Atefeh Alaei1,2,3, Mahmoud Mahmoudi1,2, Maryam Sahebari4, Zohreh Vahidi5, Nafiseh Tabasi1, Maryam Rastin1,2*

1Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
2Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
3Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
4Rheumatic Disease Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
5Division of Inflammation and Inflammatory Diseases, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran

Keywords: Cytokines, immunomodulatory, Lactobacillus delbrueckii, Lactobacillus rhamnosus, systemic lupus erythematosus.

Abstract

Objectives: This study aimed to assess the ex vivo impact of Lactobacillus delbrueckii (L. delbrueckii) and Lactobacillus rhamnosus (L. rhamnosus) on inflammatory and anti-inflammatory cytokines as well as their related molecules on the peripheral blood mononuclear cells (PBMCs) of systemic lupus erythematosus (SLE) patients.

Patients and methods: This study was conducted with 20 newly diagnosed SLE patients (18 females, 2 males; mean age: 33.3±12.4 years; range, 18 to 68 years) between September 2017 and September 2018. Extracted PBMCs from each patient were divided into 4 cell groups in our study. Three cell groups act as treatment groups receiving L. rhamnosus (107 CFU/mL), L. delbrueckii (105 CFU/mL) or a mixture of both, and one group act as our untreated control group in the absence of any probiotic agents. All cell groups were cultured in RPMI 1460 medium for 48 h. Then, total RNA was extracted, and cDNA was synthesized.

Results: The gene expression levels of forkhead box P3 (FOXP3), transforming growth factor beta (TGF-β), interleukin (IL)-6, IL-10, and IL-2 were evaluated by a quantitative real-time polymerase chain reaction. The results revealed that expression levels of FOXP3, TGF-β, IL-10, and IL-2 increased and the level of IL-6 decreased in probiotics-receiving groups compared to the control group. Lactobacillus delbrueckii and L. rhamnosus enhanced the expression of regulatory T cell-related molecules such as FOXP3 and IL-2 and also increased the expression of IL-10. These probiotics also reduced the expression of IL-6 as proinflammatory cytokines in the PBMCs of SLE patients.

Conclusion: The results of the present study show that these probiotics could be effective in regulating the balance of cytokine gene expression ex vivo , and due to their beneficial effects, they can be an intriguing option in the production of new complement drugs for SLE

Introduction

Systemic lupus erythematosus (SLE) is a heterogenic, prototypic, and chronic autoimmune disease characterized by breakdowns of immune tolerance and production of autoantibodies against numerous self-antigens.[1] Production of autoantibodies leads to the generation of inflammatory mediators, immune complexes, autoreactive T cells, and vast organ damage.[2] Due to the heterogeneous nature of the disease, there is a broad spectrum of clinical manifestations.[3] The etiology of SLE has not been clarified, but many factors, including genetics, hormones, infections, and environmental factors, play an important role in SLE pathogenesis.[4,5]

Regulatory T cells (Tregs) have a critical role in the maintenance of tolerance and hemostasis in the immune system; thus, reduction and dysfunction of Tregs are associated with the induction and development of autoimmune diseases.[6,7]

Forkhead box P3 (FOXP3) is a transcription factor that is expressed by Tregs and is essential for regulatory functions. Inhibitory cytokines, including transforming growth factor (TGF)-β and interleukin (IL)-10, play critical roles in the maintenance of immune hemostasis. TGF-β cytokine is involved in controlling proliferation and apoptosis.[8,9] Serum levels of TGF-β are reduced in SLE.[10]

Many studies have shown that the levels of IL-10 increased in the plasma of SLE patients. IL-10 is implicated in the apoptosis of cluster of differentiation (CD)4+ and CD8+ T cells. However, IL-10 cytokines contribute to the differentiation and stimulation of B cells. On the other hand, other studies have shown IL-10 is effective in the treatment of SLE disease. [11,12] All these suggest that the function of these cytokines is complicated.[13]

Interleukin-6 is a pleiotropic cytokine in the regulation of the immune system. IL-6 and TGF-β contribute to the differentiation of naïve CD4+ T cells into T helper (Th)17 cells. Thus, IL-6 is correlated with disease activity.[14] Furthermore, studies have indicated that TGF-β and IL-2 are involved in the differentiation of naïve CD4+ T cells into Tregs,[15] and they are also necessary for function, survival, and development of Tregs.[16] Recent studies have reported that in SLE patients, the level of IL-2 is reduced, which could affect FOXP3 expression and cause dysfunction of Tregs.[9,10,17]

Additionally, the high rate of mortality and morbidity of SLE patients is not only due to the pathogenesis of the disease but also the toxic effects of the drugs used. Probiotics are living microorganisms known as commensal microflora. Two studies have reported the beneficial effects of the probiotics on the host health and regulation of immune responses.[18,19] Today, Lactobacillus is known as the most important species of probiotics that can modulate the immune response.[20] Tolerogenic probiotics, through the regulation of responses of inflammatory cells or promotion of regulatory cell responses, act as a modulator of immune system responses to maintain hemostasis. Some tolerogenic probiotics have a positive effect on increasing the number of Tregs, while negatively reducing the proinflammatory and inflammatory cytokines. The use of Lactobacillus has been effective in the treatment and prevention of rheumatoid arthritis, atopic dermatitis, asthma, allergies, rhinitis, and infectious diseases.[17]

