1
|
Nasser M, Wadie M, Farid A, Amir AE. The effect of pro-inflammatory cytokines on the development of atherosclerosis in systemic lupus erythematosus patients: ultrasonographic assessment of intimal medial thickness and resistive index. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2025; 52:1. [DOI: 10.1186/s43166-024-00298-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 12/16/2024] [Indexed: 01/12/2025] Open
Abstract
Abstract
Background
Systemic lupus erythematosus (SLE) is a debilitating rheumatic condition that results in the dysfunction of multiple organs in the body. Atherosclerosis is a common occurrence in people with SLE and is exacerbated by an overabundant of various cytokines, such as tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6), and their soluble receptors, such as soluble tumor necrosis factor receptor I (sTNFR I) and soluble interleukin-6 receptor (sIL-6R) (known as SLE risk factors) and high concentrations of lipids (known as dyslipidemia risk factors). The intimal medial thickness (IMT) of the internal carotid artery (ICA) is a sonographic measurement that assesses the extent of atherosclerosis. On the other hand, the resistive index (RI) is primarily employed to evaluate the severity of chronic kidney diseases (CKDs), but it is seldom utilized to estimate atherosclerosis in ICA. So, the objective of this study was to conduct retrospective cohort study among SLE patients to define which risk factor, inflammation or dyslipidemia, correlated with the development of atherosclerosis in SLE and which ultrasound assessment, CIMT or RI, is more useful in identifying atherosclerosis.
Results
TNF-α, sTNFR I, IL-6, and sIL-6R were significantly elevated (P < 0.0001) in SLE patients (n = 75) compared to the controls (n = 15); also, both CIMT and RI showed significant higher levels in patient’s group (P = 0.001 and 0.0025, respectively). Systemic lupus disease activity index (SLEDAI) (P = 0.002), total cholesterol (TC) (P = 0.025), CIMT (P = 0.00045), TNF-α (P < 0.0001), IL-6 (P < 0.0001), sTNFR I (P = 0.006), and sIL-6R (P < 0.0001) rates were significantly higher in atherosclerotic SLE patients (n = 27) than in non-atherosclerotic patients (n = 48). There were clear and meaningful positive correlations (r = 0.82, P = 0.003) observed between CIMT and SLEDAI, as well as between investigated cytokines and their soluble receptors. RI showed no significant differences between two studied groups of patients and also no significant correlations with the studied parameters except with age (r = 0.45, P = 0.035).
Conclusion
Inflammation is a more prevalent cause of atherosclerosis than dyslipidemia in SLE, thereby making it a recognized risk factor for SLE. In comparison to RI, CIMT is a valuable sonographic measure for identifying incidence of atherosclerosis.
Collapse
|
2
|
Wu D, Li X, Guo T, Feng X, Li X, Wang Y, Chen W. Unmasking the Hidden Threat: The Role of Left Ventricular Subendocardial Involvement in Autoimmune Rheumatic Disease. Clin Cardiol 2025; 48:e70069. [PMID: 39743740 PMCID: PMC11693844 DOI: 10.1002/clc.70069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 11/13/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Late gadolinium enhancement (LGE) has been found in patients with autoimmune rheumatic disease (ARD). However, the prognostic implications of some specific LGE patterns in ARD patients remain unclear. PURPOSE To investigate the prevalence and prognostic significance of left ventricular (LV) subendocardium-involved LGE (LGEse) in a cohort of ARD patients. MATERIALS AND METHODS This retrospective study evaluated 176 patients diagnosed with ARD with clinically suspected cardiac involvement between 2018 and 2023. LV LGEse was defined as LGE involving the LV subendocardium that did not correspond to a coronary vascular distribution. The endpoints included a composite of cardiac death, heart failure-related admission, cardiogenic shock, and appropriate pacemaker or implantable cardioverter-defibrillator therapy. RESULTS Of the 176 consecutive patients, LV LGEse was observed in 22 patients (13%). During a median follow-up of 776 days (interquartile range, 395-1405 days), 20 patients (11%) experienced a composite endpoint. Compared with those without LV LGEse, the LV LGEse group had a greater proportion of men (64% vs. 14%; p < 0.001), lower LV ejection fraction (50% vs. 60%; p = 0.001), greater LV end-diastolic volume index (78 vs. 75; p = 0.043), and more adverse outcomes (32% vs. 8%; p = 0.005). In the univariable and multivariable Cox regression analyses, the LV LGEse showed independent prognostic value. In the sensitivity analyses, the prognostic difference in terms of LV subendocardial involvement remained. CONCLUSION In our cohort, LV subendocardial involvement, an underrecognized LGE pattern, was observed in 13% of all patients with autoimmune disease and indicated a worse prognosis.
Collapse
Affiliation(s)
- Danni Wu
- Department of CardiologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Xiao Li
- Department of RadiologyPeking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical CollegeBeijingChina
| | - Tianchen Guo
- Department of CardiologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Xiaojin Feng
- Department of CardiologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Xinhao Li
- Department of CardiologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Yining Wang
- Department of RadiologyPeking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical CollegeBeijingChina
| | - Wei Chen
- Department of CardiologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| |
Collapse
|
3
|
Richter P, Cardoneanu A, Dima N, Bratoiu I, Rezus C, Burlui AM, Costin D, Macovei LA, Rezus E. Interstitial Lung Disease in Systemic Lupus Erythematosus and Systemic Sclerosis: How Can We Manage the Challenge? Int J Mol Sci 2023; 24:9388. [PMID: 37298342 PMCID: PMC10253395 DOI: 10.3390/ijms24119388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Interstitial lung disease (ILD) is a severe and frequent manifestation of connective tissue diseases (CTD). Due to its debilitating potential, it requires serious evaluation and treatment. The prevalence of ILD in systemic lupus erythematosus (SLE) is still controversial. Therefore, in order to establish the diagnosis of ILD, an overlap syndrome must be excluded. Increasing the identification of SLE-associated ILD cases should become a target. To treat this complication, various therapies are now being proposed. To date, no placebo-controlled studies were conducted. Regarding another CTD, systemic sclerosis (SSc), SSc-associated ILD is considered one of the leading causes of mortality. The incidence of ILD varies among disease subtypes, being influenced by diagnostic method, but also by disease duration. Due to the high prevalence of this complication, all SSc patients should be investigated for ILD at the time of SSc diagnosis and during the course of the disease. Fortunately, progress was made in terms of treatment. Nintedanib, a tyrosine kinases inhibitor, showed promising results. It appeared to decrease the rate of progression of ILD compared to placebo. This review aimed to provide up-to-date findings related to SLE-associated ILD and SSc-associated ILD, in order to raise awareness of their diagnosis and management.
Collapse
Affiliation(s)
- Patricia Richter
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Anca Cardoneanu
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Nicoleta Dima
- Department of Internal Medicine, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- “Sf. Spiridon” Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Ioana Bratoiu
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- “Sf. Spiridon” Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Alexandra Maria Burlui
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Damiana Costin
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Luana Andreea Macovei
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| |
Collapse
|
4
|
Zheng F, Tan L, Zhang F, Li S, Lai Z, Xu H, Xiong Z, Dai Y. The circRNA-miRNA-mRNA regulatory network in plasma and peripheral blood mononuclear cells and the potential associations with the pathogenesis of systemic lupus erythematosus. Clin Rheumatol 2023:10.1007/s10067-023-06560-5. [PMID: 36862342 DOI: 10.1007/s10067-023-06560-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023]
Abstract
OBJECTIVES This study aimed to explore the possible role of plasma and peripheral blood mononuclear cells (PBMCs) circular RNA (circRNA) in systemic lupus erythematosus (SLE). METHOD Total RNA was extracted from blood plasma samples obtained from 10 patients with SLE and 10 healthy controls and subjected to microarray analysis to define the profile of circRNA expression. The quantitative reverse transcription-polymerase chain reaction (qRT-PCR) amplification was conducted. The overlapped circRNA between PBMCs and plasma was performed, the interactions with microRNAs were predicted, the miRNA target mRNA was predicted, and the GEO database was used. The Gene ontology and pathway analysis was performed. RESULTS One hundred thirty-one upregulated and 314 significantly downregulated circRNAs were identified in the plasma of patients with SLE by the Fold change criteria (≥ 2.0) and P < 0.05. The qRT-PCR results showed that the expression of has-circRNA-102531, has-circRNA-103984, and has-circRNA-104262 was increased in plasma of SLE, and the expression of has-circRNA-102972, has-circRNA-102006, has-circRNA-104313 was decreased in plasma of SLE. Twenty-eight upregulated circRNAs and 119 downregulated circRNAs were overlapped from PBMCs and plasma, and ubiquitination was enriched. Furthermore, the circRNA-miRNA-mRNA network was constructed in SLE after analyzing dataset GSE61635 from GEO. The circRNA-miRNA-mRNA network comprises 54 circRNAs, 41 miRNAs, and 580 mRNAs. In addition, the TNF signaling pathway and the MAPK pathway were enriched from the mRNA of the miRNA target. CONCLUSION We first revealed the differentially expressed circRNAs in plasma and PBMCs, and then the circRNA-miRNA-mRNA network was constructed. The network's circRNAs could be a potential diagnostic biomarker and potentially play an important role in the pathogenesis and development of SLE. Key Points • This study analyzed the circRNAs expression profiles combined with the plasma and PBMCs, which provided a comprehensive overview of circRNAs expression patterns in SLE. • The network of the circRNA-miRNA-mRNA in SLE was constructed, which contributes to a better understanding of the pathogenesis and development of SLE.
Collapse
Affiliation(s)
- Fengping Zheng
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Lishan Tan
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Fan Zhang
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Sanmu Li
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Zhiwei Lai
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Huixuan Xu
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, 518020, People's Republic of China
| | - Zuying Xiong
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China.
| | - Yong Dai
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, 518020, People's Republic of China.
| |
Collapse
|
5
|
Karati D, Mahadik KR, Trivedi P, Kumar D. The Emerging Role of Janus Kinase Inhibitors in the Treatment of Cancer. Curr Cancer Drug Targets 2022; 22:221-233. [PMID: 35232350 DOI: 10.2174/1568009622666220301105214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/15/2021] [Accepted: 12/24/2021] [Indexed: 11/22/2022]
Abstract
Cancer is a leading cause of death worldwide. The Janus kinase (JAK) signal transducer and activator of transcription (STAT) signalling pathway is activated abnormally, which promotes carcinogenesis. Several cytokines are important cancer drivers. These proteins bind to receptors and use the Janus kinase (JAK) and STAT pathways to communicate their responses. Cancer risks are linked to genetic differences in the JAK-STAT system. JAK inhibitors have shown to reduce STAT initiation, tissue propagation, and cell existence in preclinical investigations in solid tumour cell line models. JAK inhibitors, notably ruxolitinib, a, JAK1 or 2 blockers, make cell lines and mouse models more susceptible to radiotherapy, biological response modifier therapy, and oncolytic viral treatment. Numerous JAK antagonists have been or are now being evaluated in cancerous patients as monotherapy or by combining with other drugs in clinical studies. In preclinical investigations, certain JAK inhibitors showed promise anticancer effects; however, clinical trials explicitly evaluating their effectiveness against the JAK/STAT system in solid tumours have yet to be completed. JAK inhibition is a promising strategy to target the JAK/STAT system in solid tumours, and it deserves to be tested further in clinical studies. The function of directing Janus kinases (JAKs), an upstream accelerator of STATs, as a technique for lowering STAT activity in various malignant circumstances is summarized in this article, which will help scientists to generate more specific drug molecules in future.
