1
|
Tatar Z, Basakci Calik B, Gur Kabul E, Dundar Ok Z, Cobankara V. Investigation of lower extremity performance, balance, and fatigue in individuals with systemic lupus erythematosus: A comparative study. Clin Biomech (Bristol, Avon) 2024; 120:106372. [PMID: 39531864 DOI: 10.1016/j.clinbiomech.2024.106372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The aim of this study was to investigate lower extremity performance, balance, fatigue and pain in individuals with Systemic Lupus Erythematosus and compare them with healthy controls. METHODS 41 participants (Systemic Lupus Erythematosus n = 21 and mean age = 38.33 ± 13.37; healthy group n = 20 and mean age = 38.95 ± 12.62 years) were included in the study. Lower extremity performance was evaluated with timed up and go test and 30 s sit-to-stand test, static and dynamic balance with Sensamove Miniboard, fatigue levels with Visual Analog Scale and Fatigue Severity Scale and pain intensity with Visual Analog Scale. FINDINGS Significant differences were found in favor of the healthy group in the sit-to-stand test (p = 0.001), timed up and go test (p = 0.001), static balance-center (p = 0.020), front (p = 0.001), back (p = 0.002), left (p = 0.001), right (p = 0.001); proprioception-left (p = 0.004), reaction time-front (p = 0.002) and left (p = 0.016); travel time-front (p = 0.001), back (p = 0.001), left (p = 0.001) and right (p = 0.001), Fatigue Severity Scale (p = 0.001); Visual Analog Scale-fatigue (p = 0.001) and Visual Analog Scale-pain (p = 0.001). In Systemic Lupus Erythematosus, timed up and go test had low correlation with travel time-back (r = -0.449; p = 0.041). Visual Analog Scale-fatigue had low correlation with proprioception-left (r = 0.484; p = 0.026) and proprioception-right (r = 0.461; p = 0.035). Visual Analog Scale-pain had moderate correlation with proprioception-back (r = 0.521; p = 0.015) and low correlation with proprioception-right (r = 0.441; p = 0.045). INTERPRETATION Compared to healthy, individuals with Systemic Lupus Erythematosus had worse lower extremity performance, static/dynamic balance, fatigue, and pain. Dynamic balance-back was related to lower extremity performance. Fatigue was related to left-right proprioception, and pain was related to back-right proprioception.
Collapse
Affiliation(s)
- Zulal Tatar
- Faculty of Physiotherapy and Rehabilitation, Pamukkale University, Denizli, Turkey
| | - Bilge Basakci Calik
- Faculty of Physiotherapy and Rehabilitation, Pamukkale University, Denizli, Turkey.
| | - Elif Gur Kabul
- Faculty of Health Sciences, Physiotherapy and Rehabilitation, Uşak University, Uşak, Turkey.
| | - Zeynep Dundar Ok
- Department of Rheumatology, Medical Faculty, Pamukkale University, Denizli, Turkey.
| | - Veli Cobankara
- Department of Rheumatology, Medical Faculty, Pamukkale University, Denizli, Turkey.
| |
Collapse
|
2
|
|
3
|
Value of Urinary Neutrophil Gelatinase-Associated Lipocalin versus Conventional Biomarkers in Predicting Response to Treatment of Active Lupus Nephritis. Int J Nephrol 2020; 2020:8855614. [PMID: 33083057 PMCID: PMC7563084 DOI: 10.1155/2020/8855614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/28/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction Lupus nephritis (LN) affects almost two-thirds of systemic lupus erythematosus (SLE) patients. Despite initial aggressive therapy, up to 25% of patients with LN will progress to permanent renal damage. Conventional serum markers for LN lack the sensitivity of an ideal biomarker. Urinary neutrophil gelatinase-associated lipocalin (UNGAL) is an excellent biomarker for early diagnosis of acute kidney injury and predicting renal outcomes. Objective To measure UNGAL among LN patients to correlate its levels with renal disease activity and to investigate its predictive performance in response to induction therapy. Patients and Methods. 40 SLE patients with biopsy-proven LN class III, IV, or V were randomly selected. The study was conducted in the internal medicine department and outpatient clinic in Ain Shams University Hospitals and completed after six months. UNGAL was measured at baseline, three-month follow-up, and after complete induction therapy. Results In LN patients at baseline, the mean serum creatinine was 2.57 ± 0.96 mg/dL and the mean UNGAL was 33.50 ± 18.34 ng/dL. Mean UNGAL levels of complete response, partial response, and nonresponse groups were 14.48 ± 2.99 ng/mL, 34.49 ± 4.09 ng/mL, and 62.07 ± 14.44 ng/mL, respectively. Based on the ROC curve, we found a better performance of baseline UNGAL to discriminate the complete response group from partial and nonresponse groups to predict response to induction, outperforming conventional biomarkers. The area under the curve was 0.943, and the best cutoff level was 26.5 ng/mL (92.31% sensitivity and 88.89% specificity). Conclusion UNGAL performed better than conventional biomarkers in predicting response to treatment of active LN.
Collapse
|
4
|
Cai Z, Xiang W, Peng X, Ding Y, Liao W, He X. MicroRNA-145 Involves in the Pathogenesis of Renal Vascular Lesions and May Become a Potential Therapeutic Target in Patients with Juvenile Lupus Nephritis. Kidney Blood Press Res 2019; 44:643-655. [PMID: 31430759 DOI: 10.1159/000500923] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/12/2019] [Indexed: 11/19/2022] Open
Abstract
AIMS The current study was conducted with the central objective of investigating the expression of microRNA-145 (miR-145) in renal vascular lesions (RVLs) in juvenile lupus nephritis (JLN) and its possible mechanism. METHODS The clinical data of 49 JLN patients confirmed by renal biopsy were collected and followed by grouping according to the RVLs score after hematoxylin-eosin staining: mild, moderate, and severe groups. In situ hybridization was used to detect the expression of miR-145 in renal vessels which was then being compared among different RVLs groups. Up-LV-miR-145 and LV-miR-NC lentiviral vectors were constructed and transfected into human vascular smooth muscle cells (HVSMCs), respectively. After HVSMCs were treated with 10.0 µg/L platelet-derived growth factor (PDGF)-BB for 24 h, the proliferation, migration, and apoptosis of endothelial cells were detected by MTT, Transwell assay, and flow cytometry, respectively. Western blot was used to detect expression of alpha-smooth muscle actin (α-SM-actin) and osteopontin (OPN). RESULTS The expression of miR-145 in renal vascular cells was statistically significant. The higher the inner membrane ratio, the lesser the miR-145 expression. After treatment with PDGF-BB, expression of miR-145 in HVSMCs decreased, proliferation and migration ability enhanced, apoptosis decreased, α-SM-actin decreased, and OPN increased. The proliferation and migration ability of HVSMCs in the LV-miR-145 group suppressed, apoptosis enhanced, α-SM-actin increased, and OPN decreased. CONCLUSIONS Our study revealed that miR-145 expression decreased with the increase of vascular damage. miR-145 can inhibit proliferation, migration, and differentiation phenotypic transformation of HVSMCs induced by PDGF-BB. miR-145 may be involved in the pathogenesis of RVLs and may be a new target for treatment of RVLs in lupus nephritis.
Collapse
Affiliation(s)
- Zhaomin Cai
- Department of Clinical Laboratory, People's Hospital of Baoan District of Shenzhen, Shenzhen, China
| | - Wei Xiang
- Department of Pediatrics, Hainan Provincial Maternal Hospital, Hainan Province, Haikou, China
| | - Xiaojie Peng
- Department of Nephrology, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Yan Ding
- Department of Dermatology, Hainan Provincial Dermatology Disease Hospital, Haikou, China
| | - Wang Liao
- Department of Cardiology, Hainan General Hospital, Haikou, China
| | - Xiaojie He
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, Central South University, Changsha, China,
| |
Collapse
|
5
|
Zeid MMH, Baddour NM, El-Neily DAEM, Elshair HS, Mamdouh M. Study of urinary interferon gamma-induced protein 10 (IP-10) and urinary soluble CD 25 (sCD25) as markers of lupus nephritis and their relation to histological class. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2018.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
| | | | | | - Heba Selim Elshair
- Internal Medicine Department, Faculty of Medicine, Alexandria University, Egypt
| | - Mohamed Mamdouh
- Internal Medicine Department, Faculty of Medicine, Alexandria University, Egypt
| |
Collapse
|
6
|
Sun J, Zhang S, Liu JS, Gui M, Zhang H. Expression of vitamin D receptor in renal tissue of lupus nephritis and its association with renal injury activity. Lupus 2019; 28:290-294. [PMID: 30691345 DOI: 10.1177/0961203319826704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective Vitamin D receptor (VDR) has potent anti-inflammatory activities. VDR gene polymorphism has been linked with systemic lupus erythematosus (SLE). However, its expression in the kidney has not been evaluated. This study aimed to investigate the relationship between VDR expression and renal pathology as well as clinical manifestations in lupus nephritis (LN). Methods A total of 20 renal biopsy specimens from 35 patients with LN were classified according to the International Society of Nephrology/Renal Pathology Society 2003 LN-type standards pathological type, and the activity index and chronicity index were determined. Five normal renal tissue samples were obtained from surrounding areas distal to nephronophthisis or renal tumors (>2 cm). The expression of VDR was assessed by immunohistochemistry. The relationships between VDR expression and histological injury index, proteinuria and Systemic Lupus International Collaborating Clinics (SLICC) renal activity scores were analyzed. Results As compared to the control group, the expression of VDR in the LN group was lower ( p < 0.001) and negatively correlated with activity index (r = –0.548, p = 0.012) but not with chronicity index (r = –0.277, p = 0.236). The expression of VDR in renal tissue was also associated with SLICC renal activity scores (r = –0.470, p = 0.037). Conclusion The down-regulation of VDR expression in renal tissues of LN patients was negatively correlated with renal activity and injury severity.