Studies have suggested that Lactobacillus rhamnosus has been effective in the treatment and prevention of autoimmune diseases. This probiotic could regulate the balance between Th1/17 cells and Tregs. It also increased IL-10 by increasing CD4+ T cells. The elevated level of IL-10 inhibits excessive inflammatory responses. Lactobacillus delbrueckii also showed a tolerogenic function and played a role in increasing tolerogenic immune responses.[20,21] This probiotic increased antimicrobial peptides and decreased proinflammatory cytokines.[22] The regulatory functions of these probiotics were seen in the production of IL-6, IL-12, and TGF-β cytokines in the colon and IL-6 and TGF-β in the spleen. Those effects also enhanced Tregs in the lymph nodes of the colitis mice model.[23,24]

In this study, we evaluated the modulatory effects of L. rhamnosus and L. delbrueckii on inflammatory and anti-inflammatory cytokines as well as their mediators in the peripheral blood mononuclear cells (PBMCs) of SLE patients ex vivo.

Patients and Methods

The study was conducted at the Mashhad University of Medical Sciences, Department of Immunology between September 2017 and September 2018. Twenty newly diagnosed SLE patients (18 females, 2 males; mean age: 33.3±12.4 years; range, 18 to 68 years) according to the American College of Rheumatology classification criteria (1997 revised criteria) were enrolled in this study. None of the patients had yet received any treatment or medications. SLE patients were sequentially enrolled in the study from the rheumatology center. The following laboratory data were evaluated: C3, C4, C-reactive protein, erythrocyte sedimentation rate, anti-dsDNA (double-strand DNA) titers, ANA (anti-nuclear antibody), hemoglobin, white blood cell count, red blood cell count, and platelet count. To evaluate the effects of two probiotics on cytokine profile in SLE patients, PBMCs were isolated and cultured in the presence or absence of probiotics. Extracted PBMCs from each patient were divided into 4 cell groups in our study; Three cell groups act as treatment groups receiving L. rhamnosus, L. delbrueckii or a mixture of both, and one group act as our untreated control group in the absence of any probiotic agents.

PBMCs Isolation

Peripheral blood samples (10 mL) of SLE patients were collected before any treatment. These samples were collected into EDTA tubes and diluted with phosphate-buffered saline (PBS; pH 7.4) in a 1;1 ratio. PBMCs were separated using Ficoll-Paque (Cedarlane, Toronto, Canada) density gradient centrifugation. After washing twice with PBS, the cells were resuspended at a concentration of 4x106 cells/mL in RPMI 1640 medium (BioSera, London, UK), where RPMI contained 10% heat-inactivated fetal bovine serum (Gibco Inc., Billings, USA).

Preparing probiotics

Lyophilized L. delbrueckii subsp. lactis (PTCC: 1743 [DSM20072]) was prepared by the Iranian Research Organization for Science and Technology and L. rhamnosus GG (ATCC: 9595) was purchased from the Pasteur institute.

Lyophilized L. delbrueckii subsp. lactis and L. rhamnosus were cultured in De Man, Rogosa, and Sharpe (MRS) broth (Biolife, Milano, Italy) under microaerobic conditions for 1 h at 37°C. Next, the probiotics were centrifuged for 2 min at 2,500 rpm. Afterward, the pellet was cultured on MRS agar (Biolife Italiana, Milan, Italy) under microaerobic conditions for 24 h at 37°C.

Thereafter, a specified colony was transferred to the MRS broth medium and placed into the incubator for 2 h at 37°C under an anaerobic condition until reaching the logarithm phase (the log phase time was characterized following previous research in our laboratory). Colony-forming units (CFU/mL) of probiotic-receiving groups were determined. Then, 108 CFU/mL of probiotics were washed twice with PBS solution and suspended in complete RPMI medium, before being added to the cell cultures.

The optimization of probiotic concentration for PBMCs treatment

To find the optimized probiotic concentration, 106 PBMCs of three SLE patients were cultured in the presence of 103, 105, 107 , and 109 CFU/mL of probiotics. The cytotoxicity effects of probiotic doses were determined by MTT assay, and the apoptotic effects of various doses of probiotics on the PBMCs were also obtained using Annexin-V/PI (Abcam, Cambridge, MA, USA) via flowcytometry. Based on the results, we chose the concentration of 107 CFU/mL for L. rhamnosus and 105 CFU/mL for L. delbrueckii in this ex vivo study.

Culture of PBMCs in the presence of probiotics

Patient-isolated PBMCs were cultured (1x106 cells/well) in the presence of L. rhamnosus (107 CFU/mL), L. delbrueckii (105 CFU/mL), a mixture of both probiotics (L. rhamnosus 107 CFU/mL and L. delbrueckii 105 CFU/mL), and in the absence of probiotics, in a total volume of 1 mL RPMI 1640 supplemented with 10% heat-inactivated fetal bovine serum (BioSera, London, UK), 100 IU/mL penicillin, 100 mg/mL streptomycin, and 2 mM L-glutamine. The cells were incubated for 48 h at 37°C in a 5% carbon dioxide humidified incubator.

RNA extraction and cDNA synthesis

Total RNA was extracted from PBMCs (1x106) of patients using Tripure reagent (Roche, Mannheim, Germany) according to the manufacturer’s instructions. Complementary DNA (cDNA) was synthesized using 9 µg of total RNA, random hexamer primer (100 µm), and reverse transcriptase enzyme (10 µm; Fermentas, Vilnius, Lithuania) according to the manufacturer’s protocol. Next, synthesized cDNAs were diluted at a 1:10 ratio for use in real-time polymerase chain reaction (PCR).