Collapse
Affiliation(s)
- Dipanjan Karati
- Poona college of Pharmacy, Bharati Vidyapeeth (Deemed to be Unoiversity), Erandwane, Pune- 411038, Maharashtra, India
| | - Kakasaheb Ramoo Mahadik
- Centre of Innovation and Translational Research, Poona College of Pharmacy, Bharati Vidyapeeth, Pune 411038, India
| | - Piyush Trivedi
- Centre of Innovation and Translational Research, Poona College of Pharmacy, Bharati Vidyapeeth, Pune 411038, India
| | - Dileep Kumar
- Poona college of Pharmacy, Bharati Vidyapeeth (Deemed to be Unoiversity), Erandwane, Pune- 411038, Maharashtra, India
| |
Collapse
|
6
|
Qiu F, Li Y, Zhu Y, Li G, Lei F, Zhang S, Luo L, Zhu J, Guo Y, Du B, Xi X. CX3CR1 might be a promising predictor of systemic lupus erythematosus patients with pulmonary fibrosis. Scand J Immunol 2021; 94:e13038. [PMID: 33665864 DOI: 10.1111/sji.13038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 11/28/2022]
Abstract
The inflammatory process in systemic lupus erythematosus (SLE) affects many organs including the lungs. Chemokines are suggested to play important roles in the pathogenesis of SLE with pulmonary fibrosis (PF). In the present study, our objective is to evaluate the correlation between chemokines and PF in SLE patients. Transcriptome sequencing analysis was used to find the different expressed genes between SLE patients with PF and without PF. Enzyme-linked immunosorbent assay (ELISA) was used to detect serum levels of chemokines in SLE patients and healthy controls. Expression of CX3CR1 was measured by real-time polymerase chain reaction (PCR) and flow cytometer. Sixteen differentially chemokine genes were found to be associated to SLE with PF. Meanwhile, the upregulation of C-X3-C motif chemokine receptor 1 (CX3CR1) and its ligand, CX3C chemokine ligand 1 (CX3CL1) were observed in SLE patients with PF than that of SLE patients without PF and healthy control. Phenotypic analysis also showed that the surface expression of CX3CR1 increased in PBMCs from SLE patients with PF. Our observations indicated that CX3CL1/CX3CR1 axis is associated with PF in SLE. CX3CR1 might be a promising predictor of SLE with PF and the interactions between CX3CL1 and CX3CR1 might provide potential candidate target for the treatment of SLE with PF.
Collapse
Affiliation(s)
- Fen Qiu
- Institute of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Youling Li
- Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Yunhe Zhu
- Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Gang Li
- Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Feifei Lei
- Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Shuang Zhang
- Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Lei Luo
- Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Jietao Zhu
- Institute of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Yang Guo
- Institute of Basic Medical Science, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Boyu Du
- Institute of Basic Medical Science, Hubei University of Medicine, Shiyan, China.,Renmin Hospital, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Xueyan Xi
- Institute of Basic Medical Science, Hubei University of Medicine, Shiyan, China.,Renmin Hospital, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
7
|
Nozaki Y. The Network of Inflammatory Mechanisms in Lupus Nephritis. Front Med (Lausanne) 2020; 7:591724. [PMID: 33240910 PMCID: PMC7677583 DOI: 10.3389/fmed.2020.591724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022] Open
Abstract
Several signaling pathways are involved in the progression of kidney disease in humans and in animal models, and kidney disease is usually due to the sustained activation of these pathways. Some of the best understood pathways are specific proinflammatory cytokine and protein kinase pathways (e.g., protein kinase C and mitogen-activated kinase pathways, which cause cell proliferation and fibrosis and are associated with angiotensin II) and transforming growth factor-beta (TGF-β) signaling pathways (e.g., the TGF-β signaling pathway, which leads to increased fibrosis and kidney scarring. It is thus necessary to continue to advance our knowledge of the pathogenesis and molecular biology of kidney disease and to develop new treatments. This review provides an update of important findings about kidney diseases (including diabetic nephropathy, lupus nephritis, and vasculitis, i.e., vasculitis with antineutrophilic cytoplasmic antibodies). New disease targets, potential pathological pathways, and promising therapeutic approaches from basic science to clinical practice are presented, and the blocking of JAK/STAT and TIM-1/TIM-4 signaling pathways as potential novel therapeutic agents in lupus nephritis is discussed.
Collapse
Affiliation(s)
- Yuji Nozaki
- Department of Hematology and Rheumatology, Faculty of Medicine, Kindai University, Osaka, Japan
| |
Collapse
|
8
|
|
9
|
Chen SK, Barbhaiya M, Solomon DH, Guan H, Yoshida K, Feldman CH, Everett BM, Costenbader KH. Atrial Fibrillation/flutter Hospitalizations among US Medicaid Recipients with and without Systemic Lupus Erythematosus. J Rheumatol 2019; 47:1359-1365. [PMID: 31676703 DOI: 10.3899/jrheum.190502] [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] [Accepted: 10/11/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a multisystem chronic inflammatory autoimmune disease with high prevalence of several risk factors for atrial fibrillation/flutter (AF). However, the incidence and risk of AF in SLE have not been well quantified. METHODS We used the United States Medicaid Analytic eXtract from 2007 to 2010 to identify beneficiaries aged 18-65 years, with prevalent SLE, each matched by age and sex to 4 non-SLE general Medicaid recipients. We estimated the incidence rates (IR) per 1000 person-years (PY) for AF hospitalizations and used multivariable Cox regression to estimate the HR for AF hospitalization. RESULTS We identified 46,876 US Medicaid recipients with SLE, and 187,504 age- and sex-matched non-SLE controls (93% female; mean age 41.5 ± 12.2 yrs). Known AF risk factors such as hypertension (HTN), cardiovascular disease (CVD), and kidney disease were more prevalent in patients with SLE. During a mean followup of 1.9 ± 1.1 years for SLE, and 1.8 ± 1.1 years for controls, the IR per 1000 PY for AF was 1.4 (95% CI 1.1-1.6) among patients with SLE and 0.7 (95% CI 0.6-0.8) among non-SLE controls. In age- and sex-matched and race-adjusted Cox models, the HR for AF was 1.79 (95% CI 1.43-2.24); after adjustment for baseline HTN and CVD, the adjusted HR was reduced to 1.17 (95% CI 0.92-1.48). CONCLUSION SLE was associated with a doubled rate of hospitalization for AF compared to age- and sex-matched general Medicaid patients. In a race-adjusted model, the risk was 80% higher. However, the AF risk factors HTN and CVD were more prevalent among patients with SLE and accounted for the excess risk.
Collapse
Affiliation(s)
- Sarah K Chen
- From the Division of Rheumatology, Immunology and Allergy, the Division of Pharmacoepidemiology and Pharmacoeconomics, and the Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, New York, USA. .,S.K. Chen, MD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; M. Barbhaiya, MD, MPH, Division of Rheumatology, Department of Medicine, Hospital for Special Surgery; D.H. Solomon, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, and Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital; H. Guan, PhD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; K. Yoshida, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; C.H. Feldman, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; B.M. Everett, MD, MPH, Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital; K.H. Costenbader, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital. B.M. Everett and K.H. Costenbader are co-senior authors.
| | - Medha Barbhaiya
- From the Division of Rheumatology, Immunology and Allergy, the Division of Pharmacoepidemiology and Pharmacoeconomics, and the Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, New York, USA.,S.K. Chen, MD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; M. Barbhaiya, MD, MPH, Division of Rheumatology, Department of Medicine, Hospital for Special Surgery; D.H. Solomon, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, and Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital; H. Guan, PhD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; K. Yoshida, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; C.H. Feldman, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; B.M. Everett, MD, MPH, Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital; K.H. Costenbader, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital. B.M. Everett and K.H. Costenbader are co-senior authors
| | - Daniel H Solomon
- From the Division of Rheumatology, Immunology and Allergy, the Division of Pharmacoepidemiology and Pharmacoeconomics, and the Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, New York, USA.,S.K. Chen, MD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; M. Barbhaiya, MD, MPH, Division of Rheumatology, Department of Medicine, Hospital for Special Surgery; D.H. Solomon, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, and Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital; H. Guan, PhD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; K. Yoshida, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; C.H. Feldman, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; B.M. Everett, MD, MPH, Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital; K.H. Costenbader, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital. B.M. Everett and K.H. Costenbader are co-senior authors
| | - Hongshu Guan
- From the Division of Rheumatology, Immunology and Allergy, the Division of Pharmacoepidemiology and Pharmacoeconomics, and the Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, New York, USA.,S.K. Chen, MD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; M. Barbhaiya, MD, MPH, Division of Rheumatology, Department of Medicine, Hospital for Special Surgery; D.H. Solomon, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, and Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital; H. Guan, PhD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; K. Yoshida, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; C.H. Feldman, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; B.M. Everett, MD, MPH, Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital; K.H. Costenbader, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital. B.M. Everett and K.H. Costenbader are co-senior authors
| | - Kazuki Yoshida
- From the Division of Rheumatology, Immunology and Allergy, the Division of Pharmacoepidemiology and Pharmacoeconomics, and the Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, New York, USA.,S.K. Chen, MD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; M. Barbhaiya, MD, MPH, Division of Rheumatology, Department of Medicine, Hospital for Special Surgery; D.H. Solomon, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, and Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital; H. Guan, PhD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; K. Yoshida, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; C.H. Feldman, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; B.M. Everett, MD, MPH, Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital; K.H. Costenbader, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital. B.M. Everett and K.H. Costenbader are co-senior authors
| | - Candace H Feldman
- From the Division of Rheumatology, Immunology and Allergy, the Division of Pharmacoepidemiology and Pharmacoeconomics, and the Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, New York, USA.,S.K. Chen, MD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; M. Barbhaiya, MD, MPH, Division of Rheumatology, Department of Medicine, Hospital for Special Surgery; D.H. Solomon, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, and Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital; H. Guan, PhD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; K. Yoshida, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; C.H. Feldman, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; B.M. Everett, MD, MPH, Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital; K.H. Costenbader, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital. B.M. Everett and K.H. Costenbader are co-senior authors
| | - Brendan M Everett
- From the Division of Rheumatology, Immunology and Allergy, the Division of Pharmacoepidemiology and Pharmacoeconomics, and the Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, New York, USA.,S.K. Chen, MD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; M. Barbhaiya, MD, MPH, Division of Rheumatology, Department of Medicine, Hospital for Special Surgery; D.H. Solomon, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, and Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital; H. Guan, PhD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; K. Yoshida, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; C.H. Feldman, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; B.M. Everett, MD, MPH, Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital; K.H. Costenbader, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital. B.M. Everett and K.H. Costenbader are co-senior authors
| | - Karen H Costenbader
- From the Division of Rheumatology, Immunology and Allergy, the Division of Pharmacoepidemiology and Pharmacoeconomics, and the Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, New York, USA.,S.K. Chen, MD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; M. Barbhaiya, MD, MPH, Division of Rheumatology, Department of Medicine, Hospital for Special Surgery; D.H. Solomon, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, and Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital; H. Guan, PhD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; K. Yoshida, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; C.H. Feldman, MD, MPH, ScD, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital; B.M. Everett, MD, MPH, Divisions of Cardiovascular and Preventive Medicine, Department of Medicine, Brigham and Women's Hospital; K.H. Costenbader, MD, MPH, Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital. B.M. Everett and K.H. Costenbader are co-senior authors
| |
Collapse
|
10
|
Omran GA. Hematological and immunological impairment following in-utero and postnatal exposure to aluminum sulfate in female offspring of albino rats. Immunopharmacol Immunotoxicol 2019; 41:40-47. [PMID: 30706732 DOI: 10.1080/08923973.2018.1533967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Aim: Aluminum (Al) is a ubiquitous element extensively utilized in many products like food additives, pharmaceuticals, and vaccines, but its hematotoxic and immunotoxic effects are not entirely clarified. The present study explored the developmental hematotoxic and immunotoxic properties of aluminum sulfate (AS) in rats' offspring. Methods: Forty female offspring (10 rats each) were given three incremental AS doses plus a control group, from conception through lactation and after weaning until reached eight weeks old (near adults). Spleen relative weights along with total and differential blood counts were evaluated. Spectroscopic Al levels in spleen and brain were analyzed. Three immunoglobulins (IgG, IgM, and IgE) and two cytokines, interferon-γ and tumor necrosis factor-α, were measured through the ELISA technique. Results: The results revealed a significant relative increase in splenic weights mostly observed in the highest AS dose treated group. Reduction in the total leukocytic count was noticed in the three AS treated groups with relative lymphocytosis. Additionally, a significant decline in RBCs counts and hemoglobin concentrations were recorded. Tumor necrosis factor-α was significantly elevated in the three Al treated groups, while, interferon- γ showed a non-significant reduction compared to the control group. A significant increment in IgG and decline in IgE concentrations with no change in IgM level among groups were observed. Conclusion: Perinatal AS exposure caused mostly non-linear dose-dependent hematotoxicity and immunological impairment especially for the acquired immunity either cellular or humoral. Further studies can examine the immunotoxic effect of Al on male offspring during different stages of immune development.
