51
|
Tailliar M, Schanstra JP, Dierckx T, Breuil B, Hanouna G, Charles N, Bascands JL, Dussol B, Vazi A, Chiche L, Siwy J, Faguer S, Daniel L, Daugas E, Jourde-Chiche N. Urinary Peptides as Potential Non-Invasive Biomarkers for Lupus Nephritis: Results of the Peptidu-LUP Study. J Clin Med 2021; 10:jcm10081690. [PMID: 33920017 PMCID: PMC8071029 DOI: 10.3390/jcm10081690] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Lupus nephritis (LN) is a severe manifestation of Systemic Lupus Erythematosus (SLE). The therapeutic strategy relies on kidney biopsy (KB) results. We tested whether urinary peptidome analysis could non-invasively differentiate active from non-active LN. Design: Urinary samples were collected from 93 patients (55 with active LN and 38 with non-active LN), forming a discovery (n = 42) and an independent validation (n = 51) cohort. Clinical characteristics were collected at inclusion and prospectively for 24 months. The urinary peptidome was analyzed by capillary-electrophoresis coupled to mass-spectrometry, comparing active LN to non-active LN, and assessing chronic lesions and response to therapy. The value of previously validated prognostic (CKD273) and differential diagnostic (LN172) signatures was evaluated. Results: Urinary peptides could not discriminate between active and non-active LN or predict early response to therapy. Tubulo-interstitial fibrosis was correlated to the CKD273. The LN172 score identified 92.5% of samples as LN. Few patients developed new-onset CKD. Conclusions: We validated the CKD273 and LN172 classifiers but did not identify a robust signature that could predict active LN and replace KB. The value of urinary peptidome to predict long-term CKD, or renal flares in SLE, remains to be evaluated.
Collapse
Affiliation(s)
- Maxence Tailliar
- AP-HM, Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, 13005 Marseille, France; (M.T.); (B.D.)
| | - Joost P. Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institut of Metabolic and Cardiovascular Disease (I2MC), 31432 Toulouse, France; (J.P.S.); (B.B.); (S.F.)
- Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Tim Dierckx
- Laboratory of Clinical and Epidemiological Virology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium;
| | - Benjamin Breuil
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institut of Metabolic and Cardiovascular Disease (I2MC), 31432 Toulouse, France; (J.P.S.); (B.B.); (S.F.)
- Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Guillaume Hanouna
- AP-HP, Service de Néphrologie, Hôpital Bichat, DMU VICTOIRE, 75018 Paris, France; (G.H.); (E.D.)
| | - Nicolas Charles
- Centre de Recherche sur l’Inflammation, Université de Paris, INSERM UMRS1149, CNRS ERL8252, Labex INFLAMEX, DHU FIRE, 75890 Paris, France;
| | - Jean-Loup Bascands
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1188-Université de La Réunion, 97490 Saint-Denis, France;
| | - Bertrand Dussol
- AP-HM, Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, 13005 Marseille, France; (M.T.); (B.D.)
- Centre d’Investigation Clinique, CHU Conception, AP-HM, 13005 Marseille, France;
| | - Alain Vazi
- Centre d’Investigation Clinique, CHU Conception, AP-HM, 13005 Marseille, France;
| | - Laurent Chiche
- Médecine Interne, Hôpital Européen, 13003 Marseille, France;
| | - Justyna Siwy
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany;
| | - Stanislas Faguer
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institut of Metabolic and Cardiovascular Disease (I2MC), 31432 Toulouse, France; (J.P.S.); (B.B.); (S.F.)
- Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
- CHU de Toulouse, Service de Néphrologie, 31300 Toulouse, France
| | - Laurent Daniel
- AP-HM, Laboratoire d’Ananatomie Pathologique, Hôpital de la Timone, 13005 Marseille, France;
- Center for CardioVascular and Nutrition Research (C2VN), Aix-Marseille University, INSERM, INRAE, 13005 Marseille, France
| | - Eric Daugas
- AP-HP, Service de Néphrologie, Hôpital Bichat, DMU VICTOIRE, 75018 Paris, France; (G.H.); (E.D.)
- Centre de Recherche sur l’Inflammation, Université de Paris, INSERM UMRS1149, CNRS ERL8252, Labex INFLAMEX, DHU FIRE, 75890 Paris, France;
| | - Noémie Jourde-Chiche
- AP-HM, Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, 13005 Marseille, France; (M.T.); (B.D.)
- Center for CardioVascular and Nutrition Research (C2VN), Aix-Marseille University, INSERM, INRAE, 13005 Marseille, France
- Correspondence:
| | | |
Collapse
|
52
|
Abdelazeem ME, Abdelhaleem MI, Mohamed RA, Abdelaleem EA. The role of Dickkopf-1 as a biomarker in systemic lupus erythematosus and active lupus nephritis. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2021. [DOI: 10.1186/s43166-021-00064-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Abstract
Background
Systemic lupus erythematosus (SLE) is a chronic disease which is mainly attributed to autoantibodies, cytokines, and immune complex deposition. Studies have demonstrated that cytokines and autoantibodies were strongly associated with renal diseases and can be used for the prediction of patients with lupus nephritis (LN). However, antibodies to dsDNA and the reduction of complements were also detected in non-LN patients as well as clinically non-active SLE patients. The current study was performed to detect the role of serum DKK-1 as a biomarker for the identification of SLE patients and patients with LN and its relation to disease activity and severity. The study was conducted on fifty clinically diagnosed SLE patients who were diagnosed according to Systemic Lupus International Collaborating Clinics (SLICC) classification criteria for SLE, in addition to thirty healthy control volunteers matched for age and sex. Assessment of SLE disease activity was done using Systemic Lupus Erythematosus Disease Activity Index (SLEDAI). Assessment of SLE disease severity was done using the Systemic Lupus International Collaborative Clinics/American College of Rheumatology (SLICC/ACR) damage index. Serum levels of DKK-1 were measured for all participants by ELISA using commercially available kits.
Results
DKK-1 serum levels were significantly higher among active lupus nephritis cases as compared with SLE cases with no LN and with healthy controls (9197.60 μg/uL ± 2939.2 μg/uL vs. 6405.15 μg/uL ± 2018.91 μg/uL vs. 2790.33 μg/uL ± 833.49 μg/uL) respectively (p-values < 0.001). DKK-1 concentration was significantly higher among SLE patients with positive as compared with negative anti-double-stranded DNA (dsDNA) antibodies (p-value < 0.001). According to receiver operating characteristic (ROC) curve analysis, serum DKK-1 level diagnosed the SLE at a statistically significant level with a 98% sensitivity and 70% specificity and serum DKK-1 level also diagnosed active lupus nephritis at a 90% sensitivity and 63% specificity.
Conclusion
DKK-1 could diagnose SLE and lupus nephritis with high sensitivity and specificity. Serum DKK-1 is a reliable biomarker for the identification of SLE and patients with LN and could be used as a key molecule for the diagnosis of SLE and as a prognostic indicator of LN.
Collapse
|
53
|
Tan L, Zhao M, Wu H, Zhang Y, Tong X, Gao L, Zhou L, Lu Q, Zeng J. Downregulated Serum Exosomal miR-451a Expression Correlates With Renal Damage and Its Intercellular Communication Role in Systemic Lupus Erythematosus. Front Immunol 2021; 12:630112. [PMID: 33643314 PMCID: PMC7906989 DOI: 10.3389/fimmu.2021.630112] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease characterized by continuous inflammation and the production of autoantibodies. Exosomes, acting as a critical tool for communication between cells, are involved in the pathogenesis of SLE, particularly in inflammation and immune imbalance. In this study, we aimed to extract and confirm the pro-inflammatory effect of serum exosomes in SLE. Then, we attempted to find differentially expressed exosomal microRNAs in the serum of healthy subjects and SLE patients via miRNA microarray analysis and validated the target exosomal microRNA, exosomal miR-451a, which expression level decreased in serum of SLE patients by RT-qPCR. Furtherly, we analyzed the correlation between exosomal miR-451a and disease activity, kidney damage and typing, and traditional medicine therapy. Finally, we investigated the intercellular communication role of exosomal miR-451a in SLE by co-culture assay in vitro. Taken together, our study demonstrated that downregulated serum exosomal miR-451a expression correlated with SLE disease activity and renal damage as well as its intercellular communication role in SLE which provided potential therapeutic strategies.
Collapse
Affiliation(s)
- Lina Tan
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yuezhong Zhang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiaoliang Tong
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Gao
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lu Zhou
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jinrong Zeng
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
54
|
Zhang T, Duran V, Vanarsa K, Mohan C. Targeted urine proteomics in lupus nephritis - a meta-analysis. Expert Rev Proteomics 2021; 17:767-776. [PMID: 33423575 DOI: 10.1080/14789450.2020.1874356] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND Proteomic approaches are central in biomarker discovery. While mass-spectrometry-based techniques are widely used, novel targeted proteomic platforms have enabled the high-throughput detection of low-abundance proteins in an affinity-based manner. Urine has gained growing attention as an ideal biofluid for monitoring renal disease including lupus nephritis (LN). METHODS Pubmed was screened for targeted proteomic studies of LN urine interrogating ≥1000 proteins. Data from the primary studies were combined and a meta-analysis was performed. Shared proteins elevated in active LN across studies were identified, and relevant pathways were elucidated using ingenuity pathway and gene ontology analysis. Urine proteomic data was cross-referenced against renal single-cell RNAseq data from LN kidneys. RESULTS Two high-throughput targeted proteomic platforms with capacity to interrogate ≥1000 proteins have been used to investigate LN urine. Twenty-three urine proteins were significantly elevated in both studies, including 10 chemokines, and proteins implicated in angiogenesis, and extracellular matrix turnover. Of these, Cathepsin S, CXCL10, FasL, ferritin, macrophage migration inhibitory factor (MIF), and resistin were also significantly elevated within LN kidneys. CONCLUSION Targeted urinary proteomics have uncovered multiple novel biomarkers for LN. Further validation in prospective cohorts and mechanistic studies are warranted to establish their clinical utility.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Biomedical Engineering, University of Houston , Houston, Texas, USA
| | - Valeria Duran
- Department of Biomedical Engineering, University of Houston , Houston, Texas, USA
| | - Kamala Vanarsa
- Department of Biomedical Engineering, University of Houston , Houston, Texas, USA
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston , Houston, Texas, USA
| |
Collapse
|
55
|
Moloi MW, Rusch JA, Omar F, Ekrikpo U, Dandara C, Bello AK, Jayne D, Okpechi IG. Urinary MCP-1 and TWEAK as non-invasive markers of disease activity and treatment response in patients with lupus nephritis in South Africa. Int Urol Nephrol 2021; 53:1865-1873. [PMID: 33459955 DOI: 10.1007/s11255-020-02780-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/26/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Treatment of patients with lupus nephritis (LN) requires judicious use of immunosuppression. Novel biomarkers may be useful for monitoring disease activity and treatment response. We assessed the utility of urinary monocyte chemoattractant protein-1 (uMCP-1) and urinary tumour necrosis factor-like weak inducer of apoptosis (uTWEAK) for disease activity and treatment response monitoring in South Africans with LN. METHODS We recruited consenting patients with active LN confirmed on kidney biopsy. Urinary levels of MCP-1 and TWEAK were assayed at baseline and after completion of induction therapy using ELISA methods. We also collected relevant demographic, clinical and biochemical data for patients included in this study. RESULTS The mean age of patients in this study was 29.8 ± 10.7 years, 60% were patients of mixed ancestry, 70% had proliferative LN and mean spot urine proteinuria at baseline was 0.37 (0.18-0.59) g/mmolCr. At completion of induction therapy, the level of uMCP-1 had reduced to 314.5 (IQR: 197.0-622) pg/mgCr from a baseline of 1092.7 (IQR 578.6-1848) pg/mgCr (P = 0.06) while uTWEAK had reduced to 36.0 (IQR 17.0-88.0) pg/mgCr from 159.0 (IQR: 88.5-295.5) pg/mgCr (P = 0.03). For patients reaching early complete or partial remission (n = 17), both biomarkers had significantly declined in their urine: uMCP-1 (P = 0.018) and uTWEAK (P = 0.015). There was no reduction of both biomarkers in patients not achieving remission and no association between uMCP-1 or uTWEAK with renal histological features. CONCLUSION Our study shows that uMCP-1 and uTWEAK are elevated in patients with active LN, correlated with the remission status (response to treatment) at the end of induction therapy and can, therefore, be useful for monitoring disease activity and treatment response.
