1
|
Chen L, Zhang L, Liu B, Liu X, Huang Z, Tang K, Chen P, Jiang X. Exposure-response relationship of mycophenolic acid in pediatric lupus nephritis patients receiving multi-target therapy: An observational cohort study. Semin Arthritis Rheum 2025; 72:152674. [PMID: 40024071 DOI: 10.1016/j.semarthrit.2025.152674] [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: 10/09/2024] [Revised: 02/05/2025] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVE To establish the effectiveness threshold of mycophenolic acid-area under the concentration-time curve between 0 h and 12 h (MPA-AUC0-12h) in pediatric lupus nephritis (LN) patients receiving multi-target therapy. METHODS This observational cohort study enrolled 48 pediatric LN patients treated with mycophenolate mofetil (MMF), tacrolimus, and prednisone. MPA-AUC0-12h was calculated using concentrations based on a limited sampling strategy. Binary logistic regression analysis was employed to investigate factors influencing efficacy. Receiver operating characteristic analysis was conducted to assess MPA-AUC0-12h threshold values. The cumulative incidence of renal remission and inactive systemic lupus erythematosus (SLE) over time was evaluated using Kaplan-Meier analysis. The t-test or Mann-Whitney test was utilized for comparisons between two groups of continuous variables. RESULTS To achieve renal remission, the MPA-AUC0-12h threshold at 6 months was determined to be 25.24 μg·h·mL⁻¹, with an area under the ROC curve (AUC) of 0.83 (P = 0.0002). At 12 months, the MPA-AUC threshold decreased to 23.52 μg·h·mL⁻¹, yielding an AUC of 0.89 (P < 0.0001). For inactive SLE, the MPA-AUC0-12h threshold at 6 months was found to be 31.16 μg·h·mL⁻¹, with an AUC of 0.80 (P = 0.0004), while at 12 months it decreased slightly to 28.87 μg·h·mL⁻¹, resulting in an AUC of 0.82 (P = 0.0012). Patients who reached target thresholds for MPA-AUC0-12h achieved renal response or inactive SLE more rapidly. CONCLUSION There is a significant correlation between MPA-AUC0-12h and treatment response in pediatric LN patients receiving multi-target therapy; therefore, it is recommended that MMF dosing be adjusted according to individual MPA-AUC0-12h levels.
Collapse
Affiliation(s)
- Lizhi Chen
- Department of Pediatric Nephrology and Rheumatology, the First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, Guangzhou, PR China
| | - Lu Zhang
- Department of Pharmacy, the First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, Guangzhou, PR China; Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China; Department of Pharmacy, Zhuhai Center for Maternal and Child Health Care (Zhuhai Women and Children's Hospital), PR China
| | - Baojing Liu
- Department of Pharmacy, the First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, Guangzhou, PR China; Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Xiaohong Liu
- Department of Pediatric Nephrology and Rheumatology, the First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, Guangzhou, PR China
| | - Zhijun Huang
- Department of Pediatric Nephrology and Rheumatology, the First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, Guangzhou, PR China
| | - Kejing Tang
- Department of Pharmacy, the First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, Guangzhou, PR China
| | - Pan Chen
- Department of Pharmacy, the First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, Guangzhou, PR China.
| | - Xiaoyun Jiang
- Department of Pediatric Nephrology and Rheumatology, the First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, Guangzhou, PR China.
| |
Collapse
|
2
|
Zhao Y, Zheng Z, Jin X, Liang S, Zhang C, Zhang M, Lang Y, Li P, Liu Z. Aurora kinase B inhibitor AZD1152: repurposing for treatment of lupus nephritis driven by the results of clinical trials. EBioMedicine 2025; 112:105553. [PMID: 39799765 PMCID: PMC11773216 DOI: 10.1016/j.ebiom.2024.105553] [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: 01/25/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Lupus nephritis (LN) is one of the most common and severe complications of systemic lupus erythematosus (SLE). Multitarget therapy (MT) achieves a 20% higher complete remission (CR) rate compared to conventional therapy in LN management. Intrigued by its excellent clinical efficacy, we aimed to develop a single-agent therapy with comparable efficacy to MT, offering a simplified treatment regimen. METHODS AZD1152, an Aurora kinase B (Aurkb) inhibitor, was identified through transcriptomic analyses and the L1000 CMap drug repurposing database. The therapeutic efficacy of AZD1152 was evaluated in MRL/lpr mice. Transcriptome sequencing and functional assays were performed to elucidate its mechanisms of action. Aurkb expression and its clinical relevance were assessed in lupus-prone mice and patients with LN. FINDINGS AZD1152 significantly attenuated systemic immune activation and renal injury in MRL/lpr mice, demonstrating efficacy comparable to MT regimens in animal studies. AZD1152 treatment modulated immune-inflammatory pathways in the kidney. Aurkb expression was upregulated in T cells infiltrating the renal interstitium in LN. Additionally, Aurkb expression levels positively correlated with the activity index (AI) and serum creatinine (Scr) in patients with LN. Mechanistic studies revealed that AZD1152 exerts therapeutic effects primarily by inhibiting T-cell proliferation. INTERPRETATION This study presents a drug development strategy that integrates clinically validated LN therapies with drug repurposing approaches. This strategy could accelerate drug development and clinical translation processes for LN. FUNDING A full list of funding sources can be found in the acknowledgements section.
Collapse
Affiliation(s)
- Yue Zhao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China
| | - Zuguo Zheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xuexiao Jin
- Institute of Immunology and Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Shaoshan Liang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China
| | - Changming Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China
| | - Yue Lang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China.
| |
Collapse
|
3
|
Tian GQ, Li ZQ. Efficacy and safety of biologics, multitarget therapy, and standard therapy for lupus nephritis: a systematic review and network meta-analysis. Ren Fail 2024; 46:2395451. [PMID: 39212247 PMCID: PMC11370699 DOI: 10.1080/0886022x.2024.2395451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/15/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE This study aimed to compare the efficacy and safety of biologics, multitarget therapy, and standard therapy for the induction of lupus nephritis. METHODS A systematic search of electronic databases (EMBASE, Web of Science, PubMed, Cochrane Library, and ClinicalTrials.gov) was conducted from inception to 30 August 2023. Our study included randomized controlled trials enrolling adult lupus nephritis patients treated with biologics or multitarget therapy, in comparison with standard therapy. The primary outcomes were the rates of complete renal remission (CRR) and serious adverse events (SAE). Stata 15.0 was used to conduct the network meta-analysis. RESULTS Ten randomized controlled trials with a total of 1989 patients met the inclusion criteria. The network meta-analysis indicated that compared with standard therapy, multitarget therapy, obinutuzumab, belimumab, and voclosporin therapy demonstrated superior efficacy in achieving complete renal remission. Among these options, multitarget therapy had the greatest effect (OR = 2.78, 95% CI = 1.81-4.26). Regarding safety, it was observed that there were no significant statistical differences among the various treatment options. Cluster analysis revealed that both obinutuzumab and belimumab exhibited good efficacy and safety. CONCLUSIONS belimumab and obinutuzumab stood out as promising treatments due to their good performance in terms of efficacy and safety. Multitarget therapy may be the most effective approach for treating lupus nephritis. However, since the study population consists exclusively of Asian patients, further research is needed to verify the efficacy of multitarget therapy in lupus nephritis patients of non-Asian descent.
Collapse
Affiliation(s)
- Gui-Qing Tian
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen-Qiong Li
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Xia L, Yang F, Hayashi N, Ma Y, Yan B, Du Y, Chen S, Xia Y, Feng F, Ma Z. Investigation of Nutritional Factors and Malnutrition Risk Prediction Model in Hospitalized Patients with Systemic Lupus Erythematosus in China. J Inflamm Res 2024; 17:8891-8904. [PMID: 39575346 PMCID: PMC11579133 DOI: 10.2147/jir.s486792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
Introduction Nutritional status is a critical indicator of overall health and immune function, significantly influencing treatment outcomes. Despite its importance, the nutritional status of patients with systemic lupus erythematosus (SLE) often receives insufficient attention. This study aims to evaluate the nutritional status of patients with SLE, identify factors associated with malnutrition, and develop a risk prediction model for malnutrition in this population. Methods We collected clinical data from a convenience sample of SLE patients at a general hospital in Ningxia Province, China, between January and December 2022. Univariate and multivariate logistic regression analyses were performed to determine the independent risk factors for malnutrition. A risk prediction model was constructed and evaluated using the receiver operating characteristic (ROC) curve. Results This study included 420 patients with SLE (mean age: 41.43 years, 91.7% women), of whom 46.2% were malnourished based on their serum albumin levels. Multivariate logistic regression analysis identified monthly income (OR=0.192, P<0.05), sleep quality (OR=2.559, P<0.05), kidney involvement (OR=4.269, P<0.05), disease activity (OR=2.743, P<0.05), leukocyte count (OR=1.576, P<0.05), lymphocyte count (OR=0.393, P<0.05), hemoglobin (OR=0.972, P<0.05), complement C3 (OR=0.802, P<0.05), and complement C4 (OR=0.493, P<0.05) as independent risk factors for malnutrition. The prediction model showed good predictive value with an area under the ROC curve of 0.895 (95% CI: 0.823-0.840), sensitivity of 0.907, and specificity of 0.827. The Hosmer-Lemeshow test indicated a good model fit (χ²=10.779, P=0.215). Discussion Malnutrition is a significant concern among SLE patients, influenced by a range of socioeconomic and clinical factors. Our risk prediction model, with its high sensitivity and specificity, provides a robust tool for early identification of malnutrition in this population. Implementing this model in clinical practice can guide healthcare providers in prioritizing at-risk patients, enabling proactive nutritional interventions that could potentially improve clinical outcomes, enhance quality of life, and reduce healthcare costs associated with SLE.
Collapse
Affiliation(s)
- Lijuan Xia
- Department of Radiotherapy, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
- Department of Oncology Nursing and Palliative Care/ Chronic Illness and Conditions Nursing, St. Luke’s International University, Tokyo, Japan
| | - Fanxing Yang
- Department of Labor Delivery Recovery Postpartum, People’s Hospital of Zhengzhou, Zhengzhou, Henan, People’s Republic of China
| | - Naoko Hayashi
- Department of Oncology Nursing and Palliative Care/ Chronic Illness and Conditions Nursing, St. Luke’s International University, Tokyo, Japan
| | - Yuan Ma
- Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
| | - Bin Yan
- Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
| | - Yingxin Du
- Department of Clinical Nutrition, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
| | - Sujuan Chen
- Department of Radiotherapy, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
| | - Yuke Xia
- Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
| | - Fang Feng
- Department of Rheumatology and Immunology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
| | - Zhifang Ma
- Department of Radiotherapy, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People’s Republic of China
| |
Collapse
|
5
|
Cheng Y, Liu L, Ye Y, He Y, Hu W, Ke H, Guo ZY, Shao G. Roles of macrophages in lupus nephritis. Front Pharmacol 2024; 15:1477708. [PMID: 39611168 PMCID: PMC11602334 DOI: 10.3389/fphar.2024.1477708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
LN is a serious complication of systemic lupus erythematosus (SLE), affecting up to 60% of patients with SLE and may lead to end-stage renal disease (ESRD). Macrophages play multifaceted roles in the pathogenesis of LN, including clearance of immune complexes, antigen presentation, regulation of inflammation, and tissue repair. Macrophages are abundant in the glomeruli and tubulointerstitium of LN patients and are positively correlated with serum creatinine levels and the severity of renal pathology. It has been shown that the infiltration of macrophages is closely associated with several clinical indicators, such as serum creatinine and complement C3 levels, anti-dsDNA antibody titers, Austin score, interstitial fibrosis and renal tubular atrophy. Moreover, cytokines expressed by macrophages were upregulated at LN onset and downregulated after remission, suggesting that macrophages may serve as markers of LN pathogenesis and remission. Therapies targeting macrophages have been shown to alleviate LN. There are two main types of macrophages in the kidney: kidney-resident macrophages (KRMs) and monocyte-derived macrophages (MDMs). KRMs and MDMs play different pathological roles in LN, with KRMs promoting leukocyte recruitment at sites of inflammation by expressing monocyte chemokines, while MDMs may exacerbate autoimmune responses by presenting immune complex antigens. Macrophages exhibit high plasticity and can differentiate into various phenotypes in response to distinct environmental stimuli. M1 (proinflammatory) macrophages are linked to the progression of active SLE, whereas the M2 (anti-inflammatory) phenotype is observed during the remission phase of LN. The polarization of macrophages in LN can be manipulated through multiple pathways, such as the modulation of signaling cascades including TLR 2/1, S1P, ERS, metabolic reprogramming, and HMGB1. This paper provides a comprehensive overview of the role of macrophages in the progression of lupus nephritis (LN), and elucidates how these cells and their secretory products function as indicators and therapeutic targets for the disease in the context of diagnosis and treatment of LN.
