Skip to main content

Immunoprofiling of cytokines, chemokines, and growth factors in female patients with systemic lupus erythematosus– a pilot study

Abstract

Systemic Lupus Erythematosus (SLE) is a chronic autoimmune disease affecting different organ systems. This study aimed to determine the concentrations of 30 different human cytokines, chemokines, and growth factors in human plasma to understand the role of these markers in the pathogenicity of SLE using Luminex Multiple Analyte Profiling (xMAP) technology. Plasma samples were obtained from patients with SLE (n = 28), osteoarthritis (OA) (n = 9), and healthy individuals (n = 12) were obtained. High levels of TNF, IL-6, IFN-γ, INF-α, IL-4, IL-5, IL-13, IL-8, IP-10, MIG, MCP-1, MIP-1β, GM-CSF, G-CSF, EGF, VEGF, IL-12, IL-1RA, and IL-10 was detected in SLE patients compared with the OA and healthy control groups. xMAP analysis has been used to address the differential regulation of clinical heterogeneity and immunological phenotypes in SLE patients. In addition, complete disease phenotyping information along with cytokine immune profiles would be useful for developing personalized treatments for patients with SLE.

Peer Review reports

Introduction

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by aberrant immune system activity, which causes different clinical symptoms such as heterogeneous renal, dermatological, and cardiovascular dysfunction [1], [2]. Genetic and environmental factors and cytokine imbalance are factors that trigger inflammation and induce organ damage [3]. In addition, a link between SLE and cytomegalovirus (CMV) has been suggested to affect disease development [4]. The SLE Disease Activity Index (SLEDAI) is one of the most commonly used disease activity measurements. It has less sensitive for changes, provides a high bar for improvement, and is based on symptom severity, regardless of the current severity in the patient [5]. The American College of Rheumatology (ACR) established the criteria for SLE classification. It contains laboratory biomarkers including anti-nuclear antibody (Ab) (ANA), anti-dsDNA Ab, complement (C2, C3, and C4), white blood cells, platelets, urinary casts, proteinuria, and hemolytic anemia [6]. The Systemic Lupus International Collaborating Clinics (SLICC) has validated an alternative system using a series of consensus exercises for symptomatology and laboratory results from real rheumatologic cases. The SLICC criteria require either meeting ≥ 4 of 17 criteria, including at least one clinical and one immunological criterion, or demonstrating biopsy for lupus nephritis proven with ANA or dsDNA [7], [8]. The SLICC classification criteria correlate with disease activity, capturing more manifestations that are also included in the SLEDAI [9]. Furthermore, although lupus is not a form of arthritis, it is one of the most common clinical features of SLE, which is usually non-erosive compared to rheumatoid arthritis (RA) [10]. Osteoarthritis (OA) is the most common type of noninflammatory arthritis. It is not rare for lupus patients to have OA, but significant conditions usually start at about age 50, which are unrelated to their lupus [11], [12].

Biomarkers play an important role in SLE diagnosis, classification, complications, and disease activity assessment. Therefore, finding an ideal biomarker for SLE is challenging, as it needs to meet different characteristics such as (i) reflecting the underlying pathophysiology, (ii) high predictive values, high sensitivity, and specificity, (iii) ability to monitor disease activity, (iv) reliability in tissues, cells, or fluid, and (v) stability and ease of detection [2], [13], [14].

Cytokines play a critical role in T cell proliferation, differentiation, activation, and function [15,16,17]. Secreted cytokines can be detected in the blood, saliva, urine, and target tissues, such as the skin and kidney. Most of these cytokines have pro-inflammatory properties; however, some have anti-inflammatory or immunomodulatory properties. However, as the pathogenesis of SLE is not well understood, it is not yet clear whether the levels of certain cytokines trigger the disease or are merely an epiphenomenon of multifunctional immune regulation, responses, cell death, or elimination of non-viable cell remnants [18]. In contrast, abnormal production of pro-inflammatory and anti-inflammatory cytokines has been reported as a potential therapeutic target for the treatment of SLE patients [11]. Cytokine profiling techniques provide a good way to evaluate the levels of various cytokines and assess their association with SLE disease progression and severity [19]. Therefore, finding an ideal biomarker for SLE is challenging, as it needs to meet different characteristics such as (i) reflecting the underlying pathophysiology, (ii) high predictive values, high sensitivity, and specificity, (iii) ability to monitor disease activity, (iv) reliability in tissues, cells, or fluid, and (v) stability and ease of detection [2], [13], [14].