Collapse
Affiliation(s)
- J Sun
- Department of Nephrology and Rheumatology, Third Xiangya Hospital, Central South University,Changsha,China
| | - S Zhang
- Department of Nephrology and Rheumatology, Third Xiangya Hospital, Central South University,Changsha,China
| | - J S Liu
- Department of Nephrology and Rheumatology, Third Xiangya Hospital, Central South University,Changsha,China
| | - M Gui
- Department of Nephrology and Rheumatology, Third Xiangya Hospital, Central South University,Changsha,China
| | - H Zhang
- Department of Nephrology and Rheumatology, Third Xiangya Hospital, Central South University,Changsha,China
| |
Collapse
|
7
|
Niederkorn A, Frühauf J, Schwantzer G, Wutte N, Painsi C, Werner S, Stradner M, Berghold A, Hermann J, Aberer E. CXCL13 is an activity marker for systemic, but not cutaneous lupus erythematosus: a longitudinal cohort study. Arch Dermatol Res 2018; 310:485-493. [DOI: 10.1007/s00403-018-1836-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/16/2018] [Accepted: 04/25/2018] [Indexed: 12/20/2022]
|
8
|
Zhang CX, Cai L, Shao K, Wu J, Zhou W, Cao LF, Chen TX. Serum IP-10 is useful for identifying renal and overall disease activity in pediatric systemic lupus erythematosus. Pediatr Nephrol 2018; 33:837-845. [PMID: 29264699 DOI: 10.1007/s00467-017-3867-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Traditional serological biomarkers often fail to assess systemic lupus erythematosus (SLE) disease activity and discriminate lupus nephritis (LN). The aim of this study was to identify novel markers for evaluating renal and overall disease activity in Chinese patients with pediatric systemic lupus erythematosus (pSLE). METHODS The study included 46 patients with pSLE (35 girls, 11 boys; average age 13.3 ± 2.6 years) and 31 matched healthy controls (22 girls, 9 boys; average age 12.3 ± 2.4 years). The SLE Disease Activity Index (SLEDAI) and renal SLEDAI were used to assess disease activity. Nine different soluble mediators in plasma, including tumor necrosis factor alpha (TNF-α), platelet-derived growth factor-BB (PDGF-BB), interferon (IFN) gamma inducible protein 10 (IP-10), interleukin (IL)-1β, IFN-γ, IL-17A, IL-2, Fas and Fas ligand, were measured by Luminex assay and compared between patients with active and inactive pSLE as well as between patients with pSLE with active and inactive renal disease. Receiver operating characteristic curve analysis was used to measure the discrimination accuracy. RESULTS Of the 46 patients with pSLE, 30 (65.2%) had LN. These patients had significantly elevated levels of serum TNF-α, PDGF-BB, IP-10 and Fas. The serum levels of IP-10 were also significantly higher in patients with active pSLE. We found that IP-10 was also more sensitive and specific than conventional laboratory parameters, including anti-double-stranded DNA and complement components C3 and C4, for distinguishing active lupus from quiescent lupus. The serum level of IP-10 was also significantly increased in children with pSLE with active renal disease relative to those with inactive renal disease. There was also a positive correlation between serum IP-10 levels and renal SLEDAI scores as well as with 24 h urine protein. CONCLUSIONS Serum IP-10 is useful for identifying renal and overall disease activity in children with pSLE.
Collapse
Affiliation(s)
- Chen-Xing Zhang
- Department of Allergy and Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University, 1678, Dongfang Road, Shanghai, China.,Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Cai
- Department of Allergy and Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University, 1678, Dongfang Road, Shanghai, China.,Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kang Shao
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Wu
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhou
- Department of Nephrology and Rheumatology, Shanghai Children's Medical Center, Shanghai Jiao Tong University, Shanghai, China
| | - Lan-Fang Cao
- Department of Pediatrics, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Tong-Xin Chen
- Department of Allergy and Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University, 1678, Dongfang Road, Shanghai, China. .,Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University, Shanghai, China. .,Department of Nephrology and Rheumatology, Shanghai Children's Medical Center, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
9
|
Xu R, Li Q, Liu R, Shen J, Li M, Zhao M, Wang M, Liao Q, Mao H, Li Z, Zhou N, Yin P, Li Y, Tang X, Wu T, Zhong Z, Wang Y, Ai Z, Wang O, Chen N, Yang X, Fang J, Fu P, Gu J, Ye K, Chen J, Dai L, Liu H, Liu Z, Liao Y, Wan J, Ding G, Zhao J, Zhang H, Fu S, Sun L, Zhang X, Yang H, Wang J, Wang J, Liu J, Li Y, Yu X. Association Analysis of the MHC in Lupus Nephritis. J Am Soc Nephrol 2017; 28:3383-3394. [PMID: 28754791 DOI: 10.1681/asn.2016121331] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 05/29/2017] [Indexed: 02/05/2023] Open
Abstract
Lupus nephritis (LN) is one of the most prevalent and serious complications of SLE, with significant effects on patient and renal survival. Although a large number of genetic variants associated with SLE have been identified, biomarkers that correlate with LN are extremely limited. In this study, we performed a comprehensive sequencing analysis of the whole MHC region in 1331 patients with LN and 1296 healthy controls and validated the independent associations in another 950 patients with LN and 1000 controls. We discovered five independent risk variants for LN within the MHC region, including HLA-DRβ1 amino acid 11 (Pomnibus<0.001), HLA-DQβ1 amino acid 45 (P<0.001; odds ratio, 0.58; 95% confidence interval, 0.52 to 0.65), HLA-A amino acid 156 (Pomnibus<0.001), HLA-DPβ1 amino acid 76 (Pomnibus<0.001), and a missense variant in PRRC2A (rs114580964; P<0.001; odds ratio, 0.38; 95% confidence interval, 0.30 to 0.49) at genome-wide significance. These data implicate aberrant peptide presentation by MHC classes 1 and 2 molecules and sex hormone modulation in the development of LN.
Collapse
Affiliation(s)
- Ricong Xu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangzhou, Guangdong, China.,Department of Nephrology, Shenzhen Second People's Hospital and the First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qibin Li
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Rongjun Liu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangzhou, Guangdong, China.,Nephrology and Rheumatology Department, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong, China
| | - Juan Shen
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Ming Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangzhou, Guangdong, China
| | - Minghui Zhao
- Renal Division, Peking University First Hospital, Peking University, Institute of Nephrology, Beijing, China
| | - Meng Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangzhou, Guangdong, China
| | - Qijun Liao
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangzhou, Guangdong, China
| | - Zhijian Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangzhou, Guangdong, China
| | - Na Zhou
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Peiran Yin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangzhou, Guangdong, China
| | - Yue Li
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Xueqing Tang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangzhou, Guangdong, China
| | - Tian Wu
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Zhong Zhong
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangzhou, Guangdong, China
| | - Yan Wang
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Zhen Ai
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangzhou, Guangdong, China
| | - Ou Wang
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Nan Chen
- Department of Nephrology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | | - Ping Fu
- Department of Nephrology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jieruo Gu
- Department of Rheumatology, The Third Affiliated Hospital and
| | - Kun Ye
- Department of Nephrology, The People's Hospital of Guangxi Autonomous Region, Nanning, Guangxi, China
| | - Jian Chen
- Department of Nephrology, Fuzhou General Hospital of Nanjing Military Command, Fuzhou, Fujian, China
| | - Lie Dai
- Department of Rheumatology and Clinical Immunology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huafeng Liu
- Institute of Nephrology, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yunhua Liao
- Department of Nephrology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Jianxin Wan
- Department of Nephrology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Guohua Ding
- Department of Nephrology, Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Jinghong Zhao
- Department of Nephrology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuxia Fu
- Department of Nephrology, The Second Hospital, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Liangdan Sun
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital and.,Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui, China
| | - Xuejun Zhang
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, Anhui, China.,Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Huanming Yang
- BGI Genomics, BGI-Shenzhen, Shenzhen, China.,James D. Watson Institute of Genome Sciences, Hangzhou, China
| | - Jian Wang
- BGI Genomics, BGI-Shenzhen, Shenzhen, China.,James D. Watson Institute of Genome Sciences, Hangzhou, China
| | - Jun Wang
- BGI Genomics, BGI-Shenzhen, Shenzhen, China.,Department of Biology, University of Copenhagen, Copenhagen, Denmark.,Institute of Dermatology and Department of Dermatology, No. 1 Hospital and
| | - Jianjun Liu
- Institute of Dermatology and Department of Dermatology, No. 1 Hospital and.,Princess Al Jawhara Albrahim Center of Excellence in the Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia; and.,School of Biological Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Yingrui Li
- BGI Genomics, BGI-Shenzhen, Shenzhen, China;
| | - Xueqing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Key Laboratory of Nephrology, Ministry of Health, Guangzhou, Guangdong, China; .,Institute of Nephrology, Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
10
|
Buyon J, Furie R, Putterman C, Ramsey-Goldman R, Kalunian K, Barken D, Conklin J, Dervieux T. Reduction in erythrocyte-bound complement activation products and titres of anti-C1q antibodies associate with clinical improvement in systemic lupus erythematosus. Lupus Sci Med 2016; 3:e000165. [PMID: 27752336 PMCID: PMC5051407 DOI: 10.1136/lupus-2016-000165] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 08/17/2016] [Accepted: 09/01/2016] [Indexed: 11/16/2022]
Abstract
Background The relationship between cell-bound complement activation products (CB-CAPs: EC4d, EC3d), anti-C1q, soluble complement C3/C4 and disease activity in systemic lupus erythematosus (SLE) was evaluated. Methods Per protocol, at baseline all SLE subjects enrolled in this longitudinal study presented with active disease and elevated CB-CAPs. At each monthly visit, the non-serological (ns) Safety of Estrogens in Lupus Erythematosus: National Assessment (SELENA-SLEDAI) and the British Isles Lupus Assessment Group (BILAG)-2004 index scores were determined as was a random urinary protein to creatinine ratio (uPCR). Short-form 36 (SF-36) questionnaires were also collected. All soluble markers were determined using immunoassays, while EC4d and EC3d were determined using flow cytometry. Statistical analysis consisted of linear mixed models with random intercept and fixed slopes. Results A total of 36 SLE subjects (mean age 34 years; 94% female) were enrolled and evaluated monthly for an average 11 visits per subject. Clinical improvements were observed during the study, with significant decreases in ns-SELENA-SLEDAI scores, BILAG-2004 index scores and uPCR, and increases in all domains of SF-36 (p<0.01). The longitudinal decrease in ns-SELENA-SLEDAI and BILAG-2004 index scores was significantly associated with reduced EC4d and EC3d levels, reduced anti-C1q titres and increased serum complement C3/C4 (p<0.05). The changes in uPCR significantly correlated with C3, C4, anti-C1q and EC4d, with EC4d outperforming C3/C4 by a multivariate analysis. The reduced EC4d or EC3d was associated with improvements in at least six out of the eight domains of SF-36 and outperformed C3/C4. Anti-dsDNA titres did not correlate with changes in disease activity. Conclusions These data indicate that CB-CAPs and anti-C1q are helpful in monitoring patients with SLE.