Collapse
Affiliation(s)
- Mothusi W Moloi
- Division of Nephrology and Hypertension, University of Cape Town, Cape Town, South Africa.,Kidney and Hypertension Research Unit, University of Cape Town Cape Town, Cape Town, South Africa.,Department of Medicine, University of Botswana, Gaborone, Botswana
| | - Jody A Rusch
- Division of Chemical Pathology, University of Cape Town and National Health Laboratory Services (NHLS), Cape Town, South Africa
| | - Fierdoz Omar
- Division of Chemical Pathology, University of Cape Town and National Health Laboratory Services (NHLS), Cape Town, South Africa
| | - Udeme Ekrikpo
- Division of Nephrology and Hypertension, University of Cape Town, Cape Town, South Africa.,Kidney and Hypertension Research Unit, University of Cape Town Cape Town, Cape Town, South Africa.,Department of Medicine, University of Uyo, Uyo, Nigeria
| | - Collet Dandara
- Division of Human Genetics, University of Cape Town, Cape Town, South Africa
| | - Aminu K Bello
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - David Jayne
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ikechi G Okpechi
- Division of Nephrology and Hypertension, University of Cape Town, Cape Town, South Africa. .,Kidney and Hypertension Research Unit, University of Cape Town Cape Town, Cape Town, South Africa. .,Department of Medicine, University of Alberta, Edmonton, Canada.
| |
Collapse
|
56
|
Mok CC, Mohan C. Urinary Biomarkers in Lupus Nephritis: Are We There Yet? Arthritis Rheumatol 2021; 73:194-196. [PMID: 32892509 DOI: 10.1002/art.41508] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 02/02/2023]
|
57
|
Liu XR, Qi YY, Zhao YF, Cui Y, Wang XY, Zhao ZZ. Albumin-to-globulin ratio (AGR) as a potential marker of predicting lupus nephritis in Chinese patients with systemic lupus erythematosus. Lupus 2021; 30:412-420. [PMID: 33407045 DOI: 10.1177/0961203320981139] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVES To evaluate a potential role of albumin-to-globulin ratio (AGR) in the development of lupus nephritis (LN) and determine the potential to use AGR as a marker for future LN in systemic lupus erythematosus (SLE) patients. METHODS 194 newly diagnosed SLE patients without renal impairment were followed. The clinical data were collected and analyzed at the time of initial diagnosis of SLE and the end of follow-up. We compared baseline characteristics between those who did or did not develop LN on follow-up. Univariate and multivariate Cox hazard analysis were used to identify predictors of lupus nephritis. RESULTS Among the 194 newly diagnosed SLE patients without renal impairment, 26 (13.40%) patients were diagnosed with LN during a median follow-up of 53.87 months. On univariate Cox analysis, patients with the history of alopecia, higher SBP, lower AGR, lower CRP, lower C3, lower C4, higher anti-dsDNA Ab, presence of ANA homogeneous patterns or higher SLEDAI had an increased probability of developing LN. In a multivariate model, the history of alopecia (adjust hazard ratio, aHR = 3.614, 95%CI 1.365-9.571 P = 0.010), lower AGR (aHR = 6.968, 95%CI 1.873-25.919, P = 0.004), lower CRP (aHR = 4.230, 95%CI 1.591-11.247, P = 0.004) and higher level of anti-dsDNA (aHR = 2.675, 95%CI 1.008-7.093, P = 0.048) were independently associated with an increased risk of developing LN after adjusting for covariates. CONCLUSION Our findings indicated that SLE patients with low AGR, low CRP, high anti-dsDNA and the history of alopecia were more likely to develop LN in the course of SLE. AGR shown the greatest hazard for developing LN among them, it may be a strong predictor.
Collapse
Affiliation(s)
- Xin-Ran Liu
- Nephrology Hospital, The First Affiliated Hospital of Zhengzhou University, Henan, China.,Institute of Nephrology, Zhengzhou University, Henan, China
| | - Yuan-Yuan Qi
- Nephrology Hospital, The First Affiliated Hospital of Zhengzhou University, Henan, China.,Institute of Nephrology, Zhengzhou University, Henan, China
| | - Ya-Fei Zhao
- Nephrology Hospital, The First Affiliated Hospital of Zhengzhou University, Henan, China.,Institute of Nephrology, Zhengzhou University, Henan, China
| | - Yan Cui
- Nephrology Hospital, The First Affiliated Hospital of Zhengzhou University, Henan, China.,Institute of Nephrology, Zhengzhou University, Henan, China
| | - Xiao-Yang Wang
- Nephrology Hospital, The First Affiliated Hospital of Zhengzhou University, Henan, China.,Institute of Nephrology, Zhengzhou University, Henan, China
| | - Zhan-Zheng Zhao
- Nephrology Hospital, The First Affiliated Hospital of Zhengzhou University, Henan, China.,Institute of Nephrology, Zhengzhou University, Henan, China
| |
Collapse
|
58
|
Wang S, Wang F, Wang X, Zhang Y, Song L. Elevated Creatinine Clearance in Lupus Nephritis patients with Normal Creatinine. Int J Med Sci 2021; 18:1449-1455. [PMID: 33628102 PMCID: PMC7893553 DOI: 10.7150/ijms.51117] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 01/07/2021] [Indexed: 11/05/2022] Open
Abstract
Objectives: The present study aimed to observe the differences in creatinine clearance (Ccr) in systemic lupus erythematosus (SLE) patients with normal serum creatinine at different levels of urinary protein. Method: The present cross-sectional study included 177 SLE patients with normal serum creatinine from Qilu Hospital of Shandong University between January 2010 and April 2020. The following data were collected: blood urea nitrogen (BUN), serum creatinine (Cr), serum total protein, serum albumin, immunoglobulin (Ig) G, IgA, IgM, complement 3, complement 4, anti-ds-DNA antibody, routine urine test, urine protein/creatinine ratio (UPCR) (g/g), and the SLE disease activity index. The estimated Ccr was calculated according to the Cockcroft formula. Results: 123 patients were with positive urinary protein (Lupus Nephritis, LN group) and 54 patients were with negative urinary protein (Non-LN group). Compared with the Non-LN group, the LN group had higher BUN (5.76±3.22 vs. 4.78±1.58, P=0.007) and Cr (62.36±19.53 vs. 54.83±11.09, P=0.001). There was a strong correlation between the UPCR and the semi-quantitative determination of urine protein in LN patients (r=0.9583, P=0.0417). The serum creatinine levels were significantly higher in patients with urine protein 3+ (72.97±25.16) or massive proteinuria (62.32±19.66) than the other groups. Patients with urinary protein ± exhibited a significantly elevated Ccr when compared to patients with urinary protein 3+ (130.6±44.15 vs. 110.5±33.50, P=0.02), and patients with UPCR<0.15 g/g had higher Ccr than other groups and showed significantly increased Ccr compared with patients with UPCR≥0.15 g/g (132.44±21.02 vs. 115.14±35.89, P=0.007). Conclusions: Early renal function impairment may be present in LN patients. The kidneys of LN patients with urinary protein ± or UPCR<0.15 g/g are in a state of hyperfunction.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Rheumatology, Qilu Hospital of Shandong University, Ji'nan 250012, Shandong, China
| | - Fang Wang
- Department of Rheumatology, Qilu Hospital of Shandong University, Ji'nan 250012, Shandong, China
| | - Xiao Wang
- Department of Rheumatology, Qilu Hospital of Shandong University, Ji'nan 250012, Shandong, China
| | - Yuxian Zhang
- Department of Rheumatology, Qilu Hospital of Shandong University, Ji'nan 250012, Shandong, China
| | - Lijun Song
- Department of Rheumatology, Qilu Hospital of Shandong University, Ji'nan 250012, Shandong, China
| |
Collapse
|
59
|
Bourse Chalvon N, Orquevaux P, Giusti D, Gatouillat G, Tabary T, Tonye Libyh M, Chrusciel J, Drame M, Stockton-Bliard G, Amoura Z, Arnaud L, Lorenz HM, Blaison G, Bonnotte B, Magy-Bertrand N, Revuz S, Voll RE, Hinschberger O, Schwarting A, Pham BN, Martin T, Pennaforte JL, Servettaz A. Absence of Anti-Glomerular Basement Membrane Antibodies in 200 Patients With Systemic Lupus Erythematosus With or Without Lupus Nephritis: Results of the GOODLUPUS Study. Front Immunol 2020; 11:597863. [PMID: 33381119 PMCID: PMC7768036 DOI: 10.3389/fimmu.2020.597863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/10/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Anti-glomerular basement membrane (GBM) antibodies are pathogenic antibodies first detected in renal-limited anti-GBM disease and in Goodpasture disease, the latter characterized by rapidly progressive crescentic glomerulonephritis combined with intra-alveolar hemorrhage. Studies have suggested that anti-GBM antibody positivity may be of interest in lupus nephritis (LN). Moreover, severe anti-GBM vasculitis cases in patients with systemic lupus erythematosus (SLE) have been described in the literature, but few studies have assessed the incidence of anti-GBM antibodies in SLE patients. Objective The main study objective was to determine if positive anti-GBM antibodies were present in the serum of SLE patients with or without proliferative renal damage and compared to a healthy control group. Methodology This retrospective study was performed on SLE patients’ sera from a Franco-German European biobank, developed between 2011 and 2014, from 17 hospital centers in the Haut-Rhin region. Patients were selected according to their renal involvement, and matched by age and gender. The serum from healthy voluntary blood donors was also tested. Anti-GBM were screened by fluorescence enzyme immunoassay (FEIA), and then by indirect immunofluorescence (IIF) in case of low reactivity detection (titer >6 U/ml). Results The cohort was composed of 100 SLE patients with proliferative LN (27% with class III, 67% with class IV, and 6% with class V), compared to 100 SLE patients without LN and 100 controls. Patients were mostly Caucasian and met the ACR 1997 criteria and/or the SLICC 2012 criteria. Among the 300 tested sera, no significant levels of anti-GBM antibodies were detected (>10 U/ml) by the automated technique, three sera were found “ambivalent” (>7 U/ml): one in the SLE with LN group and two in the SLE without LN group. Subsequent IIF assays did not detect anti-GBM antibodies. Conclusion Anti-GBM antibodies were not detected in the serum of Caucasian patients with SLE, even in case of renal involvement, a situation favoring the antigenic exposure of glomerular basement membranes. Our results reaffirm the central role of anti-GBM antibodies as a specific diagnostic biomarker for Goodpasture vasculitis and therefore confirm that anti-GBM antibody must not be carried out in patients with SLE (with or without LN) in the absence of disease-suggestive symptoms.
Collapse
Affiliation(s)
- Nellie Bourse Chalvon
- Département de médecine interne, Centre Hospitalier Universitaire de Reims, Reims, France
| | - Pauline Orquevaux
- Département de médecine interne, Centre Hospitalier Universitaire de Reims, Reims, France
| | - Delphine Giusti
- Département d'immunologie biologique (laboratoire d'immunologie), Centre Hospitalier Universitaire de Reims, Reims, France
| | - Gregory Gatouillat
- Département d'immunologie biologique (laboratoire d'immunologie), Centre Hospitalier Universitaire de Reims, Reims, France
| | - Thierry Tabary
- Département d'immunologie biologique (laboratoire d'immunologie), Centre Hospitalier Universitaire de Reims, Reims, France
| | - Marcelle Tonye Libyh
- Département d'immunologie biologique (laboratoire d'immunologie), Centre Hospitalier Universitaire de Reims, Reims, France
| | - Jan Chrusciel
- Département d'information médicale et d'évaluation des performances, santé publique, Centre Hospitalier de Troyes, Troyes, France
| | - Moustapha Drame
- Département de Délégation à la Recherche Clinique et à l'Innovation, University Hospital of Martinique, Fort-de-France, Martinique
| | | | - Zahir Amoura
- Service de Médecine interne, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - Laurent Arnaud
- Service de Rhumatologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Clinic for Hematology, Oncology and Rheumatology, Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Gilles Blaison
- Département de médecine interne, Hôpital Pasteur, Colmar, France
| | - Bernard Bonnotte
- Département de Médecine Interne et d'immunologie Clinique, Centre Hospitalier Regional Universitaire De Dijon, Dijon, France
| | - Nadine Magy-Bertrand
- Département de médecine interne, Centre Hospitalier Universitaire de Besançon, Besancon, France
| | - Sabine Revuz
- Département de médecine interne, Hôpital Belle-Isle, Metz, France
| | - Reinhard Edmund Voll
- Department of Rheumatology and Clinical Immunology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Oliver Hinschberger
- Département de médecine interne, Groupe Hospitalier de la Région de Mulhouse et Sud Alsace (GHRMSA), Mulhouse, France
| | - Andreas Schwarting
- Universitäres Centrum für Autoimmunität Mainz, Johannes Gutenberg-Universität Mainz, Mainz, Germany
| | - Bach Nga Pham
- Département d'immunologie biologique (laboratoire d'immunologie), Centre Hospitalier Universitaire de Reims, Reims, France
| | - Thierry Martin
- Immunologie Clinique et Médecine Interne, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jean-Loup Pennaforte
- Département de médecine interne, Centre Hospitalier Universitaire de Reims, Reims, France
| | - Amelie Servettaz
- Département de médecine interne, Centre Hospitalier Universitaire de Reims, Reims, France
| |
Collapse
|
60
|
González LA, Ugarte-Gil MF, Alarcón GS. Systemic lupus erythematosus: The search for the ideal biomarker. Lupus 2020; 30:181-203. [PMID: 33307987 DOI: 10.1177/0961203320979051] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
During the last decades, there has been an increased interest in the discovery and validation of biomarkers that reliably reflect specific aspects of lupus. Although many biomarkers have been developed, few of them have been validated and used in clinical practice, but with unsatisfactory performances. Thus, there is still a need to rigorously validate many of these novel promising biomarkers in large-scale longitudinal studies and also identify better biomarkers not only for lupus diagnosis but also for monitoring and predicting upcoming flares and response to treatment. Besides serological biomarkers, urinary and cerebrospinal fluid biomarkers have emerged for assessing both renal and central nervous system involvement in systemic lupus erythematosus, respectively. Also, novel omics techniques help us to understand the molecular basis of the disease and also allow the identification of novel biomarkers which may be potentially useful for guiding new therapeutic targets.
