Journal of Peking University (Health Sciences) ›› 2023, Vol. 55 ›› Issue (6): 975-981. doi: 10.19723/j.issn.1671-167X.2023.06.004

Previous Articles     Next Articles

Interferon-α mediating the functional damage of CD56dimCD57+natural killer cells in peripheral blood of systemic lupus erythematosuss

Xiang-ge ZHAO,Jia-qing LIU,Hui-na HUANG,Zhi-min LU,Zi-ran BAI,Xia LI,Jing-jing QI*()   

  1. Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, Liaoning, China
  • Received:2023-07-06 Online:2023-12-18 Published:2023-12-11
  • Contact: Jing-jing QI E-mail:jingjingqi_nju@126.com
  • Supported by:
    the National Natural Science Foundation of China(82201990);the National Natural Science Foundation of China(82071834);the National Natural Science Foundation of China(82271839);Science and Technology Talent Innovation Support Project of Dalian(2022RQ070)

Abstract:

Objective: To investigate the regulatory effect of interferon-α (IFN-α) on the apoptosis and killing function of CD56dimCD57+ natural killer (NK) cells in systemic lupus erythematosus (SLE) patients, and to explore the specific mechanism. Methods: A total of sixty-four newly treated SLE patients and sixteen healthy controls (HC) enrolled in the Second Hospital of Dalian Medical University were selected as the research subjects. And the gene expression levels of molecules related to NK cell-killing function were detected by real-time quantitative polymerase chain reaction. CD56dimCD57+ NK cells were co-cultured with the K562 cells, and the apoptotic K562 cells were labeled with Annexin-Ⅴ and 7-amino-actinomycin D. Peripheral blood mononuclear cells were treated with 20, 40, and 80 μmol/L hydrogen peroxide (H2O2), and treated without H2O2 as control, the expression level of perforin (PRF) was detected by flow cytometry. The concentration of IFN-α in serum was determined by enzyme linked immunosorbent assay. The expression levels of IFN-α receptors (IFNAR) on the surface of CD56dimCD57+ NK cells were detected by flow cytometry, and were represented by mean fluorescence intensity (MFI). CD56dimCD57+ NK cells were treated with 1 000 U/mL IFN-α for 24, 48 and 72 h, and no IFN-α treatment was used as the control, the apoptosis and the expression levels of mitochondrial reactive oxygen species (mtROS) were measured by flow cytometry and represented by MFI. Results: Compared with HC(n=3), the expression levels of PRF1 gene in peripheral blood NK cells of the SLE patients (n=3) were decreased (1.24±0.41 vs. 0.57±0.12, P=0.05). Compared with HC(n=5), the ability of peripheral blood CD56dimCD57+ NK cells in the SLE patients (n=5) to kill K562 cells was significantly decreased (58.61%±10.60% vs. 36.74%±6.27%, P < 0.01). Compared with the control (n=5, 97.51%±1.67%), different concentrations of H2O2 treatment significantly down-regulated the PRF expression levels of CD56dimCD57+ NK cells in a dose-dependent manner, the 20 μmol/L H2O2 PRF was 83.23%±8.48% (n=5, P < 0.05), the 40 μmol/L H2O2 PRF was 79.53%±8.56% (n=5, P < 0.01), the 80 μmol/L H2O2 PRF was 76.67%±7.16% (n=5, P < 0.01). Compared to HC (n=16), the serum IFN-α levels were significantly increased in the SLE patients (n=45) with moderate to high systemic lupus erythematosus disease activity index (SLEDAI≥10) [(55.07±50.36) ng/L vs. (328.2±276.3) ng/L, P < 0.001]. Meanwhile, compared with HC (n=6), IFNAR1 expression in peripheral blood CD56dimCD57+ NK cells of the SLE patients (n=6) were increased (MFI: 292.7±91.9 vs. 483.2±160.3, P < 0.05), and compared with HC (n=6), IFNAR2 expression in peripheral blood CD56dimCD57+ NK cells of the SLE patients (n=7) were increased (MFI: 643.5±113.7 vs. 919.0±246.9, P < 0.05). Compared with control (n=6), the stimulation of IFN-α (n=6) significantly promoted the apoptosis of CD56dimCD57+ NK cells (20.48%±7.01% vs. 37.82%±5.84%, P < 0.05). In addition, compared with the control (n=4, MFI: 1 049±174.5), stimulation of CD56dimCD57+ NK cells with IFN-α at different times significantly promoted the production of mtROS in a time-dependent manner, 48 h MFI was 3 437±1 472 (n=4, P < 0.05), 72 h MFI was 6 495±1 089 (n=4, P < 0.000 1), but there was no significant difference at 24 h of stimulation. Conclusion: High serum IFN-α level in SLE patients may induce apoptosis by promoting mtROS production and inhibit perforin expression, which can down-regulate CD56dimCD57+ NK killing function.

