Original article

Investigating correlation between gut microbiota and peripheral lymphocyte subsets in patients with systemic lupus erythematosus

  • CEN Xing ,
  • ZHAO Chunmiao ,
  • BU Yujie ,
  • ZHAO Guifang ,
  • YANG Jinhua ,
  • CHEN Junwei
Expand
  • 1. Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan 030001, China
    2. Beijing Jishuitan Hospital, Capital Medical University, Beijing 102208,China
    3. Department of Internal Medicine, Xinghualing District Central Hospital, Taiyuan 030001, China

Received date: 2024-09-09

  Online published: 2025-07-08

Abstract

Objective To investigate the correlation between the distribution of critical differential bacterial populations and lymphocyte subsets in systemic lupus erythematosus (SLE) patients by detecting the abundance changes of intestinal microbiota and lymphocyte subsets. Methods Twenty-three SLE patients and 16 healthy controls (HC) were included. Flow cytometry was used to detect the number of peripheral blood lymphocyte subsets, and 16S rRNA technology was used to detect the diversity and abundance of intestinal microbiota. The correlation between peripheral blood lymphocyte subsets and intestinal microbiota was investigated, and the experimental data were analyzed using SPSS 26.0. Results Compared with HC, the numbers of regulatory T(Treg) cells, initial CD4+T cells, and interferon(IFN)γ-secreting γδT cells in SLE patients were significantly reduced (t=-2.284, P=0.022; t=-2.084, P=0.037; t=-2.370, P=0.017), and follicular regulatory T (Tfr), follicular helper T (Tfh), germinal center B (GC-B)and B lymphocytes showed a downward trend. The relative abundance of Firmicutes in SLE patients was significantly decreased (t=-2.323, P=0.020) and was positively correlated with Tfr (r=0.544, P=0.029) and GC-B cells (r=0.518, P=0.040). The abundance of Bacteroidetes was positively correlated with Tfr cells (r=0.521, P=0.039). Conclusions The peripheral blood lymphocyte subsets and intestinal microbiota levels of SLE patients are significantly different from those of HC, there was a correlation between the intestinal flora and lymphocyte subpopulations in the intestinal tract of SLE patients, especially the Firmicutes and Bacteroidetes are positively correlated with Tfr, Tfh and GC-B cells, suggesting that the structural changes of intestinal microbiota may affect lymphocyte subsets and mediate the progression of the disease.

Cite this article

CEN Xing , ZHAO Chunmiao , BU Yujie , ZHAO Guifang , YANG Jinhua , CHEN Junwei . Investigating correlation between gut microbiota and peripheral lymphocyte subsets in patients with systemic lupus erythematosus[J]. Journal of Internal Medicine Concepts & Practice, 2025 , 20(02) : 140 -145 . DOI: 10.16138/j.1673-6087.2025.02.07

