), also affect adiponectin levels [11].
Adiponectin is expressed in healthy adipocytes, whereas adipose tissue in obesity is hypoxic and inflamed, and as such adipocytes produce less adiponectin [18]. Reduced adiponectin levels are also observed in obesity-related diseases such as type 2 diabetes, cardiovascular disease (CVD) [13] and obesity-associated cancers [19]. Another illustration of this phenomenon are diseases in which adipose tissue is reduced, such as anorexia nervosa, and these usually have elevated serum adiponectin levels [20]. It should be noted that the local loss of adipocytes can also have a major impact on disease pathogenesis. In the case of systemic sclerosis (SSc), dedifferentiation of adipocytes in the intradermal fat depot leads to a local decrease in adiponectin synthesis. The inhibitory effect of adiponectin on fibroblasts is lost, allowing the excessive production of the extracellular matrix, which leads to fibrosis [21,22].

2. Adiponectin in Systemic Autoimmune Rheumatic Diseases

Systemic autoimmune rheumatic diseases (SARDs) are heterogeneous disorders with prominent autoimmune dysregulation leading to multiorgan involvement and varying degrees of inflammation. Their etiology is unknown, but a genetic predisposition has been found [22,23]. SARDs include RA (the most frequent disease), connective tissue diseases such as systemic lupus erythematosus (SLE), Sjögren’s syndrome (SS), systemic sclerosis (SSc), and antiphospholipid syndrome (APS). Since there is no cure for these diseases, long-term pharmacotherapy is required and significant morbidity and mortality are observed. There are many studies focusing on adiponectin serum levels in various SARDs. The general circulating values calculated in meta-analyses of some of these are presented in Figure 3.

2.1. Rheumatoid Arthritis

RA is characterized by chronic inflammation of the synovium, leading to joint deformation. Small and large joints are affected, characterized by synovial hyperplasia, bone edema progressing into a joint space and bone erosion. Systemic inflammation and extra-articular manifestations may also occur affecting the skin, eyes, heart, lung, kidneys, and nervous and gastrointestinal systems. Additionally RA patients have a high risk of CVD [24]. Obesity increases the probability of developing RA and is associated with poorer disease outcomes and impaired responses to treatment [25].

2.1.1. Serum

A meta-analysis from 2017 (12 studies, RA n = 784, healthy controls (HCs) n = 655) revealed that circulating adiponectin levels are significantly higher in RA patients than in HCs (SMD = 1.5) [7]. Several additional studies published later (RA n = 168, HC n = 90) showed a similar trend [26,27,28] and some found no difference (RA n = 131, HC n = 131) [29,30], while one study reported lower levels (RA n = 95, HC n = 95) [31]. However, adiponectin levels were not changed between RA (n = 820) and osteoarthritis or undifferentiated arthritis controls (n = 298), [30,32,33,34,35]. Also, no changes were seen when comparing only early RA patients (n = 97) and HC (n = 96) [29,36].
Circulating adiponectin levels in RA are associated with several clinically relevant characteristics as shown in Table 1. RA disease activity, most commonly measured as Disease Activity Score of 28 joints (DAS28), showed positive correlations with serum adiponectin levels in the majority of the studies. Higher adiponectin levels are associated with radiographic severity and progression and are independently related to poorer bone outcomes and reduced muscle mass. However, the status of adiponectin in carotid atherosclerosis is not clear. Lower serum levels of adiponectin were found in RA smokers compared to never smokers [37]. Two studies reported lower adiponectin in obese RA patients [38,39], while one found similar levels compared to non-obese patients [40]. In a cohort of subjects who had suffered from obesity (followed for up to 29 years), high serum levels of adiponectin at baseline were associated with an increased risk for RA [41].

2.1.2. Synovial Fluid

Adiponectin levels in the synovial fluid of RA patients (n = 39) were lower than in serum and correlated positively with disease activity [46]. Specifically, compared to serum levels the HMW adiponectin levels in synovial fluid were lower but the LMW levels were higher. No significant difference was found for MMW levels (n = 7) [58].

2.1.3. Cells/Tissues

Synovium with articular adipose tissue, in particular synoviocytes and articular adipocytes from RA and osteoarthritis patients, express adiponectin strongly both at the transcriptional and the protein level [59]. In RA tissue explants, the synovial membrane produces twice as much adiponectin as the articular adipose tissue regardless of activation status [60]. Also, subcutaneous abdominal adipose tissue of RA patients secretes more adiponectin than the corresponding osteoarthritis tissue [33,34].

2.2. Systemic Lupus Erythematosus

SLE is characterized by the formation of immune complexes that are deposited in the body and cause inflammation and complement activation. The heterogeneous presentation of the disease includes neuropsychiatric, gastrointestinal, hematologic, renal, pulmonary and cardiovascular manifestations and cardiovascular disease (CVD) occurs in more than 50% of SLE patients. With atherosclerosis being an important long-term complication, patients tend to develop carotid intima media thickening (IMT) and carotid plaques [61]. Lupus nephritis (LN), a form of glomerulonephritis, is one of the most serious organ involvements in SLE, and despite the advances in understanding its pathology and in improving treatment options, it remains a significant cause of death in SLE patients [62]. Disease activity is measured by the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) [63].

2.2.1. Serum

Circulating adiponectin levels are significantly higher in SLE than in HC (SMD = 0.547), as shown by a meta-analysis from 2020 (12 studies, SLE n = 1024, HC n = 720) [4]. Disease activity did not seem to correlate with serum adiponectin as evidenced by a meta-analysis in 2017 (SLE n = 782, HC n = 550) [4], but higher concentrations were associated with the presence of LN and correlated with the severity of proteinuria [64,65,66,67].
Studies conducted in SLE generally show a positive association of adiponectin with atherosclerotic development. Higher concentrations were found in patients with carotid plaque (plaque n = 118, no plaque n = 186) [68,69,70]; however, two studies from the same research group could not demonstrate any association [71,72]. A positive correlation between adiponectin levels and the presence of carotid plaque was independent of age, disease duration or SLE treatment [68]. Therefore, adiponectin, in combination with other markers, may be a potential biomarker for plaque prediction. In SLE patients with carotid IMT ≤ 0.8, adiponectin levels corresponded with IMT [73], while a negative correlation between adiponectin and vascular stiffness parameters was found [74].

2.2.2. Urine

Several studies suggest that adiponectin is a potential urine biomarker to discriminate LN SLE patients. Adiponectin levels and the adiponectin-to-creatinine ratio are significantly higher in the urine of SLE patients with renal involvement (renal involvement n = 25, no renal involvement n = 25) [75] and in active LN SLE (n = 33) compared to active non-LN SLE (n = 16), or patients with only LN history (n = 30) [76]. Similarly, adiponectin urine concentrations are significantly elevated in active LN (n = 125) compared to inactive LN (n = 31), SLE without LN (n = 36), or HC (n = 55) [77]. The adiponectin-to-creatinine ratio in patients with LN (n = 27) was also increased compared to normal controls (n = 8) [66]. There were differences in urine adiponectin levels based on LN activity status, as observed in a renal biopsy; levels were lower in patients with low-to-moderate LN activity than in patients with high LN activity [78,79]. Adiponectin is also included as a biomarker for the calculation of the Pediatric Renal Activity Index for Lupus (p-RAIL), which reflects histological LN activity [78,80].

2.2.3. Cells

Adiponectin expression in peripheral blood mononuclear cells (PBMCs) was significantly higher in SLE patients (n = 46) compared to HC (n = 51). However, there were no differences in expression between SLE patients with and without LN and no associations with major clinical and laboratory parameters [81].

2.3. Ankylosing Spondylitis

Ankylosing spondylitis (AS) is a disease of the sacroiliac joint and the spine and the adjacent soft tissues such as tendons and ligaments. It is associated with the presence of the major histocompatibility complex class I allele HLA-B27 and the interleukin 23/17 axis [82].

Serum

Adiponectin levels do not differ significantly in AS patients compared to HC, as concluded by meta-analysis in 2017 (6 studies, AS n = 273, HC n = 202) [6] and one additional study (AS n = 20, HC n = 11) [83]. It needs to be mentioned that the lack of significance might be due to the sample size because the meta-analysis showed SMD = 0.460. Adiponectin levels did not change in longitudinal monitoring over the two-year period [83], and no correlation with disease activity or functional indices was found [84]. Clinically, baseline serum levels were lower in patients who showed radiographic progression of the spine after two years. There was also a significant inverse correlation between radiographic progression of the spine and HMW adiponectin isoform levels [85].

2.4. Systemic Sclerosis

SSc is characterized by microvascular damage, changes in the immune system and extensive fibrosis of the skin and organs such as lungs, kidneys, gastrointestinal tract and heart. Skin fibrosis, usually quantified by the modified Rodnan skin score (mRSS), is a distinguishing feature of SSc [86]. Pulmonary complications are another hallmark, as about 40% of patients develop interstitial lung disease (ILD); about 15% have pulmonary arterial hypertension; and the majority of patients also have pathological changes to the gastrointestinal tract [86]. The early stage of the disease is usually characterized by microvascular pathology that can manifest itself as skin ulcers, and the late stage is characterized by abnormal fibroblast activation leading to tissue fibrosis [87]. Based on the extent of skin fibrosis and involvement of internal organs, SSc patients can be divided into two subgroups: limited cutaneous SSc (lcSSc) and diffuse cutaneous SSc (dcSSc), the latter being associated with the more severe disease course and poorer outcomes [86].

2.4.1. Serum

Circulating adiponectin levels are lower in SSc patients compared to HC (SMD = −0.638), as confirmed by a meta-analysis published in 2017 (11 studies, SSc n = 511, HC n = 341) [88] and a later published study (SSc n = 100, HC n = 20) [89]. The same meta-analysis showed, although not significantly, associations of low adiponectin levels in Caucasian and Asian ethnic groups [88]. Additionally, two other studies (SSc n = 246), found that patients with dcSSc had significantly lower concentrations than patients with lcSSc or HC [88,90,91,92]; thus, the downregulation in certain studies depended on the percentage of dcSSc and lcSSc patients included in each study. In dcSSc serum, adiponectin was the seventeenth most downregulated protein, out of 228 determined proteins [93].
Negative correlations between adiponectin levels and disease activity and progression, measured as Valentini disease activity index [94] and mRSS, were reported [87,95,96], with only one study reporting a positive correlation between adiponectin and mRRS in dcSSc patients [97]. One study found that patients with reduced concentrations showed a higher prevalence of pitting scars and pulmonary fibrosis [96].
The results on differences in adiponectin levels between early versus late SSc are not consistent. The majority (early SSc n = 49, late SSc n = 99) agree that the levels are lower in early SSc [95,97]. However, two studies reported either an opposite trend (early SSc n = 20, late SSc n = 16) [98] or no differences at all (early SSc n = 13, late SSc n = 16) [94]. The results are difficult to compare due to varying definitions of early SSc and late SSc from <18 months to 5 years for early SSc and >2 to 5 years of disease duration for late SSc. In addition, some studies included all SSc patients, while others focused only on dcSSc patients.

2.4.2. Skin

SSc skin biopsies were examined for the expression of adiponectin and adiponectin-regulated genes. Mean relative transcript for adiponectin in skin tissue differed in the SSc subsets because the values in dcSSc patients (n = 5) were reduced compared to lcSSc patients (n = 7) and HC (n = 7) [96]. An inverse correlation of mRNA adiponectin levels with mRSS was found in lesional skin biopsies of early dcSSc (n = 15) and lcSSc patients (n = 6) [95]. Dermal levels of cellular phosphorylated AMPK, a molecule downstream of adiponectin signaling, were significantly reduced in SSc skin (n = 19) compared to HC (n = 4). In addition, lower or absent phosphorylated AMPK in myofibroblasts, were observed in the lesional skin of patients. Changes in gene expression were also found in SSc skin (n = 70) compared to HC (n = 22), with reduced scores of adiponectin signaling pathways [22].
Skin fibrosis is usually accompanied by a consistent decrease in dermal white adipose tissue (dWAT), an important source of adiponectin. The SSc patient skin biopsies showed a reduction in adipocyte number and size, as well as dWAT replacement by a fibrous matrix. This caused a local decrease in adiponectin synthesis, which resulted in the loss of its inhibitory signals on fibroblasts, allowing their activation into myofibroblasts. Adipocytes may be an important source of myofibroblasts as they are capable of undergoing adipocyte mesenchymal transition (AMT) (Figure 4). In this process, adipocytes lose expression of their distinct markers and begin to express myofibroblast markers, such as α-SMA, suggesting that they actively contribute to the extensive accumulation of extracellular matrix components that lead to fibrosis [21,99].

2.4.3. Other

Overall, adiponectin expression in histologically stained lung tissue samples from patients with SSc and patients with idiopathic pulmonary fibrosis was significantly reduced compared to controls. However, no reduction of adiponectin was found in the study of early stage fibrotic bronchoalveolar lavage and lung protein lysates. In the same study, adiponectin was examined in gastroscopic biopsies, and they showed lower adiponectin levels in SSc gastritis compared to gastritis not associated with SSc [100].

2.5. Sjögren Syndrome

In Sjögren syndrome (SS), inflammation of exocrine glandular tissue, usually the lachrymal and salivary glands is present, leading to xerostomia, keratoconjunctivitis sicca and enlargement of the parotid gland. Salivary glandular epithelial cells (SGEC) are important in the pathogenesis of SS [101]. SS often overlaps with other SARDs and may also affect other organ systems and cause polyarthritis, cutaneous vasculitis, peripheral neuropathy, lung disorders, nephritis, optic neuritis, multiple sclerosis-like diseases. In addition, SS patients have an increased risk of lymphoma. Histopathologically, focal lymphocytic infiltrates, which are mainly located around the glandular ducts, are seen [102].

2.5.1. Serum

Studies of serum concentrations of adiponectin in SS patients are scarce and they report either similar or higher circulating levels. Concentrations of adiponectin in primary SS (n = 29), SS associated with rheumatoid arthritis (n = 30), patients with non-autoimmune sicca syndrome (n = 17), and HC (n = 15) were at similar levels [103]. However, in another study, concentrations were higher in primary SS patients (n = 71) than in HC (n = 71) [102].

2.5.2. Salivary Gland and Saliva

Adiponectin expression in minor salivary glands is among the highest tissue expression levels in the organism as shown in Figure 1. Adiponectin is additionally upregulated in the SGEC of SS patients, exerting a protective function against apoptosis [104]. In saliva, adiponectin levels normalized to total protein are higher in patients with SS (n = 17) than in HC (n = 13) or non-SS sicca patients (n = 19) and correlate with the xerostomia inventory, a scale for evaluating dry mouth [105].

