1
|
Giuliani KTK, Nag P, Adams BC, Wang X, Hong S, Grivei A, Johnston RL, Waddell N, Ho KKC, Tian Y, Khan MA, Kim CS, Ng MSY, Gobe G, Ungerer JPJ, Forbes JM, Healy HG, Kassianos AJ. Human proximal tubular epithelial cell interleukin-1 receptor signalling triggers G2/M arrest and cellular senescence during hypoxic kidney injury. Cell Death Dis 2025; 16:61. [PMID: 39890773 PMCID: PMC11785723 DOI: 10.1038/s41419-025-07386-6] [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: 09/23/2024] [Revised: 01/12/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Hypoxia and interleukin (IL)-1β are independent mediators of tubulointerstitial fibrosis, the histological hallmark of chronic kidney disease (CKD). Here, we examine how hypoxia and IL-1β act in synergy to augment maladaptive proximal tubular epithelial cell (PTEC) repair in human CKD. Ex vivo patient-derived PTECs were cultured under normoxic (21% O2) or hypoxic (1% O2) conditions in the absence or presence of IL-1β and examined for maladaptive repair signatures. Hypoxic PTECs incubated with IL-1β displayed a discrete transcriptomic profile distinct from PTECs cultured under hypoxia alone, IL-1β alone or under normoxia. Hypoxia+IL-1β-treated PTECs had 692 'unique' differentially expressed genes (DEGs) compared to normoxic PTECs, with 'cell cycle' the most significantly enriched KEGG pathway based on 'unique' down-regulated DEGs (including CCNA2, CCNB1 and CCNB2). Hypoxia+IL-1β-treated PTECs displayed signatures of cellular senescence, with reduced proliferation, G2/M cell cycle arrest, increased p21 expression, elevated senescence-associated β-galactosidase (SA-β-gal) activity and increased production of pro-inflammatory/fibrotic senescence-associated secretory phenotype (SASP) factors compared to normoxic conditions. Treatment of Hypoxia+IL-1β-treated PTECs with either a type I IL-1 receptor (IL-1RI) neutralizing antibody or a senolytic drug combination, quercetin+dasatinib, attenuated senescent cell burden. In vitro findings were validated in human CKD bio-specimens (kidney tissue, urine), with elevated PTEC IL-1RI expression and senescence (SA-β-gal activity) detected in fibrotic kidneys and numbers of senescent (SA-β-gal+) urinary PTECs correlating with urinary IL-1β levels and severity of interstitial fibrosis. Our data identify a mechanism whereby hypoxia in combination with IL-1β/IL-1RI signalling trigger PTEC senescence, providing novel therapeutic and diagnostic check-points for restoring tubular regeneration in human CKD.
Collapse
Affiliation(s)
- Kurt T K Giuliani
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Purba Nag
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Benjamin C Adams
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Xiangju Wang
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Seokchan Hong
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Centre, Seoul, Republic of Korea
| | - Anca Grivei
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | | | - Nicola Waddell
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kenneth K C Ho
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Yilin Tian
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Muhammad Ali Khan
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Kidney Disease Research Collaborative, Princess Alexandra Hospital and University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Dhaka, Bangladesh
| | - Chang Seong Kim
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Department of Internal Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Monica S Y Ng
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Glenda Gobe
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Kidney Disease Research Collaborative, Princess Alexandra Hospital and University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Jacobus P J Ungerer
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Josephine M Forbes
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Mater Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Helen G Healy
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Andrew J Kassianos
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia.
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
- Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.
| |
Collapse
|
2
|
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
|
3
|
Xu Q, Liang R, Luo J, Zhang Y. Development and validation of a predictive model for end-stage renal disease in systemic lupus erythematosus patients. Rheumatol Int 2024; 44:1941-1958. [PMID: 39168871 DOI: 10.1007/s00296-024-05686-2] [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: 06/25/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024]
Abstract
Systemic lupus erythematosus (SLE) affects many populations. This study aims to develop a predictive model and create a nomogram for assessing the risk of end-stage renal disease (ESRD) in patients diagnosed with SLE. Data from electronic health records of SLE patients treated at the Affiliated Hospital of North Sichuan Medical College between 2013 and 2023 were collected. The dataset underwent thorough cleaning and variable assignment procedures. Subsequently, variables were selected using one-way logistic regression and lasso logistic regression methods, followed by multifactorial logistic regression to construct nomograms. The model's performance was assessed using calibration, receiver operating characteristic (ROC), and decision curve analysis (DCA) curves. Statistical significance was set at P < 0.05. The predictive variables for ESRD development in SLE patients included anti-GP210 antibody presence, urinary occult blood, proteinuria, white blood cell count, complement 4 levels, uric acid, creatinine, total protein, globulin, glomerular filtration rate, pH, specific gravity, very low-density lipoprotein, homocysteine, apolipoprotein B, and absolute counts of cytotoxic T cells. The nomogram exhibited a broad predictive range. The ROC area under the curve (AUC) was 0.886 (0.858-0.913) for the training set and 0.840 (0.783-0.897) for the testing set, indicating good model performance. The model demonstrated both applicability and significant clinical benefits. The developed model presents strong predictive capabilities and considerable clinical utility in estimating the risk of ESRD in patients with SLE.
Collapse
Affiliation(s)
- Qiang Xu
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Rui Liang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Jiesi Luo
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Yonglin Zhang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.
| |
Collapse
|
4
|
Chernova I. Lupus Nephritis: Immune Cells and the Kidney Microenvironment. KIDNEY360 2024; 5:1394-1401. [PMID: 39120952 PMCID: PMC11441818 DOI: 10.34067/kid.0000000000000531] [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: 06/05/2024] [Accepted: 07/29/2024] [Indexed: 08/11/2024]
Abstract
Lupus nephritis (LN) is the most common major organ manifestation of the autoimmune disease SLE (lupus), with 10% of those afflicted progressing to ESKD. The kidney in LN is characterized by a significant immune infiltrate and proinflammatory cytokine milieu that affects intrinsic renal cells and is, in part, responsible for the tissue damage observed in LN. It is now increasingly appreciated that LN is not due to unidirectional immune cell activation with subsequent kidney damage. Rather, the kidney microenvironment influences the recruitment, survival, differentiation, and activation of immune cells, which, in turn, modify kidney cell function. This review covers how the biochemical environment of the kidney ( i.e ., low oxygen tension and hypertonicity) and unique kidney cell types affect the intrarenal immune cells in LN. The pathways used by intrinsic renal cells to interact with immune cells, such as antigen presentation and cytokine production, are discussed in detail. An understanding of these mechanisms can lead to the design of more kidney-targeted treatments and the avoidance of systemic immunosuppressive effects and may represent the next frontier of LN therapies.