Collapse
Affiliation(s)
- Luis Alonso González
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Universidad de Antioquia, Hospital Universitario de San Vicente Fundación, Medellín, Colombia
| | - Manuel Francisco Ugarte-Gil
- Rheumatology Department, Hospital Guillermo Almenara Irigoyen, EsSalud, Lima, Perú.,School of Medicine, Universidad Científica del Sur, Lima, Perú
| | - Graciela S Alarcón
- Division of Clinical Immunology and Rheumatology, Department of Medicine, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Medicine, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Perú
| |
Collapse
|
61
|
Iwata Y, Kitajima S, Yamahana J, Shimomura S, Yoneda-Nakagawa S, Sakai N, Furuichi K, Ogura H, Sato K, Toyama T, Yamamura Y, Miyagawa T, Hara A, Shimizu M, Ohkawa R, Kurano M, Yatomi Y, Wada T. Higher serum levels of autotaxin and phosphatidylserine-specific phospholipase A 1 in patients with lupus nephritis. Int J Rheum Dis 2020; 24:231-239. [PMID: 33314787 DOI: 10.1111/1756-185x.14031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/03/2020] [Accepted: 11/07/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Recent studies revealed that lysophospholipids (LPLs) and related molecules, such as autotaxin (ATX) and phosphatidylserine-specific phospholipase A1 (PS-PLA1 ), are candidates for novel biomarkers in melanoma, glaucoma and diabetic nephropathy. However, it is not clear whether serum levels of ATX/ PS-PLA1 would be associated with pathological and clinical findings of lupus nephritis (LN). METHODS In this retrospective cohort study, serum samples were collected from 39 patients with LN and 37 patients with other glomerular diseases. The serum levels of ATX and PS-PLA1 were evaluated for an association with renal pathology and clinical phenotypes of LN. RESULTS The serum levels of ATX and PS-PLA1 were higher in the patients with LN as compared to those with other glomerular diseases. Among the classes of LN, the patients with class IV showed the trend of lower serum levels of ATX. Moreover, the patients with lower levels of ATX exhibited higher scores of activity index (AI) and chronicity index (CI). The level of ATX tended to be negatively correlated with AI and CI. These results might be explained by the effect of treatment, because the serum levels of ATX and PS-PLA1 were inversely correlated with the daily amount of oral prednisolone. Moreover, they did not reflect the level of proteinuria or kidney survival in LN patients. CONCLUSION Although the serum levels of ATX and PS-PLA1 were affected by the treatment, these levels were higher in the patients with LN. The potential clinical benefits of these markers need to be clarified in further studies.
Collapse
Affiliation(s)
- Yasunori Iwata
- Division of Infection Control, Kanazawa University, Kanazawa, Japan.,Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Shinji Kitajima
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | | | - Shuji Shimomura
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | | | - Norihiko Sakai
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan.,Division of Blood Purification, Kanazawa University, Kanazawa, Japan
| | - Kengo Furuichi
- Division of Nephrology, Kanazawa Medical University School of Medicine, Ishikawa, Japan
| | - Hisayuki Ogura
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Koichi Sato
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Tadashi Toyama
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Yuta Yamamura
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Taro Miyagawa
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Akinori Hara
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Miho Shimizu
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Ryunosuke Ohkawa
- Department of Analytical Laboratory Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, University of Tokyo, Tokyo, Japan
| | - Takashi Wada
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
62
|
Luo Q, Li X, Fu B, Zhang L, Fang L, Qing C, Guo Y, Huang Z, Li J. Expression profile and diagnostic value of circRNAs in peripheral blood from patients with systemic lupus erythematosus. Mol Med Rep 2020; 23:1. [PMID: 33169172 PMCID: PMC7673322 DOI: 10.3892/mmr.2020.11639] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
Circular RNAs (circRNAs) have gained attention due to their performance in disease diagnosis. However, the characteristics of circRNAs in peripheral blood from patients with systemic lupus erythematosus (SLE) remain unknown. Therefore, the aim of the present study was to determine the expression profile and diagnostic potential of circRNAs in peripheral blood from patients with SLE. The global circRNA expression in the peripheral blood of patients with SLE and healthy controls (HCs) was detected using a circRNA microarray. Then, the expression levels of three upregulated circRNAs were selected for further validation by reverse transcription‑quantitative PCR (RT‑qPCR) in a training set. Moreover, the diagnostic value of these circRNAs was assessed by constructing a receiver operating characteristic curve, and then verified in a blind testing set. In total, 1,566 circRNAs were identified to be dysregulated between patients with SLE and HCs (≥2 fold change, P<0.05). Furthermore, the RT‑qPCR results were consistent with the microarray data, in that all three selected circRNAs, hsa_circ_0082688, hsa_circ_0082689 and hsa_circ_0008675, were significantly upregulated in patients with SLE (P<0.05). Results from the training set demonstrated that the combination of hsa_circ_0082688‑hsa_circ_0082689 may provide the most beneficial diagnostic potential. Moreover, the blind test results indicated that the combination model of hsa_circ_0082688‑hsa_circ_0082689 could effectively discriminate between patients with SLE from patients with rheumatoid arthritis and HCs, with a sensitivity of 91.30%, a specificity of 78.57% and an accuracy of 82.28%. Moreover, the combination model of hsa_circ_0082688‑hsa_circ_0082689 + anti‑dsDNA could more effectively discriminated the SLE group from the control groups, with a sensitivity of 95.65%, a specificity of 100.00% and an accuracy of 98.73%. In addition, correlation analysis results suggested that all three circRNAs in patients with SLE did not correlate with the SLE disease activity index. In conclusion, the expression levels of hsa_circ_0082688‑hsa_circ_0082689 may serve as potential biomarkers for SLE diagnosis.
Collapse
Affiliation(s)
- Qing Luo
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xue Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Biqi Fu
- Department of Rheumatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 710065, P.R. China
| | - Lu Zhang
- Department of Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Le Fang
- Department of Blood Transfusion, 521 Hospital of Ordnance Industry, Xi'an, Shanxi 710065, P.R. China
| | - Cheng Qing
- Department of Intensive Care Unit, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yang Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zikun Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Junming Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
63
|
Gamez-Nava JI, Diaz-Rizo V, Perez-Guerrero EE, Muñoz-Valle JF, Saldaña-Cruz AM, Fajardo-Robledo NS, Jacobo-Cuevas H, Nava-Valdivia CA, Alcaraz-Lopez MF, Trujillo X, Huerta M, Cardona-Muñoz EG, Gonzalez-Lopez L. Assessment of serum macrophage migration inhibitory factor (MIF), adiponectin, and other adipokines as potential markers of proteinuria and renal dysfunction in lupus nephritis: a cross-sectional study. Biomark Res 2020; 8:55. [PMID: 33133605 PMCID: PMC7594329 DOI: 10.1186/s40364-020-00236-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/15/2020] [Indexed: 12/15/2022] Open
Abstract
Background To date, the association of serum macrophage migration inhibitory factor (MIF) and serum adipokines with lupus nephritis is controversial. Objective To assess the utility of serum MIF, leptin, adiponectin and resistin levels as markers of proteinuria and renal dysfunction in lupus nephritis. Methods Cross-sectional study including 196 systemic lupus erythematosus (SLE) patients and 52 healthy controls (HCs). Disease activity was assessed by Systemic Lupus Erythematosus Disease Activity Index (SLEDAI). Renal SLE involvement was investigated by renal-SLEDAI. MIF, adiponectin, leptin and resistin levels were quantified by ELISA. We assessed the correlations of quantitative variables by Spearman correlation (rs). Multivariable linear regression adjusted the variables associated with the severity of proteinuria. Results SLE patients had higher MIF (p = 0.02) and adiponectin (p < 0.001) than HCs. Patients with renal SLE involvement (n = 43) had higher adiponectin (19.0 vs 13.3 μg/mL, p = 0.002) and resistin (10.7 vs 8.9 ng/mL, p = 0.01) than patients with non-renal SLE (n = 153). Proteinuria correlated with high adiponectin (rs = 0.19, p < 0.009) and resistin (rs = 0.26, p < 0.001). MIF (rs = 0.27, p = 0.04). Resistin correlated with increased creatinine (rs = 0.18, p = 0.02). High renal-SLEDAI correlated with adiponectin (rs = 0.21, p = 0.004). Multiple linear regression showed that elevated adiponectin (p = 0.02), younger age (p = 0.04) and low MIF (p = 0.02) were associated with the severity of proteinuria. Low MIF and high adiponectin levels interacted to explain the association with the severity of proteinuria (R2 = 0.41). Conclusions High adiponectin combined with low MIF concentrations int+eract to explain the severity of proteinuria in renal SLE. These findings highlight the relevance of adiponectin, resistin and MIF as markers of LN.
Collapse
Affiliation(s)
- Jorge Ivan Gamez-Nava
- Programa de Doctorado en Farmacología del Departamento de Fisiología y Programa de Doctorado en Salud del Pública Depatamento de Salud Pública, Universidad de Guadalajara, Centro Universitario de Ciencias de la Salud, Sierra Mojada 950, Colonia Independencia, 44340 Guadalajara, Jalisco Mexico.,Centro Medico Nacional de Occidente, Unidad de Investigacion Biomedica 02, Instituto Mexicano del Seguro Social, Hospital de Especialidades, 44340 Guadalajara, Jalisco Mexico
| | - Valeria Diaz-Rizo
- Departamento de Disciplinas Filosófico, Metodológico e Instrumentales, Universidad de Guadalajara, Centro Universitario de Ciencias de la Salud, 44340 Guadalajara, Jalisco Mexico
| | - Edsaul Emilio Perez-Guerrero
- Universidad de Guadalajara, Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas, 44340 Guadalajara, Jalisco Mexico
| | - Jose Francisco Muñoz-Valle
- Universidad de Guadalajara, Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas, 44340 Guadalajara, Jalisco Mexico
| | - Ana Miriam Saldaña-Cruz
- Departamento de Fisiología, Universidad de Guadalajara, Centro Universitario de Ciencias de la Salud, Sierra Mojada 950, Colonia Independencia, 44340 Guadalajara, Jalisco Mexico
| | - Nicte Selene Fajardo-Robledo
- Laboratorio de Investigación y Desarrollo Farmacéutico, Universidad de Guadalajara, Centro Universitario de Ciencias Exactas e Ingenierías, 44430 Guadalajara, Mexico
| | - Heriberto Jacobo-Cuevas
- Programa de Doctorado en Farmacología del Departamento de Fisiología y Programa de Doctorado en Salud del Pública Depatamento de Salud Pública, Universidad de Guadalajara, Centro Universitario de Ciencias de la Salud, Sierra Mojada 950, Colonia Independencia, 44340 Guadalajara, Jalisco Mexico
| | - Cesar Arturo Nava-Valdivia
- Departamento de Microbiología y Patología, Universidad de Guadalajara, Centro Universitario de Ciencias de la Salud, 44340 Guadalajara, Jalisco Mexico
| | | | - Xochitl Trujillo
- Universidad de Colima, Centro Universitario de Investigaciones Biomédicas, 28040 Colima, Mexico
| | - Miguel Huerta
- Universidad de Colima, Centro Universitario de Investigaciones Biomédicas, 28040 Colima, Mexico
| | - Ernesto German Cardona-Muñoz
- Departamento de Fisiología, Universidad de Guadalajara, Centro Universitario de Ciencias de la Salud, Sierra Mojada 950, Colonia Independencia, 44340 Guadalajara, Jalisco Mexico
| | - Laura Gonzalez-Lopez
- Programa de Doctorado en Farmacología del Departamento de Fisiología y Programa de Doctorado en Salud del Pública Depatamento de Salud Pública, Universidad de Guadalajara, Centro Universitario de Ciencias de la Salud, Sierra Mojada 950, Colonia Independencia, 44340 Guadalajara, Jalisco Mexico.,Departamento de Medicina InternaReumatología, Instituto Mexicano del Seguro Social (IMSS), Hospital General Regional 110, 44716 Guadalajara, Jalisco Mexico
| |
Collapse
|
64
|
Vanarsa K, Soomro S, Zhang T, Strachan B, Pedroza C, Nidhi M, Cicalese P, Gidley C, Dasari S, Mohan S, Thai N, Truong VTT, Jordan N, Saxena R, Putterman C, Petri M, Mohan C. Quantitative planar array screen of 1000 proteins uncovers novel urinary protein biomarkers of lupus nephritis. Ann Rheum Dis 2020; 79:1349-1361. [PMID: 32651195 PMCID: PMC7839323 DOI: 10.1136/annrheumdis-2019-216312] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The goal of these studies is to discover novel urinary biomarkers of lupus nephritis (LN). METHODS Urine from systemic lupus erythematosus (SLE) patients was interrogated for 1000 proteins using a novel, quantitative planar protein microarray. Hits were validated in an independent SLE cohort with inactive, active non-renal (ANR) and active renal (AR) patients, in a cohort with concurrent renal biopsies, and in a longitudinal cohort. Single-cell renal RNA sequencing data from LN kidneys were examined to deduce the cellular origin of each biomarker. RESULTS Screening of 1000 proteins revealed 64 proteins to be significantly elevated in SLE urine, of which 17 were ELISA validated in independent cohorts. Urine Angptl4 (area under the curve (AUC)=0.96), L-selectin (AUC=0.86), TPP1 (AUC=0.84), transforming growth factor-β1 (TGFβ1) (AUC=0.78), thrombospondin-1 (AUC=0.73), FOLR2 (AUC=0.72), platelet-derived growth factor receptor-β (AUC=0.67) and PRX2 (AUC=0.65) distinguished AR from ANR SLE, outperforming anti-dsDNA, C3 and C4, in terms of specificity, sensitivity and positive predictive value. In multivariate regression analysis, urine Angptl4, L-selectin, TPP1 and TGFβ1 were highly associated with disease activity, even after correction for demographic variables. In SLE patients with serial follow-up, urine L-selectin (followed by urine Angptl4 and TGFβ1) were best at tracking concurrent or pending disease flares. Importantly, several proteins elevated in LN urine were also expressed within the kidneys in LN, either within resident renal cells or infiltrating immune cells, based on single-cell RNA sequencing analysis. CONCLUSION Unbiased planar array screening of 1000 proteins has led to the discovery of urine Angptl4, L-selectin and TGFβ1 as potential biomarker candidates for tracking disease activity in LN.