Accordingly, this study aimed to determine the concentrations of 30 different human cytokines, chemokines, and growth factors in human plasma to understand the role of these markers in SLE pathogenicity using Luminex Multiple Analyte Profiling (xMAP) technology. Healthy controls and OA samples were used as comparators to gain insight into the nature of cytokine levels in SLE patients.

Materials and methods

Sample collection

Peripheral blood samples were obtained from healthy volunteers (n = 12), patients with knee osteoarthritis (OA) (n = 9), and patients with SLE (n = 28). The samples included in this study were from consecutive SLE patients who had no clinical evidence of OA, and the knee OA patients included in this study were examined clinically. The SLICC classification criteria [8] were determined at the last visit, and samples were collected. Disease activity was assessed using the SLEDAI, and a score of 3 or 4 was considered an active disease. Patient samples were collected from the Rheumatology Clinic of the King Abdulaziz University Hospital (KAUH), Jeddah, KSA, after obtaining written informed consent.

Sample processing

Plasma samples were obtained by centrifugation of the blood at 3,000 ×g for 10 min and stored immediately at -80 °C until further use.

Multiple analyte profiling (xMAP)

Plasma samples isolated from normal controls, OA patients, and SLE patients were analyzed within 6 months for a variety of cytokines, chemokines, cytokine receptors, and growth factors using the Human Cytokine Magnetic 30-Plex Panel (Novex®) (Invitrogen, USA) according to the manufacturer’s instructions, as previously described (11). Briefly, magnetic beads coupled with antibodies for 30 different analytes were added and washed twice with 1X wash buffer. The standard was prepared by mixing the 16plex and 14plex solutions provided by the manufacturer, and a serial dilution protocol was used to prepare a range of standard solutions (1:3 serial dilution). Both standards and serum samples (1:2 dilution) were prepared and added to the washed beads in the designated wells of the Mylar plates and incubated in an orbital shaker at 500 rpm for 2 h. Between incubations with different antibodies, the plate was washed twice with the wash buffer. The plate was then incubated with secondary antibodies for 1 h and with streptavidin-RPE-coupled detection antibodies for 30 min. The plate was washed three times, resuspended in wash buffer, and analyzed using a MAGPIX® instrument (Luminex Corporation, USA).

Statistical analysis

The raw data obtained for individual analytes were analyzed using the Luminex xPONENT® multiplex assay analysis software (Luminex Corporation, USA) to calculate the absolute concentration in the normal control, OA, and SLE groups. In addition, the concentration of each analyte was further analyzed using GraphPad Prism v.9 (GraphPad Software, USA) to compute the statistical significance using the Kruskal-Wallis test and Student’s t-test. Data was shown as mean ± SD and median (Interquartile range) for non-parametric data. All results are shown with 95%, 99%, and 99.9% accuracy for both the Kruskal–Wallis test and Student’s t-tests.

Results

Demographics, medications, and laboratory parameters

All the participants in this study were female (28 patients with SLE, 9 with OA, and 12 healthy controls). The median ages of patients with SLE, OA, and controls were 37 (with interquartile range 36–38), 50 (with interquartile range 49–51), and 43 (with interquartile range 42–44) years respectively. The clinical details for SLE were as follows: the mean duration of disease was 8,5 years; the disease activity was calculated using the SLE Disease Activity Index (SLEDAI), and the SLEDAI score to define active disease was 3 or 4. Fifteen patients were active, and 13 patients were inactive. Approximately 71% of SLE patients showed the presence of ANA and high levels of dsDNA. The patient characteristics, clinical details, and treatment regimens are shown in Table 1.

Table 1 Characteristics and clinical information of SLE patients

Th cytokine profiles

Plasma levels of Th1 (TNF, IL-6, IL-1β, IFN-α, and IFN-γ) and Th2 (IL-4, IL-5, and IL-13) cytokines in the SLE, OA, and healthy control groups were quantified using a magnetic 30-plex panel based on Luminex xMAP technology. Here, we observed high expression levels of TNF (a), IL-6 (b), IFN-α (d), IL-4 (f), IL-5 (g), and IL-13 (h) in SLE patients compared to controls (healthy and OA). No expression of IL-6, IK-1β, or IL-4 was observed in the control group. However, there was no significant difference in the expression of IL-1β (c) and IFN-γ (e) (Th1) between all groups. In addition, the comparison between the groups showed that there was a significant difference between the OA and SLE groups in the levels of TNF- and IFN-α and between the control and OA groups in the levels of IFN-α and IL-4 (Fig. 1).