Gene expression determined by quantitative real-time PCR

Real-time PCR was performed using the SYBR Green PCR Master Mix (Takara, Shiga, Japan) using a Rotor-Gene 6000 thermal cycler (QIAGEN, Hilden, Germany) with forward and reverse specific primers according to the manufacturer’s instructions (Table 1). In a clean 9 µL tube of SYBR Green Master mix, 1 µL diluted cDNA and 3 µL of RNase-free water mix were added and underwent cycling conditions; 95°C for 15 min, followed by 40 cycles of 15, 30, and 30 sec at 94°C, 55 °C, and 70°C, respectively. Thereafter, 0.5 mM of each specific primer was used for FOXP3, TGF-β, IL-6, IL-10, IL-2, and glyceraldehyde3-phosphate dehydrogenase as an endogenous housekeeping internal control. Quantification of the target gene in the probiotics treated sample was expressed as a fold change compared to the untreated sample.

∆CT T= CT Target - CT HKG

∆CT C= CT Control - CT HKG

∆∆CT= ∆Ct T- ∆Ct C

Fold change= 2-∆∆Ct

Statistical analysis

All results were presented as mean ± SEM (standard error of the mean) and were analyzed with Graph Pad Prism 6 software (Graph Pad Software Inc., San Diego, CA, USA). Statistical analyses for significant differences were performed according to parametric and nonparametric tests. A p-value <0.05 was considered statistically significant.

Results

The results showed that the Systemic Lupus Erythematosus Disease Activity Index score was 21.3±11.7, which indicated a high level of disease activity in the study population. Skin rashes were the most common clinical disorders (75%), followed by joint involvement (65%). ANA was present in 70% of the patients, and 90% of the patients were positive for anti-dsDNA. A low level of C4 (<15 mg/dL) and C3 (<85 mg/dL) was detected in 55% (11/20) of patients. In addition, fever, headache, oral wound, renal diseases, and hematological disorders were reported to a lower extent in patients (Table 2).

The expression level of FOXP3 in all probiotic-receiving groups increased compared to the untreated group, and this increase was significant in the treatment group of L. delbrueckii. The expression amount of TGF-β in L. rhamnosus and the mixture of probiotics-receiving groups increased in the untreated group. Furthermore, the level of TGF-β in the group treated with L. rhamnosus and mixed probiotics (L. rhamnosus and L. delbrueckii) was higher than the L. delbrueckii-receiving group, but these increases were not significant. In the PBMCs groups receiving probiotics, we did not observe any significant decrease in the level of IL-6 compared to the untreated group. The expression level of IL-2 in all probiotic-receiving groups increased in comparison to the untreated group, and this rise was significant in the L. delbrueckii-treated group.

Expression of FOXP3 in the presence of probiotics

Peripheral blood mononuclear cells treatment with L. delbrueckii significantly raised the expression levels of FOXP3 mRNA in PBMCs of the treatment group compared to the untreated group of SLE patients (p=0.02, Figure 1). Additionally, treatment with L. rhamnosus and a mixture of probiotics increased the amount expression of FOXP3 mRNA in PBMCs of the treatment group compared to the untreated group of SLE patients, but it was not significant (p=0.5 and p=0.4, respectively; Figure 1).

Expression of TGF-β in the presence of probiotics

Treatment with L. rhamnosus and a mix of both probiotics increased the expression levels of TGF-β mRNA in PBMCs of the treatment group compared to the untreated group of SLE patients (not significant; p=0.3 and p=0.5, respectively; Figure 2).

Expression of IL-6 in the presence of probiotics

Treatment with L. delbrueckii, L. rhamnosus, and a mixture of both of them reduced the expression levels of IL-6 mRNA in PBMCs of the treatment group compared to the untreated group of SLE patients, but these reductions were not statistically significant (p=0.4, p=0.2, and p=0.3, respectively; Figure 3).

Expression of IL-10 in the presence of probiotics

Treatment with L. delbrueckii significantly enhanced the expression levels of IL-10 mRNA in PBMCs of the treated group compared to the untreated group of SLE patients (p=0.03, Figure 4). Moreover, treatment with L. rhamnosus and a mixture of both probiotics augmented the expression levels of IL-10 mRNA in the treatment group compared to the untreated group of SLE patients (not significant; p=0.8 and p=0.9, respectively; Figure 4).

Expression of IL-2 in the presence of probiotics

The treatment with L. delbrueckii significantly enhanced the expression levels of IL-2 mRNA in PBMCs of the treatment group compared to the untreated group of SLE patients (p=0.05, Figure 5). Additionally, treatment with L. rhamnosus and a mixture of both probiotics elevated the expression level of IL-2 mRNA in PBMCs of treated groups compared to untreated groups of SLE patients (not significant; p=0.8 and p=0.7, respectively; Figure 5).