Collapse
Affiliation(s)
- Yaqian Cheng
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, China
| | - Lulu Liu
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yufei Ye
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yingxue He
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, China
| | - Wenwen Hu
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, China
| | - Haiyan Ke
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, China
| | - Zhi-Yong Guo
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guojian Shao
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, China
| |
Collapse
|
6
|
Zago CCFB, Oliveira BDF, Uehara G, da Silva ALC, Rocha LP, Custódio FB, Araújo LS, da Silva CA, Reis MA, Machado JR. Influence of Podocyte Injury on the Development of Class IV Lupus Nephritis. Int J Nephrol Renovasc Dis 2024; 17:215-225. [PMID: 39381781 PMCID: PMC11460275 DOI: 10.2147/ijnrd.s473616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/27/2024] [Indexed: 10/10/2024] Open
Abstract
Purpose In the kidneys, Systemic Lupus Erythematosus leads to Lupus Nephritis (LN), a form of glomerulonephritis. There is evidence that patients with LN may present activation of specific pathways for podocyte injury. This injury can occur through different mechanisms such as loss of podocyte adhesion to the glomerular basement membrane, cell death or dedifferentiation. Podocyturia with consequent podocytopenia has been described in some nephropathies such as LN, highlighting the importance of studying podocyte injuries in this condition. Evaluating in situ morphological characteristics of podocytes becomes relevant for a better understanding of the processes involved in their pathogenesis. This study investigated podocytes in different classes of LN in renal biopsies performed by the Kidney Research Center at the Federal University of Triângulo Mineiro. Patients and Methods Twenty control cases and 29 biopsy cases diagnosed with LN were selected, divided according to the histopathological classes of the disease. Podocyte density was assessed through immunohistochemistry for Wilms tumor 1 protein and the evaluation of foot process effacement was performed by transmission electron microscopy. Results Podocyte density was lower in the LN and this reduction was observed in all analyzed classes when compared to the control group. More foot process effacement was observed in the LN group, with more effacement in classes I/II and class IV compared to the control group. The class IV group showed more foot process effacement than the class III group and presented higher proteinuria levels compared to the classes I/II group. A strong, positive, and significant correlation was observed between the activity index and foot process effacement in the class IV group. Conclusion Podocytes play an important role in the development of LN, and possibly, injuries to these cells are more closely related to the inflammatory/diffuse proliferative cellular process developed in class IV LN.
Collapse
Affiliation(s)
- Cyntia Camilo Franco Borges Zago
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Bruna de Freitas Oliveira
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Giovanna Uehara
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Ana Laura Carvalho da Silva
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Laura Penna Rocha
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | | | - Liliane Silvano Araújo
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Crislaine Aparecida da Silva
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Marlene Antônia Reis
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Juliana Reis Machado
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| |
Collapse
|
7
|
Zhu C, Liu Y, Xu J, Yang H, Zhao Y, Liu Y. Anti-vasodilator-stimulated phosphoprotein (VASP) antibodies are associated with neuropsychiatric disorders in systemic lupus erythematosus. Heliyon 2024; 10:e37110. [PMID: 39296110 PMCID: PMC11407959 DOI: 10.1016/j.heliyon.2024.e37110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024] Open
Abstract
Background Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by multi-organ involvement and the presence of autoantibodies, pathogenic factors that can serve as diagnostic biomarkers. The current research has been focusing on exploring specific autoantigens with clinical relevance for SLE subtypes. In line with this objective, this study investigated potential antigenic targets associated with specific phenotypes in SLE by leveraging an omics-based approach combined with immunoassay techniques. Methods A transcriptomic analysis was conducted in a cohort of 70 SLE patients to identify genes significantly correlated to the relevant phenotype. Epitope mapping and sequence analysis techniques were used to predict autoantigens, and the corresponding antibodies were subsequently quantified by enzyme-linked immunosorbent assay (ELISA) and validated by Western blot. Results Transcriptomic data analysis revealed a group of hub genes exhibiting a significant correlation with the neuropsychiatric phenotype and a positive relationship with platelets. Subsequent epitope prediction for the corresponding proteins highlighted vasodilator-stimulated phosphoprotein (VASP) as a potential autoantigen. Moreover, ELISA and immunoblotting confirmed that the anti-VASP antibody present in the serum was significantly elevated in SLE patients with neuropsychiatric involvement and positively associated with demyelination. Conclusion VASP harbors autoantigenic epitopes associated with neuropsychiatric phenotype, especially the demyelination symptom in SLE, and its antibodies may serve as promising biomarkers in this disease.
Collapse
Affiliation(s)
- Chenxi Zhu
- Department of Rheumatology and Immunology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiayi Xu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hang Yang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Zhao
- Department of Rheumatology and Immunology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Gowda S, Rana K, Kumar A, Prajapati PK, Patel N, Pandya S, Srivastava R. Extracellular mitochondrial components as new biomarkers for lupus nephritis. Lupus 2024; 33:779-786. [PMID: 38621786 DOI: 10.1177/09612033241247622] [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] [Indexed: 04/17/2024]
Abstract
Major reason for mortality among systemic lupus erythematosus patients is renal failure due to the deposition of immune complexes in the glomeruli. Being a chronic disease with multiple relapses and remissions across the lifespan, it's important to know the degree of nephritis for diagnosis as well as the long-term clinical management of the patients. Currently, renal biopsy is being used as the gold standard to diagnose and define the stages of the disease. However, renal biopsy being invasive only provides a localized picture of nephritis, and has the risk of bleeding. Additionally, it is also cost-intensive. Hence, a reliable, non-invasive biomarker is required for lupus nephritis. This study has evaluated extracellular mitochondrial components, including cell-free mitochondria, and cell-free mitochondrial DNA as probable biomarkers of the degree of nephritis. Both showed a significant correlation with proteinuria and protein-creatinine ratio. Our study substantiates their usage as clinical biomarkers of nephritis upon their validation in a larger cohort of lupus nephritis patients and other forms of nephritis. Although the current data suggest using cell-free mitochondria as a biomarker of lupus nephritis is better than the cell-free mitochondrial DNA.
Collapse
Affiliation(s)
- Sharath Gowda
- Department of Microbiology and Biotechnology Centre, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Khushboo Rana
- Department of Microbiology and Biotechnology Centre, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Ankit Kumar
- Department of Microbiology and Biotechnology Centre, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | | | | | - Sapan Pandya
- V. S. Hospital Medical College, Ahmedabad, India
| | - Ratika Srivastava
- Department of Microbiology and Biotechnology Centre, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| |
Collapse
|
9
|
Shen J, Li F, Han X, Fu D, Xu Y, Zhu C, Liang Z, Tang Z, Zheng R, Hu X, Lin R, Pei Q, Nie J, Luo N, Li X, Chen W, Mao H, Zhou Y, Yu X. Gasdermin D deficiency aborts myeloid calcium influx to drive granulopoiesis in lupus nephritis. Cell Commun Signal 2024; 22:308. [PMID: 38831451 PMCID: PMC11149269 DOI: 10.1186/s12964-024-01681-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
Gasdermin D (GSDMD) is emerging as an important player in autoimmune diseases, but its exact role in lupus nephritis (LN) remains controversial. Here, we identified markedly elevated GSDMD in human and mouse LN kidneys, predominantly in CD11b+ myeloid cells. Global or myeloid-conditional deletion of GSDMD was shown to exacerbate systemic autoimmunity and renal injury in lupus mice with both chronic graft-versus-host (cGVH) disease and nephrotoxic serum (NTS) nephritis. Interestingly, RNA sequencing and flow cytometry revealed that myeloid GSDMD deficiency enhanced granulopoiesis at the hematopoietic sites in LN mice, exhibiting remarkable enrichment of neutrophil-related genes, significant increases in total and immature neutrophils as well as granulocyte/macrophage progenitors (GMPs). GSDMD-deficient GMPs and all-trans-retinoic acid (ATRA)-stimulated human promyelocytes NB4 were further demonstrated to possess enhanced clonogenic and differentiation abilities compared with controls. Mechanistically, GSDMD knockdown promoted self-renewal and granulocyte differentiation by restricting calcium influx, contributing to granulopoiesis. Functionally, GSDMD deficiency led to increased pathogenic neutrophil extracellular traps (NETs) in lupus peripheral blood and bone marrow-derived neutrophils. Taken together, our data establish that GSDMD deletion accelerates LN development by promoting granulopoiesis in a calcium influx-regulated manner, unraveling its unrecognized critical role in LN pathogenesis.
Collapse
Affiliation(s)
- Jiani Shen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Feng Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Xu Han
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Dongying Fu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yiping Xu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Changjian Zhu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Zhou Liang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Ziwen Tang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Ruilin Zheng
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Xinrong Hu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Ruoni Lin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Qiaoqiao Pei
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Jing Nie
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ning Luo
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Xiaoyan Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.
| | - Yi Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.
| | - Xueqing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.
- Department of Nephrology, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China.
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
10
|
Rosetti F, Madera-Salcedo IK, Crispín JC. Relevance of acquired T cell molecular defects in the immunopathogenesis of SLE. Clin Immunol 2024; 263:110225. [PMID: 38642784 DOI: 10.1016/j.clim.2024.110225] [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: 12/14/2023] [Revised: 03/07/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
Systemic lupus erythematosus (SLE) and other autoimmune diseases are thought to develop in genetically predisposed individuals when triggered by environmental factors. This paradigm does not fully explain disease development, as it fails to consider the delay between birth and disease expression. In this review, we discuss observations described in T cells from patients with SLE that are not related to hereditary factors and have therefore been considered secondary to the disease process itself. Here, we contextualize some of those observations and argue that they may represent a pathogenic layer between genetic factors and disease development. Acquired changes in T cell phenotype and function in the setting of SLE may affect the immune system, creating a predisposition towards a more inflammatory and pathogenic system that amplifies autoimmunity and facilitates disease development.
Collapse
Affiliation(s)
- Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Mexico City 14080, Mexico
| | - Iris K Madera-Salcedo
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Mexico City 14080, Mexico
| | - José C Crispín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Mexico City 14080, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Eugenio Garza Sada 2501, Monterrey, N.L. 64849, Mexico.
| |
Collapse
|
11
|
Amo L, Kole HK, Scott B, Borrego F, Qi CF, Wang H, Bolland S. Purification and analysis of kidney-infiltrating leukocytes in a mouse model of lupus nephritis. Methods Cell Biol 2024; 188:131-152. [PMID: 38880521 DOI: 10.1016/bs.mcb.2024.03.007] [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] [Indexed: 06/18/2024]
Abstract
Renal injury often occurs as a complication in autoimmune diseases such as systemic lupus erythematosus (SLE). It is estimated that a minimum of 20% SLE patients develop lupus nephritis, a condition that can be fatal when the pathology progresses to end-stage renal disease. Studies in animal models showed that incidence of immune cell infiltrates in the kidney was linked to pathological injury and correlated with severe lupus nephritis. Thus, preventing immune cell infiltration into the kidney is a potential approach to impede the progression to an end-stage disease. A requirement to investigate the role of kidney-infiltrating leukocytes is the development of reproducible and efficient protocols for purification and characterization of immune cells in kidney samples. This chapter describes a detailed methodology that discriminates tissue-resident leukocytes from blood-circulating cells that are found in kidney. Our protocol was designed to maximize cell viability and to reduce variability among samples, with a combination of intravascular staining and magnetic bead separation for leukocyte enrichment. Experiments included as example were performed with FcγRIIb[KO] mice, a well-characterized murine model of SLE. We identified T cells and macrophages as the primary leukocyte subsets infiltrating into the kidney during severe nephritis, and we extensively characterized them phenotypically by flow cytometry.
Collapse
Affiliation(s)
- Laura Amo
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| | - Hemanta K Kole
- Laboratory of Immunogenetics, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, United States
| | - Bethany Scott
- Laboratory of Immunogenetics, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, United States
| | - Francisco Borrego
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Chen-Feng Qi
- Laboratory of Immunogenetics, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, United States
| | - Hongsheng Wang
- Laboratory of Immunogenetics, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, United States
| | - Silvia Bolland
- Laboratory of Immunogenetics, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD, United States
| |
Collapse
|
12
|
Ichinose K. The role of podocytes in lupus nephritis: Insights and implications. Clin Immunol 2024; 262:110180. [PMID: 38462157 DOI: 10.1016/j.clim.2024.110180] [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: 01/01/2024] [Revised: 02/09/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024]
Abstract
Lupus nephritis (LN) is a severe complication of systemic lupus erythematosus, with high mortality rates despite medical advancements. The complexity of its pathogenesis, including the pivotal role of podocytes - kidney-localized cells - remains a challenge, lacking effective treatments and biomarkers. Recent studies highlight the significant contribution of these cells to LN's development, particularly through their immune-related functions and interaction with other kidney cells. This new understanding opens possibilities for targeted therapies aimed at these cellular mechanisms. This review aims to summarize these recent developments, shedding light on the intricate involvement of podocytes in LN and potential avenues for innovative treatments.
