1
|
ElFeky DS, Omar NM, Shaker OG, Abdelrahman W, Gheita TA, Nada MG. Circulatory microRNAs and proinflammatory cytokines as predictors of lupus nephritis. Front Immunol 2024; 15:1449296. [PMID: 39464895 PMCID: PMC11502402 DOI: 10.3389/fimmu.2024.1449296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/25/2024] [Indexed: 10/29/2024] Open
Abstract
Introduction Lupus nephritis (LN) is one of the most prevalent severe organ manifestations of systemic lupus erythematosus (SLE), impacting 70% of SLE patients. MicroRNAs (miRNAs), are small non-coding RNA molecules which influence the expression of approximately one-third of human genes after the process of transcription. Dysregulation of miRNAs was documented in numerous disorders, including SLE and LN. Cytokines are the orchestrators of the immune response in autoimmune diseases. Our study aims to explore the variation in the levels of circulating miRNAs and proinflammatory cytokines as potential diagnostic biomarkers among LN and SLE patients without LN in comparison to controls. Methods The study involved 20 LN patients, 20 SLE patients without LN, and 10 healthy controls. Serum levels of IL-12 and IL-21 in addition to miR-124, miR-146a, miR-199a, and miR-21 were assessed using the enzyme-linked immunosorbent assay (ELISA) for cytokines and quantitative real-time PCR for miRNAs. Results A significant downregulation in miR-124 (p<0.001) and a significant overexpression of miR-146a (p=0.005) were found in SLE patients without LN in comparison to controls. In comparison to SLE patients without LN and the control group, miR-199a, miR-21, and miR-146a were significantly upregulated in LN patients (p=<0.001) with high diagnostic values of these miRNAs in discriminating LN from SLE patients without LN according to Receiver operating curve (ROC) analysis. Logistic regression analysis revealed that only miR-199a is an independent predictor of LN (OR 1.69; 95% CI: 1.1-2.6). The expression of miR-124 was reduced in LN patients in comparison to the control but increased in LN patients in comparison to SLE patients without LN. However, there was no statistically significant difference in either scenario. In comparison to both SLE patients without LN and controls, LN patients exhibited the highest serum levels of IL-12 and IL-21, with no statistically significant difference. Regression analysis revealed that only miR-146a was associated with creatinine levels and SLEDAI score (p= 0.009 and 0.03, respectively), while miR-124 was associated with hemoglobin level (p=0.03). Conclusion MiR-199a is an independent predictor for LN and might be used as a diagnostic biomarker for this disease. MiR-146a might play an important role in LN pathophysiology.
Collapse
Affiliation(s)
- Dalia Saad ElFeky
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Cairo
University, Cairo, Egypt
| | - Noha Mohamed Omar
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Cairo
University, Cairo, Egypt
| | - Olfat Gamil Shaker
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Walaa Abdelrahman
- Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Tamer A. Gheita
- Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mona Gamal Nada
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Cairo
University, Cairo, Egypt
| |
Collapse
|
2
|
Tang YY, Wang DC, Chen YY, Xu WD, Huang AF. Th1-related transcription factors and cytokines in systemic lupus erythematosus. Front Immunol 2023; 14:1305590. [PMID: 38164134 PMCID: PMC10757975 DOI: 10.3389/fimmu.2023.1305590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an inflammatory disorder related to immunity dysfunction. The Th1 cell family including Th1 cells, transcription factor T-bet, and related cytokines IFNγ, TNFα, IL-2, IL-18, TGF-β, and IL-12 have been widely discussed in autoimmunity, such as SLE. In this review, we will comprehensively discuss the expression profile of the Th1 cell family in both SLE patients and animal models and clarify how the family members are involved in lupus development. Interestingly, T-bet-related age-associated B cells (ABCs) and low-dose IL-2 treatment in lupus were emergently discussed as well. Collection of the evidence will better understand the roles of the Th1 cell family in lupus pathogenesis, especially targeting IL-2 in lupus.
Collapse
Affiliation(s)
- Yang-Yang Tang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Da-Cheng Wang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - You-Yue Chen
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
3
|
Fang Z, Sun H, Wang Y, Sun Z, Yin M. Discovery of WD-890: A novel allosteric TYK2 inhibitor for the treatment of multiple autoimmune diseases. Biomed Pharmacother 2023; 167:115611. [PMID: 37778274 DOI: 10.1016/j.biopha.2023.115611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023] Open
Abstract
Tyrosine kinase 2 (TYK2) as a member of Janus kinase (JAK) family, mainly mediates the signaling of type I interferons (IFN), interleukin-12 (IL-12) and interleukin-23 (IL-23), which has become an attractive target for treatment of immune and inflammatory diseases. However, the development of selective TYK2 inhibitors is challenging due to the high homology of the catalytic kinase domain among the JAK family members. Here, we report a novel and potent allosteric inhibitor, WD-890, which binds to the pseudokinase domain of TYK2 with high selectivity and inhibits its function. We accomplished a series of preclinical studies to demonstrate the therapeutic efficacy of WD-890 in four animal models: systemic lupus erythematosus (SLE), psoriasis, psoriatic arthritis (PsA), and inflammatory bowel disease (IBD). The pharmacokinetic and toxicology results further indicate that WD-890 has favorable absorption, distribution, metabolism, and excretion (ADME) properties and tolerable toxicity. In conclusion, our study shows that WD-890 could be a promising oral TYK2 inhibitor for future treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Zhiqin Fang
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongyin Sun
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
| | - Yutong Wang
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhenliang Sun
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, China.
| | - Mingzhu Yin
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center (CRC), Medical Pathology Center (MPC), Cancer Early Detection and Treatment Center (CEDTC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China; Translational Medicine Research Center (TMRC), School of Medicine Chongqing University, Shapingba, Chongqing, China.
| |
Collapse
|
4
|
Spinelli FR, Berti R, Farina G, Ceccarelli F, Conti F, Crescioli C. Exercise-induced modulation of Interferon-signature: a therapeutic route toward management of Systemic Lupus Erythematosus. Autoimmun Rev 2023; 22:103412. [PMID: 37597604 DOI: 10.1016/j.autrev.2023.103412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Systemic Lupus Erythematosus (SLE) is a multisystemic autoimmune disorder characterized by flares-ups/remissions with a complex clinical picture related to disease severity and organ/tissue injury, which, if left untreated, may result in permanent damage. Enhanced fatigue and pain perception, worsened quality of life (QoL) and outcome are constant, albeit symptoms may differ. An aberrant SLE immunoprofiling, note as "interferon (IFN)α-signature", is acknowledged to break immunotolerance. Recently, a deregulated "IFNγ-signature" is suggested to silently precede/trigger IFNα profile before clinical manifestations. IFNα- and IFNγ-over-signaling merge in cytokine/chemokine overexpression exacerbating autoimmunity. Remission achievement and QoL improvement are the main goals. The current therapy (i.e., corticosteroids, immunosuppressants) aims to downregulate immune over-response. Exercise could be a safe treatment due to its ever-emerging ability to shape and re-balance immune system without harmful side-effects; in addition, it improves cardiorespiratory capacity and musculoskeletal strength/power, usually impaired in SLE. Nevertheless, exercise is not yet included in SLE care plans. Furthermore, due to the fear to worsening pain/fatigue, SLE subjects experience kinesiophobia and sedentary lifestyle, worsening physical health. Training SLE patients to exercise is mandatory to fight inactive behavior and ameliorate health. This review aims to focus the attention on the role of exercise as a non-pharmacological therapy in SLE, considering its ability to mitigate IFN-signature and rebalance (auto)immune response. To this purpose, the significance of IFNα- and IFNγ-signaling in SLE etiopathogenesis will be addressed first and discussed thereafter as biotarget of exercise. Comments are addressed on the need to make aware all SLE care professional figures to promote exercise for health patients.
Collapse
Affiliation(s)
- Francesca Romana Spinelli
- Sapienza Università di Roma, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Roma, Italy
| | - Riccardo Berti
- University of Rome Foro Italico, Department of Movement, Human and Health Sciences, Rome, Italy
| | - Gabriele Farina
- University of Rome Foro Italico, Department of Movement, Human and Health Sciences, Rome, Italy
| | - Fulvia Ceccarelli
- Sapienza Università di Roma, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Roma, Italy
| | - Fabrizio Conti
- Sapienza Università di Roma, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Roma, Italy
| | - Clara Crescioli
- University of Rome Foro Italico, Department of Movement, Human and Health Sciences, Rome, Italy.
| |
Collapse
|
5
|
Cho JM, Koh JH, Kim SG, Lee S, Kim Y, Cho S, Kim K, Kim YC, Han SS, Lee H, Lee JP, Joo KW, Lim CS, Kim YS, Kim DK, Park S. Mendelian randomization uncovers a protective effect of interleukin-1 receptor antagonist on kidney function. Commun Biol 2023; 6:722. [PMID: 37452175 PMCID: PMC10349143 DOI: 10.1038/s42003-023-05091-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023] Open
Abstract
Interleukins (ILs), key cytokine family of inflammatory response, are closely associated with kidney function. However, the causal effect of various ILs on kidney function needs further investigation. Here we show two-sample summary-level Mendelian randomization (MR) analysis that examined the causality between serum IL levels and kidney function. Genetic variants with strong association with serum IL levels were obtained from a previous genome-wide association study meta-analysis. Summary-level data for estimated glomerular filtration rate (eGFR) were obtained from CKDGen database. As a main MR analysis, multiplicative random-effects inverse-variance weighted method was performed. Pleiotropy-robust MR analysis, including MR-Egger with bootstrapped error and weighted median methods, were also implemented. We tested the causal estimates from nine ILs on eGFR traits. Among the results, higher genetically predicted serum IL-1 receptor antagonist level was significantly associated with higher eGFR values in the meta-analysis of CKDGen and the UK Biobank data. In addition, the result was consistent towards eGFR decline phenotype of the outcome database. Otherwise, nonsignificant association was identified between other genetically predicted ILs and eGFR outcome. These findings support the clinical importance of IL-1 receptor antagonist-associated pathway in relation to kidney function in the general individuals, particularly highlighting the importance of IL-1 receptor antagonist.
Collapse
Affiliation(s)
- Jeong Min Cho
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jung Hun Koh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seong Geun Kim
- Department of Internal Medicine, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - Soojin Lee
- Department of Internal Medicine, Uijeongbu Eulji University Medical Center, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yaerim Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Semin Cho
- Department of Internal Medicine, Chung-Ang University Gwangmyeong Hospital, Gyeonggi-do, Korea
| | - Kwangsoo Kim
- Transdisciplinary Department of Medicine & Advanced Technology, Seoul National University Hospital, Seoul, Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Sehoon Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
6
|
Nawata A, Nakayamada S, Hisano S, Miyazaki Y, Miyamoto T, Shiba E, Hisaoka M, Tanaka Y. Differential expression of IFN-α, IL-12 and BAFF on renal immune cells and its relevance to disease activity and treatment responsiveness in patients with proliferative lupus nephritis. Lupus Sci Med 2023; 10:e000962. [PMID: 37460249 PMCID: PMC10357699 DOI: 10.1136/lupus-2023-000962] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/13/2023] [Indexed: 07/20/2023]
Abstract
OBJECTIVE Since molecularly targeted therapies are emerging for treating lupus nephritis (LN), this study aimed to assess the immunohistochemical findings of the cytokines in renal tissue and their pathological and clinical relevance in LN. METHODS Fifty patients with proliferative LN formed the case group; 5 with LN class II, IgA nephropathy and 10 with idiopathic haematuria were enrolled as controls. Immunohistochemical analysis for CD3, CD20, interferon (IFN)-α, interleukin (IL)-12/p40 and B-cell activating factor (BAFF) was performed by scoring the number of positive cells/area of the cortex. All immunohistochemical investigations were performed on formalin-fixed paraffin-embedded renal tissue. Proliferative LN cases were grouped by the dominant expression of IFN-α, IL-12/p40 and BAFF, and subsequently, clinicopathological features were compared. RESULTS Clinical data of patients with proliferative LN included urine protein creatinine ratio, 2.2 g/gCre; anti-double-stranded DNA antibody, 200.9 IU/mL; total complement activity (CH50), 21.9 U/mL and SLE Disease Activity Index, 19.8 points. Proliferative LN cases, including class III (n=18) and IV (n=32), were classified into three subgroups according to the immunohistochemical score based on the dominancy of IFN-α (n=17), IL-12 (n=16) and BAFF group (n=17) proteins. Hypocomplementaemia and glomerular endocapillary hypercellularity were significantly increased in the IFN-α group, whereas chronic lesions were significantly higher in the IL-12 group (p<0.05). The IFN-α group had a poorer renal prognosis in treatment response after 52 weeks. CONCLUSIONS The immunohistochemistry (IHC) of IFN-α, IL-12 and BAFF for proliferative LN enabled grouping. Especially, the IFN-α and IL-12 groups showed different clinicopathological features and renal prognoses. The results indicated the possibility of stratifying cases according to the IHC of target molecules, which might lead to precision medicine.