Discussion

Systemic lupus erythematosus is a chronic inflammatory autoimmune disease where the production of proinflammatory and inflammatory cytokines has a determinative role in pathogenesis and severity of the disease.[25] The immunomodulatory effects of some probiotics, particularly the Lactobacillus species, on T cells and regulatory effects on immune and inflammatory responses have been shown in previous studies.[26,27]

In the present study, we showed the effects of L. rhamnosus and L. delbrueckii on inflammatory and anti-inflammatory cytokines related to Tregs in newly diagnosed SLE patients. Tregs cells express the FOXP3 transcription factor and play an important role in the development of these cells.[28,29] Tregs are also involved in maintaining tolerance and preventing autoimmune diseases.[30] Some studies have reported that the expression level of FOXP3 molecule in patients with active lupus was reduced,[31,32] while the increased expression level of FOXP3 gene has been shown in a Lactobacillus-treated induced colitis murine model.[33] Another study revealed that treatment with Lactobacillus in the experimental autoimmune encephalomyelitis mice model and experimental myasthenia gravis increased in CD4+ CD25+ FOXP3+ Tregs.[30] Researchers reported that treatment with Lactobacillus reuteri in entering the colitis mice model increased the number of Tregs and tolerogenic dendritic cells and decreased the levels of the proinflammatory cytokines.[34] In another study, L. rhamnosus and L. delbrueckii enhanced the expansion of CD4+ CD25+ FOXP3+ Tregs cells in the spleen and also decreased the frequency of Th1 cell and inflammatory cytokines in a pristane model mice.[18,35] Additionally, a mixture of Lactobacillus paracasei and L. reuteri increased Tregs in a SLE NZB/WF1 mice model.[36] Furthermore, the expansion of Tregs occurred in the atopic dermatitis mice model by Lactobacillus acidophilus. [37] Moreover, treatment with L. rhamnosus increased Tregs in allergic diseases.[38] This study showed that L. rhamnosus, L. delbrueckii, and the combination of both increase the expression of the FOXP3 molecule in the culture of SLE patients’ PBMCs.

According to previous studies, TGF-β plays an important role in the differentiation of Tregs and inhibition of inflammation.[16,18] The drop of this cytokine in lupus patients is associated with a reduction of disease activity.[39,40] A study found that the administration of L. rhamnosus increased the level of TGF-β in children with allergic rhinitis.[41] In addition, Nawaz et al.[42] showed that in mice with inflammatory bowel disease Lactobacillus casei treatment increased the level of TGF-β and lowered the production of inflammatory cytokines in the intestinal tissue. Furthermore, treatment with L. delbrueckii led to an increased TGF-β level in the spleen and intestinal tissue of the colitis mice model. Lactobacillus also increased the number of Tregs in the mesenteric lymph nodes of the animal.[23] Contrary to these results, in several studies evaluating the antiallergic effect of L. rhamnosus in an experimental murine model, the antiallergic effects of this probiotic reduced the level of TGF-β. [42] Furthermore, treatment with L. rhamnosus in the allergic disease reduced the levels of TGF-β cytokines in infants.[43] In this study, our results showed the expression level of TGF-β cytokines was higher in patients receiving L. rhamnosus, L. delbrueckii, and their mixture compared to the untreated control group.

The level of IL-6 in patients with lupus is associated with the severity and activity of the disease and the production of anti-dsDNA.[44] Thus, a decrease in inflammatory cytokine is a way to decrease inflammation in SLE patients. Furthermore, a study on a SLENZB/W mice model revealed that mixture strains of L. paracasei and L. reuteri reduced the level of IL-6 and tumor necrosis factor.[36] In the same vein, the treatment of inflammatory bowel disease with L. paracasei decreased the level of IL-6 and reduced the production of inflammatory cytokines.[45] We also showed that the expression level of IL-6 cytokine decreased in all groups receiving probiotics compared to the control group.

IL-10, as an anti-inflammatory cytokine, has an important effect on the commitment and function of Tregs, which can be effective in treating SLE.[40] Lavasani et al.[46] stated in 2010 that the administration of L. rhamnosus increased the level of IL-10 and induction of Tregs in the experimental autoimmune encephalomyelitis mice model. Another study has shown that L. rhamnosus treatment in rotavirus infected BALB/c mice reduced the level of IL-10.[47] In our study, IL-10 cytokine expression level was higher in PBMCs of patients receiving L. rhamnosus compared to the untreated control group.

Additionally, IL-2 cytokine plays an important role in the survival and function of Tregs. This cytokine along with TGF-β can differentiate CD4+T cells to Tregs.[15] Some studies found that the level of IL-2 cytokine in lupus patients was reduced.[48] Treatment with mixture strains of Lactobacillus in the atopic dermatitis mice model increased the level of IL-2.[49] Furthermore, the administration of L. rhamnosus as a solution in healthy people enhanced the level of IL-2.[27] The results of the current study showed that IL-2 cytokine expression level was significantly higher in PBMCs of patients receiving L. delbrueckii and a mixture of both probiotics compared to the untreated control group.