Collapse
Affiliation(s)
- Kunihiro Ichinose
- Department of Rheumatology, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo 693-8501, Japan; Integrated Kidney Research and Advance, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo 693-8501, Japan.
| |
Collapse
|
13
|
Wang CM, Jan Wu YJ, Zheng JW, Huang LY, Tan KP, Chen JY. T cell expressions of aberrant gene signatures and Co-inhibitory receptors (Co-IRs) as predictors of renal damage and lupus disease activity. J Biomed Sci 2024; 31:41. [PMID: 38650001 PMCID: PMC11034032 DOI: 10.1186/s12929-024-01024-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 04/01/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is distinguished by an extensive range of clinical heterogeneity with unpredictable disease flares and organ damage. This research investigates the potential of aberrant signatures on T cell genes, soluble Co-IRs/ligands, and Co-IRs expression on T cells as biomarkers for lupus disease parameters. METHODS Comparative transcriptome profiling analysis of non-renal and end-stage renal disease (ESRD) phenotypes of SLE was performed using CD4 + and CD8 + cDNA microarrays of sorted T cells. Comparing the expression of Co-IRs on T cells and serum soluble mediators among healthy and SLE phenotypes. RESULTS SLE patients with ESRD were downregulated CD38, PLEK, interferon-γ, CX3CR1, FGFBP2, and SLCO4C1 transcripts on CD4 + and CD8 + T cells simultaneously and NKG7, FCRL6, GZMB/H, FcγRIII, ITGAM, Fas ligand, TBX21, LYN, granulysin, CCL4L1, CMKLR1, HLA-DRβ, KIR2DL3, and KLRD1 in CD8 T cells. Pathway enrichment and PPI network analyses revealed that the overwhelming majority of Differentially Expressed Genes (DEGs) have been affiliated with novel cytotoxic, antigen presentation, and chemokine-cell migration signature pathways. CD8 + GZMK + T cells that are varied in nature, including CD161 + Mucosal-associated invariant T (MAIT) cells and CD161- aged-associated T (Taa) cells and CD161-GZMK + GZMB + T cells might account for a higher level of GZMK in CD8 + T cells associated with ESRD. SLE patients have higher TIGIT + , PD1 + , and lower CD127 + cell percentages on CD4 + T cells, higher TIM3 + , TIGIT + , HLA-DR + cell frequency, and lower MFI expression of CD127, CD160 in CD8 T cells. Co-IRs expression in T cells was correlated with soluble PD-1, PDL-2, and TIM3 levels, as well as SLE disease activity, clinical phenotypes, and immune-therapy responses. CONCLUSION The signature of dysfunctional pathways defines a distinct immunity pattern in LN ESRD patients. Expression levels of Co-IRs in peripheral blood T cells and serum levels of soluble PD1/PDL-2/TIM3 can serve as biomarkers for evaluating clinical parameters and therapeutic responses.
Collapse
Affiliation(s)
- Chin-Man Wang
- Department of Rehabilitation, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin St. Kwei-Shan, Taoyuan, Republic of China
| | - Yeong-Jian Jan Wu
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan, No. 5, Fu-Shin St. Kwei-Shan, Republic of China
| | - Jian-Wen Zheng
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan, No. 5, Fu-Shin St. Kwei-Shan, Republic of China
| | - Li Yu Huang
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan, No. 5, Fu-Shin St. Kwei-Shan, Republic of China
| | - Keng Poo Tan
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan, No. 5, Fu-Shin St. Kwei-Shan, Republic of China
| | - Ji-Yih Chen
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan, No. 5, Fu-Shin St. Kwei-Shan, Republic of China.
| |
Collapse
|
14
|
Li F, Liang Z, Zhong H, Hu X, Tang Z, Zhu C, Shen J, Han X, Lin R, Zheng R, Tang R, Peng H, Zheng X, Mo C, Chen P, Wang X, Wen Q, Li J, Xia X, Ye H, Qiu Y, Yu J, Fu D, Liu J, Wang R, Xie H, Guo Y, Li X, Fan J, Liu Q, Mao H, Chen W, Zhou Y. Group 3 Innate Lymphoid Cells Exacerbate Lupus Nephritis by Promoting B Cell Activation in Kidney Ectopic Lymphoid Structures. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302804. [PMID: 37915129 PMCID: PMC10724443 DOI: 10.1002/advs.202302804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/28/2023] [Indexed: 11/03/2023]
Abstract
Group 3 innate lymphoid cells (ILC3s) represent a new population in immune regulation, yet their role in lupus nephritis (LN) remains elusive. In the present work, systemic increases in ILC3s, particularly in the kidney, are observed to correlate strongly with disease severity in both human and murine LN. Using MRL/lpr lupus mice and a nephrotoxic serum-induced LN model, this study demonstrates that ILC3s accumulated in the kidney migrate predominantly from the intestine. Furthermore, intestinal ILC3s accelerate LN progression, manifested by exacerbated autoimmunity and kidney injuries. In LN kidneys, ILC3s are located adjacent to B cells within ectopic lymphoid structures (ELS), directly activating B cell differentiation into plasma cells and antibody production in a Delta-like1 (DLL1)/Notch-dependent manner. Blocking DLL1 attenuates ILC3s' effects and protects against LN. Altogether, these findings reveal a novel pathogenic role of ILC3s in B cell activation, renal ELS formation and autoimmune injuries during LN, shedding light on the therapeutic value of targeting ILC3s for LN.
Collapse
Affiliation(s)
- Feng Li
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Zhou Liang
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Haojie Zhong
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital, Shenzhen UniversityShenzhen518000China
| | - Xinrong Hu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Ziwen Tang
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Changjian Zhu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Jiani Shen
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Xu Han
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Ruoni Lin
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Ruilin Zheng
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Ruihan Tang
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Huajing Peng
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Xunhua Zheng
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Chengqiang Mo
- Department of UrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
| | - Peisong Chen
- Department of Laboratory MedicineThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
| | - Xin Wang
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Qiong Wen
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Jianbo Li
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Xi Xia
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Hongjian Ye
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Yagui Qiu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Jianwen Yu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Dongying Fu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Jiaqi Liu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Rong Wang
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Huixin Xie
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Yun Guo
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Xiaoyan Li
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Jinjin Fan
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Qinghua Liu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Haiping Mao
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Wei Chen
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Yi Zhou
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| |
Collapse
|
15
|
Li F, Wang X, Shi J, Wu S, Xing W, He Y. Anti-inflammatory effect of dental pulp stem cells. Front Immunol 2023; 14:1284868. [PMID: 38077342 PMCID: PMC10701738 DOI: 10.3389/fimmu.2023.1284868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Dental pulp stem cells (DPSCs) have received a lot of attention as a regenerative medicine tool with strong immunomodulatory capabilities. The excessive inflammatory response involves a variety of immune cells, cytokines, and has a considerable impact on tissue regeneration. The use of DPSCs for controlling inflammation for the purpose of treating inflammation-related diseases and autoimmune disorders such as supraspinal nerve inflammation, inflammation of the pulmonary airways, systemic lupus erythematosus, and diabetes mellitus is likely to be safer and more regenerative than traditional medicines. The mechanism of the anti-inflammatory and immunomodulatory effects of DPSCs is relatively complex, and it may be that they themselves or some of the substances they secrete regulate a variety of immune cells through inflammatory immune-related signaling pathways. Most of the current studies are still at the laboratory cellular level and animal model level, and it is believed that through the efforts of more researchers, DPSCs/SHED are expected to be transformed into excellent drugs for the clinical treatment of related diseases.
Collapse
Affiliation(s)
- FenYao Li
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - XinXin Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Jin Shi
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - ShuTing Wu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - WenBo Xing
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Pennesi M, Benvenuto S. Lupus Nephritis in Children: Novel Perspectives. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1841. [PMID: 37893559 PMCID: PMC10607957 DOI: 10.3390/medicina59101841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023]
Abstract
Childhood-onset systemic lupus erythematosus is an inflammatory and autoimmune condition characterized by heterogeneous multisystem involvement and a chronic course with unpredictable flares. Kidney involvement, commonly called lupus nephritis, mainly presents with immune complex-mediated glomerulonephritis and is more frequent and severe in adults. Despite a considerable improvement in long-term renal prognosis, children and adolescents with lupus nephritis still experience significant morbidity and mortality. Moreover, current literature often lacks pediatric-specific data, leading clinicians to rely exclusively on adult therapeutic approaches. This review aims to describe pediatric lupus nephritis and provide an overview of the novel perspectives on the pathogenetic mechanisms, histopathological classification, therapeutic approach, novel biomarkers, and follow-up targets in children and adolescents with lupus nephritis.
Collapse
Affiliation(s)
- Marco Pennesi
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, 34137 Trieste, Italy
| | - Simone Benvenuto
- Department of Medicine, Surgery, and Health Sciences, University of Trieste, 34127 Trieste, Italy
| |
Collapse
|
17
|
Liu S, Zhou W. Research progress in functional magnetic resonance imaging assessment of lupus nephritis kidney injury. Lupus 2023; 32:1143-1154. [PMID: 37556364 DOI: 10.1177/09612033231193790] [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] [Indexed: 08/11/2023]
Abstract
Lupus nephritis is one of the most common and severe complications of systemic lupus erythematosus and is also a major predictor of poor prognosis and mortality. Lupus nephritis has the characteristics of insidious onset, complex pathological types, rapid progression of organ damage, and easy recurrence. Currently, kidney damage in lupus nephritis is usually assessed based on urine analysis, renal biopsy, and glomerular filtration rates. However, they all have certain limitations, making it difficult to diagnose lupus nephritis early and assess its severity and progression. With the rapid development of functional magnetic resonance, multiple functional imaging techniques are expected to provide more useful information for the pathophysiological development, early diagnosis, progression, prognosis, and renal function evaluation of lupus nephritis. This article reviews the principle of multiple functional magnetic resonance imaging and the research status of evaluating renal function in lupus nephritis.
Collapse
Affiliation(s)
- Shuangjiao Liu
- Department of Radiology, YueYang Central Hospital, Yueyang, China
| | - Wenming Zhou
- Department of Radiology, YueYang Central Hospital, Yueyang, China
| |
Collapse
|
18
|
Keskinyan VS, Lattanza B, Reid-Adam J. Glomerulonephritis. Pediatr Rev 2023; 44:498-512. [PMID: 37653138 DOI: 10.1542/pir.2021-005259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Glomerulonephritis (GN) encompasses several disorders that cause glomerular inflammation and injury through an interplay of immune-mediated mechanisms, host characteristics, and environmental triggers, such as infections. GN can manifest solely in the kidney or in the setting of a systemic illness, and presentation can range from chronic and relatively asymptomatic hematuria to fulminant renal failure. Classic acute GN is characterized by hematuria, edema, and hypertension, the latter 2 of which are the consequence of sodium and water retention in the setting of renal impairment. Although presenting signs and symptoms and a compatible clinical history can suggest GN, serologic and urinary testing can further refine the differential diagnosis, and renal biopsy can be used for definitive diagnosis. Treatment of GN can include supportive care, renin-angiotensin-aldosterone system blockade, immunomodulatory therapy, and renal transplant. Prognosis is largely dependent on the underlying cause of GN and can vary from a self-limited course to chronic kidney disease. This review focuses on lupus nephritis, IgA nephropathy, IgA vasculitis, and postinfectious GN.
Collapse
|
19
|
Cheng C, Zhu R, Liu M, Yang H, Guo F, Du Q, Wang X, Li M, Song G, Qin R, Liu S. Kunxian capsule alleviates renal damage by inhibiting the JAK1/STAT1 pathway in lupus nephritis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 310:116349. [PMID: 36924861 DOI: 10.1016/j.jep.2023.116349] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/20/2023] [Accepted: 03/01/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kunxian capsule (KXC) is a new traditional Chinese medicine drug included in "The key science and technology achievements" in the Ninth Five Year Plan of China. KXC has been clinically used for more than 10 years in the treatment of lupus nephritis (LN). However, the underlying role and molecular mechanism of KXC in LN remain unclear. AIM OF THE STUDY This study aimed to explore the efficacy and potential mechanisms of KXC through pharmacological network, in vitro and in vivo studies. MATERIALS AND METHODS Pharmacological network analysis of KXC treatment in LN was performed using data acquired from the Traditional Chinese Medicine System Pharmacology Database and Analysis Platform (TCMSP, https://old.tcmsp-e.com/tcmsp.php) and NCBI Gene Expression Omnibus (GEO, https://www.ncbi.nlm.nih.gov/geo/database). HK-2 cells were chosen as an in vitro model of the tubular immune response by simulation with interferon γ (IFN-γ). MRL/lpr mice were used to explore the mechanism of KXC in vivo. Finally, the specific active molecules of KXC were further analyzed by molecular docking. RESULTS The pharmacological network analysis showed that STAT1 is a key factor in the effects of KXC. In vitro and in vivo experiments confirmed the therapeutic effect of KXC on LN renal function and tubular inflammation. The protective effect of KXC is mediated by STAT1 blockade, which further reduces T-cell infiltration and improves the renal microenvironment in LN. Two main components of KXC, Tripterygium hypoglaucum (H.Lév.) Hutch (Shanhaitang) and Epimedium brevicornu Maxim (Yinyanghuo) could block JAK1-STAT1 activation. Furthermore, we found 8 molecules that could bind to the ATP pocket of JAK1 with high affinities by performing docking analysis. CONCLUSIONS KXC inhibits renal damage and T-cell infiltration in LN by blocking the JAK1-STAT1 pathway.