Chemokine profiles

Similar to the cytokines, some chemokines were highly expressed in patients with SLE compared to the control or OA groups, such as IL-8 (a), IP-10 (b), MIG (e), MCP-1 (f), and MIP-1α (h). However, RANTES (c) increased in OA compared to healthy controls. The MIP-1β (h) levels were significantly higher in the control and SLE groups than in the OA group. No significant difference was observed in the expression of Eotaxin (d) or MIP-1α (g) between the groups. In addition, a comparison between groups showed that there was a significant difference in the expression of IL-8 and RANTES between the control and SLE groups (Fig. 2).

Fig. 1
figure 1

Levels of Th1 (TNF, IL-6, IL-1β, IFN-α, IFN-γ), and Th2 (IL-4, IL-5, IL-13) cytokines in the plasma of control, OA, and SLE groups. Graph shows the median by Kruskal Wallis test, where *= P < 0.05, **=P < 0.01, ***=P < 0.001, ****=P < 0.0001 0001 and Student’s t-test #=P < 0.05, ##=P < 0.01, ###=P < 0.001

Fig. 2
figure 2

Levels of chemokines (IL-8, IP-10, RANTES, Eotaxin, MIG, MCP-1, MIP-1α, MIP-1β) in the plasma of control, OA, and SLE groups. Graph shows the median by Kruskal Wallis test, where *= P < 0.05, **=P < 0.01, ***=P < 0.001, ****=P < 0.0001 0001 and Student’s t-test #=P < 0.05, ##=P < 0.01, ###=P < 0.001

Growth factors profiles

Among the growth factor measurements, GM-CSF (a), G-CSF (b), EGF (e), and VEGF (f) levels were significantly higher in the SLE group than in the controls. HGF (c) and FGF (d) levels did not differ between the groups. In addition, the comparison between the groups showed that there was a significant difference in the levels of GM-CSF between the control and OA groups. G-CSF, EGF, VEGF, and FGF markers were not detected in control and some OA patients (Fig. 3).

Fig. 3
figure 3

Levels of growth factors (GM-CSF, G-CSF, HGF, FGF, EGF, VEGF) in the plasma of control, OA, and SLE groups. Graph shows the median by Kruskal Wallis test, where *= P < 0.05, **=P < 0.01, ***=P < 0.001, ****=P < 0.0001 and Student’s t-test #=P < 0.05, ##=P < 0.01, ###=P < 0.001

Anti-inflammatory and other pro-inflammatory cytokine profiles

IL-12 (d) was highly expressed in SLE patients compared to the control groups, unlike IL-2R (a), IL-2 (b), IL-7 (c), IL-15 (e), and IL-17 (f). The IL-1RA (g) and IL-10 (h) mediators were higher in the SLE group than in the OA and control groups. IL-2, IL-15, IL-17 and IL-7 were not detected in control and some OA or SLE patients. In addition, the comparison between the groups showed a significant difference between the control and SLE groups in the levels of IL -2 and between the control and OA groups in the levels of IL -2R, IL -12, IL -10, and the levels of IL -2 and IL -12 between the OA and SLE groups (Fig. 4).

Fig. 4
figure 4

Levels of pro-inflammatory (IL-2R, IL-2, IL-7, IL-15, IL-17) and anti-inflammatory (IL-1RA, IL-10) mediators in the plasma of control, OA, and SLE groups. Graph shows the median by Kruskal Wallis test, where *= P < 0.05, **=P < 0.01, ***=P < 0.001, ****=P < 0.0001 and Student’s t-test #=P < 0.05, ##=P < 0.01, ###=P < 0.001