As these probiotic agents affect a wide array of immunoregulator cells and cytokines, understanding their mechanism of action is key to unlocking their full therapeutic potential. Although such studies are lacking for a large portion of probiotic agents, several studies have demonstrated the effects on key pathways and modulators, including nuclear factor kappa B, mitogen-activated protein kinase, Wnt/BAR-1, Toll-like receptor 2, FOXP3, and dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin, altering the inflammatory profile and immune cell populations.[45,50-53]

It is important to note that although the current literature predominantly supports the beneficial effects of probiotics, their safety should be thoroughly investigated before widespread use as a therapeutic agent. Sporadic studies have reported that these agents may cause severe life-threatening infections, specifically in immunocompromised patients. This shows that despite the obvious advantages of probiotic use, universal guidelines for their dosage, handling, administration, and possible severe side effects are desperately needed.[54-57]

In conclusion, the results of our study indicated that the use of probiotics can be helpful to elevate the level of the immunomodulatory molecules, such as FOXP3 (transcription factor of Tregs), TGF-β and IL-10 (cytokines related to Tregs), IL-2 (key cytokine for production and function of Tregs), and to reduce the level of IL-6 (a proinflammatory cytokine) in highly active naïve SLE patients. Note that Tregs function by secreting cytokines and transcription factors, and they are also effective in controlling the disease. Thus, these probiotics could be effective in regulating the balance of gene expression of cytokines profile in vitro, and they can be used to produce new complement drugs due to their beneficial effects. Further in vivo studies are needed to clarify the exact mechanisms, while animal and clinical trial studies are also suggested in order for these results to be confirmed.

Citation: Alaei A, Mahmoudi M, Sahebari M, Vahidi Z, Tabasi N, Rastin M. The effects of Lactobacillus delbrueckii and Lactobacillus rhamnosus on cytokines and their related molecules: An ex vivo study on patients with systemic lupus erythematosus. Arch Rheumatol 2023;38(4):642-652. doi: 10.46497/ ArchRheumatol.2023.9941

Ethics Committee Approval

The study protocol was approved by the Mashhad University of Medical Sciences Ethics Committee (date: 14.06.2017, no: IR.MUMS.sm.REC.1396.158). The study was conducted in accordance with the principles of the Declaration of Helsinki.

Author Contributions

Idea/concept, design: M.R., M.M.; Control/supervision, references and fundings, materials: M.R.; Data collection and/or processing: A.A., M.S., N.T., Z.V.; Analysis and/or interpretation: M.M.; Literature review: A.A., M.R.; Writing the article: A.A.; Critical review: M.R., M.M.

Conflict of Interest

The authors declared no conflicts of interest with respect to the authorship and/or publication of this article.

Financial Disclosure

This work was supported by Mashhad University of Medical Sciences (MUMS) under Grant number 951444.