Collapse
Affiliation(s)
- Jill Buyon
- NYU School of Medicine , New York, New York , USA
| | - Richard Furie
- Hofstra Northwell School of Medicine , New York, New York , USA
| | - Chaim Putterman
- Albert Einstein College of Medicine and Montefiore Medical Center , Bronx, New York , USA
| | | | | | | | | | | |
Collapse
|
11
|
El-Shereef RR, Lotfi A, Abdel-Naeam EA, Tawfik H. Serum and Urinary Interleukin-6 in Assessment of Renal Activity in Egyptian Patients with Systemic Lupus Erythematosus. CLINICAL MEDICINE INSIGHTS-ARTHRITIS AND MUSCULOSKELETAL DISORDERS 2016; 9:29-36. [PMID: 26966395 PMCID: PMC4782971 DOI: 10.4137/cmamd.s32269] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/08/2015] [Accepted: 11/15/2015] [Indexed: 11/13/2022]
Abstract
AIM OF THE WORK This study investigates whether serum and urinary interleukin-6 (IL-6) represent an early marker of kidney involvement and assesses the difference between them and renal biopsy in lupus nephritis (LN). PATIENTS AND METHODS A total of 60 systemic lupus erythematosus (SLE) patients were compared to 20 healthy controls. Urinary and serum IL-6 were measured in both patients and controls. In addition, renal biopsy was done prior or shortly after urine and blood sampling; the results were classified according to the International Society of Nephrology/Renal Pathology Society classification of LN by recording the activity score and chronicity score for each sample. RESULTS There was a significant higher level of urinary IL-6 in the SLE patients with biopsy-proven LN than in those without LN and those of the control group. However, no significant difference was reported between the three groups as regards serum IL-6. A strong positive correlation was found between urinary IL-6 and renal disease activity based on the renal SLE disease activity index (SLEDAI) score with no significant correlation regarding the extra renal SLEDAI. Urinary IL-6 was positively correlated with renal biopsy results and with its activity scores but weakly correlated with the chronicity scores. CONCLUSION Urinary IL-6 may provide a simple noninvasive potential marker of disease activity of renal involvement in adult patients with SLE.
Collapse
Affiliation(s)
- Rawhya R El-Shereef
- Department of Rheumatology and Rehabilitation, Minia University, Minia, Egypt
| | - Ahmed Lotfi
- Department of Rheumatology and Rehabilitation, Minia University, Minia, Egypt
| | | | - Heba Tawfik
- Department of Histopathology, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
12
|
L'Imperio V, Smith A, Chinello C, Pagni F, Magni F. Proteomics and glomerulonephritis: A complementary approach in renal pathology for the identification of chronic kidney disease related markers. Proteomics Clin Appl 2016; 10:371-83. [DOI: 10.1002/prca.201500075] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/16/2015] [Accepted: 12/02/2015] [Indexed: 12/25/2022]
Affiliation(s)
| | - Andrew Smith
- Department of Health Sciences; University Milan Bicocca; Monza Italy
| | - Clizia Chinello
- Department of Health Sciences; University Milan Bicocca; Monza Italy
| | - Fabio Pagni
- Department of Pathology; University Milan Bicocca; Monza Italy
| | - Fulvio Magni
- Department of Health Sciences; University Milan Bicocca; Monza Italy
| |
Collapse
|
13
|
Affiliation(s)
- Jose Manuel Monroy Trujillo
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Derek Michael Fine
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
14
|
Závada J, Uher M, Svobodová R, Olejárová M, Hušáková M, Ciferská H, Hulejová H, Tomčík M, Šenolt L, Vencovský J. Serum tenascin-C discriminates patients with active SLE from inactive patients and healthy controls and predicts the need to escalate immunosuppressive therapy: a cohort study. Arthritis Res Ther 2015; 17:341. [PMID: 26608564 PMCID: PMC4660660 DOI: 10.1186/s13075-015-0862-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/13/2015] [Indexed: 01/13/2023] Open
Abstract
INTRODUCTION The aim of this study was to examine whether circulating levels of the proinflammatory glycoprotein tenascin-C (TNC) are useful as an activity-specific or predictive biomarker in systemic lupus erythematosus (SLE). METHODS Serum TNC levels were determined by enzyme-linked immunosorbent assay at inception visit in a prospective cohort of 59 SLE patients, and in 65 healthy controls (HC). SLE patients were followed for a mean of 11 months, disease activity was assessed using the Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2 K) and British Isles Lupus Assessment Group disease activity index (BILAG-2004), clinical and laboratory data were recorded every 3-6 months, and changes in glucocorticoids (GC) and immunosuppressants (IS) were recorded serially. We examined cross-sectionally the relationships between serum concentrations of TNC and SLE status, SLEDAI-2 K scores, strata of disease activity, and levels of conventional biomarkers [anti-double-stranded DNA (dsDNA), anti-nucleosome antibodies, C3 and C4]. We also explored the utility of TNC levels for predicting disease flares, defined as (i) new/increased GC, (ii) new/increased GC or IS, and (iii) increase in SLEDAI by ≥3 or (iv) BILAG A or B flare. RESULTS There was no significant difference in the mean levels of TNC between the SLE patients and HC. However, in SLE patients with active disease (SLEDAI ≥6), the TNC levels were significantly higher than in the HC (p = 0.004) or in patients with no/low disease activity (p = 0.004). In SLE patients, TNC levels were significantly associated with positivity of anti-dsDNA (p = 0.03) and anti-nucleosome antibodies (p = 0.008). Flares defined by a need to escalate immunosuppressive therapy were captured more frequently and earlier than flares defined by standard activity indices. Higher baseline levels of serum TNC presented a significantly greater risk of flare (i) [hazard ratio (HR) 1.39, 95% confidence interval (CI) 1.11-1.73] or (ii) (HR 1.25, 95% CI 1.02-1.52) but not of flares (iii) or (iv). The baseline serum TNC level was the single most important independent predictor of flare (i) compared with conventional biomarkers. CONCLUSIONS TNC is not disease-specific, but it seems to indicate the activity of SLE and may predict the need to escalate immunosuppressive therapy. TNC levels may thus serve as a useful activity-specific and predictive biomarker in SLE.
Collapse
Affiliation(s)
- Jakub Závada
- Institute of Rheumatology, Prague, and Department of Rheumatology, First Faculty of Medicine, Charles University in Prague, Na Slupi 4, Praha 2, 12850, Prague, Czech Republic.
| | - Michal Uher
- Institute of Biostatistics and Analyses, Masaryk University, Brno, Czech Republic.
| | - Radka Svobodová
- Institute of Rheumatology, Prague, and Department of Rheumatology, First Faculty of Medicine, Charles University in Prague, Na Slupi 4, Praha 2, 12850, Prague, Czech Republic.
| | - Marta Olejárová
- Institute of Rheumatology, Prague, and Department of Rheumatology, First Faculty of Medicine, Charles University in Prague, Na Slupi 4, Praha 2, 12850, Prague, Czech Republic.
| | - Markéta Hušáková
- Institute of Rheumatology, Prague, and Department of Rheumatology, First Faculty of Medicine, Charles University in Prague, Na Slupi 4, Praha 2, 12850, Prague, Czech Republic.
| | - Hana Ciferská
- Institute of Rheumatology, Prague, and Department of Rheumatology, First Faculty of Medicine, Charles University in Prague, Na Slupi 4, Praha 2, 12850, Prague, Czech Republic.
| | - Hana Hulejová
- Institute of Rheumatology, Prague, and Department of Rheumatology, First Faculty of Medicine, Charles University in Prague, Na Slupi 4, Praha 2, 12850, Prague, Czech Republic.
| | - Michal Tomčík
- Institute of Rheumatology, Prague, and Department of Rheumatology, First Faculty of Medicine, Charles University in Prague, Na Slupi 4, Praha 2, 12850, Prague, Czech Republic.
| | - Ladislav Šenolt
- Institute of Rheumatology, Prague, and Department of Rheumatology, First Faculty of Medicine, Charles University in Prague, Na Slupi 4, Praha 2, 12850, Prague, Czech Republic.
| | - Jiří Vencovský
- Institute of Rheumatology, Prague, and Department of Rheumatology, First Faculty of Medicine, Charles University in Prague, Na Slupi 4, Praha 2, 12850, Prague, Czech Republic.
| |
Collapse
|
15
|
Pretorius E, Kell DB. Diagnostic morphology: biophysical indicators for iron-driven inflammatory diseases. Integr Biol (Camb) 2014; 6:486-510. [PMID: 24714688 DOI: 10.1039/c4ib00025k] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Most non-communicable diseases involve inflammatory changes in one or more vascular systems, and there is considerable evidence that unliganded iron plays major roles in this. Most studies concentrate on biochemical changes, but there are important biophysical correlates. Here we summarize recent microscopy-based observations to the effect that iron can have major effects on erythrocyte morphology, on erythrocyte deformability and on both fibrinogen polymerization and the consequent structure of the fibrin clots formed, each of which contributes significantly and negatively to such diseases. We highlight in particular type 2 diabetes mellitus, ischemic thrombotic stroke, systemic lupus erythematosus, hereditary hemochromatosis and Alzheimer's disease, while recognizing that many other diseases have co-morbidities (and similar causes). Inflammatory biomarkers such as ferritin and fibrinogen are themselves inflammatory, creating a positive feedback that exacerbates disease progression. The biophysical correlates we describe may provide novel, inexpensive and useful biomarkers of the therapeutic benefits of successful treatments.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Private Bag x323, Arcadia 0007, South Africa.
| | | |
Collapse
|
16
|
Li T, Prokopec SD, Morrison S, Lou W, Reich H, Gladman D, Urowitz M, Scholey J, Fortin PR, Boutros PC, Wither J, Landolt-Marticorena C. Anti-nucleosome antibodies outperform traditional biomarkers as longitudinal indicators of disease activity in systemic lupus erythematosus. Rheumatology (Oxford) 2014; 54:449-57. [PMID: 25193804 DOI: 10.1093/rheumatology/keu326] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE The aim of this study was to determine whether anti-nucleosome antibodies function as activity-specific biomarkers in SLE. METHODS Fifty-one patients were recruited and followed prospectively with periodic clinical and biochemical assessments over a 14-month period. Disease activity was determined by the SLEDAI-2K. Anti-nucleosome antibody levels were measured by an ELISA and its utility as an activity-specific biomarker as compared with that of anti-dsDNA antibodies and C3 was assessed both at baseline and in longitudinal analysis. RESULTS Anti-nucleosome antibodies were significantly elevated in SLE patients vs controls and showed a moderate positive correlation with disease activity. The utility of anti-nucleosome antibodies in identifying patients with active disease in a cross-sectional analysis was comparable to that of anti-dsDNA antibodies and C3. Analysis of variance demonstrated that the level of anti-nucleosome antibodies and C3 varied significantly with changes in disease activity over time. Changes in clinical state were not mirrored by changes in anti-dsDNA antibodies. In time-dependent analysis, anti-nucleosome antibodies showed a better fit over time than anti-dsDNA antibodies and C3. In pairwise comparisons, C3 and anti-nucleosome antibodies outperformed other models, including the conventional pairing of C3 and anti-dsDNA antibodies, however, no biomarker alone or as a group accurately predicted impending remissions or exacerbations. CONCLUSION Anti-nucleosome antibodies demonstrate greater fidelity as a biomarker for changes in SLE disease activity than traditional biomarkers, supporting the routine monitoring of this antibody in clinical practice.