Key words: Systemic lupus erythematosus, Natural killer cells, Interferon-α, Reactive oxygen species, Perforin

CLC Number: 

  • R593.2

Figure 1

Expression level of total NK cell killing functional molecules in peripheral blood of SLE patients HC, healthy control; SLE, systemic lupus erythematosus; NK, natural killer."

Figure 2

Comparison of cytotoxic function of peripheral blood CD56dimCD57+ NK cell to kill K562 cells between HC and SLE patients 7-AAD, 7-amino-actinomycin D; HC, healthy control; SLE, systemic lupus erythematosus; NK, natural killer. **P < 0.01."

Figure 3

Effects of H2O2and NAC on PRF expression in CD56dimCD57+ NK cell A, expression of perforin in CD56dimCD57+NK cells after peripheral blood PBMC of HC were exposed to different concentrations of H2O2 (20, 40 and 80 μmol/L) for 24 h. *P < 0.05; **P < 0.01. B, expression of perforin in CD56dimCD57+NK cells cultured with NAC (5 mmol/L) in peripheral blood of SLE patients for 24 h.**P < 0.01. NAC, N-acetyl cysteine; NK, natural killer; PBMC, peripheral blood mononuclear cell; H2O2, hydrogen peroxide; HC, healthy control; SLE, systemic lupus erythematosus; PRF, perforin."

Figure 4

Level of IFN-α and expression of IFNAR on CD56dimCD57+ NK cell in peripheral blood of HC and SLE patients A, IFN-α levels in peripheral blood serum of HC and SLE; ***P < 0.001. B, the expression level of IFNAR1 on the surface of CD56dimCD57+NK cell in peripheral blood of HC and SLE; *P < 0.05. C, the expression level of IFNAR2 on the surface of CD56dimCD57+NK cell in peripheral blood of HC and SLE. *P < 0.05; IFN-α, interferon-α; SLEDAI, systemic lupus erythematosus disease activity index; HC, healthy control; SLE, systemic lupus erythematosus; IFNAR, interferon-α receptor; NK, natural killer; MFI, mean fluorescence intensity."

Figure 5

Effects of IFN-α on apoptosis and mtROS production of CD56dimCD57+NK cells A, peripheral blood NK cells of HC were sorted and stimulated with IFN-α (1 000 U/mL) for 72 h to detect apoptosis of CD56dimCD57+NK cells, *P < 0.05; B, peripheral blood NK cells of HC were sorted and stimulated with IFN-α (1 000 U/mL) for 24, 48 and 72 h, mtROS expression in CD56dimCD57+NK cells was detected by flow cytometry, *P < 0.05, ****P < 0.000 1.7-AAD, 7-amino-actinomycin D; IFN-α, interferon-α; MFI, mean fluorescence intensity; mtROS, mitochondrial reactive oxygen species; NK, natural killer; HC, healthy control."