Collapse
Affiliation(s)
- Chen Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Rongrong Zhu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mingjian Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, China
| | - Hao Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fangfang Guo
- Center of Clinical Laboratory, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Qunqun Du
- Baiyunshan Chenliji Pharmaceutical Co., Ltd, Guangzhou, 510288, China
| | - Xiaolan Wang
- Baiyunshan Chenliji Pharmaceutical Co., Ltd, Guangzhou, 510288, China
| | - Minmin Li
- Center of Clinical Laboratory, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Gaopeng Song
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, China.
| | - Renan Qin
- Baiyunshan Chenliji Pharmaceutical Co., Ltd, Guangzhou, 510288, China.
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
20
|
Lin Z, Yu N, Cheng C, Jin B, Zhang Q, Zhuang H, Jiang X. Serum levels and significance of soluble B-cell maturation antigen in childhood-onset systemic lupus erythematosus with renal involvement. Lupus 2023; 32:680-687. [PMID: 36914971 DOI: 10.1177/09612033231164633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
OBJECTIVE The aim of this study was to investigate serum levels of soluble B-cell maturation antigen (sBCMA) in childhood-onset systemic lupus erythematous (cSLE) patients with renal involvement, and to elucidate their association with clinical characteristics. METHODS 116 cases of cSLE patients with renal involvement (84 females and 32 males; median age 11.6 (10.1, 12.9) years) hospitalized in Department of Pediatric Nephrology and Rheumatology, the First Affiliated Hospital, Sun Yat-sen University and 31 healthy controls (HCs) were enrolled. Serum concentrations of sBCMA were determined using enzyme-linked immunosorbent assay (ELISA). Clinical and laboratory information of cSLE patients were retrospectively analyzed. RESULTS Serum sBCMA levels were significantly increased in primary cSLE when compared with treated cSLE patients and HCs, whereas there was no significant difference between treated cSLE patients and HCs. Patients with high disease activity displayed higher serum sBCMA levels compared with those with no or mild to moderate disease activity. Positive correlation was observed between serum sBCMA levels and systemic lupus erythematosus disease activity index-2K (SLEDAI-2K), antinuclear antibody titers, anti-double-stranded DNA titers, erythrocyte sedimentation rate, and immunoglobulin G levels, while sBCMA levels were negatively correlated with blood white blood cell count, hemoglobin, platelet count, complement C3 and C4 levels. Serum sBCMA levels decreased as disease ameliorated after treatments among 11 cases with follow-up examinations. CONCLUSIONS In cSLE patients with renal involvement, serum sBCMA levels correlated significantly with disease activity, immunological, and hematological parameters, but not with renal parameters. Our results suggest the potential and significance of serum sBCMA as a biomarker in cSLE patients.
Collapse
Affiliation(s)
- Zhilang Lin
- Department of Pediatric Nephrology and Rheumatology, 71068Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Nannan Yu
- Department of Pediatric Nephrology and Rheumatology, 71068Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Cheng Cheng
- Department of Pediatric Nephrology and Rheumatology, 71068Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Bei Jin
- Department of Pediatric Nephrology and Rheumatology, 71068Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Qiufang Zhang
- Department of Pediatric Nephrology and Rheumatology, 71068Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Hongjie Zhuang
- Department of Pediatric Nephrology and Rheumatology, 71068Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Xiaoyun Jiang
- Department of Pediatric Nephrology and Rheumatology, 71068Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| |
Collapse
|
21
|
Roointan A, Gholaminejad A, Shojaie B, Hudkins KL, Gheisari Y. Candidate MicroRNA Biomarkers in Lupus Nephritis: A Meta-analysis of Profiling Studies in Kidney, Blood and Urine Samples. Mol Diagn Ther 2023; 27:141-158. [PMID: 36520403 DOI: 10.1007/s40291-022-00627-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2022] [Indexed: 12/16/2022]
Abstract
CONTEXT Lupus nephritis (LN) is a kidney disease caused by systemic lupus erythematosus in which kidneys are attacked by the immune system. So far, various investigations have reported altered miRNA expression profiles in LN patients and different miRNAs have been introduced as biomarkers and/or therapeutic targets in LN. The aim of this study was to introduce a consensus panel of potential miRNA biomarkers by performing a meta-analysis of miRNA profiles in the LN patients. MATERIALS AND METHODS A comprehensive literature review approach was performed to find LN-related miRNA expression profiles in renal tissues, blood, and urine samples. After selecting the eligible studies and performing the data extraction, meta-analysis was done based on the vote-counting rank strategy as well as meta-analysis of p-values. The meta-miRNAs and their related genes were subjected to functional enrichment analyses and network construction. RESULTS The results of the meta-analysis of 41 studies were three lists of consensus miRNAs with altered expression profiles in the various tissue samples of LN patients (meta-analysis of p-values < 0.05). Of the 13 studies on kidney tissue, the meta-miRNAs were let-7a, miR-198, let-7e, miR-145, and miR-26a. In addition, meta-miRNAs of miR-199a, miR-21, miR-423, miR-1260b, miR-589, miR-150, miR-155, miR-146a, and miR-183 from 21 studies on blood samples, and miR-146a, miR-204, miR-30c, miR-3201, and miR-1273e from 11 studies on urine samples can be considered as non-invasive biomarker panels for LN. Functional enrichment analysis on the meta-miRNA lists confirmed the involvement of their target genes in nephropathy-related signaling pathways. CONCLUSION Using a meta-analytical approach, our study proposes three meta-miRNA panels that could be the target of further research to assess their potential as therapeutic targets/biomarkers in LN disease.
Collapse
Affiliation(s)
- Amir Roointan
- Faculty of Medicine, Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Hezar jarib St, Isfahan, 81746-73461, Iran
| | - Alieh Gholaminejad
- Faculty of Medicine, Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Hezar jarib St, Isfahan, 81746-73461, Iran.
| | - Behrokh Shojaie
- Faculty of Medicine, Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Hezar jarib St, Isfahan, 81746-73461, Iran
| | - Kelly L Hudkins
- Department of Pathology, School of Medicine, University of Washington, Seattle, USA
| | - Yousof Gheisari
- Faculty of Medicine, Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Hezar jarib St, Isfahan, 81746-73461, Iran
| |
Collapse
|
22
|
Favor OK, Chauhan PS, Pourmand E, Edwards AM, Wagner JG, Lewandowski RP, Heine LK, Harkema JR, Lee KSS, Pestka JJ. Lipidome modulation by dietary omega-3 polyunsaturated fatty acid supplementation or selective soluble epoxide hydrolase inhibition suppresses rough LPS-accelerated glomerulonephritis in lupus-prone mice. Front Immunol 2023; 14:1124910. [PMID: 36875087 PMCID: PMC9978350 DOI: 10.3389/fimmu.2023.1124910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/17/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Lipopolysaccharide (LPS)-accelerated autoimmune glomerulonephritis (GN) in NZBWF1 mice is a preclinical model potentially applicable for investigating lipidome-modulating interventions against lupus. LPS can be expressed as one of two chemotypes: smooth LPS (S-LPS) or rough LPS (R-LPS) which is devoid of O-antigen polysaccharide sidechain. Since these chemotypes differentially affect toll-like receptor 4 (TLR4)-mediated immune cell responses, these differences may influence GN induction. Methods We initially compared the effects of subchronic intraperitoneal (i.p.) injection for 5 wk with 1) Salmonella S-LPS, 2) Salmonella R-LPS, or 3) saline vehicle (VEH) (Study 1) in female NZBWF1 mice. Based on the efficacy of R-LPS in inducing GN, we next used it to compare the impact of two lipidome-modulating interventions, ω-3 polyunsaturated fatty acid (PUFA) supplementation and soluble epoxide hydrolase (sEH) inhibition, on GN (Study 2). Specifically, effects of consuming ω-3 docosahexaenoic acid (DHA) (10 g/kg diet) and/or the sEH inhibitor 1-(4-trifluoro-methoxy-phenyl)-3-(1-propionylpiperidin-4-yl) urea (TPPU) (22.5 mg/kg diet ≈ 3 mg/kg/day) on R-LPS triggering were compared. Results In Study 1, R-LPS induced robust elevations in blood urea nitrogen, proteinuria, and hematuria that were not evident in VEH- or S-LPS-treated mice. R-LPS-treated mice further exhibited kidney histopathology including robust hypertrophy, hyperplasia, thickened membranes, lymphocytic accumulation containing B and T cells, and glomerular IgG deposition consistent with GN that was not evident in VEH- or SLPS-treated groups. R-LPS but not S-LPS induced spleen enlargement with lymphoid hyperplasia and inflammatory cell recruitment in the liver. In Study 2, resultant blood fatty acid profiles and epoxy fatty acid concentrations reflected the anticipated DHA- and TPPU-mediated lipidome changes, respectively. The relative rank order of R-LPS-induced GN severity among groups fed experimental diets based on proteinuria, hematuria, histopathologic scoring, and glomerular IgG deposition was: VEH/CON< R-LPS/DHA ≈ R-LPS/TPPU<<< R-LPS/TPPU+DHA ≈ R-LPS/CON. In contrast, these interventions had modest-to- negligible effects on R-LPS-induced splenomegaly, plasma antibody responses, liver inflammation, and inflammation-associated kidney gene expression. Discussion We show for the first time that absence of O-antigenic polysaccharide in R-LPS is critical to accelerated GN in lupus-prone mice. Furthermore, intervention by lipidome modulation through DHA feeding or sEH inhibition suppressed R-LPS-induced GN; however, these ameliorative effects were greatly diminished upon combining the treatments.
Collapse
Affiliation(s)
- Olivia K. Favor
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Preeti S. Chauhan
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Elham Pourmand
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - Angel M. Edwards
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - James G. Wagner
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - Ryan P. Lewandowski
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - Lauren K. Heine
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Jack R. Harkema
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - Kin Sing Stephen Lee
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - James J. Pestka
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
23
|
Tampe D, Hakroush S, Tampe B. Molecular signatures of intrarenal complement receptors C3AR1 and C5AR1 correlate with renal outcome in human lupus nephritis. Lupus Sci Med 2022; 9:9/1/e000831. [PMID: 36521939 PMCID: PMC9756185 DOI: 10.1136/lupus-2022-000831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Lupus nephritis is one of the most common and serious complications of systemic lupus erythematosus (SLE). Lupus nephritis is a major cause of kidney failure in patients with SLE, attributed to increased morbidity and mortality. The in situ deposition of intrarenal immune complexes promotes the accumulation of inflammatory cells and causes kidney injury. METHODS We here extracted transcriptome array datasets for expression of complement molecules in human lupus nephritis. Furthermore, we performed gene set enrichment analysis to identify molecular signatures associated with follow-up kidney function in lupus nephritis. RESULTS Within the glomerular compartment, intrarenal mRNA expression levels of C3AR1 (p=0.0333) and C5AR1 (p=0.0167) correlated with treatment success reflected by kidney function recovery specifically in class III lupus nephritis, while no such association was observed in class II or class IV lupus nephritis. Interestingly, mRNA expression levels of either glomerular C3AR1 or C5AR1 resulted in identical gene set and signalling pathways enrichments in human lupus nephritis, including interferon signalling and signalling by interleukins. Direct comparison of C3AR1 and C5AR1 confirmed a strong association between glomerular mRNA expression levels of both complement receptors (r=0.8955, p<0.0001). CONCLUSIONS This study provides additional insights into signalling pathways associated with intrarenal synthesis of complement components in lupus nephritis that might be also affected by targeted therapy of the complement system. These results require confirmation but may contribute to a personalised treatment approach in distinct classes of human lupus nephritis.
Collapse
Affiliation(s)
- Désirée Tampe
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - Samy Hakroush
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Björn Tampe
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
24
|
Yu C, Li P, Dang X, Zhang X, Mao Y, Chen X. Lupus nephritis: new progress in diagnosis and treatment. J Autoimmun 2022; 132:102871. [PMID: 35999111 DOI: 10.1016/j.jaut.2022.102871] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 02/07/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic multifactorial autoimmune disease that affects many organs, including the kidney. Lupus nephritis (LN) is a common manifestation characterized by heterogeneous clinical and histopathological findings, and often associates with poor prognosis. The diagnosis and treatment of LN is challenging, depending largely on renal biopsy, and there is no reliable non-invasive LN biomarker. Up to now, the complete remission rate of LN is only 20%∼30% after receiving six months of standard treatment, which is far from satisfactory. Moreover, adverse reactions to immunosuppressants, especially glucocorticoids, further compromise the prognosis of LN. Biological reagents targetting autoimmune responses and inflammatory pathways, bring hope to the treatment of intractable lupus. The European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) and KDIGO (Kidney Disease: Improving Global Outcomes) have been working on and launched the recommendations for the management of LN. In this review, we update our knowledge in the pathogenesis, diagnosis, and management of LN and prospect for the future potential targets in the management of LN.
Collapse
Affiliation(s)
- Chen Yu
- Department of Nephrology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xin Dang
- Department of Nephrology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yonghui Mao
- Department of Nephrology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China.
| |
Collapse
|
25
|
Mei X, Jin H, Zhao M, Lu Q. Association of Immune-Related Genetic and Epigenetic Alterations with Lupus Nephritis. KIDNEY DISEASES (BASEL, SWITZERLAND) 2022; 8:286-296. [PMID: 36157263 PMCID: PMC9386430 DOI: 10.1159/000524937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 05/02/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The familial clustering phenomenon together with environmental influences indicates the presence of a genetic and epigenetic predisposition to systematic lupus erythematosus (SLE). Interestingly, regarding lupus nephritis (LN), the worst complication of SLE, mortality, and morbidity were not consistent with SLE in relation to sexuality and ethnicity. SUMMARY Genetic and epigenetic alterations in LN include genes and noncoding RNAs that are involved in antigen-presenting, complements, immune cell infiltration, interferon pathways, and so on. Once genetic or epigenetic change occurs alone or simultaneously, they will promote the formation of immune complexes with autoantibodies that target various autoantigens, which results in inflammatory cytokines and autoreactive immune cells colonizing renal tissues and contributing to LN. KEY MESSAGES Making additional checks for immunopathology-related heredity and epigenetic factors may lead to a more holistic perspective of LN.