Collapse
Affiliation(s)
- Ghada A Omran
- a Faculty of Medicine, Forensic Medicine and Clinical toxicology department , Assiut University , Assiut , Egypt
| |
Collapse
|
11
|
Aynacıoğlu AŞ, Bilir A, Tuna MY. Involvement of midkine in autoimmune and autoinflammatory diseases. Mod Rheumatol 2018; 29:567-571. [DOI: 10.1080/14397595.2018.1523701] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Ahmet Şükrü Aynacıoğlu
- Department of Medical Pharmacology, Istanbul Aydin University Medical Faculty, Istanbul, Turkey
| | - Ayhan Bilir
- Department of Histology and Embryology, Istanbul Aydin University Medical Faculty, Istanbul, Turkey
| | - Mehmet Yakup Tuna
- Department of Anatomy, Istanbul Aydin University Medical Faculty, Istanbul, Turkey
| |
Collapse
|
12
|
Li R, Tian C, Postlethwaite A, Jiao Y, Garcia-Godoy F, Pattanaik D, Wei D, Gu W, Li J. Rheumatoid arthritis and periodontal disease: What are the similarities and differences? Int J Rheum Dis 2018; 20:1887-1901. [DOI: 10.1111/1756-185x.13240] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Rongbin Li
- Center of Integrative Research; The First Hospital of Qiqihaer City; Qiqihaer Heilongjiang China
- Department of Orthopedic Surgery and BME-Campbell Clinic; University of Tennessee Health Science Center; Memphis TN USA
| | - Cheng Tian
- Department of Orthopedic Surgery and BME-Campbell Clinic; University of Tennessee Health Science Center; Memphis TN USA
| | - Arnold Postlethwaite
- Division of Connective Tissue Diseases; Department of Medicine; University of Tennessee Health Science Center; Memphis TN USA
- Department of Veterans Affairs Medical Center; University of Tennessee Health Science Center; Memphis TN USA
| | - Yan Jiao
- Department of Orthopedic Surgery and BME-Campbell Clinic; University of Tennessee Health Science Center; Memphis TN USA
| | - Franklin Garcia-Godoy
- Bioscience Research Center; College of Dentistry; University of Tennessee Health Science Center; Memphis TN USA
| | - Debendra Pattanaik
- Division of Connective Tissue Diseases; Department of Medicine; University of Tennessee Health Science Center; Memphis TN USA
- Department of Veterans Affairs Medical Center; University of Tennessee Health Science Center; Memphis TN USA
| | - Dongmei Wei
- Center of Integrative Research; The First Hospital of Qiqihaer City; Qiqihaer Heilongjiang China
| | - Weikuan Gu
- Department of Orthopedic Surgery and BME-Campbell Clinic; University of Tennessee Health Science Center; Memphis TN USA
- Department of Veterans Affairs Medical Center; University of Tennessee Health Science Center; Memphis TN USA
| | - Jianwei Li
- Center of Integrative Research; The First Hospital of Qiqihaer City; Qiqihaer Heilongjiang China
| |
Collapse
|
13
|
Role of vitamin D deficiency in systemic lupus erythematosus incidence and aggravation. AUTOIMMUNITY HIGHLIGHTS 2017; 9:1. [PMID: 29280010 PMCID: PMC5743852 DOI: 10.1007/s13317-017-0101-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/01/2017] [Indexed: 12/31/2022]
Abstract
Vitamin D is one of the main groups of sterols; playing an important role in phospho-calcic metabolism. The conversion of 7-dehydrocholesterol to pre- vitamin D3 in the skin, through solar ultraviolet B radiation, is the main source of vitamin D. Since lupus patients are usually photosensitive, the risk of developing vitamin D deficiency in is high in this population. Although evidences showed the connotation between systemic lupus erythematosus (SLE) and vitamin D through which SLE can lead to lower vitamin D levels, it is also important to consider the possibility that vitamin D deficiency may have a causative role in SLE etiology. This paper analyzes existing data from various studies to highlight the role of vitamin D deficiency in SLE occurrence and aggravation and the probable efficacy of vitamin D supplementation on SLE patients. We searched “Science Direct” and “Pub Med” using “Vitamin D” and “SLE” for finding the studies focusing on the association between vitamin D deficiency and SLE incidence and consequences. Evidences show that vitamin D plays an important role in the pathogenesis and progression of SLE and vitamin D supplementation seems to ameliorate inflammatory and hemostatic markers; so, can improve clinical subsequent.
Collapse
|
14
|
Stabilized β-Catenin Ameliorates ALPS-Like Symptoms of B6/ lpr Mice. J Immunol Res 2017; 2017:3469108. [PMID: 29250557 PMCID: PMC5700472 DOI: 10.1155/2017/3469108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 08/15/2017] [Indexed: 01/05/2023] Open
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is an incurable disease mainly caused by the defect of Fas-mediated apoptosis and characterized by nonmalignant autoimmune lymphoproliferation. Stabilized β-catenin could not only potentiate Fas-mediated T cell apoptosis via upregulating the expression of Fas on activated T cells, but also potentiate T cell apoptosis via intrinsic apoptotic pathway. In the present study, we introduced β-catTg into lpr/lpr mice and aimed to explore the potential role of stabilized β-catenin (β-catTg) in the development of ALPS-like phenotypes of lpr/lpr mice. We found that the total splenocyte cells and some compositions were slightly downregulated in β-catTglpr/lpr mice, especially the CD4 and CD8 TEM cells were significantly reduced. Meanwhile, stabilized β-catenin obviously decreased the numbers of spleen TCRβ+CD4−CD8− T (DNT) cells, and the levels of some serum proinflammatory factors also were lowered in β-catTglpr/lpr mice. Beyond that, stabilized β-catenin slightly lowered the levels of the serum autoantibodies and the scores of kidney histopathology of β-catTglpr/lpr mice compared with lpr/lpr mice. Our study suggested that stabilized β-catenin ameliorated some ALPS-like symptoms of lpr/lpr mice by potentiating Fas-independent signal-mediated T cell apoptosis, which might uncover a potential novel therapeutic direction for ALPS.
Collapse
|
15
|
Unmet Needs in the Pathogenesis and Treatment of Cardiovascular Comorbidities in Chronic Inflammatory Diseases. Clin Rev Allergy Immunol 2017; 55:254-270. [PMID: 28741263 DOI: 10.1007/s12016-017-8624-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The developments that have taken place in recent decades in the diagnosis and therapy of a number of diseases have led to improvements in prognosis and life expectancy. As a consequence, there has been an increase in the number of patients affected by chronic diseases and who can face new pathologies during their lifetime. The prevalence of chronic heart failure, for example, is approximately 1-2% of the adult population in developed countries, rising to ≥10% among people >70 years of age; in 2015, more than 85 million people in Europe were living with some sort of cardiovascular disease (CVD) (Lubrano and Balzan World J Exp Med 5:21-32, 5; Takahashi et al. Circ J 72:867-72, 8; Kaptoge et al. Lancet 375:132-40, 9). Chronic disease can become, in turn, a major risk factor for other diseases. Furthermore, several new drugs have entered clinical practice whose adverse effects on multiple organs are still to be evaluated. All this necessarily involves a multidisciplinary vision of medicine, where the physician must view the patient as a whole and where collaboration between the various specialists plays a key role. An example of what has been said so far is the relationship between CVD and chronic inflammatory diseases (CIDs). Patients with chronic CVD may develop a CID within their lifetime, and, vice versa, a CID can be a risk factor for the development of CVD. Moreover, drugs used for the treatment of CIDs may have side effects involving the cardiovascular system and thus may be contraindicated. The purpose of this paper is to investigate the close relationship between these two groups of diseases and to provide recommendations on the diagnostic approach and treatments in light of the most recent scientific data available.
Collapse
|
16
|
Xu X, Yu B, Cai W, Huang Z. TCF1 deficiency ameliorates autoimmune lymphoproliferative syndrome (ALPS)-like phenotypes oflpr/lprmice. Scand J Immunol 2017; 85:406-416. [DOI: 10.1111/sji.12546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/07/2017] [Indexed: 12/12/2022]
Affiliation(s)
- X. Xu
- Institute of Human Virology; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou China
- Key Laboratory of Tropical Diseases Control; Ministry of Education in China; Sun Yat-sen University; Guangzhou China
| | - B. Yu
- Key Laboratory for Major Obstetric Diseases of Guangdong Province; Third Affiliated Hospital of Guangzhou Medical University; Guangzhou China
| | - W. Cai
- Department of Biochemistry; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou China
| | - Z. Huang
- Institute of Human Virology; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou China
- Key Laboratory of Tropical Diseases Control; Ministry of Education in China; Sun Yat-sen University; Guangzhou China
- Department of Biochemistry; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou China
| |
Collapse
|
17
|
Regulatory immune cells and functions in autoimmunity and transplantation immunology. Autoimmun Rev 2017; 16:435-444. [DOI: 10.1016/j.autrev.2017.03.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 01/26/2017] [Indexed: 12/15/2022]
|
18
|
Kuhn A, Aberer E, Bata-Csörgő Z, Caproni M, Dreher A, Frances C, Gläser R, Klötgen HW, Landmann A, Marinovic B, Nyberg F, Olteanu R, Ranki A, Szepietowski JC, Volc-Platzer B. S2k guideline for treatment of cutaneous lupus erythematosus - guided by the European Dermatology Forum (EDF) in cooperation with the European Academy of Dermatology and Venereology (EADV). J Eur Acad Dermatol Venereol 2016; 31:389-404. [PMID: 27859683 DOI: 10.1111/jdv.14053] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 10/26/2016] [Indexed: 12/31/2022]
Abstract
Cutaneous lupus erythematosus (CLE) is a rare inflammatory autoimmune disease with heterogeneous clinical manifestations. To date, no therapeutic agents have been licensed specifically for patients with this disease entity, and topical and systemic drugs are mostly used 'off-label'. The aim of the present guideline was to achieve a broad consensus on treatment strategies for patients with CLE by a European subcommittee, guided by the European Dermatology Forum (EDF) and supported by the European Academy of Dermatology and Venereology (EADV). In total, 16 European participants were included in this project and agreed on all recommendations. Topical corticosteroids remain the mainstay of treatment for localized CLE, and further topical agents, such as calcineurin inhibitors, are listed as alternative first-line or second-line topical therapeutic option. Antimalarials are recommended as first-line and long-term systemic treatment in all CLE patients with severe and/or widespread skin lesions, particularly in patients with a high risk of scarring and/or the development of systemic disease. In addition to antimalarials, systemic corticosteroids are recommended as first-line treatment in highly active and/or severe CLE. Second- and third-line systemic treatments include methotrexate, retinoids, dapsone and mycophenolate mofetil or mycophenolate acid, respectively. Thalidomide should only be used in selected therapy-refractory CLE patients, preferably in addition to antimalarials. Several new therapeutic options, such as B-cell- or interferon α-targeted agents, need to be further evaluated in clinical trials to assess their efficacy and safety in the treatment of patients with CLE.