Collapse
Affiliation(s)
- Timothy Li
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Stephenie D Prokopec
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Stacey Morrison
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Wendy Lou
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Heather Reich
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Dafna Gladman
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University
| | - Murray Urowitz
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University
| | - James Scholey
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Paul R Fortin
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Paul C Boutros
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University
| | - Joan Wither
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University
| | - Carolina Landolt-Marticorena
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network (UHN), Ontario Institute for Cancer Research, University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, University Health Network, Division of Biostatistics, Dalla Lana School of Public Health, Department of Medicine, University of Toronto, Arthritis Centre of Excellence, Division of Health Care and Outcomes Research, Toronto Western Hospital Research Institute, University Health Network, Toronto, ON, Centre de recherche du CHU de Québec, CHU de Québec, Division of Rheumatology, Department of Medicine, CHU de Québec and Faculty of Medicine, Université Laval, Quebec City, QC, Department of Immunology, Department of Medical Biophysics and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University
| |
Collapse
|
17
|
Yang X, Gao Y, Wang H, Zhao X, Gong X, Wang Q, Zhang X. Increased urinary interleukin 22 binding protein levels correlate with lupus nephritis activity. J Rheumatol 2014; 41:1793-1800. [PMID: 25086075 DOI: 10.3899/jrheum.131292] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Interleukin 22 (IL-22) plays an important role in the promotion of antimicrobial immunity. However, dysregulated IL-22 action leads to inflammation and is involved in autoimmune diseases, including systemic lupus erythematosus (SLE). IL-22 binding protein (IL-22BP) is a soluble inhibitory IL-22 receptor and may represent a crucial regulator of IL-22. We investigated the expression and potential significance of serum and urinary IL-22BP levels in patients with SLE. METHODS A total of 112 patients with SLE and healthy control subjects participated in our study. Patients were classified according to kidney involvement and disease activity based on clinical and laboratory measures such as urinary sediment, proteinuria, kidney function, complement factor 3 (C3), C4, anti-dsDNA, disease activity index, and renal SLE disease activity index. The concentrations of IL-22BP and IL-22 were measured by ELISA. The expression of IL-22BP in the renal tissue was detected by immunohistochemistry. RESULTS Patients with active renal disease had urinary levels of IL-22BP higher than (1) patients with active SLE but no renal involvement, (2) patients with a history of lupus nephritis in remission with no systemic disease activity and no history of renal involvement, and (3) control subjects. There was no difference in serum levels of IL-22BP among the groups. Urinary levels of IL-22BP in patients with active renal disease were positively correlated with SLE Disease Activity Index, Systemic Lupus International Collaborating Clinics renal activity score, and histological activity index. IL-22BP was highly expressed in renal tissue of patients with active renal disease. After 6 months of treatment, urinary IL-22BP levels decreased significantly in patients with complete response, but remained unchanged in those with partial or no response. CONCLUSION Urinary but not serum IL-22BP levels were associated with active renal disease. Urinary levels of IL-22BP might be a potential marker for the presence of renal involvement in patients with SLE.
Collapse
Affiliation(s)
- Xuyan Yang
- From the Department of Rheumatology, Second Affiliated Hospital, College of Medicine, and Department of Immunology, Institute of Basic Medical Sciences, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.X.Y. Yang, MD; Y. Gao, MD, Department of Rheumatology; H.Y. Wang, MD, PhD, Department of Allergy and Clinical Immunology; X.Y. Zhao, MD, Department of Hematology; X.B. Gong, MD, Laboratory of Bone Marrow, Second Affiliated Hospital, College of Medicine, Zhejiang University; Q.Q. Wang, MD, PhD, Department of Immunology, Institute of Basic Medical Sciences; X.F. Zhang, MD, Department of Clinical Epidemiology and Biostatistics, Second Affiliated Hospital, College of Medicine, Zhejiang University
| | - Yin Gao
- From the Department of Rheumatology, Second Affiliated Hospital, College of Medicine, and Department of Immunology, Institute of Basic Medical Sciences, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.X.Y. Yang, MD; Y. Gao, MD, Department of Rheumatology; H.Y. Wang, MD, PhD, Department of Allergy and Clinical Immunology; X.Y. Zhao, MD, Department of Hematology; X.B. Gong, MD, Laboratory of Bone Marrow, Second Affiliated Hospital, College of Medicine, Zhejiang University; Q.Q. Wang, MD, PhD, Department of Immunology, Institute of Basic Medical Sciences; X.F. Zhang, MD, Department of Clinical Epidemiology and Biostatistics, Second Affiliated Hospital, College of Medicine, Zhejiang University
| | - Huiying Wang
- From the Department of Rheumatology, Second Affiliated Hospital, College of Medicine, and Department of Immunology, Institute of Basic Medical Sciences, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.X.Y. Yang, MD; Y. Gao, MD, Department of Rheumatology; H.Y. Wang, MD, PhD, Department of Allergy and Clinical Immunology; X.Y. Zhao, MD, Department of Hematology; X.B. Gong, MD, Laboratory of Bone Marrow, Second Affiliated Hospital, College of Medicine, Zhejiang University; Q.Q. Wang, MD, PhD, Department of Immunology, Institute of Basic Medical Sciences; X.F. Zhang, MD, Department of Clinical Epidemiology and Biostatistics, Second Affiliated Hospital, College of Medicine, Zhejiang University
| | - Xiaoying Zhao
- From the Department of Rheumatology, Second Affiliated Hospital, College of Medicine, and Department of Immunology, Institute of Basic Medical Sciences, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.X.Y. Yang, MD; Y. Gao, MD, Department of Rheumatology; H.Y. Wang, MD, PhD, Department of Allergy and Clinical Immunology; X.Y. Zhao, MD, Department of Hematology; X.B. Gong, MD, Laboratory of Bone Marrow, Second Affiliated Hospital, College of Medicine, Zhejiang University; Q.Q. Wang, MD, PhD, Department of Immunology, Institute of Basic Medical Sciences; X.F. Zhang, MD, Department of Clinical Epidemiology and Biostatistics, Second Affiliated Hospital, College of Medicine, Zhejiang University.
| | - Xubo Gong
- From the Department of Rheumatology, Second Affiliated Hospital, College of Medicine, and Department of Immunology, Institute of Basic Medical Sciences, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.X.Y. Yang, MD; Y. Gao, MD, Department of Rheumatology; H.Y. Wang, MD, PhD, Department of Allergy and Clinical Immunology; X.Y. Zhao, MD, Department of Hematology; X.B. Gong, MD, Laboratory of Bone Marrow, Second Affiliated Hospital, College of Medicine, Zhejiang University; Q.Q. Wang, MD, PhD, Department of Immunology, Institute of Basic Medical Sciences; X.F. Zhang, MD, Department of Clinical Epidemiology and Biostatistics, Second Affiliated Hospital, College of Medicine, Zhejiang University
| | - Qingqing Wang
- From the Department of Rheumatology, Second Affiliated Hospital, College of Medicine, and Department of Immunology, Institute of Basic Medical Sciences, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.X.Y. Yang, MD; Y. Gao, MD, Department of Rheumatology; H.Y. Wang, MD, PhD, Department of Allergy and Clinical Immunology; X.Y. Zhao, MD, Department of Hematology; X.B. Gong, MD, Laboratory of Bone Marrow, Second Affiliated Hospital, College of Medicine, Zhejiang University; Q.Q. Wang, MD, PhD, Department of Immunology, Institute of Basic Medical Sciences; X.F. Zhang, MD, Department of Clinical Epidemiology and Biostatistics, Second Affiliated Hospital, College of Medicine, Zhejiang University
| | - Xiaofei Zhang
- From the Department of Rheumatology, Second Affiliated Hospital, College of Medicine, and Department of Immunology, Institute of Basic Medical Sciences, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.X.Y. Yang, MD; Y. Gao, MD, Department of Rheumatology; H.Y. Wang, MD, PhD, Department of Allergy and Clinical Immunology; X.Y. Zhao, MD, Department of Hematology; X.B. Gong, MD, Laboratory of Bone Marrow, Second Affiliated Hospital, College of Medicine, Zhejiang University; Q.Q. Wang, MD, PhD, Department of Immunology, Institute of Basic Medical Sciences; X.F. Zhang, MD, Department of Clinical Epidemiology and Biostatistics, Second Affiliated Hospital, College of Medicine, Zhejiang University
| |
Collapse
|
18
|
Nozaki Y, Kinoshita K, Yano T, Shiga T, Hino S, Niki K, Kishimoto K, Funauchi M, Matsumura I. Estimation of kidney injury molecule-1 (Kim-1) in patients with lupus nephritis. Lupus 2014; 23:769-77. [DOI: 10.1177/0961203314526292] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 02/04/2014] [Indexed: 02/01/2023]
Abstract
Objective Biomarkers of disease activity in lupus nephritis (LN) are needed. Ideally, such biomarkers would be capable of detecting early sub-clinical disease and could be used to gauge response to therapy, thus obviating the need for serial renal biopsies. Much of the focus in the search for LN biomarkers has been on the measurement of urinary chemokines and cytokines in LN patients. However, these have yet to be widely implemented in clinical practice. Kidney injury molecule-1 (Kim-1) is expressed in damaged tubules, but whether urinary (u) and tubular (t)-Kim-1 could serve as a biomarker of active LN is unknown. To investigate the disease activity and histological findings in LN, we evaluated u-Kim-1 levels and t-Kim-1 cells in patients with systemic lupus erythematosus (SLE). Method We measured u-Kim-1 levels and stained t-Kim-1 expression in 57 patients with LN using an ELISA and immunohistochemistry staining. Patients were classified into two groups (active LN, n = 37; inactive LN, n = 20) based on the presence of active renal disease according to the renal SLE disease activity index. correlations of clinical, laboratory data, and histological findings with urinary and t-Kim-1 expression were assessed. Result The u-Kim-1 levels were significantly correlated with the expression of t-Kim-1 ( R = 0.64; P = 0.004) in the SLE patients. The active LN patients exhibited elevated u-Kim-1 levels compared to the inactive LN patients. The number of t-Kim-1 cells was also correlated with histological findings (both glomerular and interstitial inflammation). The u-Kim-1 levels were also correlated with proteinuria and tubular damage in the active LN group. The number of t-Kim-1 cells at baseline was significantly correlated with the estimated glomerular filtration rate ( R = 0.72; P = 0.005) and serum creatinine ( R = 0.53; P = 0.005) after 6–8 months of treatment. Conclusion These data suggest the potential use of the u-Kim-1 levels to screen for active LN and for the estimation of t-Kim-1 expression in renal biopsies to predict renal damage, ongoing glomerular nephritis and tubulointerstitial inflammation, and tubular atrophy.