Collapse
Affiliation(s)
- Xiaole Mei
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immunological Dermatology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| | - Hui Jin
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| | - Ming Zhao
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immunological Dermatology, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| |
Collapse
|
26
|
Jiang B, Zhang Y, Li Y, Chen Y, Sha S, Zhao L, Li D, Wen J, Lan J, Lou Y, Su H, Zhang C, Zhu J, Tao J. A Tissue-Tended Mycophenolate-Modified Nanoparticle Alleviates Systemic Lupus Erythematosus in MRL/Lpr Mouse Model Mainly by Promoting Local M2-Like Macrophagocytes Polarization. Int J Nanomedicine 2022; 17:3251-3267. [PMID: 35924257 PMCID: PMC9342721 DOI: 10.2147/ijn.s361400] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/20/2022] [Indexed: 11/23/2022] Open
Abstract
Background Mycophenolate mofetil (MMF), for which the bioactive metabolite is mycophenolic acid (MPA), is a frequently used immunosuppressant for systemic lupus erythematosus (SLE). However, its short half-life and poor biodistribution into cells and tissues hinder its clinical efficacy. Our dextran mycophenolate-based nanoparticles (MPA@Dex-MPA NPs) have greatly improved the pharmacokinetics of MMF/MPA. We here tested the therapeutic efficacy of MPA@Dex-MPA NPs against SLE and investigated the underlying mechanism. Methods The tissue and immune cell biodistributions of MPA@Dex-MPA NPs were traced using live fluorescence imaging system and flow cytometry, respectively. Serological proinflammatory mediators and kidney damage were detected to assess the efficacy of MPA@Dex-MPA NPs treatments of MRL/lpr lupus-prone mice. Immune cell changes in the kidney and spleen were further analyzed post-treatment via flow cytometry. Bone marrow-derived macrophages were used to investigate the potential mechanism. Results MPA@Dex-MPA NPs exhibited superior therapeutic efficacy and safety in the MRL/lpr mice using significantly lower administration dosage (one-fifth) and frequency (once/3 days) compared to MMF/MPA used in ordinary practice. The overall prognosis of the mice was improved as they showed lower levels of serological proinflammatory mediators. Moreover, kidney injury was alleviated with reduced pathological signs and decreased urine protein-creatinine ratio. Further investigations of the underlying mechanism revealed a preferential penetration and persistent retention of MPA@Dex-MPA NPs in the spleen and kidney, where they were mostly phagocytosed by macrophages. The macrophages were found to be polarized towards a CD206+ M2-like phenotype, with a downregulation of surface CD80 and CD40, and reduced TNF-α production in the spleen and kidney and in vitro. The expansion of T cells was also significantly inhibited in these two organs. Conclusion Our research improved the efficacy of MPA for MRL/lpr mice through synthesizing MPA@Dex-MPA NPs to enhance its tissue biodistribution and explored the possible mechanism, providing a promising strategy for SLE therapy.
Collapse
Affiliation(s)
- Biling Jiang
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Yamin Zhang
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Yuce Li
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, HUST, Wuhan, People’s Republic of China
| | - Yu Chen
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, HUST, Wuhan, People’s Republic of China
| | - Shanshan Sha
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Liang Zhao
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Danqi Li
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Jingjing Wen
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Jiajia Lan
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Yuchen Lou
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical College, HUST, Wuhan, People’s Republic of China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, HUST, Wuhan, People’s Republic of China
| | - Jintao Zhu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, HUST, Wuhan, People’s Republic of China
| | - Juan Tao
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
- Correspondence: Juan Tao; Jintao Zhu, Email ;
| |
Collapse
|
27
|
Tampe D, Hakroush S, Tampe B. Dissecting signalling pathways associated with intrarenal synthesis of complement components in lupus nephritis. RMD Open 2022; 8:rmdopen-2022-002517. [PMID: 35906025 PMCID: PMC9345095 DOI: 10.1136/rmdopen-2022-002517] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022] Open
Abstract
Lupus nephritis is one of the most common and serious complications of systemic lupus erythematosus, attributed to increased morbidity and mortality. The in situ deposition of intrarenal immune complexes promote the accumulation of inflammatory cells and cause kidney injury in lupus nephritis. Among potential sources of intrarenal complement deposits, the concept of intrarenal complement synthesis has been described more than three decades ago in experimental lupus nephritis. By using transcriptome datasets, we here identified accelerated intrarenal synthesis of distinct classical and alternative complement pathway components, most associated with impaired kidney function. Contrasting to this, no such induction of intrarenal complement synthesis was observed in disease controls, further supporting relevance of intrarenal complement synthesis especially in human lupus nephritis. Gene set enrichment identified that glomerular complement synthesis predominantly associated with interferon signalling and signalling by interleukins in human lupus nephritis, whereas tubulointerstitial complement synthesis with aberrant T-cell receptor signalling. Because the pathomechanistic involvement of complement system activation contributed to recent advances in targeted therapy in lupus nephritis, this study provides additional insights into signalling pathways associated with intrarenal synthesis of complement components in lupus nephritis that might be also affected by targeted therapy of the complement system.
Collapse
Affiliation(s)
- Desiree Tampe
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - Samy Hakroush
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Björn Tampe
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
28
|
Mysore V, Tahir S, Furuhashi K, Arora J, Rosetti F, Cullere X, Yazbeck P, Sekulic M, Lemieux ME, Raychaudhuri S, Horwitz BH, Mayadas TN. Monocytes transition to macrophages within the inflamed vasculature via monocyte CCR2 and endothelial TNFR2. J Exp Med 2022; 219:e20210562. [PMID: 35404389 PMCID: PMC9006314 DOI: 10.1084/jem.20210562] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 11/16/2021] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Monocytes undergo phenotypic and functional changes in response to inflammatory cues, but the molecular signals that drive different monocyte states remain largely undefined. We show that monocytes acquire macrophage markers upon glomerulonephritis and may be derived from CCR2+CX3CR1+ double-positive monocytes, which are preferentially recruited, dwell within glomerular capillaries, and acquire proinflammatory characteristics in the nephritic kidney. Mechanistically, the transition to immature macrophages begins within the vasculature and relies on CCR2 in circulating cells and TNFR2 in parenchymal cells, findings that are recapitulated in vitro with monocytes cocultured with TNF-TNFR2-activated endothelial cells generating CCR2 ligands. Single-cell RNA sequencing of cocultures defines a CCR2-dependent monocyte differentiation path associated with the acquisition of immune effector functions and generation of CCR2 ligands. Immature macrophages are detected in the urine of lupus nephritis patients, and their frequency correlates with clinical disease. In conclusion, CCR2-dependent functional specialization of monocytes into macrophages begins within the TNF-TNFR2-activated vasculature and may establish a CCR2-based autocrine, feed-forward loop that amplifies renal inflammation.
Collapse
Affiliation(s)
- Vijayashree Mysore
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Suhail Tahir
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Kazuhiro Furuhashi
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Jatin Arora
- Center for Data Sciences, Brigham and Women’s Hospital, Boston, MA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Xavier Cullere
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Pascal Yazbeck
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Miroslav Sekulic
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | | | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women’s Hospital, Boston, MA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Centre for Genetics and Genomics Versus Arthritis, The University of Manchester, Manchester, UK
| | - Bruce H. Horwitz
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA
| | - Tanya N. Mayadas
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
29
|
Regulation of activated T cell survival in rheumatic autoimmune diseases. Nat Rev Rheumatol 2022; 18:232-244. [PMID: 35075294 DOI: 10.1038/s41584-021-00741-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2021] [Indexed: 12/29/2022]
Abstract
Adaptive immune responses rely on the proliferation of T lymphocytes able to recognize and eliminate pathogens. The magnitude and duration of the expansion of activated T cell clones are finely regulated to minimize immunopathology and avoid autoimmunity. In patients with rheumatic autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis, activated lymphocytes survive and exert effector functions for prolonged periods, defying the mechanisms that normally curb their capacities during acute and chronic infections. Here, we review the molecular mechanisms that limit the duration of immune responses in health and discuss the factors that alter such regulation in the setting of systemic lupus erythematosus and rheumatoid arthritis. We highlight defects that could contribute to the development and progression of autoimmune disease and describe how chronic inflammation can alter the regulation of activated lymphocyte survival, promoting its perpetuation. These concepts might contribute to the understanding of the mechanisms that underlie the chronicity of inflammation in the context of autoimmunity.
Collapse
|
30
|
Lavine N, Ohayon A, Mahroum N. Renal autoimmunity: The role of bacterial and viral infections, an extensive review. Autoimmun Rev 2022; 21:103073. [PMID: 35245692 DOI: 10.1016/j.autrev.2022.103073] [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: 02/12/2022] [Accepted: 02/27/2022] [Indexed: 02/08/2023]
Abstract
Autoimmunity is a process by which the loss of self-tolerance results in an immune attack against the body own tissues and organs. For autoimmunity to occur, various elements serving as triggers were described by which infections are considered one of the leading factors. In turn, renal involvement in autoimmune diseases, whether by an organ-specific attack, or as part of a systemic disease process, is well known. As bacterial and viral infections are considered to be common triggers for autoimmunity in general, we aimed to study their association with renal autoimmunity in particular. We performed an extensive search of the recent and relevant medical literature regarding renal autoimmunity syndromes such as infection-associated glomerulonephritis and vasculitis, associated with bacterial and viral infections. By utilizing PubMed and Google Scholar search engines, over 200 articles and case reports were reviewed. Among other mechanisms, direct infection of the renal parenchyma, molecular mimicry, induction of B-cells or secretion of superantigens, bacterial and viral pathogens were found to correlate with the development of renal autoimmunity. Nevertheless, this was not true for all pathogens, as some mimic autoimmune diseases and others show a surprisingly protective effect. The exact immunopathogenesis is yet to be determined, however. For conclusion, bacterial and viral infections are linked to renal autoimmunity by both direct damage and as mediators of systemic diseases. Further research particularly on the immunopathogenetic mechanisms of renal autoimmunity associated with infections is required.
Collapse
Affiliation(s)
- Noy Lavine
- St. George School of Medicine, University of London, London, UK; Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel.
| | - Aviran Ohayon
- St. George School of Medicine, University of London, London, UK; Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel
| | - Naim Mahroum
- Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel; International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
31
|
Rajasinghe LD, Bates MA, Benninghoff AD, Wierenga KA, Harkema JR, Pestka JJ. Silica Induction of Diverse Inflammatory Proteome in Lungs of Lupus-Prone Mice Quelled by Dietary Docosahexaenoic Acid Supplementation. Front Immunol 2022; 12:781446. [PMID: 35126352 PMCID: PMC8813772 DOI: 10.3389/fimmu.2021.781446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/06/2021] [Indexed: 11/18/2022] Open
Abstract
Repeated short-term intranasal instillation of lupus-prone mice with crystalline silica (cSiO2) induces inflammatory gene expression and ectopic lymphoid neogenesis in the lung, leading to early onset of systemic autoimmunity and rapid progression to glomerulonephritis. These responses are suppressed by dietary supplementation with the ω-3 polyunsaturated fatty acid docosahexaenoic acid (DHA). Here, we tested the hypothesis that dietary DHA supplementation suppresses cSiO2-induced inflammatory proteins in bronchoalveolar alveolar lavage fluid (BALF) and plasma of lupus-prone mice. Archived tissue fluid samples were used from a prior investigation in which 6 wk-old lupus-prone female NZBWF1 mice were fed isocaloric diets containing 0 or 10 g/kg DHA for 2 wks and then intranasally instilled with 1 mg cSiO2 or vehicle once weekly for 4 wks. Cohorts were terminated at 1, 5, 9 or 13 wk post-instillation (PI). BALF and plasma from each cohort were analyzed by high density multiplex array profiling of 200 inflammatory proteins. cSiO2 time-dependently induced increases in the BALF protein signatures that were highly reflective of unresolved lung inflammation, although responses in the plasma were much less robust. Induced proteins in BALF included chemokines (e.g., MIP-2, MCP-5), enzymes (e.g., MMP-10, granzyme B), adhesion molecules (e.g., sE-selectin, sVCAM-1), co-stimulatory molecules (e.g., sCD40L, sCD48), TNF superfamily proteins (e.g., sTNFRI, sBAFF-R), growth factors (e.g., IGF-1, IGFBP-3), and signal transduction proteins (e.g., MFG-E8, FcgRIIB), many of which were blocked or delayed by DHA supplementation. The BALF inflammatory proteome correlated positively with prior measurements of gene expression, pulmonary ectopic lymphoid tissue neogenesis, and induction of autoantibodies in the lungs of the control and treatment groups. Ingenuity Pathway Analysis (IPA) revealed that IL-1β, TNF-α, and IL-6 were among the top upstream regulators of the cSiO2-induced protein response. Furthermore, DHA's effects were associated with downregulation of cSiO2-induced pathways involving i) inhibition of ARE-mediated mRNA decay, ii) bacterial and viral pattern recognition receptor activation, or iii) TREM1, STAT3, NF-κB, and VEGF signaling and with upregulation of PPAR, LXR/RXR and PPARα/RXRα signaling. Altogether, these preclinical findings further support the contention that dietary DHA supplementation could be applicable as an intervention against inflammation-driven autoimmune triggering by cSiO2 or potentially other environmental agents.