2.6. Psoriatic Arthritis

Psoriatic arthritis (PsA) is an inflammatory arthropathy that affects some patients diagnosed with psoriasis, an immune-mediated disease of the skin and nails. It is the most common form of peripheral spondyloarthritis and it is associated with increased mortality from CVD. Obesity is an important comorbidity in PsA patients, and adiponectin may play a role in PsA, along with other pro- and anti-inflammatory mediators [106].

Serum

Results regarding circulating adiponectin levels in PsA are not consistent. PsA patients (n = 28) had significantly higher serum levels than HC (n = 39) in the study of Dikbas et al. [107]. Xue et al. proved the opposite, with PsA patients (n = 41) having lower adiponectin levels than HC (n = 24) and psoriasis patients (n = 20), and a negative correlation with osteoclast precursor numbers [108]. In another study PsA patients (n = 203) had higher adiponectin serum levels than patients with psoriasis without arthritis (n = 155) [109]. Finally, there were two studies in which no difference was found between PsA (n = 109 and n = 77, respectively) compared to HC (n = 32) [110] and osteoarthritis groups (n = 76) [111].

2.7. Antiphospholipid Syndrome

Antiphospholipid syndrome (APS) is a disorder, characterized by thrombosis, miscarriages and other pregnancy-related complications, in combination with persistent presence of antiphospholipid antibodies, among which are anti-β2GPI antibodies. Hematological, cutaneous, non-thrombotic cardiac and pulmonary, neurological, and renal manifestations may also be present [112].

Serum

In a single study, circulating adiponectin levels in primary APS patients (n = 56) were slightly but not significantly higher than in HC (n = 72). There was a positive correlation with anti-β2GPI IgG concentration [113].
In summary, circulating adiponectin levels are higher in RA and SLE, lower in SSc, while in AS patients the increase is not statistically significant. Due to a lack of studies and inconsistent results, the levels in SS, PsA and APS are still not well defined. Serum adiponectin in RA is positively associated with clinical manifestations, such as disease activity, radiographic severity and progression, poorer bone outcomes and reduced muscle mass. Negative associations are seen in SSc with disease activity and in AS with spinal radiographic disease progression. The role of adiponectin in CVD is not clear, there are studies that link adiponectin to increased mortality in CVD [2], and studies that report of its vasoprotective role [13]. A positive correlation of adiponectin with atherosclerotic plaques in SLE was found, which places SLE along studies reporting unfavorable adiponectin CVD effects. Unexpectedly, the association with CVD is not clear in RA. In general, adiponectin levels in the disease-affected tissues and cells reflect the levels in the bloodstream. Adiponectin is strongly expressed in RA synoviocytes and articular adipocytes, also levels in synovial fluid are associated with disease activity. The concentrations in urine strongly correlate with lupus nephritis in SLE, while in SS it is strongly expressed in the salivary gland, and normalized salivary levels correlate with dry mouth. In SSc, adiponectin expression is reduced not only in serum, but also in organs such as the skin, lung and gastroscopic sample tissue with less adiponectin present in the dcSSc subset with poorer clinical outcomes.

3. Could SARDs Treatment Change Serum Adiponectin?

Since SARDs are chronic diseases, patients need lifelong treatment which might affect adiponectin serum levels. We have investigated publications since 2010 collecting the effects of therapy on circulating adiponectin levels in SARDs patients. Most patients have received multidrug therapy, making the effects of a particular drug difficult to judge.

3.1. Anti-TNF

Anti-tumor necrosis factor (TNF) drugs were the first targeted biologicals approved for RA. Today, their use has spread to other chronic inflammatory diseases such as Crohn’s disease, ulcerative colitis, psoriasis, psoriatic arthritis, ankylosing spondylitis and juvenile RA [114].
As presented in Table 2, the majority of studies on RA, PsA and AS exhibit no change in adiponectin levels when anti-TNF therapy was used. However, three RA studies reported an increase in adiponectin. In the first study, the increase was presented only as the ratio of serum adiponectin to the body mass index [115]. The second study reported an increase at 3–12 months, but not during the 24-month period following treatment [116]. In the last report, an increase was only observed in patients who responded to treatment, but the measured adiponectin levels were 1000 times lower than in other reports [117].
Interestingly, the 3D-protein structure of the adiponectin C1q domain proved to be homologous to the TNF-α structure (Figure 5) in their identical folding topologies, key residue conservations and similarity of trimer interfaces. However, no homology was found in the primary amino acid sequence [128,129]. While anti-TNF therapy might increase adiponectin levels by inhibiting the inflammatory TNF-α signaling, the similar structure raises the question whether anti-TNF agents could bind to adiponectin and cause inactivation or reduction in circulation levels. Because serum levels after anti-TNF therapy remained unchanged, anti-TNF therapy did not seem to interfere with adiponectin detection. A study on AS patients treated with infliximab showed that adiponectin levels remained unchanged 120 min after therapy [127], suggesting that the clearance of adiponectin due to anti-TNF did not occur either. Also in vitro experiments confirmed that anti-TNF (adalimumab, etanercept) do not bind to adiponectin. However, they alter its signaling as shown in RA synovial fibroblasts (RASF), where adiponectin induction of IL-6 and matrix metalloproteinase-1, was reduced with application of anti-TNF. The preincubation of anti-TNF with adiponectin did not reduce this effect [59]. As shown later, RASF stimulated with adiponectin also did not increase TNF-α [130]. This suggests that observed anti-TNF effects inhibiting adiponectin proinflammatory signaling may be due to the binding of anti-TNF to transmembrane TNF-α, eliciting reverse transmembrane signaling.

3.2. Glucocorticoids

Glucocorticoids (GC) in SARDs are used as very effective tools for reducing disease activity and achieving clinical remission in the short term and reducing structural progression, disability and systemic manifestations in the medium term. They are widely used but have major drawbacks because of side effects [131,132]. Although they have been extensively studied in vitro, in animal models and in clinical trials, there is no consensus on the effects of GC on adiponectin expression [133]. In particular, GC is usually not used as a monotherapy, so its direct effect on adiponectin levels is difficult to discern. However, according to the studies, adiponectin serum levels generally tended to increase with GC treatment, but no changes were observed in the use of long-term, high-dose combination therapy. Elevated adiponectin levels after combined treatment with GC were associated with improved insulin resistance and endothelial function with a better lipid profile, and adiponectin may therefore play a role in cardiovascular protection (Table 3) [134,135].
There are two possible explanations for the association we see between GC and adiponectin levels, the first being the indirect GC affect through the decrease in inflammatory mediators. The second could be influenced by a GC-receptor binding site on the adiponectin gene promoter found in several cistromic studies in mice and humans [136]; however, this has not yet been well characterized or confirmed by other experiments.

3.3. Disease-Modifying Antirheumatic Drugs

Non-biological disease-modifying antirheumatic drugs (DMARDs) are a group of drugs that includes methotrexate (MTX), sulfasalazine (SSA), leflunomide, hydroxychloroquine (HCQ), azathioprine and some others. They are widely used in treatment of rheumatic diseases and are often prescribed in combination [138,139].
The increase in adiponectin after DMARD treatment was seen in most of the studies with RA patients. One report included patients (n = 27) treated with DMARDs in combination with prednisolone (the dose was reduced to 5 mg/d within one month) for 3 months [32]. Another study (n = 14) showed an increase in 24-month treatment with MTX to up to 20 mg/week with the addition of sulfasalazine 2000 mg/day and HCQ 400 mg daily [116]. In the third case, patients (n = 15) received MTX 0.2 mg/kg/week with prednisone 10 mg/day for 6 months [135]. In the fourth case, monotherapy or combination therapy with methotrexate, sulfasalazine and HCQ was used for 6 months (n = 40). Additionally, some patients received an anti-TNF agent (n = 16) [117]. In the latter case, adiponectin levels were significantly elevated in the third month of therapy with MTX 10–15 mg/week and prednisolone 7.5–15.0 mg/day (n = 65) [134]. Only one study (n = 46 early RA) reported a slight decrease in adiponectin levels after 6 months of DMARD treatment (MTX median dose 17.5 mg/week, sulfasalazine and leflunomide at standard doses) [36]. Additionally, one report reported no changes in circulating adiponectin levels after treatment with one or more of the following DMARDs: azathioprine, HCQ, leflunomide, MTX, SSA with prednisone at doses of 10 mg/day or less after 6 months (n = 127), 1 year (n = 91) and 2 years [137]. The great variety of different drugs in this group and their use in combination therapy make it difficult to investigate associations with clinical manifestations.

3.4. Tocilizumab

Tocilizumab (TCZ), a monoclonal antibody against the interleukin-6 receptor, in monotherapy or combination is recommended for treating moderate to severe RA, which had insufficient response to other DMARDs or to anti-TNF treatment [138].
In RA patients treated with TCZ, most studies report that adiponectin levels increased significantly after treatment as shown in Table 4. The only decrease was shown in one study where adiponectin was measured at 4 months, while in the other studies a time point of 6 months was used.
Studies also suggest TCZ may have protective role on CVD in RA patients, as beneficial changes in lipid profile are observed after treatment in most cases, but no correlation was found with adiponectin [27,121,140].

3.5. JAK Inhibitors

Janus kinases (JAKs) are enzymes transducing pro-inflammatory cytokine signals in cells, which contribute to an immune or inflammatory response in the cell. Their inhibition therefore leads to anti-inflammatory effects [142]. Baricitinib, a JAK 1/2 inhibitor, has been recommended for RA patients who do not respond to initial treatment with MTX or other conventional synthetic DMARDs [132]. In a recent study published on RA, adiponectin levels decreased significantly in patients treated with baricitinib monotherapy at 4 mg/day (n = 11) or in combination with other DMARDs (n = 4) for 6 months. The decrease in systemic inflammation was observed but not studied for adiponectin association [143].

3.6. Interleukin (IL)-1-Receptor Antagonists

Another important pro-inflammatory cytokine, IL-1, is targeted with biologic therapy. Patients treated daily with 100 mg of anakinra (n = 15), an interleukin-1 receptor antagonist, showed a significant improvement in type 2 diabetes-related metabolic parameters, but no changes in serum adiponectin concentrations over a treatment period of 6 months [124].

3.7. Anti-Interleukin-17A

IL-17 is an important pro-inflammatory cytokine produced by T helper 17 (Th17) cells upon stimulation with IL-23. In PsA patients, an increased number of polyfunctional circulating Th17 memory cells was found that produced IL-17 [144]. Adiponectin was shown to decrease the synthesis of IL-17 by acting on murine γδ-T-cells and human CD4+ and CD8+ T-cells, which lead to decreased inflammation in the skin [145]. Furthermore, lymphocytes from myelin-immunized adiponectin-deficient mice produced higher amounts of IL-17, which decreased after treatment with globular adiponectin [146]. The entanglement of adiponectin and the IL-17 signaling pathway led to the question of how inhibition of IL-17 could result in circulating adiponectin levels. Patients with PsA (n = 28) treated with secukinumab, a monoclonal antibody that binds to the protein IL-17A, with doses of 75 to 150 mg per month, showed no effect on serum adiponectin levels after the first, third and sixth month of therapy [147].

3.8. Cyclophosphamide

Cyclophosphamide (CY) metabolic intermediates are alkylating agents that cross-link DNA, causing cell death, the modulation of lymphocytes and the impairment of inflammatory responses [148]. CY’s main active metabolite, phosphoramide mustard, is produced in cells with low aldehyde dehydrogenases (ALDH) enzyme, thus enabling relatively cell-specific effects [149]. The expression of ALDH1A1 is abundant in adipose tissue and lungs, while expression is low in cultured fibroblasts, whole blood, heart, and skin [150], making them susceptible to CY effects [151], but leaving adipocytes, the main producers of adiponectin, unaffected.
CY is the most common treatment for SSc-ILD [152], based on a study that showed a significant improvement in forced vital capacity (FVC) in patients receiving cyclophosphamide compared to a placebo as a percentage of predicted FVC after a one-year follow-up [153]. Serum adiponectin levels in a group of SSc patients with active ILD (n = 8) increased more than five-fold after completion of three to six courses of intravenous pulse cyclophosphamide therapy and correlated significantly with a decrease in ILD [154].

3.9. n-3 Fatty Acids

Among other beneficial effects on cardiovascular health, n-3 fatty acids increase adiponectin levels by activating peroxisome-proliferator-activated receptor gamma (PPAR-γ) [155]. CVD is one of the major causes of death in SLE [156]. Two studies have been published on the effects of dietary supplements containing n-3 fatty acids on adiponectin levels in SLE, both using the same dose, 1800 mg eicosapentaenoic acid and 1200 mg docosahexaenoic acid daily. The 12-week study (SLE n = 22, HC n = 27) found no changes in adiponectin levels [157], while the 17-week study (SLE n = 41, HC n = 21) reported a significant increase and improvement in the disease activity score but did not include an adiponectin correlation [158].
To summarize, while GC, TCZ, CY and DMARD therapies are generally associated with the upregulation of adiponectin levels, anti-TNF therapy does not appear to cause changes in serum adiponectin levels, although these drugs successfully down-regulate inflammation. This may be partially due to the crosstalk and similarities in the structure of adiponectin and TNF-α. However, no binding of anti-TNF to adiponectin has been observed, so possible underlying mechanisms that could be related to signalization pathways will need to be investigated.

4. Divergent Changes in the Activation or Suppression of Adiponectin Gene Regulation by Transcription Factors Can Affect Its Serum Levels

Adiponectin promoters consist of nuclear receptor sites (PPARG, LRH, RXR), transcription factor sites that enhance adiponectin gene expression (C/EBPα, SREBP1c, TFAP2B, FOXO1, SP1) and co-regulators of transcription factors (SIRT1, NCOR1, NCOR2), among others (Figure 6) [159].

4.1. PPAR-γ

Adiponectin expression is best characterized by PPAR-γ regulator, a ligand activated by binding to unsaturated fatty acids and eicosanoids such as 15-deoxy-Δ12,14-prostaglandin J2, TZD, and cannabinoids. PPAR-γ heteromerizes with the retionid X-receptor and activates adiponectin expression. Contrary to expectations, in SARDs with high adiponectin serum levels, such as RA, low levels of PPAR-γ expression or activity is present and also beneficial effects of PPAR-γ activation have been demonstrated [160,161]. This indicates that adiponectin serum levels are elevated in these disorders for reasons other than PPAR-γ activation. In SLE, elevated PPAR-γ levels have been shown [162], but the beneficial effects of PPAR-γ activation have also been demonstrated in cell experiments and animal models [163,164]. The PPAR-γ activators TZDs (pioglitazone and rosiglitazone) are approved by the FDA for type 2 diabetes [165], but are thought to exert beneficial effects on SARDs with altered PPAR-γ activity [160]. Clinical trials with TZDs have been conducted in RA and in SLE patients, with the focus on inflammation control, cardiovascular protection and skeletal muscle dysfunction [165]. In vitro and in vivo studies have shown positive effects of PPAR-γ activators also in SSc skin and lung fibrosis, as well as in many other fibrotic disorders [166]. However, the clinical study with IVA337—Lanifibranor, a pan-PPAR agonist, has failed to demonstrate a significant improvement in SSc treatment [165]. This means that other factors (PPAR and non-PPAR related) that contribute to the pathology of these diseases also play an important role in regulating adiponectin levels. One of these factors could be the PPAR-γ nuclear corepressor NCoR, which is aberrantly activated in SSc skin [167].