Collapse
Affiliation(s)
- Y Nozaki
- Department of Hematology and Rheumatology, Kinki University School of Medicine, Osaka, Japan
| | - K Kinoshita
- Department of Hematology and Rheumatology, Kinki University School of Medicine, Osaka, Japan
| | - T Yano
- Department of Hematology and Rheumatology, Kinki University School of Medicine, Osaka, Japan
| | - T Shiga
- Department of Hematology and Rheumatology, Kinki University School of Medicine, Osaka, Japan
| | - S Hino
- Department of Hematology and Rheumatology, Kinki University School of Medicine, Osaka, Japan
| | - K Niki
- Department of Hematology and Rheumatology, Kinki University School of Medicine, Osaka, Japan
| | - K Kishimoto
- Department of Hematology and Rheumatology, Kinki University School of Medicine, Osaka, Japan
| | - M Funauchi
- Department of Hematology and Rheumatology, Kinki University School of Medicine, Osaka, Japan
| | - I Matsumura
- Department of Hematology and Rheumatology, Kinki University School of Medicine, Osaka, Japan
| |
Collapse
|
19
|
Abstract
The last decade has witnessed an explosion in efforts to discover and validate lupus biomarkers. The currently steep trajectory of this progress is unprecedented. However, advances in the lupus biomarker field remain fewer and slower than physicians, patients, and pharmaceutical companies have hoped for. This chapter will review the challenges confronted by physicians and scientists in pursuit of lupus biomarkers and will present our experience on this path and specific efforts to surmount some of the obstacles in this endeavor. A comprehensive review of the current landscape in lupus biomarker research has recently been published elsewhere (Ahearn et al. Transl Res 159:326-342, 2012; Liu et al. Ther Adv Musculoskelet Dis 5:210-233, 2013; Liu and Ahearn Best Pract Res Clin Rheumatol 23:507-523, 2009; Liu et al. Curr Opin Rheumatol 17:543-549, 2005).
Collapse
Affiliation(s)
- Joseph M Ahearn
- Asthma Allergy and Autoimmunity Institute, Pittsburgh, PA, USA
| | | | | |
Collapse
|
20
|
Cottrell I, Khan A, Maqsood S, Thornes J, Eggleton P. Meta-analysis as a diagnostic tool for predicting disease onset and/or activity in systemic lupus erythematosus. Methods Mol Biol 2014; 1134:249-259. [PMID: 24497368 DOI: 10.1007/978-1-4939-0326-9_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Systemic lupus erythematous (SLE) is a relatively rare disorder with prevalence rates between 5 and 50 per 100,000 population. This means performing any epidemiological analysis in a specific research center is difficult, due to the low number of cases within any one location. There is a need for biomarkers and diagnostic aids to monitor SLE disease activity and severity prior, during, and after treatment. Many specialist lupus clinics worldwide have published trials following in detail small numbers of patients that have been monitored for a disease biomarker, e.g., an autoantibody against a self-molecule in prospective and retrospective studies. They have then attempted to correlate autoantibody levels against an autoantigen with disease activity, e.g., nephritis development. The results are often inconclusive with the conclusion "the autoantibody may be useful in monitoring disease activity." Meta-analysis is a statistical technique that can be used for combining the findings of multiple studies to add power to any tentative conclusion proposed by individual studies. Here, we describe a method for analyzing biomarkers of interest as predictors of disease activity, using anti-C1q autoantibodies as an example.
Collapse
|
21
|
Li Y, Fang X, Li QZ. Biomarker profiling for lupus nephritis. GENOMICS PROTEOMICS & BIOINFORMATICS 2013; 11:158-65. [PMID: 23732627 PMCID: PMC4357827 DOI: 10.1016/j.gpb.2013.05.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Revised: 05/26/2013] [Accepted: 05/26/2013] [Indexed: 12/04/2022]
Abstract
Lupus nephritis (LN) is one of the most severe manifestations of systemic lupus erythematosus (SLE), which is associated with significant morbidity and mortality of SLE patients. The pathogenesis of LN involves multiple factors, including genetic predisposition, epigenetic regulation and environmental interaction. Over the last decade, omics-based techniques have been extensively utilized for biomarker screening and a wide variety of variations which are associated with SLE and LN have been identified at the levels of genomics, transcriptomics and proteomics. These studies and discoveries have expanded our understanding of the molecular basis of the disease and are important for identification of potential therapeutic targets for disease prediction and early treatment. In this review, we summarize some of the recent studies targeted at the identification of LN-associated biomarkers using genomics and proteomic approaches.
Collapse
Affiliation(s)
- Yajuan Li
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiangdong Fang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Quan-Zhen Li
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Corresponding author.
| |
Collapse
|
22
|
Meester I, Solis-Soto JM. Cytokines: monitors of disease severity for the clinic. ACTA ACUST UNITED AC 2013; 3:143-55. [PMID: 23485161 DOI: 10.1517/17530050802708999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Cytokines communicate between the cells of the immune system and its targets to maintain homeostasis after injury or pathogenic events. They are involved in almost any pathological situation imaginable. OBJECTIVE To verify the importance of cytokines as biomarkers in current preclinical (aetiopathogenic, development of new therapies) and clinical studies (diagnosis, disease severity, prognosis and response to therapy). METHOD/RESULTS A Medline search with the query 'cytokine' AND 'biomarker' AND a variable for a variety of biomedical fields, followed by deeper-level searches, demonstrated the immense popularity of cytokines as biomarkers in almost any biomedical field. CONCLUSION As cytokines are not disease-specific they do not serve as single diagnostic biomarkers. The strength of the cytokines resides in monitoring disease severity, prognosis and response to treatment.
Collapse
Affiliation(s)
- Irene Meester
- Faculty of Medicine Department of Immunology, UANL, Gonzalitos 235, Mitras Centro, Monterrey, NL, Mexico, CP64460
| | | |
Collapse
|
23
|
Jovanović V, Abdul Aziz N, Lim YT, Ng Ai Poh A, Jin Hui Chan S, Ho Xin Pei E, Lew FC, Shui G, Jenner AM, Bowen L, McKinney EF, Lyons PA, Kemeny MD, Smith KGC, Wenk MR, MacAry PA. Lipid anti-lipid antibody responses correlate with disease activity in systemic lupus erythematosus. PLoS One 2013; 8:e55639. [PMID: 23409013 PMCID: PMC3567138 DOI: 10.1371/journal.pone.0055639] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 12/28/2012] [Indexed: 12/17/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder characterized by broad clinical manifestations including cardiovascular and renal complications with periodic disease flares and significant morbidity and mortality. One of the main contributing factors to the pathology of SLE is the accumulation and impaired clearance of immune complexes of which the principle components are host auto-antigens and antibodies. The contribution of host lipids to the formation of these autoimmune complexes remains poorly defined. The aim of the present study was to identify and analyze candidate lipid autoantigens and their corresponding anti-lipid antibody responses in a well-defined SLE patient cohort using a combination of immunological and biophysical techniques. Disease monitoring in the SLE cohort was undertaken with serial British Isles Lupus Assessment Group (BILAG) scoring. Correlations between specific lipid/anti-lipid responses were investigated as disease activity developed from active flares to quiescent during a follow up period. We report a significant negative correlation between anti-lipid antibodies for 24S-hydroxycholesterol, cardiolipin and phosphatidylserine with SLE disease activity. Taken together, these data suggest that lipid autoantigens represent a new family of biomarkers that can be employed to monitor disease activity plus the efficacy of therapeutic intervention in SLE.
Collapse
Affiliation(s)
- Vojislav Jovanović
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Nurhuda Abdul Aziz
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Yan Ting Lim
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Amanda Ng Ai Poh
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Sherlynn Jin Hui Chan
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Eliza Ho Xin Pei
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Fei Chuin Lew
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Guanghou Shui
- Department of Biochemistry, National University of Singapore, Singapore
| | - Andrew M. Jenner
- School of Biological Sciences, Illawara Health and Medical Research Institute, University of Wollongong, Australia
| | - Li Bowen
- Department of Biochemistry, National University of Singapore, Singapore
| | - Eoin F. McKinney
- Cambridge Institute for Medical Research, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Paul A. Lyons
- Cambridge Institute for Medical Research, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Michael D. Kemeny
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| | - Kenneth G. C. Smith
- Cambridge Institute for Medical Research, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Markus R. Wenk
- Department of Biochemistry, National University of Singapore, Singapore
| | - Paul A. MacAry
- Immunology Programme and Department of Microbiology, National University of Singapore, Singapore
| |
Collapse
|
24
|
Batal I, Liang K, Bastacky S, Kiss LP, McHale T, Wilson NL, Paul B, Lertratanakul A, Ahearn JM, Manzi SM, Kao AH. Prospective assessment of C4d deposits on circulating cells and renal tissues in lupus nephritis: a pilot study. Lupus 2011; 21:13-26. [PMID: 21959138 DOI: 10.1177/0961203311422093] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Complement activation plays a key role in the pathogenesis of lupus nephritis (LN), a severe complication of systemic lupus erythematosus (SLE). We prospectively evaluated 15 LN subjects and two control groups: 13 non-SLE renal subjects (control A) and 239 SLE subjects without LN (control B). All had C4d levels on circulating erythrocytes (E-C4d), reticulocytes (R-C4d) and platelets (P-C4d) measured by flow cytometry, while C4d deposition in renal tissue was semiquantitatively assessed in LN subjects and control A using immunoperoxidase staining. Compared with control A, LN biopsies had higher glomerular-C4d scores (p = 0.003), which were associated with more frequent granular glomerular immunofluorescence staining and electron dense deposits (p < 0.001). Compared with control A and B groups, LN subjects had higher E-C4d (p = 0.002 and p = 0.005) and R-C4d levels (p = 0.002 and p = 0.008), respectively. LN subjects were more likely to have P-C4d compared with control A (p = 0.016). In LN, only E-C4d correlated with National Institutes of Health (NIH) activity index (r = 0.55, p = 0.04). In conclusion, LN biopsies showed frequent glomerular-C4d staining associated with immune complex deposits. LN subjects had higher E-C4d and R-C4d levels compared with both control groups. E-C4d levels also correlated with NIH activity index. These findings suggest a potential role of C4d on circulating cells as a biomarker for lupus nephritis.