Collapse
Affiliation(s)
- Lichchavi D. Rajasinghe
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Melissa A. Bates
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Abby D. Benninghoff
- Department of Animal, Dairy and Veterinary Sciences, School of Veterinary Medicine, Utah State University, Logan, UT, United States
| | - Kathryn A. Wierenga
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Jack R. Harkema
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - James J. Pestka
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
32
|
Koga T, Ichinose K, Tsokos GC. Tissue resident cell processes determine organ damage in systemic lupus erythematosus. Clin Immunol 2022; 234:108919. [PMID: 34974170 DOI: 10.1016/j.clim.2021.108919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 12/25/2021] [Indexed: 11/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that affects almost any organ. Multiple immunological abnormalities involving every domain of the immune system contribute to the expression of the disease. It is now recognized that elements of the immune system instigate processes in tissue resident cells which execute organ damage. Although correction of ongoing immune aberrations is important in the control of disease activity, targeting tissue specific injurious processes may prove desirable in limiting organ damage.
Collapse
Affiliation(s)
- Tomohiro Koga
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - Kunihiro Ichinose
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - George C Tsokos
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
33
|
Pan BP, Feng ZJ, Li XL, He LP, Jiang D, Zhang RX, Cao WT, Cai M, Bi YF, Wang WF, Luan CY. An Analysis of the Correlation Between Clinical Indexes and Pathological Classifications in 202 Patients with Lupus Nephritis. J Inflamm Res 2021; 14:6917-6927. [PMID: 34938094 PMCID: PMC8687693 DOI: 10.2147/jir.s339744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/05/2021] [Indexed: 11/23/2022] Open
Affiliation(s)
- Bang-Pin Pan
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhi-Jian Feng
- Department of Nephrology, The Second Affiliated Hospital of Kunming Medical University, Branch of National Clinical Research Center For Kidney Disease, Kunming, People’s Republic of China
| | - Xiao-Lan Li
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
- Correspondence: Xiao-Lan Li Tel +86 87163402212Fax +86 87165334416 Email
| | - Li-Ping He
- Department of Epidemiology and Health Statistics, School of Public Health, Kunming Medical University, Kunming, People’s Republic of China
| | - Dong Jiang
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Rui-Xian Zhang
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Wen-Ting Cao
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Mei Cai
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Yun-Feng Bi
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Wen-Fang Wang
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Chun-Yan Luan
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| |
Collapse
|
34
|
Owen KA, Grammer AC, Lipsky PE. Deconvoluting the heterogeneity of SLE: The contribution of ancestry. J Allergy Clin Immunol 2021; 149:12-23. [PMID: 34857396 DOI: 10.1016/j.jaci.2021.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/23/2022]
Abstract
Systemic lupus erythematosus (SLE) is a multiorgan autoimmune disorder with a prominent genetic component. Evidence has shown that individuals of non-European ancestry experience the disease more severely, exhibiting an increased incidence of cardiovascular disease, renal involvement, and tissue damage compared with European ancestry populations. Furthermore, there seems to be variability in the response of individuals within different ancestral groups to standard medications, including cyclophosphamide, mycophenolate, rituximab, and belimumab. Although the widespread application of candidate gene, Immunochip, and genome-wide association studies has contributed to our understanding of the link between genetic variation (typically single nucleotide polymorphisms) and SLE, despite decades of research it is still unclear why ancestry remains a key determinant of poorer outcome in non-European-ancestry patients with SLE. Here, we will discuss the impact of ancestry on SLE disease burden in patients from diverse backgrounds and highlight how research efforts using novel bioinformatic and pathway-based approaches have begun to disentangle the complex genetic architecture linking ancestry to SLE susceptibility. Finally, we will illustrate how genomic and gene expression analyses can be combined to help identify novel molecular pathways and drug candidates that might uniquely impact SLE among different ancestral populations.
Collapse
|
35
|
Level and avidity of antineutrophil cytoplasmic antibodies specific to lactoferrin are useful biomarkers in systemic lupus erythematosus. Clin Rheumatol 2021; 41:709-720. [PMID: 34618258 DOI: 10.1007/s10067-021-05926-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 08/27/2021] [Accepted: 09/16/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVES To evaluate specificity, level, and avidity of antineutrophil cytoplasmic antibodies (ANCA) in systemic lupus erythematosus (SLE). There are no studies of ANCA avidity in SLE. METHODS Level (ELISA) and avidity (ELISA) of myeloperoxidase (MPO-), proteinase 3 (PR3-), lactoferrin (LF-), cathepsin G, elastase (EL-), and bactericidal/permeability increasing protein (BPI)-ANCA in 142 SLE patients were studied. SLE activity was measured by SLEDAI-2 K. 25/40 ANCA-positive patients were immunoserologically followed (12 ± 2 months). RESULTS 40/142 (28.2%) SLE patients were ANCA-positive: LF- (21/40), MPO- (19/40), EL- (6/40), PR3- (3/40), and BPI-ANCA (1/40). Only LF-ANCA were associated with renal manifestations (p < 0.05), and positive predictive value for renal involvement in ANCA-positive SLE was 76.2%. LF-ANCA-positive patients had higher SLEDAI-2 K (p < 0.05) and more frequently had anti-dsDNA (p < 0.05), low C3 (p < 0.001), and low C4 (p < 0.05) than LF-ANCA-negative patients. LF-ANCA level was in a positive correlation with SLEDAI-2 K, anti-dsDNA, and anti-C1q (p < 0.01) and in a negative correlation with C3 and C4 (p < 0.05). LF-ANCA avidity was higher than MPO-, EL-, PR3-, and BPI-ANCA avidity (p < 0.01). In LF-ANCA-positive patients, renal manifestations were associated with higher LF-ANCA level (p < 0.01) and avidity (p < 0.05). Based on LF-ANCA level and avidity, the receiver operating characteristic curves for discriminating patients with and without renal involvement had areas under the curves of 0.988 (95% CI: 0.949-1.00) and 0.813 (95% CI: 0.607-1.00), respectively. After the follow-up period, number of LF-ANCA-positive patients decreased (p < 0.01). CONCLUSIONS In contrast to other ANCAs, only LF-ANCA level correlated with activity and standard serological SLE markers. LF-ANCA level and avidity might be biomarkers of renal involvement in SLE. LF-ANCA are promising serological marker in SLE. Key Points • LF- and MPO-ANCA were most frequently found, while EL-, PR3-, and BPI-ANCA were rarely detected in SLE. • In contrast to other ANCAs, only LF-ANCA were associated with renal involvement, and their level correlated with the activity and standard serological markers of SLE. • LF-ANCA avidity was higher than other ANCAs' avidity; LF-ANCA level and avidity might be useful biomarkers of renal manifestations in SLE. • Detection of ANCA specificity, level, and avidity may help in the diagnosis of particular clinical SLE phenotypes.
Collapse
|
36
|
Zhang LH, Jiang SZ, Guo X, Xiao B, Li Q, Chen JY, Huang JR, Rao H. MiR-146b-5p targets IFI35 to inhibit inflammatory response and apoptosis via JAK1/STAT1 signalling in lipopolysaccharide-induced glomerular cells. Autoimmunity 2021; 54:430-438. [PMID: 34435525 DOI: 10.1080/08916934.2020.1864730] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The dysregulated microRNAs (miRNAs) are implicated in the malignancy of lupus nephritis (LN). This work aims to analyse the effect and mechanism of miR-146b-5p in lipopolysaccharides (LPS)-induced model of LN in vitro. The serum samples of LN patients and normal volunteers were collected. HK-2 cells were challenged via LPS. miR-146b-5p and interferon-induced protein 35 (IFI35) abundances were detected via quantitative real-time polymerase chain reaction (qRT-PCR) or western blot. The inflammatory response was assessed via inflammatory cytokines levels via qRT-PCR and enzyme-linked immunosorbent assay. Cell apoptosis was analysed via flow cytometry and apoptotic protein levels. The protein levels of JAK1/STAT1 signalling were detected via western blot. The relationship of miR-146b-5p and IFI35 was analysed via bioinformatics and dual-luciferase reporter assays. This study revealed that miR-146b-5p level was declined and IFI35 abundance was elevated in serum of LN patients and LPS-challenged HK-2 cells. Functionally, IFI35 overexpression promoted LPS-caused inflammatory response and cell apoptosis, and knockdown of IFI35 caused an opposite trend. Meanwhile, miR-146b-5p targeted IFI35 to suppress inflammatory response and cell inflammatory response and apoptosis via inactivating the JAK1/STAT1 pathway. MiR-146b-5p suppressed inflammatory response and cell apoptosis by IFI35 mediated-JAK1/STAT1 signalling in HK-2 cells, which provided a new mechanism for understanding the pathogenesis of LN.
Collapse
Affiliation(s)
- Li-Hua Zhang
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, P. R. China
| | - Sheng-Zhi Jiang
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, P. R. China
| | - Xia Guo
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, P. R. China
| | - Bin Xiao
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, P. R. China
| | - Qiao Li
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, P. R. China
| | - Jian-Ying Chen
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, P. R. China
| | - Jie-Rou Huang
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, P. R. China
| | - Hui Rao
- Department of Rheumatology and Immunology, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, P. R. China
| |
Collapse
|
37
|
Liu T, Yang M, Xia Y, Jiang C, Li C, Jiang Z, Wang X. Microarray-based analysis of renal complement components reveals a therapeutic target for lupus nephritis. Arthritis Res Ther 2021; 23:223. [PMID: 34433493 PMCID: PMC8385907 DOI: 10.1186/s13075-021-02605-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
Background Screening abnormal pathways and complement components in the kidneys of patients with lupus nephritis (LN) and NZB/W mice may help to identify complement-related therapeutic targets for LN. Methods KEGG and GO enrichment assays were used to analyze kidney microarray data of LN patients and NZB/W mice. Immunohistochemistry and immunofluorescence assays were used to measure renal expression of complement-related proteins and TGFβ1. Cytokines were measured using RT-qPCR and ELISA. Results We screened the renal pathogenic pathways present in LN patients and NZB/W mice and selected the complement activation pathway for further study. The results indicated greater renal expression of C1qa, C1qb, C3, C3aR1, and C5aR1 at the mRNA and protein levels. C3 appeared to be a key factor in LN and the renal signaling downstream of C1 was inhibited. There were significant correlations between the expression of TGFβ1 and C3. Analysis of primary cell cultures indicated that TGFβ1 promoted the expression of C3 and that a TGFβ1 antagonist decreased the levels of C3 and C3aR. TGFβ1 inhibition significantly inhibited the deposition of complement-related factors in the kidneys of NZB/W mice. Conclusions At the onset of LN, there are significant increases in the renal levels of C3 and other complement pathway-related factors in patients with LN and NZB/W mice. C3 may lead to albuminuria and participate in the pathogenesis of LN. TGFβ1 promotes C3 synthesis, and TGFβ1 inhibition may block the progression of LN by inhibiting the synthesis of C3 and other complement components. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02605-9.
Collapse
Affiliation(s)
- Tao Liu
- Department of Rheumatology and Immunology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Mingyue Yang
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Ying Xia
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Chuan Jiang
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Chenxu Li
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zhenyu Jiang
- Department of Rheumatology and Immunology, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Xiaosong Wang
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
38
|
Chi M, Ma K, Li Y, Quan M, Han Z, Ding Z, Liang X, Zhang Q, Song L, Liu C. Immunological Involvement of MicroRNAs in the Key Events of Systemic Lupus Erythematosus. Front Immunol 2021; 12:699684. [PMID: 34408748 PMCID: PMC8365877 DOI: 10.3389/fimmu.2021.699684] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an archetype autoimmune disease characterized by a myriad of immunoregulatory abnormalities that drives injury to multiple tissues and organs. Due to the involvement of various immune cells, inflammatory cytokines, and related signaling pathways, researchers have spent a great deal of effort to clarify the complex etiology and pathogenesis of SLE. Nevertheless, current understanding of the pathogenesis of SLE is still in the early stages, and available nonspecific treatment options for SLE patients remain unsatisfactory. First discovered in 1993, microRNAs (miRNAs) are small RNA molecules that control the expression of 1/3 of human genes at the post-transcriptional level and play various roles in gene regulation. The aberrant expression of miRNAs in SLE patients has been intensively studied, and further studies have suggested that these miRNAs may be potentially relevant to abnormal immune responses and disease progression in SLE. The aim of this review was to summarize the specific miRNAs that have been observed aberrantly expressed in several important pathogenetic processes in SLE, such as DCs abnormalities, overactivation and autoantibody production of B cells, aberrant activation of CD4+ T cells, breakdown of immune tolerance, and abnormally increased production of inflammatory cytokines. Our summary highlights a novel perspective on the intricate regulatory network of SLE, which helps to enrich our understanding of this disorder and ignite future interest in evaluating the molecular regulation of miRNAs in autoimmunity SLE.