4.2. Id3

Id3 is a repressor of adiponectin expression that acts on SREBP, an adiponectin transcription promoter in adipocytes [15,168]. It has been found upregulated in lung tissue and fibroblasts of SSc and idiopathic pulmonary fibrosis, thereby maintaining fibroblasts in a dedifferentiated, hyperproliferative and apoptosis-resistant state [169]. Id3-deficient mice develop an autoimmune disease similar to human SS [170], but Id3 has been shown to be elevated in RA synovium [171] and correlates with the SLEDAI in SLE [172].

4.3. ATF3

Another repressor of adiponectin gene expression, activating transcription factor 3 (ATF3), suppresses the expression of inflammatory cytokines/chemokines in immune cells after various stimuli [173]. ATF3 was elevated in skin/fibroblasts of SSc, in association with the profibrotic signal transduction of TGF-β [166].

4.4. SIRT1, FoXO1 and C/EBPα

Experiments suggest that C/EBPα is required to fully activate adiponectin gene expression, although the physiological significance is not clear. FoXO1 binds to C/EBPα and its activity is regulated by insulin, IGF-1 and SIRT1. Adiponectin gene expression induced by SIRT1 is likely to occur through FoxO1 deacetylation [15]. Overexpression of SIRT1 is found in synovial RA tissue [174] and in LN [175], but SIRT1 has been shown to be reduced in peripheral blood mononuclear cells of SSc patients with pulmonary fibrosis and in lung tissues of bleomycin-induced lung fibrosis mice [176]. This could be an additional explanation for diverging adiponectin levels in SARDs.

5. How Inflammatory Cytokines IL-6 and TNF-α Regulate Adiponectin Levels in SARDs

Inflammatory cytokines, IL-6 and TNF-α, downregulate adiponectin levels [177,178,179], which was demonstrated at the mRNA and protein level in 3T3-L1 adipocytes and in a mouse model. Adiponectin gene expression was dose- (0.5–100 ng/mL) and time-dependent (0–24 h) on IL-6 stimulation [177,178,179]. Paradoxically, in some SARDs, such as RA and SLE, adiponectin levels remain elevated [4,6,88] despite persistent inflammation and higher concentrations of IL-6 and TNF-α [4,6,88,138,180,181]. Of note, the IL-6–TNF-α-adiponectin relation has been also observed in the opposite direction: adiponectin reduces the release of IL-6 and TNF-α in adipocytes and stromal-vascular cells [11].
Both TNF-α and IL-6 inhibit adiponectin synthesis by downregulating PPAR-γ activity. TNF-α activates JNK that inhibits PPAR-γ DNA binding trough phosphorylation of PPAR-γ, and shares a pathway with IL-6 through the STAT-SIRT1-FoxO1 pathway. This inhibits occupancy of peroxisome proliferator response element (PPRE) trough interaction of FoxO1 and PPAR-γ [182,183]. IL-6, on the other hand, also inhibits adiponectin expression through the Erk1/2—NFAT pathway, which is PPAR-γ unrelated (Figure 6) [15]. Differences in PPAR-γ activation and the non-PPAR-γ dependent regulation of adiponectin expression could explain why we observe a serum adiponectin increase after anti-IL-6 treatment, but no change after an anti-TNF treatment in SARDs.
Adiponectin in certain SARDs is produced abundantly in cells other than adipocytes, such as synoviocytes in RA [59]. Some of these cell types have insignificant PPAR-γ expression, which could be the reason that IL-6 and TNF-α, through suppression of PPAR-γ activity, cannot sufficiently decrease serum adiponectin levels. PPAR-γ is the product of one gene, but has been reported to have 2 splice variants. While PPAR-γ1 is present in macrophages, colonic epithelium, endothelium and vascular smooth muscle cells, PPAR-γ2 is predominantly present in adipose tissue [160]. PPAR-γ expression is increased in adipogenesis and in monocytes, when transdifferentiating into macrophages [184]. Interestingly, RNA expression is very low in normal, unstimulated circulating blood cells [151], although the PPAR-γ gene was originally cloned from bone marrow cDNA library [185]. Activation of PPAR-γ promotes both M2 polarization in macrophages and Th2 polarization in T cells [186] and in RA, which lacks PPAR-γ activity. M1 and Th1 are the predominant cell types.
In summary, first, the lack of PPAR-γ activity in RA and SLE was proven in vitro and in vivo models, minimizing the ability of TNF-α and IL-6 to downregulate adiponectin levels via this pathway. Second, TNF-α- and IL-6-related adiponectin downregulation was mainly characterized in cells expressing high levels of PPAR-γ, and could be efficient in obesity, for example. However, this mechanism is not as efficient in RA or SLE, where serum adiponectin levels depend on synthesis in other cells with intrinsically low PPAR-γ and thus cannot be further downregulated via this pathway. Altogether, this points to the reason that TNF-α and IL-6 do not inhibit serum adiponectin levels in RA and SLE patients.

6. Conclusions

The deregulation of adiponectin in SARDs was seen in both upregulated and downregulated mechanisms. We concluded that diseases with less prominent inflammation (SSc), where adipocytes have been shown to be reduced at the site of disease pathology, have decreased adiponectin levels and a negative correlation with disease activity. On the other hand, highly inflammatory diseases (RA, SLE) show increased adiponectin levels and a positive correlation with clinical manifestations. Treatment used in SARDs is associated with either decreased adiponectin levels, as with JAK inhibitors, or increased levels, as in cases where GC, CY, TCZ and DMARDs are used (Figure 7). The paradox of why increased adiponectin levels are present in RA and SLE despite high levels of proinflammatory cytokines IL-6 and TNF-α could be explained at the level of adiponectin gene regulation. In general, IL-6 and TNF-α inhibit adiponectin via the suppression of PPAR-γ, which is highly expressed in adipocytes. However, in some SARDs, adiponectin is also abundantly produced by other cells that lack PPAR-γ activity, leaving adiponectin production intact. Differences in PPAR-γ activation, as well as other non-PPAR-γ-dependent signaling pathways regulating adiponectin expression, may provide an answer to why adiponectin levels are associated with anti-IL-6 treatment, but not with anti-TNF therapy. However, the molecular mechanisms need further investigation.
In SARDs, where elevated adiponectin levels are associated with deleterious manifestations, blocking adiponectin with antagonists (ADP400) [179] and adiponectin-targeting agents (monoclonal antibodies KH7-33, KH4-8) is suggested. In contrast, various AdipoR agonists (AdipoRon, ADP355) and PPAR-γ activators (thiazolinediones) could be useful in SARDs that lack adiponectin. In general, it seems very important to ensure normal or optimal levels of adiponectin and its activity, as any deregulation is associated with many pathologies.

Author Contributions

K.L. and N.B. conceptualized the paper and wrote the first edition of the manuscript. N.B. reviewed the literature. K.P.-P., S.S.-Š., S.Č., and J.V. contributed their expertise to the manuscript and read, edited, and approved the submitted version. All authors have read and agreed to the published version of the manuscript.

Funding

The study was funded by the Slovenian Research Agency ARRS, with the post-doctoral project Z3-9261, project J7-8276 and the National Research Program #P3-0314.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The gene expression figure was obtained from the GTEx Portal, Analysis Release V8 on 05/11/20. Figures were created with BioRender.com. Molecular graphics figure was produced using the UCSF Chimera package from the Computer Graphics Laboratory, University of California, San Francisco (supported by NIH P41 RR-01081).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ALDHAldehyde dehydrogenases
AMPKAMP-activated protein kinase
AMTAdipocyte mesenchymal transition
APSAntiphospholipid syndrome
ASAnkylosing spondylitis
ATF3Activating transcription factor 3
ATVAtorvastatin
CVDCardiovascular disease
CYCyclophosphamide
DAS28Disease Activity Score of 28 joints
dcSScDiffuse cutaneous SSc
DMARDDisease-modifying antirheumatic drugs
dWATDermal white adipose tissue
ESRErythrocyte sedimentation rate
FVCForced vital capacity
GCGlucocorticoids
HCHealthy controls
HCQHydroxychloroquine
HMWHigh molecular weight
ILInterleukin
ILDInterstitial lung disease
JAKJanus kinase
lcSScLimited cutaneous SSc
LMWLow molecular weight
LNLupus nephritis
MHAQMultidimensional Health Assessment Questionnaire
MMWMedium molecular weight
mRSSModified Rodnan skin score
MTXMethotrexate
PBMCsPeripheral blood mononuclear cells
PPAR-γPeroxisome-proliferator-activated receptor gamma
PPAR-αPeroxisome-proliferator-activated receptor alpha
PPREPeroxisome proliferator response element
PsAPsoriatic arthritis
RARheumatoid arthritis
RAPID3Routine Assessment of Patient Index Data 3
RASF RA synovial fibroblasts
SARDsSystemic autoimmune rheumatic diseases
SGECSalivary glandular epithelial cells
SHSSharp-van der Heijde Score
SLESystemic lupus erythematosus
SLEDAISystemic Lupus Erythematosus Disease Activity Index
SMDStandard mean difference
SSSjögren’s syndrome
SSASulfasalazine
SScSystemic sclerosis
TCZTocilizumab
Th17T helper 17
TNF-αTumor necrosis factor α
TZDThiazolidinediones