Collapse
Affiliation(s)
- I Batal
- Department of Pathology, University of Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Renal involvement in patients with systemic lupus erythematosus in the form of severe lupus nephritis is associated with a significant burden of morbidity and mortality. Conventional laboratory biomarkers in current use have not been very successful in anticipating disease flares, predicting renal histology, or decreasing unwanted outcomes. Since early treatment is associated with improved clinical results, it is thus essential to identify new biomarkers with substantial predictive power to reduce the serious sequelae of this difficult to control lupus manifestation. Indeed, considerable efforts and progress have been made over the last few years in the search for novel biomarkers. Since urinary biomarkers are more easily obtainable with much less risk to the patient than repeat renal biopsies, and these may more accurately discern between renal disease and other organ manifestations than their serum counterparts, there has been tremendous interest in studying new candidate urine biomarkers. Below, we review several promising urinary biomarkers under investigation, including total proteinuria and microalbuminuria, urinary proteomic signatures, and the individual inflammatory mediators interleukin-6, vascular cell adhesion molecule-1, CXCL16, IP-10, and tumor necrosis factor-like weak inducer of apoptosis.
Collapse
Affiliation(s)
- Joyce Reyes-Thomas
- Division of Rheumatology, Albert Einstein College of Medicine, Forchheimer Building, Room 701N, 1300 Morris Park Ave, Bronx, New York, NY 10461, USA
| | | | | |
Collapse
|
26
|
Julkunen H, Ekblom-Kullberg S, Miettinen A. Nonrenal and renal activity of systemic lupus erythematosus: a comparison of two anti-C1q and five anti-dsDNA assays and complement C3 and C4. Rheumatol Int 2011; 32:2445-51. [DOI: 10.1007/s00296-011-1962-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 05/22/2011] [Indexed: 10/18/2022]
|
27
|
Carlsson A, Wuttge DM, Ingvarsson J, Bengtsson AA, Sturfelt G, Borrebaeck CAK, Wingren C. Serum protein profiling of systemic lupus erythematosus and systemic sclerosis using recombinant antibody microarrays. Mol Cell Proteomics 2011; 10:M110.005033. [PMID: 21350050 PMCID: PMC3098590 DOI: 10.1074/mcp.m110.005033] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Systemic lupus erythematosus (SLE) and systemic sclerosis (SSc) are two severe autoimmune connective tissue diseases. The fundamental knowledge about their etiology is limited and the conditions display complex pathogenesis, multifaceted presentations, and unpredictable courses. Despite significant efforts, the lack of fully validated biomarkers enabling diagnosis, classification, and monitoring of disease activity represents significant unmet clinical needs. In this discovery study, we have for the first time used recombinant antibody microarrays for miniaturized, multiplexed serum protein profiling of SLE and SSc, targeting mainly immunoregulatory proteins. The data showed that several candidate SLE-associated multiplexed serum biomarker signatures were delineated, reflecting disease (diagnosis), disease severity (phenotypic subsets), and disease activity. Selected differentially expressed markers were validated using orthogonal assays and a second, independent patient cohort. Further, biomarker signatures differentiating SLE versus SSc were demonstrated, and the observed differences increased with severity of SLE. In contrast, the data showed that the serum profiles of SSc versus healthy controls were more similar. Hence, we have shown that affinity proteomics could be used to de-convolute crude, nonfractionated serum proteomes, extracting molecular portraits of SLE and SSc, further enhancing our fundamental understanding of these complex autoimmune conditions.
Collapse
Affiliation(s)
- Anders Carlsson
- Department of Immunotechnology, BMC D13, Lund University, SE-221 84 Lund, Sweden
| | | | | | | | | | | | | |
Collapse
|
28
|
WANG GANG, TAM LAISHAN, LI EDMUNDKWOKMING, KWAN BONNIECHINGHA, CHOW KAIMING, LUK CATHYCHOIWAN, LI PHILIPKAMTAO, SZETO CHEUKCHUN. Serum and Urinary Cell–free MiR-146a and MiR-155 in Patients with Systemic Lupus Erythematosus. J Rheumatol 2010; 37:2516-22. [DOI: 10.3899/jrheum.100308] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective.Recent studies showed that micro-RNA play important roles in the pathogenesis of autoimmune diseases. We studied the levels of miR-146a and miR-155 in the serum and urinary supernatant of patients with systemic lupus erythematosus (SLE).Methods.The serum and urinary supernatant levels of miR-146a and miR-155 were determined by real-time quantitative polymerase chain reaction in 40 patients with SLE and 30 healthy controls.Results.Compared to controls, serum miR-146a and miR-155 levels were lower, and the urinary level of miR-146a was higher, in SLE. Estimated glomerular filtration rate (eGFR) correlated with both serum miR-146a (r = 0.519, p = 0.001) and miR-155 (r = 0.384, p = 0.014). Serum miR-146a inversely correlated with proteinuria (r = −0.341, p = 0.031) and the SLE Disease Activity Index (r = −0.465, p = 0.003). Serum miR-146a and miR-155 levels also correlated with red blood cell count, platelet count, and lymphocyte count. After treatment with calcitriol for 6 months, serum miR-146a level of SLE patients increased significantly (p < 0.001), and its change inversely correlated with the level of calcium-phosphate product (r = −0.466, p = 0.003).Conclusion.The results suggested that serum miR-146a and miR-155 participate in the pathophysiology of SLE and might be used as biomarkers of SLE.
Collapse
|
29
|
Abstract
Biomarkers have the potential to be useful tools for noninvasively evaluating and managing patients with lupus nephritis. Many candidate biomarkers have been identified, but they require validation in larger cohorts. It is likely that combinations or biomarker profiles, rather than individual markers, will emerge to help better predict the severity of inflammation, the extent of fibrosis, degree of drug responsiveness, and other variables. This approach has the potential to reduce the use of the renal biopsy, improve therapeutic efficacy, and limit toxicity. We predict algorithms based on genotype and biomarkers combined with clinical presentation will emerge to help guide physicians in management. Assays that show the most potential include serum erythrocyte bound complement C4d, interleukin 17, interleukin 23, interferon score/chemokine score ratio, and anti-C1q antibodies. Such urinary biomarkers as fractional excretion of endothelial-1, monocyte chemoattractant protein-1, vascular cell adhesion molecule-1, and TWEAK (tumor necrosis factor-like weak inducer of apoptosis) may also be useful but require validations.
Collapse
Affiliation(s)
- Anup Manoharan
- Department of Medicine, Nephrology and Kidney Transplantation Section, Medical College of Georgia, 1120 15th Street, BA 9413, Augusta, GA 30912-3140, USA.
| | | |
Collapse
|
30
|
Gaffney PM, Moser KL, Baechler EC. Defining a new molecular basis of systemic lupus erythematosus through transcriptional profiling. Expert Rev Clin Immunol 2010; 3:913-23. [PMID: 20477140 DOI: 10.1586/1744666x.3.6.913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Data generated using high-throughput DNA microarrays are changing the way we think about systemic lupus erythematosus (SLE). The identification of an interferon gene-expression signature in the majority of patients with SLE, especially those with severe SLE, has stimulated substantial interest in targeting the interferon pathway for the treatment SLE and has catalyzed new inquiries into the utility of interferon signaling as a diagnostic and prognostic biomarker for SLE. As these genomic datasets enlarge and mature, new signatures are being identified that implicate other pathways dysregulated in SLE, including oxidative phosphorylation, immunoglobulin production and granulocyte maturation. Highly anticipated longitudinal studies will be important in defining how this information will ultimately change the way SLE is managed in the clinical setting.
Collapse
Affiliation(s)
- Patrick M Gaffney
- Oklahoma Medical Research Foundation, Arthritis and Immunology Program, 825 N.E. 13th Street, Oklahoma City, OK 73104, USA.
| | | | | |
Collapse
|
31
|
Rubinstein T, Pitashny M, Levine B, Schwartz N, Schwartzman J, Weinstein E, Pego-Reigosa JM, Lu TYT, Isenberg D, Rahman A, Putterman C. Urinary neutrophil gelatinase-associated lipocalin as a novel biomarker for disease activity in lupus nephritis. Rheumatology (Oxford) 2010; 49:960-71. [PMID: 20144927 DOI: 10.1093/rheumatology/kep468] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES Clinical and laboratory markers in current use have limited specificity and sensitivity for predicting the development of renal disease in lupus patients. In this longitudinal study, we investigated whether urinary neutrophil gelatinase-associated lipocalin (uNGAL) predicts active nephritis and renal flares in lupus patients with and without a history of biopsy-proven lupus nephritis. METHODS Renal disease activity and flare status was determined by SLEDAI and BILAG scores. Random effects models were used to determine whether uNGAL was a significant predictor for renal disease activity in SLE patients, and for renal flares in patients with established nephritis. To assess the predictive performance of uNGAL, receiver operating characteristic (ROC) curves were constructed using the previous visit's uNGAL level. These curves were then compared with curves constructed with currently used biomarkers. Cut-offs determined by ROC curves were tested in an independent validation cohort. RESULTS uNGAL was found to be a significant predictor of renal disease activity in all SLE patients, and a significant predictor for flare in patients with a history of biopsy-proven nephritis, in multivariate models adjusting for age, race, sex and anti-double-stranded (ds)DNA antibody titres. As a predictor of renal flare in patients with biopsy-proven nephritis, uNGAL outperformed anti-dsDNA antibody titres. These results were confirmed in an independent validation cohort. CONCLUSIONS uNGAL predicts renal flare in patients with a history of biopsy-proven nephritis with high sensitivity and specificity. Furthermore, uNGAL is a more sensitive and specific forecaster of renal flare in patients with a history of lupus nephritis than anti-dsDNA antibody titres.