Discussion

The abnormal activity of cytokines plays an important role in autoimmune disease (e.g., SLE) pathogenicity, and multiplex assays help to assess the abnormal biological activity of cytokines, chemokines, and growth factor levels [19,20,21,22]. Activation of the major components of the innate and adaptive immune systems has been observed in patients with SLE. Increased levels of various cytokines are also observed during the disease course [12]. In the current study, an array of cytokines, chemokines, and growth factor levels were evaluated using the Human Cytokine Magnetic 30-Plex Panel in 28 SLE patients, 9 with OA, and 12 healthy controls. OA or arthralgia occurs in 95% of patients with SLE at some point during the course of the disease [19], [22]. Therefore, in this study, we compared SLE and OA patients to identify common markers that may play a role in the development of OA in SLE patients. Patients with SLE had significantly increased levels of Th1 and Th2 associated cytokines including TNF, IL-6, IFN-γ, IL-4, IL-5, and IL-13. A comparison between the groups also showed significant differences in the levels of TNF and IFN-α. TNF can either promote or modulate autoimmunity; however, it has been shown to be upregulated in SLE patients compared to healthy controls and is correlated with disease activity [23,24,25]. A previous study by Xiang et al. showed high expression level of IL-1β, IL-18, IL-6, IFN-α, IL-10, and TNF compared to healthy controls. In the same study, they found that TGF-β1 levels in SLE patients were significantly lower than those in controls [26]. Another study by Hashemi et al. found that the serum levels of TNF-α, IL-2, IL-4, and IL-5 were significantly decreased in SLE patients who underwent an 8-weeks exercise training program compared to those in the control group. In addition, the levels of IL-10, IL-13, and IL-22 were significantly higher in the control group than in the intervention group after 8-weeks of exercises [27]. Although OA is a noninflammatory form of arthritis, we found that pro-inflammatory cytokines TNF-α and IL -6 were significantly higher in OA compared with healthy controls. Molnar et al. reviewed in detail the cytokines and chemokines involved in the pathogenesis of OA and highlighted the studies showing the systemic increase in pro-inflammatory cytokines and chemokines in OA patients compared to healthy controls [28].

Chemokines are a group of cytokines with low molecular weights that direct the chemotaxis of target cells. They play a role in the pathogenesis of SLE and related complications, especially lupus nephritis [29]. The concentrations of IL-8, IP-10, MIG, and MCP-1 were significantly higher is SLE patients compering than in the OA and control groups. On the other hand, here, the level of RANTES was higher in OA group compering to SLE group However, comparisons within groups showed that there was a significant difference between the control and SLE groups in IL-8 and RANTES. Similar to Yoshio et al., the mean concentrations of IL-6, IL-8, IP-10, MCP-1, G-CSF, and GM-CSF were higher in central neuropsychiatric SLE than in the sera, and the level of RANTES was much lower than that in the control [30]. Eotaxin and MIP-1α levels were not significantly different between groups. However, a study by Novikov et al. showed that the expression of otaxin and MIP-1α was significantly lower in patients with SLE than in those with rheumatoid arthritis (RA) group [31].

For growth factors expression, the GM-CSF and G-CSF are the key survival factors for granulocytes [32]. They have been shown to inhibit neutrophil apoptosis in healthy subjects in vitro at various concentrations [33]. GM-CSF deficiency in mice leads to the development of an SLE-like disease that correlates with impaired apoptotic cell phagocytosis [34]. EGF has been suggested for monitoring kidney injury in the setting of demonstrations of SLE [35]. Previous studies have shown that high VEGF levels may correlate with disease activity in SLE and lupus nephritis. However, high urinary VEGF expression may represent the ongoing process of fibrosis and tissue repair in lupus nephritis kidneys [36], [37]. A study by Adhya et al. showed that VEGF levels could be a useful marker to detect disease activity in the urine of lupus nephritis patients compared to non-renal SLE patients [38]. VEGF, TNF-α, HGF, and FGF are potent mitogens with angiogenic activity [39], [40]. Hrycek et al. showed that FGF levels were low in patients with newly diagnosed untreated SLE [39]. In the present study, GM-CSF, G-CSF, EGF, and VEGF levels were significantly higher in the SLE group than in the OA and healthy control groups. There was a significant difference in the levels of GM-CSF. However, HGF and FGF levels did not differ between the groups.