1 Humbel M , Bellanger F , Fluder N , et al. Restoration of NK cell cytotoxic function with elotuzumab and daratumumab promotes elimination of circulating plasma cells in patients with SLE[J]. Front Immunol, 2021, 12, 645478.
doi: 10.3389/fimmu.2021.645478
2 Cheng Q , Chen X , Xu J , et al. lncRNA X-inactive-specific transcript is a potential biomarker related to changes in CD4+T cell levels in systemic lupus erythematosus[J]. Rheumatol Autoimmun, 2022, 2 (3): 159- 174.
doi: 10.1002/rai2.12048
3 Lin SJ , Kuo ML , Hsiao HS , et al. Cytotoxic function and cytokine production of natural killer cells and natural killer T-like cells in systemic lupus erythematosis regulation with interleukin-15[J]. Mediators Inflamm, 2019, 2019, 4236562.
4 Sordo-Bahamonde C , Lorenzo-Herrero S , Payer ÁR , et al. Mechanisms of apoptosis resistance to NK cell-mediated cytotoxicity in cancer[J]. Int J Mol Sci, 2020, 21 (10): 3726.
doi: 10.3390/ijms21103726
5 Lorenzo-Herrero S , Sordo-Bahamonde C , Gonzalez S , et al. CD107a degranulation assay to evaluate immune cell antitumor activity[J]. Methods Mol Biol, 2019, 1884, 119- 130.
6 Myers JA , Miller JS . Exploring the NK cell platform for cancer immunotherapy[J]. Nat Rev Clin Oncol, 2021, 18 (2): 85- 100.
doi: 10.1038/s41571-020-0426-7
7 Quatrini L , Della Chiesa M , Sivori S , et al. Human NK cells, their receptors and function[J]. Eur J Immunol, 2021, 51 (7): 1566- 1579.
doi: 10.1002/eji.202049028
8 Kucuksezer UC , Aktas Cetin E , Esen F , et al. The role of natural killer cells in autoimmune diseases[J]. Front Immunol, 2021, 12, 622306.
doi: 10.3389/fimmu.2021.622306
9 Bryceson YT , March ME , Barber DF , et al. Cytolytic granule polarization and degranulation controlled by different receptors in resting NK cells[J]. J Exp Med, 2005, 202 (7): 1001- 1012.
doi: 10.1084/jem.20051143
10 Nielsen CM , White MJ , Goodier MR , et al. Functional significance of CD57 expression on human NK cells and relevance to disease[J]. Front Immunol, 2013, 4, 422.
11 Lu Z , Tian Y , Bai Z , et al. Increased oxidative stress contributes to impaired peripheral CD56dimCD57+ NK cells from patients with systemic lupus erythematosus[J]. Arthritis Res Ther, 2022, 24 (1): 48.
doi: 10.1186/s13075-022-02731-y
12 Banchereau R , Hong S , Cantarel B , et al. Personalized immunomonitoring uncovers molecular networks that stratify lupus patients[J]. Cell, 2016, 165 (3): 551- 565.
doi: 10.1016/j.cell.2016.03.008
13 Li P , Jiang M , Li K , et al. Glutathione peroxidase 4-regulated neutrophil ferroptosis induces systemic autoimmunity[J]. Nat Immunol, 2021, 22 (9): 1107- 1117.
doi: 10.1038/s41590-021-00993-3
14 Furie R , Werth VP , Merola JF , et al. Monoclonal antibody targeting BDCA2 ameliorates skin lesions in systemic lupus erythematosus[J]. J Clin Invest, 2019, 129 (3): 1359- 1371.
doi: 10.1172/JCI124466
15 Chatham WW , Furie R , Saxena A , et al. Long-term safety and efficacy of anifrolumab in adults with systemic lupus erythematosus: Results of a phase Ⅱ open-label extension study[J]. Arthritis Rheumatol, 2021, 73 (5): 816- 825.
doi: 10.1002/art.41598
16 Kirou KA , Mavragani CP , Crow MK . Activation of type I inter-feron in systemic lupus erythematosus[J]. Expert Rev Clin Immunol, 2007, 3 (4): 579- 588.
doi: 10.1586/1744666X.3.4.579
17 Le Bon A , Thompson C , Kamphuis E , et al. Cutting edge: Enhancement of antibody responses through direct stimulation of B and T cells by type I IFN[J]. J Immunol, 2006, 176 (4): 2074- 2078.
doi: 10.4049/jimmunol.176.4.2074