Collapse
Affiliation(s)
- Irene Chernova
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
5
|
Zou Y, Wang D, Sun W, Wu Q, Liu S, Ren Z, Li Y, Zhao T, Li Z, Li X, Cao W, Han J, Guo X, Ren G. Fibroblast growth factor 21 mitigates lupus nephritis progression via the FGF21/Irgm 1/NLRP3 inflammasome pathway. Int Immunopharmacol 2024; 131:111875. [PMID: 38508095 DOI: 10.1016/j.intimp.2024.111875] [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: 01/04/2024] [Revised: 03/01/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
As an endocrine cytokine, fibroblast growth factor 21 (FGF21) exhibits anti-inflammatory properties. With the development of lupus nephritis (LN), which is tightly related to pathogenic factors, including inflammation and immune cell dysregulation, we explored the impact of Fibroblast Growth Factor 21 (FGF21) as well as its underlying mechanism. We induced an in vivo LN model using pristane in both wild-type C57BL/6 and FGF21 knockout (FGF21-/-) mice. LN serum obtained from 32-week-old wild-type LN mice was used to stimulate RAW264.7 and human renal tubular epithelial (HK-2) cells to mimic an in vitro LN model. Moreover, our findings revealed that FGF21-/- mice showed more severe kidney injury compared to wild-type mice, as evidenced by increased levels of renal function markers, inflammatory factors, and fibrosis markers. Notably, exogenous administration of FGF21 to wild-type LN mice markedly mitigated these adverse effects. Additionally, we used tandem mass tag (TMT)-based quantitative proteomics to detect differentially expressed proteins following FGF21 treatment. Results indicated that 121 differentially expressed proteins influenced by FGF21 were involved in biological processes such as immune response and complement activation. Significantly upregulated protein Irgm 1, coupled with modulated inflammatory response, appeared to contribute to the beneficial effects of FGF21. Furthermore, Western blot analysis demonstrated that FGF21 upregulated Irgm 1 while inhibiting nucleotide-binding oligomerization domain-like receptors family pyrin domain including 3 (NLRP3) inflammasome expression. Silencing Irgm 1, in turn, reversed FGF21's inhibitory effect on NLRP3 inflammasome. In summary, FGF21 can potentially alleviate pristane-induced lupus nephritis in mice, possibly through the FGF21/Irgm 1/NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Yimeng Zou
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Dan Wang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Wenying Sun
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Qing Wu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Shijie Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Zeheng Ren
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Yanan Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Tianqi Zhao
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Zhitong Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Xinyu Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Weiyue Cao
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Jiachi Han
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Xiaochen Guo
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Guiping Ren
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China; Research Center of Genetic Engineering of Pharmaceuticals of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
6
|
Liu R, Wen X, Peng X, Zhao M, Mi L, Lei J, Xu K. Immune podocytes in the immune microenvironment of lupus nephritis (Review). Mol Med Rep 2023; 28:204. [PMID: 37711069 PMCID: PMC10540031 DOI: 10.3892/mmr.2023.13091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disorder caused by the loss of tolerance to endogenous nuclear antigens such as double‑stranded DNA, leading to the proliferation of T cells and subsequent activation of B cells, which results in serious organ damage and life‑threatening complications such as lupus nephritis. Lupus nephritis (LN) develops as a frequent complication of SLE, accounting for >60% of SLE cases, and is characterized by proteinuria and heterogeneous histopathological findings. Glomerular injury serves a role in proteinuria as podocyte damage is the leading contributor. Numerous studies have reported that podocytes are involved in the immune response that promotes LN progression. In LN, immune complex deposition stimulates dendritic cells to secrete inflammatory cytokines that activate T cells and B cells. B cells secrete autoantibodies that attack and damage the renal podocytes, leading to renal podocyte injury. The injured podocytes trigger inflammatory cells through the expression of toll‑like receptors and trigger T cells through major histocompatibility complexes and CD86, thereby participating in the local immune response and the exacerbation of podocyte injury. Based on the existing literature, the present review summarizes the research progress of podocytes in LN under the local immune microenvironment of the kidney, explores the mechanism of podocyte injury under the immune microenvironment, and evaluates podocytes as a potential therapeutic target for LN.
Collapse
Affiliation(s)
- Ruiling Liu
- Department of Microbiology and Immunology, Basic Medical College, Shanxi Medical University, Jinzhong, Shanxi 030619, P.R. China
| | - Xiaoting Wen
- Department of Rheumatology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Xinyue Peng
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Miaomiao Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Liangyu Mi
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Jiamin Lei
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Ke Xu
- Department of Rheumatology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| |
Collapse
|
7
|
Shankland SJ, Jefferson JA, Wessely O. Repurposing drugs for diseases associated with podocyte dysfunction. Kidney Int 2023; 104:455-462. [PMID: 37290603 PMCID: PMC11088848 DOI: 10.1016/j.kint.2023.05.018] [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: 02/11/2023] [Revised: 04/02/2023] [Accepted: 05/11/2023] [Indexed: 06/10/2023]
Abstract
The majority of podocyte disorders are progressive in nature leading to chronic kidney disease and often kidney failure. The scope of current therapies is typically nonspecific immunosuppressant medications, which are accompanied by unwanted and serious side effects. However, many exciting clinical trials are underway to reduce the burden of podocyte diseases in our patients. Major advances and discoveries have recently been made experimentally in our understanding of the molecular and cellular mechanisms underlying podocyte injury in disease. This begs the question of how best to take advantage of these impressive strides. One approach to consider is the repurposing of therapeutics that have already been approved by the Food and Drug Administration, European Medicines Agency, and other regulatory agencies for indications beyond the kidney. The advantages of therapy repurposing include known safety profiles, drug development that has already been completed, and overall reduced costs for studying alternative indications for selected therapies. The purpose of this mini review is to examine the experimental literature of podocyte damage and determine if there are mechanistic targets in which prior approved therapies can be considered for repurposing to podocyte disorders.
Collapse
Affiliation(s)
- Stuart J Shankland
- Division of Nephrology, University of Washington, Seattle, Washington, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington, USA.
| | - J Ashley Jefferson
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Oliver Wessely
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA.
| |
Collapse
|
8
|
Xiao L, Matharoo J, Chi J, Ma J, Chen M, Manley B, Xu P, Shi W, Felder RA, Sung SSJ, Jin L, Li X. Transient depletion of macrophages alters local inflammatory response at the site of disc herniation in a transgenic mouse model. Osteoarthritis Cartilage 2023; 31:894-907. [PMID: 36754251 PMCID: PMC10272080 DOI: 10.1016/j.joca.2023.01.574] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 10/06/2022] [Accepted: 01/10/2023] [Indexed: 02/10/2023]
Abstract
OBJECTIVE Macrophages are abundantly detected at sites of disc herniation, however, their function in the disease progression is unclear. We aim to investigate the functions of macrophages in acute disc herniation using a macrophage Fas-induced apoptosis (MaFIA) transgenic mouse strain. METHOD To transiently deplete macrophages, a dimerizer, AP20187, or vehicle solution was administered via intraperitoneal injection to MaFIA mice immediately, day 1 and 2 after annular puncture induced disc herniation. Local infiltrated tissues at disc hernia and DRGs at corresponding levels were harvested to analyze immune cells and neuroinflammation on postoperative day (POD) 6 by flow cytometry and/or immunostaining. Mouse spines were harvested to analyze structures of degenerated discs and adjacent vertebrae and to assess osteoclast activity by histology and tartrate-resistant acid phosphatase (TRAP) staining on POD 6, 13, and 20, respectively. RESULTS On POD 6, abundant macrophages were confirmed at disc hernia sites. Compared to vehicle control, AP20187 significantly reduced GFP+ cells in blood, spleen, and local inflammatory tissue. At disc hernia sites, AP20187 markedly reduced macrophages (CD11b+, F4/80+, GFP+CD11b+, CD11b+F4/80+) while increasing neutrophils and B cells. Transient macrophage depletion decreased ectopic bone formation and osteoclast activity in herniated discs and adjacent cortical bones for up to 20 days post herniation. Disc herniation elevated expressions of TNF-α, IL-6, substance P, calcitonin gene-related peptide, accompanied by increasing GFP+, CD11b+ and F4/80+ macrophages. Macrophage depletion did not attenuate these markers of neuroinflammation. CONCLUSIONS Transient depletion of macrophages altered local inflammatory response at the site of disc herniation.
