Skip to main content
  • Research note
  • Open access
  • Published:

Epigenetic mechanisms shape the underlining expression regulatory mechanisms of the STAT3 in multiple sclerosis disease

Abstract

Objectives

Immunological tolerance is mediated by CD4+CD25+ regulatory T (Treg) cells. Studies have shown that thymic and peripheral generations of Treg cells depend on the CD28 signaling pathway. T helper 17 (Th17) cells are involved in the pathophysiology of various inflammatory diseases. Cytokines, such as interleukin (IL)-6 and TGF-β, regulate the reciprocal development of Th17 and Treg cells. In CD4+ T cells, signal transducer and activator of transcription 3 (STAT3) play a critical role in the induction of Th17 cell differentiation and inhibition of Treg cell development.

Results

In this study, we investigated the STAT3 methylation and gene expression status in patients with MS. Our study demonstrated that the level of STAT3 methylation decreased in relapsing–remitting MS patient compared to control groups, which the decreases were statistically significant. STAT3 gene expression increased in patient group relative to healthy one, and the increases were found to be statistically significant. According to our findings, it can be suggested that DNA hypermethylation of STAT3 affects the gene expression. In addition, there is a strong and significant negative correlation between the methylation status and mRNA level of STAT3.

Introduction

Multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system (CNS), has been evidenced to cause demyelination and axonal degeneration within the brain and spinal cord [1, 2]. The exact etiopathology of MS has not yet been clarified, but most studies have recognized MS as an autoimmune disease mediated by autoreactive CD4+ T cells [3].

Immunological tolerance is a critical factor in the prevention of chronic infection, cancer, and autoimmune diseases [4]. Central tolerance operates in the thymus where autoreactive T cells with high affinity for self-antigens are negatively deleted [5]. Given that not all antigens are present in the thymus, self-reactive T cells can enter the peripheral blood [5]. Therefore, central tolerance alone is insufficient, and peripheral tolerance mechanisms are required [6]. CD4+CD25+ regulatory T (Treg) cells are major suppressor T lymphocytes and mediate peripheral tolerance [7, 8]. Forkhead box P3 (FOXP3) transcription factor is also necessary for the differentiation of Treg cells [9, 10]. Although Treg cells differentiate naturally in the thymus, these cells can also be generated from CD4+CD25+ naive T cells into adaptive Tregs in the periphery [11,12,13]. Adaptive Treg cells can be induced in the periphery when encountered with repeated antigens [14].

Researchers have suggested that the CD28/B7 costimulatory molecule is essential for the expression of CD25 and FOXP3 on Tregs [15,16,17]. It has also been indicated that in the absence of the CD28 costimulatory pathway, the peripheral number of Tregs decreases [16]. Besides, Lck-binding motif in the cytosolic tail of CD28 is required for Tregs generation [18]. However, it is uncertain how CD28 leads to the FOXP3 expression and Treg development [16]. Treg cells are involved in maintaining anergic state and exert suppressive function in various inflammation and autoimmune diseases, including rheumatoid arthritis, systemic lupus erythematosus, and MS [17, 19,20,21].

In MS patients, autoreactive CD4+ T cells display mainly T helper 17 (Th17) phenotype [3]. Cytokines, such as TGF-β and interleukin (IL)-6, play a key role in regulating Th17 cell differentiation [3, 22]. Retinoic acid receptor-related orphan receptor (ROR) γt transcription factor induces the differentiation of naive CD4+ T cells into Th17 cells [22, 23], which are pathogenic in MS due to the production of cytokines such as IL-17, IL-21, and IL-22 [24]. In MS, IL-17 leads to blood–brain barrier disruption and clinical disease activity and symptoms [25]. The upregulation of RORγt is dependent on STAT3 [26]. Following the binding of IL-6 to IL-6R, STAT3 is phosphorylated on Tyr705, dimerizes, moves into the nucleus and regulates the gene expression [27, 28]. STAT3 functions distinctly in the Th17 development and regulation of the Th17/Treg balance [23], and STAT3 deficiency impairs RORɣt expression, giving rise to the increased expression of FOXP3 [29]. Therefore, dysregulation of STAT3 results in the development of various inflammatory diseases, and loss of STAT3 in naïve CD4+ T cells inhibits the development of CNS inflammatory diseases [30, 31]. Several studies have introduced STAT3 as a risk factor allele for MS disease susceptibility [32,33,34]. These observations persuaded us to investigate whether the hypo- or hyper-methylation of STAT3 in CD4+ T cells is associated with the susceptibility of MS.