References

  1. Choi, H.M.; Doss, H.M.; Kim, K.S. Multifaceted physiological roles of adiponectin in inflammation and diseases. Int. J. Mol. Sci. 2020, 21, 1219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Menzaghi, C.; Trischitta, V. Erratum. The adiponectin paradox for all-cause and cardiovascular mortality. Diabetes 2018, 67, 12–22. [Google Scholar] [CrossRef] [Green Version]
  3. Zhang, M.Y.; Dini, A.A.; Yang, X.K.; Li, L.J.; Wu, G.C.; Leng, R.X.; Pan, H.F.; Ye, D.Q. Association between serum/plasma adiponectin levels and immune-mediated diseases: A meta-analysis. Arch. Derm. Res. 2017, 309, 625–635. [Google Scholar] [CrossRef] [PubMed]
  4. Dini, A.A.; Wang, P.; Ye, D.-Q. Serum adiponectin levels in patients with systemic lupus erythematosus: A Meta-analysis. J. Clin. Rheumatol. 2017, 23, 361–367. [Google Scholar] [CrossRef] [PubMed]
  5. Zhao, J.-H.; Huang, X.-L.; Duan, Y.; Wang, Y.-J.; Chen, S.-Y.; Wang, J. Serum adipokines levels in patients with systemic sclerosis: A meta-analysis. Mod. Rheumatol. 2017, 27, 298–305. [Google Scholar] [CrossRef]
  6. Yang, J.; Zhang, X.; Ma, Y.; Wu, M.; Hu, X.; Han, R.; Yuan, Y.; Wang, M.; Chen, M.; Jiang, S.; et al. Serum levels of leptin, adiponectin and resistin in patients with ankylosing spondylitis: A systematic review and meta-analysis. Int. Immunopharmacol. 2017, 52, 310–317. [Google Scholar] [CrossRef]
  7. Lee, Y.H.; Bae, S.C. Circulating adiponectin and visfatin levels in rheumatoid arthritis and their correlation with disease activity: A meta-analysis. Int. J. Rheum. Dis. 2018, 21, 664–672. [Google Scholar] [CrossRef]
  8. Toussirot, E.; Binda, D.; Gueugnon, C.; Dumoulin, G. Adiponectin in autoimmune diseases. Curr. Med. Chem. 2012, 19, 5474–5480. [Google Scholar] [CrossRef] [PubMed]
  9. Krysiak, R.; Handzlik-Orlik, G.; Okopien, B. The role of adipokines in connective tissue diseases. Eur. J. Nutr. 2012, 51, 513–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Scotece, M.; Conde, J.; Lopez, V.; Gomez, R.; Lago, F.; Gomez Reino, J.J.; Gualillo, O. Adipokines and systemic rheumatic diseases: Linking inflammation, immunity and metabolism. In Insights and Perspectives in Rheumatology; Harrison, A., Ed.; InTech: London, UK, 2012; pp. 21–38. [Google Scholar]
  11. Fang, H.; Judd, R.L. Adiponectin regulation and function. Compr. Physiol. 2018, 8, 1031–1063. [Google Scholar]
  12. Ye, J.; Bian, X.; Lim, J.; Medzhitov, R. Adiponectin and related C1q/TNF-related proteins bind selectively to anionic phospholipids and sphingolipids. Proc. Natl. Acad. Sci. USA 2020, 117, 17381–17388. [Google Scholar] [CrossRef]
  13. Achari, A.E.; Jain, S.K. Adiponectin, a therapeutic target for obesity, diabetes, and endothelial dysfunction. Int. J. Mol. Sci. 2017, 18, 1321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Azamar-Llamas, D.; Hernandez-Molina, G.; Ramos-Avalos, B.; Furuzawa-Carballeda, J. Adipokine contribution to the pathogenesis of osteoarthritis. Mediat. Inflamm. 2017, 2017, 5468023. [Google Scholar] [CrossRef] [Green Version]
  15. Liu, M.; Liu, F. Transcriptional and post-translational regulation of adiponectin. Biochem. J. 2010, 425, 41–52. [Google Scholar] [CrossRef]
  16. Blaak, E. Gender differences in fat metabolism. Curr. Opin. Clin. Nutr. Metab. Care 2001, 4, 499–502. [Google Scholar] [CrossRef] [Green Version]
  17. Furukawa, S.; Fujita, T.; Shimabukuro, M.; Iwaki, M.; Yamada, Y.; Nakajima, Y.; Nakayama, O.; Makishima, M.; Matsuda, M.; Shimomura, I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J. Clin. Investig. 2004, 114, 1752–1761. [Google Scholar] [CrossRef]
  18. Burhans, M.S.; Hagman, D.K.; Kuzma, J.N.; Schmidt, K.A.; Kratz, M. Contribution of adipose tissue inflammation to the development of type 2 diabetes mellitus. Compr. Physiol. 2018, 9, 1–58. [Google Scholar]
  19. Katira, A.; Tan, P.H. Evolving role of adiponectin in cancer-controversies and update. Cancer Biol. Med. 2016, 13, 101–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Khalil, R.B.; El Hachem, C. Adiponectin in eating disorders. Eat. Weight Disord. 2014, 19, 3–10. [Google Scholar] [CrossRef]
  21. Marangoni, R.G.; Masui, Y.; Fang, F.; Korman, B.; Lord, G.; Lee, J.; Lakota, K.; Wei, J.; Scherer, P.E.; Otvos, L.; et al. Adiponectin is an endogenous anti-fibrotic mediator and therapeutic target. Sci. Rep. 2017, 7, 4397. [Google Scholar] [CrossRef] [PubMed]
  22. Marangoni, R.G.; Lu, T.T. The roles of dermal white adipose tissue loss in scleroderma skin fibrosis. Curr. Opin. Rheumatol. 2017, 29, 585–590. [Google Scholar] [CrossRef]
  23. Goldblatt, F.; O’Neill, S.G. Clinical aspects of autoimmune rheumatic diseases. Lancet 2013, 382, 797–808. [Google Scholar] [CrossRef]
  24. MᵃᶜDonald, I.; Liu, S.-C.; Huang, C.-C.; Kuo, S.-J.; Tsai, C.-H.; Tang, C.-H. Associations between adipokines in arthritic disease and implications for obesity. Int. J. Mol. Sci. 2019, 20, 1505. [Google Scholar] [CrossRef] [Green Version]
  25. Versini, M.; Jeandel, P.Y.; Rosenthal, E.; Shoenfeld, Y. Obesity in autoimmune diseases: Not a passive bystander. Autoimmun. Rev. 2014, 13, 981–1000. [Google Scholar] [CrossRef]
  26. Khajoei, S.; Hassaninevisi, M.; Kianmehr, N.; Seif, F.; Khoshmirsafa, M.; Shekarabi, M.; Samei, A.; Haghighi, A. Serum levels of adiponectin and vitamin D correlate with activity of Rheumatoid Arthritis. Mol. Biol. Rep. 2019, 46, 2505–2512. [Google Scholar] [CrossRef]
  27. Hoffman, E.; Rahat, M.A.; Feld, J.; Elias, M.; Rosner, I.; Kaly, L.; Lavie, I.; Gazitt, T.; Zisman, D. Effects of tocilizumab, an anti-interleukin-6 receptor antibody, on serum lipid and adipokine levels in patients with rheumatoid arthritis. Int. J. Mol. Sci. 2019, 20, 4633. [Google Scholar] [CrossRef] [Green Version]
  28. Qian, J.; Xu, L.; Sun, X.; Wang, Y.; Xuan, W.; Zhang, Q.; Zhao, P.; Wu, Q.; Liu, R.; Che, N.; et al. Adiponectin aggravates bone erosion by promoting osteopontin production in synovial tissue of rheumatoid arthritis. Arthritis Res. Ther. 2018, 20, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Chaparro-Sanabria, J.A.; Bautista-Molano, W.; Bello-Gualtero, J.M.; Chila-Moreno, L.; Castillo, D.M.; Valle-Oñate, R.; Chalem, P.; Romero-Sánchez, C. Association of adipokines with rheumatic disease activity indexes and periodontal disease in patients with early rheumatoid arthritis and their first-degree relatives. Int. J. Rheum. Dis. 2019, 22, 1990–2000. [Google Scholar] [CrossRef] [PubMed]
  30. DeClercq, V.; Cui, Y.; Forbes, C.; Grandy, S.A.; Keats, M.; Parker, L.; Sweeney, E.; Yu, Z.M.; Dummer, T.J.B. Adiposity measures and plasma adipokines in females with rheumatoid and osteoarthritis. Mediat. Inflamm. 2017, 2017, 4302412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Zhao, Y.-L.; Zhang, T.-P.; Wu, J.; Li, B.-Z.; Li, X.-M.; Pan, H.-F.; Ye, D.-Q. Association of adiponectin and adiponectin receptor gene polymorphisms with rheumatoid arthritis in a Chinese population. Postgrad. Med. J. 2020, 96, 149–155. [Google Scholar] [CrossRef] [PubMed]
  32. Cansu, B.; Cansu, D.U.; Kaşifoğlu, T.; Gülbas, Z.; Korkmaz, C. Disease-modifying antirheumatic drugs increase serum adiponectin levels in patients with rheumatoid arthritis. J. Clin. Rheumatol. 2011, 17, 14–17. [Google Scholar] [CrossRef]
  33. Kontny, E.; Zielińska, A.; Skalska, U.; Księżopolska-Orłowska, K.; Głuszko, P.; Maśliński, W. Distinct secretory activity and clinical impact of subcutaneous abdominal adipose tissue in women with rheumatoid arthritis and osteoarthritis. Inflammation 2017, 40, 106–116. [Google Scholar] [CrossRef] [PubMed]
  34. Kontny, E.; Zielińska, A.; Księżopolska-Orłowska, K.; Głuszko, P. Secretory activity of subcutaneous abdominal adipose tissue in male patients with rheumatoid arthritis and osteoarthritis—Association with clinical and laboratory data. Reumatol. /Rheumatol. 2016, 5, 227–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Meyer, M.; Sellam, J.; Fellahi, S.; Kotti, S.; Bastard, J.-P.; Meyer, O.; Lioté, F.; Simon, T.; Capeau, J.; Berenbaum, F. Serum level of adiponectin is a surrogate independent biomarker of radiographic disease progression in early rheumatoid arthritis: Results from the ESPOIR cohort. Arthritis Res. Ther. 2013, 15, R210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Manrique-Arija, S.; Ureña, I.; Valdivielso, P.; Rioja, J.; Jiménez-Núñez, F.G.; Irigoyen, M.V.; Fernández-Nebro, A. Insulin resistance and levels of adipokines in patients with untreated early rheumatoid arthritis. Clin. Rheumatol. 2016, 35, 43–53. [Google Scholar] [CrossRef] [Green Version]
  37. Erlandsson, M.C.; Doria Medina, R.; Töyrä Silfverswärd, S.; Bokarewa, M.I. Smoking Functions as a negative regulator of IGF1 and impairs adipokine network in patients with rheumatoid arthritis. Mediat. Inflamm. 2016, 2016, 3082820. [Google Scholar] [CrossRef] [Green Version]
  38. Minamino, H.; Katsushima, M.; Yoshida, T.; Hashimoto, M.; Fujita, Y.; Shirakashi, M.; Yamamoto, W.; Murakami, K.; Murata, K.; Nishitani, K.; et al. Increased circulating adiponectin is an independent disease activity marker in patients with rheumatoid arthritis: A cross-sectional study using the KURAMA database. PLoS ONE 2020, 15, e0229998. [Google Scholar] [CrossRef]
  39. Oranskiy, S.P.; Yeliseyeva, L.N.; Tsanaeva, A.V.; Zaytseva, N.V. Body composition and serum levels of adiponectin, vascular endothelial growth factor, and interleukin-6 in patients with rheumatoid arthritis. Croat. Med. J. 2012, 53, 350–356. [Google Scholar] [CrossRef] [Green Version]
  40. Guimarães, M.F.B.d.R.; de Andrade, M.V.M.; Machado, C.J.; Vieira, É.L.M.; Pinto, M.R.d.C.; Júnior, A.L.T.; Kakehasi, A.M. Leptin as an obesity marker in rheumatoid arthritis. Rheumatol. Int. 2018, 38, 1671–1677. [Google Scholar] [CrossRef]
  41. Zhang, Y.; Peltonen, M.; Andersson-Assarsson, J.; Svensson, P.A.; Herder, C.; Rudin, A.; Carlsson, L.; Maglio, C. Elevated adiponectin predicts the development of rheumatoid arthritis in subjects with obesity. Scand. J. Rheumatol. 2020, 49. [Google Scholar] [CrossRef]
  42. Ozgen, M.; Koca, S.S.; Dagli, N.; Balin, M.; Ustundag, B.; Isik, A. Serum adiponectin and vaspin levels in rheumatoid arthritis. Arch. Med. Res. 2010, 41, 457–463. [Google Scholar] [CrossRef] [PubMed]
  43. Bustos Rivera-Bahena, C.; Xibillé-Friedmann, D.-X.; González-Christen, J.; Carrillo-Vázquez, S.M.; Montiel-Hernández, J.L. Peripheral blood leptin and resistin levels as clinical activity biomarkers in Mexican Rheumatoid Arthritis patients. Reumatol. Clínica 2016, 12, 323–326. [Google Scholar] [CrossRef] [PubMed]
  44. Li, B.T.; Zhang, F.Z.; Xu, T.S.; Ding, R.; Li, P. Increasing production of matrix metalloproteinases, tumor necrosis factor-α, vascular endothelial growth factor and prostaglandin E2 in rheumatoid arthritis synovial fibroblasts by different adiponectin isoforms in a concentration-dependent manner. Cell. Mol. Biol. 2015, 61, 27–32. [Google Scholar] [PubMed]
  45. El-Hini, S.H.; Mohamed, F.I.; Hassan, A.A.; Ali, F.; Mahmoud, A.; Ibraheem, H.M. Visfatin and adiponectin as novel markers for evaluation of metabolic disturbance in recently diagnosed rheumatoid arthritis patients. Rheumatol. Int. 2013, 33, 2283–2289. [Google Scholar] [CrossRef] [PubMed]
  46. Alkady, E.A.M.; Ahmed, H.M.; Tag, L.; Abdou, M.A. Adiponectin, resistin und visfatin in serum und gelenkflüssigkeit bei patienten mit rheumatoider arthritis: Zusammenhang mit der krankheitsaktivität. Z. Für Rheumatol. 2011, 70, 602–608. [Google Scholar] [CrossRef]
  47. Targońska-Stępniak, B.; Dryglewska, M.; Majdan, M. Adiponectin and leptin serum concentrations in patients with rheumatoid arthritis. Rheumatol. Int. 2010, 30, 731–737. [Google Scholar] [CrossRef] [Green Version]
  48. Klein-Wieringa, I.R.; Andersen, S.N.; Herb-van Toorn, L.; Kwekkeboom, J.C.; van der Helm-van Mil, A.H.M.; Meulenbelt, I.; Huizinga, T.W.J.; Kloppenburg, M.; Toes, R.E.M.; Ioan-Facsinay, A. Are baseline high molecular weight adiponectin levels associated with radiographic progression in rheumatoid arthritis and osteoarthritis? J. Rheumatol. 2014, 41, 853–857. [Google Scholar] [CrossRef]
  49. Park, Y.-J.; Cho, C.-S.; Emery, P.; Kim, W.-U. LDL Cholesterolemia as a novel risk factor for radiographic progression of rheumatoid arthritis: A single-center prospective study. PLoS ONE 2013, 8, e68975. [Google Scholar] [CrossRef] [Green Version]
  50. Klein-Wieringa, I.R.; van der Linden, M.P.M.; Knevel, R.; Kwekkeboom, J.C.; van Beelen, E.; Huizinga, T.W.J.; van der Helm-van Mil, A.; Kloppenburg, M.; Toes, R.E.M.; Ioan-Facsinay, A. Baseline serum adipokine levels predict radiographic progression in early rheumatoid arthritis. Arthritis Rheum. 2011, 63, 2567–2574. [Google Scholar] [CrossRef]
  51. Giles, J.T.; van der Heijde, D.M.; Bathon, J.M. Association of circulating adiponectin levels with progression of radiographic joint destruction in rheumatoid arthritis. Ann. Rheum. Dis. 2011, 70, 1562–1568. [Google Scholar] [CrossRef] [Green Version]