Collapse
Affiliation(s)
- Aya Nawata
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
- Department of Pathology and Oncology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shingo Nakayamada
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Satoshi Hisano
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yusuke Miyazaki
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tetsu Miyamoto
- Kidney Center, University of Occupational and Environmental Health Hospital, Kitakyushu, Japan
| | - Eisuke Shiba
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masanori Hisaoka
- Department of Pathology and Oncology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
7
|
Nakayamada S, Tanaka Y. Novel JAK inhibitors under investigation for systemic lupus erythematosus: - where are we now? Expert Opin Investig Drugs 2023; 32:901-908. [PMID: 37753834 DOI: 10.1080/13543784.2023.2264172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/25/2023] [Indexed: 09/28/2023]
Abstract
INTRODUCTION Glucocorticoids and immunosuppressants are used to treat systemic lupus erythematosus (SLE). However, patients with SLE have poor long-term prognoses. This can be attributed to organ damage caused by flare-ups and drug toxicity due to the administration of nonspecific treatment. Therefore, SLE should be treated using therapeutic agents specific to its pathology. Janus kinase (JAK) inhibitors exert multitargeted effects by blocking the signaling of multiple cytokines. The use of JAK inhibitors has been approved to treat several inflammatory autoimmune diseases. Several clinical trials of JAK inhibitors for SLE treatment are ongoing. AREA COVERED This review summarizes the basic and clinical significance of JAK inhibitors for treating SLE and the current status of the development of JAK inhibitors based on recent reports. EXPERT OPINION SLE is a clinically and immunologically heterogeneous disease. Therefore, drugs targeting a single molecule require precision medicine to exert maximal therapeutic efficacy. JAK inhibitors can probably fine-tune the immune network via various mechanisms and broadly regulate complex immune-mediated pathologies in SLE. However, evidence is required to address some safety concerns associated with the use of JAK inhibitors in patients with SLE, including infections (particularly herpes zoster) and thromboembolism (particularly in the presence of concomitant antiphospholipid syndrome).
Collapse
Affiliation(s)
- Shingo Nakayamada
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan
| |
Collapse
|
8
|
Abstract
Systemic lupus erythematosus (SLE) is a typical autoimmune disease with a complex pathogenesis and genetic predisposition. With continued understanding of this disease, it was found that SLE is related to the interferon gene signature. Most studies have emphasized the important role of IFN-α in SLE, but our previous study suggested a nonnegligible role of IFN-γ in SLE. Some scholars previously found that IFN-γ is abnormally elevated as early as before the classification of SLE and before the emergence of autoantibodies and IFN-α. Due to the large overlap between IFN-α and IFN-γ, SLE is mostly characterized by expression of the IFN-α gene after onset. Therefore, the role of IFN-γ in SLE may be underestimated. This article mainly reviews the role of IFN-γ in SLE and focuses on the nonnegligible role of IFN-γ in SLE to gain a more comprehensive understanding of the disease.
Collapse
|
9
|
Longitudinal comparison of IL-6, IL-10, and IL-12 cytokine profiles in adult and childhood-onset systemic lupus erythematosus. J Transl Autoimmun 2022; 5:100158. [PMID: 36683864 PMCID: PMC9846463 DOI: 10.1016/j.jtauto.2022.100158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 01/25/2023] Open
Abstract
Objective To compare the levels of Th1 (IL-12) and Th2 (IL-6 and IL10) cytokines over a two-year period among systemic lupus erythematosus patients with childhood-onset (cSLE), adult-onset (sSLE), and healthy controls, and correlate with their clinical, laboratory, and treatment manifestations. Methods The study included 63 patients with cSLE [57 (90%) women; mean age 19.7 ± 4.3 years (range = 10-29); mean disease duration 7.3 ± 4.2 years (range 2-15)], 67 patients with aSLE [65 (97%) women; mean age of 39.9 ± 11.8 years (range 21-68); disease duration 7.7 ± 3.1 years (range 4-16)], and 40 healthy controls [36 (90%) women; mean age of 29.6 ± 10 years (range 12-49)]. cSLE and aSLE patients were paired by disease duration. Clinical and laboratory manifestations, disease activity (SLEDAI), cumulative damage (SDI), and current drug exposures were evaluated. Symptoms of anxiety and depression were evaluated by the Beck inventory (BAI and BDI, respectively). Th1 (IL-12) and Th2 (IL-6 and IL-10) cytokines were measured by the ELISA test. Data were collected at four different time points (TI, TII, TIII, and TIV) and compared by non-parametric tests. Results IL-6 levels were significantly higher in aSLE patients compared to healthy controls at times I, II, and III (TI p = 0.013, TII p = 0.015, TIII p = 0.004, and TIV p = 0.634). However, no difference was observed between cSLE patients and healthy controls (TI p = 0.223, TII p = 0.613, TIII p = 0.341, and TIV p = 0.977). In addition, no difference was observed between aSLE and cSLE patients (TI p = 0.377, TII p = 0.123, TIII p = 0.105, and TIV p = 0.591). The levels of IL-12 were significantly higher in cSLE patients compared to healthy controls at all time points (TI p = 0.04, TII p < 0.001, TIII p = 0.015, and TIV p = 0.021). aSLE patients showed significantly elevated levels when compared to healthy controls at time III and IV (TI p = 0.752, TII p = 0.827, TIII p = 0.011*, and TIV p < 0.001*). cSLE patients showed significantly higher levels than aSLE patients at times I and II (TI p = 0.07*, TII p < 0.001*, TIII p = 0.998, and TIV p = 0.140). In aSLE patients, IL-6 was associated with headache (p = 0.006), arthritis (p = 0.044), and nephritis (p = 0.012); IL-10 was associated with nephritis (p = 0.043), hypocomplementemia (p = 0.001), and disease activity (p = 0.001); in these patients, IL-12 was associated with alopecia (p = 0.025) and leukopenia (p = 0.044). In cSLE patients, IL-6 was associated with arthritis (p = 0.022) and malar rash (p = 0.012). Conclusion aSLE and cSLE patients with long disease duration present similar levels of cytokines, despite differences in clinical activity patterns over time.
Collapse
|
10
|
Li J, Luo M, Li B, Lou Y, Zhu Y, Bai X, Sun B, Lu X, Luo P. Immunomodulatory Activity of Mesenchymal Stem Cells in Lupus Nephritis: Advances and Applications. Front Immunol 2022; 13:843192. [PMID: 35359961 PMCID: PMC8960601 DOI: 10.3389/fimmu.2022.843192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 02/17/2022] [Indexed: 12/29/2022] Open
Abstract
Lupus nephritis (LN) is a significant cause of various acute and chronic renal diseases, which can eventually lead to end-stage renal disease. The pathogenic mechanisms of LN are characterized by abnormal activation of the immune responses, increased cytokine production, and dysregulation of inflammatory signaling pathways. LN treatment is an important issue in the prevention and treatment of systemic lupus erythematosus. Mesenchymal stem cells (MSCs) have the advantages of immunomodulation, anti-inflammation, and anti-proliferation. These unique properties make MSCs a strong candidate for cell therapy of autoimmune diseases. MSCs can suppress the proliferation of innate and adaptive immune cells, such as natural killer cells (NKs), dendritic cells (DCs), T cells, and B cells. Furthermore, MSCs suppress the functions of various immune cells, such as the cytotoxicity of T cells and NKs, maturation and antibody secretion of B cells, maturation and antigen presentation of DCs, and inhibition of cytokine secretion, such as interleukins (ILs), tumor necrosis factor (TNF), and interferons (IFNs) by a variety of immune cells. MSCs can exert immunomodulatory effects in LN through these immune functions to suppress autoimmunity, improve renal pathology, and restore kidney function in lupus mice and LN patients. Herein, we review the role of immune cells and cytokines in the pathogenesis of LN and the mechanisms involved, as well as the progress of research on the immunomodulatory role of MSCs in LN.
Collapse
Affiliation(s)
- Jicui Li
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Manyu Luo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Bing Li
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Yan Lou
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Yuexin Zhu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Xue Bai
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Baichao Sun
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Xuehong Lu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Spontaneous CD4+ T Cell Activation and Differentiation in Lupus-Prone B6.Nba2 Mice Is IFNAR-Independent. Int J Mol Sci 2022; 23:ijms23020874. [PMID: 35055071 PMCID: PMC8778657 DOI: 10.3390/ijms23020874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 11/21/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by dysregulated T and B lymphocytes. Type I interferons (IFN-I) have been shown to play important pathogenic roles in both SLE patients and mouse models of lupus. Recent studies have shown that B cell intrinsic responses to IFN-I are enough to drive B cell differentiation into autoantibody-secreting memory B cells and plasma cells, although lower levels of residual auto-reactive cells remain present. We speculated that IFN-I stimulation of T cells would similarly drive specific T-cell associated lupus phenotypes including the upregulation of T follicular helper cells and Th17, thereby affecting autoantibody production and the development of glomerulonephritis. Using the B6.Nba2 mouse model of lupus, we evaluated disease parameters in T cell specific IFN-I receptor (IFNAR)-deficient mice (cKO). Surprisingly, all measured CD4+ T cell abnormalities and associated intra-splenic cytokine levels (IFNγ, IL-6, IL-10, IL-17, IL-21) were unchanged and thus independent of IFN-I. In contrast B6.Nba2 cKO mice displayed reduced levels of effector CD8+ T cells and increased levels of Foxp3+ CD8+ regulatory T cells, suggesting that IFN-I induced signaling specifically affecting CD8+ T cells. These data suggest a role for both pathogenic and immunosuppressive CD8+ T cells in Nba2-driven autoimmunity, providing a model to further evaluate the role of these cell subsets during lupus-like disease development in vivo.
Collapse
|
12
|
Fotheringham AK, Solon-Biet SM, Bielefeldt-Ohmann H, McCarthy DA, McMahon AC, Ruohonen K, Li I, Sullivan MA, Whiddett RO, Borg DJ, Cogger VC, Ballard WO, Turner N, Melvin RG, Raubenheimer D, Le Couteur DG, Simpson SJ, Forbes JM. Kidney disease risk factors do not explain impacts of low dietary protein on kidney function and structure. iScience 2021; 24:103308. [PMID: 34820603 PMCID: PMC8602032 DOI: 10.1016/j.isci.2021.103308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/29/2021] [Accepted: 10/15/2021] [Indexed: 02/06/2023] Open
Abstract
The kidneys balance many byproducts of the metabolism of dietary components. Previous studies examining dietary effects on kidney health are generally of short duration and manipulate a single macronutrient. Here, kidney function and structure were examined in C57BL/6J mice randomized to consume one of a spectrum of macronutrient combinations (protein [5%–60%], carbohydrate [20%–75%], and fat [20%–75%]) from weaning to late-middle age (15 months). Individual and interactive impacts of macronutrients on kidney health were modeled. Dietary protein had the greatest influence on kidney function, where chronic low protein intake decreased glomerular filtration rates and kidney mass, whereas it increased kidney immune infiltration and structural injury. Kidney outcomes did not align with cardiometabolic risk factors including glucose intolerance, overweight/obesity, dyslipidemia, and hypertension in mice with chronic low protein consumption. This study highlights that protein intake over a lifespan is an important determinant of kidney function independent of cardiometabolic changes. Chronic high macronutrient intake from any source increases kidney function (GFR) Low protein intake led to greater kidney tubular structural injury and inflammation Lower protein intake decreased kidney mass and glomerular filtration capacity Kidney outcomes did not align with longevity or cardiometabolic outcomes
Collapse
Affiliation(s)
- Amelia K Fotheringham
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane 4072, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane 4067, QLD, Australia
| | - Samantha M Solon-Biet
- Charles Perkins Centre, University of Sydney, Sydney 2006, NSW, Australia.,School of Medical Sciences, University of Sydney, Sydney 2006, NSW, Australia
| | - Helle Bielefeldt-Ohmann
- School of Veterinary Science, University of Queensland, Gatton Campus, Gatton 4343, QLD, Australia.,School of Chemistry & Molecular Biosciences, University of Queensland, Brisbane 4067, QLD, Australia
| | - Domenica A McCarthy
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane 4072, QLD, Australia
| | - Aisling C McMahon
- Charles Perkins Centre, University of Sydney, Sydney 2006, NSW, Australia.,Centre for Education and Research on Aging, and Aging and Alzheimer's Institute, Concord Hospital, Sydney 2139, NSW, Australia.,ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney 2139, NSW, Australia
| | - Kari Ruohonen
- Animal Nutrition and Health, Cargill, Sandnes, Norway
| | - Isaac Li
- Faculty of Medicine, University of Queensland, Brisbane 4067, QLD, Australia
| | - Mitchell A Sullivan
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane 4072, QLD, Australia
| | - Rani O Whiddett
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane 4072, QLD, Australia
| | - Danielle J Borg
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane 4072, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane 4067, QLD, Australia
| | - Victoria C Cogger
- Charles Perkins Centre, University of Sydney, Sydney 2006, NSW, Australia.,Centre for Education and Research on Aging, and Aging and Alzheimer's Institute, Concord Hospital, Sydney 2139, NSW, Australia.,ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney 2139, NSW, Australia
| | - William O Ballard
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney 2052, NSW, Australia
| | - Nigel Turner
- Department of Pharmacology, School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, NSW 2052, Australia
| | - Richard G Melvin
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, Duluth 55812, MN, USA
| | - David Raubenheimer
- Charles Perkins Centre, University of Sydney, Sydney 2006, NSW, Australia.,School of Life and Environmental Sciences, University of Sydney, NSW, Australia
| | - David G Le Couteur
- Charles Perkins Centre, University of Sydney, Sydney 2006, NSW, Australia.,Centre for Education and Research on Aging, and Aging and Alzheimer's Institute, Concord Hospital, Sydney 2139, NSW, Australia.,ANZAC Research Institute, Concord Hospital, University of Sydney, Sydney 2139, NSW, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, University of Sydney, Sydney 2006, NSW, Australia.,School of Life and Environmental Sciences, University of Sydney, NSW, Australia
| | - Josephine M Forbes
- Glycation and Diabetes Complications Group, Mater Research Institute-The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Brisbane 4072, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane 4067, QLD, Australia.,Department of Medicine, University of Melbourne, Heidelberg, VIC 3084, Australia
| |
Collapse
|
13
|
Noor A, Assiri A. A novel computational drug repurposing approach for Systemic Lupus Erythematosus (SLE) treatment using Semantic Web technologies. Saudi J Biol Sci 2021; 28:3886-3892. [PMID: 34220244 PMCID: PMC8241633 DOI: 10.1016/j.sjbs.2021.03.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/20/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022] Open
Affiliation(s)
- Adeeb Noor
- Department of Information Technology, Faculty of Computing and Information Technology, King Abdulaziz University, Jeddah 80221, Saudi Arabia
| | - Abdullah Assiri
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| |
Collapse
|
14
|
Liu C, Lin J, Langevine C, Smith D, Li J, Tokarski JS, Khan J, Ruzanov M, Strnad J, Zupa-Fernandez A, Cheng L, Gillooly KM, Shuster D, Zhang Y, Thankappan A, McIntyre KW, Chaudhry C, Elzinga PA, Chiney M, Chimalakonda A, Lombardo LJ, Macor JE, Carter PH, Burke JR, Weinstein DS. Discovery of BMS-986202: A Clinical Tyk2 Inhibitor that Binds to Tyk2 JH2. J Med Chem 2020; 64:677-694. [PMID: 33370104 DOI: 10.1021/acs.jmedchem.0c01698] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A search for structurally diversified Tyk2 JH2 ligands from 6 (BMS-986165), a pyridazine carboxamide-derived Tyk2 JH2 ligand as a clinical Tyk2 inhibitor currently in late development for the treatment of psoriasis, began with a survey of six-membered heteroaryl groups in place of the N-methyl triazolyl moiety in 6. The X-ray co-crystal structure of an early lead (12) revealed a potential new binding pocket. Exploration of the new pocket resulted in two frontrunners for a clinical candidate. The potential hydrogen bonding interaction with Thr599 in the pocket was achieved with a tertiary amide moiety, confirmed by the X-ray co-crystal structure of 29. When the diversity search was extended to nicotinamides, a single fluorine atom addition was found to significantly enhance the permeability, which directly led to the discovery of 7 (BMS-986202) as a clinical Tyk2 inhibitor that binds to Tyk2 JH2. The preclinical studies of 7, including efficacy studies in mouse models of IL-23-driven acanthosis, anti-CD40-induced colitis, and spontaneous lupus, will also be presented.