Data Sharing Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Lleo A, Invernizzi P, Gao B, Podda M, Gershwin ME. Definition of human autoimmunity--autoantibodies versus autoimmune disease. Autoimmun Rev 2010;9:A259-66. doi: 10.1016/j.autrev.2009.12.002. DOI: 10.1016/j.autrev.2009.12.002
  2. Liu J, Zhang X, Cao X. Dendritic cells in systemic lupus erythematosus: From pathogenesis to therapeutic applications. J Autoimmun 2022;132:102856. doi: 10.1016/j.jaut.2022.102856. DOI: 10.1016/j.jaut.2022.102856
  3. Yeoh SA, Dias SS, Isenberg DA. Advances in systemic lupus erythematosus. Medicine 2018;46:84-92. doi: 10.1016/j.mpmed.2017.11.010. DOI: 10.1016/j.mpmed.2017.11.010
  4. Lavi Arab F, Faraji F, Rastin M, Mahmoudi M, Haghmorad D, Nosratabadi R, et al. Assessment of vitamin D effects in lupus- like syndrome induced mice model. Front Immunol 2013. doi: 10.3389/conf. fimmu.2013.02.00234.
  5. Faraji F, Rastin M, Arab FL, Kalantari MR, Rabe SZ, Tabasi N, et al. Effects of 1,25-dihydroxyvitamin D3 on IL-17/IL-23 axis, IFN-g and IL-4 expression in systemic lupus erythematosus induced mice model. Iran J Basic Med Sci 2016;19:374-80.
  6. Wakamatsu E, Omori H, Ohtsuka S, Ogawa S, Green JM, Abe R. Regulatory T cell subsets are differentially dependent on CD28 for their proliferation. Mol Immunol 2018;101:92-101. doi: 10.1016/j. molimm.2018.05.021. DOI: 10.1016/j.molimm.2018.05.021
  7. Chavele KM, Ehrenstein MR. Regulatory T-cells in systemic lupus erythematosus and rheumatoid arthritis. FEBS Lett 2011;585:3603-10. doi: 10.1016/j.febslet.2011.07.043. DOI: 10.1016/j.febslet.2011.07.043
  8. Prado C, de Paz B, López P, Gómez J, RodríguezCarrio J, Suárez A. Relationship between FOXP3 positive populations and cytokine production in systemic lupus erythematosus. Cytokine 2013;61:90- 6. doi: 10.1016/j.cyto.2012.08.033. DOI: 10.1016/j.cyto.2012.08.033
  9. Robinson ES, Werth VP. The role of cytokines in the pathogenesis of cutaneous lupus erythematosus. Cytokina 2015;73(2):326-34. doi: 10.1016/j. cyto.2015.01.031. DOI: 10.1016/j.cyto.2015.01.031
  10. Metawie SA, ElRefai RM, ElAdle SS, Shahin RMH. Transforming growth factor-b1 in systemic lupus erythematosus patients and its relation to organ damage and disease activity. Egypt Rheumatol 2015;37 Suppl 4: S49-54. doi:10.1016/j.ejr.2015.02.001. DOI: 10.1016/j.ejr.2015.02.001
  11. Wu YR, Hsing CH, Chiu CJ, Huang HY, Hsu YH. Roles of IL-1 and IL-10 family cytokines in the progression of systemic lupus erythematosus: Friends or foes? IUBMB Life 2022;74:143-156. doi: 10.1002/ iub.2568. DOI: 10.1002/iub.2568
  12. Geginat J, Vasco M, Gerosa M, Tas SW, Pagani M, Grassi F, et al. IL-10 producing regulatory and helper T-cells in systemic lupus erythematosus. Semin Immunol 2019;44:101330. doi: 10.1016/j. smim.2019.101330. DOI: 10.1016/j.smim.2019.101330
  13. McCarthy EM, Smith S, Lee RZ, Cunnane G, Doran MF, Donnelly S, et al. The association of cytokines with disease activity and damage scores in systemic lupus erythematosus patients. Rheumatology (Oxford) 2014;53:1586-94. doi: 10.1093/rheumatology/ ket428. DOI: 10.1093/rheumatology/ket428
  14. Jacob N, Stohl W. Cytokine disturbances in systemic lupus erythematosus. Arthritis Res Ther 2011;13:228. doi: 10.1186/ar3349. DOI: 10.1186/ar3349
  15. El-Maraghy N, Ghaly MS, Dessouki O, Nasef SI, Metwally L. CD4+CD25-Foxp3+ T cells as a marker of disease activity and organ damage in systemic lupus erythematosus patients. Arch Med Sci 2018;14:1033- 40. doi: 10.5114/aoms.2016.63597. DOI: 10.5114/aoms.2016.63597
  16. Davidson TS, DiPaolo RJ, Andersson J, Shevach EM. Cutting Edge: IL-2 is essential for TGF-betamediated induction of Foxp3+ T regulatory cells. J Immunol 2007;178:4022-6. doi: 10.4049/ jimmunol.178.7.4022. DOI: 10.4049/jimmunol.178.7.4022
  17. Zhao Z, Zhang X, Su L, Xu L, Zheng Y, Sun J. Fine tuning subsets of CD4+ T cells by low-dosage of IL-2 and a new therapeutic strategy for autoimmune diseases. Int Immunopharmacol 2018;56:269-76. doi: 10.1016/j.intimp.2018.01.042. DOI: 10.1016/j.intimp.2018.01.042
  18. Khorasani S, Mahmoudi M, Kalantari MR, Lavi Arab F, Esmaeili SA, Mardani F, et al. Amelioration of regulatory T cells by Lactobacillus delbrueckii and Lactobacillus rhamnosus in pristane-induced lupus mice model. J Cell Physiol 2019;234:9778-86. doi: 10.1002/jcp.27663. DOI: 10.1002/jcp.27663
  19. Calvo-Barreiro L, Eixarch H, Ponce-Alonso M, Castillo M, Lebrón-Galán R, Mestre L, et al. A commercial probiotic induces tolerogenic and reduces pathogenic responses in experimental autoimmune encephalomyelitis. Cells 2020;9:906. doi: 10.3390/ cells9040906. DOI: 10.3390/cells9040906
  20. Wells JM. Immunomodulatory mechanisms of lactobacilli. Microb Cell Fact 2011;10 Suppl 1:S17. doi: 10.1186/1475-2859-10-S1-S17. DOI: 10.1186/1475-2859-10-S1-S17