Collapse
Affiliation(s)
- L Xiao
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - J Matharoo
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - J Chi
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - J Ma
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - M Chen
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - B Manley
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - P Xu
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - W Shi
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22908, USA; Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - R A Felder
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - S-S J Sung
- Department of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - L Jin
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - X Li
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904, USA.
| |
Collapse
|
9
|
Tsai CY, Li KJ, Shen CY, Lu CH, Lee HT, Wu TH, Ng YY, Tsao YP, Hsieh SC, Yu CL. Decipher the Immunopathological Mechanisms and Set Up Potential Therapeutic Strategies for Patients with Lupus Nephritis. Int J Mol Sci 2023; 24:10066. [PMID: 37373215 PMCID: PMC10298725 DOI: 10.3390/ijms241210066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Lupus nephritis (LN) is one of the most severe complications in patients with systemic lupus erythematosus (SLE). Traditionally, LN is regarded as an immune complex (IC) deposition disease led by dsDNA-anti-dsDNA-complement interactions in the subendothelial and/or subepithelial basement membrane of glomeruli to cause inflammation. The activated complements in the IC act as chemoattractants to chemically attract both innate and adaptive immune cells to the kidney tissues, causing inflammatory reactions. However, recent investigations have unveiled that not only the infiltrating immune-related cells, but resident kidney cells, including glomerular mesangial cells, podocytes, macrophage-like cells, tubular epithelial cells and endothelial cells, may also actively participate in the inflammatory and immunological reactions in the kidney. Furthermore, the adaptive immune cells that are infiltrated are genetically restricted to autoimmune predilection. The autoantibodies commonly found in SLE, including anti-dsDNA, are cross-reacting with not only a broad spectrum of chromatin substances, but also extracellular matrix components, including α-actinin, annexin II, laminin, collagen III and IV, and heparan sulfate proteoglycan. Besides, the glycosylation on the Fab portion of IgG anti-dsDNA antibodies can also affect the pathogenic properties of the autoantibodies in that α-2,6-sialylation alleviates, whereas fucosylation aggravates their nephritogenic activity. Some of the coexisting autoantibodies, including anti-cardiolipin, anti-C1q, anti-ribosomal P autoantibodies, may also enhance the pathogenic role of anti-dsDNA antibodies. In clinical practice, the identification of useful biomarkers for diagnosing, monitoring, and following up on LN is quite important for its treatments. The development of a more specific therapeutic strategy to target the pathogenic factors of LN is also critical. We will discuss these issues in detail in the present article.
Collapse
Affiliation(s)
- Chang-Youh Tsai
- Division of Immunology & Rheumatology, Department of Medicine, Fu Jen Catholic University Hospital & College of Medicine, Fu Jen Catholic University, New Taipei City 24352, Taiwan
| | - Ko-Jen Li
- Division of Rheumatology, Immunology & Allergy, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 106319, Taiwan; (K.-J.L.); (C.-Y.S.); (C.-H.L.); (S.-C.H.)
| | - Chieh-Yu Shen
- Division of Rheumatology, Immunology & Allergy, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 106319, Taiwan; (K.-J.L.); (C.-Y.S.); (C.-H.L.); (S.-C.H.)
| | - Cheng-Hsun Lu
- Division of Rheumatology, Immunology & Allergy, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 106319, Taiwan; (K.-J.L.); (C.-Y.S.); (C.-H.L.); (S.-C.H.)
| | - Hui-Ting Lee
- MacKay Memorial Hospital & MacKay Medical College, New Taipei City 25245, Taiwan;
| | - Tsai-Hung Wu
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital and Faculty of Medicine, National Yang-Ming Chiao-Tung University, Taipei 112304, Taiwan;
| | - Yee-Yung Ng
- Department of Medicine, Fu Jen Catholic University Hospital & College of Medicine, Fu Jen Catholic University, New Taipei City 24352, Taiwan;
| | - Yen-Po Tsao
- Division of Holistic and Multidisciplinary Medicine, Department of Medicine, Taipei Veterans General Hospital and Faculty of Medicine, National Yang-Ming Chiao-Tung University, Taipei 112304, Taiwan;
| | - Song-Chou Hsieh
- Division of Rheumatology, Immunology & Allergy, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 106319, Taiwan; (K.-J.L.); (C.-Y.S.); (C.-H.L.); (S.-C.H.)
| | - Chia-Li Yu
- Division of Rheumatology, Immunology & Allergy, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 106319, Taiwan; (K.-J.L.); (C.-Y.S.); (C.-H.L.); (S.-C.H.)
| |
Collapse
|
10
|
Lin W, Lin Z, Lin X, Peng Z, Liang X, Wei S. Integrated analysis and clinical correlation analysis of hub genes, immune infiltration, and potential therapeutic agents related to lupus nephritis. Lupus 2023; 32:633-643. [PMID: 36912500 DOI: 10.1177/09612033231161587] [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: 03/14/2023]
Abstract
BACKGROUND Lupus nephritis (LN) is the most common complication of systemic lupus erythematosus (SLE). This study aimed to explore biomarkers, mechanisms, and potential novel agents regarding LN through bioinformatic analysis. METHOD Four expression profiles were downloaded from the Gene Expression Omnibus (GEO) database and differentially expressed genes (DEGs) were acquired. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGGs) pathway enrichment analyses of DEGs were performed using the R software. The protein-protein interaction (PPI) network was developed using the STRING database. Additionally, five algorithms were used to screen out the hub genes. Expression of the hub genes were validated using Nephroseq v5. CIBERSORT was used to evaluate the infiltration of immune cells. Finally, The Drug-Gene Interaction Database was used to predict potential targeted drugs. RESULT FOS and IGF1 were identified as hub genes, with excellent specificity and sensitivity diagnosis of LN. FOS was also related to renal injury. LN patients had lower activated and resting dendritic cells (DCs) and higher M1 macrophages and activated NK cells than healthy control (HC). FOS had a positive correlation with activated mast cells and a negative correlation with resting mast cells. IGF1 had a positive correlation with activated DCs and a negative correlation with monocytes. The targeted drugs were dusigitumab and xentuzumab target for IGF1. CONCLUSION We analyzed the transcriptomic signature of LN along with the landscape of the immune cell. FOS and IGF1 are promising biomarkers for diagnosing and evaluating the progression of LN. The drug-gene interaction analyses provide a list of candidate drugs for the precise treatment of LN.
Collapse
Affiliation(s)
- Weiyi Lin
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Zien Lin
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Xiaobing Lin
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Zhishen Peng
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Xiaofeng Liang
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Shanshan Wei
- Department of Dermatology, Zhujiang Hospital, 70570Southern Medical University, Guangzhou, The People's Republic of China
| |
Collapse
|
11
|
Daamen AR, Wang H, Bachali P, Shen N, Kingsmore KM, Robl RD, Grammer AC, Fu SM, Lipsky PE. Molecular mechanisms governing the progression of nephritis in lupus prone mice and human lupus patients. Front Immunol 2023; 14:1147526. [PMID: 36936908 PMCID: PMC10016352 DOI: 10.3389/fimmu.2023.1147526] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/15/2023] [Indexed: 03/04/2023] Open
Abstract
Introduction Pathologic inflammation is a major driver of kidney damage in lupus nephritis (LN), but the immune mechanisms of disease progression and risk factors for end organ damage are poorly understood. Methods To characterize molecular profiles through the development of LN, we carried out gene expression analysis of microdissected kidneys from lupus-prone NZM2328 mice. We examined male mice and the congenic NZM2328.R27 strain as a means to define mechanisms associated with resistance to chronic nephritis. Gene expression profiles in lupus mice were compared with those in human LN. Results NZM2328 mice exhibited progress from acute to transitional and then to chronic glomerulonephritis (GN). Each stage manifested a unique molecular profile. Neither male mice nor R27 mice progressed past the acute GN stage, with the former exhibiting minimal immune infiltration and the latter enrichment of immunoregulatory gene signatures in conjunction with robust kidney tubule cell profiles indicative of resistance to cellular damage. The gene expression profiles of human LN were similar to those noted in the NZM2328 mouse suggesting comparable stages of LN progression. Conclusions Overall, this work provides a comprehensive examination of the immune processes involved in progression of murine LN and thus contributes to our understanding of the risk factors for end-stage renal disease. In addition, this work presents a foundation for improved classification of LN and illustrates the applicability of murine models to identify the stages of human disease.