Collapse
Affiliation(s)
- A Kuhn
- Interdisciplinary Center for Clinical Trials (IZKS), University Medical Center Mainz, Mainz, Germany.,Division of Immunogenetics, Tumor Immunology Program, German Cancer Research Center, Heidelberg, Germany
| | - E Aberer
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Z Bata-Csörgő
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - M Caproni
- Department of Medical and Surgical Critical Care Section of Dermatology, University of Florence, Florence, Italy
| | - A Dreher
- Evidence-Based Medicine Frankfurt, Institute for General Practice, Goethe-University Frankfurt, Frankfurt, Germany
| | - C Frances
- Department of Dermatology and Allergology, Hôpital Tenon, Paris, France
| | - R Gläser
- Department of Dermatology, Venerology and Allergology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - H-W Klötgen
- Department of Dermatology, Inselspital Bern - University Hospital, Bern, Switzerland
| | - A Landmann
- Division of Immunogenetics, Tumor Immunology Program, German Cancer Research Center, Heidelberg, Germany
| | - B Marinovic
- Department of Dermatology and Venereology, University Hospital Center Zagreb and School of Medicine University of Zagreb, Zagreb, Croatia
| | - F Nyberg
- Institution for Clinical Sciences, Unit for Dermatology, Karolinska Institutet at Danderyd Hospital (KIDS), Stockholm, Sweden
| | - R Olteanu
- Department of Dermatology, Colentina Clinical Hospital, Bucharest, Romania
| | - A Ranki
- Department of Skin and allergic diseases, Inflammation Center, Helsinki University Central Hospital, Helsinki, Finland
| | - J C Szepietowski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - B Volc-Platzer
- Department of Dermatology, Donauspital, University affiliated Hospital, Vienna, Austria
| |
Collapse
|
19
|
Nakken B, Bodolay E, Szodoray P. Cytokine Milieu in Undifferentiated Connective Tissue Disease: a Comprehensive Review. Clin Rev Allergy Immunol 2016; 49:152-62. [PMID: 25274451 DOI: 10.1007/s12016-014-8452-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Undifferentiated connective tissue disease (UCTD) is a unique clinical entity, a potential forerunner of well-established systemic autoimmune/rheumatic diseases. UCTD is characterized by the presence of various clinical symptoms, as well as a diverse repertoire of autoantibodies, resembling systemic autoimmune diseases. Since approximately one third of these patients consequently transform into a full-blown systemic autoimmune/rheumatic disease, it is of major importance to assess pathogenic factors leading to this progression. In view of the fact that the serological and clinical picture of UCTD and systemic autoimmune diseases are very similar, it is assumed that analogous pathogenic factors perpetuate both disease entities. In systemic autoimmune conditions, a quantitative and qualitative impairment of regulatory T cells has been shown previously, and in parallel, a relative dominance of pro-inflammatory Th17 cells has been introduced. Moreover, the imbalance between regulatory and Th17 cells plays a pivotal role in the initiation and propagation of UCTD. Additionally, we depict a cytokine imbalance, which give raise to a biased T cell homeostasis from the UCTD phase throughout the fully developed systemic autoimmune disease stage. The levels of interleukin (IL)-6, IL-12, IL-17, IL-23, and interferon (IFN)-γ were pathologically increased with a parallel reduction of IL-10. We believe that the assessment of Th17/Treg cell ratio, as well as the simultaneous quantitation of cytokines may give a useful diagnostic tool at the early UCTD stage to identify patients with a higher chance of consecutive disease progression toward serious systemic autoimmune diseases. Moreover, the early targeted immunomodulating therapy in these patients may decelerate, or even stop this progression, before the development of serious autoimmune conditions with organ damage.
Collapse
Affiliation(s)
- Britt Nakken
- Institute of Immunology, Rikshospitalet, Oslo University Hospital, Sognsvannsveien 20, Oslo, Norway, N-0027
| | - Edit Bodolay
- Department of Clinical Immunology, Institute of Medicine, University of Debrecen Medical and Health Science Centre, Debrecen, Hungary
| | - Peter Szodoray
- Institute of Immunology, Rikshospitalet, Oslo University Hospital, Sognsvannsveien 20, Oslo, Norway, N-0027.
| |
Collapse
|
20
|
Abstract
The treatment of systemic lupus erythematosus (SLE) has been refined over the years, with the recognition that a fine balance lies between aggressive and prompt therapy and attendant complications brought upon by immunosuppressive therapy itself. However, there has been limited change to the repertoire of drugs available to treat this challenging disease. The current standard therapy for severe manifestations of SLE includes the use of high-dose corticosteroids and cytotoxic agents such as cyclophosphamide (CYC), which have been associated with an increased risk of serious and opportunistic infections. The need for safer, more targeted therapies has been recognized and now, with the exponential increase in the understanding of immunopathogenic mechanisms in SLE, the way has been paved for the development of biologic or targeted therapies in SLE. Although the potential immunosuppression, long-term safety issues and cost-effectiveness remain unclear. These targeted therapies may range from small molecules that specifically inhibit inflammatory processes at an intracellular, cell-cell or cell-matrix level to monoclonal antibodies, soluble receptors or natural antagonists that interfere with cytokine function, cellular activation and inflammatory gene transcription.
Collapse
Affiliation(s)
- S Vasoo
- The Lupus Research Unit, The Rayne Institute, St Thomas' Hospital London, UK
| | | |
Collapse
|
21
|
Use of the VH6-1 gene segment to code for anti-interleukin-18 autoantibodies in multiple sclerosis. Immunogenetics 2016; 68:237-46. [PMID: 26743536 DOI: 10.1007/s00251-015-0895-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/23/2015] [Indexed: 12/12/2022]
Abstract
We investigated whether levels and repertoires of anti-interleukin-18 (IL-18) autoantibodies (auto-Abs) differ in multiple sclerosis (MS) patients and healthy donors (HDs). IL-18 concentration in MS patients' sera was higher than in HD, but the level of anti-IL-18 auto-Abs was lower in MS patients. Correlation patterns of IL-18/anti-IL-18 auto-Abs system differed in HD and MS patients, so we have compared segment composition of the anti-IL-18 single-chain variable fragments (scFvs) selected from MS and naïve phage display libraries. Considerable differences between anti-IL-18 auto-Abs of these libraries were found. MS panel contained auto-Abs displaying both signs of "fetal" and somatically hypermutated repertoires. Naïve panel mainly contained the naïve antibodies. These variations from the norm are possible results of abnormal regulation of the repertoire perhaps determined by remodeling of the molecular mechanisms involved in the V(D)J recombination and/or abnormal selection by antigen in MS pathogenesis.
Collapse
|
22
|
Wang Y, Ewart D, Crabtree JN, Yamamoto A, Baechler EC, Fazeli P, Peterson EJ. PTPN22 Variant R620W Is Associated With Reduced Toll-like Receptor 7-Induced Type I Interferon in Systemic Lupus Erythematosus. Arthritis Rheumatol 2015; 67:2403-14. [PMID: 26018863 DOI: 10.1002/art.39211] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 05/19/2015] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Protein tyrosine phosphatase nonreceptor type 22 (PTPN22) is associated with an increased risk of systemic lupus erythematosus (SLE). PTPN22 encodes Lyp, and a disease-associated coding variant bears an R620W substitution (LypW). LypW carriage is associated with impaired production of type I interferon (IFN) by myeloid cells following Toll-like receptor (TLR) engagement. The aim of this study was to investigate the effects of LypW carriage on TLR signaling in patients with SLE. METHODS Plasma IFNα concentrations and whole-blood IFN gene scores were compared in SLE patients who were LypW carriers and those who were noncarriers. TLR-7 agonist R848-stimulated IFNα and tumor necrosis factor levels, IFN-dependent gene expression, and STAT-1 activation were determined in peripheral blood mononuclear cells (PBMCs) and/or plasmacytoid dendritic cells (PDCs) obtained from these patients. The effect of LypW expression on the systemic type I IFN response to R848 stimulation in vivo was assessed in transgenic mice. RESULTS Plasma IFNα levels and whole-blood IFN gene signatures were comparable in SLE patients who were LypW carriers and those who were noncarriers. However, PBMCs from LypW carriers produced less IFNα and showed reduced IFN-dependent gene up-regulation and STAT-1 activation after R848 stimulation. The frequency of PDCs producing IFNα2 and the per-cell IFNα2 levels were significantly reduced in LypW carriers. LypW-transgenic mice displayed reduced TLR-7-induced circulating type I IFN responses. CONCLUSION PDCs from SLE patients carrying the disease-associated PTPN22 variant LypW showed a reduced capacity for TLR-7 agonist-induced type I IFN production, even though LypW carriers displayed systemic type I IFN activation comparable with that observed in noncarriers. LypW carriage identifies SLE patients who may harbor defects in TLR- and PDC-dependent host defense or antiinflammatory functions.
Collapse
Affiliation(s)
- Yaya Wang
- University of Minnesota, Minneapolis
| | | | | | | | | | | | | |
Collapse
|
23
|
Selected Aspects in the Pathogenesis of Autoimmune Diseases. Mediators Inflamm 2015; 2015:351732. [PMID: 26300591 PMCID: PMC4537751 DOI: 10.1155/2015/351732] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/24/2015] [Indexed: 11/26/2022] Open
Abstract
Autoimmune processes can be found in physiological circumstances. However, they are quenched with properly functioning regulatory mechanisms and do not evolve into full-blown autoimmune diseases. Once developed, autoimmune diseases are characterized by signature clinical features, accompanied by sustained cellular and/or humoral immunological abnormalities. Genetic, environmental, and hormonal defects, as well as a quantitative and qualitative impairment of immunoregulatory functions, have been shown in parallel to the relative dominance of proinflammatory Th17 cells in many of these diseases. In this review we focus on the derailed balance between regulatory and Th17 cells in the pathogenesis of autoimmune diseases. Additionally, we depict a cytokine imbalance, which gives rise to a biased T-cell homeostasis. The assessment of Th17/Treg-cell ratio and the simultaneous quantitation of cytokines, may give a useful diagnostic tool in autoimmune diseases. We also depict the multifaceted role of dendritic cells, serving as antigen presenting cells, contributing to the development of the pathognomonic cytokine signature and promote cellular and humoral autoimmune responses. Finally we describe the function and role of extracellular vesicles in particular autoimmune diseases. Targeting these key players of disease progression in patients with autoimmune diseases by immunomodulating therapy may be beneficial in future therapeutic strategies.