  52. Burgmaier, M.; Hoppe, S.; Krüger, T.; Mahnken, A.H.; Ketteler, M.; Reith, S.; Mühlenbruch, G.; Marx, N.; Brandenburg, V. Serum levels of C-peptide are associated with coronary artery calcification in patients with rheumatoid arthritis. Rheumatol. Int. 2015, 35, 1541–1547. [Google Scholar] [CrossRef] [PubMed]
  53. Dessein, P.H.; Tsang, L.; Solomon, A.; Woodiwiss, A.J.; Millen, A.M.E.; Norton, G.R. Adiponectin and atherosclerosis in rheumatoid arthritis. Mediat. Inflamm. 2014, 2014, 358949. [Google Scholar] [CrossRef] [Green Version]
  54. Kang, Y.; Park, H.-J.; Kang, M.-I.; Lee, H.-S.; Lee, S.-W.; Lee, S.-K.; Park, Y.-B. Adipokines, inflammation, insulin resistance, and carotid atherosclerosis in patients with rheumatoid arthritis. Arthritis Res. Ther. 2013, 15, R194. [Google Scholar] [CrossRef] [Green Version]
  55. Dessein, P.H.; Woodiwiss, A.J.; Norton, G.R.; Tsang, L.; Solomon, A. Independent associations of total and high molecular weight adiponectin with cardiometabolic risk and surrogate markers of enhanced early atherogenesis in black and white patients with rheumatoid arthritis: A cross-sectional study. Arthritis Res. Ther. 2013, 15, R128. [Google Scholar] [CrossRef] [Green Version]
  56. Baker, J.F.; Long, J.; Mostoufi-Moab, S.; Denburg, M.; Jorgenson, E.; Sharma, P.; Zemel, B.S.; Taratuta, E.; Ibrahim, S.; Leonard, M.B. Muscle deficits in rheumatoid arthritis contribute to inferior cortical bone structure and trabecular bone mineral density. J. Rheumatol. 2017, 44, 1777–1785. [Google Scholar] [CrossRef]
  57. Baker, J.F.; Von Feldt, J.M.; Mostoufi-Moab, S.; Kim, W.; Taratuta, E.; Leonard, M.B. Insulin-like growth factor 1 and adiponectin and associations with muscle deficits, disease characteristics, and treatments in rheumatoid arthritis. J. Rheumatol. 2015, 42, 2038–2045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Chedid, P.; Hurtado-Nedelec, M.; Marion-Gaber, B.; Bournier, O.; Hayem, G.; Gougerot-Pocidalo, M.-A.; Frystyk, J.; Flyvbjerg, A.; El Benna, J.; Marie, J.-C. Adiponectin and its globular fragment differentially modulate the oxidative burst of primary human phagocytes. Am. J. Pathol. 2012, 180, 682–692. [Google Scholar] [CrossRef] [PubMed]
  59. Ehling, A.; Schaffler, A.; Herfarth, H.; Tarner, I.H.; Anders, S.; Distler, O.; Paul, G.; Distler, J.; Gay, S.; Scholmerich, J.; et al. The potential of adiponectin in driving arthritis. J. Immunol. 2006, 176, 4468–4478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Kontny, E.; Plebanczyk, M.; Lisowska, B.; Olszewska, M.; Maldyk, P.; Maslinski, W. Comparison of rheumatoid articular adipose and synovial tissue reactivity to proinflammatory stimuli: Contribution to adipocytokine network. Ann. Rheum. Dis. 2012, 71, 262–267. [Google Scholar] [CrossRef]
  61. Cojocaru, M.; Cojocaru, I.M.; Silosi, I.; Vrabie, C.D. Manifestations of systemic lupus erythematosus. Maedica 2011, 6, 330–336. [Google Scholar]
  62. Anders, H.-J.; Saxena, R.; Zhao, M.-h.; Parodis, I.; Salmon, J.E.; Mohan, C. Lupus nephritis. Nat. Rev. Dis. Primers 2020, 6, 7. [Google Scholar] [CrossRef]
  63. Yang, H.; Liu, H.; Zhou, Z.; Zhao, L.; Fei, Y.; Chen, H.; Zhang, F.; Zhang, X. Management of severe refractory systemic lupus erythematosus: Real-world experience and literature review. Clin. Rev. Allergy Immunol. 2020, 60, 17–30. [Google Scholar] [CrossRef]
  64. Kuo, C.Y.; Tsai, T.Y.; Huang, Y.C. Insulin resistance and serum levels of adipokines in patients with systemic lupus erythematosus: A systematic review and meta-analysis. Lupus 2020, 29, 1078–1084. [Google Scholar] [CrossRef]
  65. Diaz-Rizo, V.; Bonilla-Lara, D.; Gonzalez-Lopez, L.; Sanchez-Mosco, D.; Fajardo-Robledo, N.S.; Perez-Guerrero, E.E.; Rodriguez-Jimenez, N.A.; Saldana-Cruz, A.M.; Vazquez-Villegas, M.L.; Gomez-Banuelos, E.; et al. Serum levels of adiponectin and leptin as biomarkers of proteinuria in lupus nephritis. PLoS ONE 2017, 12, e0184056. [Google Scholar] [CrossRef] [Green Version]
  66. Hutcheson, J.; Ye, Y.; Han, J.; Arriens, C.; Saxena, R.; Li, Q.Z.; Mohan, C.; Wu, T. Resistin as a potential marker of renal disease in lupus nephritis. Clin. Exp. Immunol. 2015, 179, 435–443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Santos, F.M.; Telles, R.W.; Lanna, C.C.; Teixeira, A.L., Jr.; Miranda, A.S.; Rocha, N.P.; Ribeiro, A.L. Adipokines, tumor necrosis factor and its receptors in female patients with systemic lupus erythematosus. Lupus 2017, 26, 10–16. [Google Scholar] [CrossRef] [PubMed]
  68. Carbone, F.; Montecucco, F.; Poggi, A.; Nobili, F.; Cacciapaglia, F.; Afeltra, A.; Moccetti, T.; Colombo, B.M. Serum adiponectin levels are associated with presence of carotid plaque in women with systemic lupus erythematosus. Nutr. Metab. Cardiovasc. Dis. 2020, 30, 1147–1151. [Google Scholar] [CrossRef]
  69. Gronwall, C.; Reynolds, H.; Kim, J.K.; Buyon, J.; Goldberg, J.D.; Clancy, R.M.; Silverman, G.J. Relation of carotid plaque with natural IgM antibodies in patients with systemic lupus erythematosus. Clin. Immunol. 2014, 153, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Reynolds, H.R.; Buyon, J.; Kim, M.; Rivera, T.L.; Izmirly, P.; Tunick, P.; Clancy, R.M. Association of plasma soluble E-selectin and adiponectin with carotid plaque in patients with systemic lupus erythematosus. Atherosclerosis 2010, 210, 569–574. [Google Scholar] [CrossRef] [Green Version]
  71. McMahon, M.; Skaggs, B.J.; Sahakian, L.; Grossman, J.; FitzGerald, J.; Ragavendra, N.; Charles-Schoeman, C.; Chernishof, M.; Gorn, A.; Witztum, J.L.; et al. High plasma leptin levels confer increased risk of atherosclerosis in women with systemic lupus erythematosus, and are associated with inflammatory oxidised lipids. Ann. Rheum. Dis. 2011, 70, 1619–1624. [Google Scholar] [CrossRef]
  72. McMahon, M.; Skaggs, B.J.; Grossman, J.M.; Sahakian, L.; Fitzgerald, J.; Wong, W.K.; Lourenco, E.V.; Ragavendra, N.; Charles-Schoeman, C.; Gorn, A.; et al. A panel of biomarkers is associated with increased risk of the presence and progression of atherosclerosis in women with systemic lupus erythematosus. Arthritis Rheumatol. 2014, 66, 130–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Hrycek, E.; Banasiewicz-Szkróbka, I.; Żurakowski, A.; Dworak, J.; Błaszczak, E.; Franek, A.; Buszman, P. Selected adipokines and thickness of the intima-media complex in patients with systemic lupus erythematosus. Kardiol. Pol. 2018, 76, 917–919. [Google Scholar] [CrossRef] [Green Version]
  74. Vadacca, M.; Zardi, E.M.; Margiotta, D.; Rigon, A.; Cacciapaglia, F.; Arcarese, L.; Buzzulini, F.; Amoroso, A.; Afeltra, A. Leptin, adiponectin and vascular stiffness parameters in women with systemic lupus erythematosus. Intern. Emerg. Med. 2013, 8, 705–712. [Google Scholar] [CrossRef] [PubMed]
  75. Loghman, M.; Haghighi, A.; Broumand, B.; Ataipour, Y.; Tohidi, M.; Marzbani, C.; Fakharran, M. Association between urinary adiponectin level and renal involvement in systemic lupus erythematous. Int. J. Rheum. Dis. 2016, 19, 678–684. [Google Scholar] [CrossRef]
  76. Landolt-Marticorena, C.; Prokopec, S.D.; Morrison, S.; Noamani, B.; Bonilla, D.; Reich, H.; Scholey, J.; Avila-Casado, C.; Fortin, P.R.; Boutros, P.C.; et al. A discrete cluster of urinary biomarkers discriminates between active systemic lupus erythematosus patients with and without glomerulonephritis. Arthritis Res. 2016, 18, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Wang, Y.; Tao, Y.; Liu, Y.; Zhao, Y.; Song, C.; Zhou, B.; Wang, T.; Gao, L.; Zhang, L.; Hu, H. Rapid detection of urinary soluble intercellular adhesion molecule-1 for determination of lupus nephritis activity. Medicine 2018, 97, e11287. [Google Scholar] [CrossRef] [PubMed]
  78. Brunner, H.I.; Bennett, M.R.; Abulaban, K.; Klein-Gitelman, M.S.; O’Neil, K.M.; Tucker, L.; Ardoin, S.P.; Rouster-Stevens, K.A.; Onel, K.B.; Singer, N.G.; et al. Development of a novel renal activity index of lupus nephritis in children and young adults: Noninvasive measurement of lupus nephritis activity. Arthritis Care Res. 2016, 68, 1003–1011. [Google Scholar] [CrossRef] [Green Version]
  79. Gulati, G.; Bennett, M.R.; Abulaban, K.; Song, H.; Zhang, X.; Ma, Q.; Brodsky, S.V.; Nadasdy, T.; Haffner, C.; Wiley, K.; et al. Prospective validation of a novel renal activity index of lupus nephritis. Lupus 2017, 26, 927–936. [Google Scholar] [CrossRef]
  80. Bennett, M.R.; Ma, Q.; Ying, J.; Devarajan, P.; Brunner, H. Effects of age and gender on reference levels of biomarkers comprising the pediatric Renal Activity Index for Lupus Nephritis (p-RAIL). Pediatr. Rheumatol. 2017, 15, 74. [Google Scholar] [CrossRef] [Green Version]
  81. Zhang, T.-P.; Zhao, Y.-L.; Li, X.-M.; Wu, C.-H.; Pan, H.-F.; Ye, D.-Q. Altered mRNA expression levels of vaspin and adiponectin in peripheral blood mononuclear cells of systemic lupus erythematosus patients. Clin. Exp. Rheumatol. 2019, 37, 458–464. [Google Scholar]
  82. Zhu, W.; He, X.; Cheng, K.; Zhang, L.; Chen, D.; Wang, X.; Qiu, G.; Cao, X.; Weng, X. Ankylosing spondylitis: Etiology, pathogenesis, and treatments. Bone Res. 2019, 7, 22. [Google Scholar] [CrossRef] [Green Version]
  83. Park, J.-H.; Lee, S.-G.; Jeon, Y.-K.; Park, E.-K.; Suh, Y.-S.; Kim, H.-O. Relationship between serum adipokine levels and radiographic progression in patients with ankylosing spondylitis: A preliminary 2-year longitudinal study. Medicine 2017, 96, e7854. [Google Scholar] [CrossRef]
  84. Syrbe, U.; Callhoff, J.; Conrad, K.; Poddubnyy, D.; Haibel, H.; Junker, S.; Frommer, K.W.; Müller-Ladner, U.; Neumann, E.; Sieper, J. Serum adipokine levels in patients with ankylosing spondylitis and their relationship to clinical parameters and radiographic spinal progression: Adipokines in ankylosing spondylitis. Arthritis Rheumatol. 2015, 67, 678–685. [Google Scholar] [CrossRef] [Green Version]
  85. Hartl, A.; Sieper, J.; Syrbe, U.; Listing, J.; Hermann, K.-G.; Rudwaleit, M.; Poddubnyy, D. Serum levels of leptin and high molecular weight adiponectin are inversely associated with radiographic spinal progression in patients with ankylosing spondylitis: Results from the ENRADAS trial. Arthritis Res. Ther. 2017, 19, 140. [Google Scholar] [CrossRef] [PubMed]
  86. Allanore, Y.; Simms, R.; Distler, O.; Trojanowska, M.; Pope, J.; Denton, C.P.; Varga, J. Systemic sclerosis. Nat. Rev. Dis. Primers 2015, 1, 15002. [Google Scholar] [CrossRef] [PubMed]
  87. Tomčík, M.; Arima, K.; Hulejová, H.; Kuklová, M.; Filková, M.; Braun, M.; Beláček, J.; Novák, M.; Bečvář, R.; Vencovský, J.; et al. Adiponectin relation to skin changes and dyslipidemia in systemic sclerosis. Cytokine 2012, 58, 165–168. [Google Scholar] [CrossRef] [PubMed]
  88. Lee, Y.H.; Song, G.G. Meta-analysis of circulating adiponectin, leptin, and resistin levels in systemic sclerosis. Z. Rheumatol. 2017, 76, 789–797. [Google Scholar] [CrossRef]
  89. Stochmal, A.; Czuwara, J.; Zaremba, M.; Rudnicka, L. Altered serum level of metabolic and endothelial factors in patients with systemic sclerosis. Arch. Dermatol. Res. 2020, 312, 453–458. [Google Scholar] [CrossRef] [Green Version]
  90. Michalska-Jakubus, M.; Sawicka, K.; Potembska, E.; Kowal, M.; Krasowska, D. Clinical associations of serum leptin and leptin/adiponectin ratio in systemic sclerosis. Adv. Dermatol. Allergol. 2019, 36, 325–328. [Google Scholar] [CrossRef]
  91. Korman, B.D.; Marangoni, R.G.; Hinchcliff, M.; Shah, S.J.; Carns, M.; Hoffmann, A.; Ramsey-Goldman, R.; Varga, J. Brief report: Association of elevated adipsin levels with pulmonary arterial hypertension in systemic sclerosis: ELEVATED ADIPSIN LEVELS IN SSc-RELATED PAH. Arthritis Rheumatol. 2017, 69, 2062–2068. [Google Scholar] [CrossRef]
  92. Zuo, W.; Wu, Z.-H.; Wu, N.; Duan, Y.-H.; Wu, J.-T.; Wang, H.; Qiu, G.-X. Adiponectin receptor 1 mediates the difference in adiponectin- induced prostaglandin E2 production in rheumatoid arthritis and osteoarthritis synovial fibroblasts. Chin. Med. J. 2011, 124, 3919–3924. [Google Scholar]
  93. Bellocchi, C.; Ying, J.; Goldmuntz, E.A.; Keyes-Elstein, L.; Varga, J.; Hinchcliff, M.E.; Lyons, M.A.; McSweeney, P.; Furst, D.E.; Nash, R.; et al. Large-scale characterization of systemic sclerosis serum protein profile: Comparison to peripheral blood cell transcriptome and correlations with skin/lung fibrosis. Arthritis Rheumatol. 2020, 73, 660–670. [Google Scholar] [CrossRef] [PubMed]
  94. Olewicz-Gawlik, A.; Danczak-Pazdrowska, A.; Kuznar-Kaminska, B.; Batura-Gabryel, H.; Katulska, K.; Wojciech, S.; Trzybulska, D.; Hrycaj, P. Circulating adipokines and organ involvement in patients with systemic sclerosis. Acta Reum. Port. 2015, 40, 156–162. [Google Scholar]