Collapse
Affiliation(s)
- Chunjian Liu
- Immunosciences Discovery Chemistry, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - James Lin
- Immunosciences Discovery Chemistry, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Charles Langevine
- Immunosciences Discovery Chemistry, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Daniel Smith
- Department of Discovery Synthesis, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Jianqing Li
- Department of Discovery Synthesis, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - John S Tokarski
- Molecular Structure and Design, Molecular Discovery Technologies, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Javed Khan
- Molecular Structure and Design, Molecular Discovery Technologies, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Max Ruzanov
- Molecular Structure and Design, Molecular Discovery Technologies, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Joann Strnad
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Adriana Zupa-Fernandez
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Lihong Cheng
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Kathleen M Gillooly
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - David Shuster
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Yifan Zhang
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Anil Thankappan
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Kim W McIntyre
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Charu Chaudhry
- Leads Discovery and Optimization, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Paul A Elzinga
- Metabolism and Pharmacokinetic Department, Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Manoj Chiney
- Metabolism and Pharmacokinetic Department, Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Anjaneya Chimalakonda
- Metabolism and Pharmacokinetic Department, Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Louis J Lombardo
- Immunosciences Discovery Chemistry, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - John E Macor
- Immunosciences Discovery Chemistry, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Percy H Carter
- Immunosciences Discovery Chemistry, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - James R Burke
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - David S Weinstein
- Immunosciences Discovery Chemistry, Bristol-Myers Squibb Research & Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| |
Collapse
|
15
|
Cytokines and Transgenic Matrix in Autoimmune Diseases: Similarities and Differences. Biomedicines 2020; 8:biomedicines8120559. [PMID: 33271810 PMCID: PMC7761121 DOI: 10.3390/biomedicines8120559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/16/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases are increasingly recognized as disease entities in which dysregulated cytokines contribute to tissue-specific inflammation. In organ-specific and multiorgan autoimmune diseases, the cytokine profiles show some similarities. Despite these similarities, the cytokines have different roles in the pathogenesis of different diseases. Altered levels or action of cytokines can result from changes in cell signaling. This article describes alterations in the JAK-STAT, TGF-β and NF-κB signaling pathways, which are involved in the pathogenesis of multiple sclerosis and systemic lupus erythematosus. There is a special focus on T cells in preclinical models and in patients afflicted with these chronic inflammatory diseases.
Collapse
|
16
|
Nozaki Y. The Network of Inflammatory Mechanisms in Lupus Nephritis. Front Med (Lausanne) 2020; 7:591724. [PMID: 33240910 PMCID: PMC7677583 DOI: 10.3389/fmed.2020.591724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022] Open
Abstract
Several signaling pathways are involved in the progression of kidney disease in humans and in animal models, and kidney disease is usually due to the sustained activation of these pathways. Some of the best understood pathways are specific proinflammatory cytokine and protein kinase pathways (e.g., protein kinase C and mitogen-activated kinase pathways, which cause cell proliferation and fibrosis and are associated with angiotensin II) and transforming growth factor-beta (TGF-β) signaling pathways (e.g., the TGF-β signaling pathway, which leads to increased fibrosis and kidney scarring. It is thus necessary to continue to advance our knowledge of the pathogenesis and molecular biology of kidney disease and to develop new treatments. This review provides an update of important findings about kidney diseases (including diabetic nephropathy, lupus nephritis, and vasculitis, i.e., vasculitis with antineutrophilic cytoplasmic antibodies). New disease targets, potential pathological pathways, and promising therapeutic approaches from basic science to clinical practice are presented, and the blocking of JAK/STAT and TIM-1/TIM-4 signaling pathways as potential novel therapeutic agents in lupus nephritis is discussed.
Collapse
Affiliation(s)
- Yuji Nozaki
- Department of Hematology and Rheumatology, Faculty of Medicine, Kindai University, Osaka, Japan
| |
Collapse
|
17
|
Elsner RA, Shlomchik MJ. IL-12 Blocks Tfh Cell Differentiation during Salmonella Infection, thereby Contributing to Germinal Center Suppression. Cell Rep 2020; 29:2796-2809.e5. [PMID: 31775046 DOI: 10.1016/j.celrep.2019.10.069] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/21/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022] Open
Abstract
Germinal centers (GC) are crucial for the formation of long-lived humoral immunity. Many pathogens suppress GC, including Salmonella enterica serovar Typhimurium (STm), but the mechanisms driving suppression remain unknown. We report that neither plasmablasts nor STm-specific B cells are required for GC suppression in mice. Rather, we identify that interleukin-12 (IL-12), but not interferon-γ (IFN-γ), directly suppresses T follicular helper (Tfh) cell differentiation of T cells intrinsically. Administering recombinant IL-12 during nitrophenyl-Chicken Gamma Globulin (NP-CGG) immunization also suppresses Tfh cell differentiation and GC B cells, indicating that IL-12 is sufficient to suppress Tfh cell differentiation independent of STm infection. Recombinant IL-12 induces high levels of T-bet, and T-bet is necessary for Tfh cell suppression. Therefore, IL-12 induced during STm infection in mice contributes to GC suppression via suppression of Tfh cell differentiation. More broadly, these data suggest that IL-12 can tailor the proportions of humoral (Tfh cell) and cellular (T helper type 1 [Th1] cell) immunity to the infection, with implications for IL-12 targeting therapies in autoimmunity and vaccination.
Collapse
Affiliation(s)
- Rebecca A Elsner
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Mark J Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15216, USA.
| |
Collapse
|
18
|
Li M, Yang C, Wang Y, Song W, Jia L, Peng X, Zhao R. The Expression of P2X7 Receptor on Th1, Th17, and Regulatory T Cells in Patients with Systemic Lupus Erythematosus or Rheumatoid Arthritis and Its Correlations with Active Disease. THE JOURNAL OF IMMUNOLOGY 2020; 205:1752-1762. [PMID: 32868411 DOI: 10.4049/jimmunol.2000222] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/31/2020] [Indexed: 01/21/2023]
Abstract
P2X7 receptor (P2X7R) is highly expressed on immune cells, triggering the release of cytokines and regulating autoimmune responses. To investigate P2X7R surface expression on T helper (Th) 1, Th17, and regulatory T (Treg) cells in patients with systemic lupus erythematosus (SLE) or rheumatoid arthritis (RA) and correlations with disease activity, 29 SLE and 29 RA patients and 18 healthy controls (HCs) were enrolled. We showed that SLE and RA patients had significantly higher levels of plasma cytokines (IFN-γ, IL-1β, IL-6, IL-17A, and IL-23), frequencies of Th1 and Th17 cells, and expression of P2X7R on Th1 and Th17 than HCs, and the Th17/Treg ratio was significantly increased, whereas Treg cell levels were significantly decreased. The Ca2+ influx increase following BzATP stimulation was significantly higher in CD4+PBMCs from SLE and RA patients than in HCs. Blood levels of shed P2X7R were increased in SLE and RA patients. Furthermore, 28-joint Disease Activity Score and SLE Disease Activity Index score showed negative correlations with Treg cell levels and positive correlations with Th17/Treg ratio and Th17 cell P2X7R expression. Interestingly, Th17 cell P2X7R expression was closely correlated with IL-1β, C-reactive protein, the erythrocyte sedimentation rate, anticyclic citrullinated peptide Abs, albumin, and C4. These data indicate that increased Th17 cell P2X7R expression is functionally and positively related to disease activity and some inflammatory mediators in SLE and RA patients, and P2X7R could be critical in promoting the Th17 immune response and contributing to the complex pathogenesis of SLE and RA.
Collapse
Affiliation(s)
- Mingxuan Li
- School of Laboratory Medicine, Weifang Medical University, Weifang 261053, Shandong, China
| | - Chuanyu Yang
- Department of Blood Transfusion, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong, China; and
| | - Yunhai Wang
- Department of Clinical Laboratory, Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong, China
| | - Wei Song
- School of Laboratory Medicine, Weifang Medical University, Weifang 261053, Shandong, China
| | - Lina Jia
- School of Laboratory Medicine, Weifang Medical University, Weifang 261053, Shandong, China
| | - Xiaoxiang Peng
- School of Laboratory Medicine, Weifang Medical University, Weifang 261053, Shandong, China;
| | - Ronglan Zhao
- School of Laboratory Medicine, Weifang Medical University, Weifang 261053, Shandong, China;
| |
Collapse
|
19
|
Ueno H. The IL-12-STAT4 axis in the pathogenesis of human systemic lupus erythematosus. Eur J Immunol 2019; 50:10-16. [PMID: 31762023 DOI: 10.1002/eji.201948134] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/05/2019] [Indexed: 12/28/2022]
Abstract
Generation of autoantibodies is a hallmark of systemic lupus erythematosus (SLE). As demonstrated in a number of lupus mouse models, recent evidence suggests that both GC and extrafollicular pathways contribute to the generation of autoantibodies also in human SLE, and that CD11c+ IgD- CD27- (double negative:DN) B cells play a central role in the latter pathway. In this mini-review, the author will first briefly summarize the features of CD11c+ DN B cells in human SLE, and discuss how the IL-12-STAT4 axis might contribute to the generation of autoantibodies in SLE. In addition, various types of CD4+ helper T cell subsets promoting the generation of autoantibodies in SLE will be described, and finally it will be discussed how these recent discoveries contribute to understanding of SLE pathogenesis and treatment of SLE patients.