Collapse
Affiliation(s)
| | - Hongyang Wang
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
- Division of Rheumatology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | | - Nan Shen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Robert D. Robl
- AMPEL BioSolutions LLC, Charlottesville, VA, United States
| | | | - Shu Man Fu
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
- Division of Rheumatology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | |
Collapse
|
12
|
Xiang C, Li H, Tang W. Targeting CSF-1R represents an effective strategy in modulating inflammatory diseases. Pharmacol Res 2023; 187:106566. [PMID: 36423789 DOI: 10.1016/j.phrs.2022.106566] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/12/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Colony-stimulating factor-1 receptor (CSF-1R), also known as FMS kinase, is a type I single transmembrane protein mainly expressed in myeloid cells, such as monocytes, macrophages, glial cells, and osteoclasts. The endogenous ligands, colony-stimulating factor-1 (CSF-1) and Interleukin-34 (IL-34), activate CSF-1R and downstream signaling pathways including PI3K-AKT, JAK-STATs, and MAPKs, and modulate the proliferation, differentiation, migration, and activation of target immune cells. Over the past decades, the promising therapeutic potential of CSF-1R signaling inhibition has been widely studied for decreasing immune suppression and escape in tumors, owing to depletion and reprogramming of tumor-associated macrophages. In addition, the excessive activation of CSF-1R in inflammatory diseases is consecutively uncovered in recent years, which may result in inflammation in bone, kidney, lung, liver and central nervous system. Agents against CSF-1R signaling have been increasingly investigated in preclinical or clinical studies for inflammatory diseases treatment. However, the pathological mechanism of CSF-1R in inflammation is indistinct and whether CSF-1R signaling can be identified as biomarkers remains controversial. With the background information aforementioned, this review focus on the dialectical roles of CSF-1R and its ligands in regulating innate immune cells and highlights various therapeutic implications of blocking CSF-1R signaling in inflammatory diseases.
Collapse
Affiliation(s)
- Caigui Xiang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Heng Li
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Wei Tang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
13
|
Liu M, Zhang L, Wang Y, Hu W, Wang C, Wen Z. Mesangial cell: A hub in lupus nephritis. Front Immunol 2022; 13:1063497. [PMID: 36591251 PMCID: PMC9795068 DOI: 10.3389/fimmu.2022.1063497] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Lupus nephritis (LN) is a severe renal disease caused by the massive deposition of the immune complexes (ICs) in renal tissue, acting as one of the significant organ manifestations of systemic lupus erythematosus (SLE) and a substantial cause of death in clinical patients. As mesangium is one of the primary sites for IC deposition, mesangial cells (MCs) constantly undergo severe damage, resulting in excessive proliferation and increased extracellular matrix (ECM) production. In addition to playing a role in organizational structure, MCs are closely related to in situ immunomodulation by phagocytosis, antigen-presenting function, and inflammatory effects, aberrantly participating in the tissue-resident immune responses and leading to immune-mediated renal lesions. Notably, such renal-resident immune responses drive a second wave of MC damage, accelerating the development of LN. This review summarized the damage mechanisms and the in situ immune regulation of MCs in LN, facilitating the current drug research for exploring clinical treatment strategies.
Collapse
Affiliation(s)
- Mengdi Liu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Lei Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Yixin Wang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Weijie Hu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Chunhong Wang
- Cyrus Tang Hematology Center, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China,*Correspondence: Zhenke Wen, ; Chunhong Wang,
| | - Zhenke Wen
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China,*Correspondence: Zhenke Wen, ; Chunhong Wang,
| |
Collapse
|
14
|
Inhibition of the chemokine signal regulator FROUNT by disulfiram ameliorates crescentic glomerulonephritis. Kidney Int 2022; 102:1276-1290. [PMID: 36049642 DOI: 10.1016/j.kint.2022.07.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 07/01/2022] [Accepted: 07/20/2022] [Indexed: 01/12/2023]
Abstract
Activated monocytes/macrophages promote glomerular injury, including crescent formation, in anti-glomerular basement membrane (GBM) glomerulonephritis. Disulfiram, an alcohol-aversion drug, inhibits monocyte/macrophage migration by inhibiting FROUNT, a cytosolic protein that enhances chemokine receptor signaling. Our study found that disulfiram at a human equivalent dose successfully blocked albuminuria and crescent formation with podocyte loss, and later stage kidney fibrotic lesions, in a rat model of anti-GBM glomerulonephritis. A disulfiram derivative, DSF-41, with more potent FROUNT inhibition activity, inhibited glomerulonephritis at a lower dose than disulfiram. Disulfiram markedly reduced the number of monocytes or macrophages at the early stage of glomerulonephritis and that of CD3+ and CD8+ lymphocytes at the established stage. Impaired pseudopodia formation was observed in the glomerular monocytes/macrophages of the disulfiram group; consistent with the in vitro observation that disulfiram blocked chemokine-dependent pseudopodia formation and chemotaxis of bone marrow-derived monocytes/macrophages. Furthermore, disulfiram suppressed macrophage activation as revealed by reduced expression of inflammatory cytokines and chemokines (TNF-α, CCL2, and CXCL9) and reduced CD86 and MHC class II expressions in monocytes/macrophages during glomerulonephritis. The dramatic reduction in monocyte/macrophage number might have resulted from disulfiram suppression of both the chemotactic response of monocytes/macrophages and their subsequent activation to produce cytokines and chemokines, which further recruit monocytes. Additionally, FROUNT was expressed in CD68+ monocytes/macrophages infiltrating the crescentic glomeruli in human anti-GBM glomerulonephritis. Thus, disulfiram can be a highly effective and safe drug for the treatment of glomerulonephritis by blocking the chemotactic responses of monocytes/macrophages and their activation status in the glomerulus.
Collapse
|
15
|
Lou H, Ling GS, Cao X. Autoantibodies in systemic lupus erythematosus: From immunopathology to therapeutic target. J Autoimmun 2022; 132:102861. [PMID: 35872103 DOI: 10.1016/j.jaut.2022.102861] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 11/26/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multiple organ inflammatory damage and wide spectrum of autoantibodies. The autoantibodies, especially anti-dsDNA and anti-Sm autoantibodies are highly specific to SLE, and participate in the immune complex formation and inflammatory damage on multiple end-organs such as kidney, skin, and central nervous system (CNS). However, the underlying mechanisms of autoantibody-induced tissue damage and systemic inflammation are still not fully understood. Single cell analysis of autoreactive B cells and monoclonal antibody screening from patients with active SLE has improved our understanding on the origin of autoreactive B cells and the antigen targets of the pathogenic autoantibodies. B cell depletion therapies have been widely studied in the clinics, but the development of more specific therapies against the pathogenic B cell subset and autoantibodies with improved efficacy and safety still remain a big challenge. A more comprehensive autoantibody profiling combined with functional characterization of autoantibodies in diseases development will shed new insights on the etiology and pathogenesis of SLE and guide a specific treatment to individual SLE patients.