Collapse
Affiliation(s)
- Kamala Vanarsa
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Sanam Soomro
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Ting Zhang
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Briony Strachan
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Claudia Pedroza
- Center for Clinical Research and Evidence-based Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Malavika Nidhi
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Pietro Cicalese
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Christopher Gidley
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Shobha Dasari
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Shree Mohan
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Nathan Thai
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Van Thi Thanh Truong
- Center for Clinical Research and Evidence-based Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nicole Jordan
- Division of Rheumatology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ramesh Saxena
- Division of Nephrology, Department of Medicine, UT Southwestern Medical, Dallas, Texas, USA
| | - Chaim Putterman
- Division of Rheumatology, Albert Einstein College of Medicine, Bronx, New York, USA
- Azrieli Faculty of Medicine, Bar-Ilan University, Zefat, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| |
Collapse
|
65
|
Brady MP, Korte EA, Caster DJ, Powell DW. TNIP1/ABIN1 and lupus nephritis: review. Lupus Sci Med 2020; 7:e000437. [PMID: 33122334 PMCID: PMC7597513 DOI: 10.1136/lupus-2020-000437] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/15/2020] [Accepted: 10/08/2020] [Indexed: 12/17/2022]
Abstract
SLE is a complex autoimmune disease with genetic, epigenetic, immune-regulatory, environmental and hormonal factors. Kidney inflammation and injury, termed lupus nephritis (LN), occurs in over half of patients with SLE and is a leading cause of disability and death. There is a high degree of short-term and long-term side effects associated with current LN therapies and they are not effective for many patients. Thus, novel therapies with reduced toxicity and improved efficacy are drastically needed. Many of the known LN susceptibility genes have functions that mediate inflammation via cytokine/chemokine production and activation of myeloid and B cells. Understanding the cellular and molecular mechanisms mediated by these variant gene products provides valuable insight for the development of improved and personalised diagnostics and therapeutics. This review describes variants in the TNIP1 (tumour necrosis factor α-induced protein 3-interacting protein 1) gene associated with risks for SLE and LN and potential roles for loss of function of its protein product ABIN1 in the activation of myeloid and B-cell-mediated injury in LN.
Collapse
Affiliation(s)
- Makayla P Brady
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Erik A Korte
- Bluewater Diagnostics Laboratory, Mt. Washington, Kentucky, USA
| | - Dawn J Caster
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - David W Powell
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
66
|
Zhang L, Zhang K, Dong W, Li R, Huang R, Zhang H, Shi W, Liu S, Li Z, Chen Y, Ye Z, Liang X, Yu X. Raised Plasma Levels of Asymmetric Dimethylarginine Are Associated with Pathological Type and Predict the Therapeutic Effect in Lupus Nephritis Patients Treated with Cyclophosphamide. KIDNEY DISEASES (BASEL, SWITZERLAND) 2020; 6:355-363. [PMID: 33490115 PMCID: PMC7745665 DOI: 10.1159/000509767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 01/30/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Lupus nephritis (LN) is one of the most serious complications of systemic lupus erythematosus (SLE). Asymmetric dimethylarginine (ADMA) has been associated with cardiovascular events in SLE patients and is a strong predictor of the progression of chronic kidney disease. However, whether ADMA can provide a predictive value for the diagnosis and treatment of LN patients remains unclear. This study evaluated the clinical significance of ADMA in LN patients. METHODS Blood samples of 114 patients with LN, 52 patients with primary glomerular disease, and 20 healthy people were collected. Plasma ADMA was measured via enzyme-linked immunosorbent assay. The relationship between plasma ADMA levels and pathological types and renal function and efficacy in LN patients were further analyzed. RESULTS There was no significant difference in plasma ADMA levels between LN and primary glomerular disease, but both were significantly higher than the values in healthy people (p < 0.05). Plasma ADMA levels in LN patients were negatively correlated with baseline estimated glomerular filtration rate (eGFR) and serum superoxide dismutase and positively correlated with serum cystatin C and serum β2-microglobulin (p < 0.05). The plasma ADMA levels of diffuse proliferative LN patients were significantly higher than those of other histopathological classes of LN. High plasma ADMA levels in LN patients (OR = 1.012; 95% CI 1.003-1.022; p = 0.010) is a risk factor for diffuse proliferative LN. The area under the receiver operating characteristic (ROC) curve of diagnosing diffuse proliferative LN by plasma ADMA was 0.707 (95% CI 0.610-0.805). The area under the ROC curve of combination with plasma ADMA, serum complement C3, and eGFR for diffuse proliferative LN was 0.796 (95% CI 0.713-0.879), which was significantly higher than that of ADMA, complement C3, and eGFR for diffuse proliferative LN alone, respectively (p < 0.05). Low plasma ADMA is an independent protective factor for proliferative LN patients achieving complete remission with cyclophosphamide as induction therapy (OR = 0.978; 95% CI 0.961-0.996; p < 0.05). CONCLUSION High plasma ADMA levels in combination with eGFR and complement C3 may be useful to diagnose diffuse proliferative LN. Low plasma ADMA may help to predict complete remission in proliferative LN patients treated with cyclophosphamide as induction therapy. Plasma ADMA may be a new biomarker to determine the pathological type of LN and predict the therapeutic effect.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xinling Liang
- Department of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | | |
Collapse
|
67
|
Cao Y, Mi X, Wang Z, Zhang D, Tang W. Bioinformatic analysis reveals that the OAS family may play an important role in lupus nephritis. J Natl Med Assoc 2020; 112:567-577. [PMID: 32622555 DOI: 10.1016/j.jnma.2020.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 05/19/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Lupus nephritis (LN) is a common complication of systemic lupus erythematosus that presents a high risk of end-stage renal disease. However, the molecular mechanisms of LN remain unclear. The lack of understanding hinders the development of specific targeted therapy for this progressive disease. OBJECTIVES In the present study, we used bioinformatics analysis of gene expression profiles from the Gene Expression Omnibus to identify novel targets and potential biomarkers for LN. MATERIAL AND METHODS A GSE32591 dataset, which included 31 LN glomerular biopsy tissues and 14 living donors' glomerular tissues, was downloaded for further analysis. Differentially expressed genes in LN were analyzed by the limma package. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed for the differentially expressed genes by using the Disease Ontology Semantic and Enrichment and the clusterProfiler software. The protein-protein interaction (PPI) network was then formed using STRING online tool. RESULTS 440 genes, including 310 upregulated genes and 130 downregulated genes, were found as differentially expressed genes. GO and KEGG analyses revealed that immune response is significantly enriched in such genes. The PPI network showed that ISG15, MX1, OAS1, OAS2, and OAS3 were the hub genes enriched in LN. Along with literature review, the OAS family genes were revealed to be closely associated with LN progression. CONCLUSIONS our studies provided new insight into the molecular pathogenesis of LN. The OAS family may play an important role in LN and act as a novel molecular candidate for the further study of LN.
Collapse
Affiliation(s)
- Yiling Cao
- Department of Nephrology, West China Hospital, Sichuan University, No.37, Guoxue Alley, Chengdu, Sichuan, China
| | - Xuhua Mi
- Department of Nephrology, West China Hospital, Sichuan University, No.37, Guoxue Alley, Chengdu, Sichuan, China
| | - Zheng Wang
- Department of Nephrology, West China Hospital, Sichuan University, No.37, Guoxue Alley, Chengdu, Sichuan, China
| | - Dongmei Zhang
- Department of Nephrology, West China Hospital, Sichuan University, No.37, Guoxue Alley, Chengdu, Sichuan, China
| | - Wanxin Tang
- Department of Nephrology, West China Hospital, Sichuan University, No.37, Guoxue Alley, Chengdu, Sichuan, China.
| |
Collapse
|
68
|
Elevated Urinary Neutrophil Gelatinase-Associated Lipocalin Is a Biomarker for Lupus Nephritis: A Systematic Review and Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2768326. [PMID: 32685458 PMCID: PMC7346103 DOI: 10.1155/2020/2768326] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/30/2020] [Indexed: 01/20/2023]
Abstract
Objective Lupus nephritis (LN) is a major and severe complication of systemic lupus erythematosus (SLE). Neutrophil gelatinase-associated lipocalin (NGAL), as a promising next-generation biomarker in clinical nephrology, has received extensive attention. However, its diagnostic performance in LN has high variability. Therefore, we performed an updated meta-analysis to further evaluate the diagnostic accuracy of urinary NGAL (uNGAL). Materials and Methods PubMed, Embase, and Cochrane Library were searched from inception to October 27, 2019. Meta-analysis was performed with a bivariate random effects model. Additionally, the summary receiver operating characteristic (SROC) curves were established. The sources of heterogeneity were explored by meta-regression, subgroup analysis, and sensitivity analysis. Publication bias was assessed using the Deeks test. Results 19 articles consisting of 21 eligible studies were included. In diagnosing LN, the estimates (95% confidence interval (CI)) were as follows: sensitivity, 0.84 (0.71-0.91); specificity, 0.91 (0.70-0.98); and the SROC-AUC value, 0.92 (0.90-0.94). In identifying active LN, the estimates were as follows: sensitivity, 0.72 (0.56-0.84); specificity, 0.71 (0.51-0.84); and the AUC value, 0.77 (0.74-0.81). With respect to predicting renal flare, the estimates were as follows: sensitivity, 0.80 (0.57-0.92); specificity, 0.67 (0.58-0.75); and the AUC value, 0.74 (0.70-0.78). For the studies to distinguish proliferative LN, the estimates were as follows: sensitivity, 0.87 (0.66-0.97), and specificity, 0.69 (0.39-0.91). Deeks' funnel plot suggested that there was no significant publication bias. Conclusions Our meta-analysis indicates that uNGAL was a useful biomarker for diagnosis, estimation of activity, and prediction of renal flare of LN. In addition, the usefulness of uNGAL to distinguish pathological types of LN needs to be further investigated.
Collapse
|
69
|
Fava A, Buyon J, Mohan C, Zhang T, Belmont HM, Izmirly P, Clancy R, Trujillo JM, Fine D, Zhang Y, Magder L, Rao DA, Arazi A, Berthier CC, Davidson A, Diamond B, Hacohen N, Wofsy D, Apruzzese W, Raychaudhuri S, Petri M. Integrated urine proteomics and renal single-cell genomics identify an IFN-γ response gradient in lupus nephritis. JCI Insight 2020; 5:138345. [PMID: 32396533 PMCID: PMC7406291 DOI: 10.1172/jci.insight.138345] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
Lupus nephritis, one of the most serious manifestations of systemic lupus erythematosus (SLE), has a heterogeneous clinical and pathological presentation. For example, proliferative nephritis identifies a more aggressive disease class that requires immunosuppression. However, the current classification system relies on the static appearance of histopathological morphology, which does not capture differences in the inflammatory response. Therefore, a biomarker grounded in the disease biology is needed in order to understand the molecular heterogeneity of lupus nephritis and identify immunologic mechanism and pathways. Here, we analyzed the patterns of 1000 urine protein biomarkers in 30 patients with active lupus nephritis. We found that patients stratify over a chemokine gradient inducible by IFN-γ. Higher values identified patients with proliferative lupus nephritis. After integrating the urine proteomics with the single-cell transcriptomics of kidney biopsies, we observed that the urinary chemokines defining the gradient were predominantly produced by infiltrating CD8+ T cells, along with natural killer and myeloid cells. The urine chemokine gradient significantly correlated with the number of kidney-infiltrating CD8+ cells. These findings suggest that urine proteomics can capture the complex biology of the kidney in lupus nephritis. Patient-specific pathways could be noninvasively tracked in the urine in real time, enabling diagnosis and personalized treatment.