Collapse
Affiliation(s)
- Hideki Ueno
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Immunology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan.,Institute for the Advanced Study of Human Biology, Kyoto University, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
20
|
Rohraff DM, He Y, Farkash EA, Schonfeld M, Tsou PS, Sawalha AH. Inhibition of EZH2 Ameliorates Lupus-Like Disease in MRL/lpr Mice. Arthritis Rheumatol 2019; 71:1681-1690. [PMID: 31106974 PMCID: PMC6764871 DOI: 10.1002/art.40931] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/14/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVE We previously identified a role for EZH2, a transcriptional regulator in inducing proinflammatory epigenetic changes in lupus CD4+ T cells. This study was undertaken to investigate whether inhibiting EZH2 ameliorates lupus-like disease in MRL/lpr mice. METHODS EZH2 expression levels in multiple cell types in lupus patients were evaluated using flow cytometry and messenger RNA expression data. Inhibition of EZH2 in MRL/lpr mice was achieved by intraperitoneal 3'-deazaneplanocin (DZNep) administration using a preventative and a therapeutic treatment model. Effects of DZNep on animal survival, anti-double-stranded DNA (anti-dsDNA) antibody production, proteinuria, renal histopathology, cytokine production, and T and B cell numbers and percentages were assessed. RESULTS EZH2 expression levels were increased in whole blood, neutrophils, monocytes, B cells, and CD4+ T cells in lupus patients. In MRL/lpr mice, inhibition of EZH2 by DZNep was confirmed by significant reduction of EZH2 and H3K27me3 in splenocytes. Inhibiting EZH2 with DZNep treatment before or after disease onset improved survival and significantly reduced anti-dsDNA antibody production. DZNep-treated mice displayed a significant reduction in renal involvement, splenomegaly, and lymphadenopathy. Lymphoproliferation and numbers of double-negative T cells were significantly reduced in DZNep-treated mice. Concentrations of circulating cytokines and chemokines, including tumor necrosis factor, interferon-γ, CCL2, RANTES/CCL5, interleukin-10 (IL-10), keratinocyte-derived chemokine/CXCL1, IL-12, IL-12p40, and CCL4/macrophage inflammatory protein 1β, were decreased in DZNep-treated mice. CONCLUSION EZH2 is up-regulated in multiple cell types in lupus patients. Therapeutic inhibition of EZH2 abrogates lupus-like disease in MRL/lpr mice, suggesting that EZH2 inhibitors may be repurposed as a novel therapeutic option for lupus patients.
Collapse
Affiliation(s)
- Dallas M. Rohraff
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Ye He
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Evan A. Farkash
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Mark Schonfeld
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Amr H. Sawalha
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Division of Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA
- Division of Rheumatology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
21
|
Kato R, Sumitomo S, Tsuchida Y, Tsuchiya H, Nakachi S, Sakurai K, Hanata N, Nagafuchi Y, Kubo K, Tateishi S, Kanda H, Okamura T, Yamamoto K, Fujio K. CD4 +CD25 +LAG3 + T Cells With a Feature of Th17 Cells Associated With Systemic Lupus Erythematosus Disease Activity. Front Immunol 2019; 10:1619. [PMID: 31354747 PMCID: PMC6640175 DOI: 10.3389/fimmu.2019.01619] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 06/28/2019] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that involves multiple immune cell subsets. We analyzed immune cell subsets in human peripheral blood mononuclear cells (PBMC) in order to identify the cells that are significantly associated with SLE disease activity and treatment. The frequencies of various subsets of CD4+ T cells, B cells, monocytes and NK cells in PBMC were assessed in 30 healthy controls (HC), 30 rheumatoid arthritis (RA) patients and 26 SLE patients using flow cytometry. The correlations between subset frequencies in SLE and clinical traits including Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) were examined. Changes in subset frequencies after the treatment in SLE patients were investigated. We focused on CD25+LAG3+ T cells and investigated their characteristics, including cytokine secretion, mRNA expression and suppression capacity. We assessed correlations between CD25+LAG3+ T cells and SLEDAI by Spearman's rank correlation coefficient. CD25+LAG3+ T cells were significantly increased in SLE whereas there were few in RA and HC groups. CD25+LAG3+ T cell frequencies were significantly correlated with SLEDAI and were increased in patients with a high SLEDAI score (> 10). CD25+LAG3+ T cells produced both IL-17 and FOXP3, expressed mRNA of both FOXP3 and RORC and lacked suppressive capacity. CD25+LAG3+ T cells were associated with disease activity of SLE. CD25+LAG3+ T cells had features of both CD25+FOXP3+ regulatory T cells (CD25+ Treg) and Th17. CD25+LAG3+ T cells could be associated with the inflammatory pathophysiology of SLE.
Collapse
Affiliation(s)
- Rika Kato
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shuji Sumitomo
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yumi Tsuchida
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruka Tsuchiya
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinichiro Nakachi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keiichi Sakurai
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Norio Hanata
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuo Nagafuchi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kanae Kubo
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shoko Tateishi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Immunotherapy Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroko Kanda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Immunotherapy Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Yamamoto
- Center for Integrative Medical Sciences, The Institute of Physical and Chemical Research, Yokohama, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
22
|
Innate lymphoid cell disturbance with increase in ILC1 in systemic lupus erythematosus. Clin Immunol 2019; 202:49-58. [PMID: 30926441 DOI: 10.1016/j.clim.2019.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 03/15/2019] [Accepted: 03/25/2019] [Indexed: 12/21/2022]
Abstract
The innate lymphoid cell (ILC) is a group of effector cells with diverse important cellular functions in both health and disease states. In comparison with healthy controls, there were increases in circulating ILC in SLE patients. The proportion of ILC1 significantly increased with significant decreases of ILC2 in SLE patients and ILC3 in SLE patients with moderate to severe activity. IL-12, IL-18, IL-25, IL-33, IL-23, IL-1β and IFN-γ were significantly increased in SLE patients. Moreover, IL-12, IL-18 and IL-1β but not IFN-γ correlated significantly with SLEDAI. Successful treatments rapidly reduced them and with certain normalization of the ILC subsets. In addition to increases in ILC1 numbers, ~ 80% of the ILC1 in SLE patients were positive for synthesis of IFN-γ. Plasma from SLE patients were shown to be potent in inducing ILC1. Thus, increased circulating ILC1 might contribute to the pathogenesis of SLE through mounting type 1 immune response.
Collapse
|
23
|
Goropevšek A, Holcar M, Pahor A, Avčin T. STAT signaling as a marker of SLE disease severity and implications for clinical therapy. Autoimmun Rev 2019; 18:144-154. [DOI: 10.1016/j.autrev.2018.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 08/13/2018] [Indexed: 12/21/2022]
|
24
|
Elshikha AS, Yuan Y, Lu Y, Chen MJ, Abboud G, Akbar MA, Plate H, Wolney H, Hoffmann T, Tagari E, Zeumer L, Morel L, Song S. Alpha 1 Antitrypsin Gene Therapy Extends the Lifespan of Lupus-Prone Mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 11:131-142. [PMID: 30547047 PMCID: PMC6258868 DOI: 10.1016/j.omtm.2018.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/13/2018] [Indexed: 12/22/2022]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease characterized by high levels of pathogenic autoantibodies and tissue damage. Multiple studies showed that dendritic cell (DC) activation plays a critical role in SLE pathogenesis. Human alpha 1 antitrypsin (hAAT) is a serine proteinase inhibitor with potent anti-inflammatory and cytoprotective properties. In this study, we first examined the effects of hAAT on the functions of DCs from lupus-prone mice, and we showed that hAAT treatment efficiently inhibited CpG- (TLR9 agonist) induced activation of bone marrow-derived conventional and plasmacytoid DCs as well as the production of pro-inflammatory cytokines. The hAAT treatment also attenuated DC help for B cell proliferation and immunoglobulin M (IgM) production. We next tested the protective effect of hAAT protein and gene therapy using recombinant adeno-associated virus 8 (rAAV8-CB-hAAT) in a spontaneous lupus mouse model, and we showed that both treatments decreased autoantibody levels. Importantly, rAAV8-CB-hAAT did not induce an immune response to its transgene product (hAAT), but it showed more pronounced therapeutic effects in reducing urine protein levels and extending the lifespan of these mice. These results indicate that AAT has therapeutic potential in the treatment of SLE in humans.
Collapse
Affiliation(s)
- Ahmed Samir Elshikha
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA.,Department of Pharmaceutics, Zagazig University, Zagazig, Sharkia, Egypt
| | - Ye Yuan
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Yuanqing Lu
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Mong-Jen Chen
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Georges Abboud
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mohammad Ahsanul Akbar
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Henrike Plate
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Hedwig Wolney
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Tanja Hoffmann
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Eleni Tagari
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Leilani Zeumer
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Laurence Morel
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Sihong Song
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
25
|
Papp K, Gordon K, Thaçi D, Morita A, Gooderham M, Foley P, Girgis IG, Kundu S, Banerjee S. Phase 2 Trial of Selective Tyrosine Kinase 2 Inhibition in Psoriasis. N Engl J Med 2018; 379:1313-1321. [PMID: 30205746 DOI: 10.1056/nejmoa1806382] [Citation(s) in RCA: 262] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Tyrosine kinase 2 (TYK2) signaling pathways, which mediate cytokine signaling, are implicated in the pathophysiology of psoriasis. Selective inhibitors of TYK2 may be effective in treating psoriasis. METHODS We conducted a phase 2, double-blind trial of a TYK2 inhibitor, BMS-986165, in adults with moderate-to-severe psoriasis, excluding patients with a previous lack of response to agents targeting cytokine signaling through the same tyrosine kinase pathway. Patients were randomly assigned to receive the drug orally at a dose of 3 mg every other day, 3 mg daily, 3 mg twice daily, 6 mg twice daily, or 12 mg daily or to receive placebo. The primary end point was a 75% or greater reduction from baseline in the Psoriasis Area and Severity Index (PASI) score at week 12 (higher scores indicate greater severity of psoriasis). RESULTS A total of 267 patients received at least one dose in an intervention group of the trial. At week 12, the percentage of patients with a 75% or greater reduction in the PASI score was 7% (3 of 45 patients) with placebo, 9% (4 of 44 patients) with 3 mg of BMS-986165 every other day (P=0.49 vs. placebo), 39% (17 of 44 patients) with 3 mg daily (P<0.001 vs. placebo), 69% (31 of 45 patients) with 3 mg twice daily (P<0.001 vs. placebo), 67% (30 of 45 patients) with 6 mg twice daily (P<0.001 vs. placebo), and 75% (33 of 44 patients) with 12 mg daily (P<0.001 vs. placebo). There were three serious adverse events in patients receiving the active drug, as well as one case of malignant melanoma 96 days after the start of treatment. CONCLUSIONS Selective inhibition of TYK2 with the oral agent BMS-986165 at doses of 3 mg daily and higher resulted in greater clearing of psoriasis than did placebo over a period of 12 weeks. Larger and longer-duration trials of this drug are required to determine its safety and durability of effect in patients with psoriasis. (Funded by Bristol-Myers Squibb; ClinicalTrials.gov number, NCT02931838 .).
Collapse
Affiliation(s)
- Kim Papp
- From K. Papp Clinical Research and Probity Medical Research, Waterloo (K.P.), the SKiN Centre for Dermatology and Probity Medical Research, Peterborough (M.G.), and Queen's University, Kingston (M.G.) - all in Ontario, Canada; Medical College of Wisconsin, Milwaukee (K.G.); the University of Luebeck, Luebeck, Germany (D.T.); Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan (A.M.); the University of Melbourne, St. Vincent's Hospital Melbourne, Skin and Cancer Foundation, and Probity Medical Research, Melbourne, VIC, Australia (P.F.); and Bristol-Myers Squibb, Princeton, NJ (I.G.G., S.K., S.B.)
| | - Kenneth Gordon
- From K. Papp Clinical Research and Probity Medical Research, Waterloo (K.P.), the SKiN Centre for Dermatology and Probity Medical Research, Peterborough (M.G.), and Queen's University, Kingston (M.G.) - all in Ontario, Canada; Medical College of Wisconsin, Milwaukee (K.G.); the University of Luebeck, Luebeck, Germany (D.T.); Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan (A.M.); the University of Melbourne, St. Vincent's Hospital Melbourne, Skin and Cancer Foundation, and Probity Medical Research, Melbourne, VIC, Australia (P.F.); and Bristol-Myers Squibb, Princeton, NJ (I.G.G., S.K., S.B.)
| | - Diamant Thaçi
- From K. Papp Clinical Research and Probity Medical Research, Waterloo (K.P.), the SKiN Centre for Dermatology and Probity Medical Research, Peterborough (M.G.), and Queen's University, Kingston (M.G.) - all in Ontario, Canada; Medical College of Wisconsin, Milwaukee (K.G.); the University of Luebeck, Luebeck, Germany (D.T.); Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan (A.M.); the University of Melbourne, St. Vincent's Hospital Melbourne, Skin and Cancer Foundation, and Probity Medical Research, Melbourne, VIC, Australia (P.F.); and Bristol-Myers Squibb, Princeton, NJ (I.G.G., S.K., S.B.)
| | - Akimichi Morita
- From K. Papp Clinical Research and Probity Medical Research, Waterloo (K.P.), the SKiN Centre for Dermatology and Probity Medical Research, Peterborough (M.G.), and Queen's University, Kingston (M.G.) - all in Ontario, Canada; Medical College of Wisconsin, Milwaukee (K.G.); the University of Luebeck, Luebeck, Germany (D.T.); Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan (A.M.); the University of Melbourne, St. Vincent's Hospital Melbourne, Skin and Cancer Foundation, and Probity Medical Research, Melbourne, VIC, Australia (P.F.); and Bristol-Myers Squibb, Princeton, NJ (I.G.G., S.K., S.B.)