In this study, we display that in CD4+ T cells, STAT3 methylation decreases in relapsing–remitting MS (RRMS), whereas the gene expression of STAT3 increases.

Main text

Methods

Study groups

A total of 50 MS patients (36 males and 14 females) aged between 19 and 65 years with clinically RRMS were collected from the Imam Reza Hospital of Tabriz University of Medical Sciences, East Azerbaijan Province, Iran. All the patients had RRMS according to the McDonald's diagnostic criteria and were in the remission clinical phase. Disease remission was defined as improvement from baseline clinical status for at least three months. The cases were weekly being treated with interferon beta. Normal controls enrolled in this study were composed of 50 age, gender, and ethnically matched healthy subjects without any clinical or laboratory signs of autoimmune or inflammatory diseases. A written informed consent was obtained from each case, and the study protocol was approved by the Ethics Committee of the Tabriz University of Medical Sciences. The clinical/pathological data of both RRMS and controls are summarized in Table 1.

Table 1 Clinical characteristics of RRMS and control subjects

Blood sampling and cell isolation

Peripheral blood samples (20 ml) were obtained from all the patients with RRMS. After the blood collection, peripheral blood mononuclear cells were isolated using Ficoll-Paque™plus gradient centrifugation (Biosera, UK) within 12 h. The isolation of CD4+ T cells from peripheral blood mononuclear cells was carried out with the Miltenyi Biotech’s MACS System. The CD4+ MACS Isolation Kit was applied to positively select CD4+ T cells. The purity of the CD4+ T cells was assessed with flow cytometry and assigned to be greater than 90%.

DNA extraction and methylation-specific quantitative polymerase chain reaction (MS-qPCR)

Total DNA isolated from the CD4+ T cells was gathered in EDTA-containing tubes by the salting-out method. STAT3 promoter sequences and data were obtained from the NCBI (National Center for Biotechnology Information) database. STAT3 expression primers were designed by the aid of the PrimerQuest Tool, and the methylation- and demethylation (DM)-specific primers for STAT3 were designed using MethPrimer online database and OLIGO software. The primer sequences and product size for STAT3 are shown in Table 2. The methylation status of STAT3 was analyzed by applying the MS‐qPCR. Power SYBR Green reagent (Thermo Fisher Scientific, USA) was utilized for MS-qPCR. The DM rate of STAT3 was calculated by a previously described formula [35, 36] in which DM is the ratio of amplification efficiency of the methylated to unmethylated samples:

$$ {\text{DM}}\% = 100/[1 + 2({\text{Ct.TG}} - {\text{Ct.CG}})]. $$
Table 2 PCR primers, melting temperature, and product size

RNA isolation and reverse transcriptase (RT)-PCR

Total RNA from the collected CD4+ T cells was isolated using TRIzol Reagent (Life Technologies, USA) based on the manufacturer’s instructions. RNA was then reverse transcribed with the Prime Script™ RT reagent Kit (Takara, Japan) as per the protocol recommended by manufacturer. Subsequently, SYBR Green reagent (Thermo Fisher Scientific) was used for quatitative real‐time (qRT‐PCR). Pfaffl method [37] was applied to calculate the relative gene expression. PCR cycles included a holding cycle at 95 °C for 15 min and held at 80 °C before the addition of 1.25 units of Taq polymerase (Invitrogen, USA). The forward and reverse primers used for STAT3 expression were comprised of 5′-TGGAGCTGCGGCAGTTTCTG-3′ and 5′-CCGCATCTGGTCCAGCGCAG-3′, respectively [38]. For STAT3, the temperature condition was as follows: 30 cycles of 95 °C for 1 min, 63 °C for 1 min, followed by one cycle of 72 °C for 5 min. The mRNA expression level was normalized against glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA. STAT3 primer sequences are listed in Table 2.

Statistical analysis

Statistical analysis was performed with SPSS software version 25 (IBM Corp., Armonk, NY, USA). All the data were presented as mean ± standard error of mean (SEM). Kolmogorov–Smirnov test with P–P plot and Q–Q plot was employed for normal distributions. The differences in the mRNA level of STAT3 between RRMS patient and control groups were evaluated by unpaired t-test. A P-value < 0.05 was considered as statistically significant difference.

Results

STAT3 expression in the study groups

Our results showed that the STAT3 expression level increased in patients in comparison with the control group (P-value < 0.0001; Fig. 1a).

Fig. 1
figure 1

The gene expression and promoter methylation level of the STAT3 gene in MS and control groups. a STAT3 gene expression in patients compared with control subjects. Mean fold change in MS patients and control group was 1.000 ± 0.024 and 0.250 ± 0.014, respectively (P-value < 0.0001). b The methylation level (% M) of STAT3 in MS patients compared with control group. The red dots are individual values, and boxes are the mean methylation for patient (30.56 ± 1.317) and control groups (59.08 ± 1.986). c, d Correlation analysis between methylation status and gene expression level of STAT3 in patients (R2 = 0.8967, P-value < 0.0001, regression analysis) and control (R2 = 0.9256, P-value < 0.0001, regression analysis) group. e Negative and positive controls for methylation assays. 5-Azacytidine and DNA sample treated with SssI methyltransferase were used as negative and positive controls, respectively [39]. (*P-value < 0.05, **P-value < 0.01, ***P-value < 0.001, ****P-value < 0.0001).

STAT3 methylation status in the study groups

Methylation level (% M) of the promoter region of STAT3 was present in 23% (12/50) of MS cases, while this level was found in 62.9% (45/50) of the controls. The decrease level was statistically significant (P-value < 0.0001). In addition, a significant and strong negative correlation was found between the STAT3 gene methylation level and mRNA expression level for the methylation assay (Figs. 1b–e).

Discussion

FOXP3 is an essential transcription factor in the differentiation of Treg cells [40, 41]. Immunodysregulation, polyendocrinopathy, enteropathy, X-linked syndrome are disorders found in patients with FOXP3 mutations [22, 40]. FOXP3-deficient mice also exhibit eosinophilia, hyperimmunoglobulinemia E syndrome, and dysregulated production of Th1 and Th2 cytokines [42]. These observations verifies the major role of FOXP3 in Treg development, control immune tolerance, and homeostasis [15]. The CD28 costimulatory molecule is another factor required for the Treg development and peripheral conversion. Th17 cells have immunopathogenic potential, and their responses have been associated with the murine models of collagen-induced arthritis and experimental autoimmune encephalitis.

STAT3 is one of the regulating factors in the reciprocal development of Th17 cells and Tregs. A previous study has been shown that STAT3 is directly involved in the FOXP3 expression and Treg development [27]. In the present study, we observed STAT3 hypormethylation in RRMS patients and found that the STAT3 gene expression increases in RRMS patients, but not in the control subjects.

The results of the STAT3 methylation level and gene expression status in our study demonstrated the decreased level of methylation and the increased mean of mRNA expression in the patient group compared to the healthy one (hypomethylated). Therefore, our findings reveal a critical novel epigenetic event and new insights into the pathogenesis of MS disease. Regulation of the STAT3 in the present study maybe a novel promising treatment for MS as it has formerly been demonstrated that highly activated Th17 activity is related to STAT3 mutations [43]. Moreover, germline mutations in STAT3 causes the lymphoproliferation and early-onset autoimmunity [44]. An earlier investigation has reflected that STAT3-targeted therapeutics prevents experimental autoimmune uveitis mediated by Th17 cells [45]. STAT3 inhibitors are also effective in CNS autoimmune diseases [46].

Taken together, these findings affirm the role of STAT3 in Th17-mediated immune diseases. However, further studies are needed to fully elucidate the exact role of STAT3 in MS disease. STAT3 induction in the autoimmune therapy protocol is recommended.

Limitations

The major concern of this study is the examination of STAT3 methylation on limited MS patients. The test of methylation on samples from various regions and in large areas in the country is suggested.

Availability of data and materials

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

Abbreviations

Treg:

Regulatory T

STAT3:

Signal transducer and activator of transcription 3

MS:

Multiple sclerosis

CNS:

Central nervous system

FOXP3:

Forkhead box P3

Th17:

T helper 17

IL:

Interleukin

RRMS:

Relapsing–remitting multiple sclerosis

NCBI:

National Center for Biotechnology Information

GAPDH:

Glyceraldehyde-3-phosphate dehydrogenase

SD:

Standard deviation

DM:

Demethylation

RT:

Reverse transcriptase

qRT:

Quantitative reverse transcriptase

SEM:

Standard error of mean

References

  1. Klineova S, Lublin FD. Clinical Course of Multiple Sclerosis. Cold Spring Harb Perspect Med. 2018. https://0-doi-org.brum.beds.ac.uk/10.1101/cshperspect.a028928.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Hosseini A, Gharibi T, Marofi F, Babaloo Z, Baradaran B. CTLA-4: From mechanism to autoimmune therapy. Int Immunopharmacol. 2020;80:106221. https://0-doi-org.brum.beds.ac.uk/10.1016/j.intimp.2020.106221.

    Article  CAS  PubMed  Google Scholar 

  3. Kaskow BJ, Baecher-Allan C. Effector T Cells in Multiple Sclerosis. Cold Spring Harb Perspect Med. 2018. https://0-doi-org.brum.beds.ac.uk/10.1101/cshperspect.a029025.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Sakaguchi S. Naturally arising CD4+ regulatory T cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol. 2004;22:531–62.

    Article  CAS  Google Scholar 

  5. Xing Y, Hogquist KA. T-cell tolerance: central and peripheral. Cold Spring Harb Perspect Biol. 2012. https://0-doi-org.brum.beds.ac.uk/10.1101/cshperspect.a006957.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Bhandoola A, Tai X, Eckhaus M, Auchincloss H, Mason K, Rubin SA, et al. Peripheral expression of self-MHC-II influences the reactivity and self-tolerance of mature CD4(+) T cells: evidence from a lymphopenic T cell model. Immunity. 2002;17(4):425–36. https://0-doi-org.brum.beds.ac.uk/10.1016/s1074-7613(02)00417-x.

    Article  CAS  PubMed  Google Scholar 

  7. Piccirillo CA, Thornton AM. Cornerstone of peripheral tolerance: naturally occurring CD4+ CD25+ regulatory T cells. Trends Immunol. 2004;25(7):374–80.

    Article  CAS  Google Scholar 

  8. Schildknecht A, Brauer S, Brenner C, Lahl K, Schild H, Sparwasser T, et al. FoxP3+ regulatory T cells essentially contribute to peripheral CD8+ T-cell tolerance induced by steady-state dendritic cells. Proc Natl Acad Sci. 2010;107(1):199–203.

    Article  CAS  Google Scholar 

  9. Ziegler SF, Buckner JH. FOXP3 and the regulation of Treg/Th17 differentiation. Microbes Infect. 2009;11(5):594–8. https://0-doi-org.brum.beds.ac.uk/10.1016/j.micinf.2009.04.002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Rudensky AY. Regulatory T cells and Foxp3. Immunol Rev. 2011;241(1):260–8. https://0-doi-org.brum.beds.ac.uk/10.1111/j.1600-065X.2011.01018.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhang X, Izikson L, Liu L, Weiner HL. Activation of CD25+ CD4+ regulatory T cells by oral antigen administration. J Immunol. 2001;167(8):4245–53.

    Article  CAS  Google Scholar 

  12. Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, et al. Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med. 2003;198(12):1875–86. https://0-doi-org.brum.beds.ac.uk/10.1084/jem.20030152.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Liang S, Alard P, Zhao Y, Parnell S, Clark SL, Kosiewicz MM. Conversion of CD4+ CD25− cells into CD4+ CD25+ regulatory T cells in vivo requires B7 costimulation, but not the thymus. J Exp Med. 2005;201(1):127–37.

    Article  CAS  Google Scholar 

  14. Taams LS, Vukmanovic-Stejic M, Smith J, Dunne PJ, Fletcher JM, Plunkett FJ, et al. Antigen-specific T cell suppression by human CD4+ CD25+ regulatory T cells. Eur J Immunol. 2002;32(6):1621–30.

    Article  CAS  Google Scholar 

  15. Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, et al. B7/CD28 costimulation is essential for the homeostasis of the CD4+ CD25+ immunoregulatory T cells that control autoimmune diabetes. Immunity. 2000;12(4):431–40.

    Article  CAS  Google Scholar 

  16. Tang Q, Henriksen KJ, Boden EK, Tooley AJ, Ye J, Subudhi SK, et al. Cutting edge: CD28 controls peripheral homeostasis of CD4+ CD25+ regulatory T cells. J Immunol. 2003;171(7):3348–52.

    Article  CAS  Google Scholar 

  17. Baecher-Allan C, Hafler DA. Human regulatory T cells and their role in autoimmune disease. Immunol Rev. 2006;212(1):203–16.

    Article  CAS  Google Scholar 

  18. Tai X, Cowan M, Feigenbaum L, Singer A. CD28 costimulation of developing thymocytes induces Foxp3 expression and regulatory T cell differentiation independently of interleukin 2. Nat Immunol. 2005;6(2):152–62. https://0-doi-org.brum.beds.ac.uk/10.1038/ni1160.

    Article  CAS  PubMed  Google Scholar 

  19. Alvarado-Sanchez B, Hernandez-Castro B, Portales-Perez D, Baranda L, Layseca-Espinosa E, Abud-Mendoza C, et al. Regulatory T cells in patients with systemic lupus erythematosus. J Autoimmun. 2006;27(2):110–8. https://0-doi-org.brum.beds.ac.uk/10.1016/j.jaut.2006.06.005.

    Article  CAS  PubMed  Google Scholar 

  20. Haas J, Fritzsching B, Trübswetter P, Korporal M, Milkova L, Fritz B, et al. Prevalence of newly generated naive regulatory T cells (Treg) is critical for Treg suppressive function and determines Treg dysfunction in multiple sclerosis. J Immunol. 2007;179(2):1322–30.

    Article  CAS  Google Scholar 

  21. Mai J, Wang H, Yang XF. T helper 17 cells interplay with CD4+ CD25highFoxp3+ Tregs in regulation of inflammations and autoimmune diseases. Front Biosci. 2010;15:986.

    Article  CAS  Google Scholar 

  22. Hosseini A, Dolati S, Hashemi V, Abdollahpour-Alitappeh M, Yousefi M. Regulatory T and T helper 17 cells: their roles in preeclampsia. J Cell Physiol. 2018;233(9):6561–73.

    Article  CAS  Google Scholar 

  23. Jhun J, Lee J, Byun J-K, Kim E-K, Woo J-W, Lee J-H, et al. Red ginseng extract ameliorates autoimmune arthritis via regulation of STAT3 pathway, Th17/Treg balance, and osteoclastogenesis in mice and human. Mediat Inflamm. 2014. https://0-doi-org.brum.beds.ac.uk/10.1155/2014/351856.

    Article  Google Scholar 

  24. Traugott U, Lebon P. Multiple sclerosis: involvement of interferons in lesion pathogenesis. Ann Neurol. 1988;24(2):243–51. https://0-doi-org.brum.beds.ac.uk/10.1002/ana.410240211.

    Article  CAS  PubMed  Google Scholar 

  25. Hofstetter H, Gold R, Hartung HP. Th17 cells in MS and experimental autoimmune encephalomyelitis. Int MS J. 2009;16(1):12–8.

    PubMed  Google Scholar 

  26. Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 cells. Annu Rev Immunol. 2009;27:485–517. https://0-doi-org.brum.beds.ac.uk/10.1146/annurev.immunol.021908.132710.

    Article  CAS  PubMed  Google Scholar 

  27. Pallandre J-R, Brillard E, Créhange G, Radlovic A, Remy-Martin J-P, Saas P, et al. Role of STAT3 in CD4+ CD25+ FOXP3+ regulatory lymphocyte generation: implications in graft-versus-host disease and antitumor immunity. J Immunol. 2007;179(11):7593–604.

    Article  CAS  Google Scholar 

  28. Hosseini A, Gharibi T, Marofi F, Javadian M, Babaloo Z, Baradaran B. Janus kinase inhibitors: a therapeutic strategy for cancer and autoimmune diseases. J Cell Physiol. 2020;239(9):5903–24.

    Article  Google Scholar 

  29. Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D, Watowich SS, et al. STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. J Biol Chem. 2007;282(13):9358–63.

    Article  CAS  Google Scholar 

  30. Liu X, Lee YS, Yu C-R, Egwuagu CE. Loss of STAT3 in CD4+ T cells prevents development of experimental autoimmune diseases. J Immunol. 2008;180(9):6070–6.

    Article  CAS  Google Scholar 

  31. Egwuagu CE. STAT3 in CD4+ T helper cell differentiation and inflammatory diseases. Cytokine. 2009;47(3):149–56. https://0-doi-org.brum.beds.ac.uk/10.1016/j.cyto.2009.07.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Cenit M, Alcina A, Márquez A, Mendoza JL, Diaz-Rubio M, de Las HV, et al. STAT3 locus in inflammatory bowel disease and multiple sclerosis susceptibility. Genes. 2010;11(3):264–8.

    CAS  Google Scholar 

  33. Jakkula E, Leppä V, Sulonen A-M, Varilo T, Kallio S, Kemppinen A, et al. Genome-wide association study in a high-risk isolate for multiple sclerosis reveals associated variants in STAT3 gene. Am J Hum Genet. 2010;86(2):285–91.

    Article  CAS  Google Scholar 

  34. Lill CM, Schjeide B-MM, Akkad DA, Blaschke P, Winkelmann A, Gerdes L-A, et al. Independent replication of STAT3 association with multiple sclerosis risk in a large German case–control sample. Neurogenetics. 2012;13(1):83–6.

    Article  CAS  Google Scholar 

  35. Cottrell S, Jung K, Kristiansen G, Eltze E, Semjonow A, Ittmann M, et al. Discovery and validation of 3 novel DNA methylation markers of prostate cancer prognosis. J Urol. 2007;177(5):1753–8.

    Article  CAS  Google Scholar 

  36. Zhuo C, Li Z, Xu Y, Wang Y, Li Q, Peng J, et al. Higher FOXP3-TSDR demethylation rates in adjacent normal tissues in patients with colon cancer were associated with worse survival. Mol Cancer. 2014;13(1):153.

    Article  Google Scholar 

  37. Pfaffl MW. Quantification strategies in real-time polymerase chain reaction. Norfolk: Caister Academic Press; 2012.

    Google Scholar 

  38. Wang Y-C, Zheng L-H, Ma B-A, Zhou Y, Zhang M-H, Zhang D-Z, et al. Clinical value of signal transducers and activators of transcription 3 (STAT3) gene expression in human osteosarcoma. Acta Histochem. 2011;113(4):402–8.