Collapse
Affiliation(s)
- Andrea Fava
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jill Buyon
- New York University School of Medicine, New York, New York, USA
| | | | - Ting Zhang
- University of Houston, Houston, Texas, USA
| | | | - Peter Izmirly
- New York University School of Medicine, New York, New York, USA
| | - Robert Clancy
- New York University School of Medicine, New York, New York, USA
| | | | - Derek Fine
- Division of Nephrology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yuji Zhang
- Department of Epidemiology and Public Health, University of Maryland, Baltimore, Maryland, USA
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Laurence Magder
- Department of Epidemiology and Public Health, University of Maryland, Baltimore, Maryland, USA
| | - Deepak A. Rao
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Arnon Arazi
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Celine C. Berthier
- Internal Medicine, Department of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - Anne Davidson
- Center for Autoimmune and Musculoskeletal Diseases, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Betty Diamond
- Center for Autoimmune and Musculoskeletal Diseases, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - David Wofsy
- Division of Rheumatology, UCSF, San Francisco, California, USA
| | - William Apruzzese
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Soumya Raychaudhuri
- Center for Data Sciences and
- Division of Rheumatology and Genetics, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
70
|
Zhang T, Li H, Vanarsa K, Gidley G, Mok CC, Petri M, Saxena R, Mohan C. Association of Urine sCD163 With Proliferative Lupus Nephritis, Fibrinoid Necrosis, Cellular Crescents and Intrarenal M2 Macrophages. Front Immunol 2020; 11:671. [PMID: 32351512 PMCID: PMC7174755 DOI: 10.3389/fimmu.2020.00671] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/25/2020] [Indexed: 12/15/2022] Open
Abstract
CD163 is a marker for alternatively activated macrophages, which have been implicated in the pathogenesis of lupus nephritis (LN). In our preliminary screening of urine proteins in LN, urine soluble CD163 (sCD163) was significantly elevated in patients with active LN. To evaluate the potential of sCD163 as a biomarker in LN, urine sCD163 was assayed in patients with active LN, active non-renal lupus patients (ANR), inactive SLE and healthy controls (HC), using ELISA and normalized to urine creatinine. The correlation of urine sCD163 with clinical parameters and renal pathological attributes was further investigated in LN patients with concurrent renal biopsies. A total of 228 SLE patients and 56 HC were included from three cohorts. Results demonstrated that urine sCD163 was significantly elevated in active LN when compared with HC, inactive SLE, or ANR in African-American, Caucasian and Asian subjects (all P < 0.001). In LN patients with concurrent renal biopsies, urine sCD163 was significantly increased in patients with proliferative LN when compared with non-proliferative LN (P < 0.001). Urine sCD163 strongly correlated with SLEDAI, rSLEDAI, activity index (AI) of renal pathology, fibrinoid necrosis, cellular crescents, and interstitial inflammation on biopsies (all P < 0.01). Macrophages, particularly M2 macrophages, the predominant cells expressing CD163 within LN kidneys, represented a potential source of elevated urine sCD163, based on single-cell RNA sequencing analysis. To conclude, urine sCD163 discriminated patients with active LN from other SLE patients and was significantly elevated in proliferative LN. It strongly correlated with concurrent AI and several specific pathological attributes, demonstrating its potential in predicting renal pathology.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Hao Li
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Kamala Vanarsa
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Gabriel Gidley
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, Hong Kong, China
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ramesh Saxena
- University Hospital Kidney & Liver Clinic, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| |
Collapse
|
71
|
You X, Zhang R, Shao M, He J, Chen J, Liu J, Zhang X, Liu X, Jia R, Sun X, Li Z. Double Negative B Cell Is Associated With Renal Impairment in Systemic Lupus Erythematosus and Acts as a Marker for Nephritis Remission. Front Med (Lausanne) 2020; 7:85. [PMID: 32318574 PMCID: PMC7155774 DOI: 10.3389/fmed.2020.00085] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/02/2020] [Indexed: 12/23/2022] Open
Abstract
Objective: Recent studies on double negative B cells (DN B cells) suggested that they have potential pathogenic roles in systemic lupus erythematosus (SLE). This study aimed to determine the circulating DN B cells in SLE patients and analyzed the clinical significance of this cell subset. Methods: Fifty-seven SLE patients and fifty healthy controls (HCs) were recruited in this study. Among the 57 SLE patients, 25 had lupus nephritis (LN). All patients were followed up for 24 weeks. Peripheral B cell subsets were analyzed by flow cytometry. Results: DN B cells were significantly elevated in the SLE patients, especially in the patients with LN (p < 0.01). DN B showed a positive correlation with 24-h urine protein excretion (24 h-UPE) levels (r = 0.444, p = 0.034) in LN patients, and inversely correlated with evaluated glomerular filtration rate (eGFR) (r = -0.351, p = 0.011). DN B cells had a positive correlation with plasma cells (r = 0.484, p < 0.001) and memory B cells (r = 0.703, p < 0.001). After treatment, decreased DN B cells were associated with LN alleviation (p = 0.002). In the follow-up, the remission rate of LN patients with decreased DN B cells was significantly higher than LN patients with increased DN B cells (83.33 vs. 25.00%, p = 0.030) at week 24. Conclusions: This study suggests that the peripheral DN B cells are positively correlated with the severity of renal damage in LN patients and may potentially be used as a prognostic marker in LN.
Collapse
Affiliation(s)
- Xujie You
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Peking University People's Hospital, Beijing, China
| | - Ruijun Zhang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Peking University People's Hospital, Beijing, China
| | - Miao Shao
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Peking University People's Hospital, Beijing, China
| | - Jing He
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Peking University People's Hospital, Beijing, China
| | - Jiali Chen
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Peking University People's Hospital, Beijing, China
| | - Jiajia Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Peking University People's Hospital, Beijing, China
| | - Xia Zhang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Peking University People's Hospital, Beijing, China
| | - Xu Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Peking University People's Hospital, Beijing, China
| | - Rulin Jia
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Peking University People's Hospital, Beijing, China
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Peking University People's Hospital, Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China.,Beijing Key Laboratory for Rheumatism and Immune Diagnosis (BZ0135), Peking University People's Hospital, Beijing, China
| |
Collapse
|
72
|
Luo Q, Zhang L, Fang L, Fu B, Guo Y, Huang Z, Li J. Circular RNAs hsa_circ_0000479 in peripheral blood mononuclear cells as novel biomarkers for systemic lupus erythematosus. Autoimmunity 2020; 53:167-176. [PMID: 32093518 DOI: 10.1080/08916934.2020.1728529] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Circular RNAs (circRNAs) are a class of non-coding RNAs that play a crucial role in diagnosis and prognosis of systemic lupus erythematosus (SLE). However, circRNAs expression profiling in SLE from different reports are different. In this study, 11 circRNAs (hsa_circ_0000479, hsa_circ_0002316, hsa_circ_0000317, hsa_circ_0082688, hsa_circ_0082689, hsa_circ_0087798, hsa_circ_0008529, hsa_circ_0000787, hsa_circ_0021727, hsa_circ_0000175, and hsa_circ_0003694) which were found to be significantly up-regulated in both peripheral blood mononuclear cells (PBMCs) from SLE patients in our previous study, and T cells from SLE patients in previous literature, were chosen for validation by quantitative reverse transcription-polymerase chain reaction in PBMCs from 50 new-onset SLE patients, 24 new-onset rheumatoid arthritis (RA) patients, 24 new-onset ankylosing spondylitis (AS) patients, and 45 age- and sex-matched healthy controls (HC). The results validated that PBMCs hsa_circ_0000479, hsa_circ_0082688, and hsa_circ_0082689 were increased, while hsa_circ_0000175 was significantly decreased in SLE patients than that in RA patients, AS patients, and HC. The correlation analysis of these confirmed differentially expressed circRNAs showed that hsa_circ_0000479 was associated with C3 level and treatment, hsa_circ_0082688 was associated with anti-dsDNA level, hsa_circ_0082689 was associated with anti-dsDNA level, anti-nuclesome frequency and treatment. Receiver operating characteristic curve anaylsis suggested that hsa_circ_0000479 has significant value in distinguishing SLE from AS patients, RA patients, and HC (AUC = 0.825, p < .001). Moreover, the hsa_circ_0000479-anti-dsDNA combination model could effectively discriminate the SLE group and the control groups (RA + AS + HC), with a sensitivity of 86.00% (43/50), a specificity of 100.00% (93/93), and an accuracy of 95.10% (136/143). This study suggested that hsa_circ_0000479 in PBMC and hsa_circ_0000479-anti-dsDNA combination model may serve as potential biomarkers for SLE diagnosis and evaluation of therapeutic effect.
Collapse
Affiliation(s)
- Qing Luo
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Lu Zhang
- Department of medical college, Nanchang University, Nanchang, Jiangxi, China
| | - Le Fang
- Department of Blood Transfusion, 521 Hospital of Ordnance Industry, Xi'an, Shanxi, China
| | - Biqi Fu
- Department of rheumatology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yang Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zikun Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Junming Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
73
|
Hoover P, Der E, Berthier CC, Arazi A, Lederer JA, James JA, Buyon J, Petri M, Belmont HM, Izmirly P, Wofsy D, Hacohen N, Diamond B, Putterman C, Davidson A. Accelerating Medicines Partnership: Organizational Structure and Preliminary Data From the Phase 1 Studies of Lupus Nephritis. Arthritis Care Res (Hoboken) 2020; 72:233-242. [PMID: 31502417 PMCID: PMC6992476 DOI: 10.1002/acr.24066] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022]
Abstract
The Accelerating Medicines Partnership (AMP) Lupus Network was established as a partnership between the National Institutes of Health, pharmaceutical companies, nonprofit stakeholders, and lupus investigators across multiple academic centers to apply high-throughput technologies to the analysis of renal tissue, urine, and blood from patients with lupus nephritis (LN). The AMP network provides publicly accessible data to the community with the goal of generating new scientific hypotheses and improving diagnostic and therapeutic tools so as to improve disease outcomes. We present here a description of the structure of the AMP Lupus Network and a summary of the preliminary results from the phase 1 studies. The successful completion of phase 1 sets the stage for analysis of a large cohort of LN samples in phase 2 and provides a model for establishing similar discovery cohorts.
Collapse
Affiliation(s)
- Paul Hoover
- Broad Institute of MIT and Harvard, Cambridge Massachusetts, USA
| | - Evan Der
- Division of Rheumatology and Department of Microbiology and Immunology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, USA
| | - Celine C. Berthier
- Internal Medicine, Department of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - Arnon Arazi
- Broad Institute of MIT and Harvard, Cambridge Massachusetts, USA
| | - James A. Lederer
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston Massachusetts, USA
| | - Judith A. James
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Jill Buyon
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, New-York, USA
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - H. Michael Belmont
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, New-York, USA
| | - Peter Izmirly
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, New-York, USA
| | - David Wofsy
- Rheumatology Division and Russell/Engleman Rheumatology Research Center, University of California San Francisco, San Francisco, California, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge Massachusetts, USA
| | - Betty Diamond
- Center for Autoimmune, Musculoskeletal and Hematologic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Chaim Putterman
- Division of Rheumatology and Department of Microbiology and Immunology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, USA
| | - Anne Davidson
- Center for Autoimmune, Musculoskeletal and Hematologic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York, USA
| |
Collapse
|
74
|
Abstract
Lupus nephritis (LN) is a form of glomerulonephritis that constitutes one of the most severe organ manifestations of the autoimmune disease systemic lupus erythematosus (SLE). Most patients with SLE who develop LN do so within 5 years of an SLE diagnosis and, in many cases, LN is the presenting manifestation resulting in the diagnosis of SLE. Understanding of the genetic and pathogenetic basis of LN has improved substantially over the past few decades. Treatment of LN usually involves immunosuppressive therapy, typically with mycophenolate mofetil or cyclophosphamide and with glucocorticoids, although these treatments are not uniformly effective. Despite increased knowledge of disease pathogenesis and improved treatment options, LN remains a substantial cause of morbidity and death among patients with SLE. Within 10 years of an initial SLE diagnosis, 5-20% of patients with LN develop end-stage kidney disease, and the multiple comorbidities associated with immunosuppressive treatment, including infections, osteoporosis and cardiovascular and reproductive effects, remain a concern. Clearly, early and accurate diagnosis of LN and prompt initiation of therapy are of vital importance to improve outcomes in patients with SLE.
Collapse
|
75
|
Rovin BH, Caster DJ, Cattran DC, Gibson KL, Hogan JJ, Moeller MJ, Roccatello D, Cheung M, Wheeler DC, Winkelmayer WC, Floege J, Alpers CE, Ayoub I, Bagga A, Barbour SJ, Barratt J, Chan DT, Chang A, Choo JCJ, Cook HT, Coppo R, Fervenza FC, Fogo AB, Fox JG, Glassock RJ, Harris D, Hodson EM, Hogan JJ, Hoxha E, Iseki K, Jennette JC, Jha V, Johnson DW, Kaname S, Katafuchi R, Kitching AR, Lafayette RA, Li PK, Liew A, Lv J, Malvar A, Maruyama S, Mejía-Vilet JM, Mok CC, Nachman PH, Nester CM, Noiri E, O'Shaughnessy MM, Özen S, Parikh SM, Park HC, Peh CA, Pendergraft WF, Pickering MC, Pillebout E, Radhakrishnan J, Rathi M, Ronco P, Smoyer WE, Tang SC, Tesař V, Thurman JM, Trimarchi H, Vivarelli M, Walters GD, Wang AYM, Wenderfer SE, Wetzels JF. Management and treatment of glomerular diseases (part 2): conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int 2020; 95:281-295. [PMID: 30665569 DOI: 10.1016/j.kint.2018.11.008] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/30/2018] [Accepted: 11/01/2018] [Indexed: 02/06/2023]
Abstract
In November 2017, the Kidney Disease: Improving Global Outcomes (KDIGO) initiative brought a diverse panel of experts in glomerular diseases together to discuss the 2012 KDIGO glomerulonephritis guideline in the context of new developments and insights that had occurred over the years since its publication. During this KDIGO Controversies Conference on Glomerular Diseases, the group examined data on disease pathogenesis, biomarkers, and treatments to identify areas of consensus and areas of controversy. This report summarizes the discussions on primary podocytopathies, lupus nephritis, anti-neutrophil cytoplasmic antibody-associated nephritis, complement-mediated kidney diseases, and monoclonal gammopathies of renal significance.
Collapse
Affiliation(s)
- Brad H Rovin
- Division of Nephrology, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA.
| | - Dawn J Caster
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Daniel C Cattran
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Keisha L Gibson
- University of North Carolina Kidney Center at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jonathan J Hogan
- Division of Nephrology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marcus J Moeller
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule, University of Aachen, Aachen, Germany
| | - Dario Roccatello
- CMID (Center of Research of Immunopathology and Rare Diseases), and Division of Nephrology and Dialysis (ERK-Net member), University of Turin, Italy
| | | | | | - Wolfgang C Winkelmayer
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Jürgen Floege
- Division of Nephrology, Rheinisch-Westfälische Technische Hochschule, University of Aachen, Aachen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Rao DA, Arazi A, Wofsy D, Diamond B. Design and application of single-cell RNA sequencing to study kidney immune cells in lupus nephritis. Nat Rev Nephrol 2019; 16:238-250. [PMID: 31853010 DOI: 10.1038/s41581-019-0232-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2019] [Indexed: 11/09/2022]
Abstract
The immune mechanisms that cause tissue injury in lupus nephritis have been challenging to define. The advent of high-dimensional cellular analyses, such as single-cell RNA sequencing, has enabled detailed characterization of the cell populations present in small biopsy samples of kidney tissue. In parallel, the development of methods that cryopreserve kidney biopsy specimens in a manner that preserves intact, viable cells, has enabled the uniform analysis of tissue samples collected at multiple sites and across many geographic areas and demographic cohorts with high-dimensional platforms. The application of these methods to kidney biopsy samples from patients with lupus nephritis has begun to define the phenotypes of both infiltrating and resident immune cells, as well as parenchymal cells, present in nephritic kidneys. The detection of similar immune cell populations in urine suggests that it might be possible to non-invasively monitor immune activation in kidneys. Once applied to large patient cohorts, these high-dimensional studies might enable patient stratification according to patterns of immune cell activation in the kidney or identify disease features that can be used as surrogate measures of efficacy in clinical trials. Applied broadly across multiple inflammatory kidney diseases, these studies promise to enormously expand our understanding of renal inflammation in the next decade.