Collapse
|
24
|
Ethnicity-stratified analysis of the association between IL-18 polymorphisms and systemic lupus erythematosus in a European population: a meta-analysis. Arch Dermatol Res 2015; 307:747-55. [PMID: 26026656 DOI: 10.1007/s00403-015-1580-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 04/10/2015] [Accepted: 05/17/2015] [Indexed: 10/23/2022]
Abstract
We performed a meta-analysis to identify the association between polymorphisms in the promoter of interleukin-18 (IL-18) and susceptibility for systemic lupus erythematosus (SLE) . Genotype data for three single-nucleotide polymorphisms (SNPs rs360719, rs1946518, and rs187238) in the IL-18 promoter were extracted from 20 studies of three different ethnicities (European, Asian, and South American). Data from each ethnicity group and their combinations were analyzed. We found distinct evidence of an association between rs360719 and SLE (P = 0.001) in the European/South American group [odds ratio (OR) 1.31 per C allele, 95% confidence interval (CI) 1.11-1.53]. Stratification analysis by ethnicity showed a significant association between rs360719 and SLE in the European population (OR 1.33 per C allele, 95% CI 1.11-1.61, P = 0.003) and a lesser effect in the same direction in the South American population (OR 1.18). A significant association was also identified between rs1946518 and SLE in the European population (OR 1.16 per A allele, 95% CI 1.03-1.30, P = 0.017), although there was no association in the Asian or the combined European/Asian population. We also examined genome-wide association study (GWAS) data from an Asian subpopulation (Chinese) for the association between rs1946518 and SLE, but found no association (P = 0.83). The third SNP, rs187238, was not significantly associated with SLE in any of the populations examined. In summary, this study identified a significant association between SLE and two SNPs within the IL-18 gene promoter region (rs360719 and rs1946518) in a European population, but not in populations of Asian origin.
Collapse
|
25
|
Fleischer SJ, Giesecke C, Mei HE, Lipsky PE, Daridon C, Dörner T. Increased frequency of a unique spleen tyrosine kinase bright memory B cell population in systemic lupus erythematosus. Arthritis Rheumatol 2015; 66:3424-35. [PMID: 25156507 DOI: 10.1002/art.38854] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 08/14/2014] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is characterized by B cell hyperactivity and autoantibody production. As spleen tyrosine kinase (Syk) is pivotal in B cell activation, these experiments aimed to examine the extent to which Syk was abnormally expressed in SLE B cells and the nature of the B cell subset that differently expressed Syk. METHODS B cells from healthy donors and SLE patients were analyzed by flow cytometry to assess basal expression of Syk and phosphorylated Syk. B cell subsets expressing higher levels of Syk were found, and their detailed phenotype, in vitro differentiation into plasmablasts/plasma cells, and Syk induction by cytokines were determined. RESULTS Syk expression was higher in CD27+ memory B cells than in naive B cells from SLE patients. However, a significantly increased frequency of CD27- B cells with bright expression of Syk (Syk++) was found in SLE patients. CD27-Syk++ B cells showed enhanced basal expression of p-Syk and stronger Syk phosphorylation upon B cell receptor (BCR) engagement as compared to CD27-Syk+ B cells. CD27-Syk++ B cells were CD38- as well as CD19++, CD20++, and mainly CD21-, with decreased ABCB1 transporter activity. In contrast to CD27-Syk+ B cells, CD27-Syk++ B cells exhibited enhanced differentiation into CD27++ IgG-secreting cells and expressed somatically mutated BCR gene rearrangements. Syk++ B cells were inducible in vitro by stimulation with interferon-γ, lipopolysaccharide, or tumor necrosis factor α. CONCLUSION SLE patients exhibit an increased frequency of hitherto unknown CD27-Syk++ memory-like B cells, indicating that intracellular Syk density could distinguish CD27- memory B cells from truly naive B cell subsets. Furthermore, the CD27-Syk++ subset is a candidate for a source of increased plasma cells in SLE.
Collapse
Affiliation(s)
- Sarah J Fleischer
- Charité University Medicine Berlin and German Rheumatism Research Center Berlin, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Morris G, Berk M, Walder K, Maes M. Central pathways causing fatigue in neuro-inflammatory and autoimmune illnesses. BMC Med 2015; 13:28. [PMID: 25856766 PMCID: PMC4320458 DOI: 10.1186/s12916-014-0259-2] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 12/17/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The genesis of severe fatigue and disability in people following acute pathogen invasion involves the activation of Toll-like receptors followed by the upregulation of proinflammatory cytokines and the activation of microglia and astrocytes. Many patients suffering from neuroinflammatory and autoimmune diseases, such as multiple sclerosis, Parkinson's disease and systemic lupus erythematosus, also commonly suffer from severe disabling fatigue. Such patients also present with chronic peripheral immune activation and systemic inflammation in the guise of elevated proinflammtory cytokines, oxidative stress and activated Toll-like receptors. This is also true of many patients presenting with severe, apparently idiopathic, fatigue accompanied by profound levels of physical and cognitive disability often afforded the non-specific diagnosis of chronic fatigue syndrome. DISCUSSION Multiple lines of evidence demonstrate a positive association between the degree of peripheral immune activation, inflammation and oxidative stress, gray matter atrophy, glucose hypometabolism and cerebral hypoperfusion in illness, such as multiple sclerosis, Parkinson's disease and chronic fatigue syndrome. Most, if not all, of these abnormalities can be explained by a reduction in the numbers and function of astrocytes secondary to peripheral immune activation and inflammation. This is also true of the widespread mitochondrial dysfunction seen in otherwise normal tissue in neuroinflammatory, neurodegenerative and autoimmune diseases and in many patients with disabling, apparently idiopathic, fatigue. Given the strong association between peripheral immune activation and neuroinflammation with the genesis of fatigue the latter group of patients should be examined using FLAIR magnetic resonance imaging (MRI) and tested for the presence of peripheral immune activation. SUMMARY It is concluded that peripheral inflammation and immune activation, together with the subsequent activation of glial cells and mitochondrial damage, likely account for the severe levels of intractable fatigue and disability seen in many patients with neuroimmune and autoimmune diseases.This would also appear to be the case for many patients afforded a diagnosis of Chronic Fatigue Syndrome.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Bryn Road seaside 87, Llanelli, SA152LW Wales UK
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Department of Psychiatry and The Florey Institute of Neuroscience and Mental Health, Orygen, The National Centre of Excellence in Youth Mental Health, The University of Melbourne, Parkville, Australia
| | - Ken Walder
- Centre for Molecular and Medical Research, School of Medicine, Deakin University, Geelong, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
27
|
Nielepkowicz-Goździńska A, Fendler W, Robak E, Kulczycka-Siennicka L, Górski P, Pietras T, Brzeziańska E, Antczak A. Exhaled IL-8 in systemic lupus erythematosus with and without pulmonary fibrosis. Arch Immunol Ther Exp (Warsz) 2014; 62:231-8. [PMID: 24492930 PMCID: PMC4024123 DOI: 10.1007/s00005-014-0270-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 10/09/2013] [Indexed: 11/25/2022]
Abstract
The purpose of this study is to evaluate the relationship between the concentration of interleukin-8 (IL-8) in exhaled breath condensate (EBC) and bronchoalveolar lavage fluid (BALF) with the disease activity score and pulmonary function of systemic lupus erythematosus (SLE) patients with and without pulmonary fibrosis. Thirty-four SLE patients and 31 healthy controls were enrolled and evaluated using high-resolution computed tomography (HRCT), pulmonary function tests, systemic lupus activity measure (SLAM), assessing BALF and EBC. IL-8 levels in BALF and EBC samples were measured with an enzyme-immunosorbent assay kit. The mean (±SEM) IL-8 concentrations in BALF and EBC were higher in SLE patients compared to healthy controls (34.84 ± 95.0 vs. 7.65 ± 21.22 pg/ml, p < 0.001; 3.82 ± 0.52 pg/m vs. 1.7 ± 1.7 pg/ml, p < 0.001, respectively). SLE patients had increased percentage of neutrophils in BALF when compared with control group (1.00 ± 5.99 vs. 0.00 ± 0.56 %, p = 0.0003). Pulmonary fibrosis in HRCT was found in 50 % of SLE patients. The disease activity scored by SLAM was significantly higher and total lung capacity was significantly lower in SLE patients with pulmonary fibrosis (8.00 ± 3.17 vs. 6.00 ± 2.31, p = 0.01; 88.00 ± 28.29 vs. 112.00 ± 21.08 % predicted, p = 0.01, respectively). In SLE patients with pulmonary fibrosis, correlations were found between SLAM and IL-8 concentration in BALF, forced expiratory volume in 1 s and forced vital capacity (r = 0.65, p = 0.006; r = −0.53, p = 0.035; r = −0.67, p = 0.006, respectively). Our results indicate that IL-8 plays an important role in the pathogenesis of SLE. An increased concentration of IL-8 according to BALF could be considered as a useful biomarker of SLE activity and pulmonary fibrosis in SLE.
Collapse
Affiliation(s)
| | - Wojciech Fendler
- Department of Pediatrics, Oncology, Hematology and Diabetology, Medical University of Lodz, Lodz, Poland
| | - Ewa Robak
- Department of Dermatology and Venereology, Medical University of Lodz, Lodz, Poland
| | | | - Paweł Górski
- Department of Pneumonology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Tadeusz Pietras
- Department of Pneumonology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Ewa Brzeziańska
- Department of Molecular Bases of Medicine, Medical University of Lodz, Lodz, Poland
| | - Adam Antczak
- Department of General and Oncological Pneumology, Medical University of Lodz, Kopcińskiego 22, 90-153 Lodz, Poland
| |
Collapse
|
28
|
Burkard T, Pfister O, Rickli H, Follath F, Hack D, Zaker R, Pittl U, Handschin R, Pfisterer M, Brunner-La Rocca HP. Prognostic impact of systemic inflammatory diseases in elderly patients with congestive heart failure. QJM 2014; 107:131-8. [PMID: 24131549 DOI: 10.1093/qjmed/hct205] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND AND AIMS Inflammation is part of the pathophysiology of congestive heart failure (CHF). However, little is known about the impact of the presence of systemic inflammatory disease (SID), defined as inflammatory syndrome with constitutional symptoms and involvement of at least two organs as co-morbidity on the clinical course and prognosis of patients with CHF. METHODS AND RESULTS This is an analysis of all 622 patients included in TIME-CHF. After an 18 months follow-up, outcomes of patients with and without SID were compared. Primary endpoint was all-cause hospitalization free survival. Secondary endpoints were overall survival and CHF hospitalization free survival. At baseline, 38 patients had history of SID (6.1%). These patients had higher N-terminal pro brain natriuretic peptide and worse renal function than patients without SID. SID was a risk factor for adverse outcome [primary endpoint: hazard ratio (HR) = 1.73 (95% confidence interval: 1.18-2.55, P = 0.005); survival: HR = 2.60 (1.49-4.55, P = 0.001); CHF hospitalization free survival: HR = 2.3 (1.45-3.65, P < 0.001)]. In multivariate models, SID remained the strongest independent risk factor for survival and CHF hospitalization free survival. CONCLUSION In elderly patients with CHF, SID is independently accompanied with adverse outcome. Given the increasing prevalence of SID in the elderly population, these findings are clinically important for both risk stratification and patient management.