Collapse
Affiliation(s)
- Mingxuan Chi
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kuai Ma
- Department of Nephrology, Osaka University, Suita, Japan
| | - Yunlong Li
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Min Quan
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhongyu Han
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhaolun Ding
- Department of Emergency Surgery, Shannxi Provincial People's Hospital, Xi'an, China
| | - Xin Liang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qinxiu Zhang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Linjiang Song
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chi Liu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Nephrology, Sichuan Clinical Research Center for Kidney Disease, Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, China
| |
Collapse
|
39
|
Cellular Response against Oxidative Stress, a Novel Insight into Lupus Nephritis Pathogenesis. J Pers Med 2021; 11:jpm11080693. [PMID: 34442337 PMCID: PMC8401250 DOI: 10.3390/jpm11080693] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023] Open
Abstract
The interaction of reactive oxygen species (ROS) with lipids, proteins, nucleic acids and hydrocarbonates promotes acute and chronic tissue damage, mediates immunomodulation and triggers autoimmunity in systemic lupus erythematous (SLE) patients. The aim of the study was to determine the pathophysiological mechanisms of the oxidative stress-related damage and molecular mechanisms to counteract oxidative stimuli in lupus nephritis. Our study included 38 SLE patients with lupus nephritis (LN group), 44 SLE patients without renal impairment (non-LN group) and 40 healthy volunteers as control group. In the present paper, we evaluated serum lipid peroxidation, DNA oxidation, oxidized proteins, carbohydrate oxidation, and endogenous protective systems. We detected defective DNA repair mechanisms via 8-oxoguanine-DNA-glycosylase (OGG1), the reduced regulatory effect of soluble receptor for advanced glycation end products (sRAGE) in the activation of AGE-RAGE axis, low levels of thiols, disulphide bonds formation and high nitrotyrosination in lupus nephritis. All these data help us to identify more molecular mechanisms to counteract oxidative stress in LN that could permit a more precise assessment of disease prognosis, as well as developing new therapeutic targets.
Collapse
|
40
|
Association Between Hypoalbuminemia, Degree of Proteinuria, and Lupus Nephritis Class: A Single-Center Cross-sectional Biopsy Study. Nephrourol Mon 2021. [DOI: 10.5812/numonthly.112883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: The current study intended to determine whether serum albumin level and urine protein/creatinine rate (uPCR) are appropriate predictors of severe lupus nephritis in childhood-onset SLE. Objectives: Following a cross-sectional single-center design, 85 LN children referred to the National Children Hospital, Ha Noi, Viet Nam, from 6/2019 to 6/2020 were recruited. Renal biopsy was performed for all participants. Methods: Following a cross-sectional single-center design, 85 LN children referred to the National Children Hospital, Ha Noi, Viet Nam, from 6/2019 to 6/2020 were recruited. Renal biopsy was performed for all participants. Results: The mean SLEDAI score of all patients was 14.69. The proportion of patients with high and very high SLEDAI was 61.2 and 17.6%, respectively. The mean concentration of serum albumin was 28.55 g/L, and the proportion of decreased albumin concentration was 55.3%. The median uPCR was 446.6 mg/mmol in which 76.5% of values were ≥ 200 mg/mmol. Pathological morphology of LN class from I to VI was observed in 0%, 17.6%, 37.6 %, 37.6%, 7.1%, and 0%, respectively. Serum albumin level and uPCR presented the predictive value for severe and active LN (class IV and V); (AUC: 0.725 P < 0.001 for both). Conclusions: Serum albumin and uPCR were appropriate predictors for severe and active LN in Vietnamese children.
Collapse
|
41
|
Melki I, Allaeys I, Tessandier N, Lévesque T, Cloutier N, Laroche A, Vernoux N, Becker Y, Benk-Fortin H, Zufferey A, Rollet-Labelle E, Pouliot M, Poirier G, Patey N, Belleannee C, Soulet D, McKenzie SE, Brisson A, Tremblay ME, Lood C, Fortin PR, Boilard E. Platelets release mitochondrial antigens in systemic lupus erythematosus. Sci Transl Med 2021; 13:13/581/eaav5928. [PMID: 33597264 DOI: 10.1126/scitranslmed.aav5928] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 03/20/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022]
Abstract
The accumulation of DNA and nuclear components in blood and their recognition by autoantibodies play a central role in the pathophysiology of systemic lupus erythematosus (SLE). Despite the efforts, the sources of circulating autoantigens in SLE are still unclear. Here, we show that in SLE, platelets release mitochondrial DNA, the majority of which is associated with the extracellular mitochondrial organelle. Mitochondrial release in patients with SLE correlates with platelet degranulation. This process requires the stimulation of platelet FcγRIIA, a receptor for immune complexes. Because mice lack FcγRIIA and murine platelets are completely devoid of receptor capable of binding IgG-containing immune complexes, we used transgenic mice expressing FcγRIIA for our in vivo investigations. FcγRIIA expression in lupus-prone mice led to the recruitment of platelets in kidneys and to the release of mitochondria in vivo. Using a reporter mouse with red fluorescent protein targeted to the mitochondrion, we confirmed platelets as a source of extracellular mitochondria driven by FcγRIIA and its cosignaling by the fibrinogen receptor α2bβ3 in vivo. These findings suggest that platelets might be a key source of mitochondrial antigens in SLE and might be a therapeutic target for treating SLE.
Collapse
Affiliation(s)
- Imene Melki
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Isabelle Allaeys
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Nicolas Tessandier
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Tania Lévesque
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Nathalie Cloutier
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Audrée Laroche
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Nathalie Vernoux
- Axe Neurosciences du Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval et Département de Médecine Moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Yann Becker
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Hadrien Benk-Fortin
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Anne Zufferey
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Emmanuelle Rollet-Labelle
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Marc Pouliot
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Guy Poirier
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Université Laval, Quebec, QC G1V 4G2, Canada
| | - Natacha Patey
- Centre Hospitalier Universitaire de Sainte-Justine, Faculté de Médecine, Département de pathologie et biologie cellulaire, Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - Clemence Belleannee
- Department of Obstetrics, Gynecology and Reproduction, Centre hospitalier universitaire de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Denis Soulet
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Université Laval, Quebec, QC G1V 4G2, Canada
| | - Steven E McKenzie
- Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alain Brisson
- UMR-CBMN CNRS-Université de Bordeaux-IPB, Pessac 33600, France
| | - Marie-Eve Tremblay
- Axe Neurosciences du Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval et Département de Médecine Moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Christian Lood
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Paul R Fortin
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada. .,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada.,Division of Rheumatology, Department of Medicine, Centre hospitalier universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Eric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada. .,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| |
Collapse
|
42
|
Atisha-Fregoso Y, Toz B, Diamond B. Meant to B: B cells as a therapeutic target in systemic lupus erythematosus. J Clin Invest 2021; 131:149095. [PMID: 34128474 PMCID: PMC8203443 DOI: 10.1172/jci149095] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
B cells have a prominent role in the pathogenesis of systemic lupus erythematosus (SLE). They are mediators of inflammation through the production of pathogenic antibodies that augment inflammation and cause direct tissue and cell damage. Multiple therapeutic agents targeting B cells have been successfully used in mouse models of SLE; however, these preclinical studies have led to approval of only one new agent to treat patients with SLE: belimumab, a monoclonal antibody targeting B cell-activating factor (BAFF). Integrating the experience acquired from previous clinical trials with the knowledge generated by new studies about mechanisms of B cell contributions to SLE in specific groups of patients is critical to the development of new treatment strategies that will help to improve outcomes in patients with SLE. In particular, a sharper focus on B cell differentiation to plasma cells is warranted.
Collapse
Affiliation(s)
- Yemil Atisha-Fregoso
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, New York, USA
| | - Bahtiyar Toz
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Queens Hospital Center, New York, New York, USA
| | - Betty Diamond
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| |
Collapse
|
43
|
Diffusion tensor imaging of renal cortex in lupus nephritis. Jpn J Radiol 2021; 39:1069-1076. [PMID: 34125367 DOI: 10.1007/s11604-021-01154-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/08/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE To evaluate the diagnostic value of diffusion tensor imaging (DTI) of renal cortex in assessment of lupus nephritis (LN) and prediction of its pathological subtypes. METHODS Prospective study was performed upon 39 female patients with pathologically proven LN and 16 sex- and age-matched healthy controls. Patients and controls underwent DTI of kidney. Mean diffusivity (MD) and fractional anisotropy (FA) of renal cortex were calculated by two radiologists. LN patients were pathologically classified into either non-proliferative (n = 15) or proliferative (n = 24). RESULTS Mean MD of renal cortex in LN was significantly lower (p = 0.001) than that of controls with cut-off (2.16 and 2.2 X10-3mm2/s), area under curve (AUC) of (0.92, 0.94) and accuracy of (91%, 89%) for both observers. Mean FA of renal cortex in LN was significantly higher (p = 0.001) than that of controls with cut-off (0.20, 0.21), AUC of (0.86, 0.82) and accuracy of (86%, 84%) for both observers. Renal cortex MD and FA in non-proliferative LN were significantly different (p = 0.001) from that of proliferative LN for both observers. There was excellent inter-observer agreement of MD and FA (ICC = 0.96 and 0.81). CONCLUSION MD and FA of renal cortex may help to assess renal affection in LN patients and predict its pathological subtypes.
Collapse
|
44
|
Schell SL, Bricker KN, Fike AJ, Chodisetti SB, Domeier PP, Choi NM, Fasnacht MJ, Luckenbill SA, Ziegler SF, Rahman ZSM. Context-Dependent miR-21 Regulation of TLR7-Mediated Autoimmune and Foreign Antigen-Driven Antibody-Forming Cell and Germinal Center Responses. THE JOURNAL OF IMMUNOLOGY 2021; 206:2803-2818. [PMID: 34039637 DOI: 10.4049/jimmunol.2001039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 04/01/2021] [Indexed: 01/03/2023]
Abstract
MicroRNAs (miRNAs) are involved in healthy B cell responses and the loss of tolerance in systemic lupus erythematosus (SLE), although the role of many miRNAs remains poorly understood. Dampening miR-21 activity was previously shown to reduce splenomegaly and blood urea nitrogen levels in SLE-prone mice, but the detailed cellular responses and mechanism of action remains unexplored. In this study, using the TLR7 agonist, imiquimod-induced SLE model, we observed that loss of miR-21 in Sle1b mice prevented the formation of plasma cells and autoantibody-producing Ab-forming cells (AFCs) without a significant effect on the magnitude of the germinal center (GC) response. We further observed reduced dendritic cell and monocyte numbers in the spleens of miR-21-deficient Sle1b mice that were associated with reduced IFN, proinflammatory cytokines, and effector CD4+ T cell responses. RNA sequencing analysis on B cells from miR-21-deficient Sle1b mice revealed reduced activation and response to IFN, and cytokine and target array analysis revealed modulation of numerous miR-21 target genes in response to TLR7 activation and type I IFN stimulation. Our findings in the B6.Sle1bYaa (Sle1b Yaa) spontaneous model recapitulated the miR-21 role in TLR7-induced responses with an additional role in autoimmune GC and T follicular helper responses. Finally, immunization with T-dependent Ag revealed a role for miR-21 in foreign Ag-driven GC and Ab, but not AFC, responses. Our data suggest a potential multifaceted, context-dependent role for miR-21 in autoimmune and foreign Ag-driven AFC and GC responses. Further study is warranted to delineate the cell-intrinsic requirements and mechanisms of miR-21 during infection and SLE development.
Collapse
Affiliation(s)
- Stephanie L Schell
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA; and
| | - Kristen N Bricker
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA; and
| | - Adam J Fike
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA; and
| | - Sathi Babu Chodisetti
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA; and
| | | | - Nicholas M Choi
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA; and
| | - Melinda J Fasnacht
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA; and
| | - Sara A Luckenbill
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA; and
| | | | - Ziaur S M Rahman
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA; and
| |
Collapse
|
45
|
Schell SL, Rahman ZSM. miRNA-Mediated Control of B Cell Responses in Immunity and SLE. Front Immunol 2021; 12:683710. [PMID: 34079558 PMCID: PMC8165268 DOI: 10.3389/fimmu.2021.683710] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/04/2021] [Indexed: 12/11/2022] Open
Abstract
Loss of B cell tolerance is central to autoimmune diseases such as systemic lupus erythematosus (SLE). As such, the mechanisms involved in B cell development, maturation, activation, and function that are aberrantly regulated in SLE are of interest in the design of targeted therapeutics. While many factors are involved in the generation and regulation of B cell responses, miRNAs have emerged as critical regulators of these responses within the last decade. To date, miRNA involvement in B cell responses has largely been studied in non-autoimmune, immunization-based systems. However, miRNA profiles have also been strongly associated with SLE in human patients and these molecules have proven critical in both the promotion and regulation of disease in mouse models and in the formation of autoreactive B cell responses. Functionally, miRNAs are small non-coding RNAs that bind to complementary sequences located in target mRNA transcripts to mediate transcript degradation or translational repression, invoking a post-transcriptional level of genetic regulation. Due to their capacity to target a diverse range of transcripts and pathways in different immune cell types and throughout the various stages of development and response, targeting miRNAs is an interesting potential therapeutic avenue. Herein, we focus on what is currently known about miRNA function in both normal and SLE B cell responses, primarily highlighting miRNAs with confirmed functions in mouse models. We also discuss areas that should be addressed in future studies and whether the development of miRNA-centric therapeutics may be a viable alternative for the treatment of SLE.