Collapse
Affiliation(s)
- Hantao Lou
- Ludwig Institute of Cancer Research, University of Oxford, Oxford, OX3 7DR, UK; Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK.
| | - Guang Sheng Ling
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xuetao Cao
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK; Nankai-Oxford International Advanced Institute, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
16
|
Quaglia M, Merlotti G, Fornara L, Colombatto A, Cantaluppi V. Extracellular Vesicles Released from Stem Cells as a New Therapeutic Strategy for Primary and Secondary Glomerulonephritis. Int J Mol Sci 2022; 23:ijms23105760. [PMID: 35628570 PMCID: PMC9142886 DOI: 10.3390/ijms23105760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/13/2022] [Accepted: 05/20/2022] [Indexed: 12/04/2022] Open
Abstract
Current treatment of primary and secondary glomerulopathies is hampered by many limits and a significant proportion of these disorders still evolves towards end-stage renal disease. A possible answer to this unmet challenge could be represented by therapies with stem cells, which include a variety of progenitor cell types derived from embryonic or adult tissues. Stem cell self-renewal and multi-lineage differentiation ability explain their potential to protect and regenerate injured cells, including kidney tubular cells, podocytes and endothelial cells. In addition, a broad spectrum of anti-inflammatory and immunomodulatory actions appears to interfere with the pathogenic mechanisms of glomerulonephritis. Of note, mesenchymal stromal cells have been particularly investigated as therapy for Lupus Nephritis and Diabetic Nephropathy, whereas initial evidence suggest their beneficial effects in primary glomerulopathies such as IgA nephritis. Extracellular vesicles mediate a complex intercellular communication network, shuttling proteins, nucleic acids and other bioactive molecules from origin to target cells to modulate their functions. Stem cell-derived extracellular vesicles recapitulate beneficial cytoprotective, reparative and immunomodulatory properties of parental cells and are increasingly recognized as a cell-free alternative to stem cell-based therapies for different diseases including glomerulonephritis, also considering the low risk for potential adverse effects such as maldifferentiation and tumorigenesis. We herein summarize the renoprotective potential of therapies with stem cells and extracellular vesicles derived from progenitor cells in glomerulonephritis, with a focus on their different mechanisms of actions. Technological progress and growing knowledge are paving the way for wider clinical application of regenerative medicine to primary and secondary glomerulonephritis: this multi-level, pleiotropic therapy may open new scenarios overcoming the limits and side effects of traditional treatments, although the promising results of experimental models need to be confirmed in the clinical setting.
Collapse
|
17
|
Jin L, Xiao L, Ding M, Pan A, Balian G, Sung SSJ, Li XJ. Heterogeneous macrophages contribute to the pathology of disc herniation induced radiculopathy. Spine J 2022; 22:677-689. [PMID: 34718176 PMCID: PMC8957503 DOI: 10.1016/j.spinee.2021.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Macrophages play important roles in the progression of intervertebral disc herniation and radiculopathy. PURPOSE To better understand the roles of macrophages in this process, we developed a new mouse model that mimics human radiculopathy. STUDY DESIGN/SETTING A preclinical randomized animal study. METHODS Three types of surgeries were performed in randomly assigned Balb/c mice. These were spinal nerve exposure, traditional anterior disc puncture, and lateral disc puncture with nerve exposure (n=16/group). For the nerve exposure group, the left L5 spinal nerve was exposed without disc injury. For the traditional anterior puncture, L5/6 disc was punctured by an anterior approach as previously established. For lateral puncture with nerve exposure, the left L5 spinal nerve was exposed by removing the psoas major muscle fibers, and the L5/6 disc was punctured laterally on the left side with a 30G needle, allowing the nucleus to protrude toward the L5 spinal nerve. Mechanical hyperalgesia (pain sensitivity) of hind paws was assessed with electronic von Frey assay on alternative day for up to 2 weeks. MRI, histology, and immunostaining were performed to confirm disc herniation and inflammation. RESULTS Ipsilateral pain in the lateral puncture with nerve exposure group was significantly greater than the other groups. Pro-inflammatory cytokines IL-1β and IL-6 were markedly elevated at the hernia sites of both puncture groups and the spinal nerve of lateral puncture with never exposure group on postoperative day 7. Heterogeneous populations of macrophages were detected in the infiltration tissue of this mouse model and in tissue from patients undergone discectomy. CONCLUSIONS We have established a new mouse model that mimics human radiculopathy and demonstrated that a mixed phenotype of macrophages contribute to the pathogenesis of acute discogenic radiculopathy. CLINICAL SIGNIFICANCE This study provides a clinically relevant in vivo animal model to elucidate complex interactions of disc herniation and radicular pain, which may present opportunities for the development of macrophage-anchored therapeutics to manage radiculopathy.
Collapse
Affiliation(s)
- Li Jin
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Li Xiao
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Mengmeng Ding
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA; Department of Anesthesiology, Shengjing hospital, China Medical University, Shenyang, China
| | - Aixing Pan
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA; Department of Orthopaedic Surgery, Chaoyang Hospital, Capital Medical School, Beijing, China
| | - Gary Balian
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA; Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Sun-Sang J Sung
- Department of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Xudong Joshua Li
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904, USA.
| |
Collapse
|
18
|
Dong Z, Dai H, Liu W, Jiang H, Feng Z, Liu F, Zhao Q, Rui H, Liu WJ, Liu B. Exploring the Differences in Molecular Mechanisms and Key Biomarkers Between Membranous Nephropathy and Lupus Nephritis Using Integrated Bioinformatics Analysis. Front Genet 2022; 12:770902. [PMID: 35047003 PMCID: PMC8762271 DOI: 10.3389/fgene.2021.770902] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 12/06/2021] [Indexed: 01/16/2023] Open
Abstract
Background: Both membranous nephropathy (MN) and lupus nephritis (LN) are autoimmune kidney disease. In recent years, with the deepening of research, some similarities have been found in the pathogenesis of these two diseases. However, the mechanism of their interrelationship is not clear. The purpose of this study was to investigate the differences in molecular mechanisms and key biomarkers between MN and LN. Method: The expression profiles of GSE99325, GSE99339, GSE104948 and GSE104954 were downloaded from GEO database, and the differentially expressed genes (DEGs) of MN and LN samples were obtained. We used Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) for enrichment analysis of DEGs. A protein-protein interaction (PPI) network of DEGs was constructed using Metascape. We filtered DEGs with NetworkAnalyst. Finally, we used receiver operating characteristic (ROC) analysis to identify the most significant DEGs for MN and LN. Result: Compared with LN in the glomerulus, 14 DEGs were up-regulated and 77 DEGs were down-regulated in MN. Compared with LN in renal tubules, 21 DEGs were down-regulated, but no up-regulated genes were found in MN. According to the result of GO and KEGG enrichment, PPI network and Networkanalyst, we screened out six genes (IFI6, MX1, XAF1, HERC6, IFI44L, IFI44). Interestingly, among PLA2R, THSD7A and NELL1, which are the target antigens of podocyte in MN, the expression level of NELL1 in MN glomerulus is significantly higher than that of LN, while there is no significant difference in the expression level of PLA2R and THSD7A. Conclusion: Our study provides new insights into the pathogenesis of MN and LN by analyzing the differences in gene expression levels between MN and LN kidney samples, and is expected to be used to prepare an animal model of MN that is more similar to human.
Collapse
Affiliation(s)
- Zhaocheng Dong
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Haoran Dai
- Shunyi Branch, Beijing Traditional Chinese Medicine Hospital, Beijing, China
| | - Wenbin Liu
- Beijing University of Chinese Medicine, Beijing, China
| | - Hanxue Jiang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Zhendong Feng
- Beijing Chinese Medicine Hospital Pinggu Hospital, Beijing, China
| | - Fei Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Qihan Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Capital Medical University, Beijing, China
| | - Hongliang Rui
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Wei Jing Liu
- Renal Research Institution of Beijing University of Chinese Medicine, and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Baoli Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Shunyi Branch, Beijing Traditional Chinese Medicine Hospital, Beijing, China
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Mesangial cells are critical for the proper function of the glomerulus, playing roles in structural support and injury repair. However, they are also early responders to glomerular immune complex deposition and contribute to inflammation and fibrosis in lupus nephritis. This review highlights recent studies identifying signaling pathways and mediators in mesangial cell response to lupus-relevant stimuli. RECENT FINDINGS Anti-dsDNA antibodies, serum, or plasma from individuals with lupus nephritis, or specific pathologic factors activated multiple signaling pathways. These pathways largely included JAK/STAT/SOCS, PI3K/AKT, and MAPK and led to induction of proliferation and expression of multiple proinflammatory cytokines, growth factors, and profibrotic factors. NFκB activation was a common mediator of response. Mesangial cells proliferate and express a wide array of proinflammatory/profibrotic factors in response to a variety of lupus-relevant pathologic stimuli. While some of the responses are similar, the mechanisms involved appear to be diverse depending on the stimulus. Future studies are needed to fully elucidate these mechanisms with respect to the diverse milieu of stimuli.