Collapse
Affiliation(s)
- T Burkard
- Department of Cardiology, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Ischemic heart disease in systemic inflammatory diseases. An appraisal. Int J Cardiol 2013; 170:286-90. [PMID: 24331863 DOI: 10.1016/j.ijcard.2013.11.048] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 11/17/2013] [Indexed: 02/07/2023]
Abstract
Systemic inflammatory diseases are inflammatory syndromes that are associated with increased cardiovascular morbidity and mortality. The link between inflammatory and cardiovascular diseases can be attributed to coexistence of classical risk factors and of inflammatory mechanisms activated in systemic inflammatory diseases and involving the immune system. Yet, clinical implications of these findings are not entirely clear and deeper knowledge and awareness of cardiac involvement in inflammatory diseases are necessary. The aims of this review are to summarize cardiac involvement in systemic inflammatory diseases and to identify areas where evidence is currently lacking that deserve further investigation in the future.
Collapse
|
31
|
Malarial infection of female BWF1 lupus mice alters the redox state in kidney and liver tissues and confers protection against lupus nephritis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:156562. [PMID: 24319531 PMCID: PMC3844167 DOI: 10.1155/2013/156562] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 09/07/2013] [Accepted: 09/30/2013] [Indexed: 01/17/2023]
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease characterized by an imbalanced redox state and increased apoptosis. Tropical infections, particularly malaria, may confer protection against SLE. Oxidative stress is a hallmark of SLE. We have measured changes in the levels of nitric oxide (NO), hydrogen peroxide (H2O2), malondialdehyde (MDA), and reduced glutathione (GSH) in both kidney and liver tissues of female BWF1 lupus mice, an experimental model of SLE, after infection with either live or gamma-irradiated malaria. We observed a decrease in NO, H2O2, and MDA levels in kidney tissues after infection of lupus mice with live malaria. Similarly, the levels of NO and H2O2 were significantly decreased in the liver tissues of lupus mice after infection with live malaria. Conversely, GSH levels were obviously increased in both kidney and liver tissues after infection of lupus mice with either live or gamma-irradiated malaria. Liver and kidney functions were significantly altered after infection of lupus mice with live malaria. We further investigated the ultrastructural changes and detected the number of apoptotic cells in kidney and liver tissues in situ by electron microscopy and TUNEL assays. Our data reveal that infection of lupus mice with malaria confers protection against lupus nephritis.
Collapse
|
32
|
Halade GV, Williams PJ, Veigas JM, Barnes JL, Fernandes G. Concentrated fish oil (Lovaza(R)) extends lifespan and attenuates kidney disease in lupus-prone short-lived (NZBxNZW)F1 mice. Exp Biol Med (Maywood) 2013; 238:610-22. [PMID: 23918873 DOI: 10.1177/1535370213489485] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
A growing number of reports indicate that anti-inflammatory actions of fish oil (FO) are beneficial against systemic lupus erythematosus (SLE). However, the majority of pre-clinical studies were performed using 5-20% FO, which is higher than the clinically relevant dose for lupus patients. The present study was performed in order to determine the effective low dose of FDA-approved concentrated FO (Lovaza®) compared to the commonly used FO-18/12 (18-Eicosapentaenoic acid [EPA]/12-Docosahexaenoic acid [DHA]). We examined the dose-dependent response of Lovaza® (1% and 4%) on an SLE mouse strain (NZBxNZW)F1 and compared the same with 1% and 4% placebo, as well as 4% FO-18/12, maintaining standard chow as the control. Results show for the first time that 1% Lovaza® extends maximal lifespan (517 d) and 4% Lovaza® significantly extends both the median (502 d) and maximal (600 d) life span of (NZBxNZW)F1 mice. In contrast, FO-18/12 extends only median lifespan (410 d) compared to standard chow diet (301 d). Additionally, 4% Lovaza® significantly decreased anti-dsDNA antibodies, reduced glomerulonephritis and attenuated lipopolysaccharide-induced pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) in splenocytes compared to placebo. 4% Lovaza® was also shown to reduce the expression of inflammatory cytokines, including IL-1β, IL-6 and TNF-α, while increasing renal anti-oxidant enzymes in comparison to placebo. Notably, NFκB activation and p65 nuclear translocation were lowered by 4% Lovaza® compared to placebo. These data indicate that 1% Lovaza® is beneficial, but 4% Lovaza® is more effective in suppressing glomerulonephritis and extending life span of SLE-prone short-lived mice, possibly via reducing inflammation signaling and modulating oxidative stress.
Collapse
Affiliation(s)
- Ganesh V Halade
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Texas Health Science Center at San Antonio, TX 78229, USA.
| | | | | | | | | |
Collapse
|
33
|
Familial aggregation of high tumor necrosis factor alpha levels in systemic lupus erythematosus. Clin Dev Immunol 2013; 2013:267430. [PMID: 24187561 PMCID: PMC3800640 DOI: 10.1155/2013/267430] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 08/19/2013] [Indexed: 11/28/2022]
Abstract
Systemic lupus erythematosus (SLE) patients frequently have high circulating tumor necrosis factor alpha (TNF-α) levels. We explored circulating TNF-α levels in SLE families to determine whether high levels of TNF-α were clustered in a heritable pattern. We measured TNF-α in 242 SLE patients, 361 unaffected family members, 23 unaffected spouses of SLE patients, and 62 unrelated healthy controls. Familial correlations and relative recurrence risk rates for the high TNF-α trait were assessed. SLE-affected individuals had the highest TNF-α levels, and TNF-α was significantly higher in unaffected first degree relatives than healthy unrelated subjects (P = 0.0025). No Mendelian patterns were observed, but 28.4% of unaffected first degree relatives of SLE patients had high TNF-α levels, resulting in a first degree relative recurrence risk of 4.48 (P = 2.9 × 10−5). Interestingly, the median TNF-α value in spouses was similar to that of the first degree relatives. Concordance of the TNF-α trait (high versus low) in SLE patients and their spouses was strikingly high at 78.2%. These data support a role for TNF-α in SLE pathogenesis, and TNF-α levels may relate with heritable factors. The high degree of concordance in SLE patients and their spouses suggests that environmental factors may also play a role in the observed familial aggregation.
Collapse
|
34
|
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
|
35
|
Postal M, Peliçari KO, Sinicato NA, Marini R, Costallat LTL, Appenzeller S. Th1/Th2 cytokine profile in childhood-onset systemic lupus erythematosus. Cytokine 2013; 61:785-91. [PMID: 23332615 DOI: 10.1016/j.cyto.2012.11.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 10/15/2012] [Accepted: 11/26/2012] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To determine the serum levels of Th1 (IL-12, IFN-γ,TNF-α) and Th2 (IL-5, IL-6 and IL-10) cytokines in childhood-onset SLE, first-degree relatives and healthy controls. To elucidate their association with disease activity, laboratory and treatment features. METHODS We included 60 consecutive childhood-onset SLE patients [median age 18 years (range 10-37)], 64 first-degree relatives [median 40 (range 28-52)] and 57 healthy [median age 19 years (range 6-30 years)] controls. Controls were age and sex-matched to SLE patients. SLE patients were assessed for clinical and laboratory SLE manifestations, disease activity (SLEDAI), damage (SDI) and current drug exposures. Mood and anxiety disorders were determined through Becks Depression (BDI) and Anxiety Inventory (BAI). Th1 (IL-12, IFN-γ,TNF-α) and Th2 (IL-5, IL-6 and IL-10) cytokines levels were measured by ELISA and compared by non-parametric tests. RESULTS Serum TNF-α (p=0.004), IL-6 (p=0.007) and IL-10 (p=0.03) levels were increased in childhood-onset SLE patients when compared to first-degree relatives and healthy controls. TNF-α levels were significantly increased in patients with active disease (p=0.014) and correlated directly with SLEDAI scores (r=0.39; p=0.002). IL-12 (p=0.042) and TNF-α (p=0.009) levels were significantly increased in patients with nephritis and TNF-α in patients with depression (p=0.001). No association between cytokine levels and SDI scores or medication was observed. CONCLUSION Th1 cytokines may play a role in the pathogenesis of neuropsychiatric and renal manifestations in childhood-onset SLE. The correlation with SLEDAI suggests that TNF-α may be a useful biomarker for disease activity in childhood-onset SLE, however longitudinal studies are necessary to determine if increase of this cytokine may predict flares in childhood-onset SLE.
Collapse
Affiliation(s)
- Mariana Postal
- Department of Medicine, Rheumatology Unit, Faculty of Medical Science, State University of Campinas, Brazil
| | | | | | | | | | | |
Collapse
|
36
|
Abou-Raya A, Abou-Raya S, Helmii M. The effect of vitamin D supplementation on inflammatory and hemostatic markers and disease activity in patients with systemic lupus erythematosus: a randomized placebo-controlled trial. J Rheumatol 2012. [PMID: 23204220 DOI: 10.3899/jrheum.111594] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a chronic multisystem inflammatory autoimmune disease. Vitamin D has potent immunomodulatory properties that support its use in the treatment of autoimmune conditions, including SLE. We assessed vitamin D status in patients with SLE and determined alterations in inflammatory and hemostatic markers and disease activity before and after vitamin D supplementation. METHODS Patients with SLE (n = 267) were randomized 2:1 to receive either oral cholecalciferol 2000 IU/day or placebo for 12 months. Outcome measures included assessment of alterations in levels of proinflammatory cytokines and hemostatic markers, and improvement in disease activity before and after 12 months of supplementation. Disease activity was measured by the SLE Disease Activity Index. Vitamin D levels were measured by Liaison immunoassay (normal 30-100 ng/ml). Serum levels between 10 and 30 ng/ml were classified as vitamin D insufficiency and levels < 10 ng/ml as vitamin D deficiency. RESULTS The mean 25(OH)D level at baseline was 19.8 ng/ml in patients compared to 28.7 ng/ml in controls. The overall prevalence of suboptimal and deficient 25(OH)D serum levels among patients with SLE at baseline was 69% and 39%, respectively. Lower 25(OH)D levels correlated significantly with higher SLE disease activity. At 12 months of therapy, there was a significant improvement in levels of inflammatory and hemostatic markers as well as disease activity in the treatment group compared to the placebo group. CONCLUSION Vitamin D supplementation in patients with SLE is recommended because increased vitamin D levels seem to ameliorate inflammatory and hemostatic markers and show a tendency toward subsequent clinical improvement. Clinical Trial Registry NCT01425775.
Collapse
Affiliation(s)
- Anna Abou-Raya
- Internal Medicine Department, Faculty of Medicine, University of Alexandria, Rheumatology Division, Alexandria, Egypt.
| | | | | |
Collapse
|
37
|
Autoantibodies specific to a peptide of β2-glycoprotein I cross-react with TLR4, inducing a proinflammatory phenotype in endothelial cells and monocytes. Blood 2012; 120:3360-70. [PMID: 22932793 DOI: 10.1182/blood-2011-09-378851] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
β(2)-glycoprotein I (β(2)GPI) is the major antigenic target for antiphospholipid Abs. Anti-β(2)GPI Abs are a heterogeneous population of Igs targeting all domains of the molecule. Abs specific to β(2)GPI domain I are strongly associated with thrombosis and obstetric complications. In the present study, we sought to understand the possible pathogenic mechanism for this subset of anti-β(2)GPI Abs, investigating their potential cross-reactivity with other self-proteins involved in inflammatory or coagulant events. We compared the amino acid sequence of the β(2)GPI domain I with human proteins in a protein databank and identified a peptide sharing 88% identity with an epitope of human TLR4. A high percentage of patients with antiphospholipid syndrome (41%) and systemic lupus erythematosus (50%) presented serum IgG specific to this peptide. Anti-β(2)GPI peptide Abs binding the TLR4 were able to induce NF-κB activation in HEK293 cells that were stably transfected with the TLR4 gene. Anti-β(2)GPI peptide Abs induced activation of TLR4 and triggered interleukin-1 receptor-associated kinase phosphorylation and NF-κB translocation, promoting VCAM expression on endothelial cells and TNF-α release by monocytes. In conclusion, our observations suggest a novel pathogenic mechanism in the TLR4 stimulation by anti-β(2)GPI peptide Abs that links adaptive immune responses with innate immunity in antiphospholipid syndrome and systemic lupus erythematosus.