18 Cucak H , Yrlid U , Reizis B , et al. Type Ⅰ interferon signaling in dendritic cells stimulates the development of lymph-node-resident T follicular helper cells[J]. Immunity, 2009, 31 (3): 491- 501.
doi: 10.1016/j.immuni.2009.07.005
19 De Groof A , Ducreux J , Aleva F , et al. STAT3 phosphorylation mediates the stimulatory effects of interferon alpha on B cell dif-ferentiation and activation in SLE[J]. Rheumatology (Oxford), 2020, 59 (3): 668- 677.
20 Hochberg MC . Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus[J]. Arthritis Rheum, 1997, 40 (9): 1725.
21 Li H , Geng L , Cao Z , et al. CD56brightCD16- to CD57+CD56dimCD16+ NK cell ratio discriminates disease activity and renal involvement in patients with systemic lupus erythematosus[J]. Clin Exp Rheumatol, 2023, 41 (9): 1768- 1776.
22 Yang Y , Day J , Souza-Fonseca Guimaraes F , et al. Natural killer cells in inflammatory autoimmune diseases[J]. Clin Transl Immunology, 2021, 10 (2): e1250.
doi: 10.1002/cti2.1250
23 Streltsova MA , Erokhina SA , Kanevskiy LM , et al. Analysis of NK cell clones obtained using interleukin-2 and gene-modified K562 cells revealed the ability of "senescent" NK cells to lose CD57 expression and start expressing NKG2A[J]. PLoS One, 2018, 13 (12): e0208469.
doi: 10.1371/journal.pone.0208469
24 Bahadorian D , Mollazadeh S , Mirazi H , et al. Regulatory NK cells in autoimmune disease[J]. Iran J Basic Med Sci, 2023, 26 (6): 609- 616.
25 Fresneda Alarcon M , McLaren Z , Wright HL . Neutrophils in the pathogenesis of rheumatoid arthritis and systemic lupus erythematosus: Same foe different M.O[J]. Front Immunol, 2021, 12, 649693.
doi: 10.3389/fimmu.2021.649693
26 Shah D , Mahajan N , Sah S , et al. Oxidative stress and its biomarkers in systemic lupus erythematosus[J]. J Biomed Sci, 2014, 21 (1): 23.
doi: 10.1186/1423-0127-21-23
27 Kennel KB , Greten FR . Immune cell-produced ROS and their impact on tumor growth and metastasis[J]. Redox Biol, 2021, 42, 101891.
doi: 10.1016/j.redox.2021.101891
28 Song H , Park H , Kim YS , et al. L-kynurenine-induced apoptosis in human NK cells is mediated by reactive oxygen species[J]. Int Immunopharmacol, 2011, 11 (8): 932- 938.
doi: 10.1016/j.intimp.2011.02.005
29 He H , Wang C , Liu G , et al. Isobavachalcone inhibits acute myeloid leukemia: Potential role for ROS-dependent mitochondrial apoptosis and differentiation[J]. Phytother Res, 2021, 35 (6): 3337- 3350.
doi: 10.1002/ptr.7054
30 Bhattacharya A , Hegazy AN , Deigendesch N , et al. Superoxide dismutase 1 protects hepatocytes from type I interferon-driven oxidative damage[J]. Immunity, 2015, 43 (5): 974- 986.
doi: 10.1016/j.immuni.2015.10.013
31 Tasdogan A , Kumar S , Allies G , et al. DNA damage-induced HSPC malfunction depends on ROS accumulation downstream of IFN-1 signaling and bid mobilization[J]. Cell Stem Cell, 2016, 19 (6): 752- 767.
doi: 10.1016/j.stem.2016.08.007
32 Buang N , Tapeng L , Gray V , et al. Type Ⅰ interferons affect the metabolic fitness of CD8+ T cells from patients with systemic lupus erythematosus[J]. Nat Commun, 2021, 12 (1): 980.
doi: 10.1038/s41467-021-21210-7
[1] Hai-hong YAO,Fan YANG,Su-mei TANG,Xia ZHANG,Jing HE,Yuan JIA. Clinical characteristics and diagnostic indicators of macrophage activation syndrome in patients with systemic lupus erythematosus and adult-onset Still's disease [J]. Journal of Peking University (Health Sciences), 2023, 55(6): 966-974.
[2] Zhi-jun LUO,Jia-jia WU,You SONG,Chun-li MEI,Rong DU. Systemic lupus erythematosus associated macrophage activation syndrome with neuropsychiatric symptoms: A report of 2 cases [J]. Journal of Peking University (Health Sciences), 2023, 55(6): 1111-1117.