  95. Lakota, K.; Wei, J.; Carns, M.; Hinchcliff, M.; Lee, J.; Whitfield, M.L.; Sodin-Semrl, S.; Varga, J. Levels of adiponectin, a marker for PPAR-gamma activity, correlate with skin fibrosis in systemic sclerosis: Potential utility as a biomarker? Arthritis Res. Ther. 2012, 14, R102. [Google Scholar] [CrossRef] [Green Version]
  96. Arakawa, H.; Jinnin, M.; Muchemwa, F.C.; Makino, T.; Kajihara, I.; Makino, K.; Honda, N.; Sakai, K.; Fukushima, S.; Ihn, H. Adiponectin expression is decreased in the involved skin and sera of diffuse cutaneous scleroderma patients: Letter to the Editor. Exp. Dermatol. 2011, 20, 764–766. [Google Scholar] [CrossRef] [PubMed]
  97. Masui, Y.; Asano, Y.; Shibata, S.; Noda, S.; Aozasa, N.; Akamata, K.; Yamada, D.; Tamaki, Z.; Tada, Y.; Sugaya, M.; et al. Serum adiponectin levels inversely correlate with the activity of progressive skin sclerosis in patients with diffuse cutaneous systemic sclerosis: Significance of serum adiponectin levels in SSc. J. Eur. Acad. Dermatol. Venereol. 2012, 26, 354–360. [Google Scholar] [CrossRef] [PubMed]
  98. Winsz-Szczotka, K.; Kuźnik-Trocha, K.; Komosińska-Vassev, K.; Kucharz, E.; Kotulska, A.; Olczyk, K. Relationship between adiponectin, leptin, IGF-1 and total lipid peroxides plasma concentrations in patients with systemic sclerosis: Possible role in disease development. Int. J. Rheum. Dis. 2016, 19, 706–714. [Google Scholar] [CrossRef]
  99. Marangoni, R.G.; Korman, B.; Varga, J. Adipocytic progenitor cells give rise to pathogenic myofibroblasts: Adipocyte-to-mesenchymal transition and its emerging role in fibrosis in multiple organs. Curr. Rheumatol. Rep. 2020, 22, 79. [Google Scholar] [CrossRef] [PubMed]
  100. Neumann, E.; Lepper, N.; Vasile, M.; Riccieri, V.; Peters, M.; Meier, F.; Hülser, M.-L.; Distler, O.; Gay, S.; Mahavadi, P.; et al. Adipokine expression in systemic sclerosis lung and gastrointestinal organ involvement. Cytokine 2019, 117, 41–49. [Google Scholar] [CrossRef]
  101. Katsiougiannis, S.; Tenta, R.; Skopouli, F.N. Autoimmune epithelitis (Sjögren’s syndrome); the impact of metabolic status of glandular epithelial cells on auto-immunogenicity. J. Autoimmun. 2019, 104, 102335. [Google Scholar] [CrossRef]
  102. Augusto, K.L.; Bonfa, E.; Pereira, R.M.R.; Bueno, C.; Leon, E.P.; Viana, V.S.T.; Pasoto, S.G. Metabolic syndrome in Sjögren’s syndrome patients: A relevant concern for clinical monitoring. Clin. Rheumatol. 2016, 35, 639–647. [Google Scholar] [CrossRef] [PubMed]
  103. Zinkevičienė, A.; Dumalakienė, I.; Mieliauskaitė, D.; Vilienė, R.; Narkevičiūtė, I.; Girkontaitė, I. sICAM-1 as potential additional parameter in the discrimination of the Sjögren syndrome and non-autoimmune sicca syndrome: A pilot study. Clin. Rheumatol. 2019, 38, 2803–2809. [Google Scholar] [CrossRef]
  104. Katsiougiannis, S.; Tenta, R.; Skopouli, F.N. Activation of AMP-activated protein kinase (AMPK) by adiponectin rescues salivary gland epithelial cells from spontaneous and IFNγ-induced apoptosis. Arthritis Rheum. 2010, 62, 414–419. [Google Scholar]
  105. Tvarijonaviciute, A.; Zamora, C.; Martinez-Subiela, S.; Tecles, F.; Pina, F.; Lopez-Jornet, P. Salivary adiponectin, but not adenosine deaminase, correlates with clinical signs in women with Sjögren’s syndrome: A pilot study. Clin. Oral Investig. 2019, 23, 1407–1414. [Google Scholar] [CrossRef]
  106. Kumthekar, A.; Ogdie, A. Obesity and psoriatic arthritis: A narrative review. Rheumatol. Ther. 2020, 7, 447–456. [Google Scholar] [CrossRef] [PubMed]
  107. Dikbas, O.; Tosun, M.; Bes, C.; Tonuk, S.B.; Aksehirli, O.Y.; Soy, M. Serum levels of visfatin, resistin and adiponectin in patients with psoriatic arthritis and associations with disease severity. Int. J. Rheum. Dis. 2016, 19, 672–677. [Google Scholar] [CrossRef] [PubMed]
  108. Xue, Y.; Jiang, L.; Cheng, Q.; Chen, H.; Yu, Y.; Lin, Y.; Yang, X.; Kong, N.; Zhu, X.; Xu, X.; et al. Adipokines in psoriatic arthritis patients: The correlations with osteoclast precursors and bone erosions. PLoS ONE 2012, 7, e46740. [Google Scholar] [CrossRef] [Green Version]
  109. Eder, L.; Jayakar, J.; Pollock, R.; Pellett, F.; Thavaneswaran, A.; Chandran, V.; Rosen, C.F.; Gladman, D.D. Serum adipokines in patients with psoriatic arthritis and psoriasis alone and their correlation with disease activity. Ann. Rheum. Dis. 2013, 72, 1956–1961. [Google Scholar] [CrossRef] [PubMed]
  110. Feld, J.; Nissan, S.; Eder, L.; Rahat, M.A.; Elias, M.; Rimar, D.; Laor, A.; Bitterman, H.; Zisman, D. Increased prevalence of metabolic syndrome and adipocytokine levels in a psoriatic arthritis cohort. J. Clin. Rheumatol. 2018, 24, 302–307. [Google Scholar] [CrossRef] [PubMed]
  111. Chandran, V.; Abji, F.; Perruccio, A.V.; Gandhi, R.; Li, S.; Cook, R.J.; Gladman, D.D. Serum-based soluble markers differentiate psoriatic arthritis from osteoarthritis. Ann. Rheum. Dis. 2019, 78, 796–801. [Google Scholar] [CrossRef]
  112. Negrini, S.; Pappalardo, F.; Murdaca, G.; Indiveri, F.; Puppo, F. The antiphospholipid syndrome: From pathophysiology to treatment. Clin. Exp. Med. 2017, 17, 257–267. [Google Scholar] [CrossRef] [PubMed]
  113. Rodrigues, C.E.M.; Vendramini, M.B.; Bueno, C.; Bonfá, E.; de Carvalho, J.F. Adipocytokines in primary antiphospholipid syndrome: Potential markers of low-grade inflammation, insulin resistance and metabolic syndrome. Clin. Exp. Rheumatol. 2012, 30, 871–878. [Google Scholar] [PubMed]
  114. Monaco, C.; Nanchahal, J.; Taylor, P.; Feldmann, M. Anti-TNF therapy: Past, present and future. Int. Immunol. 2015, 27, 55–62. [Google Scholar] [CrossRef] [PubMed]
  115. Corrado, A.; Colia, R.; Rotondo, C.; Sanpaolo, E.; Cantatore, F.P. Changes in serum adipokines profile and insulin resistance in patients with rheumatoid arthritis treated with anti-TNF-α. Curr. Med. Res. Opin. 2019, 35, 2197–2205. [Google Scholar] [CrossRef] [PubMed]
  116. Engvall, I.-L.; Tengstrand, B.; Brismar, K.; Hafström, I. Infliximab therapy increases body fat mass in early rheumatoid arthritis independently of changes in disease activity and levels of leptin and adiponectin: A randomised study over 21 months. Arthritis Res. Ther. 2010, 12, R197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Kim, K.S.; Choi, H.-M.; Ji, H.-I.; Song, R.; Yang, H.-I.; Lee, S.-K.; Yoo, M.C.; Park, Y.-B. Serum adipokine levels in rheumatoid arthritis patients and their contributions to the resistance to treatment. Mol. Med. Rep. 2014, 9, 255–260. [Google Scholar] [CrossRef] [Green Version]
  118. Ferraz-Amaro, I.; Arce-Franco, M.; Muñiz, J.; López-Fernández, J.; Hernández-Hernández, V.; Franco, A.; Quevedo, J.; Martínez-Martín, J.; Díaz-González, F. Systemic blockade of TNF-α does not improve insulin resistance in humans. Horm. Metab. Res. 2011, 43, 801–808. [Google Scholar] [CrossRef] [PubMed]
  119. Klaasen, R.; Herenius, M.M.J.; Wijbrandts, C.A.; de Jager, W.; van Tuyl, L.H.; Nurmohamed, M.T.; Prakken, B.J.; Gerlag, D.M.; Tak, P.P. Treatment-specific changes in circulating adipocytokines: A comparison between tumour necrosis factor blockade and glucocorticoid treatment for rheumatoid arthritis. Ann. Rheum. Dis. 2012, 71, 1510–1516. [Google Scholar] [CrossRef]
  120. Peters, M.J.L.; Watt, P.; Cherry, L.; Welsh, P.; Henninger, E.; Dijkmans, B.A.C.; McInnes, I.B.; Nurmohamed, M.T.; Sattar, N. Lack of effect of TNF blockade therapy on circulating adiponectin levels in patients with autoimmune disease: Results from two independent prospective studies. Ann. Rheum. Dis. 2010, 69, 1687–1690. [Google Scholar] [CrossRef]
  121. Virone, A.; Bastard, J.-P.; Fellahi, S.; Capeau, J.; Rouanet, S.; Sibilia, J.; Ravaud, P.; Berenbaum, F.; Gottenberg, J.-E.; Sellam, J. Comparative effect of tumour necrosis factor inhibitors versus other biological agents on cardiovascular risk-associated biomarkers in patients with rheumatoid arthritis. RMD Open 2019, 5, e000897. [Google Scholar] [CrossRef] [Green Version]
  122. Toussirot, É.; Mourot, L.; Dehecq, B.; Wendling, D.; Grandclément, É.; Dumoulin, G. TNFα blockade for inflammatory rheumatic diseases is associated with a significant gain in android fat mass and has varying effects on adipokines: A 2-year prospective study. Eur. J. Nutr. 2014, 53, 951–961. [Google Scholar] [CrossRef]
  123. Sikorska, D.; Rutkowski, R.; Łuczak, J.; Samborski, W.; Witowski, J. Serum adiponectin as a predictor of laboratory response to anti-TNF-α therapy in rheumatoid arthritis. Cent. Eur. J. Immunol. 2018, 43, 289–294. [Google Scholar] [CrossRef] [PubMed]
  124. Ruscitti, P.; Ursini, F.; Cipriani, P.; Greco, M.; Alvaro, S.; Vasiliki, L.; Di Benedetto, P.; Carubbi, F.; Berardicurti, O.; Gulletta, E.; et al. IL-1 inhibition improves insulin resistance and adipokines in rheumatoid arthritis patients with comorbid type 2 diabetes: An observational study. Medicine 2019, 98, e14587. [Google Scholar] [CrossRef]
  125. Wagner, C.L.; Visvanathan, S.; Elashoff, M.; McInnes, I.B.; Mease, P.J.; Krueger, G.G.; Murphy, F.T.; Papp, K.; Gomez-Reino, J.J.; Mack, M.; et al. Markers of inflammation and bone remodelling associated with improvement in clinical response measures in psoriatic arthritis patients treated with golimumab. Ann. Rheum. Dis. 2013, 72, 83–88. [Google Scholar] [CrossRef]
  126. Derdemezis, C.S.; Filippatos, T.D.; Voulgari, P.V.; Tselepis, A.D.; Drosos, A.A.; Kiortsis, D.N. Leptin and adiponectin levels in patients with ankylosing spondylitis. The effect of infliximab treatment. Clin. Exp. Rheumatol. 2010, 28, 880–883. [Google Scholar]
  127. Miranda-Filloy, J.A.; López-Mejias, R.; Genre, F.; Carnero-López, B.; Ochoa, R.; Diaz de Terán, T.; González-Juanatey, C.; Blanco, R.; Llorca, J.; González-Gay, M.A. Adiponectin and resistin serum levels in non-diabetic ankylosing spondylitis patients undergoing TNF-α antagonist therapy. Clin. Exp. Rheumatol. 2013, 31, 365–371. [Google Scholar] [PubMed]
  128. Shapiro, L.; Scherer, P.E. The crystal structure of a complement-1q family protein suggests an evolutionary link to tumor necrosis factor. Curr. Biol. 1998, 8, 335–340. [Google Scholar] [CrossRef] [Green Version]
  129. Kishore, U.; Gaboriaud, C.; Waters, P.; Shrive, A.K.; Greenhough, T.J.; Reid, K.B.M.; Sim, R.B.; Arlaud, G.J. C1q and tumor necrosis factor superfamily: Modularity and versatility. Trends Immunol. 2004, 25, 551–561. [Google Scholar] [CrossRef]
  130. Frommer, K.W.; Schäffler, A.; Büchler, C.; Steinmeyer, J.; Rickert, M.; Rehart, S.; Brentano, F.; Gay, S.; Müller-Ladner, U.; Neumann, E. Adiponectin isoforms: A potential therapeutic target in rheumatoid arthritis? Ann. Rheum. Dis. 2012, 71, 1724–1732. [Google Scholar] [CrossRef] [Green Version]
  131. Hua, C.; Buttgereit, F.; Combe, B. Glucocorticoids in rheumatoid arthritis: Current status and future studies. RMD Open 2020, 6, e000536. [Google Scholar] [CrossRef] [Green Version]
  132. Smolen, J.S.; Landewé, R.; Bijlsma, J.; Burmester, G.; Chatzidionysiou, K.; Dougados, M.; Nam, J.; Ramiro, S.; Voshaar, M.; van Vollenhoven, R.; et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann. Rheum. Dis. 2017, 76, 960–977. [Google Scholar] [CrossRef] [PubMed]
  133. Sukumaran, S.; DuBois, D.C.; Jusko, W.J.; Almon, R.R. Glucocorticoid effects on adiponectin expression. In Vitamins & Hormones; Elsevier: Amsterdam, The Netherlands, 2012; Volume 90, pp. 163–186. [Google Scholar]
  134. Yaşar Bilge, N.Ş.; Kaşifoğlu, N.; Kaşifoğlu, T.; Şahin, F.; Gönüllü, E.; Korkmaz, C. The role of methotrexate and low-dose prednisolone on adiponectine levels and insulin resistance in patients with rheumatoid arthritis naïve to disease-modifying antirheumatic drugs. Int. J. Rheum. Dis. 2016, 19, 665–671. [Google Scholar] [CrossRef] [PubMed]
  135. El-Barbary, A.M.; Hussein, M.S.; Rageh, E.M.; Hamouda, H.E.; Wagih, A.A.; Ismail, R.G. Effect of atorvastatin on inflammation and modification of vascular risk factors in rheumatoid arthritis. J. Rheumatol. 2011, 38, 229–235. [Google Scholar] [CrossRef]
  136. The Signaling Pathways Project. Available online: https://www.signalingpathways.org/index.jsf (accessed on 23 November 2020).
  137. Xibillé-Friedmann, D.-X.; Ortiz-Panozo, E.; Bustos Rivera-Bahena, C.; Sandoval-Ríos, M.; Hernández-Góngora, S.-E.; Dominguez-Hernandez, L.; Montiel-Hernández, J.-L. Leptin and adiponectin as predictors of disease activity in rheumatoid arthritis. Clin. Exp. Rheumatol. 2015, 33, 471–477. [Google Scholar]
  138. Kim, G.W.; Lee, N.R.; Pi, R.H.; Lim, Y.S.; Lee, Y.M.; Lee, J.M.; Jeong, H.S.; Chung, S.H. IL-6 inhibitors for treatment of rheumatoid arthritis: Past, present, and future. Arch. Pharm. Res. 2015, 38, 575–584. [Google Scholar] [CrossRef] [PubMed]
  139. Kim, K.S.; Lee, Y.-A.; Ji, H.-I.; Song, R.; Kim, J.Y.; Lee, S.-H.; Hong, S.-J.; Yoo, M.C.; Yang, H.-I. Increased expression of endocan in arthritic synovial tissues: Effects of adiponectin on the expression of endocan in fibroblast-like synoviocytes. Mol. Med. Rep. 2015, 11, 2695–2702. [Google Scholar] [CrossRef] [Green Version]