Collapse
Affiliation(s)
- Tamar Rubinstein
- Department of Medicine, Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Bauer JW, Petri M, Batliwalla FM, Koeuth T, Wilson J, Slattery C, Panoskaltsis-Mortari A, Gregersen PK, Behrens TW, Baechler EC. Interferon-regulated chemokines as biomarkers of systemic lupus erythematosus disease activity: a validation study. ACTA ACUST UNITED AC 2009; 60:3098-107. [PMID: 19790071 DOI: 10.1002/art.24803] [Citation(s) in RCA: 228] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by unpredictable flares of disease activity and irreversible damage to multiple organ systems. An earlier study showed that SLE patients carrying an interferon (IFN) gene expression signature in blood have elevated serum levels of IFN-regulated chemokines. These chemokines were associated with more-severe and active disease and showed promise as SLE disease activity biomarkers. This study was designed to validate IFN-regulated chemokines as biomarkers of SLE disease activity in 267 SLE patients followed up longitudinally. METHODS To validate the potential utility of serum chemokine levels as biomarkers of disease activity, we measured serum levels of CXCL10 (IFNgamma-inducible 10-kd protein), CCL2 (monocyte chemotactic protein 1), and CCL19 (macrophage inflammatory protein 3beta) in an independent cohort of 267 SLE patients followed up longitudinally over 1 year (1,166 total clinic visits). RESULTS Serum chemokine levels correlated with lupus activity at the current visit (P = 2 x 10(-10)), rising at the time of SLE flare (P = 2 x 10(-3)) and decreasing as disease remitted (P = 1 x 10(-3)); they also performed better than the currently available laboratory tests. Chemokine levels measured at a single baseline visit in patients with a Systemic Lupus Erythematosus Disease Activity Index of < or =4 were predictive of lupus flare over the ensuing year (P = 1 x 10(-4)). CONCLUSION Monitoring serum chemokine levels in SLE may improve the assessment of current disease activity, the prediction of future disease flares, and the overall clinical decision-making.
Collapse
|
33
|
Smith MF, Hiepe F, Dörner T, Burmester G. Biomarkers as tools for improved diagnostic and therapeutic monitoring in systemic lupus erythematosis. Arthritis Res Ther 2009; 11:255. [PMID: 19939293 PMCID: PMC3003542 DOI: 10.1186/ar2834] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
One of the major challenges in rheumatology is to overcome the classification criteria that previously defined systemic lupus erythematosis, since the heterogeneity of the disease(s) appears to represent a complexity that probably substantially contributed to the failure of a number of recent trials. For those engaged in clinical trials, validated disease activity biomarkers that respond rapidly to treatment and are predictive of clinical response would greatly facilitate early decision-making around futility and dose selection. Likewise, use of validated patient stratification biomarkers possibly in conjunction with autoantibody profiles and disease manifestations will result in the recruitment of more homogeneous patient populations during later stage clinical studies, thereby decreasing size, costs, and risks in pivotal studies.
Collapse
Affiliation(s)
- Michael F Smith
- Wyeth Research, Discovery Translational Medicine, Collegeville, PA 19426, USA.
| | | | | | | |
Collapse
|
34
|
Lipocalin 2 regulation by thermal stresses: Protective role of Lcn2/NGAL against cold and heat stresses. Exp Cell Res 2009; 315:3140-51. [DOI: 10.1016/j.yexcr.2009.08.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2009] [Revised: 08/22/2009] [Accepted: 08/25/2009] [Indexed: 11/20/2022]
|
35
|
Sandrin-Garcia P, Junta CM, Fachin AL, Mello SS, Baião AMT, Rassi DM, Ferreira MCT, Trevisan GL, Sakamoto-Hojo ET, Louzada-Júnior P, Passos GAS, Donadi EA. Shared and unique gene expression in systemic lupus erythematosus depending on disease activity. Ann N Y Acad Sci 2009; 1173:493-500. [PMID: 19758191 DOI: 10.1111/j.1749-6632.2009.04636.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Patients presenting with active systemic lupus erythematosus (SLE) manifestations may exhibit distinct pathogenetic features in relation to inactive SLE. Also, cDNA microarrays may potentially discriminate the gene expression profile of a disease or disease variant. Therefore, we evaluated the expression profile of 4500 genes in peripheral blood lymphocytes (PBL) of SLE patients. We studied 11 patients with SLE (seven with active SLE and four with inactive SLE) and eight healthy controls. Total RNA was isolated from PBL, reverse transcribed into cDNA, and postlabeled with Cy3 fluorochrome. These probes were then hybridized to a glass slide cDNA microarray containing 4500 human IMAGE cDNA target sequences. An equimolar amount of total RNA from human cell lines served as reference. The microarray images were quantified, normalized, and analyzed using the R environment (ANOVA, significant analysis of microarrays, and cluster-tree view algorithms). Disease activity was assessed by the SLE disease activity index. Compared to the healthy controls, 104 genes in active SLE patients (80 repressed and 24 induced) and 52 genes in nonactive SLE patients (31 induced and 21 repressed) were differentially expressed. The modulation of 12 genes, either induced or repressed, was found in both disease variants; however, each disease variant had differential expression of different genes. Taken together, these results indicate that the two lupus variants studied have common and unique differentially expressed genes. Although the biological significance of the differentially expressed genes discussed above has not been completely understood, they may serve as a platform to further explore the molecular basis of immune deregulation in SLE.
Collapse
Affiliation(s)
- Paula Sandrin-Garcia
- Molecular Immunogenetics Group (Department of Genetics), Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Landolt-Marticorena C, Bonventi G, Lubovich A, Ferguson C, Unnithan T, Su J, Gladman DD, Urowitz M, Fortin PR, Wither J. Lack of association between the interferon-alpha signature and longitudinal changes in disease activity in systemic lupus erythematosus. Ann Rheum Dis 2009; 68:1440-6. [PMID: 18772188 DOI: 10.1136/ard.2008.093146] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To study the longitudinal expression of interferon (IFN)-inducible genes in systemic lupus erythematosus (SLE) and determine their suitability as disease biomarkers. METHODS RNA was isolated from the peripheral blood of 94 patients with SLE and 11 controls and reverse transcribed into cDNA. The expression levels of five IFN-responsive genes (LY6E, OAS1, IFIT1, ISG15 and MX1) were determined by quantitative PCR, normalised to GAPDH and summed to generate a global IFN score. Patients were followed longitudinally for a period of 3-12 months, and the association between disease activity, as measured by the SLE disease activity index (SLEDAI-2K), and other clinical and laboratory variables was examined. RESULTS The expression of all five IFN-responsive genes was significantly higher in patients with SLE than in controls. The expression of LY6E, OAS1, IFIT1 and the global IFN score was associated with high disease activity. The global IFN score was also associated with active renal disease, a decreased C3, and the presence of anti-dsDNA or anti-RNA binding protein antibodies at a single point in time. However, there was a poor correlation between changes in this score and changes in disease activity, C3 or anti-dsDNA antibody levels in patients followed longitudinally. In most patients the levels of IFN-induced gene expression remained relatively stable over 3-12 months despite marked changes in disease activity. Nevertheless, in patients with low/moderate disease activity, those with high IFN scores had a more recent history of sustained high disease activity. CONCLUSION The findings indicate that IFN-induced gene expression has limited clinical utility as a biomarker of acute changes in disease activity.
Collapse
Affiliation(s)
- C Landolt-Marticorena
- Arthritis Centre of Excellence, Division of Genetics and Development, Toronto Western Hospital Research Institute, University Health Network, Toronto, Ontario M5T 2S8, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Yang DH, Chang DM, Lai JH, Lin FH, Chen CH. Usefulness of erythrocyte-bound C4d as a biomarker to predict disease activity in patients with systemic lupus erythematosus. Rheumatology (Oxford) 2009; 48:1083-7. [DOI: 10.1093/rheumatology/kep161] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
38
|
Suh CH, Kim HA. Cytokines and their receptors as biomarkers of systemic lupus erythematosus. Expert Rev Mol Diagn 2008; 8:189-98. [PMID: 18366305 DOI: 10.1586/14737159.8.2.189] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Systemic lupus erythematosus is the most clinically diverse autoimmune disease. Owing to its heterogeneous presentation, clinical management of systemic lupus erythematosus remains as one of the greatest challenges. Therefore, there is a great need to assess disease activity accurately. Biomarkers can be objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes or pharmacologic responses to a therapeutic intervention, and may also predict the risk of the disease, confirm diagnosis, monitor disease activity and provide prognostic information. Cytokines play an important and diverse role in the immune dysregulation in systemic lupus erythematosus. Measuring serum levels of soluble IL-2 receptor, IL-6, IL-10, soluble TNF receptor and IFN-alpha/IFN-induced genes may be promising biomarkers of disease activity in systemic lupus erythematosus.
Collapse
Affiliation(s)
- Chang-Hee Suh
- Department of Allergy and Rheumatology, Ajou University School of Medicine, Woncheon-dong, San5, Youngtong-gu, Suwon, 443-721 South Korea.
| | | |
Collapse
|
39
|
Magalhães MB, da Silva LM, Voltarelli JC, Donadi EA, Louzada-Junior P. Lymphocytotoxic antibodies in systemic lupus erythematosus are associated with disease activity irrespective of the presence of neuropsychiatric manifestations. Scand J Rheumatol 2008; 36:442-7. [PMID: 18092265 DOI: 10.1080/03009740701482768] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVES To evaluate the association of the presence of lymphocytotoxic, anti-beta2-glycoprotein I (anti-beta2-GPI) and anti-ribosomal P (anti-P) antibodies in patients with systemic lupus erythematosus (SLE), presenting or not neuropsychiatric (NP) manifestations, stratified according to the activity of the disease. METHODS A total of 138 patients with SLE (59 with active NPSLE, 49 with active non-NPSLE, and 30 with inactive disease) and 57 healthy controls were studied. Disease activity was assessed by the SLE Disease Activity Index (SLEDAI). The presence of lymphocytotoxic antibodies was assessed using a complement-dependent lymphocytotoxicity assay. The presence of anti-beta2-GPI and anti-P antibodies was detected by enzyme-linked immunosorbent assay (ELISA). RESULTS Lymphocytotoxic antibodies were detected primarily in patients with active disease, that is in 35 out of 59 (59.3%) NPSLE and 23 out of 49 (46.9%) non-NPSLE patients, whereas only four out of 30 (13.3%) inactive SLE patients and none of the healthy controls exhibited the autoantibody. The frequency of lymphocytotoxic antibodies in active SLE patients, considered as a whole or stratified into NPSLE or non-NPSLE, was significantly increased in relation to inactive SLE patients (p<0.001 for each comparison). No significant difference was observed when comparing active NPSLE with non-NPSLE patients. No associations were observed between the presence of anti-beta2-GPI or anti-P antibodies and the activity of SLE or the presence of lymphocytotoxic antibodies. CONCLUSIONS Lymphocytotoxic antibodies occurred more frequently in patients with active SLE than in patients with inactive disease, irrespective of the presence of NP manifestations, a finding that is similar to classical biomarkers of lupus activity (anti-dsDNA and complement). These results indicate that the assessment of the presence of lymphocytotoxic antibodies may be an additional useful tool for the evaluation of SLE activity.