Collapse
Affiliation(s)
- Deepak A Rao
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Arnon Arazi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David Wofsy
- Rheumatology Division and Russell/Engleman Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Betty Diamond
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA.
| |
Collapse
|
77
|
Shi M, Luo W, Feng X, Jin L, Yang M, Wu L, Yang Z, Su C, Li Y, Su H, Wang G, Cao W. Urinary Angiotensinogen Predicts Renal Disease Activity in Lupus Nephritis. Antioxid Redox Signal 2019; 31:1289-1301. [PMID: 31264479 DOI: 10.1089/ars.2019.7782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Aims: A noninvasive indicator of renal histological lesions and disease activity in lupus nephritis (LN) is needed for timely and targeted treatment before overt renal injury. Here, we tested the utility of urinary angiotensinogen (UAGT) to predict renal disease activity in LN. Results: A prospective, three-stage study was performed in patients with LN. In stage I, UAGT was measured in 140 newly diagnosed LN patients. UAGT significantly increased in LN patients, correlating well with kidney angiotensinogen expression and histological activity. Patients with LN class IV exhibited the highest UAGT compared with other histopathological classes of LN. For identifying LN class IV, a particularly aggressive type of LN, UAGT outperformed the conventional clinical measures and improved their performance. In stage II, UAGT was monitored in 61 subjects from stage I for up to 12 months. UAGT decreased after induction therapy and remained low in patients with LN remission during follow-up. For predicting therapy success at month 12, the area under the receiver operating characteristics curve of UAGT reduction at month 4 was 0.83, outperforming that of 24-h proteinuria. In stage III, UAGT was monitored in 12 LN patients before, during, and after the onset of renal flares. An elevation in UAGT predicted recurrence of LN, and a decline in UAGT after a renal flare heralded the remission of disease before conventional clinical measures. Innovation and Conclusion: UAGT in LN is a promising indicator for dynamic surveillance of renal disease activity and prediction of renal flares. Antioxid. Redox Signal. 31, 1289-1301.
Collapse
Affiliation(s)
- Meng Shi
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangzhou, People's Republic of China
| | - Weihong Luo
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangzhou, People's Republic of China
| | - Xiaodan Feng
- Division of Nephrology, Guangzhou Development District Hospital, Guangzhou, People's Republic of China
| | - Lingwei Jin
- Division of Nephrology, The 2nd Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Manqiu Yang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangzhou, People's Republic of China
| | - Liling Wu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangzhou, People's Republic of China
| | - Zhichen Yang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangzhou, People's Republic of China
| | - Cailing Su
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangzhou, People's Republic of China
| | - Yajing Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangzhou, People's Republic of China
| | - Huanjuan Su
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangzhou, People's Republic of China
| | - Guobao Wang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangzhou, People's Republic of China
| | - Wei Cao
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangzhou, People's Republic of China
| |
Collapse
|
78
|
Abstract
Is systemic lupus erythematosus (SLE) is occurring more frequently now than in decades past? Despite improvements in the identification of patients with SLE, the development of new classification criteria, and the recognition of several biomarkers used alone or in combination, the diagnosis of SLE is still a challenge for clinicians, in particular early in the course of the disease, which makes the recognition of secular trends difficult to ascertain. Lacking a uniform definition of preclinical lupus or incomplete lupus, it is difficult to predict accurately which patients would go on to develop SLE. We will briefly review the classification criteria, early or preclinical SLE, the epidemiology of SLE, antinuclear antibodies-negative SLE, and biomarkers of the disease.
Collapse
Affiliation(s)
- M F Ugarte-Gil
- Rheumatology Department, Hospital Guillermo Almenara Irigoyen, EsSalud, Lima, Perú
- School of Medicine, Universidad Científica del Sur, Lima, Perú
| | - L A González
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - G S Alarcón
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, USA
- School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Perú
| |
Collapse
|
79
|
Becker Y, Marcoux G, Allaeys I, Julien AS, Loignon RC, Benk-Fortin H, Rollet-Labelle E, Rauch J, Fortin PR, Boilard E. Autoantibodies in Systemic Lupus Erythematosus Target Mitochondrial RNA. Front Immunol 2019; 10:1026. [PMID: 31134086 PMCID: PMC6524553 DOI: 10.3389/fimmu.2019.01026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/23/2019] [Indexed: 12/22/2022] Open
Abstract
The mitochondrion supplies energy to the cell and regulates apoptosis. Unlike other mammalian organelles, mitochondria are formed by binary fission and cannot be directly produced by the cell. They contain numerous copies of a compact circular genome that encodes RNA molecules and proteins involved in mitochondrial oxidative phosphorylation. Whereas, mitochondrial DNA (mtDNA) activates the innate immune system if present in the cytosol or the extracellular milieu, it is also the target of circulating autoantibodies in systemic lupus erythematosus (SLE). However, it is not known whether mitochondrial RNA is also recognized by autoantibodies in SLE. In the present study, we evaluated the presence of autoantibodies targeting mitochondrial RNA (AmtRNA) in SLE. We quantified AmtRNA in an inducible model of murine SLE. The AmtRNA were also determined in SLE patients and healthy volunteers. AmtRNA titers were measured in both our induced model of murine SLE and in human SLE, and biostatistical analyses were performed to determine whether the presence and/or levels of AmtRNA were associated with clinical features expressed by SLE patients. Both IgG and IgM classes of AmtRNA were increased in SLE patients (n = 86) compared to healthy controls (n = 30) (p < 0.0001 and p = 0.0493, respectively). AmtRNA IgG levels correlated with anti-mtDNA-IgG titers (rs = 0.54, p < 0.0001) as well as with both IgG and IgM against β-2-glycoprotein I (anti-β2GPI; rs = 0.22, p = 0.05), and AmtRNA-IgG antibodies were present at higher levels when patients were positive for autoantibodies to double-stranded-genomic DNA (p < 0.0001). AmtRNA-IgG were able to specifically discriminate SLE patients from healthy controls, and were negatively associated with plaque formation (p = 0.04) and lupus nephritis (p = 0.03). Conversely, AmtRNA-IgM titers correlated with those of anti-β2GPI-IgM (rs = 0.48, p < 0.0001). AmtRNA-IgM were higher when patients were positive for anticardiolipin antibodies (aCL-IgG: p = 0.01; aCL-IgM: p = 0.002), but AmtRNA-IgM were not associated with any of the clinical manifestations assessed. These findings identify mtRNA as a novel mitochondrial antigen target in SLE, and support the concept that mitochondria may provide an important source of circulating autoantigens in SLE.
Collapse
Affiliation(s)
- Yann Becker
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Geneviève Marcoux
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Isabelle Allaeys
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Anne-Sophie Julien
- Département de mathématiques et statistiques, Université Laval, Québec City, QC, Canada
| | - Renée-Claude Loignon
- Division de Rhumatologie, Département de Médecine, CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Hadrien Benk-Fortin
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Emmanuelle Rollet-Labelle
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Joyce Rauch
- Division of Rheumatology, Department of Medicine, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Paul R Fortin
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.,Division de Rhumatologie, Département de Médecine, CHU de Québec-Université Laval, Québec City, QC, Canada.,Axe maladies infectieuses et inflammatoires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Eric Boilard
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.,Axe maladies infectieuses et inflammatoires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| |
Collapse
|
80
|
Pinheiro SVB, Dias RF, Fabiano RCG, Araujo SDA, Silva ACSE. Pediatric lupus nephritis. J Bras Nefrol 2019; 41:252-265. [PMID: 30465590 PMCID: PMC6699445 DOI: 10.1590/2175-8239-jbn-2018-0097] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 09/05/2018] [Indexed: 12/18/2022] Open
Abstract
Involvement of the kidneys by lupus nephritis (LN) is one of the most severe clinical manifestations seen in individuals with systemic lupus erythematosus (SLE). LN is more frequent and severe in pediatric patients and has been associated with higher morbidity and mortality rates. This narrative review aimed to describe the general aspects of LN and its particularities when affecting children and adolescents, while focusing on the disease's etiopathogenesis, clinical manifestations, renal tissue alterations, and treatment options.
Collapse
Affiliation(s)
- Sergio Veloso Brant Pinheiro
- Universidade Federal de Minas
GeraisHospital das ClínicasUnidade de Nefrologia
PediátricaBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais,
Hospital das Clínicas, Unidade de Nefrologia Pediátrica, Belo Horizonte, MG,
Brasil.
| | - Raphael Figuiredo Dias
- Universidade Federal de Minas
GeraisHospital das ClínicasUnidade de Nefrologia
PediátricaBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais,
Hospital das Clínicas, Unidade de Nefrologia Pediátrica, Belo Horizonte, MG,
Brasil.
| | - Rafaela Cabral Gonçalves Fabiano
- Universidade Federal de Minas
GeraisHospital das ClínicasUnidade de Nefrologia
PediátricaBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais,
Hospital das Clínicas, Unidade de Nefrologia Pediátrica, Belo Horizonte, MG,
Brasil.
| | - Stanley de Almeida Araujo
- Universidade Federal de Minas
GeraisHospital das ClínicasUnidade de Nefrologia
PediátricaBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais,
Hospital das Clínicas, Unidade de Nefrologia Pediátrica, Belo Horizonte, MG,
Brasil.
| | - Ana Cristina Simões e Silva
- Universidade Federal de Minas
GeraisHospital das ClínicasUnidade de Nefrologia
PediátricaBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais,
Hospital das Clínicas, Unidade de Nefrologia Pediátrica, Belo Horizonte, MG,
Brasil.
| |
Collapse
|
81
|
Gasparin AA, Pamplona Bueno de Andrade N, Hax V, Tres GL, Veronese FV, Monticielo OA. Urinary biomarkers for lupus nephritis: the role of the vascular cell adhesion molecule-1. Lupus 2019; 28:265-272. [PMID: 30712490 DOI: 10.1177/0961203319826695] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Renal involvement is one of the main complications of systemic lupus erythematosus, causing a significant impact on patients' morbidity and mortality. Renal biopsy is still the gold standard of diagnosis, but it has many limitations. In this sense, several recent studies aim to identify biomarkers that not only predict disease activity and renal histology, but also lead to earlier treatment. In previous studies, the soluble vascular cell adhesion molecule-1 measured in urine showed a strong association with the presence of lupus nephritis, with clinical and histological activity indexes of the disease and with more severe renal lesions. This paper reviews the main urinary biomarkers of lupus nephritis that have been studied, with special emphasis on vascular cell adhesion molecule-1 results.
Collapse
Affiliation(s)
- A A Gasparin
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | | | - V Hax
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - G Leví Tres
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - F V Veronese
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - O A Monticielo
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
82
|
Ouyang Q, Huang Q, Jiang Z, Zhao J, Shi GP, Yang M. Using plasma circRNA_002453 as a novel biomarker in the diagnosis of lupus nephritis. Mol Immunol 2018; 101:531-538. [DOI: 10.1016/j.molimm.2018.07.029] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/28/2018] [Accepted: 07/25/2018] [Indexed: 02/07/2023]
|
83
|
Two-Parametric Immunological Score Development for Assessing Renal Involvement and Disease Activity in Systemic Lupus Erythematosus. J Immunol Res 2018; 2018:1294680. [PMID: 30246032 PMCID: PMC6136571 DOI: 10.1155/2018/1294680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 06/05/2018] [Accepted: 06/13/2018] [Indexed: 01/09/2023] Open
Abstract
Objective Anti-double-stranded (ds) DNA and anti-C1q autoantibodies are useful tools in the assessment of disease activity and nephritis in systemic lupus erythematosus (SLE) patients. This study aimed to explore the utility of these antibodies along with anti-Ku antibodies in an oligoparametric model approach for the assessment of disease activity and lupus nephritis. Methods Samples from 261 well-characterized SLE patients were tested using chemiluminescent immunoassays (CIA) for anti-dsDNA and anti-Ku antibodies as well as by anti-C1q antibody ELISA (Inova Diagnostics, USA). Of these SLE patients, 26.4% had lupus nephritis (LN) at the time of blood draw or had a history of LN, and modified SLE disease activity index-2K (SLEDAI) scores were used to assess disease activity. Results All three antibodies demonstrated higher prevalence and higher antibody levels in active versus inactive SLE patients and in LN versus non-LN patients. When oligoparametric analysis was performed, the likelihood of LN and patients with active disease increased with dual and triple positivity. Conclusions Anti-dsDNA and anti-C1q antibodies are useful tools to identify disease activity and/or renal involvement in SLE patients. In addition, the combination of those antibodies in a two-parametric score might improve the clinical utility of those markers.