| | - Melinda Gooderham
- From K. Papp Clinical Research and Probity Medical Research, Waterloo (K.P.), the SKiN Centre for Dermatology and Probity Medical Research, Peterborough (M.G.), and Queen's University, Kingston (M.G.) - all in Ontario, Canada; Medical College of Wisconsin, Milwaukee (K.G.); the University of Luebeck, Luebeck, Germany (D.T.); Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan (A.M.); the University of Melbourne, St. Vincent's Hospital Melbourne, Skin and Cancer Foundation, and Probity Medical Research, Melbourne, VIC, Australia (P.F.); and Bristol-Myers Squibb, Princeton, NJ (I.G.G., S.K., S.B.)
| | - Peter Foley
- From K. Papp Clinical Research and Probity Medical Research, Waterloo (K.P.), the SKiN Centre for Dermatology and Probity Medical Research, Peterborough (M.G.), and Queen's University, Kingston (M.G.) - all in Ontario, Canada; Medical College of Wisconsin, Milwaukee (K.G.); the University of Luebeck, Luebeck, Germany (D.T.); Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan (A.M.); the University of Melbourne, St. Vincent's Hospital Melbourne, Skin and Cancer Foundation, and Probity Medical Research, Melbourne, VIC, Australia (P.F.); and Bristol-Myers Squibb, Princeton, NJ (I.G.G., S.K., S.B.)
| | - Ihab G Girgis
- From K. Papp Clinical Research and Probity Medical Research, Waterloo (K.P.), the SKiN Centre for Dermatology and Probity Medical Research, Peterborough (M.G.), and Queen's University, Kingston (M.G.) - all in Ontario, Canada; Medical College of Wisconsin, Milwaukee (K.G.); the University of Luebeck, Luebeck, Germany (D.T.); Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan (A.M.); the University of Melbourne, St. Vincent's Hospital Melbourne, Skin and Cancer Foundation, and Probity Medical Research, Melbourne, VIC, Australia (P.F.); and Bristol-Myers Squibb, Princeton, NJ (I.G.G., S.K., S.B.)
| | - Sudeep Kundu
- From K. Papp Clinical Research and Probity Medical Research, Waterloo (K.P.), the SKiN Centre for Dermatology and Probity Medical Research, Peterborough (M.G.), and Queen's University, Kingston (M.G.) - all in Ontario, Canada; Medical College of Wisconsin, Milwaukee (K.G.); the University of Luebeck, Luebeck, Germany (D.T.); Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan (A.M.); the University of Melbourne, St. Vincent's Hospital Melbourne, Skin and Cancer Foundation, and Probity Medical Research, Melbourne, VIC, Australia (P.F.); and Bristol-Myers Squibb, Princeton, NJ (I.G.G., S.K., S.B.)
| | - Subhashis Banerjee
- From K. Papp Clinical Research and Probity Medical Research, Waterloo (K.P.), the SKiN Centre for Dermatology and Probity Medical Research, Peterborough (M.G.), and Queen's University, Kingston (M.G.) - all in Ontario, Canada; Medical College of Wisconsin, Milwaukee (K.G.); the University of Luebeck, Luebeck, Germany (D.T.); Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan (A.M.); the University of Melbourne, St. Vincent's Hospital Melbourne, Skin and Cancer Foundation, and Probity Medical Research, Melbourne, VIC, Australia (P.F.); and Bristol-Myers Squibb, Princeton, NJ (I.G.G., S.K., S.B.)
| |
Collapse
|
26
|
Ma X, Nakayamada S, Kubo S, Sakata K, Yamagata K, Miyazaki Y, Yoshikawa M, Kitanaga Y, Zhang M, Tanaka Y. Expansion of T follicular helper-T helper 1 like cells through epigenetic regulation by signal transducer and activator of transcription factors. Ann Rheum Dis 2018; 77:1354-1361. [PMID: 29853448 DOI: 10.1136/annrheumdis-2017-212652] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 05/06/2018] [Accepted: 05/07/2018] [Indexed: 01/31/2023]
Abstract
OBJECTIVES T follicular helper (Tfh) cells are critical in the development and progression of systemic lupus erythematosus (SLE). To assess the characteristics and mechanisms of differentiation of Tfh cells, we investigated the phenotype of T helper cells in patients with SLE and underlying epigenetic modifications by cytokine-induced signal transducer and activators of transcription (STAT) family factors. METHODS Peripheral blood mononuclear cells from patients and healthy donors were analysed by flow cytometry. CD4+ T cells were isolated and cultured under various stimulations. Expression of characteristic markers and phosphorylation of STATs were analysed by flow cytometry and quantitative PCR. Histone modifications were analysed by chromatin immunoprecipitation (ChIP)-PCR. RESULTS Differentiation of CD4+CXCR5+CXCR3+Bcl-6+T-bet+IL-21+IFN-γ+Tfh-Th1-like cells was induced by interleukin (IL)-12-induced activation of STAT1 and STAT4 simultaneously. The loci of Bcl-6 and T-bet at STAT binding sites were marked by bivalent histone modifications. After IL-12 stimulation, both STAT1 and STAT4 directly bound on BCL6 and TBX21 gene loci accompanied by suppression of repressive histone mark trimethylated histone 3 lysine 27. Levels of serum IL-12 and interferon (IFN)-γ, expression of IL-12 receptors and proportion of CXCR5+CXCR3+ activated Tfh-Th1-like cells were increased in patients with SLE. Furthermore, the level of pSTAT1, pSTAT4 and T-bet were higher in activated Tfh-Th1-like cells than non-Tfh-Th1 cells. CONCLUSION Our findings suggest that IL-12-mediated co-activation of STAT1 and STAT4 alters histone modification, resulting in differentiation of Tfh-Th1-like cells that are characteristically expanded in patients with SLE. This could be one of the underlying mechanisms responsible for expansion of Tfh-Th1-like cells and potentially helpful towards development of cell-specific treatment for SLE.
Collapse
Affiliation(s)
- Xiaoxue Ma
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan.,Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Shingo Nakayamada
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Satoshi Kubo
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Kei Sakata
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan.,Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Kaoru Yamagata
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Yusuke Miyazaki
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Maiko Yoshikawa
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Yukihiro Kitanaga
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan.,Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Japan
| | - Mingzeng Zhang
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan.,Department of Hematology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yoshiya Tanaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| |
Collapse
|
27
|
Hagberg N, Joelsson M, Leonard D, Reid S, Eloranta ML, Mo J, Nilsson MK, Syvänen AC, Bryceson YT, Rönnblom L. The STAT4 SLE risk allele rs7574865[T] is associated with increased IL-12-induced IFN-γ production in T cells from patients with SLE. Ann Rheum Dis 2018; 77:1070-1077. [PMID: 29475858 PMCID: PMC6029643 DOI: 10.1136/annrheumdis-2017-212794] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/30/2018] [Accepted: 02/03/2018] [Indexed: 01/26/2023]
Abstract
Objectives Genetic variants in the transcription factor STAT4 are associated with increased susceptibility to systemic lupus erythematosus (SLE) and a more severe disease phenotype. This study aimed to clarify how the SLE-associated intronic STAT4 risk allele rs7574865[T] affects the function of immune cells in SLE. Methods Peripheral blood mononuclear cells (PBMCs) were isolated from 52 genotyped patients with SLE. Phosphorylation of STAT4 (pSTAT4) and STAT1 (pSTAT1) in response to interferon (IFN)-α, IFN-γ or interleukin (IL)-12, total levels of STAT4, STAT1 and T-bet, and frequency of IFN-γ+ cells on IL-12 stimulation were determined by flow cytometry in subsets of immune cells before and after preactivation of cells with phytohaemagglutinin (PHA) and IL-2. Cellular responses and phenotypes were correlated to STAT4 risk allele carriership. Janus kinase inhibitors (JAKi) selective for TYK2 (TYK2i) or JAK2 (JAK2i) were evaluated for inhibition of IL-12 or IFN-γ-induced activation of SLE PBMCs. Results In resting PBMCs, the STAT4 risk allele was neither associated with total levels of STAT4 or STAT1, nor cytokine-induced pSTAT4 or pSTAT1. Following PHA/IL-2 activation, CD8+ T cells from STAT4 risk allele carriers displayed increased levels of STAT4 resulting in increased pSTAT4 in response to IL-12 and IFN-α, and an augmented IL-12-induced IFN-γ production in CD8+ and CD4+ T cells. The TYK2i and the JAK2i efficiently blocked IL-12 and IFN-γ-induced activation of PBMCs from STAT4 risk patients, respectively. Conclusions T cells from patients with SLE carrying the STAT4 risk allele rs7574865[T] display an augmented response to IL-12 and IFN-α. This subset of patients may benefit from JAKi treatment.
Collapse
Affiliation(s)
- Niklas Hagberg
- Department of Medical Sciences, Rheumatology and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Martin Joelsson
- Department of Medical Sciences, Rheumatology and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Dag Leonard
- Department of Medical Sciences, Rheumatology and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Sarah Reid
- Department of Medical Sciences, Rheumatology and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Maija-Leena Eloranta
- Department of Medical Sciences, Rheumatology and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - John Mo
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Magnus K Nilsson
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Ann-Christine Syvänen
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| | - Yenan T Bryceson
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Department of Clinical Sciences, Broegelmann Research Laboratory, University of Bergen, Bergen, Norway
| | - Lars Rönnblom
- Department of Medical Sciences, Rheumatology and Science for Life Laboratories, Uppsala University, Uppsala, Sweden
| |
Collapse
|
28
|
Arriens C, Wren JD, Munroe ME, Mohan C. Systemic lupus erythematosus biomarkers: the challenging quest. Rheumatology (Oxford) 2017; 56:i32-i45. [PMID: 28013203 DOI: 10.1093/rheumatology/kew407] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Indexed: 01/01/2023] Open
Abstract
SLE, a multisystem heterogeneous disease, is characterized by production of antibodies to cellular components, with activation of both the innate and the adaptive immune system. Decades of investigation of blood biomarkers has resulted in incremental improvements in the understanding of SLE. Owing to the heterogeneity of immune dysregulation, no single biomarker has emerged as a surrogate for disease activity or prediction of disease. Beyond identification of surrogate biomarkers, a multitude of clinical trials have sought to inhibit elevated SLE biomarkers for therapeutic benefit. Armed with new -omics technologies, the necessary yet daunting quest to identify better surrogate biomarkers and successful therapeutics for SLE continues with tenacity.
Collapse
Affiliation(s)
- Cristina Arriens
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation.,Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Jonathan D Wren
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation
| | - Melissa E Munroe
- Department of Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| |
Collapse
|
29
|
Caster DJ, Powell DW, Miralda I, Ward RA, McLeish KR. Re-Examining Neutrophil Participation in GN. J Am Soc Nephrol 2017; 28:2275-2289. [PMID: 28620081 DOI: 10.1681/asn.2016121271] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Significant advances in understanding the pathogenesis of GN have occurred in recent decades. Among those advances is the finding that both innate and adaptive immune cells contribute to the development of GN. Neutrophils were recognized as key contributors in early animal models of GN, at a time when the prevailing view considered neutrophils to function as nonspecific effector cells that die quickly after performing antimicrobial functions. However, advances over the past two decades have shown that neutrophil functions are more complex and sophisticated. Specifically, research has revealed that neutrophil survival is regulated by the inflammatory milieu and that neutrophils demonstrate plasticity, mediate microbial killing through previously unrecognized mechanisms, demonstrate transcriptional activity leading to the release of cytokines and chemokines, interact with and regulate cells of the innate and adaptive immune systems, and contribute to the resolution of inflammation. Therefore, neutrophil participation in glomerular diseases deserves re-evaluation. In this review, we describe advances in understanding classic neutrophil functions, review the expanded roles of neutrophils in innate and adaptive immune responses, and summarize current knowledge of neutrophil contributions to GN.
Collapse
Affiliation(s)
- Dawn J Caster
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, .,Nephrology Section, Medicine Service, Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, and
| | - David W Powell
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky
| | - Irina Miralda
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Richard A Ward
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky
| | - Kenneth R McLeish
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky.,Nephrology Section, Medicine Service, Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, and
| |
Collapse
|
30
|
Gao S, Gong Y, Ji J, Yuan L, Han L, Guo Y, Fan X, Hou Y, Hua C. A new benzenediamine derivative modulates Toll-like receptors-induced myeloid dendritic cells activation and ameliorates lupus-like syndrome in MRLlpr/lpr mice. Eur J Pharmacol 2017; 803:94-102. [PMID: 28342978 DOI: 10.1016/j.ejphar.2017.03.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 11/29/2022]
Abstract
Modulators of the over-activation of myeloid dendritic cells (mDCs) by Toll-like receptors (TLRs) have an advantage in the treatment of systemic lupus erythematosus (SLE). This study was designed to evaluate the effects of FC-99, a novel benzenediamine derivative, on TLR-induced activation of mDCs, and to assess the efficacy of FC-99 in a murine model of SLE. In vitro, FC-99 inhibited the phenotypic (CD40 and MHC-II) and functional activation (IL-12 and CXCL10) of mDCs induced by TLR ligands. In vivo, MRLlpr/lpr mice displayed renal diseases associated with increased levels of proteinuria and immunoglobulin, which were ameliorated by FC-99. Enhanced accumulation and activation of mDCs in lymphoid organs was also impaired by FC-99. Additionally, FC-99 inhibited the activation of IκB-α and upregulated the expression of TNFα-induced protein 3 (TNFAIP3) in vitro and in vivo. These results indicate that FC-99 modulates TLR-induced activation of mDCs and ameliorates lupus-like syndrome in MRLlpr/lpr mice. This effect is closely associated with the inhibition of IκB-α and upregulation of TNFAIP3.