Collapse
|
38
|
Rana A, Minz RW, Aggarwal R, Anand S, Pasricha N, Singh S. Gene expression of cytokines (TNF-α, IFN-γ), serum profiles of IL-17 and IL-23 in paediatric systemic lupus erythematosus. Lupus 2012; 21:1105-12. [PMID: 22759859 DOI: 10.1177/0961203312451200] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Paediatric systemic lupus erythematosus (pSLE) exhibits an aggressive clinical phenotype and severe complications commonly renal involvement. This could be reflective of the ongoing chronic pro-inflammatory cytokine milieu. We examined relative gene expression of tumour necrosis factor-alpha (TNF-α), interferon-γ (IFN-γ) and serum levels of interleukin-17 (IL-17) and IL-23 and their association with SLEDAI (SLE disease activity index) score and organ manifestations in pSLE. METHODS We enrolled 40 pSLE patients (age 5-16 years, on treatment) and 20 age-matched healthy controls. Relative gene expression levels of IFN-γ and TNF-α in the peripheral blood were determined by quantitative real-time reverse transcriptase polymerase chain reaction (RT-PCR). β actin gene was used for normalization of gene expression. Serum levels of IL-17 and IL-23 were determined by solid phase sandwich ELISA. Statistical analysis were carried out for comparing (Mann-Whitney U test) and correlating data (Univariate, multivariate analysis and Pearson correlation test) with SLEDAI scores and clinical manifestations. RESULTS Over-expression of TNF-α and IFN-γ was found in 90% (36/40) and 80% (32/40) of pSLE patients, respectively. The relative gene expression of TNF-α and IFN-γ were significantly correlated with renal manifestations (p < 0.05). Further, relative expression of IFN-γ gene correlated significantly with skin manifestations and SLEDAI (p < 0.05). Serum levels of IL-17 (766.95 ± 357.83 pg/ml) and IL-23 (135.4 ± 54.23 pg/ml) in pSLE were significantly higher than in controls (IL-17, 172.7 ± 39.19 pg/ml and IL-23, 21.15 ± 10.99 pg/ml) (p < 0.05). Patients with cutaneous (p = 0.002) and haematological involvement (p = 0.003) had high serum IL-17 levels. Serum IL-17 levels correlated with SLEDAI (r = 0.447; p < 0.05). CONCLUSIONS In this preliminary study, we observed a persistent, strong pro-inflammatory cytokine milieu in pSLE patients which reflects ongoing inflammatory damage in different organs. The gene expression profile of these cytokines may be used for assessing organ involvement in pSLE. IL-17 may also serve as a prognostic marker in pSLE. However, longitudinal studies on treatment of naïve patients are required to corroborate these findings.
Collapse
Affiliation(s)
- A Rana
- Department of Immunopathology, Post Graduate Institute of Medical Education and Research, India
| | | | | | | | | | | |
Collapse
|
39
|
Choubey D, Moudgil KD. Interferons in autoimmune and inflammatory diseases: regulation and roles. J Interferon Cytokine Res 2012; 31:857-65. [PMID: 22149411 DOI: 10.1089/jir.2011.0101] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Several lines of evidence strongly implicate type I interferons (IFN-α and β) and IFN-signaling in the pathogenesis of certain autoimmune inflammatory diseases. Accordingly, genome-wide association studies have identified polymorphisms in the type I IFN-signaling pathways. Other studies also indicate that a feed-forward loop of type I IFN production, which involves sensing of cytoplasmic nucleic acids by sensors, contributes to the development of immunopathology. In addition, a mutually positive regulatory feedback loop between type I IFNs and estrogen receptor-α may contribute to a gender bias, thus resulting in an increased production of type I IFNs and associated immunopathology in women. Increased levels of type I IFNs have numerous immunomodulatory functions for both the innate and adaptive immune responses. Given that the IFN-β also has some anti-inflammatory roles, identifying molecular links among certain genotypes, cytokine profiles, and associated phenotypes in patients with autoimmune inflammatory diseases is likely to improve our understanding of autoimmunity-associated pathogenesis and suboptimal outcomes following standard therapies.
Collapse
Affiliation(s)
- Divaker Choubey
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio 45267, USA.
| | | |
Collapse
|
40
|
Tomljenovic L, Shaw CA. Mechanisms of aluminum adjuvant toxicity and autoimmunity in pediatric populations. Lupus 2012; 21:223-30. [DOI: 10.1177/0961203311430221] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immune challenges during early development, including those vaccine-induced, can lead to permanent detrimental alterations of the brain and immune function. Experimental evidence also shows that simultaneous administration of as little as two to three immune adjuvants can overcome genetic resistance to autoimmunity. In some developed countries, by the time children are 4 to 6 years old, they will have received a total of 126 antigenic compounds along with high amounts of aluminum (Al) adjuvants through routine vaccinations. According to the US Food and Drug Administration, safety assessments for vaccines have often not included appropriate toxicity studies because vaccines have not been viewed as inherently toxic. Taken together, these observations raise plausible concerns about the overall safety of current childhood vaccination programs. When assessing adjuvant toxicity in children, several key points ought to be considered: (i) infants and children should not be viewed as “small adults” with regard to toxicological risk as their unique physiology makes them much more vulnerable to toxic insults; (ii) in adult humans Al vaccine adjuvants have been linked to a variety of serious autoimmune and inflammatory conditions (i.e., “ASIA”), yet children are regularly exposed to much higher amounts of Al from vaccines than adults; (iii) it is often assumed that peripheral immune responses do not affect brain function. However, it is now clearly established that there is a bidirectional neuro-immune cross-talk that plays crucial roles in immunoregulation as well as brain function. In turn, perturbations of the neuro-immune axis have been demonstrated in many autoimmune diseases encompassed in “ASIA” and are thought to be driven by a hyperactive immune response; and (iv) the same components of the neuro-immune axis that play key roles in brain development and immune function are heavily targeted by Al adjuvants. In summary, research evidence shows that increasing concerns about current vaccination practices may indeed be warranted. Because children may be most at risk of vaccine-induced complications, a rigorous evaluation of the vaccine-related adverse health impacts in the pediatric population is urgently needed.
Collapse
Affiliation(s)
- L Tomljenovic
- Neural Dynamics Research Group, Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - CA Shaw
- Departments of Ophthalmology and Visual Sciences and Experimental Medicine and the Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
41
|
Seavey MM, Lu LD, Stump KL, Wallace NH, Ruggeri BA. Novel, orally active, proteasome inhibitor, delanzomib (CEP-18770), ameliorates disease symptoms and glomerulonephritis in two preclinical mouse models of SLE. Int Immunopharmacol 2012; 12:257-70. [DOI: 10.1016/j.intimp.2011.11.019] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 10/20/2011] [Accepted: 11/29/2011] [Indexed: 10/14/2022]
|
42
|
Choubey D. DNA-responsive inflammasomes and their regulators in autoimmunity. Clin Immunol 2011; 142:223-31. [PMID: 22245264 DOI: 10.1016/j.clim.2011.12.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 12/08/2011] [Accepted: 12/10/2011] [Indexed: 01/31/2023]
Abstract
Upon sensing microbial and self-derived DNA, DNA sensors initiate innate immune responses. These sensors include the interferon (IFN)-inducible Toll-like receptor 9 (TLR9) and PYHIN proteins. Upon sensing DNA, cytosolic (murine Aim2 and human AIM2) and nuclear (IFI16) PYHIN proteins recruit an adaptor protein (ASC) and pro-caspase-1 to form an inflammasome, which activates caspase-1. The activated caspase-1 cleaves pro-IL-1β and pro-IL-18 to generate active forms. However, upon sensing cytosolic DNA, the IFI16 protein recruits STING to induce the expression of type I IFN. Recognition of self DNA by innate immune cells contributes to the production of increased levels of type I IFN. Given that the type I IFNs modulate the expression of inflammasome proteins and that the IFN-inducible proteins inhibit the activity of DNA-responsive inflammasomes, an improved understanding of the molecular mechanisms that regulate the activity of DNA-responsive inflammasomes is likely to identify new therapeutic targets to treat autoimmune diseases.
Collapse
Affiliation(s)
- Divaker Choubey
- Department of Environmental Health, University of Cincinnati, 3223 Eden Avenue, P. O. Box 670056, Cincinnati, OH 45267, USA.
| |
Collapse
|
43
|
Seavey MM, Dobrzanski P. The many faces of Janus kinase. Biochem Pharmacol 2011; 83:1136-45. [PMID: 22209716 DOI: 10.1016/j.bcp.2011.12.024] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 12/15/2011] [Accepted: 12/16/2011] [Indexed: 12/12/2022]
Abstract
Janus kinases have proved to be essential for many immunological processes but there is growing evidence that they also play a critical role in pathogenesis of many diseases including inflammatory diseases and cancer where they promote multiple steps of tumorigenesis. Several companies are in late stage clinical programs for the development of JAK kinase inhibitors and the first small molecule JAK inhibitor, Jakafi® (ruxolitinib) has been just approved for treatment of myeloproliferative neoplasms. Several other molecules are on the rise to treat arthritis, psoriasis and multiple types of cancer. This commentary will provide a review of the JAK kinase field as it pertains to small molecule inhibition for the treatment of cancer and autoimmune diseases with an emphasis on JAK2. The use of experimental and clinical inhibitors of JAK will be discussed for solid tumor and hematological malignancies, lupus, arthritis, colitis, neurological disorders, pain, diabetes and cardiovascular disease. In addition, it will review current paradigms in the field and treatment programs which could be complemented by small molecule inhibitors of Janus kinase.