Collapse
|
84
|
Colliard S, Jourde-Chiche N, Clavarino G, Sarrot-Reynauld F, Gout E, Deroux A, Fougere M, Bardin N, Bouillet L, Cesbron JY, Thielens NM, Dumestre-Pérard C. Autoantibodies Targeting Ficolin-2 in Systemic Lupus Erythematosus Patients With Active Nephritis. Arthritis Care Res (Hoboken) 2018; 70:1263-1268. [PMID: 29045037 DOI: 10.1002/acr.23449] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 10/10/2017] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a multisystem inflammatory disease characterized by the production of various autoantibodies. The aim of this study was to investigate the presence of anti-ficolin-2 antibodies in SLE patients and to evaluate the association between the levels of these autoantibodies, clinical manifestations, and disease activity. METHODS This is a comparative study using a cohort of 165 SLE patients and 48 healthy subjects. SLE patients were further divided into 2 groups (low disease activity [SLE Disease Activity Index (SLEDAI) score ≤4, n = 88] and high disease activity [SLEDAI score >4, n = 77]). Clinical manifestations were defined according to the physician in charge. Active lupus nephritis (LN) was documented by kidney biopsy. Detection of anti-ficolin-2 antibodies was performed by enzyme-linked immunosorbent assay. RESULTS Levels of anti-ficolin-2 autoantibodies were significantly higher in SLE patients as compared to healthy subjects and associated with SLEDAI score. They were found to be positive in 61 of 165 SLE patients (37%). The presence of anti-ficolin-2 antibodies was significantly related only to renal involvement, with a very high prevalence (86%) of anti-ficolin-2 antibodies in SLE patients with active LN. Patients with active proliferative LN had significantly more positive anti-ficolin-2 antibodies than those with nonproliferative LN. The combination of anti-ficolin-2, anti-ficolin-3, and anti-C1q demonstrated a very high specificity (98%) for the diagnosis of active LN. CONCLUSION Our results support the usefulness of anti-ficolin-2 as a complementary serologic biomarker for the diagnosis of active lupus with renal manifestations.
Collapse
Affiliation(s)
- Sophie Colliard
- Laboratoire d'Immunologie, Pôle de Biologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Cedex 9, France
| | - Noémie Jourde-Chiche
- Aix-Marseille Université, Assistance-Publique Hôpitaux de Marseille, Centre de Néphrologie et Transplantation Rénale, Centre Hospitalier Universitaire de la Conception, and Aix-Marseille Université, Vascular Research Center of Marseille, Marseille, France
| | - Giovanna Clavarino
- Laboratoire d'Immunologie, Pôle de Biologie, Centre Hospitalier Universitaire Grenoble Alpes and CNRS-Université Grenoble Alpes, Grenoble Cedex 9, France
| | - Françoise Sarrot-Reynauld
- Clinique Universitaire de Médecine Interne, Pôle Pluridisciplinaire de Médecine et de Gérontologie Clinique, Centre Hospitalier Universitaire, Grenoble Alpes, Grenoble Cedex 9, France
| | - Evelyne Gout
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Alban Deroux
- Clinique Universitaire de Médecine Interne, Pôle Pluridisciplinaire de Médecine et de Gérontologie Clinique, Centre Hospitalier Universitaire, Grenoble Alpes, Grenoble Cedex 9, France
| | - Mélanie Fougere
- Laboratoire d'Immunologie, Pôle de Biologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Cedex 9, France
| | - Nathalie Bardin
- Aix-Marseille Université, UMR_S 1076, Vascular Research Center of Marseille, Hôpital de la Conception, Marseille, Aix-Marseille Université, Marseille, France
| | - Laurence Bouillet
- Clinique Universitaire de Médecine Interne, Pôle Pluridisciplinaire de Médecine et de Gérontologie Clinique, Centre Hospitalier Universitaire, Grenoble Alpes, Grenoble Cedex 9, France
| | - Jean-Yves Cesbron
- Laboratoire d'Immunologie, Pôle de Biologie, Centre Hospitalier Universitaire Grenoble Alpes and CNRS-Université Grenoble Alpes, Grenoble Cedex 9, France
| | | | - Chantal Dumestre-Pérard
- Laboratoire d'Immunologie, Pôle de Biologie, Centre Hospitalier Universitaire Grenoble Alpes and CNRS-Université Grenoble Alpes, Grenoble Cedex 9, France
| |
Collapse
|
85
|
Dumestre-Pérard C, Clavarino G, Colliard S, Cesbron JY, Thielens NM. Antibodies targeting circulating protective molecules in lupus nephritis: Interest as serological biomarkers. Autoimmun Rev 2018; 17:890-899. [PMID: 30009962 DOI: 10.1016/j.autrev.2018.03.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 03/15/2018] [Indexed: 12/15/2022]
Abstract
Lupus nephritis (LN) is one of the most frequent and severe manifestations of systemic lupus erythematosus (SLE), considered as the major predictor of poor prognosis. An early diagnosis of LN is a real challenge in the management of SLE and has an important implication in guiding treatments. In clinical practice, conventional parameters still lack sensitivity and specificity for detecting ongoing disease activity in lupus kidneys and early relapse of nephritis. LN is characterized by glomerular kidney injury, essentially due to deposition of immune complexes involving autoantibodies against cellular components and circulating proteins. One of the possible mechanisms of induction of autoantibodies in SLE is a defect in apoptotic cells clearance and subsequent release of intracellular autoantigens. Autoantibodies against soluble protective molecules involved in the uptake of dying cells, including complement proteins and pentraxins, have been described. In this review, we present the main autoantibodies found in LN, with a focus on the antibodies against these protective molecules. We also discuss their pathogenic role and conclude with their potential interest as serological biomarkers in LN.
Collapse
Affiliation(s)
- Chantal Dumestre-Pérard
- Laboratoire d'Immunologie, Pôle de Biologie, CHU Grenoble Alpes, CS 10217, 38043 Grenoble Cedex 9, France; BNI TIMC-IMAG, UMR5525, CNRS-Université Grenoble Alpes, BP170, 38042 Grenoble Cedex 9, France.
| | - Giovanna Clavarino
- Laboratoire d'Immunologie, Pôle de Biologie, CHU Grenoble Alpes, CS 10217, 38043 Grenoble Cedex 9, France; BNI TIMC-IMAG, UMR5525, CNRS-Université Grenoble Alpes, BP170, 38042 Grenoble Cedex 9, France
| | - Sophie Colliard
- Laboratoire d'Immunologie, Pôle de Biologie, CHU Grenoble Alpes, CS 10217, 38043 Grenoble Cedex 9, France
| | - Jean-Yves Cesbron
- Laboratoire d'Immunologie, Pôle de Biologie, CHU Grenoble Alpes, CS 10217, 38043 Grenoble Cedex 9, France; BNI TIMC-IMAG, UMR5525, CNRS-Université Grenoble Alpes, BP170, 38042 Grenoble Cedex 9, France
| | | |
Collapse
|
86
|
Jakiela B, Kosałka J, Plutecka H, Węgrzyn AS, Bazan-Socha S, Sanak M, Musiał J. Urinary cytokines and mRNA expression as biomarkers of disease activity in lupus nephritis. Lupus 2018; 27:1259-1270. [DOI: 10.1177/0961203318770006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction Renal involvement is one of the most serious manifestations of systemic lupus erythematosus, but non-invasive assessment of inflammatory response in kidneys is challenging. In this study we aimed to validate markers of active lupus nephritis (LN) using urine immune profiling. Methods Urine and serum cytokines (17-plex array) and urine mRNA expression (∼40 immune and glomerular injury genes) were measured in LN patients with active disease ( n = 17) during remission ( n = 16) and in healthy subjects ( n = 18). Results Urine and serum levels of CCL2, CCL5 and CXCL10 were elevated in active LN as compared with disease remission (best discrimination for urine CXCL10 and CCL2) and correlated with LN activity. In the active disease, urinary cell transcriptome showed marked upregulation of proinflammatory cytokines (e.g. TNF, CCL2, CCL5, CXCL10), and type-1 immunity-related genes (e.g. CD3G, CD4, TBX21, IFNG). An active pattern of gene expression was also observed in four patients in remission, who had moderately increased urinary leucocyte count. Two patients from this group developed renal exacerbation during the following 3 months. Markers of type-17 immune axis (e.g. IL-17A) were not significantly increased in active LN. Conclusions Active LN patients were characterized by marked increase of proinflammatory mediators in the urine. Urine cytokines (CCL2 and CXCL10) and type-1 T-cell-related gene markers in the urine sediment had similar diagnostic performance in detection of active LN.
Collapse
Affiliation(s)
- B Jakiela
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - J Kosałka
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - H Plutecka
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - A S Węgrzyn
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Wroclaw Research Centre IET+, Department of Nanobioengineering, Wroclaw, Poland
| | - S Bazan-Socha
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - M Sanak
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - J Musiał
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
87
|
da Rosa Franchi Santos LF, Stadtlober NP, Costa Dall'Aqua LG, Scavuzzi BM, Guimarães PM, Flauzino T, Batisti Lozovoy MA, Mayumi Iriyoda TV, Vissoci Reiche EM, Dichi I, Maes M, Colado Simão A. Increased adhesion molecule levels in systemic lupus erythematosus: relationships with severity of illness, autoimmunity, metabolic syndrome and cortisol levels. Lupus 2018; 27:380-388. [DOI: 10.1177/0961203317723716] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background This study was performed to assess adhesion molecules in systemic lupus erythematosus (SLE). Methods This case-control study examined 126 SLE patients and 48 healthy individuals. Blood levels of six adhesion molecules, cortisol, nuclear autoantibody (ANA) and anti-double stranded DNA (anti-dsDNA) titers were measured, while disease activity was assessed using the SLE Disease Activity Index (SLEDAI) score. Results Platelet endothelial cell adhesion molecule 1 (PECAM-1), vascular cell adhesion molecule 1 (VCAM-1), E-selectin, P-selectin, and plasminogen activator inhibitor type-1 (PAI-1) were significantly higher in SLE patients than in controls. Binary logistic regression analysis showed that PECAM-1 and PAI-1 predicted SLE with a sensitivity of 86.5% and a specificity of 81.3%. ANA titers were significantly and positively associated with PECAM-1, VCAM-1, E-selectin, and PAI-1, whereas there were no associations between anti-dsDNA titers and adhesion molecules. Cortisol was negatively associated with PCAM-1 and ICAM-1. There were significant associations between metabolic syndrome (MetS) and E-selectin and PAI-1. 14.8% of the variance in the SLEDAI score was explained by the regression on PECAM-1 and MetS. Conclusions Our data show that adhesion molecules, especially PECAM-1, are significantly associated with SLE and disease activity, suggesting that they play a role in SLE pathophysiology. While MetS, ANA titers and cortisol levels modulate adhesion molecule levels, these associations do not explain the increased levels of adhesion molecules in SLE. Increased levels of adhesion molecules are new drug targets in SLE.
Collapse
Affiliation(s)
| | - N P Stadtlober
- Graduate Program in Pathology, Clinical Analysis and Toxicology, University of Londrina, Brazil
| | - L G Costa Dall'Aqua
- Graduate Program in Pathology, Clinical Analysis and Toxicology, University of Londrina, Brazil
| | - B M Scavuzzi
- Graduate Program in Health Sciences, University of Londrina, Brazil
| | - P M Guimarães
- Graduate Program in Health Sciences, University of Londrina, Brazil
| | - T Flauzino
- Graduate Program in Health Sciences, University of Londrina, Brazil
| | - M A Batisti Lozovoy
- Department of Pathology, Clinical Analysis and Toxicology, University of Londrina, Brazil
| | | | - E M Vissoci Reiche
- Department of Pathology, Clinical Analysis and Toxicology, University of Londrina, Brazil
| | - I Dichi
- Department of Internal Medicine, University of Londrina, Brazil
| | - M Maes
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - A Colado Simão
- Department of Pathology, Clinical Analysis and Toxicology, University of Londrina, Brazil
| |
Collapse
|
88
|
Liang Y, Xie SB, Wu CH, Hu Y, Zhang Q, Li S, Fan YG, Leng RX, Pan HF, Xiong HB, Ye DQ. Coagulation cascade and complement system in systemic lupus erythematosus. Oncotarget 2017; 9:14862-14881. [PMID: 29599912 PMCID: PMC5871083 DOI: 10.18632/oncotarget.23206] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/16/2017] [Indexed: 12/26/2022] Open
Abstract
This study was conducted to (1) characterize coagulation cascade and complement system in systemic lupus erythematosus (SLE); (2) evaluate the associations between coagulation cascade, complement system, inflammatory response and SLE disease severity; (3) test the diagnostic value of a combination of D-dimer and C4 for lupus activity. Transcriptomics, proteomics and metabolomics were performed in 24 SLE patients and 24 healthy controls. The levels of ten coagulations, seven complements and three cytokines were measured in 112 SLE patients. Clinical data were collected from 2025 SLE patients. The analysis of multi-omics data revealed the common links for the components of coagulation cascade and complement system. The results of ELISA showed coagulation cascade and complement system had an interaction effect on SLE disease severity, this effect was pronounced among patients with excess inflammation. The analysis of clinical data revealed a combination of D-dimer and C4 provided good diagnostic performance for lupus activity. This study suggested that coagulation cascade and complement system become 'partners in crime', contributing to SLE disease severity and identified the diagnostic value of D-dimer combined with C4for lupus activity.