  21. Chen RC, Xu LM, Du SJ, Huang SS, Wu H, Dong JJ, et al. Lactobacillus rhamnosus GG supernatant promotes intestinal barrier function, balances Treg and TH17 cells and ameliorates hepatic injury in a mouse model of chronic-binge alcohol feeding. Toxicol Lett 2016;241:103-10. doi: 10.1016/j.toxlet.2015.11.019. DOI: 10.1016/j.toxlet.2015.11.019
  22. Moro-García MA, Alonso-Arias R, Baltadjieva M, Fernández Benítez C, Fernández Barrial MA, Díaz Ruisánchez E, et al. Oral supplementation with Lactobacillus delbrueckii subsp. bulgaricus 8481 enhances systemic immunity in elderly subjects. Age (Dordr) 2013;35:1311-26. doi: 10.1007/s11357-012-9434-6. DOI: 10.1007/s11357-012-9434-6
  23. Santos Rocha C, Gomes-Santos AC, Garcias Moreira T, de Azevedo M, Diniz Luerce T, Mariadassou M, et al. Local and systemic immune mechanisms underlying the anti-colitis effects of the dairy bacterium Lactobacillus delbrueckii. PLoS One 2014;9:e85923. doi: 10.1371/journal.pone.0085923. DOI: 10.1371/journal.pone.0085923
  24. Vahidi Z, Samadi M, Mahmoudi M, RezaieYazdi Z, Sahebari M, Tabasi N, et al. Lactobacillus rhamnosus and Lactobacillus delbrueckii ameliorate the expression of miR-155 and miR-181a in SLE patients. J Funct Foods 2018;48:228-33. DOI: 10.1016/j.jff.2018.07.025
  25. La Paglia GMC, Leone MC, Lepri G, Vagelli R, Valentini E, Alunno A, et al. One year in review 2017: Systemic lupus erythematosus. Clin Exp Rheumatol 2017;35:551-61.
  26. Mu Q, Zhang H, Luo XM. SLE: Another autoimmune disorder influenced by microbes and diet? Front Immunol 2015;6:608. doi: 10.3389/ fimmu.2015.00608. DOI: 10.3389/fimmu.2015.00608
  27. Ludwig IS, Broere F, Manurung S, Lambers TT, van der Zee R, van Eden W. Lactobacillus rhamnosus GG-derived soluble mediators modulate adaptive immune cells. Front Immunol 2018;9:1546. doi: 10.3389/fimmu.2018.01546. DOI: 10.3389/fimmu.2018.01546
  28. Yousefi F, Lavi Arab F, Jaafari MR, Rastin M, Tabasi N, Hatamipour M, et al. Immunoregulatory, proliferative and anti-oxidant effects of nanocurcuminoids on adipose-derived mesenchymal stem cells. EXCLI J 2019;18:405-21. doi: 10.17179/excli2019-1366.
  29. Heidari AR, Boroumand-Noughabi S, Nosratabadi R, Lavi Arab F, Tabasi N, Rastin M, et al. Acylated and deacylated quillaja saponin-21 adjuvants have opposite roles when utilized for immunization of C57BL/6 mice model with MOG35-55 peptide. Mult Scler Relat Disord 2019;29:68-82. doi: 10.1016/j. msard.2019.01.025. DOI: 10.1016/j.msard.2019.01.025
  30. Ohl K, Tenbrock K. Regulatory T cells in systemic lupus erythematosus. Eur J Immunol 2015;45:344- 55. doi: 10.1002/eji.201344280. DOI: 10.1002/eji.201344280
  31. Valencia X, Yarboro C, Illei G, Lipsky PE. Deficient CD4+CD25high T regulatory cell function in patients with active systemic lupus erythematosus. J Immunol 2007;178:2579-88. doi: 10.4049/ jimmunol.178.4.2579. DOI: 10.4049/jimmunol.178.4.2579
  32. Suen JL, Chiang BL. CD4(+)FoxP3(+) regulatory T-cells in human systemic lupus erythematosus. J Formos Med Assoc 2012;111:465-70. doi: 10.1016/j. jfma.2012.05.013. DOI: 10.1016/j.jfma.2012.05.013
  33. Di Giacinto C, Marinaro M, Sanchez M, Strober W, Boirivant M. Probiotics ameliorate recurrent Th1-mediated murine colitis by inducing IL-10 and IL-10-dependent TGF-beta-bearing regulatory cells. J Immunol 2005;174:3237-46. doi: 10.4049/ jimmunol.174.6.3237. DOI: 10.4049/jimmunol.174.6.3237
  34. Hoang TK, He B, Wang T, Tran DQ, Rhoads JM, Liu Y. Protective effect of Lactobacillus reuteri DSM 17938 against experimental necrotizing enterocolitis is mediated by Toll-like receptor 2. Am J Physiol Gastrointest Liver Physiol 2018;315:G231-40. doi: 10.1152/ajpgi.00084.2017. DOI: 10.1152/ajpgi.00084.2017
  35. Mardani F, Mahmoudi M, Esmaeili SA, Khorasani S, Tabasi N, Rastin M. In vivo study: Th1-Th17 reduction in pristane-induced systemic lupus erythematosus mice after treatment with tolerogenic Lactobacillus probiotics. J Cell Physiol 2018;234:642-9. doi: 10.1002/jcp.26819. DOI: 10.1002/jcp.26819
  36. Tzang BS, Liu CH, Hsu KC, Chen YH, Huang CY, Hsu TC. Effects of oral Lactobacillus administration on antioxidant activities and CD4+CD25+forkhead box P3 (FoxP3)+ T cells in NZB/W F1 mice. Br J Nutr 2017;118:333-42. doi: 10.1017/ S0007114517002112. DOI: 10.1017/S0007114517002112
  37. Shah MM, Saio M, Yamashita H, Tanaka H, Takami T, Ezaki T, et al. Lactobacillus acidophilus strain L-92 induces CD4(+)CD25(+)Foxp3(+) regulatory T cells and suppresses allergic contact dermatitis. Biol Pharm Bull 2012;35:612-6. doi: 10.1248/ bpb.35.612. DOI: 10.1248/bpb.35.612
  38. Zhang J, Ma JY, Li QH, Su H, Sun X. Lactobacillus rhamnosus GG induced protective effect on allergic airway inflammation is associated with gut microbiota. Cell Immunol 2018;332:77-84. doi: 10.1016/j. cellimm.2018.08.002. DOI: 10.1016/j.cellimm.2018.08.002
  39. Xing Q, Su H, Cui J, Wang B. Role of Treg cells and TGF-b1 in patients with systemic lupus erythematosus: A possible relation with lupus nephritis. Immunol Invest 2012;41:15-27. doi: 10.3109/08820139.2011.578189. DOI: 10.3109/08820139.2011.578189