  140. Fioravanti, A.; Tenti, S.; Bacarelli, M.R.; Damiani, A.; Li Gobbi, F.; Bandinelli, F.; Cheleschi, S.; Galeazzi, M.; Benucci, M. Tocilizumab modulates serum levels of adiponectin and chemerin in patients with rheumatoid arthritis: Potential cardiovascular protective role of IL-6 inhibition. Clin. Exp. Rheumatol. 2019, 37, 293–300. [Google Scholar]
  141. Choi, I.A.; Sagawa, A.; Lee, E.Y.; Lee, E.B.; Song, Y.W. Tocilizumab Increases body weight and serum adipokine levels in patients with rheumatoid arthritis independently of their treatment response: A retrospective cohort study. J. Korean Med. Sci. 2020, 35, e155. [Google Scholar] [CrossRef] [PubMed]
  142. Kubler, P. Experimental and clinical pharmacology: Janus kinase inhibitors: Mechanisms of action. Aust. Prescr. 2014, 37, 154–157. [Google Scholar] [CrossRef]
  143. d’Alessandro, M.; Perillo, F.; Metella Refini, R.; Bergantini, L.; Bellisai, F.; Selvi, E.; Cameli, P.; Manganelli, S.; Conticini, E.; Cantarini, L.; et al. Efficacy of baricitinib in treating rheumatoid arthritis: Modulatory effects on fibrotic and inflammatory biomarkers in a real-life setting. Int. Immunopharmacol. 2020, 86, 106748. [Google Scholar] [CrossRef]
  144. Jandus, C.; Bioley, G.; Rivals, J.-P.; Dudler, J.; Speiser, D.; Romero, P. Increased numbers of circulating polyfunctional Th17 memory cells in patients with seronegative spondylarthritides. Arthritis Rheum. 2008, 58, 2307–2317. [Google Scholar] [CrossRef]
  145. Shibata, S.; Tada, Y.; Hau, C.S.; Mitsui, A.; Kamata, M.; Asano, Y.; Sugaya, M.; Kadono, T.; Masamoto, Y.; Kurokawa, M.; et al. Adiponectin regulates psoriasiform skin inflammation by suppressing IL-17 production from γδ-T cells. Nat. Commun. 2015, 6, 7687. [Google Scholar] [CrossRef] [Green Version]
  146. Piccio, L.; Cantoni, C.; Henderson, J.G.; Hawiger, D.; Ramsbottom, M.; Mikesell, R.; Ryu, J.; Hsieh, C.-S.; Cremasco, V.; Haynes, W.; et al. Lack of adiponectin leads to increased lymphocyte activation and increased disease severity in a mouse model of multiple sclerosis: Immunomodulation. Eur. J. Immunol. 2013, 43, 2089–2100. [Google Scholar] [CrossRef] [Green Version]
  147. Fassio, A.; Gatti, D.; Gisondi, P.; Girolomoni, G.; Viapiana, O.; Giollo, A.; Zamboni, M.; Rossini, M.; Idolazzi, L. Effects of secukinumab on serum adipocytokines: Preliminary data. Reumatismo 2017, 69, 105. [Google Scholar] [CrossRef] [Green Version]
  148. Hall, A.G.; Tilby, M.J. Mechanisms of action of, and modes of resistance to, alkylating agents used in the treatment of haematological malignancies. Blood Rev. 1992, 6, 163–173. [Google Scholar] [CrossRef]
  149. Iqubal, A.; Iqubal, M.K.; Sharma, S.; Ansari, M.A.; Najmi, A.K.; Ali, S.M.; Ali, J.; Haque, S.E. Molecular mechanism involved in cyclophosphamide-induced cardiotoxicity: Old drug with a new vision. Life Sci. 2019, 218, 112–131. [Google Scholar] [CrossRef]
  150. GTEx Portal. Available online: https://gtexportal.org/home/gene/ALDH1A1 (accessed on 15 November 2020).
  151. Available online: https://www.gtexportal.org/home/gene/PPARG (accessed on 15 November 2020).
  152. Huscher, D.; Siegert, E.; Allanore, Y.; Czirják, L.; DelGaldo, F.; Denton, C.P.; Distler, O.; Frerix, M.; Matucci-Cerinic, M.; EUSTAR co-workers on behalf of the DeSScipher Project Research Group within the EUSTAR Network; et al. Systemic sclerosis associated interstitial lung disease—Individualized immunosuppressive therapy and course of lung function: Results of the EUSTAR group. Arthritis Res. Ther. 2018, 20, 17. [Google Scholar]
  153. Tashkin, D.P.; Elashoff, R.; Clements, P.J.; Goldin, J.; Roth, M.D.; Furst, D.E.; Arriola, E.; Silver, R.; Strange, C.; Bolster, M.; et al. Cyclophosphamide versus placebo in scleroderma lung disease. N. Engl. J. Med. 2006, 354, 2655–2666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Masui, Y.; Asano, Y.; Takahashi, T.; Shibata, S.; Akamata, K.; Aozasa, N.; Noda, S.; Taniguchi, T.; Ichimura, Y.; Toyama, T.; et al. Clinical significance of monitoring serum adiponectin levels during intravenous pulse cyclophosphamide therapy in interstitial lung disease associated with systemic sclerosis. Mod. Rheumatol. 2013, 23, 323–329. [Google Scholar] [CrossRef] [PubMed]
  155. Banga, A.; Unal, R.; Tripathi, P.; Pokrovskaya, I.; Owens, R.J.; Kern, P.A.; Ranganathan, G. Adiponectin translation is increased by the PPARγ agonists pioglitazone and ω-3 fatty acids. Am. J. Physiol. Endocrinol. Metab. 2009, 296, E480–E489. [Google Scholar] [CrossRef] [Green Version]
  156. Bernatsky, S.; Boivin, J.F.; Joseph, L.; Manzi, S.; Ginzler, E.; Gladman, D.D.; Urowitz, M.; Fortin, P.R.; Petri, M.; Barr, S.; et al. Mortality in systemic lupus erythematosus. Arthritis Rheum. 2006, 54, 2550–2557. [Google Scholar] [CrossRef]
  157. Curado Borges, M.; De Miranda Moura Dos Santos, F.; Weiss Telles, R.; Melo De Andrade, M.V.; Toulson Davisson Correia, M.I.; Lanna, C.C.D. Omega-3 fatty acids, inflammatory status and biochemical markers of patients with systemic lupus erythematosus: A pilot study. Rev. Bras. Reumatol. 2017, 57, 526–534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Lozovoy, M.; Simão, A.; Morimoto, H.; Scavuzzi, B.; Iriyoda, T.; Reiche, E.; Cecchini, R.; Dichi, I. Fish oil N-3 fatty acids increase adiponectin and decrease leptin levels in patients with systemic lupus erythematosus. Mar. Drugs 2015, 13, 1071–1083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Kedenko, L.; Lamina, C.; Kiesslich, T.; Kapur, K.; Bergmann, S.; Waterworth, D.; Heid, I.M.; Wichmann, H.E.; Kedenko, I.; Kronenberg, F.; et al. Genetic polymorphisms of the main transcription factors for adiponectin gene promoter in regulation of adiponectin levels: Association analysis in three european cohorts. PLoS ONE 2012, 7, e52497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Liu, Y.; Wang, J.; Luo, S.; Zhan, Y.; Lu, Q. The roles of PPARgamma and its agonists in autoimmune diseases: A comprehensive review. J. Autoimmun 2020, 113, 102510. [Google Scholar] [CrossRef]
  161. Li, X.F.; Sun, Y.Y.; Bao, J.; Chen, X.; Li, Y.H.; Yang, Y.; Zhang, L.; Huang, C.; Wu, B.M.; Meng, X.M.; et al. Functional role of PPAR-gamma on the proliferation and migration of fibroblast-like synoviocytes in rheumatoid arthritis. Sci. Rep. 2017, 7, 12671. [Google Scholar] [CrossRef] [Green Version]
  162. Oxer, D.S.; Godoy, L.C.; Borba, E.; Lima-Salgado, T.; Passos, L.A.; Laurindo, I.; Kubo, S.; Barbeiro, D.F.; Fernandes, D.; Laurindo, F.R.; et al. PPARgamma expression is increased in systemic lupus erythematosus patients and represses CD40/CD40L signaling pathway. Lupus 2011, 20, 575–587. [Google Scholar] [CrossRef]
  163. Zhao, W.; Berthier, C.C.; Lewis, E.E.; McCune, W.J.; Kretzler, M.; Kaplan, M.J. The peroxisome-proliferator activated receptor-gamma agonist pioglitazone modulates aberrant T cell responses in systemic lupus erythematosus. Clin. Immunol. 2013, 149, 119–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Aprahamian, T.R.; Bonegio, R.G.; Weitzner, Z.; Gharakhanian, R.; Rifkin, I.R. Peroxisome proliferator-activated receptor gamma agonists in the prevention and treatment of murine systemic lupus erythematosus. Immunology 2014, 142, 363–373. [Google Scholar] [CrossRef] [PubMed]
  165. FDA. Available online: https://www.fda.gov/drugs/drug-safety-and-availability/medication-guides (accessed on 13 November 2020).
  166. Mallano, T.; Palumbo-Zerr, K.; Zerr, P.; Ramming, A.; Zeller, B.; Beyer, C.; Dees, C.; Huang, J.; Hai, T.; Distler, O.; et al. Activating transcription factor 3 regulates canonical TGFbeta signalling in systemic sclerosis. Ann. Rheum. Dis. 2016, 75, 586–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Korman, B.; Marangoni, R.G.; Lord, G.; Olefsky, J.; Tourtellotte, W.; Varga, J. Adipocyte-specific repression of PPAR-gamma by NCoR contributes to scleroderma skin fibrosis. Arthritis Res. 2018, 20, 145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Rahmouni, K.; Sigmund, C.D. Id3, E47, and SREBP-1c: Fat factors controlling adiponectin expression. Circ. Res. 2008, 103, 565–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Lindahl, G.E.; Stock, C.J.; Shi-Wen, X.; Leoni, P.; Sestini, P.; Howat, S.L.; Bou-Gharios, G.; Nicholson, A.G.; Denton, C.P.; Grutters, J.C.; et al. Microarray profiling reveals suppressed interferon stimulated gene program in fibroblasts from scleroderma-associated interstitial lung disease. Respir Res. 2013, 14, 80. [Google Scholar] [CrossRef] [Green Version]
  170. Guo, Z.; Li, H.; Han, M.; Xu, T.; Wu, X.; Zhuang, Y. Modeling Sjögren’s syndrome with Id3 conditional knockout mice. 2011, 135, 34–42. Immunol. Lett. 2011, 135, 34–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Sakurai, D.; Yamaguchi, A.; Tsuchiya, N.; Yamamoto, K.; Tokunaga, K. Expression of ID family genes in the synovia from patients with rheumatoid arthritis. Biochem. Biophys. Res. Commun. 2001, 284, 436–442. [Google Scholar] [CrossRef]
  172. Liu, C.; Yu, S.; Jin, R.; Long, Y.; Lu, S.; Song, Y.; Sun, X.; Sun, X.H.; Zhang, Y. Correlation of the levels of DNA-binding inhibitor Id3 and regulatory T cells with SLE disease severity. J. Autoimmun. 2020, 113, 102498. [Google Scholar] [CrossRef]
  173. Hai, T.; Wolford, C.C.; Chang, Y.S. ATF3, a hub of the cellular adaptive-response network, in the pathogenesis of diseases: Is modulation of inflammation a unifying component? Gene Expr. 2010, 15, 1–11. [Google Scholar] [CrossRef]
  174. Engler, A.; Tange, C.; Frank-Bertoncelj, M.; Gay, R.E.; Gay, S.; Ospelt, C. Regulation and function of SIRT1 in rheumatoid arthritis synovial fibroblasts. J. Mol. Med. 2016, 94, 173–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Olivares, D.; Perez-Hernandez, J.; Forner, M.J.; Perez-Soriano, C.; Tormos, M.C.; Saez, G.T.; Chaves, F.J.; Redon, J.; Cortes, R. Urinary levels of sirtuin-1 associated with disease activity in lupus nephritis. Clin. Sci. 2018, 132, 569–579. [Google Scholar] [CrossRef] [PubMed]
  176. Chu, H.; Jiang, S.; Liu, Q.; Ma, Y.; Zhu, X.; Liang, M.; Shi, X.; Ding, W.; Zhou, X.; Zou, H.; et al. Sirtuin1 protects against systemic sclerosis-related pulmonary fibrosis by decreasing proinflammatory and profibrotic processes. Am. J. Respir Cell Mol. Biol. 2018, 58, 28–39. [Google Scholar] [CrossRef]
  177. Fasshauer, M.; Kralisch, S.; Klier, M.; Lossner, U.; Bluher, M.; Klein, J.; Paschke, R. Adiponectin gene expression and secretion is inhibited by interleukin-6 in 3T3-L1 adipocytes. Biochem. Biophys. Res. Commun. 2003, 301, 1045–1050. [Google Scholar] [CrossRef]
  178. He, Y.; Lu, L.; Wei, X.; Jin, D.; Qian, T.; Yu, A.; Sun, J.; Cui, J.; Yang, Z. The multimerization and secretion of adiponectin are regulated by TNF-alpha. Endocrine 2016, 51, 456–468. [Google Scholar] [CrossRef] [PubMed]
  179. Lim, J.Y.; Kim, W.H.; Park, S.I. GO6976 prevents TNF-alpha-induced suppression of adiponectin expression in 3T3-L1 adipocytes: Putative involvement of protein kinase C. FEBS Lett. 2008, 582, 3473–3478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Farrugia, M.; Baron, B. The role of TNF-α in rheumatoid arthritis: A focus on regulatory T cells. J. Clin. Transl. Res. 2016, 2, 84. [Google Scholar] [CrossRef] [PubMed]
  181. Postal, M.; Appenzeller, S. The role of Tumor Necrosis Factor-alpha (TNF-α) in the pathogenesis of systemic lupus erythematosus. Cytokine 2011, 56, 537–543. [Google Scholar] [CrossRef]
  182. Shehzad, A.; Iqbal, W.; Shehzad, O.; Lee, Y.S. Adiponectin: Regulation of its production and its role in human diseases. Hormones 2012, 11, 8–20. [Google Scholar] [CrossRef] [PubMed]
  183. Fan, W.; Imamura, T.; Sonoda, N.; Sears, D.D.; Patsouris, D.; Kim, J.J.; Olefsky, J.M. FOXO1 transrepresses peroxisome proliferator-activated receptor gamma transactivation, coordinating an insulin-induced feed-forward response in adipocytes. J. Biol. Chem. 2009, 284, 12188–12197. [Google Scholar] [CrossRef] [Green Version]
  184. Standiford, T.J.; Keshamouni, V.G.; Reddy, R.C. Peroxisome proliferator-activated receptor-{gamma} as a regulator of lung inflammation and repair. Proc. Am. Thorac. Soc. 2005, 2, 226–231. [Google Scholar] [CrossRef]
  185. Zhang, X.; Young, H.A. PPAR and immune system—What do we know? Int. Immunopharmacol. 2002, 2, 1029–1044. [Google Scholar] [CrossRef]
  186. Chinetti, G.; Griglio, S.; Antonucci, M.; Torra, I.P.; Delerive, P.; Majd, Z.; Fruchart, J.-C.; Chapman, J.; Najib, J.; Staels, B. Activation of proliferator-activated receptors α and γ induces apoptosis of human monocyte-derived macrophages. J. Biol. Chem. 1998, 273, 25573–25580. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Gene expression of adiponectin—ADIPOQ (ENSG00000181092.9) in human tissues from the Genotype-Tissue Expression (GTEx) portal. The expression values are given in transcripts per million (log10 (TPM + 1)), calculated from a gene model with isoforms collapsed to a single gene. No other normalization steps were applied. Box plots are shown as median and as 25th and 75th percentile. Only tissues with the median TPM > 0.1 are presented.