Collapse
Affiliation(s)
- M B Magalhães
- Division of Clinical Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | | | | | | | | |
Collapse
|
40
|
Rubinstein T, Pitashny M, Putterman C. The novel role of neutrophil gelatinase-B associated lipocalin (NGAL)/Lipocalin-2 as a biomarker for lupus nephritis. Autoimmun Rev 2008; 7:229-34. [DOI: 10.1016/j.autrev.2007.11.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Wu YL, Savelli SL, Yang Y, Zhou B, Rovin BH, Birmingham DJ, Nagaraja HN, Hebert LA, Yu CY. Sensitive and specific real-time polymerase chain reaction assays to accurately determine copy number variations (CNVs) of human complement C4A, C4B, C4-long, C4-short, and RCCX modules: elucidation of C4 CNVs in 50 consanguineous subjects with defined HLA genotypes. THE JOURNAL OF IMMUNOLOGY 2007; 179:3012-25. [PMID: 17709516 DOI: 10.4049/jimmunol.179.5.3012] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent comparative genome hybridization studies revealed that hundreds to thousands of human genomic loci can have interindividual copy number variations (CNVs). One of such CNV loci in the HLA codes for the immune effector protein complement component C4. Sensitive, specific, and accurate assays to interrogate the C4 CNV and its associated polymorphisms by using submicrogram quantities of genomic DNA are needed for high throughput epidemiologic studies of C4 CNVs in autoimmune, infectious, and neurological diseases. Quantitative real-time PCR (qPCR) assays were developed using TaqMan chemistry and based on sequences specific for C4A and C4B genes, structural characteristics corresponding to the long and short forms of C4 genes, and the breakpoint region of RP-C4-CYP21-TNX (RCCX) modular duplication. Assignments for gene copy numbers were achieved by relative standard curve methods using cloned C4 genomic DNA covering 6 logs of DNA concentrations for calibrations. The accuracies of test results were cross-confirmed internally in each sample, as the sum of C4A plus C4B equals to the sum of C4L plus C4S or the total copy number of RCCX modules. These qPCR assays were applied to determine C4 CNVs from samples of 50 consanguineous subjects who were mostly homozygous in HLA genotypes. The results revealed eight HLA haplotypes with single C4 genes in monomodular RCCX that are associated with multiple autoimmune and infectious diseases and 32 bimodular, 4 trimodular, and one quadrimodular RCCX. These C4 qPCR assays are proven to be robust, sensitive, and reliable, as they have contributed to the elucidation of C4 CNVs in >1000 human samples with autoimmune and neurological diseases.
Collapse
Affiliation(s)
- Yee Ling Wu
- Center for Molecular and Human Genetics, Columbus Children's Research Institute, 700 Children's Drive, Columbus, OH 43205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Pitashny M, Schwartz N, Qing X, Hojaili B, Aranow C, Mackay M, Putterman C. Urinary lipocalin-2 is associated with renal disease activity in human lupus nephritis. ACTA ACUST UNITED AC 2007; 56:1894-903. [PMID: 17530720 DOI: 10.1002/art.22594] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Pathogenic monoclonal anti-double-stranded DNA (anti-dsDNA) antibodies up-regulate the expression of lipocalin-2 in glomerular mesangial cells. This study was undertaken to investigate whether polyclonal anti-dsDNA antibodies promote the local secretion of lipocalin-2 in the kidneys of patients with systemic lupus erythematosus (SLE), and whether urinary lipocalin-2 represents a marker of kidney involvement in SLE. METHODS Hispanic, African American, and white patients with SLE and normal healthy control subjects from affiliated hospitals of the Albert Einstein College of Medicine were recruited for this cross-sectional study. Patients were classified based on the presence of active renal disease according to the SLE Disease Activity Index (SLEDAI). Correlations of clinical and laboratory data with urinary and serum levels of lipocalin-2 were assessed. RESULTS Among SLE patients, urinary lipocalin-2 levels were significantly higher in those with lupus nephritis (LN) (median 17.1 ng/mg creatinine, interquartile range [IQR] 10.3-45.4; n = 32) than in those without LN (median 11.2 ng/mg creatinine, IQR 3.1-20.3; n = 38) (P = 0.023). Compared with the values in normal controls (median 4 ng/ml, IQR 0-11.1; n = 14), urinary levels of lipocalin-2 in SLE patients were significantly higher (non-normalized median 19.3 ng/ml, IQR 8-34.2) (P = 0.004). The presence of lipocalin-2 in the urine of patients with LN correlated significantly with the renal SLEDAI score (r = 0.452, P = 0.009), but not with extrarenal disease activity. CONCLUSION The high prevalence of LN in SLE patients and the prognostic significance of kidney disease support the need for identifying early biomarkers to assess the risk of nephritis development and for following up patients with established disease. These findings indicate that urinary lipocalin-2 is a potential marker of the presence and severity of renal involvement in adult patients with SLE.
Collapse
Affiliation(s)
- Milena Pitashny
- Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Chun HY, Chung JW, Kim HA, Yun JM, Jeon JY, Ye YM, Kim SH, Park HS, Suh CH. Cytokine IL-6 and IL-10 as biomarkers in systemic lupus erythematosus. J Clin Immunol 2007; 27:461-6. [PMID: 17587156 DOI: 10.1007/s10875-007-9104-0] [Citation(s) in RCA: 286] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Accepted: 05/04/2007] [Indexed: 11/28/2022]
Abstract
There is a great deal of interest in the identification of biomarkers that are closely associated with disease activity in systemic lupus erythematosus (SLE), but few biomarkers have been validated. Cytokines play an important role in the pathogenesis of SLE. Therefore, we evaluated the levels of cytokines and their possible association with disease activity. In the present study, we found that the SLE patients had higher IL-6, IL-10, IL-12, and IFN-gamma levels, but lower IL-2, than normal controls. Serum IL-6 level was significantly elevated in active SLE patients and correlated with the SLE activity index (SLEDAI), erythrocyte sedimentation rate (ESR), and C-reactive protein (CRP). Serum IL-10 level was also significantly elevated in active SLE patients and revealed positive correlation with SLEDAI and anti-double-stranded DNA (dsDNA) titer and negative correlation with C3, C4, and lymphocyte counts. No significant differences in the levels of cytokines were observed between SLE patients with nephritis and those without nephritis. These data suggest that IL-6 and IL-10 may be a useful biomarker for disease activity in SLE.
Collapse
Affiliation(s)
- Hye-Young Chun
- Department of Allergy and Rheumatology, Ajou University School of Medicine, Woncheon-dong, San5, Youngtong-gu, Suwon 443-721, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Schwartz N, Su L, Burkly LC, Mackay M, Aranow C, Kollaros M, Michaelson JS, Rovin B, Putterman C. Urinary TWEAK and the activity of lupus nephritis. J Autoimmun 2007; 27:242-50. [PMID: 17257812 DOI: 10.1016/j.jaut.2006.12.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2006] [Revised: 12/07/2006] [Accepted: 12/07/2006] [Indexed: 10/23/2022]
Abstract
The TNF superfamily cytokine TWEAK induces mesangial cells, podocytes, and endothelial cells to secrete pro-inflammatory chemokines including MCP-1, IP-10 and RANTES, which are crucial in the pathogenesis of lupus nephritis (LN). As TWEAK regulates the secretion of these inflammatory mediators, we studied whether urinary TWEAK (uTWEAK) levels might be predictive and/or diagnostic in LN. In a cross-sectional study of a large, multi-center cohort of systemic lupus erythematosus (SLE) patients, uTWEAK levels were higher in patients with active as compared to never or non-active nephritis (median (IQR): 16.3 (9.9-23.0) versus 5.5 (2.3-16.8) pg/mg creatinine, p=0.001), and levels of uTWEAK correlated with the renal SLE disease activity index (rSLEDAI) score (r=0.405, p<0.001). uTWEAK levels were higher in patients undergoing a flare as compared to patients with chronic stable disease (11.1 (8.1-18.2) and 5.2 (2.3-15.3) pg/mg creatinine, respectively; p=0.036). Moreover, uTWEAK levels were significantly higher in patients undergoing a renal flare, as opposed to a non-renal flare (12.4 (9.1-18.2) and 5.2 (3.0-11.9) pg/mg creatinine, respectively; p=0.029). An accurate, non-invasive method to repeatedly assess kidney disease in lupus would be very helpful in managing these often challenging patients. Our study indicates that urinary TWEAK levels may be useful as a novel biomarker in LN.
Collapse
Affiliation(s)
- Noa Schwartz
- Division of Rheumatology, Forchheimer 701N, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Browning JL. B cells move to centre stage: novel opportunities for autoimmune disease treatment. Nat Rev Drug Discov 2006; 5:564-76. [PMID: 16816838 DOI: 10.1038/nrd2085] [Citation(s) in RCA: 205] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The B-cell arm of the immune system has long been appreciated for its crucial role in pathogen resistance, but in the study of many autoimmune diseases, T cells have dominated the limelight for decades. However, the development of the B-cell-depleting antibody rituximab as a lymphoma therapy has provided a tool to probe the contribution made by B cells in several immune disorders. Recently, the success of B-cell depletion with rituximab in the treatment of rheumatoid arthritis has stimulated investigation of its effects in several other immune disorders, and considerable interest in the potential of drugs that can modulate B-cell function for the treatment of such diseases in general. This article discusses the role of B cells in a range of autoimmune disorders, including rheumatoid arthritis and systemic lupus erythematosus, and analyses approaches to therapeutic B-cell manipulation.
Collapse
Affiliation(s)
- Jeffrey L Browning
- Department of Immunobiology, Biogen Idec, 12 Cambridge Center, Cambridge, Massachusetts 02445, USA.
| |
Collapse
|
46
|
Abstract
Susceptibility to the autoimmune phenotype of systemic lupus erythematosus (SLE) is heritable. Linkage analysis and recent advances in the field of single nucleotide polymorphisms (SNPs) have resulted in the identification of several genetic loci and functional allelic variants of signaling proteins which have become the mainstay of understanding disease susceptibility and exploring the basis of autoimmunity in SLE. However, genetic heterogeneity and possible epistatic interactions among genetic elements have precluded replication of these findings in multiple population groups and thus complicated their interpretation. In this regard, the discovery that a plethora of normal signaling proteins are expressed in abnormal amounts in immune cells from patients with SLE has gained significance. Thus, the key to precise elucidation of the pathologic basis of autoimmunity in SLE lies in tying genetics and disease biology. This review highlights recent discoveries of important functional genetic variants and altered expression of normal signaling proteins that network together to disrupt peripheral tolerance and initiate the autoimmune process in SLE.
Collapse
Affiliation(s)
- Sandeep Krishnan
- Department of Cellular Injury, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| | | | | |
Collapse
|