IL-12 is a pro-inflammatory cytokine that induces Th cell differentiation into Th1 cells [41]. It also induces different cytotoxicity pathways and the expression of cytotoxic mediators by amplifying the production of IFN-γ [42]. High levels of IL-12 have been observed in patients with SLE compared with controls [43]. IL-2 is a proinflammatory mediator that plays an important role in immune cell activation and peripheral tolerance. It has been suggested to exhibit therapeutic efficacy in patients [44], [45]. IL-15 plays a role in B cell antibody production, which is increased in the serum of SLE patients [46]. Moreover, IL-2R is correlated with other markers that are potential targets of IL-15 [47]. IL-7 plays an important role in T-cell development, homeostasis, and immune tolerance, suggesting the use of the IL-7/IL-7Rα pathway as a target for autoimmune disease treatment [48]. IL-17 and Th17 cells are involved in SLE pathogenesis [49]. However, previous studies have shown no increase in IL-17 expression and no correlation with disease activity [50], [51]. In the current study, IL-12 was the only pro-inflammatory mediator that showed high expression in SLE patients compared to that in healthy controls. On the other hand, IL-2R, IL-2, IL-7, IL-15, and IL-17 mediators did not show any significant differences compared with the control group. IL-1RA and IL-10 mediators were higher in the SLE group than in the OA and control groups. In addition, the comparison between the control and OA groups showed significant differences in the values of IL-2R, IL-2, IL-12, and IL-10, and in the values of IL-2R, IL-2, and IL-12 between OA and SLE, as well as in the values of IL-2 between the control and OA groups. Similarly, Toit et al. showed high expression of IL -1RA in SLE patients with myocardial damage [52], and IL -10 level were higher in SLE patients than in controls [16].

Conclusions

In conclusion, the current study demonstrates that SLE is a complex disease, and the underlying disease mechanism remines unclear. Further analysis is required to understand whether there is any relationship between the high expression of some markers in SLE and OA patients, but not in controls. This indicates that patients with SLE may be able to develop OA at some stage during their disease course. A better understanding of these biological factors and their expression may provide essential clues to the pathogenic pathways and open new avenues for more effective therapeutics. However, the small sample size in the present study may be considered a bias, and further studies are needed to understand the cytokine network and patterns both ex vivo in human SLE samples and in vivo in animal models. Additionally, more information on disease phenotyping would be helpful in planning personalized treatment.

Data Availability

The datasets used and/or analyzed during the current study available from the corresponding author on reasonable request.

Abbreviations

IL-1RA:

Interleukin 1-receptor antagonist

IL-2R:

Interleukin-2 receptor

ACR:

American College of Rheumatology

ALT:

Alanine aminotransferase

ANA:

Anti-nuclear antibody

AST:

Aspartate aminotransferase

CBC:

Complete blood count

CCL11:

C-C motif chemokine Ligand 11 or Eotaxin

CMV:

Cytomegalovirus

EGF:

Epidermal growth factor

FGF:

Fibroblast growth factor

G-CSF:

Granulocyte-colony stimulating factor

GM-CSF:

Granulocyte Macrophage -Colony Stimulating factor

HB:

Hemoglobin

HGF:

Hepatocyte growth factor

IFN-α:

Interferon alpha

IFN-γ:

Interferon gamma

IL-1β:

Interleukin-1 beta

IL:

Interleukin

IP-10:

Interferon γ-inducible protein-10

MCH:

Mean corpuscular hemoglobin

MCP-1:

Monocyte chemotactic protein-1

MCV:

Mean corpuscular volume

MIG:

Monokine induced by gamma interferon

MIP-1α:

Macrophage inflammatory protein alpha

MIP-1β:

Macrophage inflammatory protein-1 beta

OA:

Osteoarthritis

PLT:

Platelet

RA:

Rheumatoid arthritis

RANTES:

regulated on activation, normal T cell expressed and secreted

RBC:

Red blood cell

SLE:

Systemic Lupus Erythematosus

SLEDAI:

SLE Disease Activity Index

SLICC:

Systemic Lupus International Collaborating Clinics

TNF-α:

Tumor necrosis factor alpha

VEGF:

Vascular endothelial growth factor

xMAP:

Luminex Multiple Analyte Profiling

References

  1. Kiriakidou M, Ching CL. Systemic Lupus Erythematosus. Ann Intern Med. 2020;172:ITC81–ITC96.

    Article  PubMed  Google Scholar 

  2. Yu H, Nagafuchi Y, Fujio K. Clinical and Immunological Biomarkers for Systemic Lupus Erythematosus. Biomolecules vol. 11 (2021).