Collapse
Affiliation(s)
- Sheng Gao
- Laboratory Animal Center, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Yongsheng Gong
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Jianjian Ji
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Linbo Yuan
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Liping Han
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Yimin Guo
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Xiaofang Fan
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China.
| |
Collapse
|
31
|
Stem cell therapy: An emerging modality in glomerular diseases. Cytotherapy 2017; 19:333-348. [PMID: 28089754 DOI: 10.1016/j.jcyt.2016.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/17/2016] [Accepted: 11/03/2016] [Indexed: 12/12/2022]
Abstract
The kidney has been considered a highly terminally differentiated organ with low proliferative potential and thus unlikely to undergo regeneration. Glomerular disease progresses to end-stage renal disease (ESRD), which requires dialysis or renal transplantation for better quality of life for patients with ESRD. Because of the shortage of implantable kidneys and complications such as immune rejection, septicemia and toxicity of immunosuppression, kidney transplantation remains a challenge. Therapeutic options available for glomerular disease include symptomatic treatment and strategies to delay progression. In an attempt to develop innovative treatments by promoting the limited capability of regeneration and repair after kidney injury and overcome the progressive pathological process that is uncontrolled with conventional treatment modalities, stem cell-based therapy has emerged as novel intervention due to its ability to inhibit inflammation and promote regeneration. Recent developments in cell therapy have demonstrated promising therapeutic outcomes in terms of restoration of renal structure and function. This review focuses on stem cell therapy approaches for the treatment of glomerular disease, including the various cell sources used and recent advances in preclinical and clinical studies.
Collapse
|
32
|
Abstract
The introduction of corticosteroids and later, cyclophosphamide dramatically improved survival in patients with proliferative lupus nephritis, and combined administration of these agents became the standard-of-care treatment for this disease. However, treatment failures were still common and the rate of progression to ESRD remained unacceptably high. Additionally, treatment was associated with significant morbidity. Therefore, as patient survival improved, the goals for advancing lupus nephritis treatment shifted to identifying therapies that could improve long-term renal outcomes and minimize treatment-related toxicity. Unfortunately, progress has been slow and the current approaches to the management of lupus nephritis continue to rely on high-dose corticosteroids plus a broad-spectrum immunosuppressive agent. Over the past decade, an improved understanding of lupus nephritis pathogenesis fueled several clinical trials of novel drugs, but none have been found to be superior to the combination of a cytotoxic agent and corticosteroids. Despite these trial failures, efforts to translate mechanistic advances into new treatment approaches continue. In this review, we discuss current therapeutic strategies for lupus nephritis, briefly review recent advances in understanding the pathogenesis of this disease, and describe emerging approaches developed on the basis of these advances that promise to improve upon the standard-of-care lupus nephritis treatments.
Collapse
Affiliation(s)
- Samir V Parikh
- Division of Nephrology, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Brad H Rovin
- Division of Nephrology, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
33
|
Blanco P, Ueno H, Schmitt N. T follicular helper (Tfh) cells in lupus: Activation and involvement in SLE pathogenesis. Eur J Immunol 2016; 46:281-90. [DOI: 10.1002/eji.201545760] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 11/17/2015] [Accepted: 11/23/2015] [Indexed: 12/28/2022]
Affiliation(s)
- Patrick Blanco
- Univ. Bordeaux; CIRID, UMR/CNRS; 5164 Bordeaux France
- CNRS; CIRID, UMR; 5164 Bordeaux France
- CHU de Bordeaux; Bordeaux France
| | - Hideki Ueno
- Baylor Institute for Immunology Research; Dallas USA
| | | |
Collapse
|
34
|
El-Karaksy SM, Raafat HA, Abadir MNY, Hanna MOF. Down-regulation of expression of retinoid acid-related orphan receptor C (RORC) in systemic lupus erythematosus. J Recept Signal Transduct Res 2015; 36:207-12. [PMID: 26498317 DOI: 10.3109/10799893.2015.1075042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Retinoic acid-related orphan receptor C (RORC), the key factor orchestrating the transcription of genes encoding interleukin 17, plays a critical role in the regulation of inflammatory responses. OBJECTIVE The objective of this study was to analyze the expression of RORC in the peripheral blood of patients with systemic lupus erythematosus (SLE) for a better understanding of the pathogenesis of SLE especially in relation to disease activity and clinical and biochemical findings. METHODS The study included 24 patients with SLE and a control group of 18 healthy gender- and age-matched individuals. Evaluation of the level of expression of RORC mRNA was performed by real-time polymerase chain reaction. RESULTS The results showed that patients with SLE had lower RORC gene expression levels compared with healthy subjects that were not correlated with disease activity. The down-regulation of RORC was significantly lower in patients with lupus nephritis in remission than active lupus nephritis and nonrenal patients. CONCLUSIONS The findings suggest that RORC plays a significant role in the dysregulated immune response associated with SLE. Deciphering the intricate regulatory network and the target genes of RORC will help unravel new specific treatments for SLE.
Collapse
Affiliation(s)
| | - Hala A Raafat
- b Department of Rheumatology and Rehabilitation , Faculty of Medicine, Cairo University , Cairo , Egypt , and
| | | | | |
Collapse
|
35
|
Schneider L, Colar da Silva AC, Werres Junior LC, Alegretti AP, Pereira dos Santos AS, Santos M, Sassi R, Heemann B, Pfaffenseller B, Tavares Brenol JC, Monticielo OA. Vitamin D levels and cytokine profiles in patients with systemic lupus erythematosus. Lupus 2015; 24:1191-7. [PMID: 25926056 DOI: 10.1177/0961203315584811] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 04/07/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To evaluate the expression of Th1, Th2, and Th17 cytokines in patients with systemic lupus erythematosus (SLE), and verify the association between serum cytokine levels and vitamin D concentration. METHODS The sample consisted of 172 patients with SLE. 25-Hydroxyvitamin D (25(OH)D) levels were measured by chemiluminescence and 25(OH)D levels <20 ng/mL were considered to reflect vitamin D deficiency. Serum cytokine levels were measured in once-thawed samples, using a Th1/Th2/Th17 CBA (cytometric beads array) kit. RESULTS One hundred and sixty-one (93.6%) patients were women and 128 (74.4%) were of European descent. Mean patient age was 40.5 ± 13.8 years, and mean age at diagnosis was 31.5 ± 13.4 years. At the time of study entry, patients had a median (IQR) SLEDAI of 2 (1-4) and SLICC of 0 (0-1). Mean 25(OH)D concentration was 25.4 ± 11.04 ng/mL. Fifty-nine (34.3%) patients had a vitamin D deficiency. No statistically significant associations were identified between cytokine and vitamin D levels. The most significant finding was a positive correlation between INF-α levels and SLEDAI (r(s) = 0.22, p = 0.04). CONCLUSION Although vitamin D deficiencies are highly prevalent in patients with SLE, vitamin D levels were not significantly associated with patient cytokine profiles. The positive correlation between IFN-α levels and SLEDAI showed in this study corroborates other findings in the literature. The present results did not replicate those of in vitro studies of the effect of vitamin D levels on cytokine profiles. Placebo-controlled intervention trials of the effect of vitamin D on cytokine profiles are still required before any definitive conclusions can be drawn regarding the association between these variables.
Collapse
Affiliation(s)
- L Schneider
- Department of Internal Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Brazil
| | - A C Colar da Silva
- Clinical Pathology Laboratory, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Brazil
| | - L C Werres Junior
- Clinical Pathology Laboratory, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Brazil
| | - A P Alegretti
- Clinical Pathology Laboratory, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Brazil
| | - A S Pereira dos Santos
- Department of Internal Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Brazil
| | - M Santos
- Department of Internal Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Brazil
| | - R Sassi
- Department of Internal Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Brazil
| | - B Heemann
- Department of Internal Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Brazil
| | - B Pfaffenseller
- Molecular Psychiatry Laboratory, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Brazil
| | - J C Tavares Brenol
- Department of Internal Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Brazil
| | - O A Monticielo
- Department of Internal Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Brazil
| |
Collapse
|
36
|
Pathogenesis of renal disease in systemic lupus erythematosus--the role of autoantibodies and lymphocytes subset abnormalities. Int J Mol Sci 2015; 16:7917-31. [PMID: 25860947 PMCID: PMC4425058 DOI: 10.3390/ijms16047917] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 03/02/2015] [Indexed: 12/19/2022] Open
Abstract
Lupus nephritis (LN) is a common and severe organ manifestation of systemic lupus erythematosus (SLE), and is associated with significant patient morbidity and mortality. Autoantibodies and aberrations in lymphocyte subsets have putative roles in the pathogenesis of SLE and LN, and might reflect disease activity and are amenable to immunosuppressive treatments. Anti-DNA is one of the well-studied autoantibodies, which correlates with disease activity and has direct nephritogenic effects on resident renal cells and various glomerular components. Other important autoantibodies in the pathogenesis of LN include anti-C1q, anti-α-actinin and anti-nucleosome antibodies. Changes in naive and memory B cells and plasma cells have been observed in SLE and LN patients. These B cell subsets exert diverse effects during pathogenesis of LN such as production of autoantibodies, secretion of proinflammatory and anti-inflammatory cytokines and presentation of auto-antigens to effector cells. Aberration of T lymphocytes, especially the T-helper subsets, is also highly pertinent in the development of LN. In this context, important T helper subsets include Th1, Th2, Th9, Th17, TReg and follicular T-helper cells. The growing knowledge on these autoantibodies and lymphocyte subset abnormalities will enhance our understanding of SLE and LN, and hence help devise better strategies for disease monitoring and treatment.
Collapse
|
37
|
Martin I, Cabán-Hernández K, Figueroa-Santiago O, Espino AM. Fasciola hepatica fatty acid binding protein inhibits TLR4 activation and suppresses the inflammatory cytokines induced by lipopolysaccharide in vitro and in vivo. THE JOURNAL OF IMMUNOLOGY 2015; 194:3924-36. [PMID: 25780044 DOI: 10.4049/jimmunol.1401182] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 02/06/2015] [Indexed: 12/11/2022]
Abstract
TLR4, the innate immunity receptor for bacterial endotoxins, plays a pivotal role in the induction of inflammatory responses. There is a need to develop molecules that block either activation through TLR4 or the downstream signaling pathways to inhibit the storm of inflammation typically elicited by bacterial LPS, which is a major cause of the high mortality associated with bacterial sepsis. We report in this article that a single i.p. injection of 15 μg fatty acid binding protein from Fasciola hepatica (Fh12) 1 h before exposure to LPS suppressed significantly the expression of serum inflammatory cytokines in a model of septic shock using C57BL/6 mice. Because macrophages are a good source of IL-12p70 and TNF-α, and are critical in driving adaptive immunity, we investigated the effect of Fh12 on the function of mouse bone marrow-derived macrophages (bmMΦs). Although Fh12 alone did not induce cytokine expression, it significantly suppressed the expression of IL-12, TNF-α, IL-6, and IL-1β cytokines, as well as inducible NO synthase-2 in bmMΦs, and also impaired the phagocytic capacity of bmMΦs. Fh12 had a limited effect on the expression of inflammatory cytokines induced in response to other TLR ligands. One mechanism used by Fh12 to exert its anti-inflammatory effect is binding to the CD14 coreceptor. Moreover, it suppresses phosphorylation of ERK, p38, and JNK. The potent anti-inflammatory properties of Fh12 demonstrated in this study open doors to further studies directed at exploring the potential of this molecule as a new class of drug against septic shock or other inflammatory diseases.