Figure 1. Gene expression of adiponectin—ADIPOQ (ENSG00000181092.9) in human tissues from the Genotype-Tissue Expression (GTEx) portal. The expression values are given in transcripts per million (log10 (TPM + 1)), calculated from a gene model with isoforms collapsed to a single gene. No other normalization steps were applied. Box plots are shown as median and as 25th and 75th percentile. Only tissues with the median TPM > 0.1 are presented.
Ijms 22 04095 g001
Figure 2. (a) Adiponectin structure and isoforms. (b) Simplified adiponectin downstream signaling. ACC: Acetyl-CoA carboxylase, AMPK: AMP-activated protein kinase, APPL1: adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1, eNOS: Endothelial NOS, NF-Κb: Nuclear factor-κB, p38MAPK: p38 mitogen-activated protein kinase, PPAR-α: Peroxisome proliferator-activated receptor α, S1P: sphingosine-1-phosphate.
Figure 2. (a) Adiponectin structure and isoforms. (b) Simplified adiponectin downstream signaling. ACC: Acetyl-CoA carboxylase, AMPK: AMP-activated protein kinase, APPL1: adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1, eNOS: Endothelial NOS, NF-Κb: Nuclear factor-κB, p38MAPK: p38 mitogen-activated protein kinase, PPAR-α: Peroxisome proliferator-activated receptor α, S1P: sphingosine-1-phosphate.
Ijms 22 04095 g002
Figure 3. Adiponectin circulating levels observed in certain SARDs. The values are presented as standard mean difference (SMD) with 95% confidence intervals (CI) and association p values. The most recent meta-analysis with the highest number of studies included was presented for each SARD. AS: Ankylosing spondylitis, HC: Healthy controls, RA: Rheumatoid arthritis, SLE: Systemic lupus erythematosus, SSc: Systemic sclerosis.
Figure 3. Adiponectin circulating levels observed in certain SARDs. The values are presented as standard mean difference (SMD) with 95% confidence intervals (CI) and association p values. The most recent meta-analysis with the highest number of studies included was presented for each SARD. AS: Ankylosing spondylitis, HC: Healthy controls, RA: Rheumatoid arthritis, SLE: Systemic lupus erythematosus, SSc: Systemic sclerosis.
Ijms 22 04095 g003
Figure 4. Local loss of dWAT in SSc skin in addition to adipocytes undergoing AMT causes decreased adiponectin secretion. Consequently, its inhibitory effects on myofibroblasts are lost, which results in fibrosis. AMT: adipocyte mesenchymal transition, dWAT: dermal white adipose tissue, EMC: Extracellular matrix, SSc: Systemic sclerosis.
Figure 4. Local loss of dWAT in SSc skin in addition to adipocytes undergoing AMT causes decreased adiponectin secretion. Consequently, its inhibitory effects on myofibroblasts are lost, which results in fibrosis. AMT: adipocyte mesenchymal transition, dWAT: dermal white adipose tissue, EMC: Extracellular matrix, SSc: Systemic sclerosis.
Ijms 22 04095 g004
Figure 5. Superposition of trimeric globular domain of adiponectin (PDB: 6U66) in blue and TNF-α (PDB:1TNF) in yellow shows high 3D-structure similarity.
Figure 5. Superposition of trimeric globular domain of adiponectin (PDB: 6U66) in blue and TNF-α (PDB:1TNF) in yellow shows high 3D-structure similarity.
Ijms 22 04095 g005
Figure 6. Simplified ADIPOQ gene regulation with TNF-α and IL-6. ATF3: Activating Transcription Factor 3, C-EBP: CCAAT-enhancer-binding protein, Erk1/2: extracellular signal-regulated kinases 1/2 FoxO1: Forkhead box protein O1, Id3: Inhibitor Of DNA Binding 3, IL: Interleukin, JAK: Janus kinase, JNK: Jun N-terminal kinase, NFAT: Nuclear factor of activated T-cells, PPAR-γ: Peroxisome-proliferator-activated receptor gamma, PPRE: Peroxisome proliferator response element, R: receptor, SIRT: Sirtuin, SREBP: Sterol regulatory element-binding protein, STAT: signal transducer and activator of transcription, TNF: Tumor necrosis factor.
Figure 6. Simplified ADIPOQ gene regulation with TNF-α and IL-6. ATF3: Activating Transcription Factor 3, C-EBP: CCAAT-enhancer-binding protein, Erk1/2: extracellular signal-regulated kinases 1/2 FoxO1: Forkhead box protein O1, Id3: Inhibitor Of DNA Binding 3, IL: Interleukin, JAK: Janus kinase, JNK: Jun N-terminal kinase, NFAT: Nuclear factor of activated T-cells, PPAR-γ: Peroxisome-proliferator-activated receptor gamma, PPRE: Peroxisome proliferator response element, R: receptor, SIRT: Sirtuin, SREBP: Sterol regulatory element-binding protein, STAT: signal transducer and activator of transcription, TNF: Tumor necrosis factor.
Ijms 22 04095 g006
Figure 7. Summary of adiponectin levels in SARDs associated with clinical manifestations, treatment and adiponectin gene regulation. AS: Ankylosing spondylitis, ATF3: Activating Transcription Factor 3, C-EBP: CCAAT-enhancer binding protein, CY: Cyclophosphamide, DMARD: Disease-modifying antirheumatic drugs, FoxO1: Forkhead box protein O1, GC: Glucocorticoids, Id3: Inhibitor Of DNA Binding 3, IL: Interleukin, JAK: Janus kinase, NFAT: Nuclear factor of activated T-cells, PBMCs: Peripheral blood mononuclear cells, PPRE: Peroxisome proliferator response element, RA: rheumatoid arthritis, SIRT: Sirtuin 1/NAD-dependent deacetylase, SLE: Systemic lupus, erythematosus, SREBP: Sterol regulatory element-binding protein, SSc: Systemic sclerosis, TCZ: Tocilizumab, TNF: Tumor necrosis factor.
Figure 7. Summary of adiponectin levels in SARDs associated with clinical manifestations, treatment and adiponectin gene regulation. AS: Ankylosing spondylitis, ATF3: Activating Transcription Factor 3, C-EBP: CCAAT-enhancer binding protein, CY: Cyclophosphamide, DMARD: Disease-modifying antirheumatic drugs, FoxO1: Forkhead box protein O1, GC: Glucocorticoids, Id3: Inhibitor Of DNA Binding 3, IL: Interleukin, JAK: Janus kinase, NFAT: Nuclear factor of activated T-cells, PBMCs: Peripheral blood mononuclear cells, PPRE: Peroxisome proliferator response element, RA: rheumatoid arthritis, SIRT: Sirtuin 1/NAD-dependent deacetylase, SLE: Systemic lupus, erythematosus, SREBP: Sterol regulatory element-binding protein, SSc: Systemic sclerosis, TCZ: Tocilizumab, TNF: Tumor necrosis factor.
Ijms 22 04095 g007
Table 1. Clinical observations associated with circulating adiponectin levels in RA patients.
Table 1. Clinical observations associated with circulating adiponectin levels in RA patients.
Clinical FeatureRA Patients(N)Serum Adiponectin Levels Associations/CorrelationsReference
Disease activity351/90/52Positive association with DAS28-ESR.[26,38,42]
51Early RA with high adiponectin levels was less likely to have MHAQ score > 3 and RAPID3 score > 12.[29]
121Stratifying according to DAS28 (low, moderate and high activity), there were no differences seen for adiponectin.[43]
180Negative correlation of total, HMW, MMW, and LMW adiponectin with the DAS28.[44]
40Negative correlation with DAS28. [45]
70Positive correlation with DAS28-ESR in active disease.[46]
80Negative correlation with the number of swollen joints.[47]
Radiographic severity/progression324Positive association of total, but not HMW adiponectin with radiographic progression. [48]
242Positive correlation with radiographic severity.[49]
632Independent association with baseline total SHS, ΔSHS ≥ 1 and predicted ΔSHS ≥ 5.[36]
253Positive association with radiographic progression over 4 years.[50]
152Positive association with radiographic progression.[51]
CV-related54No correlation with coronary artery calcification.[52]
210In RA patients with abdominal obesity or no clinically evident joint damage associated with decreased carotid atherosclerosis.[53]
192Leptin: adiponectin ratio associated with common carotid artery resistive index.[54]
210Positive associations of total and HMW adiponectin with increased blood pressure parameters, and in white patients additionally with endothelial activation.[55]
Bone-related112Negative association with trabecular volumetric bone mineral density and cortical thickness.[56]
38Positive correlation with osteopontin in serum. [28]
Muscle-related50Negative association with appendicular lean mass index and muscle cross-sectional area.[57]
DAS28: Disease Activity Score of 28 joints, ESR: erythrocyte sedimentation rate, HMW: High-molecular-weight, LMW: Low-molecular-weight, MHAQ: Multidimensional Health Assessment Questionnaire, MMW: Medium-molecular-weight, RA: Rheumatoid arthritis, RAPID3: Routine Assessment of Patient Index Data 3, SHS: Sharp-van der Heijde Score.
Table 2. Anti-TNF effects on circulating adiponectin levels in rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis.
Table 2. Anti-TNF effects on circulating adiponectin levels in rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis.
SARDPatients (N)Anti-TNF Treatment RegimenStudy DurationInfluence on Adiponectin LevelsRef.
RA16adalimumab40 mg every 2 weeks1 yearNo change[118]
etanercept25 mg twice a week
Infliximab3 mg/kg every 8 weeks
48adalimumab40 mg every 2 weeks16 weeksNo change[119]
171adalimumab40 mg every 2 weeks16 weeksNo change[120]
96adalimumabat approved doses24 weeksNo change[121]
certolizumabat approved doses
etanerceptat approved doses
infliximabat approved doses
8adalimumab40 mg every 2 weeks2 yearsNo change[122]
etanercept50 mg every week
infliximab3 mg/kg
21adalimumabat approved doses12 weeksNo change[123]
certolizumabat approved doses
etanerceptat approved doses
golimumabat approved doses
infliximabat approved doses
15anti-TNFat approved doses6 monthsNo change[124]
33adalimumab40 mg every 2 weeks12 andIncrease[115]
etanercept50 mg every week24 weeks
infliximab5 mg/kg every 8 weeks
16infliximab3 mg/kg in weeks 0, 2 and 6 and every 8 weeks after24 months No change[116]
3–12 monthsIncrease
16adalimumab-6 monthsIncrease[117]
eternacept-
infliximab -
PsA126onercept50 mg or 100 mg three times a week12 weeksNo change[120]
405golimumab50 mg or 100 mg every 4 weeks14 weeksNo change[125]
AS30infliximab5 mg/kg in weeks 0, 2, 6 and every 8 weeks after6 monthsNo change[126]
29infliximabInfusion (120 min)before and right after No change[127]
12adalimumab40 mg every 2 weeks2 yearsNo change[122]
etanercept50 mg every week
infliximab5 mg/kg
AS: Ankylosing spondylitis, PsA: Psoriatic arthritis, RA Rheumatoid arthritis.
Table 3. Glucocorticoids (prednisolone) effects on circulating adiponectin levels in rheumatoid arthritis.
Table 3. Glucocorticoids (prednisolone) effects on circulating adiponectin levels in rheumatoid arthritis.
Patients (N)Treatment RegimenAdditional TherapyAdditional Therapy RegimenStudy DurationInfluence on APN LevelsRef.
657.5–15.0 mg/dayMTX10–15 mg/week3 monthsIncrease[134]
1510 mg/dayMTX0.2 mg/kg/week6 monthsIncrease[135]
1510 mg/dayMTX + ATV0.2 mg/kg/week
40 mg/day
6 monthsIncrease[135]
960 mg/day (week 1);
40 mg/day (week 2)
--2 weeksIncrease[119]
19Tapered high dose:
60 mg/day (week 1);
40 mg/day (week 2);
30 mg/day (week 3);
20 mg/day (week 4);
15 mg/day (week 5);
10 mg/day (week 6);
7.5 mg/day (thereafter)
HCQ,
SSA
MTX
400 mg/day
2 g/day
10 mg/week
22 weeksNo change[119]
12710 mg/day or lessDMARDStable therapy6 monthsNo change[137]
91 1 year
52 2 years
ATV: Atorvastatin, DMARD: Disease-modifying antirheumatic drugs, HCQ: Hydroxychloroquine, MTX: Methotrexate, SSA: Sulfasalazine.
Table 4. Tocilizumab effects on circulating adiponectin levels in rheumatoid arthritis.
Table 4. Tocilizumab effects on circulating adiponectin levels in rheumatoid arthritis.
N of PatientsTreatment RegimenAdditional TherapyStudy DurationInfluence on APN LevelsRef.
418 mg/kg± MTX, GC, NSAID6 monthsIncrease[141]
408 mg/kg every 4 weeks± MTX, SSA, HCQ, Leflunomide, GC, statins, anti-diabetics4 monthsDecrease[27]
208 mg/kg every 4 weeks± NSAID, coxibs, GC6 monthsIncrease[140]
248 mg/kg every 4 weeksMTX (± NSAID, coxibs, GC)6 monthsIncrease[140]
47--6 monthsIncrease[121]
GC: Glucocorticoids, HCQ: Hydroxychloroquine, MTX: Methotrexate, NSAID: Nonsteroidal anti-inflammatory drugs, SSA: Sulphasalazine.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Brezovec, N.; Perdan-Pirkmajer, K.; Čučnik, S.; Sodin-Šemrl, S.; Varga, J.; Lakota, K. Adiponectin Deregulation in Systemic Autoimmune Rheumatic Diseases. Int. J. Mol. Sci. 2021, 22, 4095. https://doi.org/10.3390/ijms22084095

AMA Style

Brezovec N, Perdan-Pirkmajer K, Čučnik S, Sodin-Šemrl S, Varga J, Lakota K. Adiponectin Deregulation in Systemic Autoimmune Rheumatic Diseases. International Journal of Molecular Sciences. 2021; 22(8):4095. https://doi.org/10.3390/ijms22084095

Chicago/Turabian Style

Brezovec, Neža, Katja Perdan-Pirkmajer, Saša Čučnik, Snežna Sodin-Šemrl, John Varga, and Katja Lakota. 2021. "Adiponectin Deregulation in Systemic Autoimmune Rheumatic Diseases" International Journal of Molecular Sciences 22, no. 8: 4095. https://doi.org/10.3390/ijms22084095

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop