CCL26 regulates the proportion of CD4+CD25+FOXP3+ Tregs and the production of inflammatory factors in peripheral blood mononuclear cells following acute ischemic stroke via the STAT5 pathway

Retraction in: /10.3892/etm.2023.11873

  • Authors:
    • Zhiqiang Dong
    • Limei Cao
    • Lan Guo
    • Yuan Hong
    • Jinxiu Cao
    • Xu Chen
  • View Affiliations

  • Published online on: July 24, 2020     https://doi.org/10.3892/etm.2020.9046
  • Pages: 3343-3351
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Acute ischemic stroke (AIS) is the most common type of stroke. Recent studies have found that AIS is closely involved in the immune regulation function of regulatory T cells (Tregs). C‑C motif chemokine ligand 26 (CCL26) is a member of the chemokine family that plays an essential role in cell activation, cell differentiation, lymphocyte homing, and inflammatory and immune responses. The present study aimed to investigate the role of CCL26 in the regulation of Tregs in AIS. Peripheral blood mononuclear cells (PBMCs) were incubated with a CCL26‑neutralizing antibody. The proportion of cluster of differentiation (CD)4+CD25+ forkhead box P3 (FOXP3)+ Tregs was increased, and the expression of FOXP3, phosphorylated signal transducer and activator of transcription 5 (p‑STAT5), and that of the immunosuppressive factors, interleukin (IL)‑10 and transforming growth factor (TGF)‑β1, was upregulated. Conversely, the expression of immune‑promoting factors, such as tumor necrosis factor (TNF)‑α and IL‑6 was significantly downregulated. Further experiments using CCL26 recombinant protein‑treated PBMCs revealed a decreased proportion of CD4+CD25+FOXP3+ Tregs and the downregulated expression of FOXP3, p‑STAT5, TGF‑β1 and IL‑10. Moreover, the expression of immunostimulatory factors, such as CX3C chemokine receptor 1, TNF‑α and IL‑6 was significantly upregulated. On the whole, these results demonstrate that CCL26 regulates the proportion of CD4+CD25+FOXP3+ Tregs and the production of inflammatory factors in PBMCs following AIS via the STAT5 pathway.

Introduction

At present, stroke has become the second most leading cause of mortality worldwide and is also one of the chief factors causing long-term disability among middle-aged and older individuals (1,2). There are approximately 17 million newly diagnosed stroke cases each year worldwide. In Western countries, stroke is the most common lethal disease ranked after cancer and acute myocardial infarction, with acute ischemic stroke (AIS) accounting for approximately 80% of all stroke cases (3,4). Recent studies have found that the severity of damage in ischemic stroke cases is closely related to the immune response elicited by the time and extent of ischemia, particularly regulatory T cells (Tregs)-induced immune responses; Tregs are a type of T cells with immunoregulatory function (5,6). Related studies have demonstrated that Tregs are of utmost importance to the regulation of the pathogenesis of immune diseases. Furthermore, studies have demonstrated that the proportion of Tregs in peripheral blood is significantly lower in patients with ischemic stroke than in healthy controls (7,8) and that the decreased proportion of Tregs induces the alleviation of its inhibitory effect on the inflammatory response, which further results in the occurrence of inflammation, finally causing an imbalance of autoimmune inhibition and promoting the occurrence of ischemic stroke.

Chemotactic cytokines are a group of secreted small-molecule proteins with cell chemotaxis that are responsible for cell directionality. At present, >50 types of chemokines have been identified, and they are mainly divided into 4 categories, CXC, CC, C and CX3C. Chemokines play a critical role in cell activation, cell differentiation, immune and inflammatory responses, and lymphocyte homing based on their regulatory functions in leukocyte migration. C-C motif chemokine ligand 26 (CCL26), whose receptor is CCR3, is a typical CC class chemokine. It is also known as macrophage inflammatory protein 4α, and it primarily plays a role in the regulation of eosinophils and T cells (9,10). Studies have confirmed that CCL26 can block the CCL2 response and that specific molecules, such as pro-inflammatory cytokines, including interleukin (IL)-4, tumor necrosis factor (TNF)-α, IL-1β and interferon-γ can promote the synthesis of CCL26 in human monocytes (11-13). Thus, CCL26 is of utmost importance to the regulation of inflammatory responses.

The present study investigated the role of CCL26 in the pathogenesis of AIS by isolating peripheral blood mononuclear cells (PBMCs) from patients with AIS and treating them with a CCL26-neutralizing antibody or recombinant protein. Thereafter, the proportion of Tregs and the expression levels of phosphorylated signal transducer and activator of transcription 5 (p-STAT5), forkhead box P3 (FOXP3), transforming growth factor (TGF)-β1, IL-10, TNF-α and IL-6 were determined.

Materials and methods

Patients

Peripheral blood was collected from 35 patients (20 males and 15 males) with acute ischemic middle cerebral artery stroke who were aged between 54 and 69 years at Shanghai Eighth People's Hospital from 2016 to 2018. The present study also included a control group that comprised 35 healthy subjects (18 males and 17 males) aged between 53 and 66 years. The diagnosis of stroke was made according to the World Health Organization criteria (14). Computed tomography was performed upon admission, and intracerebral hemorrhage and other possible causes of focal neurological symptoms were excluded in each patient. Patients with diabetes were excluded from the study, and other exclusion criteria were as follows: Abnormal urinalysis findings (hematuria, leukocyturia and proteinuria of 1,300 mg/24 h); clinical or laboratory signs or symptoms of complications following stroke; renal or hepatic failure; glomerulonephritis; and known concomitant neoplastic disease. Investigators recorded data on sex, age and the presence of risk factors for stroke (hypertension, ischemic heart disease and cigarette smoking) for each patient. Neurological deficit was estimated using the 58-point Scandinavian stroke scale (15). Blood samples were obtained upon admission, and serum and PBMCs were separated for performing enzyme-linked immunosorbent assay (ELISA) and for the detection of the proportion of Tregs. The present study was approved by the Ethics Committee of Shanghai Eighth People's Hospital, and written informed consent was obtained from each subject.

ELISA Patient serum processing

Serum was dissolved at 37˚C and commercial ELISA kits were used to determine the concentrations of IL-10 (#H009), TGF-β1 (#H034), TNF-α (#H052) and IL-6 (#H007) (all from Nanjing Jiancheng Bio-company). Briefly, 100 µl of serum samples were added to each well, and 8 standards were set to draw the corresponding standard curves. The samples were incubated at 37˚C for 2 h and washed 5 times with phosphate-buffered saline (PBS). Subsequently, 100 µl of enzyme-labeled antibody were added to each group, and the mixture was incubated at 37˚C for 1 h, washed with PBS for 5 times, and incubated again with a chemiluminescent substrate at 37˚C for 30 min. The reaction was terminated, and the absorbance was measured with a microplate reader (7170S, HITA-CHI, Japan) at 490 nm. The absolute concentrations of IL-10, TGF-β1, TNF-α and IL-6 were calculated with reference to the standard curve.

Cell supernatant assay

PBMCs were isolated from peripheral blood samples collected from each subject in tubes containing K2 ethylenediamine tetraacetic acid as the anticoagulant, as previously described (16). They were treated with various concentrations of recombinant human CCL26 (ab243255, Abcam) (Rep CCL26; 0, 5, 25 and 100 ng/ml) or with 200 mg/l anti-CCL26 neutralizing antibody (ab109612, Abcam) in the absence or presence of 1 µM the STAT5 inhibitor for 24 h, STAT5-IN-1 (CAS: 285986-31-4, EMD Millipore). Following treatment, the PBMC culture medium was centrifuged at 1,000 x g/min, and the supernatant was collected to perform ELISA, as mentioned above.

Flow cytometric analysis

PBMCs were treated with various concentrations of anti-CCL26 neutralizing antibody (ab109612, Abcam) (0, 100, 200 and 500 mg/l), Rep CCL26 (0, 5, 25, and 100 ng/ml), or 200 mg/l of anti-CCL26 neutralizing antibody in the absence or presence of 1 µM STAT5-IN-1. Following treatment, the PBMCs were counted, diluted to 1x106 cells/ml with PBS, and then incubated for 30 min at 4˚C in darkness with anti-human cluster of differentiation CD4 (11-0048-42, Ebioscience), anti-human CD25 (17-0257-42, Ebioscience) and anti-human Foxp3 antibodies, according to the instructions provided with the Anti-Human Foxp3 Staining kit (560133; BD Biosciences). Appropriate isotype controls-human IgG1 kappa antibody (ab206198, Abcam) were included in each experiment. Subsequently, the fluorescence intensities were determined using the BD FACSCalibur flow cytometry system (BD Biosciences).

Western blot analysis

Following experimental treatment, total protein was extracted from the PBMCs using radioimmunoprecipitation assay buffer (JRDUN Biotech Co., Shanghai, China). The total protein was quantified using BCA protein assay kit (23227, Thermo Fisher Scientific, Inc.). And protein expression was detected using an enhanced chemiluminescence system (35055, Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Equal amounts of protein (35 µg) were separated on a 12% gel using sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred to nitrocellulose membranes (JRDUN Biotech Co.). After blocking with 5% skimmed milk, the membranes were incubated overnight with anti-CCL26 antibody (ab190669, Abcam) at 4˚C and followed by secondary antibody anti-IgG antibody (A0208; Beyotime Institute of Biotechnology) for 1 h at 25˚C. Anti-GAPDH antibody (ab9485, Abcam) was used as a loading control in all experiments. Anti-CX3C chemokine receptor 1 (CX3CR1) (ab88577, Abcam), anti-CCL26 (ab190669, Abcam), anti-FOXP3 (ab450, Abcam), anti-STAT5 (ab230670, Abcam) and anti-p-STAT5 (ab32364, Abcam) were diluted using a blocking buffer at 1:1,000 into a working solution prior to use in further analysis, and anti-GAPDH was diluted using a blocking buffer at 1:2,500 prior to use in further analysis. Finally, the protein bands were detected by the imaging software of an electrophoresis gel imaging system (Tanon 6600, Tanon Science & Technology Co., Ltd.).

Statistical analysis

Statistical analyses were performed using GraphPad Prism 6.0 and SPSS17.0 statistical software. All experiments were performed in triplicate. Data for flow cytometric analysis were analyzed with FCAP Array v2.0.2 software (BD Biosciences, MN, USA). Comparisons of continuous variables were performed by the Mann-Whitney U test or one-way analysis of variance (ANOVA) followed by Tukey's multiple comparisons test. Comparisons of categorical variables were performed by the Chi-squared test. A P-value <0.05 was considered to indicate a statistically significant difference.

Results

Abnormal CCL26 expression level and proportion of Tregs in patients with AIS

The baseline characteristics and prevalence of risk factors for stroke in the study groups are presented in Table I. No significant differences were observed between patients with AIS and the healthy subjects. The peripheral blood of 35 patients with AIS and 35 healthy subjects was collected, and the expression level of CCL26 in the serum was detected by ELISA. As shown in Fig. 1A, the expression level of CCL26 in serum was significantly higher in patients with AIS than in the healthy subjects. Tregs are an immunosuppressive subset of CD4+ T cells that constitutively express CD25 on their surfaces. The lineage-defining transcription factor, FOXP3, is the most reliable marker of Tregs and is responsible for the maintenance of the function and differentiation of Tregs; therefore, it is possible to define Tregs more strictly as CD4+CD25+FOXP3+ cells (17). Simultaneously, the proportion of CD4+CD25+FOXP3+ Tregs in PBMCs, as revealed by flow cytometric analysis, was found to be significantly lower in 10 patients with AIS than in the healthy subjects (Fig. 1B).

Table I

Characteristics of the patients enrolled in the present study.

Table I

Characteristics of the patients enrolled in the present study.

CharacteristicControls (n=35)Patients with AIS (n=35)
Age, years (means ± SD)c58.2±2.858.8±2.5
Male sex, %a,d51.457.1
Hypertension, %a,d53.971.0
Ischemic heart disease, %a,d32.057.2
Smoking, %a,d17.332.6
Obesity, %a,d17.239.4
Neurological deficit on admission (SSS score)bN/A37 (24-45)
Alberta stroke program early CT score (ASPECTS)bN/A8.7 (7-12)
Glucose (mmol/l)b,c5.0 (4.5-5.8)5.3 (4.7-5.9)
TG (mmol/l)b,c1.2 (0.7-1.5)1.1 (0.9-1.6)
TC (mmol/l)b,c4.9 (3.7-5.4)4.4 (3.7-5.2)
LDL-C (mmol/l)b,c2.5 (2.1-3.0)2.7 (2.1-3.5)
HDL-C (mmol/l)b,c1.1 (0.9-1.3)1.2 (1.0-1.4)
WBC count(109/l)b,c6.9 (5.1-7.1)6.7 (4.9-7.9)

[i] TG, triglyceride; TC, total cholesterol; LDL-C, low-density lipoprotein cholesterol; HDL-C, high-density lipoprotein cholesterol; AIS, acute ischemic stroke; WBC, white blood cell count.

[ii] aPercentages indicative of the number of patients;

[iii] bdata are expressed as the median and interquartile range;

[iv] cno significant differences were found between groups (Mann-Whitney U test);

[v] dno significant differences were found between groups (Chi-squared test).

CCL26-neutralizing antibody affects the expression levels of CCL26 and CX3CR1, and the proportion of Tregs in the PBMCs of patients with AIS

The proportion of Tregs in PBMCs was detected by flow cytometric analysis, and the results revealed that with the increasing concentration of CCL26-neutralizing antibody, the proportion of CD4+CD25+FOXP3+ Tregs exhibited an increasing trend (Fig. 2A). Moreover, the results of western blot analysis revealed that the expression levels of CCL26 and its receptor, CX3CR1, were gradually decreased as the concentration of CCL26-neutralizing antibody increased (Fig. 2B).

STAT5 inhibitor reverses the effects of CCL26-neutralizing antibody in the PBMCs of patients with AIS

Given that activated STAT5 binds to the promoter, FOXP3, to promote the differentiation of Tregs (18), in the present study, the expression level of STAT5 was determined in CCL26-mediated Tregs using 200 µg/l of CCL26-neutralizing antibody and/or 1 µM of the STAT5 inhibitor, STAT5-IN-1, in the PBMCs. The proportion of Tregs in the PBMCs of patients with AIS was measured and compared with those PBMCs without antibody treatment, and the results revealed that the proportion of CD4+CD25+FOXP3+ Tregs in the PBMCs was increased following treatment with CCL26-neutralizing antibody. Conversely, the proportion of CD4+CD25+FOXP3+ Tregs was decreased in the presence of STAT5-IN-1. Following co-treatment with CCL26-neutralizing antibody and STAT5-IN-1, an increasing trend was observed in the proportion of CD4+CD25+FOXP3+ Tregs in the PBMCs; however, this proportion was lower in the PBMCs treated with the CCL26-neutralizing antibody and STAT5-IN-1 than in those treated with the CCL26-neutralizing antibody alone (Fig. 3A).

The expression levels of IL-10, TGF-β1, TNF-α and IL-6 were measured by ELISA, and the results indicated that the expression levels of TGF-β1 and IL-10 were upregulated following treatment with CCL26-neutralizing antibody. On the contrary, the levels IL-6 and TNF-α were downregulated in PBMCs without antibody treatment. However, following treatment with STAT5-IN-1, the expression levels of IL-10 and TGF-β1 were found to be downregulated, whereas those of IL-6 and TNF-α were upregulated. The expression levels of IL-10, TGF-β1, IL-6 and TNF-α were upregulated at varying degrees following co-treatment with the CCL26-neutralizing antibody and STAT5-IN-1 in PBMCs without antibody treatment; however, compared with the PBMCs treated with the CCL26-neutralizing antibody alone, those treated with the CCL26-neutralizing antibody and STAT5-IN-1 were found to have lower expression levels of IL-10 and TGF-β1, and higher expression levels of IL-6 and TNF-α, as shown in Fig. 3B.

Subsequently, the expression levels of p-STAT5, STAT5 and FOXP3 were detected by western blot analysis. Compared with PBMCs without antibody treatment, the expression levels of FOXP3 and the p-STAT5/STAT5 ratio were significantly increased following treatment with CCL26-neutralizing antibody, whereas they were decreased following treatment with STAT5-IN-1 in patients with AIS. When PBMCs were co-treated with the CCL26-neutralizing antibody and STAT5-IN-1, the expression levels of FOXP3 and p-STAT5/STAT5 ratio also exhibited an increasing trend; however, the promoting effect was not so significant compared with that observed in PBMCs treated with the CCL26-neutralizing antibody treatment alone. In addition, no significant changes were observed in the expression level of STAT5 between the experimental group and the control group (Fig. 3C).

CCL26 recombinant protein affects the proportion of Tregs and the expression levels of several proteins in PBMCs of patients with AIS in a dose-dependent manner

After the PBMCs were treated with gradient concentrations of CCL26 recombinant protein at 0, 5, 25 and 100 ng/ml, the proportion of CD4+CD25+FOXP3+ Tregs was determined and found to gradually decrease with the increasing concentration of the CCL26 recombinant protein (Fig. 4A).

The expression levels of TGF-β1 and IL-10 also decreased gradually as the concentration of CCL26 recombinant protein increased. However, the expression levels of TNF-α and IL-6 increased gradually as the concentration of CCL26 recombinant protein increased (Fig. 4B).

In addition, the expression levels of CCL26, CX3CR1, FOXP3, STAT5 and p-STAT5 were detected by western blot analysis. The expression levels of CCL26 and CX3CR1 were increased with an increasing concentration of CCL26 recombinant protein. The expression levels of FOXP3 and p-STAT5/STAT5 ratio were decreased with an increasing concentration of CCL26 recombinant protein, and no marked differences were noted in the expression level of STAT5 between the experimental group and the control group (Fig. 4C).

Discussion

Although a number of clinical treatment and prevention measures have been reported for acute ischemic stroke, the outcomes have been unsatisfactory, indicating that the critical internal pathogenesis of AIS is far from being fully clarified (19,20). Studies have found that the infiltration of inflammatory cells and the secretion of the corresponding immune factors play an essential role in acute ischemic brain injury (21-23). Tregs have two primary functions, immune response and immunosuppression; they can secrete a variety of anti-inflammatory cytokines and play a key role in inflammation inhibition in immune regulation (8,24). At present, CCL26 is the most selective and active chemotactic agent, which is mainly expressed on the surface of eosinophils, mast cells and Th2 lymphocytes (25). In certain inflammation processes, CCL26 can even promote eosinophil chemotaxis and the recruitment to the inflammatory foci, further promoting degranulation and releasing eosinophil cationic proteins, as well as other cytotoxic substances, which in turn causes a series of inflammatory cascades and finally results in inflammatory damage to brain tissues (26,27).

In the present study, it was found that the expression level of CCL26 in the serum of patients with AIS was markedly higher compared with that observed in healthy subjects, and that the proportion of CD4+CD25+FOXP3+ Tregs in PBMCs was markedly downregulated. Other studies have demonstrated that the substantial damage caused by AIS is mainly due to the upregulation of inflammatory factors in the damaged area, the activation of local microglia and systemic lymphocytes, and the invasion of granulocytes that increase the damaged area of the brain (28,29). All these phenomena are directly related to the significant decreases observed in the proportion of Tregs and the expression levels of inhibitory inflammatory factors (30). Due to excessive activation of acute pro-inflammatory factors and effector T cells, the proportion of Tregs is relatively decreased and the secretion of TGF-β1, as well as the activation and differentiation of Tregs is inhibited (31); these findings are entirely consistent with the clinical test data of the present study. Therefore, based on these findings, it was hypothesized that CCL26 affects the development of AIS by regulating the proportion and activity of Tregs.

In addition to binding to CCR3, CCL26 also binds to CX3CR1, thereby accumulating not only CX3CR1-expressing cells, such as terminally differentiated CD16+ natural killer cells and CD8+ T cells, but also CCR3-expressing cells, such as Th2 cells, eosinophils and mast cells (32). Similarly, it was found that CCL26 promoted the expression of CX3CR1 in the PBMCs of patients with AIS, suggesting the involvement of CCL26/CX3CR1 in the immune response during AIS. IL-10 and TGF-β induce Treg differentiation, which is critical to immune regulation, whereas IL-6 and TNF-α inhibit Treg differentiation (33,34). In line with the findings of these previous studies, the present study found that CCL26 decreased both the proportion of CD4+CD25+FOXP3+ Tregs in PBMCs, and the expression levels of TGF-β and IL-10, whereas it increased the expression levels of IL-6 and TNF-α. FOXP3 is a key regulator of the development and function of Tregs, and its expression is considered to be T cell-restricted. Indeed, CCL26 decreased not only the proportion of CD4+CD25+FOXP3+ Tregs, but also the expression level of FOXP3 and the activation of STAT5, which binds to the promoter FOXP3 and subsequently drives the active transcription of this locus in Tregs (35). This suggests that CCL26 regulates Tregs by targeting FOXP3 expression via the STAT5 pathway. IL-6 inhibits the upregulation of membrane-bound TGF-β on CD4+ T cells (36), whereas TGF-β inhibits the production of TNF-α, which impairs the differentiation and function of TGF-β-induced Tregs and inhibits the production of IL-10 on CD4+ T cells (37-39). The expression of TGF-β-mediated FOXP3 is cooperatively regulated by STAT5(40). The activation of STAT5 can counteract IL-6-induced T-helper cell differentiation and promote the induction of Treg generation; however, IL-6 also activates STAT5 on CD4+ T cells (41), suggesting a feedback regulation between IL-6 and the STAT5 pathway in T cells. These data suggest that CCL26 regulates Tregs by targeting FOXP3 expression via the TGF-β or IL-6-dependent STAT5 pathway.

In conclusion, the present study demonstrates that CCL26 can decrease the proportion of CD4+CD25+FOXP3+ Tregs and regulates the production of inflammatory factors via inactivating the STAT5 pathway in vitro in patients with AIS, indicating that targeting CCL26 with an antibody, an STAT5 inhibitor, or a combination of both has a broad application prospect as a novel, effective, and safe therapeutic approach for treating AIS.

Acknowledgements

Not applicable.

Funding

The present study was funded by the Project Of Medical Technology of Xuhui District, Shanghai (SHXH201710).

Availability of data and materials

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

Authors' contributions

ZD wrote the manuscript. ZD and LC conceived and designed the study. LG and YH were responsible for the collection and analysis of the experimental data. JC and X fC interpreted the data and drafted the manuscript. LG and JC revised the manuscript critically for important intellectual content. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Shanghai Eighth People's Hospital, China. Patients who participated in the study signed the informed consent and had complete clinical data. Signed written informed consent was obtained from the patients and/or their guardians.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Mikulik R and Wahlgren N: Treatment of acute stroke: An update. J Int Med. 278:145–165. 2015.PubMed/NCBI View Article : Google Scholar

2 

Wang X and Wang Y, Wang C, Zhao X, Xian Y, Wang D, Liu L, Luo Y, Liu G and Wang Y: Association between estimated glomerular filtration rate and clinical outcomes in patients with acute ischaemic stroke: Results from China national stroke registry. Age Ageing. 43:839–845. 2014.PubMed/NCBI View Article : Google Scholar

3 

Powers WJ, Derdeyn CP, Biller J, Coffey CS, Hoh BL, Jauch EC, Johnston KC, Johnston SC, Khalessi AA, Kidwell CS, et al: 2015 American heart association/American stroke association focused update of the 2013 guidelines for the early management of patients with acute ischemic stroke regarding endovascular treatment: A guideline for healthcare professionals from the American heart association/American stroke association. Stroke. 46:3020–3035. 2015.PubMed/NCBI View Article : Google Scholar

4 

Sundström BW, Herlitz J, Hansson PO and Brink P: Comparison of the university hospital and county hospitals in western Sweden to identify potential weak links in the early chain of care for acute stroke: Results of an observational study. BMJ Open. 5(e008228)2015.PubMed/NCBI View Article : Google Scholar

5 

Vidale S, Consoli A, Arnaboldi M and Consoli D: Postischemic inflammation in acute stroke. J Clin Neurol. 13:1–9. 2017.PubMed/NCBI View Article : Google Scholar

6 

Kim JY, Park J, Chang JY, Kim SH and Lee JE: Inflammation after ischemic stroke: The role of leukocytes and glial cells. Exp Neurobiol. 25:241–251. 2016.PubMed/NCBI View Article : Google Scholar

7 

Liesz A and Kleinschnitz C: Regulatory T cells in post-stroke immune homeostasis. Transl Stroke Res. 7:313–321. 2016.PubMed/NCBI View Article : Google Scholar

8 

Pang X and Qian W: Changes in regulatory T-cell levels in acute cerebral ischemia. J Neurol Surg A Cent Eur Neurosurg. 78:374–379. 2017.PubMed/NCBI View Article : Google Scholar

9 

Stubbs VE, Power C and Patel KD: Regulation of eotaxin-3/CCL26 expression in human monocytic cells. Immunology. 130:74–82. 2010.PubMed/NCBI View Article : Google Scholar

10 

Ahmadi Z, Hassanshahi G, Khorramdelazad H, Zainodini N and Koochakzadeh L: An overlook to the characteristics and roles played by eotaxin network in the pathophysiology of food allergies: Allergic asthma and atopic dermatitis. Inflammation. 39:1253–1267. 2016.PubMed/NCBI View Article : Google Scholar

11 

Tong Y, Yang T, Wang J, Zhao T, Wang L, Kang Y, Cheng C and Fan Y: Elevated plasma chemokines for eosinophils in neuromyelitis optica spectrum disorders during remission. Front Neurol. 9(44)2018.PubMed/NCBI View Article : Google Scholar

12 

Yang T, Li Y, Lyu Z, Huang K, Corrigan CJ, Ying S, Wang W and Wang C: Characteristics of proinflammatory cytokines and chemokines in airways of asthmatics: Relationships with disease severity and infiltration of inflammatory cells. Chin Med J (Engl). 130:2033–2040. 2017.PubMed/NCBI View Article : Google Scholar

13 

Liu F, Ding X, Yang Y, Li J, Tang M, Yuan M, Hu A, Zhan Z, Li Z and Lu L: Aqueous humor cytokine profiling in patients with wet AMD. Mol Vis. 22:352–361. 2016.PubMed/NCBI

14 

Stroke-1989. Recommendations on stroke prevention diagnosis and therapy. Report of the WHO task force on stroke and other cerebrovascular disorders. Stroke. 20:1407–1431. 1989.PubMed/NCBI View Article : Google Scholar

15 

Multicenter trial of hemodilution in ischemic stroke-background and study protocol. Scandinavian stroke study group. Stroke. 16:885–890. 1985.

16 

Gonzalez Rivas E, Ximenez C, Nieves-Ramirez ME, Moran Silva P, Partida-Rodriguez O, Hernandez EH, Velázquez LR, Vázquez AS and Nuñez UM: Entamoeba histolytica calreticulin induces the expression of cytokines in peripheral blood mononuclear cells isolated from patients with amebic liver abscess. Front Cell Infect Microbiol. 8(358)2018.PubMed/NCBI View Article : Google Scholar

17 

Mizukami Y, Kono K, Kawaguchi Y, Akaike H, Kamimura K, Sugai H and Fujii H: CCL17 and CCL22 chemokines within tumor microenvironment are related to accumulation of Foxp3+ regulatory T cells in gastric cancer. Int J Cancer. 122:2286–2293. 2008.PubMed/NCBI View Article : Google Scholar

18 

Yang TT, Song SJ, Xue HB, Shi DF, Liu CM and Liu H: Regulatory T cells in the pathogenesis of type 2 diabetes mellitus retinopathy by miR-155. Eur Rev Med Pharmacol Sci. 19:2010–2015. 2015.PubMed/NCBI

19 

Fransen PS, Berkhemer OA, Lingsma HF, Beumer D, van den Berg LA, Yoo AJ, Schonewille WJ, Vos JA, Nederkoorn PJ, Wermer MJH, et al: Time to reperfusion and treatment effect for acute ischemic stroke: A randomized clinical trial. JAMA Neurol. 73:190–196. 2016.PubMed/NCBI View Article : Google Scholar

20 

Touma L, Filion KB, Sterling LH, Atallah R, Windle SB and Eisenberg MJ: Stent retrievers for the treatment of acute ischemic stroke: A systematic review and meta-analysis of randomized clinical trials. JAMA Neurol. 73:275–281. 2016.PubMed/NCBI View Article : Google Scholar

21 

Toptas M, Akkoc I, Savas Y, Uzman S, Toptas Y and Can MM: Novel hematologic inflammatory parameters to predict acute mesenteric ischemia. Blood Coagul Fibrinolysis. 27:127–130. 2016.PubMed/NCBI View Article : Google Scholar

22 

Wang J, Yuan J, Cai Y, Fu S, Li M, Hong H, Lu S and Zhu B: Effects of electroacupuncture on inflammatory response of cardiac muscle tissue in mice with acute myocardial ischemia. Zhongguo Zhen Jiu. 38:5133–5138. 2018.PubMed/NCBI View Article : Google Scholar : (In Chinese).

23 

Parsa H, Faghihi M, Kardar GA and Imani A: Acute sleep deprivation induces cardioprotection against ischemia/reperfusion injury through reducing inflammatory responses: The role of central GABA-A receptors. Gen Physiol Biophys. 37:345–352. 2018.PubMed/NCBI View Article : Google Scholar

24 

Chen G, Tang L, Wei W, Li Z, Li Y, Duan X and Chen H: mTOR regulates neuroprotective effect of immunized CD4+Foxp3+ T cells in optic nerve ischemia. Sci Rep. 6(37805)2016.PubMed/NCBI View Article : Google Scholar

25 

Jeong J, Kim YJ, Yoon SY, Kim YJ, Kim JH, Sohn KY, Kim HJ, Han YH, Chong S and Kim JW: PLAG (1-Palmitoyl-2-Linoleoyl-3-Acetyl-rac-Glycerol) modulates eosinophil chemotaxis by regulating CCL26 expression from epithelial cells. PLoS One. 11(e0151758)2016.PubMed/NCBI View Article : Google Scholar

26 

Kagami S, Saeki H, Komine M, Kakinuma T, Tsunemi Y, Nakamura K, Sasaki K, Asahina A and Tamaki K: Interleukin-4 and interleukin-13 enhance CCL26 production in a human keratinocyte cell line, HaCaT cells. Clin Exp Immunol. 141:459–466. 2005.PubMed/NCBI View Article : Google Scholar

27 

Günther C, Wozel G, Meurer M and Pfeiffer C: Up-regulation of CCL11 and CCL26 is associated with activated eosinophils in bullous pemphigoid. Clin Exp Immunol. 166:145–153. 2011.PubMed/NCBI View Article : Google Scholar

28 

Yi JH, Park SW, Kapadia R and Vemuganti R: Role of transcription factors in mediating post-ischemic cerebral inflammation and brain damage. Neurochem Int. 50:1014–1027. 2007.PubMed/NCBI View Article : Google Scholar

29 

Tureyen K, Brooks N, Bowen K, Svaren J and Vemuganti R: Transcription factor early growth response-1 induction mediates inflammatory gene expression and brain damage following transient focal ischemia. J Neurochem. 105:1313–1324. 2008.PubMed/NCBI View Article : Google Scholar

30 

Wu H, Li P, Shao N, Ma J, Ji M, Sun X, Ma D and Ji C: Aberrant expression of Treg-associated cytokine IL-35 along with IL-10 and TGF-β in acute myeloid leukemia. Oncol Lett. 3:1119–1123. 2012.PubMed/NCBI View Article : Google Scholar

31 

Beal AM, Ramos-Hernández N, Riling CR, Nowelsky EA and Oliver PM: TGF-β induces the expression of the adaptor Ndfip1 to silence IL-4 production during iTreg cell differentiation. Nat Immunol. 13:77–85. 2011.PubMed/NCBI View Article : Google Scholar

32 

Shou J, Peng J, Zhao Z, Huang X, Li H, Li L, Gao X, Xing Y and Liu H: CCL26 and CCR3 are associated with the acute inflammatory response in the CNS in experimental autoimmune encephalomyelitis. J Neuroimmunol. 333(576967)2019.PubMed/NCBI View Article : Google Scholar

33 

Costa VS, Mattana TC and da Silva ME: Unregulated IL-23/IL-17 immune response in autoimmune diseases. Diabetes Res Clin Pract. 88:222–226. 2010.PubMed/NCBI View Article : Google Scholar

34 

Xu G, Liu G, Xiong S, Liu H, Chen X and Zheng B: The histone methyltransferase Smyd2 is a negative regulator of macrophage activation by suppressing interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) production. J Biol Chem. 290:5414–5423. 2015.PubMed/NCBI View Article : Google Scholar

35 

Lu L, Barbi J and Pan F: The regulation of immune tolerance by FOXP3. Nat Rev Immunol. 17:703–717. 2017.PubMed/NCBI View Article : Google Scholar

36 

Kuhn C, Rezende RM, M'Hamdi H, da Cunha AP and Weiner HL: IL-6 inhibits upregulation of membrane-bound TGF-β 1 on CD4+ T cells and blocking IL-6 enhances oral tolerance. J Immunol. 198:1202–1209. 2017.PubMed/NCBI View Article : Google Scholar

37 

Zare-Bidaki M, Assar S, Hakimi H, Abdollahi SH, Nosratabadi R, Kennedy D and Arababadi MK: TGF-β in toxoplasmosis: Friend or foe? Cytokine. 86:29–35. 2016.PubMed/NCBI View Article : Google Scholar

38 

Evans HG, Roostalu U, Walter GJ, Gullick NJ, Frederiksen KS, Roberts CA, Sumner J, Baeten DL, Gerwien JG, Cope AP, et al: TNF-α blockade induces IL-10 expression in human CD4+ T cells. Nat Commun. 5(3199)2014.PubMed/NCBI View Article : Google Scholar

39 

Zhang Q, Cui F, Fang L, Hong J, Zheng B and Zhang JZ: TNF-α impairs differentiation and function of TGF-β-induced treg cells in autoimmune diseases through Akt and Smad3 signaling pathway. J Mol Cell Biol. 5:85–98. 2013.PubMed/NCBI View Article : Google Scholar

40 

Ogawa C, Tone Y, Tsuda M, Peter C, Waldmann H and Tone M: TGF-β-mediated Foxp3 gene expression is cooperatively regulated by Stat5, Creb, and AP-1 through CNS2. J Immunol. 192:475–483. 2014.PubMed/NCBI View Article : Google Scholar

41 

Tormo AJ, Letellier MC, Sharma M, Elson G, Crabe S and Gauchat JF: IL-6 activates STAT5 in T cells. Cytokine. 60:575–582. 2012.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

October-2020
Volume 20 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Dong Z, Cao L, Guo L, Hong Y, Cao J and Chen X: CCL26 regulates the proportion of CD4+CD25+FOXP3+ Tregs and the production of inflammatory factors in peripheral blood mononuclear cells following acute ischemic stroke via the STAT5 pathway Retraction in /10.3892/etm.2023.11873. Exp Ther Med 20: 3343-3351, 2020
APA
Dong, Z., Cao, L., Guo, L., Hong, Y., Cao, J., & Chen, X. (2020). CCL26 regulates the proportion of CD4+CD25+FOXP3+ Tregs and the production of inflammatory factors in peripheral blood mononuclear cells following acute ischemic stroke via the STAT5 pathway Retraction in /10.3892/etm.2023.11873. Experimental and Therapeutic Medicine, 20, 3343-3351. https://doi.org/10.3892/etm.2020.9046
MLA
Dong, Z., Cao, L., Guo, L., Hong, Y., Cao, J., Chen, X."CCL26 regulates the proportion of CD4+CD25+FOXP3+ Tregs and the production of inflammatory factors in peripheral blood mononuclear cells following acute ischemic stroke via the STAT5 pathway Retraction in /10.3892/etm.2023.11873". Experimental and Therapeutic Medicine 20.4 (2020): 3343-3351.
Chicago
Dong, Z., Cao, L., Guo, L., Hong, Y., Cao, J., Chen, X."CCL26 regulates the proportion of CD4+CD25+FOXP3+ Tregs and the production of inflammatory factors in peripheral blood mononuclear cells following acute ischemic stroke via the STAT5 pathway Retraction in /10.3892/etm.2023.11873". Experimental and Therapeutic Medicine 20, no. 4 (2020): 3343-3351. https://doi.org/10.3892/etm.2020.9046