[3] Xiu-rui LIANG,Xue-chun SHAN,Jing GUAN,Rui ZHANG,Jing YANG,Yi ZHANG,Jia-qi JIN,Yu-xin ZHANG,Fan XU,Ji-hua FU. Role of hyperglycemia-induced 5-hydroxytryptamine degradation of hepatic stellate cells in hepatic inflammation and fibrosis induced by type 2 diabetes mellitus [J]. Journal of Peking University (Health Sciences), 2022, 54(6): 1141-1150.
[4] Min LI,Lin-qing HOU,Yue-bo JIN,Jing HE. Clinical and immunological characteristics of systemic lupus erythematosus with retinopathy [J]. Journal of Peking University (Health Sciences), 2022, 54(6): 1106-1111.
[5] Miao SHAO,Hui-fang GUO,Ling-yan LEI,Qing ZHAO,Yan-jie DING,Jin LIN,Rui WU,Feng YU,Yu-cui LI,Hua-li MIAO,Li-yun ZHANG,Yan DU,Rui-ying JIAO,Li-xia PANG,Li LONG,Zhan-guo LI,Ru LI. A multicenter study on the tolerance of intravenous low-dose cyclophosphamide in systemic lupus erythematosus [J]. Journal of Peking University (Health Sciences), 2022, 54(6): 1112-1116.
[6] Lin-qi ZHANG,Jing ZHAO,Hong-yan WANG,Zong-yi WANG,Ying-ni LI,Ji-yang TANG,Si-ying LI,Jin-feng QU,Ming-wei ZHAO. Relationship between anti-ENO1 antibody and systemic lupus erythematosus patients with retinopathy [J]. Journal of Peking University (Health Sciences), 2022, 54(6): 1099-1105.
[7] Jian-mei ZOU,Li-jun WU,Cai-nan LUO,Ya-mei SHI,Xue WU. Relationship of serum 25- hydroxy vitamin D and systemic lupus erythematosus [J]. Journal of Peking University (Health Sciences), 2021, 53(5): 938-941.
[8] MA Xiang-bo,ZHANG Xue-wu,JIA Ru-lin,GAO Ying,LIU Hong-jiang,LIU Yu-fang,LI Ying-ni. Application of lymphocytes test in peripheral blood of patients with systemic sclerosis during the treatment [J]. Journal of Peking University (Health Sciences), 2021, 53(4): 721-727.
[9] XIA Fang-fang,LU Fu-ai,LV Hui-min,YANG Guo-an,LIU Yuan. Clinical characteristics and related factors of systemic lupus erythematosus with interstitial pneumonia [J]. Journal of Peking University (Health Sciences), 2021, 53(2): 266-272.
[10] Dong YAN,Wen-jie ZHENG. Progress in interferon: A treatment of Behcet syndrome [J]. Journal of Peking University (Health Sciences), 2020, 52(6): 1166-1170.
[11] Yan GENG,Bo-rui LI,Zhuo-li ZHANG. Musculoskeletal ultrasound findings of symptomatic joints in patients with systemic lupus erythematosus [J]. Journal of Peking University(Health Sciences), 2020, 52(1): 163-168.
[12] Yu-hua WANG,Guo-hua ZHANG,Ling-ling ZHANG,Jun-li LUO,Lan GAO. Adrenal hemorrhage in a patient with systemic lupus erythematosus [J]. Journal of Peking University(Health Sciences), 2019, 51(6): 1178-1181.
[13] Ying-ni LI,Xiao-hong XIANG,Jing ZHAO,Yun LI,Feng SUN,Hong-yan WANG,Ru-lin JIA,Fan-lei HU. Significance of anti-carbamylated fibrinogen antibodies in systemic lupus erythematosus [J]. Journal of Peking University(Health Sciences), 2019, 51(6): 1019-1024.
[14] Yun-bo XIE,Ji-yuan ZHANG,Mei-ling DU,Fan-ping MENG,Jun-liang FU,Li-min LIU,Song-shan WANG,Rui QU,Fang LIAN,Fei QIAO,Yang-liu CHEN,Ying-ying GAO,Ruo-nan XU,Ming SHI,Fu-sheng WANG. Efficacy and peripheral immunity analysis of allogeneic natural killer cells therapy in patients with hepatocellular carcinoma [J]. Journal of Peking University(Health Sciences), 2019, 51(3): 591-595.
[15] Fan YANG,Yun-shan ZHOU,Yuan JIA. Systemic lupus erythematosus with acquired hemophilia A: a case report [J]. Journal of Peking University(Health Sciences), 2018, 50(6): 1108-1111.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed   
No Suggested Reading articles found!