Collapse
Affiliation(s)
- Tamara K Nowling
- Department of Medicine, Division of Rheumatology, Medical University of South Carolina, 96 Jonathan Lucas St. CSB 822 MSC 637, Charleston, SC, 29425-6370, USA.
| |
Collapse
|
20
|
Uprety LP, Park YH, Jang YJ. Autoantigen spermatid nuclear transition protein 1 enhances pro-inflammatory cytokine production stimulated by anti-DNA autoantibodies in macrophages. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221131792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Introduction Lupus nephritis (LN), a severe manifestation of systemic lupus erythematosus (SLE), is associated with high fatality rate in patients. The pathogenesis of lupus nephritis is complex and has not been fully elucidated. Kidney inflammation, renal cell damage, and accumulation of immune complexes in the glomerular basement membrane often occur in patients with lupus nephritis. Spermatid nuclear transition protein 1 (TNP1) might be a potentially interesting autoantigen in exploring the pathogenesis and therapy of lupus nephritis. Objective This study aimed to explore the effect of TNP1 and its complexes with anti-double-stranded DNA antibodies on the levels of interleukin-6 (IL-6) and interferon-α (IFN-α) in vitro. Methods We studied the effect of the synthetic peptide of the autoantigen on the pathogenic characteristics of the G2-6 and G5-8 antibodies in mouse macrophages, using enzyme-linked immunosorbent assay, quantitative RT-PCR, western blotting, and flow cytometry. Results The antibodies exhibited cross-reactivity to spermatid TNP1 in direct-binding and competitive enzyme-linked immunosorbent assay. Results of quantitative RT-PCR and western blotting revealed that the antibodies alone enhanced the levels of IL-6 and IFN-α transcripts and proteins, respectively. Flow cytometry revealed that treatment with the autoantigen enhanced the cell-penetrating activities of G2-6 and G5-8 and remarkably increased the cytokine levels. Conclusion TNP1 enhanced the cell-penetrating activities of anti-dsDNA auto-Abs, G2-6 and G5-8, and remarkably increased the levels of IL-6 and IFN-α in macrophages, suggesting that TNP1 and cell-penetrating pathogenic anti-dsDNA auto-Abs is potential targets for future therapeutic approaches to treat LN/SLE.
Collapse
Affiliation(s)
- Laxmi Prasad Uprety
- Department of Medical Sciences, Graduate School of Ajou University, Suwon, South Korea
- Department of Microbiology, School of Medicine, Ajou University, Suwon, South Korea
| | - Yong Hwan Park
- Department of Medical Sciences, Graduate School of Ajou University, Suwon, South Korea
- Department of Microbiology, School of Medicine, Ajou University, Suwon, South Korea
| | - Young-Ju Jang
- Department of Medical Sciences, Graduate School of Ajou University, Suwon, South Korea
- Department of Microbiology, School of Medicine, Ajou University, Suwon, South Korea
| |
Collapse
|
21
|
Shen L, Sun W, Zhang Q, Wei M, Xu H, Luo X, Wang G, Zhou F. Deep Learning-Based Model Significantly Improves Diagnostic Performance for Assessing Renal Histopathology in Lupus Glomerulonephritis. KIDNEY DISEASES 2022; 8:347-356. [PMID: 36157261 PMCID: PMC9386416 DOI: 10.1159/000524880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 05/02/2022] [Indexed: 01/08/2023]
Abstract
<b><i>Background:</i></b> Assessment of glomerular lesions and structures plays an essential role in understanding the pathological diagnosis of glomerulonephritis and prognostic evaluation of many kidney diseases. Renal pathophysiological assessment requires novel high-throughput tools to conduct quantitative, unbiased, and reproducible analyses representing a central readout. Deep learning may be an effective tool for glomerulonephritis pathological analysis. <b><i>Methods:</i></b> We developed a murine renal pathological system (MRPS) model to objectify the pathological evaluation via the deep learning method on whole-slide image (WSI) segmentation and feature extraction. A convolutional neural network model was used for accurate segmentation of glomeruli and glomerular cells of periodic acid-Schiff-stained kidney tissue from healthy and lupus nephritis mice. To achieve a quantitative evaluation, we subsequently filtered five independent predictors as image biomarkers from all features and developed a formula for the scoring model. <b><i>Results:</i></b> Perimeter, shape factor, minimum internal diameter, minimum caliper diameter, and number of objects were identified as independent predictors and were included in the establishment of the MRPS. The MRPS showed a positive correlation with renal score (<i>r</i> = 0.480, <i>p</i> < 0.001) and obtained great diagnostic performance in discriminating different score bands (Obuchowski index, 0.842 [95% confidence interval: 0.759, 0.925]), with an area under the curve of 0.78–0.98, sensitivity of 58–93%, specificity of 72–100%, and accuracy of 74–94%. <b><i>Conclusion:</i></b> Our MRPS for quantitative assessment of renal WSIs from MRL/lpr lupus nephritis mice enables accurate histopathological analyses with high reproducibility, which may serve as a useful tool for glomerulonephritis diagnosis and prognosis evaluation.
Collapse
Affiliation(s)
- Luping Shen
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Wenyi Sun
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qixiang Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Mengru Wei
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Huanke Xu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xuan Luo
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- *Guangji Wang,
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- **Fang Zhou,
| |
Collapse
|
22
|
Murakami Y, Fukui R, Tanaka R, Motoi Y, Kanno A, Sato R, Yamaguchi K, Amano H, Furukawa Y, Suzuki H, Suzuki Y, Tamura N, Yamashita N, Miyake K. Anti-TLR7 Antibody Protects Against Lupus Nephritis in NZBWF1 Mice by Targeting B Cells and Patrolling Monocytes. Front Immunol 2021; 12:777197. [PMID: 34868046 PMCID: PMC8632649 DOI: 10.3389/fimmu.2021.777197] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/25/2021] [Indexed: 01/27/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by autoantibody production and multiple organ damage. Toll-like receptor 7 (TLR7), an innate immune RNA sensor expressed in monocytes/macrophages, dendritic cells (DCs), and B cells, promotes disease progression. However, little is known about the cellular mechanisms through which TLR7 drives lupus nephritis. Here, we show that the anti-mouse TLR7 mAb, but not anti-TLR9 mAb, protected lupus-prone NZBWF1 mice from nephritis. The anti-TLR7 mAb reduced IgG deposition in glomeruli by inhibiting the production of autoantibodies to the RNA-associated antigens. We found a disease-associated increase in Ly6Clow patrolling monocytes that expressed high levels of TLR7 and had upregulated expression of lupus-associated IL-10, CD115, CD31, and TNFSF15 in NZBWF1 mice. Anti-TLR7 mAb abolished this lupus-associated increase in patrolling monocytes in the circulation, spleen, and glomeruli. These results suggested that TLR7 drives autoantibody production and lupus-associated monocytosis in NZBWF1 mice and, that anti-TLR7 mAb is a promising therapeutic tool targeting B cells and monocytes/macrophages.