  3. Ohl K, Tenbrock K. Inflammatory cytokines in systemic lupus erythematosus. J. Biomed. Biotechnol. 2011, 432595 (2011).

  4. Krasselt M, Kippenhahn A, Baerwald C, Pietsch C, Seifert O. Relationship between cytomegalovirus prevalence and markers of disease activity in systemic lupus erythematosus. Rheumatology. 2022;61:1288–90.

    Article  PubMed  Google Scholar 

  5. et al. Scoring systemic lupus erythematosus (SLE) disease activity with simple, rapid outcome measures. Lupus Sci. & Med. 6, e000365 (2019).

  6. Tan EM, et al. The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1982;25:1271–7.

    Article  CAS  PubMed  Google Scholar 

  7. Petri M. Review of classification criteria for systemic lupus erythematosus. Rheum Dis Clin North Am. 2005;31:245–54. vi.

    Article  PubMed  Google Scholar 

  8. Petri M, et al. Derivation and validation of the systemic Lupus International collaborating clinics classification criteria for systemic lupus erythematosus. Arthritis Rheum. 2012;64:2677–86.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Anić F, Zuvić-Butorac M, Stimac D, Novak S. New classification criteria for systemic lupus erythematosus correlate with disease activity. Croat Med J. 2014;55:514–9.

    Article  PubMed Central  Google Scholar 

  10. Sippl N, et al. Arthritis in systemic lupus erythematosus is characterized by local IL-17A and IL-6 expression in synovial fluid. Clin Exp Immunol. 2021;205:44–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Katsuyama T, Tsokos GC, Moulton VR, Aberrant. T cell signaling and subsets in systemic Lupus Erythematosus. Front Immunol vol. 9 (2018).

  12. La Cava A. Anticytokine therapies in systemic lupus erythematosus. Immunotherapy. 2010;2:575–82.

    Article  PubMed  Google Scholar 

  13. González LA, Ugarte-Gil MF, Alarcón GS. Systemic lupus erythematosus: the search for the ideal biomarker. Lupus. 2020;30:181–203.

    Article  PubMed  Google Scholar 

  14. Liu C-C, Ahearn JM. The search for lupus biomarkers. Best Pract Res Clin Rheumatol. 2009;23:507–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Jin S, Yu C, Yu B. Changes of serum IL-6, IL-10 and TNF-α levels in patients with systemic lupus erythematosus and their clinical value. Am J Transl Res. 2021;13:2867–74.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Wu Y-R, Hsing C-H, Chiu C-J, Huang H-Y, Hsu Y-H. Roles of IL-1 and IL-10 family cytokines in the progression of systemic lupus erythematosus: friends or foes? IUBMB Life. 2022;74:143–56.

    Article  CAS  PubMed  Google Scholar 

  17. Rzeszotarska E et al. IL-1β, IL-10 and TNF-α polymorphisms may affect systemic lupus erythematosus risk and phenotype. Clin Exp Rheumatol (2022).

  18. Idborg H, Oke V. Cytokines as biomarkers in systemic Lupus Erythematosus: value for diagnosis and drug therapy. Int J Mol Sci. 2021;22:11327.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Park J, et al. Cytokine clusters as potential diagnostic markers of disease activity and renal involvement in systemic lupus erythematosus. J Int Med Res. 2020;48:300060520926882.

    Article  CAS  PubMed  Google Scholar 

  20. Pacheco Y, et al. Cytokine and autoantibody clusters interaction in systemic lupus erythematosus. J Transl Med. 2017;15:239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Reynolds JA, et al. Cytokine profiling in active and quiescent SLE reveals distinct patient subpopulations. Arthritis Res Ther. 2018;20:173.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Bahlas S, et al. Rapid immunoprofiling of cytokines, chemokines and growth factors in patients with active rheumatoid arthritis using Luminex multiple analyte profiling technology for precision medicine. Clin Exp Rheumatol. 2019;37:112–9.

    PubMed  Google Scholar 

  23. Cigni A, et al. Interleukin 1, interleukin 6, Interleukin 10, and Tumor Necrosis factor α in active and quiescent systemic Lupus Erythematosus. J Investig Med. 2014;62:825LP–829.