    Article  CAS  Google Scholar 

  39. Milani L, Lundmark A, Kiialainen A, Nordlund J, Flaegstad T, Forestier E, et al. DNA methylation for subtype classification and prediction of treatment outcome in patients with childhood acute lymphoblastic leukemia. Blood J Am Soc Hematol. 2010;115(6):1214–25.

    CAS  Google Scholar 

  40. Wildin RS, Freitas A. IPEX and FOXP3: clinical and research perspectives. J Autoimmun. 2005;25(Suppl):56–62. https://0-doi-org.brum.beds.ac.uk/10.1016/j.jaut.2005.04.008.

    Article  CAS  PubMed  Google Scholar 

  41. Gavin MA, Rasmussen JP, Fontenot JD, Vasta V, Manganiello VC, Beavo JA, et al. Foxp3-dependent programme of regulatory T-cell differentiation. Nature. 2007;445(7129):771–5.

    Article  CAS  Google Scholar 

  42. Lin W, Truong N, Grossman WJ, Haribhai D, Williams CB, Wang J, et al. Allergic dysregulation and hyperimmunoglobulinemia E in Foxp3 mutant mice. J Allergy. 2005;116(5):1106–15.

    CAS  Google Scholar 

  43. Wienke J, Janssen W, Scholman R, Spits H, van Gijn M, Boes M, et al. A novel human STAT3 mutation presents with autoimmunity involving Th17 hyperactivation. Oncotarget. 2015;6(24):20037–42. https://0-doi-org.brum.beds.ac.uk/10.18632/oncotarget.5042.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Milner JD, Vogel TP, Forbes L, Ma CA, Stray-Pedersen A, Niemela JE, et al. Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations. Blood. 2015;125(4):591–9. https://0-doi-org.brum.beds.ac.uk/10.1182/blood-2014-09-602763.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Yu C-R, Lee YS, Mahdi RM, Surendran N, Egwuagu CE. Therapeutic targeting of STAT3 (signal transducers and activators of transcription 3) pathway inhibits experimental autoimmune uveitis. PLoS ONE. 2012;7(1):e29742.

    Article  CAS  Google Scholar 

  46. Egwuagu CE, Larkin IJ. Therapeutic targeting of STAT pathways in CNS autoimmune diseases. JAKSTAT. 2013;2(1):e24134. https://0-doi-org.brum.beds.ac.uk/10.4161/jkst.24134.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This project was provided by the Student Research Committee for Scientific Research, Tabriz University of Medical Sciences, Tabriz, Iran No. 62387.

Funding

This study has been supported and financed by the Student Research Committee for Scientific Research (No. 41090), Tabriz University of Medical Sciences, Tabriz, Iran to AH. The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Conceived and designed the experiments: conceptualization; AH. BB, ZB, and HA. Data curation; AH, SR, SN. Formal analysis; AH, TG, FM, and DS. Investigation; methodology; project administration; AH, VH. Software; NS, MA. Supervision; BB. Roles/writing—original draft; Writing: AH. Review & editing: AH, ZB, TG, NS, FM, VH, HA, MA, SR, SN, DS, and BB. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Behzad Baradaran.

Ethics declarations

Ethics approval and consent to participate

A written informed consent was obtained from each case, and the study protocol was approved by the Ethics Committee of the Tabriz University of Medical Sciences.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

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

Hosseini, A., Babaloo, Z., Gharibi, T. et al. Epigenetic mechanisms shape the underlining expression regulatory mechanisms of the STAT3 in multiple sclerosis disease. BMC Res Notes 13, 568 (2020). https://0-doi-org.brum.beds.ac.uk/10.1186/s13104-020-05427-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13104-020-05427-1

Keywords