  40. Okamoto A, Fujio K, Okamura T, Yamamoto K. Regulatory T-cell-associated cytokines in systemic lupus erythematosus. J Biomed Biotechnol 2011;2011:463412. doi: 10.1155/2011/463412. DOI: 10.1155/2011/463412
  41. Jerzynska J, Stelmach W, Balcerak J, Woicka-Kolejwa K, Rychlik B, Blauz A, et al. Effect of Lactobacillus rhamnosus GG and vitamin D supplementation on the immunologic effectiveness of grass-specific sublingual immunotherapy in children with allergy. Allergy Asthma Proc 2016;37:324-34. doi: 10.2500/ aap.2016.37.3958. DOI: 10.2500/aap.2016.37.3958
  42. Nawaz M, Ma C, Basra MA, Wang J, Xu J. Amelioration of ovalbumin induced allergic symptoms in Balb/c mice by potentially probiotic strains of lactobacilli. Benef Microbes 2015;6:669-78. doi: 10.3920/BM2014.0141. DOI: 10.3920/BM2014.0141
  43. Rose MA, Stieglitz F, Köksal A, Schubert R, Schulze J, Zielen S. Efficacy of probiotic Lactobacillus GG on allergic sensitization and asthma in infants at risk. Clin Exp Allergy 2010;40:1398-405. doi: 10.1111/j.1365- 2222.2010.03560.x. DOI: 10.1111/j.1365-2222.2010.03560.x
  44. Lee HM, Sugino H, Nishimoto N. Cytokine networks in systemic lupus erythematosus. J Biomed Biotechnol 2010;2010:676284. doi: 10.1155/2010/676284. DOI: 10.1155/2010/676284
  45. Thakur BK, Saha P, Banik G, Saha DR, Grover S, Batish VK, et al. Live and heat-killed probiotic Lactobacillus casei Lbs2 protects from experimental colitis through Toll-like receptor 2-dependent induction of T-regulatory response. Int Immunopharmacol 2016;36:39-50. doi: 10.1016/j. intimp.2016.03.033. DOI: 10.1016/j.intimp.2016.03.033
  46. Lavasani S, Dzhambazov B, Nouri M, Fåk F, Buske S, Molin G, et al. A novel probiotic mixture exerts a therapeutic effect on experimental autoimmune encephalomyelitis mediated by IL-10 producing regulatory T cells. PLoS One 2010;5:e9009. doi: 10.1371/journal.pone.0009009. DOI: 10.1371/journal.pone.0009009
  47. Jiang Y, Ye L, Cui Y, Yang G, Yang W, Wang J, et al. Effects of Lactobacillus rhamnosus GG on the maturation and differentiation of dendritic cells in rotavirus-infected mice. Benef Microbes 2017;8:645- 56. doi: 10.3920/BM2016.0157. DOI: 10.3920/BM2016.0157
  48. Chun HY, Chung JW, Kim HA, Yun JM, Jeon JY, Ye YM, et al. Cytokine IL-6 and IL-10 as biomarkers in systemic lupus erythematosus. J Clin Immunol 2007;27:461-6. doi: 10.1007/s10875-007-9104-0. DOI: 10.1007/s10875-007-9104-0
  49. Kim HW, Hong R, Choi EY, Yu K, Kim N, Hyeon JY, et al. A probiotic mixture regulates T sell balance and reduces atopic dermatitis symptoms in mice. Front Microbiol 2018;9:2414. doi: 10.3389/ fmicb.2018.02414. DOI: 10.3389/fmicb.2018.02414
  50. Sang Y, Ren J, Aballay A. The transcription factor HLH-26 controls probiotic-mediated protection against intestinal infection through up-regulation of the Wnt/ BAR-1 pathway. PLoS Biol 2022;20:e3001581. doi: 10.1371/journal.pbio.3001581. DOI: 10.1371/journal.pbio.3001581
  51. Smits HH, Engering A, van der Kleij D, de Jong EC, Schipper K, van Capel TM, et al. Selective probiotic bacteria induce IL-10-producing regulatory T cells in vitro by modulating dendritic cell function through dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin. J Allergy Clin Immunol 2005;115:1260-7. doi: 10.1016/j.jaci.2005.03.036. DOI: 10.1016/j.jaci.2005.03.036
  52. Feleszko W, Jaworska J, Rha RD, Steinhausen S, Avagyan A, Jaudszus A, et al. Probiotic-induced suppression of allergic sensitization and airway inflammation is associated with an increase of T regulatory-dependent mechanisms in a murine model of asthma. Clin Exp Allergy 2007;37:498-505. doi: 10.1111/j.1365-2222.2006.02629.x. DOI: 10.1111/j.1365-2222.2006.02629.x
  53. Yu HS, Lee NK, Choi AJ, Choe JS, Bae CH, Paik HD. Anti-inflammatory potential of probiotic strain Weissella cibaria JW15 isolated from kimchi through regulation of NF-kB and MAPKs pathways in LPSinduced RAW 264.7 cells. J Microbiol Biotechnol 2019;29:1022-32. doi: 10.4014/jmb.1903.03014. DOI: 10.4014/jmb.1903.03014
  54. Doron S, Snydman DR. Risk and safety of probiotics. Clin Infect Dis 2015;60 Suppl 2:S129-34. doi: 10.1093/cid/civ085. DOI: 10.1093/cid/civ085
  55. Roy U, Jessani LG, Rudramurthy SM, Gopalakrishnan R, Dutta S, Chakravarty C, et al. Seven cases of Saccharomyces fungaemia related to use of probiotics. Mycoses 2017;60:375-80. doi: 10.1111/myc.12604. DOI: 10.1111/myc.12604
  56. Vanichanan J, Chávez V, Wanger A, De Golovine AM, Vigil KJ. Carbapenem-resistant Lactobacillus intraabdominal infection in a renal transplant recipient with a history of probiotic consumption. Infection 2016;44:793-6. doi: 10.1007/s15010-016-0903-1. DOI: 10.1007/s15010-016-0903-1
  57. Guo X, Yang X, Li Q, Shen X, Zhong H, Yang Y. The microbiota in systemic lupus erythematosus: An update on the potential function of probiotics. Front Pharmacol 2021;12:759095. doi: 10.3389/ fphar.2021.759095. DOI: 10.3389/fphar.2021.759095