Collapse
Affiliation(s)
- Ivelisse Martin
- Laboratory of Immunology and Molecular Parasitology, Department of Microbiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico 00936-5067
| | - Kimberly Cabán-Hernández
- Laboratory of Immunology and Molecular Parasitology, Department of Microbiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico 00936-5067
| | - Olgary Figueroa-Santiago
- Laboratory of Immunology and Molecular Parasitology, Department of Microbiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico 00936-5067
| | - Ana M Espino
- Laboratory of Immunology and Molecular Parasitology, Department of Microbiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico 00936-5067
| |
Collapse
|
38
|
Varada S, Gottlieb AB, Merola JF, Saraiya AR, Tintle SJ. Treatment of coexistent psoriasis and lupus erythematosus. J Am Acad Dermatol 2015; 72:253-60. [DOI: 10.1016/j.jaad.2014.10.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 10/27/2014] [Accepted: 10/28/2014] [Indexed: 11/26/2022]
|
39
|
Hu J, Zhang Y, Liu X, Wei L, Zhang D. Relationship between certain T helper cytokines and ANA staining: Who is the helper? Int Immunopharmacol 2015; 24:208-210. [DOI: 10.1016/j.intimp.2014.12.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 11/24/2014] [Accepted: 12/09/2014] [Indexed: 01/15/2023]
|
40
|
Lieberman LA, Tsokos GC. Lupus-prone mice fail to raise antigen-specific T cell responses to intracellular infection. PLoS One 2014; 9:e111382. [PMID: 25360768 PMCID: PMC4216089 DOI: 10.1371/journal.pone.0111382] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 09/23/2014] [Indexed: 01/08/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is characterized by multiple cellular abnormalities culminating in the production of autoantibodies and immune complexes, resulting in tissue inflammation and organ damage. Besides active disease, the main cause of morbidity and mortality in SLE patients is infections, including those from opportunistic pathogens. To understand the failure of the immune system to fend off infections in systemic autoimmunity, we infected the lupus-prone murine strains B6.lpr and BXSB with the intracellular parasite Toxoplasma gondii and survival was monitored. Furthermore, mice were sacrificed days post infection and parasite burden and cellular immune responses such as cytokine production and cell activation were assessed. Mice from both strains succumbed to infection acutely and we observed greater susceptibility to infection in older mice. Increased parasite burden and a defective antigen-specific IFN-gamma response were observed in the lupus-prone mice. Furthermore, T cell:dendritic cell co-cultures established the presence of an intrinsic T cell defect responsible for the decreased antigen-specific response. An antigen-specific defect in IFN- gamma production prevents lupus-prone mice from clearing infection effectively. This study reveals the first cellular insight into the origin of increased susceptibility to infections in SLE disease and may guide therapeutic approaches.
Collapse
Affiliation(s)
- Linda A. Lieberman
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - George C. Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
41
|
Maier-Moore JS, Horton CG, Mathews SA, Confer AW, Lawrence C, Pan Z, Coggeshall KM, Farris AD. Interleukin-6 deficiency corrects nephritis, lymphocyte abnormalities, and secondary Sjögren's syndrome features in lupus-prone Sle1.Yaa mice. Arthritis Rheumatol 2014; 66:2521-31. [PMID: 24891301 PMCID: PMC4146640 DOI: 10.1002/art.38716] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 05/15/2014] [Indexed: 02/01/2023]
Abstract
OBJECTIVE To assess disease features in Sle1.Yaa mice with genetic interleukin-6 (IL-6) deficiency. METHODS Sera and tissues were collected from C57BL/6 (B6), Sle1.Yaa, and Sle1.Yaa.IL-6(-/-) mice and analyzed for various features of disease. Using serum samples, autoantibody specificities were determined by enzyme-linked immunosorbent assay (ELISA) and indirect immunofluorescence, cytokine production was analyzed by Luminex and ELISA, and levels of blood urea nitrogen were determined by ELISA. Renal, lung, and salivary gland tissue sections were evaluated for pathologic changes. Lymphocyte phenotypes, including CD4+ T cell cytokine production, and those of follicular and extrafollicular T helper subsets, germinal center B cells, and plasma cells, were determined using flow cytometry. RESULTS IL-6 deficiency not only ameliorated autoantibody production and renal disease in this model, but also effectively reduced inflammation of lungs and salivary glands. Furthermore, IL-6 deficiency abrogated differentiation of Th1 and extrafollicular T helper cells, germinal center B cells, and plasma cells in the spleen and eliminated renal T cells with IL-17, interferon-γ, and IL-21 production potential. CONCLUSION Our findings highlight IL-6-mediated T cell aberrations in Yaa-driven autoimmunity and support the concept of therapeutic IL-6/IL-6 receptor blockade in systemic lupus erythematosus and Sjögren's syndrome by impairing the production of autoantibodies and lymphocytic infiltration of the kidneys, lungs, and salivary glands.
Collapse
Affiliation(s)
- Jacen S. Maier-Moore
- Department of Clinical Laboratory Sciences, University of Texas at El Paso, El Paso, Texas
- The Department of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK, 73014
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104
| | - Christopher G. Horton
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104
- The Department of Microbiology and Immunology, OUHSC, Oklahoma City, OK, 73014
| | - Shirley A. Mathews
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104
| | - Anthony W. Confer
- The Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, 74078
| | - Christina Lawrence
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104
| | - Zijian Pan
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104
| | | | - A. Darise Farris
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK, 73104
- The Department of Microbiology and Immunology, OUHSC, Oklahoma City, OK, 73014
| |
Collapse
|
42
|
Hodge DL, Berthet C, Coppola V, Kastenmüller W, Buschman MD, Schaughency PM, Shirota H, Scarzello AJ, Subleski JJ, Anver MR, Ortaldo JR, Lin F, Reynolds DA, Sanford ME, Kaldis P, Tessarollo L, Klinman DM, Young HA. IFN-gamma AU-rich element removal promotes chronic IFN-gamma expression and autoimmunity in mice. J Autoimmun 2014; 53:33-45. [PMID: 24583068 PMCID: PMC4148478 DOI: 10.1016/j.jaut.2014.02.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 01/17/2014] [Accepted: 02/05/2014] [Indexed: 12/31/2022]
Abstract
We generated a mouse model with a 162 nt AU-rich element (ARE) region deletion in the 3' untranslated region (3'UTR) of the interferon-gamma (IFN-γ) gene that results in chronic circulating serum IFN-γ levels. Mice homozygous for the ARE deletion (ARE-Del) (-/-) present both serologic and cellular abnormalities typical of patients with systemic lupus erythematosus (SLE). ARE-Del(-/-) mice display increased numbers of pDCs in bone marrow and spleen. Addition of IFN-γ to Flt3-ligand (Flt3L) treated in vitro bone marrow cultures results in a 2-fold increase in pDCs with concurrent increases in IRF8 expression. Marginal zone B (MZB) cells and marginal zone macrophages (MZMs) are absent in ARE-Del(-/-) mice. ARE-Del(+/-) mice retain both MZB cells and MZMs and develop no or mild autoimmunity. However, low dose clodronate treatment in ARE-Del(+/-) mice specifically eliminates MZMs and promotes anti-DNA antibody development and glomerulonephritis. Our findings demonstrate the consequences of a chronic IFN-γ milieu on B220(+) cell types and in particular the impact of MZB cell loss on MZM function in autoimmunity. Furthermore, similarities between disease states in ARE-Del(-/-) mice and SLE patients suggest that IFN-γ may not only be a product of SLE but may be critical for disease onset and progression.
Collapse
Affiliation(s)
- Deborah L Hodge
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Cyril Berthet
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702, USA; Oncodesign, 20 Rue Jean Mazen, Dijon 21076, France.
| | - Vincenzo Coppola
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702, USA; Department of Molecular Virology, Immunology and Medical Genetics at The Ohio State University, 460 W. 12th Street, Columbus, OH 43210, USA.
| | - Wolfgang Kastenmüller
- Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Cellular Interactions and Immunimaging Institutes of Molecular Medicine and Experimental Immunology (IMMEI), University of Bonn, Sigmund-Freud Str. 25, Bonn 53105, Germany.
| | - Matthew D Buschman
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA; Division of Endocrinology and Metabolism, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Paul M Schaughency
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA; Department of Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hidekazu Shirota
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA; Department of Clinical Oncology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Anthony J Scarzello
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Jeff J Subleski
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Miriam R Anver
- Laboratory Animal Science Program (LASP), Science Applications International Corporation (SAIC), National Cancer Institute, Frederick, MD 21702, USA.
| | - John R Ortaldo
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Fanching Lin
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Della A Reynolds
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Michael E Sanford
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Philipp Kaldis
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702, USA; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Proteos, 61 Biopolis Drive, Singapore 138673, Republic of Singapore.
| | - Lino Tessarollo
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702, USA.
| | - Dennis M Klinman
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| | - Howard A Young
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, MD 21702, USA.
| |
Collapse
|
43
|
Qin M, Guo Y, Jiang L, Wang X. Elevated levels of serum sCXCL16 in systemic lupus erythematosus; potential involvement in cutaneous and renal manifestations. Clin Rheumatol 2014; 33:1595-601. [PMID: 25015061 DOI: 10.1007/s10067-014-2741-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 07/01/2014] [Accepted: 07/03/2014] [Indexed: 11/28/2022]
Abstract
The aim of this study was to investigate the levels and clinical significance of serum soluble chemokine (C-X-C motif) ligand 16 (sCXCL16) in patients with systemic lupus erythematosus (SLE), as well as the sCXCL16 molecule's associations with disease activity and organ damage. Thirty-five patients with SLE, 16 patients with rheumatoid arthritis (RA), and 15 healthy controls were included in this study. The demographic and clinical features of the patients were recorded. The serum levels of sCXCL16 were determined. Disease activity was assessed using the SLE Disease Activity Index (SLEDAI), and organ damage was evaluated with the Systemic Lupus International Collaborating Clinics/American College of Rheumatology (SLICC/ACR) Damage Index (SDI). The serum levels of sCXCL16 in the patients with SLE were higher than those in the patients with RA (P = 0.002) or healthy controls (P < 0.0001). The levels in the patients with active SLE were higher than those in the disease inactive patients (P = 0.008). Positive correlations were identified between serum sCXCL16 concentrations and both SLEDAI (r = 0.564; P < 0.0001) and SDI scores (r = 0.396; P = 0.018). Both SLEDAI (P = 0.021) and serum levels of CXCL16 (P = 0.023) decreased after conventional treatment in 12 initial onset cases of SLE patients. Elevated serum sCXCL16 levels were discovered in the SLE patients with cutaneous (P = 0.006) and renal involvement (P = 0.032). Soluble CXCL16 may become a useful serological marker of disease activity and skin and renal involvement in SLE patients; thus, it may be used for evaluation of therapeutic interventions.
Collapse
Affiliation(s)
- Muting Qin
- Department of Rheumatology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | | | | | | |
Collapse
|
44
|
Stem cell-based cell therapy for glomerulonephritis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:124730. [PMID: 25003105 PMCID: PMC4070530 DOI: 10.1155/2014/124730] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 03/27/2014] [Indexed: 01/09/2023]
Abstract
Glomerulonephritis (GN), characterized by immune-mediated inflammatory changes in the glomerular, is a common cause of end stage renal disease. Therapeutic options for glomerulonephritis applicable to all cases mainly include symptomatic treatment and strategies to delay progression. In the attempt to yield innovative interventions fostering the limited capability of regeneration of renal tissue after injury and the uncontrolled pathological process by current treatments, stem cell-based therapy has emerged as novel therapy for its ability to inhibit inflammation and promote regeneration. Many basic and clinical studies have been performed that support the ability of various stem cell populations to ameliorate glomerular injury and improve renal function. However, there is a long way before putting stem cell-based therapy into clinical practice. In the present article, we aim to review works performed with respect to the use of stem cell of different origins in GN, and to discuss the potential mechanism of therapeutic effect and the challenges for clinical application of stem cells.
Collapse
|
45
|
Kato H, Perl A. Mechanistic target of rapamycin complex 1 expands Th17 and IL-4+ CD4-CD8- double-negative T cells and contracts regulatory T cells in systemic lupus erythematosus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:4134-44. [PMID: 24683191 PMCID: PMC3995867 DOI: 10.4049/jimmunol.1301859] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is activated in CD4(-)CD8(-) double-negative (DN) T cells and its blockade is therapeutic in systemic lupus erythematosus (SLE) patients. Murine studies showed the involvement of mTOR complex 1 (mTORC1) and 2 (mTORC2) in the differentiation of Th1/Th17 cells and Th2 cells, respectively. In this study, we investigated the roles of mTORC1 and mTORC2 in T cell lineage development in SLE and matched healthy control (HC) subjects. mTORC1 activity was increased, whereas mTORC2 was reduced, as assessed by phosphorylation of their substrates phosphorylated S6 kinase 1 or phosphorylated S6 ribosomal protein and phosphorylated Akt, respectively. Rapamycin inhibited mTORC1 and enhanced mTORC2. IL-4 expression was increased in freshly isolated CD8(+) lupus T cells (SLE: 8.09 ± 1.93%, HC: 3.61 ± 0.49%; p = 0.01). DN T cells had greater IL-4 expression than CD4(+) or CD8(+) T cells of SLE patients after 3-d in vitro stimulation, which was suppressed by rapamycin (control: 9.26 ± 1.48%, rapamycin: 5.03 ± 0.66%; p < 0.001). GATA-3 expression was increased in CD8(+) lupus T cells (p < 0.01) and was insensitive to rapamycin treatment. IFN-γ expression was reduced in all lupus T cell subsets (p = 1.0 × 10(-5)) and also resisted rapamycin. IL-17 expression was increased in CD4(+) lupus T cells (SLE: 3.62 ± 0.66%, HC: 2.29 ± 0.27%; p = 0.019), which was suppressed by rapamycin (control: 3.91 ± 0.79%, rapamycin: 2.22 ± 0.60%; p < 0.001). Frequency of regulatory T cells (Tregs) was reduced in SLE (SLE: 1.83 ± 0.25%, HC: 2.97 ± 0.27%; p = 0.0012). Rapamycin inhibited mTORC1 in Tregs and promoted their expansion. Neutralization of IL-17, but not IL-4, also expanded Tregs in SLE and HC subjects. These results indicate that mTORC1 expands IL-4(+) DN T and Th17 cells, and contracts Tregs in SLE.