References

[1] Tian J, Zhang D, Yao X, et al. Global epidemiology of systemic lupus erythematosus: a comprehensive systematic analysis and modelling study[J]. Ann Rheum Dis, 2023, 82(3): 351-356.
[2] Chen S, Hu D, Shi X, et al. The relationship between Th1/Th2-type cells and disease activity in patients with systemic lupus erythematosus[J]. Chin Med J (Engl), 2000, 113(10):877-880.
[3] Yang J, Chu Y, Yang X, et al. Th17 and natural Treg cell population dynamics in systemic lupus erythematosus[J]. Arthritis Rheum, 2009, 60(5): 1472-1483.
[4] Deng J, Wei Y, Fonseca VR, et al. T follicular helper cells and T follicular regulatory cells in rheumatic diseases[J]. Nat Rev Rheumatol, 2019, 15(8): 475-490.
[5] Song W, Craft J. T follicular helper cell heterogeneity: time, space, and function[J]. Immunol Rev, 2019, 288(1):85-96.
[6] Hao H, Nakayamada S, Yamagata K, et al. Conversion of T follicular helper cells to T follicular regulatory cells by interleukin-2 through transcriptional regulation in systemic lupus erythematosus[J]. Arthritis Rheumatol, 2021, 73(1): 132-142.
[7] Fan H, Liu F, Dong G, et al. Activation-induced necroptosis contributes to B-cell lymphopenia in active systemic lupus erythematosus[J]. Cell Death Dis, 2014, 5(9): e1416.
[8] Krustev E, Clarke AE, Barber MRW. B cell depletion and inhibition in systemic lupus erythematosus[J]. Expert Rev Clin Immunol, 2023, 19(1): 55-70.
[9] Choi VM, Herrou J, Hecht AL, et al. Activation of bacteroides fragilis toxin by a novel bacterial protease contributes to anaerobic sepsis in mice[J]. Nat Med, 2016, 22(5): 563-567.
[10] Zaiss MM, Joyce Wu HJ, Mauro D, et al. The gut-joint axis in rheumatoid arthritis[J]. Nat Rev Rheumatol, 2021, 17(4): 224-237.
[11] Gao B, Wang Z, Wang K, et al. Relationships among gut microbiota, plasma metabolites, and juvenile idiopathic arthritis: a mediation Mendelian randomization study[J]. Front Microbiol, 2024, 15: 1363776.
[12] Bianchimano P, Britton GJ, Wallach DS, et al. Mining the microbiota to identify gut commensals modulating neuroinflammation in a mouse model of multiple sclerosis[J]. Microbiome, 2022, 10(1): 174.
[13] Wang Y, Wei J, Zhang W, et al. Gut dysbiosis in rheumatic diseases: a systematic review and meta-analysis of 92 observational studies[J]. EBioMedicine, 2022, 80:104055.
[14] Xiang K, Wang P, Xu Z, et al. Causal effects of gut microbiome on systemic lupus erythematosus: a two-sample Mendelian randomization study[J]. Front Immunol, 2021,12: 667097.
[15] Azzouz D, Omarbekova A, Heguy A, et al. Lupus nephritis is linked to disease-activity associated expansions and immunity to a gut commensal[J]. Ann Rheum Dis, 2019, 78(7): 947-956.
[16] Wang T, Sternes PR, Guo XK, et al. Autoimmune diseases exhibit shared alterations in the gut microbiota[J]. Rheumatology (Oxford), 2024, 63(3): 856-865.
[17] Hevia A, Milani C, López P, et al. Intestinal dysbiosis associated with systemic lupus erythematosus[J]. mBio, 2014, 5(5): e01548-14.
[18] Bates NA, Li A, Fan T, et al. Gut commensal segmented filamentous bacteria fine-tune T follicular regulatory cells to modify the severity of systemic autoimmune arthritis[J]. J Immunol, 2021, 206(5): 941-952.
[19] Liang M, Liwen Z, Jianguo S, et al. Fecal microbiota transplantation controls progression of experimental autoimmune hepatitis in mice by modulating the TFR/TFH immune imbalance and intestinal microbiota composition[J]. Front Immunol, 2021, 12: 728723.
[20] Zhang H, Liao X, Sparks JB, et al. Dynamics of gut microbiota in autoimmune lupus[J]. Appl Environ Microbiol, 2014, 80(24): 7551-7560.
[21] Van de Wiele T, Van Praet JT, Marzorati M, et al. How the microbiota shapes rheumatic diseases[J]. Nat Rev Rheumatol, 2016, 12(7): 398-411.
[22] Tyagi AM, Yu M, Darby TM, et al. The microbial metabolite butyrate stimulates bone formation via T regulatory cell-mediated regulation of WNT10B expression[J]. Immunity, 2018, 49(6): 1116-1131.
[23] Faliti CE, Mesina M, Choi J, et al. Interleukin-2-secreting T helper cells promote extra-follicular B cell maturation via intrinsic regulation of a B cell mTOR-AKT-Blimp-1 axis[J]. Immunity, 2024, 57(12): 2772-2789.
[24] Zhang SX, Wang J, Chen JW, et al. The level of peripheral regulatory T cells is linked to changes in gut commensal microflora in patients with systemic lupus erythematosus[J]. Ann Rheum Dis, 2021, 80(11): e177.
[25] Wu R, Wang D, Cheng L, et al. Impaired immune tolerance mediated by reduced Tfr cells in rheumatoid arthritis linked to gut microbiota dysbiosis and altered metabolites[J]. Arthritis Res Ther, 2024, 26(1): 21.
[26] Wu JL, Zou JY, Hu ED, et al. Sodium butyrate ameliorates S100/FCA-induced autoimmune hepatitis through regulation of intestinal tight junction and toll-like receptor 4 signaling pathway[J]. Immunol Lett, 2017, 190:169-176.
[27] Xu MQ, Cao HL, Wang WQ, et al. Fecal microbiota transplantation broadening its application beyond intestinal disorders[J]. World J Gastroenterol, 2015, 21(1):102-111.
[28] Hsu CL, Schnabl B. The gut-liver axis and gut microbiota in health and liver disease[J]. Nat Rev Microbiol, 2023, 21(11): 719-733.
Outlines

/