Collapse
Affiliation(s)
- Yan Liang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | | | - Chang-Hao Wu
- Department of Biochemical Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Yuan Hu
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qin Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui, PR China
| | - Si Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui, PR China
| | - Yin-Guang Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui, PR China
| | - Rui-Xue Leng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui, PR China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui, PR China
| | - Hua-Bao Xiong
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui, PR China
| |
Collapse
|
89
|
Increased lipid and protein oxidation and lowered anti-oxidant defenses in systemic lupus erythematosus are associated with severity of illness, autoimmunity, increased adhesion molecules, and Th1 and Th17 immune shift. Immunol Res 2017; 66:158-171. [DOI: 10.1007/s12026-017-8960-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
90
|
Affiliation(s)
- Chaim Putterman
- Division of Rheumatology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA; Department of Microbiology and Immunology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
91
|
Liao W, Zheng H, Wu S, Zhang Y, Wang W, Zhang Z, Zhou C, Wu H, Min J. The Systemic Activation of Programmed Death 1-PD-L1 Axis Protects Systemic Lupus Erythematosus Model from Nephritis. Am J Nephrol 2017; 46:371-379. [PMID: 29069649 DOI: 10.1159/000480641] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/14/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is characterized by abnormal activated T cells, autoreactive B cells, and massive cytokines. The CD4+ T cells determined B-cells differentiation and cytokines production. The programmed death 1 (PD-1) is the checkpoint immunoinhibitory receptor of activated T cells, and its engagement could exhaust T cells. In this study, we investigated the role of PD-1 systemic engagement with PD-L1-Ig in lupus-like nephritis in SLE mice. METHODS The murine PD-L1-Ig was injected into SLE-prone mice. The proteinuria and survival ratio were monitored. The production of anti-dsDNA autoantibodies and cytokines in serum were measured by enzyme-linked immunosorbent assay. The cytokine-producing T cells (interferon-γ, IFN-γ and IL-17α) in kidney and spleen were detected with flowcytometry. The pathological evaluation of the Ig deposition in the glomeruliand was determined with immunofluorescence. Lymphocytes in 24-h urine were detected with flowcytometry. RESULTS The systemic administration of PD-L1-Ig activated PD-1-PD-L1 axis of CD4+ T lymphocytes, suppressed Th17 formation in many organs, including the spleen and the kidney, demolished abnormal production of cytokines (IFN-γ, IL-17, and IL-10) and anti-dsDNA autoantibodies in serum, inhibited immunoglobulin G deposition in the glomeruli with the decrease of proteinuria, and activated T cells in urine. Accordingly, the systemic conjugation of PD-L1-PD-1 impaired renal autoimmune injure and prolonged survival time. CONCLUSION Our research demonstrated that the protective function of systemic activation of PD-1-PD-L1 axis with PD-L1-Ig attenuates the nephritis in SLE-prone mice, which facilitates us to understand the suppressive function of PD-1-PD-L1 axis in the pathogenesis and progress of the lupus nephritis, and to explore a possible effective therapeutic strategy to SLE.
Collapse
Affiliation(s)
- Wenjun Liao
- Department of Nephrology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Bennett MR, Ma Q, Ying J, Devarajan P, Brunner H. Effects of age and gender on reference levels of biomarkers comprising the pediatric Renal Activity Index for Lupus Nephritis (p-RAIL). Pediatr Rheumatol Online J 2017; 15:74. [PMID: 29029629 PMCID: PMC5640910 DOI: 10.1186/s12969-017-0202-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/05/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Systemic Lupus Erythematosus (SLE) is a multisystem autoimmune disease that disproportionately effects women and children of minorities. Renal involvement (lupus nephritis, or LN) occurs in up to 80% of children with SLE and is a major determinant of poor prognosis. We have developed a non-invasive pediatric Renal Activity Index for Lupus (p-RAIL) that consists of laboratory measures that reflect histologic LN activity. These markers are neutrophil gelatinase associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), monocyte chemotactic protein (MCP-1), adiponectin (APN), ceruloplasmin (CP) and hemopexin (HPX). A major gap in the knowledge base and a barrier to clinical utility is how these markers behave in healthy children. We set out to establish a reference range for the p-RAIL markers in a population of healthy children, and to determine if levels of these markers fluctuate with age or gender. METHODS Urine was collected from 368 healthy children presenting to Cincinnati Children's primary care clinic for well child visits and assayed for NGAL, KIM-1, MCP-1, APN, CP and HPX using commercially available kits or assay materials. RESULTS Specimens were grouped by age (0-5 years (n = 94); 5-10 (n = 89); 10-15 (n = 93); 15-20 (n = 91)) and gender (M = 184, F = 184). For age and gender comparisons, values were log transformed prior to analysis. The medians (minimums, maximums) of each marker in the combined population were as follows: NGAL 6.65 (0.004, 391.52) ng/ml, KIM-1416.84 (6.22, 2512.43) pg/ml, MCP-1209.36 (9.49, 2237.06) pg/ml, APN 8.05 (0.07, 124.50) ng/ml, CP 465.15 (8.02, 7827.00) ng/ml, HPX 588.70 (6.85, 17,658.40)ng/ml. All p-RAIL biomarkers but adiponectin had weak but significant positive correlations with age, with NGAL being the strongest (r = 0.33, p < 0.001). For gender comparisons, NGAL, CP and HPX were elevated in females vs males (86%, p < 0.0001; 3%, p = 0.007, and 5%, p = 0.0005 elevation of the log transformed mean, respectively). CONCLUSIONS We have established a reference range for the p-RAIL biomarkers and have highlighted age and gender differences. This information is essential for rational interpretation of studies and clinical trials utilizing the p-RAIL algorithm.
Collapse
Affiliation(s)
- Michael R. Bennett
- 0000 0000 9025 8099grid.239573.9Division Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Qing Ma
- 0000 0000 9025 8099grid.239573.9Division Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Jun Ying
- 0000 0001 2179 9593grid.24827.3bEnvironmental Health, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Prasad Devarajan
- 0000 0000 9025 8099grid.239573.9Division Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Hermine Brunner
- 0000 0000 9025 8099grid.239573.9Rheumatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| |
Collapse
|
93
|
Abstract
Systemic lupus erythematosus (SLE) is a complex and highly heterogeneous disease. By now, no novel drug has been approved by the US FDA in the past 50 years, except Belimumab, a monoclonal antibody to inhibit B-cell activating factor. The stagnating drug development of lupus may be due to our limited understanding of disease etiopathogenesis and the extreme heterogeneity of patient population. Thus, the individualized treatment for SLE becomes necessary. Recently, biomarkers have shown potential in individualized treatment. This review comprehensively summarizes novel potential biomarkers, discusses their current status in preclinical studies and clinical use, sensitivity to treatments and correlation with the disease activity, and provides an insight into the possibility of biomarkers in the utilization of individualized treatment for SLE.
Collapse
Affiliation(s)
- Jinrong Zeng
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
94
|
Yung S, Yap DYH, Chan TM. Recent advances in the understanding of renal inflammation and fibrosis in lupus nephritis. F1000Res 2017; 6:874. [PMID: 28663794 PMCID: PMC5473406 DOI: 10.12688/f1000research.10445.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/12/2017] [Indexed: 01/08/2023] Open
Abstract
Lupus nephritis is a potentially reversible cause of severe acute kidney injury and is an important cause of end-stage renal failure in Asians and patients of African or Hispanic descent. It is characterized by aberrant exaggerated innate and adaptive immune responses, autoantibody production and their deposition in the kidney parenchyma, triggering complement activation, activation and proliferation of resident renal cells, and expression of pro-inflammatory and chemotactic molecules leading to the influx of inflammatory cells, all of which culminate in destruction of normal nephrons and their replacement by fibrous tissue. Anti-double-stranded DNA (anti-dsDNA) antibody level correlates with disease activity in most patients. There is evidence that apart from mediating pathogenic processes through the formation of immune complexes, pathogenic anti-dsDNA antibodies can bind to resident renal cells and induce downstream pro-apoptotic, pro-inflammatory, or pro-fibrotic processes or a combination of these. Recent data also highlight the critical role of macrophages in acute and chronic kidney injury. Though clinically effective, current treatments for lupus nephritis encompass non-specific immunosuppression and the anti-inflammatory action of high-dose corticosteroids. The clinical and histological impact of novel biologics targeting pro-inflammatory molecules remains to be investigated. Insight into the underlying mechanisms that induce inflammatory and fibrotic processes in the kidney of lupus nephritis could present opportunities for more specific novel treatment options to improve clinical outcomes while minimizing off-target untoward effects. This review discusses recent advances in the understanding of pathogenic mechanisms leading to inflammation and fibrosis of the kidney in lupus nephritis in the context of established standard-of-care and emerging therapies.
Collapse
Affiliation(s)
- Susan Yung
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| | - Desmond YH Yap
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| | - Tak Mao Chan
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
95
|
Moulton VR, Suarez-Fueyo A, Meidan E, Li H, Mizui M, Tsokos GC. Pathogenesis of Human Systemic Lupus Erythematosus: A Cellular Perspective. Trends Mol Med 2017. [PMID: 28623084 DOI: 10.1016/j.molmed.2017.05.006] [Citation(s) in RCA: 297] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease affecting multiple organs. A complex interaction of genetics, environment, and hormones leads to immune dysregulation and breakdown of tolerance to self-antigens, resulting in autoantibody production, inflammation, and destruction of end-organs. Emerging evidence on the role of these factors has increased our knowledge of this complex disease, guiding therapeutic strategies and identifying putative biomarkers. Recent findings include the characterization of genetic/epigenetic factors linked to SLE, as well as cellular effectors. Novel observations have provided an improved understanding of the contribution of tissue-specific factors and associated damage, T and B lymphocytes, as well as innate immune cell subsets and their corresponding abnormalities. The intricate web of involved factors and pathways dictates the adoption of tailored therapeutic approaches to conquer this disease.
Collapse
Affiliation(s)
- Vaishali R Moulton
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Abel Suarez-Fueyo
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Esra Meidan
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Department of Rheumatology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hao Li
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
96
|
Aggarwal A, Gupta R, Negi VS, Rajasekhar L, Misra R, Singh P, Chaturvedi V, Sinha S. Urinary haptoglobin, alpha-1 anti-chymotrypsin and retinol binding protein identified by proteomics as potential biomarkers for lupus nephritis. Clin Exp Immunol 2017; 188:254-262. [PMID: 28120479 DOI: 10.1111/cei.12930] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2017] [Indexed: 12/15/2022] Open
Abstract
The study was aimed at identification by proteomics and validation by enzyme-linked immunosorbent assay (ELISA) of potential urinary biomarkers for lupus nephritis. Study subjects comprised 88 systemic lupus erythematosus (SLE) patients and 60 controls (rheumatoid arthritis, diabetes mellitus and healthy individuals). Based on the SLE disease activity index (SLEDAI), patients were classified as active renal (AR), active non-renal (ANR) or inactive disease (ID). Urinary proteins from a group of patients with AR or ID were resolved by two-dimensional gel electrophoresis and identified by matrix-assisted laser desorption ionization-time of flight-mass spectrometry (MALDI-TOF-MS/MS). The selected biomarkers were validated by ELISA using samples from all patients and controls. AR patients were followed-up for 12 months after start of therapy. Three urinary proteins, alpha-1 anti-chymotrypsin (ACT), haptoglobin (HAP) and retinol binding protein (RBP), were detected in patients with AR and not ID. Upon validation, ACT levels were higher in AR patients than the other groups (P < 0·001) and showed good correlation with renal SLEDAI (r = 0·577, P < 0·001) as well as SLEDAI (r = 0·461, P < 0·001). Similarly, HAP levels were > 10-fold higher in AR than other groups (P < 0·001) and correlated well with renal SLEDAI (r = 0·594, P < 0·001) and SLEDAI (r = 0·371, P < 0·01). RBP levels were also higher in AR patients than in other groups (P < 0·05), except diabetes, and showed moderate correlation with renal SLEDAI (r = 0·284, P < 0·008) and SLEDAI (r = 0·316, P < 0·003). Upon follow-up with treatment, levels of all three proteins declined at 6 and 12 months (P < 0·01). Multiple logistic regression identified ACT as the best marker to differentiate AR from ANR. Urinary HAP, ACT and RBP are potential biomarkers for lupus nephritis activity.
Collapse
Affiliation(s)
- A Aggarwal
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - R Gupta
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - V S Negi
- Department of Clinical Immunology, JIPMER, Puducherry, India
| | - L Rajasekhar
- Department of Rheumatology, Nizam Institute of Medical Sciences, Hyderabad, India
| | - R Misra
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - P Singh
- Biochemistry Division, Central Drug Research Institute, Lucknow, India
| | - V Chaturvedi
- Biochemistry Division, Central Drug Research Institute, Lucknow, India
| | - S Sinha
- Department of Clinical Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.,Biochemistry Division, Central Drug Research Institute, Lucknow, India
| |
Collapse
|
97
|
Wu H, Zeng J, Yin J, Peng Q, Zhao M, Lu Q. Organ-specific biomarkers in lupus. Autoimmun Rev 2017; 16:391-397. [PMID: 28212922 DOI: 10.1016/j.autrev.2017.02.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/16/2017] [Indexed: 12/12/2022]
Abstract
Systemic lupus erythematosus (SLE) is a complex and highly heterogeneous disease, which affects multiple organs, including joints, skin, kidneys, heart, hematopoietic system, and nerve system. While the etiopathogenesis of SLE still remains unclear, genetic susceptibilities and aberrant epigenetic modifications are believed to be involved. For precision therapy, it is necessary to assess accurately and objectively organ involvements and disease activity, which is difficult by current clinical laboratory tests. Biomarkers, which are a biologic, genetic, epigenetic or a chemical characteristic and conveniently detectable, serve as measures of disease diagnosis, activity, prognosis, and manifestation prediction, thereby providing instruction for individualized therapy. In addition, biomarkers differ according to different manifestations, since the disease activity index and treatments vary significantly. For example, unlike other non-renal SLE, lupus nephritis requires significant immunosuppressive drugs. Over the past decades, the research on biomarkers in lupus has been strengthened and numerous promising biomarkers have been identified at levels of genomics, transcriptomics and proteomics. In this review, we summarize the conventional and novel biomarkers in the tissue-specific manner, and discuss their roles in specific organ diagnosis, future manifestation prediction, disease activity assessment and their correlation with histology results. By doing so, it aims to shed a light on individualized treatment.
Collapse
Affiliation(s)
- Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Jinrong Zeng
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Jinghua Yin
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Qiao Peng
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China.
| |
Collapse
|
98
|
Doria A, Gershwin ME, Selmi C. From old concerns to new advances and personalized medicine in lupus: The end of the tunnel is approaching. J Autoimmun 2016; 74:1-5. [DOI: 10.1016/j.jaut.2016.08.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 08/23/2016] [Indexed: 12/11/2022]
|