Collapse
Affiliation(s)
- Hiroshi Kato
- Division of Rheumatology, Departments of Medicine, Microbiology and Immunology, and Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York 13210
| | - Andras Perl
- Division of Rheumatology, Departments of Medicine, Microbiology and Immunology, and Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York 13210
| |
Collapse
|
46
|
McCarthy EM, Smith S, Lee RZ, Cunnane G, Doran MF, Donnelly S, Howard D, O'Connell P, Kearns G, Ní Gabhann J, Jefferies CA. The association of cytokines with disease activity and damage scores in systemic lupus erythematosus patients. Rheumatology (Oxford) 2014; 53:1586-94. [PMID: 24706988 DOI: 10.1093/rheumatology/ket428] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE The aim of this study was to explore the role of cytokines in the pathogenesis of SLE in a genetically homogeneous Caucasian SLE patient population. METHODS Serum levels of the following cytokines were determined by ELISA in SLE patients (diagnosed as per ACR diagnostic criteria): IL-1β, IL-10, IL-12p70 and TNF-α. Demographic data, disease activity as per the SLEDAI and damage scores (SLICC) at the 5-year follow-up were calculated. RESULTS Enhanced production of TNF-α, IL-1 and IL-10 were observed in SLE patients compared with controls. A strong positive correlation was seen between levels of IL-12p70 and IL-10. In addition, IL-10, TNF-α and IL-1 demonstrated a significant relationship with disease activity. Interestingly, elevated levels of IL-10 were observed in SLE patients with CNS involvement while patients with elevated levels of TNF-α were more likely to have renal involvement and sustain damage over the follow-up period. Additionally, the ratio of all cytokines assayed to IL-12p70 levels were significantly higher in SLE patients when compared with controls, with an association seen between damage accrual and the IL-1β/IL-12p70 ratio (r = 0.431, P = 0.003), IL-10/IL-12p70 ratio (r = 0.351, P = 0.018) and TNF-α/IL-12p70 ratio (r = 0.33, P = 0.028). When the respective ratios were analysed for organ-specific disease, significant differences were observed for the IL-1β/IL-12p70 ratio (0.79 vs 0.47, P = 0.036), IL-10/IL-12p70 ratio (4.29 vs 1.87, P = 0.018) and TNF-α/IL-12p70 ratio (7.49 vs 5.21, P = 0.018) with respect to renal involvement. CONCLUSION Increased levels of a number of immunomodulatory cytokines relative to IL-12p70 in this Caucasian SLE patient population are seen in patients with renal involvement and are associated with increased accrual of damage at the 5-year follow-up.
Collapse
Affiliation(s)
- Eoghan M McCarthy
- Department of Rheumatology, Beaumont Hospital, Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Department of Rheumatology, St James Hospital and Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland.Department of Rheumatology, Beaumont Hospital, Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Department of Rheumatology, St James Hospital and Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Siobhán Smith
- Department of Rheumatology, Beaumont Hospital, Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Department of Rheumatology, St James Hospital and Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Ruth Z Lee
- Department of Rheumatology, Beaumont Hospital, Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Department of Rheumatology, St James Hospital and Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Gaye Cunnane
- Department of Rheumatology, Beaumont Hospital, Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Department of Rheumatology, St James Hospital and Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Michele F Doran
- Department of Rheumatology, Beaumont Hospital, Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Department of Rheumatology, St James Hospital and Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Suzanne Donnelly
- Department of Rheumatology, Beaumont Hospital, Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Department of Rheumatology, St James Hospital and Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Donough Howard
- Department of Rheumatology, Beaumont Hospital, Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Department of Rheumatology, St James Hospital and Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Paul O'Connell
- Department of Rheumatology, Beaumont Hospital, Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Department of Rheumatology, St James Hospital and Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Grainne Kearns
- Department of Rheumatology, Beaumont Hospital, Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Department of Rheumatology, St James Hospital and Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Joan Ní Gabhann
- Department of Rheumatology, Beaumont Hospital, Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Department of Rheumatology, St James Hospital and Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Caroline A Jefferies
- Department of Rheumatology, Beaumont Hospital, Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Department of Rheumatology, St James Hospital and Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland.
| |
Collapse
|
47
|
Tsai YG, Lee CY, Lin TY, Lin CY. CD8⁺ Treg cells associated with decreasing disease activity after intravenous methylprednisolone pulse therapy in lupus nephritis with heavy proteinuria. PLoS One 2014; 9:e81344. [PMID: 24475019 PMCID: PMC3903465 DOI: 10.1371/journal.pone.0081344] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 10/11/2013] [Indexed: 01/26/2023] Open
Abstract
We focus on the role of CD8+ Treg cell in Intravenous methyl-prednisolone (IVMP) pulse therapy in forty patients with active Class III/IV childhood lupus nephritis (LN) with heavy proteinuria. IVMP therapy for five days. From peripheral blood mononuclear cells (PBMCs) and renal tissues, we saw IVMP therapy definitely restoring both CD4+CD25+FoxP3+ and CD8+CD25+Foxp3+ Treg cell number plus greater expression with intracellular IL-10 and granzyme B in CD8+FoxP3+ Treg from PBMCs. IVMP-treated CD8+CD25+ Treg cells directly suppressed CD4+ T proliferation and induced CD4+CD45RO+ apoptosis. Histologically, CD4+FoxP3+ as well as CD8+FoxP3+ Treg cells appeared in renal tissue of LN patients before IVMP by double immunohistochemical stain. CD8+FoxP3+ Treg cells increased in 10 follow-up renal biopsy specimens after IVMP. Reverse correlation of serum anti-C1q antibody and FoxP3+ Treg cells in PBMNCs (r = −0.714, P<0.01). After IVMP, serum anti-C1q antibody decrease accompanied increase of CD4+FoxP3+ Treg cells. CD8+Treg cells reduced interferon-r response in PBMCs to major peptide autoepitopes from nucleosomes after IVMP therapy; siRNA of FoxP3 suppressed granzyme B expression while decreasing CD8+CD25+Treg-induced CD4+CD45RO+ apoptosis. Renal activity of LN by SLEDAI-2k in childhood LN was significantly higher than two weeks after IVMP (P<0.01). CD8+FoxP3+ Treg cells return in post-IVMP therapy and exert crucial immune modulatory effect to control autoimmune response in LN.
Collapse
Affiliation(s)
- Yi-Giien Tsai
- Departments of Pediatrics, Changhua Christian Hospital, Changhua, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Ying Lee
- Division of Pediatric Nephrology, China Medical University Hospital, Taichung, Taiwan
| | - Tze-Yi Lin
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | - Ching-Yuang Lin
- Division of Pediatric Nephrology, China Medical University Hospital, Taichung, Taiwan
- Clinical Immunology Center, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
48
|
Rovin BH, Parikh SV. Lupus nephritis: the evolving role of novel therapeutics. Am J Kidney Dis 2014; 63:677-90. [PMID: 24411715 DOI: 10.1053/j.ajkd.2013.11.023] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Accepted: 11/15/2013] [Indexed: 11/11/2022]
Abstract
Immune complex accumulation in the kidney is the hallmark of lupus nephritis and triggers a series of events that result in kidney inflammation and injury. Cytotoxic agents and corticosteroids are standard of care for lupus nephritis treatment, but are associated with considerable morbidity and suboptimal outcomes. Recently, there has been interest in using novel biologic agents and small molecules to treat lupus nephritis. These therapies can be broadly categorized as anti-inflammatory (laquinamod, anti-tumor necrosis factor-like weak inducer of apotosis, anti-C5, and retinoids), antiautoimmunity (anti-CD20, anti-interferon α, and costimulatory blockers), or both (anti-interleukin 6 and proteasome inhibitors). Recent lupus nephritis clinical trials applied biologics or small molecules of any category to induction treatment, seeking short-term end points of complete renal response. These trials in general have not succeeded. When lupus nephritis comes to clinical attention during the inflammatory stage of the disease, the autoimmune stage leading to kidney inflammation will have been active for some time. The optimal approach for using novel therapies may be to initially target kidney inflammation to preserve renal parenchyma, followed by suppression of autoimmunity. In this review, we discuss novel lupus nephritis therapies and how they fit into a combinatorial treatment strategy based on the pathogenic stage.
Collapse
Affiliation(s)
- Brad H Rovin
- Division of Nephrology, Ohio State University Wexner Medical Center, Columbus, OH.
| | - Samir V Parikh
- Division of Nephrology, Ohio State University Wexner Medical Center, Columbus, OH
| |
Collapse
|
49
|
Postal M, Peliçari KO, Sinicato NA, Marini R, Costallat LTL, Appenzeller S. Th1/Th2 cytokine profile in childhood-onset systemic lupus erythematosus. Cytokine 2013; 61:785-91. [PMID: 23332615 DOI: 10.1016/j.cyto.2012.11.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 10/15/2012] [Accepted: 11/26/2012] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To determine the serum levels of Th1 (IL-12, IFN-γ,TNF-α) and Th2 (IL-5, IL-6 and IL-10) cytokines in childhood-onset SLE, first-degree relatives and healthy controls. To elucidate their association with disease activity, laboratory and treatment features. METHODS We included 60 consecutive childhood-onset SLE patients [median age 18 years (range 10-37)], 64 first-degree relatives [median 40 (range 28-52)] and 57 healthy [median age 19 years (range 6-30 years)] controls. Controls were age and sex-matched to SLE patients. SLE patients were assessed for clinical and laboratory SLE manifestations, disease activity (SLEDAI), damage (SDI) and current drug exposures. Mood and anxiety disorders were determined through Becks Depression (BDI) and Anxiety Inventory (BAI). Th1 (IL-12, IFN-γ,TNF-α) and Th2 (IL-5, IL-6 and IL-10) cytokines levels were measured by ELISA and compared by non-parametric tests. RESULTS Serum TNF-α (p=0.004), IL-6 (p=0.007) and IL-10 (p=0.03) levels were increased in childhood-onset SLE patients when compared to first-degree relatives and healthy controls. TNF-α levels were significantly increased in patients with active disease (p=0.014) and correlated directly with SLEDAI scores (r=0.39; p=0.002). IL-12 (p=0.042) and TNF-α (p=0.009) levels were significantly increased in patients with nephritis and TNF-α in patients with depression (p=0.001). No association between cytokine levels and SDI scores or medication was observed. CONCLUSION Th1 cytokines may play a role in the pathogenesis of neuropsychiatric and renal manifestations in childhood-onset SLE. The correlation with SLEDAI suggests that TNF-α may be a useful biomarker for disease activity in childhood-onset SLE, however longitudinal studies are necessary to determine if increase of this cytokine may predict flares in childhood-onset SLE.
Collapse
Affiliation(s)
- Mariana Postal
- Department of Medicine, Rheumatology Unit, Faculty of Medical Science, State University of Campinas, Brazil
| | | | | | | | | | | |
Collapse
|
50
|
Effect of Sanqi Oral Liquid on the expressions of CD4⁺, CD8⁺ and CD68⁺ cells in 5/6 nephrectomized rats with chronic renal failure. Chin J Integr Med 2012; 19:589-95. [PMID: 23212567 DOI: 10.1007/s11655-012-1233-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To explore the mechanisms of Chinese herbal medicine Sanqi Oral Liquid, composed of Astragalus membranaceus and Panpax notoginseng, in alleviating renal injury by observing its effect on the expressions of CD4(+), CD8(+) and CD68(+) cells in 5/6 nephrectomized rats with chronic renal failure. METHODS A total of 102 SD rats were randomly divided into six groups: three treatment groups were administrated with high, medium and low dosage of Sanqi Oral Liquid respectively by gavage; a normal group, a 5/6 nephrectomized model group, and a group treated with coated aldehyde oxygenstarch were used as controls. Following oral administration of Sanqi Oral Liquid for 12 weeks, the general condition and renal pathological changes were observed, and the renal function, platelet count (PLT) and the expressions of CD4(+), CD8(+) and CD68(+) cells were determined for each group. RESULTS There were proliferation of mesangial matrix, renaltubularnecrosis and obvious tubulointerstitial fibrosis in the model group, and they were much milder in the treatment groups. Compared with the model group, the amounts of blood urea nitrogen (BUN), serum creatinine (Scr) and PLT in the treatment groups decreased (P<0.05 for all); and in the group administrated of medium dosage of Sanqi Oral Liquid, the expression of CD4(+) cells was up-regulated and those of CD8(+) and CD68(+) cells were down-regulated (P<0.05 for all), leading to an increased ratio of CD4(+)/CD8(+)(P<0.01). CONCLUSION Sanqi Oral Liquid has a significant effect on regulating lymphocyte subsets, reducing the infiltration of macrophages in renal tissues and alleviating tubulointerstitial fibrosis, and this may be one of mechanisms of Sanqi Oral Liquid in delaying the progression of chronic kidney diseases.
Collapse
|