    Article  Google Scholar 

  24. Idborg H, et al. TNF-α and plasma albumin as biomarkers of disease activity in systemic lupus erythematosus. Lupus Sci Med. 2018;5:e000260.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Weckerle CE, et al. Large-scale analysis of tumor necrosis factor α levels in systemic lupus erythematosus. Arthritis Rheum. 2012;64:2947–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Xiang N, et al. Expression profile of PU.1 in CD4(+)T cells from patients with systemic lupus erythematosus. Clin Exp Med. 2021;21:621–32.

    Article  CAS  PubMed  Google Scholar 

  27. Hashemi S, Habibagahi Z, Heidari M, Abdollahpour-Alitappeh M, Karimi MH. Effects of combined aerobic and anaerobic exercise training on cytokine profiles in patients with systemic lupus erythematosus (SLE); a randomized controlled trial. Transpl Immunol. 2022;70:101516.

    Article  CAS  PubMed  Google Scholar 

  28. Molnar V et al. Cytokines and Chemokines involved in Osteoarthritis Pathogenesis. Int J Mol Sci 22, (2021).

  29. Ghafouri-Fard S, Shahir M, Taheri M, Salimi A. A review on the role of chemokines in the pathogenesis of systemic lupus erythematosus. Cytokine. 2021;146:155640.

    Article  CAS  PubMed  Google Scholar 

  30. Yoshio T, et al. IL-6, IL-8, IP-10, MCP-1 and G-CSF are significantly increased in cerebrospinal fluid but not in sera of patients with central neuropsychiatric lupus erythematosus. Lupus. 2016;25:997–1003.

    Article  CAS  PubMed  Google Scholar 

  31. Novikov A, et al. Cytokine profiles in systemic Lupus Erythematosus and Rheumatoid Arthritis. Ann Rheum Dis. 2016;75:907LP–907.

    Article  Google Scholar 

  32. Armstrong DJ, Whitehead EM, Crockard AD, Bell AL. Distinctive effects of G-CSF, GM-CSF and TNFalpha on neutrophil apoptosis in systemic lupus erythematosus. Clin Exp Rheumatol. 2005;23:152–8.

    CAS  PubMed  Google Scholar 

  33. Stanford SJ, Pepper JR, Burke-Gaffney A, Mitchell JA. Cytokine-activated human vascular smooth muscle delays apoptosis of neutrophils: relevance of interactions between cyclo-oxygenase-2 and colony-stimulating factors. FASEB J Off Publ Fed Am Soc Exp Biol. 2001;15:1813–5.

    CAS  Google Scholar 

  34. Enzler T, et al. Deficiencies of GM-CSF and Interferon γ Link inflammation and Cancer. J Exp Med. 2003;197:1213–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ngamjanyaporn P, et al. Predicting treatment response and clinicopathological findings in lupus nephritis with urine epidermal growth factor, monocyte chemoattractant protein-1 or their ratios. PLoS ONE. 2022;17:e0263778.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Avihingsanon Y, et al. Measurement of urinary chemokine and growth factor messenger RNAs: a noninvasive monitoring in lupus nephritis. Kidney Int. 2006;69:747–53.

    Article  CAS  PubMed  Google Scholar 

  37. Robak E, Woźniacka A, Sysa-Jedrzejowska A, Stepień H, Robak T. Serum levels of angiogenic cytokines in systemic lupus erythematosus and their correlation with disease activity. Eur Cytokine Netw. 2001;12:445–52.

    CAS  PubMed  Google Scholar 

  38. Adhya Z, et al. Soluble TNF-R1, VEGF and other cytokines as markers of disease activity in systemic lupus erythematosus and lupus nephritis. Lupus. 2019;28:713–21.

    Article  CAS  PubMed  Google Scholar 

  39. Hrycek A, Janowska J, Cieślik P. Selected angiogenic cytokines in systemic lupus erythematosus patients. Autoimmunity. 2009;42:459–66.

    Article  CAS  PubMed  Google Scholar 

  40. Robak E, Woźniacka A, Sysa-Jedrzejowska A, Stepień H, Robak T. Circulating angiogenesis inhibitor endostatin and positive endothelial growth regulators in patients with systemic lupus erythematosus. Lupus. 2002;11:348–55.