Collapse
Affiliation(s)
- Matthew M Seavey
- Cephalon, Inc., Drug Discovery Research, 145 Brandywine Parkway, West Chester, PA 19380, USA.
| | | |
Collapse
|
44
|
Postal M, Appenzeller S. The role of Tumor Necrosis Factor-alpha (TNF-α) in the pathogenesis of systemic lupus erythematosus. Cytokine 2011; 56:537-43. [PMID: 21907587 DOI: 10.1016/j.cyto.2011.08.026] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 07/16/2011] [Accepted: 08/15/2011] [Indexed: 11/17/2022]
Abstract
The Tumor Necrosis Factor-alpha (TNF-α) is a pleiotropic cytokine that produces different stimuli in various physiological and pathological conditions. TNF-α contributes importantly to the development of T cells, B cells, and dendritic cells. However, TNF-α is also a potent inflammatory mediator and apoptosis inducer. The significance of the TNF-α involvement in the pathogenesis of systemic lupus erythematosus (SLE) remains controversial. From the genetic standpoint, a number of studies suggest that the TNF-α gene polymorphism is involved in the susceptibility of SLE. Moreover, there is a close association between the TNF-α gene expression and clinical manifestations. In addition, the increased serum level of TNF-α is observed in SLE patients and associated with disease activity and certain systemic manifestations. Treatment with anti-TNF agents is, however, controversial in SLE since induction of antinuclear antibodies, anti-dsDNA, anticardiolipin antibodies, and cases of drug-induced lupus have been observed in rheumatoid arthritis patients. In this context, this study reviewed the importance of TNF-α in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Mariana Postal
- Department of Medicine, Rheumatology Unit, Faculty of Medical Science, State University of Campinas, Cidade Universitária, Campinas SP, CEP 13083-970, Brazil
| | | |
Collapse
|
45
|
Choubey D, Panchanathan R, Duan X, Liu H, Liu H. Emerging roles for the interferon-inducible p200-family proteins in sex bias in systemic lupus erythematosus. J Interferon Cytokine Res 2011; 31:893-906. [PMID: 21902548 DOI: 10.1089/jir.2011.0073] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease involving multiple organs. The disease is characterized by the production of pathogenic autoantibodies to DNA and certain nuclear antigens, chronic inflammation, and immune dysregulation. Genetic studies involving SLE patients and mouse models have indicated that multiple lupus susceptible genes contribute to the disease phenotype. Notably, the development of SLE in patients and in certain mouse models exhibits a strong sex bias. In addition, several lines of evidence indicates that activation of interferon-α (IFN-α) signaling in immune cells and alterations in the expression of certain immunomodulatory cytokines contribute to lupus pathogenesis. Studies have implicated factors, such as the X chromosomal gene dosage effect and the sex hormones, in gender bias in SLE. However, the molecular mechanisms remain unclear. Additionally, it remains unclear whether these factors influence the "IFN-signature," which is associated with SLE. In this regard, a mutually positive regulatory feedback loop between IFNs and estrogen receptor-α (ERα) has been identified in immune cells. Moreover, studies indicate that the expression of certain IFN-inducible p200-family proteins that act as innate immune sensors for cytosolic DNA is differentially regulated by sex hormones. In this review, we discuss how the modulation of the expression of the p200-family proteins in immune cells by sex hormones and IFNs contributes to sex bias in SLE. An improved understanding of the regulation and roles of the p200-family proteins in immune cells is critical to understand lupus pathogenesis as well as response (or the lack of it) to various therapies.
Collapse
Affiliation(s)
- Divaker Choubey
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio 45267, USA.
| | | | | | | | | |
Collapse
|
46
|
Lu LD, Stump KL, Wallace NH, Dobrzanski P, Serdikoff C, Gingrich DE, Dugan BJ, Angeles TS, Albom MS, Mason JL, Ator MA, Dorsey BD, Ruggeri BA, Seavey MM. Depletion of autoreactive plasma cells and treatment of lupus nephritis in mice using CEP-33779, a novel, orally active, selective inhibitor of JAK2. THE JOURNAL OF IMMUNOLOGY 2011; 187:3840-53. [PMID: 21880982 DOI: 10.4049/jimmunol.1101228] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Accumulating evidence suggests that autoreactive plasma cells play an important role in systemic lupus erythematosus (SLE). In addition, several proinflammatory cytokines promote autoreactive B cell maturation and autoantibody production. Hence, therapeutic targeting of such cytokine pathways using a selective JAK2 inhibitor, CEP-33779 (JAK2 enzyme IC(50) = 1.3 nM; JAK3 enzyme IC(50)/JAK2 enzyme IC(50) = 65-fold), was tested in two mouse models of SLE. Age-matched, MRL/lpr or BWF1 mice with established SLE or lupus nephritis, respectively, were treated orally with CEP-33779 at 30 mg/kg (MRL/lpr), 55 mg/kg or 100 mg/kg (MRL/lpr and BWF1). Studies included reference standard, dexamethasone (1.5 mg/kg; MRL/lpr), and cyclophosphamide (50 mg/kg; MRL/lpr and BWF1). Treatment with CEP-33779 extended survival and reduced splenomegaly/lymphomegaly. Several serum cytokines were significantly decreased upon treatment including IL-12, IL-17A, IFN-α, IL-1β, and TNF-α. Anti-nuclear Abs and frequencies of autoantigen-specific, Ab-secreting cells declined upon CEP-33779 treatment. Increased serum complement levels were associated with reduced renal JAK2 activity, histopathology, and spleen CD138(+) plasma cells. The selective JAK2 inhibitor CEP-33779 was able to mitigate several immune parameters associated with SLE advancement, including the protection and treatment of mice with lupus nephritis. These data support the possibility of using potent, orally active, small-molecule inhibitors of JAK2 to treat the debilitative disease SLE.
Collapse
Affiliation(s)
- Lily D Lu
- Cephalon, Inc., Worldwide Discovery Research, West Chester, PA 19380, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Tomljenovic L, Shaw CA. Do aluminum vaccine adjuvants contribute to the rising prevalence of autism? J Inorg Biochem 2011; 105:1489-99. [PMID: 22099159 DOI: 10.1016/j.jinorgbio.2011.08.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 08/13/2011] [Accepted: 08/14/2011] [Indexed: 12/26/2022]
Abstract
Autism spectrum disorders (ASD) are serious multisystem developmental disorders and an urgent global public health concern. Dysfunctional immunity and impaired brain function are core deficits in ASD. Aluminum (Al), the most commonly used vaccine adjuvant, is a demonstrated neurotoxin and a strong immune stimulator. Hence, adjuvant Al has the potential to induce neuroimmune disorders. When assessing adjuvant toxicity in children, two key points ought to be considered: (i) children should not be viewed as "small adults" as their unique physiology makes them much more vulnerable to toxic insults; and (ii) if exposure to Al from only few vaccines can lead to cognitive impairment and autoimmunity in adults, is it unreasonable to question whether the current pediatric schedules, often containing 18 Al adjuvanted vaccines, are safe for children? By applying Hill's criteria for establishing causality between exposure and outcome we investigated whether exposure to Al from vaccines could be contributing to the rise in ASD prevalence in the Western world. Our results show that: (i) children from countries with the highest ASD prevalence appear to have the highest exposure to Al from vaccines; (ii) the increase in exposure to Al adjuvants significantly correlates with the increase in ASD prevalence in the United States observed over the last two decades (Pearson r=0.92, p<0.0001); and (iii) a significant correlation exists between the amounts of Al administered to preschool children and the current prevalence of ASD in seven Western countries, particularly at 3-4 months of age (Pearson r=0.89-0.94, p=0.0018-0.0248). The application of the Hill's criteria to these data indicates that the correlation between Al in vaccines and ASD may be causal. Because children represent a fraction of the population most at risk for complications following exposure to Al, a more rigorous evaluation of Al adjuvant safety seems warranted.
Collapse
Affiliation(s)
- Lucija Tomljenovic
- Neural Dynamics Research Group, Department of Ophthalmology and Visual Sciences, University of British Columbia, 828 W. 10th Ave, Vancouver, BC, Canada V5Z 1L8.
| | | |
Collapse
|
48
|
Wu X, Gao H, Ke W, Giese RW, Zhu Z. The homeobox transcription factor VentX controls human macrophage terminal differentiation and proinflammatory activation. J Clin Invest 2011; 121:2599-613. [PMID: 21670496 DOI: 10.1172/jci45556] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 04/27/2011] [Indexed: 12/24/2022] Open
Abstract
Macrophages are critical players in both innate and adaptive immunity. While the exogenous signaling events leading to the terminal differentiation of macrophages from monocytes have been studied extensively, the underlying intracellular transcriptional mechanisms remain poorly understood. Here we report that the homeobox transcription factor VentX plays a pivotal role in human macrophage terminal differentiation and proinflammatory function. Our study showed that VentX expression was upregulated upon human primary monocyte-to-macrophage differentiation induced by cytokines such as M-CSF, GM-CSF, and IL-3. Moreover, ablation of VentX expression in primary monocytes profoundly impaired their differentiation to macrophages, and ectopic expression of VentX in a myeloid progenitor cell line triggered its differentiation with prominent macrophage features. Further analysis revealed that VentX was pivotal for the proinflammatory response of terminally differentiated macrophages. Mechanistically, VentX was found to control expression of proteins key to macrophage differentiation and activation, including M-CSF receptor. Importantly, preliminary analysis of gene expression in leukocytes from patients with autoimmune diseases revealed a strong correlation between levels of VentX and those of proinflammatory cytokines. Our results provide mechanistic insight into the crucial roles of VentX in macrophage differentiation and proinflammatory activation and suggest that dysregulation of VentX may play a role in the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
- Xiaoming Wu
- Department of Medicine, Gastroenterology Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
49
|
Takeda N, Nojima T, Terao C, Yukawa N, Kawabata D, Ohmura K, Usui T, Fujii T, Ito Y, Iinuma Y, Mimori T. Interferon-gamma release assay for diagnosing Mycobacterium tuberculosis infections in patients with systemic lupus erythematosus. Lupus 2011; 20:792-800. [DOI: 10.1177/0961203310397966] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Our aim was to analyze the performance of an interferon-gamma release assay, QuantiFERON-TB Gold (QFT-2G), for diagnosing Mycobacterium tuberculosis (MTB) infection in patients with systemic lupus erythematosus (SLE). We performed the QFT-2G and tuberculin skin test (TST) in 71 SLE patients. The QFT-2G results of 279 patients with other connective tissue diseases (CTD) and 35 healthy controls were analyzed. Of the 71 SLE patients, two (2.8%) were positive and 46 (64.8%) were negative by QFT-2G. All SLE patients had no evidence of active MTB infection, apart from one. QFT-2G produced a significantly higher number of indeterminate results in patients with SLE (23/71, 32.4%) compared with those with other CTD (5.7%) or healthy controls (0%) ( p < 0.0001 and p < 0.0001). Decreased lymphocyte counts and high SLEDAI scores in SLE patients were shown to be risk factors for indeterminate results by multivariate analysis ( p = 0.02 and p = 0.04). Among all patients with CTD, SLE itself and lymphocytopenia were found to be independent risks for indeterminate results ( p = 0.00000625 and p = 0.000107). In conclusion, QFT-2G may have more potential to assist in the diagnosis of active and latent MTB infection than TST in SLE patients. However, because of the high frequency of indeterminate results, caution must be used when interpreting the results of QFT-2G among SLE patients, especially those who have parallel or subsequent flares.
Collapse
Affiliation(s)
- N Takeda
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - T Nojima
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - C Terao
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - N Yukawa
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - D Kawabata
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - K Ohmura
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - T Usui
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - T Fujii
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Y Ito
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Y Iinuma
- Department of Clinical Infection Disease, Kanazawa Medical University, Ishikawa, Japan
| | - T Mimori
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
50
|
Yildirim-Toruner C, Diamond B. Current and novel therapeutics in the treatment of systemic lupus erythematosus. J Allergy Clin Immunol 2011; 127:303-12; quiz 313-4. [PMID: 21281862 DOI: 10.1016/j.jaci.2010.12.1087] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Accepted: 12/14/2010] [Indexed: 01/12/2023]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease with significant clinical heterogeneity. Recent advances in our understanding of the genetic, molecular, and cellular bases of autoimmune diseases and especially SLE have led to the application of novel and targeted treatments. Although many treatment modalities are effective in lupus-prone mice, the situation is more complex in human subjects. This article reviews the general approach to the therapy of SLE, focusing on current approved therapies and novel approaches that might be used in the future.
Collapse
Affiliation(s)
- Cagri Yildirim-Toruner
- Department of Pediatrics, Division of Pediatric Rheumatology, Morgan Stanley Children's Hospital of New York Presbyterian, Columbia University Medical Center, New York, New York, USA
| | | |
Collapse
|