Collapse
Affiliation(s)
- Yusuke Murakami
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Faculty of Pharmacy, Department of Pharmaceutical Sciences & Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Reika Tanaka
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuji Motoi
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Atsuo Kanno
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryota Sato
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hirofumi Amano
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hitoshi Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Naoto Tamura
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Naomi Yamashita
- Faculty of Pharmacy, Department of Pharmaceutical Sciences & Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Innate Immunity, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
23
|
Du Y, Shi X, Li J, Jia Y. MicroRNA-98-5p inhibits human mesangial cell proliferation and TNF-α and IL-6 secretion by targeting BTB and CNC homology 1. Exp Ther Med 2021; 22:1436. [PMID: 34721678 PMCID: PMC8549099 DOI: 10.3892/etm.2021.10871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
MicroRNA (miR)-98-5p has been reported to be involved in the development of lupus nephritis (LN); however, its specific role in LN remains unclear. The present study aimed to investigate the effect of miR-98-5p on human mesangial cell proliferation and the secretion of TNF-α and IL-6. Reverse transcription-quantitative PCR (RT-qPCR) and western blotting were used to analyze the level of gene and protein expression, respectively. Cellular proliferation was assessed using a Cell Counting Kit-8 (CCK-8) assay. ELISA was used to detect the secretion of TNF-α and IL-6 by human mesangial cells. RT-qPCR analysis revealed that miR-98-5p expression was downregulated in LN renal tissues compared with control renal tissues. Overexpression of miR-98-5p inhibited human mesangial cell proliferation and the secretion of TNF-α and IL-6, whereas miR-98-5p-knockdown demonstrated the opposite effect. Dual luciferase reporter assays demonstrated that miR-98-5p directly targeted BTB and CNC homology 1 (BACH1). BACH1-overexpression promoted human mesangial cell proliferation and the secretion of TNF-α and IL-6, whereas BACH1-knockdown demonstrated the opposite effect. Notably, co-transfection with miR-98-5p mimic inhibited BACH1-overexpression induced human mesangial cell proliferation and the secretion of TNF-α and IL-6. The results of the present study indicated that miR-98-5p inhibited human mesangial cell proliferation and the secretion of TNF-α and IL-6 by targeting BACH1. Therefore, miR-98-5p and BACH1 may represent potential therapeutic targets for LN.
Collapse
Affiliation(s)
- Yong Du
- Department of Rheumatology and Immunology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Xiaoqiang Shi
- Department of Urology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Jing Li
- Department of Nephrology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Yanhui Jia
- Department of Rheumatology and Immunology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| |
Collapse
|
24
|
Davidson A. Renal Mononuclear Phagocytes in Lupus Nephritis. ACR Open Rheumatol 2021; 3:442-450. [PMID: 34060247 PMCID: PMC8280821 DOI: 10.1002/acr2.11269] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/29/2021] [Indexed: 01/16/2023] Open
Abstract
Renal mononuclear phagocytes are a highly pleiotropic group of immune cells of myeloid origin that play multiple protective and pathogenic roles in tissue homeostasis, inflammation, repair, and fibrosis. Infiltration of kidneys with these cells is a hallmark of lupus nephritis and is associated with more severe disease and with increased risk of progression to end‐stage renal disease. This review presents current knowledge of the diversity of these cells and their involvement in kidney inflammation and resolution and describes how they contribute to the chronic inflammation of lupus nephritis. A better understanding of the subset heterogeneity and diverse functions of mononuclear phagocytes in the lupus nephritis kidney should provide fertile ground for the development of new therapeutic approaches that promote the differentiation and survival of protective subsets while targeting pathogenic cell subsets that cause inflammation and fibrosis.
Collapse
Affiliation(s)
- Anne Davidson
- Feinstein Institutes for Medical Research, Manhasset, New York
| |
Collapse
|
25
|
Rodgers J, Sundararaj K, Bruner E, Wolf B, Nowling TK. The role of neuraminidase 1 (NEU1) in cytokine release by primary mouse mesangial cells and disease outcomes in murine lupus nephritis. Autoimmunity 2021; 54:163-175. [PMID: 33749450 DOI: 10.1080/08916934.2021.1897978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The importance of altered glycosphingolipid (GSL) metabolism is increasingly gaining attention as a characteristic of multiple chronic kidney diseases. Previously, we reported elevated levels of GSLs and neuraminidase (NEU) enzyme activity/expression in the urine or kidney of lupus patients and lupus-prone mice, and demonstrated NEU activity mediates the production of cytokines by lupus-prone mouse primary mesangial cells. This mediation occurs in part through TLR4 and p38/ERK MAPK signalling in response to lipopolysaccharide (LPS) and lupus serum (LS). However, the precise role of NEU1, the most abundant NEU in the kidney, is incompletely known. In this study, we investigated the effect of genetically reduced Neu1 levels in vitro and in vivo. Mesangial cells from non-autoimmune prone Neu1+/- C57BL/6 mice had significantly reduced NEU activity, cytokine expression and cytokine secretion in response to LS and LPS, thereby suggesting reducing Neu1 expression may reduce the inflammatory response in lupus nephritis. Disease was assessed in female B6.SLE1/2/3 lupus-prone mice with genetically reduced levels (Neu1+/-) or wild-type levels (Neu1+/+) of Neu1 from 28 to 44 weeks of age along with aged-matched C57BL/6 controls. Renal disease was unexpectedly mild in all B6.SLE1/2/3 mice despite evidence of systemic disease. B6.SLE1/2/3 Neu1+/- mice exhibited significantly reduced levels of renal NEU1 expression and changes in renal α-2,6 linked sialylated N-glycans compared to the Neu1+/+ or healthy C57BL/6 mice, but measures of renal and systemic disease were similar between the B6.SLE1/2/3 Neu1+/+ and Neu1+/- mice. We conclude that NEU1 is the NEU largely responsible for mediating cytokine release by mesangial cells, at least in vitro, but may not be involved in modulating renal GSL levels in vivo or impact onset of nephritis in lupus-prone mice. However, the effect of reduced NEU1 levels on disease may not be appreciated in the mild disease expression in our colony of B6.SLE1/2/3 mice. The impact of the altered renal sialylated N-glycan levels and potential role of NEU1 with respect to established nephritis (late disease) in lupus-prone mice bears further investigation.
Collapse
Affiliation(s)
- Jessalyn Rodgers
- Department of Medicine, Division of Rheumatology, Medical University of South Carolina, Charleston, SC, USA
| | - Kamala Sundararaj
- Department of Medicine, Division of Rheumatology, Medical University of South Carolina, Charleston, SC, USA
| | - Evelyn Bruner
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Bethany Wolf
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Tamara K Nowling
- Department of Medicine, Division of Rheumatology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
26
|
Thanou A, Jupe E, Purushothaman M, Niewold TB, Munroe ME. Clinical disease activity and flare in SLE: Current concepts and novel biomarkers. J Autoimmun 2021; 119:102615. [PMID: 33631651 DOI: 10.1016/j.jaut.2021.102615] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 12/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is a complex and heterogeneous systemic autoimmune disease associated with innate and adaptive immune dysregulation. SLE occurs primarily in females of childbearing age, with increased prevalence and severity in minority populations. Despite improvements in treatment modalities, SLE patients frequently experience periods of heightened disease activity and flare that can lead to permanent organ damage, increased morbidity, and early mortality. Such outcomes impair quality of life and inflict a significant socioeconomic burden. Predicting changes in SLE disease activity could allow for closer monitoring and preemptive treatment, but existing clinical, demographic and serologic markers have been only modestly predictive. Novel, proactive approaches to clinical disease management are thus critically needed. Panels of blood biomarkers can detect a breadth of immune pathway dysregulation that captures SLE heterogeneity and disease activity. Alterations in the balance of pro-inflammatory and regulatory soluble mediators have been associated with changes in clinical disease activity and are detectable several weeks prior to clinical flare occurrence. A soluble mediator score has been highly predictive of impending flare in both European American and African American SLE patients, and this score does not require a priori knowledge of specific pathway activation in the patient. We review current concepts of disease activity and flare in SLE, focusing on the potential of novel blood biomarkers to characterize and predict changes in disease activity. Measuring the disordered immune response in SLE in this way promises to improve disease management and prevent organ damage in SLE.