    Article  CAS  PubMed  Google Scholar 

  41. Manetti R, et al. Natural killer cell stimulatory factor (interleukin 12 [IL-12]) induces T helper type 1 (Th1)-specific immune responses and inhibits the development of IL-4-producing th cells. J Exp Med. 1993;177:1199–204.

    Article  CAS  PubMed  Google Scholar 

  42. Nembrini C, Abel B, Kopf M, Marsland BJ. Strong TCR signaling, TLR ligands, and cytokine redundancies ensure robust development of type 1 effector T cells. J Immunol. 2006;176:7180–8.

    Article  CAS  PubMed  Google Scholar 

  43. Uzrail AH, Assaf AM, Abdalla SS. Correlations of Expression Levels of a Panel of Genes (IRF5, STAT4, TNFSF4, MECP2, and TLR7) and Cytokine Levels (IL-2, IL-6, IL-10, IL-12, IFN-γ, and TNF-α) with Systemic Lupus Erythematosus Outcomes in Jordanian Patients. Biomed Res. Int. 2019, 1703842 (2019).

  44. H J Y, et al. Homeostatic imbalance of regulatory and effector T cells due to IL-2 deprivation amplifies murine lupus. Proc Natl Acad Sci. 2010;107:204–9.

    Article  Google Scholar 

  45. Mizui M et al. IL-2 Protects Lupus-Prone Mice from Multiple End-Organ Damage by Limiting CD4 – CD8 – IL-17–Producing T Cells. J. Immunol. 193, 2168 LP – 2177 (2014).

  46. Armitage RJ, Macduff BM, Eisenman J, Paxton R, Grabstein KH. IL-15 has stimulatory activity for the induction of B cell proliferation and differentiation. J Immunol. 1995;154:483–90.

    Article  CAS  PubMed  Google Scholar 

  47. Aringer M, et al. Serum interleukin-15 is elevated in systemic lupus erythematosus. Rheumatology. 2001;40:876–81.

    Article  CAS  PubMed  Google Scholar 

  48. Dooms H. Interleukin-7: fuel for the autoimmune attack. J Autoimmun. 2013;45:40–8.

    Article  CAS  PubMed  Google Scholar 

  49. Robert M, Miossec P. Interleukin-17 and lupus: enough to be a target? For which patients? Lupus. 2019;29:6–14.

    Article  PubMed  Google Scholar 

  50. Ndongo-Thiam N, Miossec P. A cell-based bioassay for circulating bioactive IL-17: application to destruction in rheumatoid arthritis. Ann. Rheum. Dis. 74, 1629 LP – 1631 (2015).

  51. Raymond W, Ostli-Eilertsen G, Griffiths S, Nossent J. IL-17A levels in systemic lupus erythematosus associated with inflammatory markers and lower rates of malignancy and heart damage: evidence for a dual role. Eur J Rheumatol. 2017;4:29–35.

    Article  PubMed  Google Scholar 

  52. du Toit R, et al. Serum cytokine levels associated with myocardial injury in systemic lupus erythematosus. Rheumatology (Oxford). 2021;60:2010–21.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

N/A.

Funding

This study received no external funding.

Author information

Authors and Affiliations

Authors

Contributions

LAD and PNP proposed this concept. LAD, ID and SB collected samples. PNP performed the experiments. LAD, ID, and PNP analyzed the data. LAD, SD and PNP wrote the manuscript. All authors have read and approved the manuscript.

Corresponding authors

Correspondence to Laila A. Damiati or Peter Natesan Pushparaj.

Ethics declarations

Ethics approval and consent to participate

The study was carried out in accordance with the Declaration of Helsinki. The study was approved by the Ethics Committee of the King Abdulaziz University Hospital (KAUH) (Reference No 140 − 37), Jeddah, Saudi Arabia. Patient samples were collected from the Rheumatology Clinic of KAUH, Jeddah, KSA, after obtaining written informed consent.

Consent for publication

N/A.

Competing interests

The authors declare that they have no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Damiati, L.A., Denetiu, I., Bahlas, S. et al. Immunoprofiling of cytokines, chemokines, and growth factors in female patients with systemic lupus erythematosus– a pilot study. BMC Immunol 24, 13 (2023). https://doi.org/10.1186/s12865-023-00551-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12865-023-00551-6

Keywords