Collapse
Affiliation(s)
- Stephanie L Schell
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Ziaur S M Rahman
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
46
|
Yariwake VY, Torres JI, Dos Santos ARP, Freitas SCF, De Angelis K, Farhat SCL, Câmara NOS, Veras MM. Chronic exposure to PM2.5 aggravates SLE manifestations in lupus-prone mice. Part Fibre Toxicol 2021; 18:15. [PMID: 33766080 PMCID: PMC7992962 DOI: 10.1186/s12989-021-00407-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Background Air pollution causes negative impacts on health. Systemic lupus erythematosus (SLE) is an autoimmune disease with diverse clinical manifestations and multifactorial etiology. Recent studies suggest that air pollution can trigger SLE and induce disease activity. However, this association has not been deeply investigated. Thus, the aim of this study was to evaluate whether exposure to fine particulate matter (PM2.5) exacerbates SLE manifestations, focusing on renal complications, in a lupus-prone animal model. Female NZBWF1 mice were exposed daily to 600 μg/m3 of inhaled concentrated ambient particles (CAP) or filtered air (FA). Survival rate, body weight, weight of organs (kidney, spleen, thymus, liver and heart), blood cell count, proteinuria, kidney stereology, renal histopathology, gene expression and oxidative stress were analyzed. Results Female NZBW mice exposed to CAP showed decreased survival, increased circulating neutrophils, early onset of proteinuria and increased kidney weight with renal cortex enlargement when compared to NZBW mice exposed to FA. Conclusions This work shows that air pollution aggravates some SLE manifestations in lupus-prone mice. These results reinforce the need of reducing air pollutant levels in order to promote a better quality of life for individuals diagnosed with SLE. Supplementary Information The online version contains supplementary material available at 10.1186/s12989-021-00407-0.
Collapse
Affiliation(s)
- Victor Yuji Yariwake
- Laboratory of Experimental Air Pollution, Department of Pathology, School of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455 - 1st floor (room 1220), São Paulo, SP, 01246-903, Brazil.
| | - Janaína Iannicelli Torres
- Laboratory of Experimental Air Pollution, Department of Pathology, School of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455 - 1st floor (room 1220), São Paulo, SP, 01246-903, Brazil
| | - Amandda Rakell Peixoto Dos Santos
- Laboratory of Experimental Cellular Immunology, Department of Medicine, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | | | - Kátia De Angelis
- Laboratory of Translational Physiology, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
| | - Sylvia Costa Lima Farhat
- Laboratory of Experimental Air Pollution, Department of Pathology, School of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455 - 1st floor (room 1220), São Paulo, SP, 01246-903, Brazil.,Pediatric Rheumatology Unit, Children's Institute of Hospital das Clínicas, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Laboratory of Experimental Cellular Immunology, Department of Medicine, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil.,Laboratory of Transplant Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mariana Matera Veras
- Laboratory of Experimental Air Pollution, Department of Pathology, School of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455 - 1st floor (room 1220), São Paulo, SP, 01246-903, Brazil
| |
Collapse
|
47
|
Fresneda Alarcon M, McLaren Z, Wright HL. Neutrophils in the Pathogenesis of Rheumatoid Arthritis and Systemic Lupus Erythematosus: Same Foe Different M.O. Front Immunol 2021; 12:649693. [PMID: 33746988 PMCID: PMC7969658 DOI: 10.3389/fimmu.2021.649693] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/12/2021] [Indexed: 12/14/2022] Open
Abstract
Dysregulated neutrophil activation contributes to the pathogenesis of autoimmune diseases including rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). Neutrophil-derived reactive oxygen species (ROS) and granule proteases are implicated in damage to and destruction of host tissues in both conditions (cartilage in RA, vascular tissue in SLE) and also in the pathogenic post-translational modification of DNA and proteins. Neutrophil-derived cytokines and chemokines regulate both the innate and adaptive immune responses in RA and SLE, and neutrophil extracellular traps (NETs) expose nuclear neoepitopes (citrullinated proteins in RA, double-stranded DNA and nuclear proteins in SLE) to the immune system, initiating the production of auto-antibodies (ACPA in RA, anti-dsDNA and anti-acetylated/methylated histones in SLE). Neutrophil apoptosis is dysregulated in both conditions: in RA, delayed apoptosis within synovial joints contributes to chronic inflammation, immune cell recruitment and prolonged release of proteolytic enzymes, whereas in SLE enhanced apoptosis leads to increased apoptotic burden associated with development of anti-nuclear auto-antibodies. An unbalanced energy metabolism in SLE and RA neutrophils contributes to the pathology of both diseases; increased hypoxia and glycolysis in RA drives neutrophil activation and NET production, whereas decreased redox capacity increases ROS-mediated damage in SLE. Neutrophil low-density granulocytes (LDGs), present in high numbers in the blood of both RA and SLE patients, have opposing phenotypes contributing to clinical manifestations of each disease. In this review we will describe the complex and contrasting phenotype of neutrophils and LDGs in RA and SLE and discuss their discrete roles in the pathogenesis of each condition. We will also review our current understanding of transcriptomic and metabolomic regulation of neutrophil phenotype in RA and SLE and discuss opportunities for therapeutic targeting of neutrophil activation in inflammatory auto-immune disease.
Collapse
Affiliation(s)
- Michele Fresneda Alarcon
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Zoe McLaren
- Liverpool University Hospitals National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| | - Helen Louise Wright
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
48
|
Hu L, Hu J, Chen L, Zhang Y, Wang Q, Yang X. Interleukin-22 From Type 3 Innate Lymphoid Cells Aggravates Lupus Nephritis by Promoting Macrophage Infiltration in Lupus-Prone Mice. Front Immunol 2021; 12:584414. [PMID: 33717066 PMCID: PMC7953152 DOI: 10.3389/fimmu.2021.584414] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/21/2021] [Indexed: 12/15/2022] Open
Abstract
Lupus nephritis (LN) is one of the most severe manifestations of systemic lupus erythematosus (SLE). Our previous studies demonstrated increased serum and renal Interleukin (IL)-22 in LN patients and MRL/lpr mice. This study investigated the role of IL-22 and its mechanism in LN. Here, we found that IL-22 was mainly produced by type 3 innate lymphoid cells (ILC3) in kidney of MRL/lpr mice. The systemic illness and local renal lesion were significantly alleviated in IL-22 or IL-22R gene knockout (KO) mice (IL-22 KO or IL-22R KO MRL/lpr mice) than control mice (MRL/lpr mice). IL-22 KO or IL-22R KO MRL/lpr mice had significantly slighter infiltration of macrophage in kidney than MRL/lpr mice. Consistently, by RNA-Seq, the expression of (CC motif) ligand 2 (CCL2) and (CXC motif) ligand 10 (CXCL10) was decreased in kidney of KO mice compared with control mice. By immunoblotting, significantly increased levels of STAT3 phosphorylation were found in the kidney of control mice compared to KO mice. In vitro, primary kidney epithelial cells from control mouse stimulated with recombinant IL-22 (rIL-22) expressed higher levels of CCL2, CXCL10, and phosphorylated STAT3. At the same time, when primary kidney epithelial cells were treated with rIL-22, transwell assay demonstrated their supernatant recruited more macrophages. In human kidney epithelial cell line (HK2) cells, when treated with rIL-22, we observed similar results with primary mouse kidney epithelial cells. Moreover, when cells were stimulated with rIL-22 following pre-treatment with STAT3 pathway inhibitor, the expression of CCL2 and CXCL10 were significantly reversed. Our findings demonstrate that IL-22 binding to IL-22R in kidney epithelial cells activated the STAT3 signaling pathway, enhanced the chemokine secretion and then promoted macrophage infiltration to the kidney of MRL/lpr mice, thus aggravated LN in lupus-prone mice. These findings indicate that IL-22 may play a pathogenic role in LN and may provide a promising novel therapeutic target for LN.
Collapse
Affiliation(s)
- Lingzhen Hu
- Department of Rheumatology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jingyi Hu
- Laboratory of Immunology, College of Medicine, Institute of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Liheng Chen
- Department of Rheumatology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Zhang
- Department of Rheumatology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Rheumatology, Hangzhou Dingqiao Hospital, Hangzhou, China
| | - Qingqing Wang
- Laboratory of Immunology, College of Medicine, Institute of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Xuyan Yang
- Department of Rheumatology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
49
|
Zhang L, Zhang M, Chen X, He Y, Chen R, Zhang J, Huang J, Ouyang C, Shi G. Identification of the tubulointerstitial infiltrating immune cell landscape and immune marker related molecular patterns in lupus nephritis using bioinformatics analysis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 8:1596. [PMID: 33437795 PMCID: PMC7791250 DOI: 10.21037/atm-20-7507] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease that commonly affects the kidneys. Research into markers that can predict the prognosis of tubulointerstitial lupus nephritis (LN) has been impeded by the lack of well-designed studies. Methods In this study, we selected and merged 3 sets of renal biopsy tubulointerstitial data from GSE32591, GSE69438, and GSE127797, including 95 LN and 15 living healthy donors. CIBERSORTx was utilized for differentially infiltrating immune cell (DIIC) analysis. Weighted Gene Co-Expression network analysis (WGCNA) was employed to explore differentially expressed gene (DEG) related modules. Combined WGCNA hub genes and protein-protein interaction (PPI) validation was used for immune marker identification. Lastly, unsupervised clustering was carried out to validate the correlation between these markers and clinical characteristics. Results Our findings unveiled TYROBP, C1QB, LAPTM5, CTSS, PTPRC as the 5 immune markers, which were negatively correlated with glomerular filtration rate (GFR). Specifically, the expression levels of TYROBP and C1QB were significantly different between proliferative LN (PLN) and membranous LN (MLN). Unsupervised clustering could aggregate LN by these immune marker expression spectrums. Conclusions This study is the first to identify infiltrating immune cells and associated molecular patterns in the tubulointerstitium of LN by utilizing bioinformatics methods. These findings contribute to a better understanding of the mechanisms behind LN, and promote more precise diagnosis.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,The Fifth Hospital of Xiamen, Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Mengqin Zhang
- Department of Rheumatology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xing Chen
- Department of Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,The Fifth Hospital of Xiamen, Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yan He
- Department of Rheumatology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Rongjuan Chen
- Department of Rheumatology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jun Zhang
- Department of Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,The Fifth Hospital of Xiamen, Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jiyi Huang
- Department of Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,The Fifth Hospital of Xiamen, Xiang'an Branch, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Chun Ouyang
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guixiu Shi
- Department of Rheumatology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| |
Collapse
|
50
|
Bagheri B, Owji SM, Torabinezhad S, Raeisi Shahraki H, Kamalinia A, Owji SH, Dehghani F. Electron microscopy study of 496 cases of lupus nephritis: A single-center experience. Lupus 2021; 30:587-596. [PMID: 33413002 DOI: 10.1177/0961203320984539] [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] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Renal involvement is seen in about 40-82% of systemic lupus erythematosus (SLE) Asian patients. The exact diagnosis and classification of lupus nephritis are important for treatment and prognosis. This study aimed to investigate the value of electron microscopy (EM) in the diagnosis and classification of lupus nephritis compared with light microscopy. METHOD In this cross-sectional referral-center 16-year study of lupus nephritis, the final diagnosis was based on the EM study. Primary light microscopy findings were compared with EM diagnosis. Moreover, Immunofluorescence patterns distribution was assessed. RESULTS From 496 patients diagnosed with lupus nephritis based on EM, 225(45.4%) of patients were categorized in class IV, followed by 98(19.7%), 93(18.8%), 46(9.3%), and 14(2.8%) who were categorized into classes of II, III, V, and VI respectively. Only 1(0.2%) patient belonged to class I, and 19(3.8%) cases were diagnosed with mixed two classes. Using EM was essential for diagnosing 25.6% of cases taking the correct classification by light microscopy into account; however, disregarding correct classification, this could change to a 7.4% contribution rate of EM. The most common cause of misdiagnosis, disregarding incorrect classification, was inadequate or wrong tissue. Positive associations were detected between tubular atrophy and interstitial fibrosis of both electron and light microscopy with different classes (P < 0.001). CONCLUSION While light microscopy is highly accurate for diagnosing lupus nephritis regardless of correct classification, EM contributes substantially to the correct classification of lupus nephritis types.
Collapse
Affiliation(s)
- Bahar Bagheri
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Owji
- Department of Pathology, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Simin Torabinezhad
- Department of Pathology, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hadi Raeisi Shahraki
- Department of Biostatistics and Epidemiology, Faculty of Health, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Seyed Hossein Owji
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Otolaryngology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farshad Dehghani
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|