Collapse
Affiliation(s)
| | - Eldon Jupe
- Progentec Diagnostics, Inc., Oklahoma City, OK, USA
| | | | - Timothy B Niewold
- Colton Center for Autoimmunity, New York University School of Medicine, New York, NY, USA
| | | |
Collapse
|
27
|
Tan L, Zhao M, Wu H, Zhang Y, Tong X, Gao L, Zhou L, Lu Q, Zeng J. Downregulated Serum Exosomal miR-451a Expression Correlates With Renal Damage and Its Intercellular Communication Role in Systemic Lupus Erythematosus. Front Immunol 2021; 12:630112. [PMID: 33643314 PMCID: PMC7906989 DOI: 10.3389/fimmu.2021.630112] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease characterized by continuous inflammation and the production of autoantibodies. Exosomes, acting as a critical tool for communication between cells, are involved in the pathogenesis of SLE, particularly in inflammation and immune imbalance. In this study, we aimed to extract and confirm the pro-inflammatory effect of serum exosomes in SLE. Then, we attempted to find differentially expressed exosomal microRNAs in the serum of healthy subjects and SLE patients via miRNA microarray analysis and validated the target exosomal microRNA, exosomal miR-451a, which expression level decreased in serum of SLE patients by RT-qPCR. Furtherly, we analyzed the correlation between exosomal miR-451a and disease activity, kidney damage and typing, and traditional medicine therapy. Finally, we investigated the intercellular communication role of exosomal miR-451a in SLE by co-culture assay in vitro. Taken together, our study demonstrated that downregulated serum exosomal miR-451a expression correlated with SLE disease activity and renal damage as well as its intercellular communication role in SLE which provided potential therapeutic strategies.
Collapse
Affiliation(s)
- Lina Tan
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yuezhong Zhang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiaoliang Tong
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Gao
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lu Zhou
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jinrong Zeng
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
28
|
Two Metal–Organic Frameworks: Luminescent Detection Performance and Treatment Activity on Chronic Glomerulus Nephritis Mediated by E. coli Abundance Regulation. J CLUST SCI 2021. [DOI: 10.1007/s10876-021-02014-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Sundararaj K, Rodgers J, Angel P, Wolf B, Nowling TK. The role of neuraminidase in TLR4-MAPK signalling and the release of cytokines by lupus serum-stimulated mesangial cells. Immunology 2021; 162:418-433. [PMID: 33314123 DOI: 10.1111/imm.13294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
Previously, we demonstrated neuraminidase (NEU) activity or NEU1 expression, specifically, is increased in the kidneys of lupus mice and urine of human patients with nephritis. Additionally, NEU activity mediates IL-6 secretion from lupus-prone MRL/lpr primary mouse mesangial cells (MCs) in response to an IgG mimic. IL-6 mediates glomerular inflammation and promotes tissue damage in patients and mouse strains with lupus nephritis. This study further elucidates the mechanisms by which NEU activity and NEU1 specifically mediates the release of IL-6 and other cytokines from lupus-prone MCs. We demonstrate significantly increased release of multiple cytokines and NEU activity in MRL/lpr MCs in response to serum from MRL/lpr mice (lupus serum). Inhibiting NEU activity significantly reduced secretion of three of those cytokines: IL-6, GM-CSF and MIP1α. Message levels of Il-6 and Gm-csf were also increased in response to lupus serum and reduced when NEU activity was inhibited. Neutralizing antibodies to cell-surface receptors and MAPK inhibitors in lupus serum- or LPS-stimulated MCs indicate TLR4 and p38 or ERK MAP kinase signalling play key roles in the NEU-mediated secretion of IL-6. Significantly reduced IL-6 release was observed in C57BL/6 (B6) Neu1+/+ primary MCs compared with wild-type (Neu1+/+) B6 MCs in response to lupus serum. Additional results show inhibiting NEU activity significantly increases sialic acid-containing N-glycan levels. Together, our novel observations support a role for NEU activity, and specifically NEU1, in mediating release of IL-6 from lupus-prone MCs in response to lupus serum through a TLR4-p38/ERK MAPK signalling pathway that likely includes desialylation of glycoproteins.
Collapse
Affiliation(s)
- Kamala Sundararaj
- Department of Medicine, Division of Rheumatology, Medical University of South Carolina, Charleston, SC, USA
| | - Jessalyn Rodgers
- Department of Medicine, Division of Rheumatology, Medical University of South Carolina, Charleston, SC, USA
| | - Peggi Angel
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Bethany Wolf
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Tamara K Nowling
- Department of Medicine, Division of Rheumatology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
30
|
Sung SSJ, Fu SM. A novel immunofluorescence detection method for renal cell-type specific in situ cytokine production by confocal microscopy. MethodsX 2020; 7:100935. [PMID: 32577408 PMCID: PMC7303990 DOI: 10.1016/j.mex.2020.100935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 05/20/2020] [Indexed: 11/25/2022] Open
Abstract
The detection of cytokines production in tissues is subjected to significant limitations: (1) Cytokine protein production frequently does not correlate with mRNA levels. (2) Cytokines are secreted rapidly and dissipate from the cellular source, thus making detection difficult. (3) The synthetic rate of many cytokines are low. (4) Tissue fixation ablates antigenic sites and diminishes detection signals. The identification of the cellular sources of cytokines poses an additional challenge because of the lack of suitable and readily available cellular markers. In our renal cytokine production studies in lupus nephritis, we have established methods to resolve problems associated with the identification of cellular sources of pertinent cytokines in the glomerulus and interstitium. Four-color confocal microscopy was used to colocalize cell-type specific markers with cytokines. The cytokine signal was amplified by the incubation of tissue slices in medium containing pan-specific stimulants plus secretion blockers. Tissue fixation was optimized to provide sharp crisp signals. Commercially available Ab suitable for fluorochrome labeling were used to establish cell-specific markers in the tubules and glomeruli. This combination of optimizations allowed us to define the cellular sources of important glomerular cytokines including TNF-α, IL-6, and IL-1β which appear to form a cytokine circuit in glomerulonephritis pathogenesis. ● Tissue stimulation and secretion blocking for cytokine detection ● Fixation optimization and Ab source identification for direct staining ● Colocalization of cytokines and renal cell-type specific markers.
Collapse
Affiliation(s)
- Sun-Sang J Sung
- Center for Immunity, Inflammation, and Regenerative Medicine
| | - Shu Man Fu
- Center for Immunity, Inflammation, and Regenerative Medicine.,Division of Rheumatology, Department of Medicine, Charlottesville, VA 22908
| |
Collapse
|
31
|
Protecting the kidney in systemic lupus erythematosus: from diagnosis to therapy. Nat Rev Rheumatol 2020; 16:255-267. [PMID: 32203285 DOI: 10.1038/s41584-020-0401-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2020] [Indexed: 12/20/2022]
Abstract
Lupus nephritis (LN) is a common manifestation of systemic lupus erythematosus that can lead to irreversible renal impairment. Although the prognosis of LN has improved substantially over the past 50 years, outcomes have plateaued in the USA in the past 20 years as immunosuppressive therapies have failed to reverse disease in more than half of treated patients. This failure might reflect disease complexity and heterogeneity, as well as social and economic barriers to health-care access that can delay intervention until after damage has already occurred. LN progression is still poorly understood and involves multiple cell types and both immune and non-immune mechanisms. Single-cell analysis of intrinsic renal cells and infiltrating cells from patients with LN is a new approach that will help to define the pathways of renal injury at a cellular level. Although many new immune-modulating therapies are being tested in the clinic, the development of therapies to improve regeneration of the injured kidney and to prevent fibrosis requires a better understanding of the mechanisms of LN progression. This mechanistic understanding, together with the development of clinical measures to evaluate risk and detect early disease and better access to expert health-care providers, should improve outcomes for patients with LN.
Collapse
|