1
|
Han Z, Wang X, Liu J, Wang R, Zhao W, Liao H. Safflower injection against obesity-induced mice podocyte injury by improving insulin resistance through increasing renal INSR and eNOS expression. Ren Fail 2025; 47:2482888. [PMID: 40259553 PMCID: PMC12016271 DOI: 10.1080/0886022x.2025.2482888] [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: 07/26/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Podocyte injury is a common pathologic mechanism in obesity-related glomerulopathy (ORG). Safflower injection (SFI), scientifically extracted and refined from safflower, is used to treat diabetic kidney disease according to clinical guideline. Our previous study confirmed that the main active compounds of SFI ameliorated high glucose-induced podocyte injury. It is uncertain whether SFI has an effect on ORG-related podocyte injury. OBJECTIVES This study aimed to explore the pharmacological effects and related mechanisms of SFI on podocyte injury of ORG mice. METHODS First, by combining ultra-high performance liquid chromatography tandem mass spectrometry analysis with online databases, the pathway enrichment, target-pathway analysis, and human protein-protein interaction network were conducted to discover the possible crucial mechanism of SFI against ORG. Then, ORG mice model was established by high-fat diet and biochemical assays, histopathology and western blot were used to explore the effects of SFI on obesity and podocyte injury. Finally, system pharmacology-based findings were evaluated in ORG mice. RESULTS The results of system pharmacology suggested that SFI could alleviate ORG through insulin resistance (IR)-related pathway by regulating insulin receptor (INSR) and endothelial nitric oxide synthase (eNOS) expressions. The in vivo experiment confirmed that SFI ameliorated obesity, lipid metabolism-related indicators, podocyte injury of ORG mice. The mechanism relationships among IR, INSR, and eNOS were further verified in ORG mice. CONCLUSIONS Our findings imply that by up-regulating the expression of renal INSR and eNOS, thereby inhibiting IR, SFI may be a promising candidate for the treatment of ORG.
Collapse
Affiliation(s)
- Zhaodi Han
- Drug Clinical Trial Institution of Shanxi Provincial People’s Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xinyu Wang
- Drug Clinical Trial Institution of Shanxi Provincial People’s Hospital Affiliated to Shanxi Medical University, Taiyuan, China
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Jing Liu
- Drug Clinical Trial Institution of Shanxi Provincial People’s Hospital Affiliated to Shanxi Medical University, Taiyuan, China
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Rui Wang
- Drug Clinical Trial Institution of Shanxi Provincial People’s Hospital Affiliated to Shanxi Medical University, Taiyuan, China
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Wenyan Zhao
- Drug Clinical Trial Institution of Shanxi Provincial People’s Hospital Affiliated to Shanxi Medical University, Taiyuan, China
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Hui Liao
- Drug Clinical Trial Institution of Shanxi Provincial People’s Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
2
|
Harvey DJ. Analysis of carbohydrates and glycoconjugates by matrix-assisted laser desorption/ionization mass spectrometry: An update for 2021-2022. MASS SPECTROMETRY REVIEWS 2025; 44:213-453. [PMID: 38925550 PMCID: PMC11976392 DOI: 10.1002/mas.21873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 06/28/2024]
Abstract
The use of matrix-assisted laser desorption/ionization (MALDI) mass spectrometry for the analysis of carbohydrates and glycoconjugates is a well-established technique and this review is the 12th update of the original article published in 1999 and brings coverage of the literature to the end of 2022. As with previous review, this review also includes a few papers that describe methods appropriate to analysis by MALDI, such as sample preparation, even though the ionization method is not MALDI. The review follows the same format as previous reviews. It is divided into three sections: (1) general aspects such as theory of the MALDI process, matrices, derivatization, MALDI imaging, fragmentation, quantification and the use of computer software for structural identification. (2) Applications to various structural types such as oligo- and polysaccharides, glycoproteins, glycolipids, glycosides and biopharmaceuticals, and (3) other general areas such as medicine, industrial processes, natural products and glycan synthesis where MALDI is extensively used. Much of the material relating to applications is presented in tabular form. MALDI is still an ideal technique for carbohydrate analysis, particularly in its ability to produce single ions from each analyte and advancements in the technique and range of applications show little sign of diminishing.
Collapse
|
3
|
Shen L, Lin JM, Lin J, Wu W. Glycosylation in Dermatology: Unveiling the Sugar Coating of Skin Disease. Exp Dermatol 2025; 34:e70098. [PMID: 40207455 DOI: 10.1111/exd.70098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Glycosylation is a common and complex post-translational modification (PTM) of proteins, involving the attachment of glycans under the regulation of various enzymes such as glycosyltransferases. Glycosylation facilitates the correct folding of peptide chains, modifies protein conformation and activity, enhances protein stability and influences inter-protein interactions. N-glycosylation and O-glycosylation are two prevalent forms, encompassing a wide range of modifications, including sialylation, fucosylation and galactosylation. In skin tumours, abnormal glycosylation promotes tumour cell proliferation, migration, invasion and metastasis, enhances anti-tumour immunity, and potentially affects immune checkpoint therapy. In inflammatory and autoimmune skin diseases, abnormal glycosylation in T and B lymphocyte subpopulations regulates antigen recognition, signal transduction, inflammatory factor secretion and immunoglobulin function, disrupting immune system homeostasis and impacting biologic therapy efficacy. Glycosylation correlates with the severity and activity of skin diseases, serving as a potential biomarker for diagnosis, condition assessment and prognosis determination. This review provides an overview of the role of protein glycosylation in melanoma, basal cell carcinoma, squamous cell carcinoma, psoriasis, systemic lupus erythematosus, dermatomyositis and skin aging. It analyses the biosynthetic process of glycosylation, elucidates functional changes in glycoproteins and their metabolism, and offers a theoretical basis for developing new targeted therapies.
Collapse
Affiliation(s)
- Linxia Shen
- Department of Dermatology, Huashan Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Jui-Ming Lin
- Department of Dermatology, Huashan Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Jinran Lin
- Department of Dermatology, Huashan Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Wenyu Wu
- Department of Dermatology, Huashan Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
- Department of Dermatology, Jing'an District Central Hospital, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Kasinath V, Tsokos GC. PI3Kα in the pathogenesis and treatment of lupus nephritis. Kidney Int 2025; 107:215-217. [PMID: 39557395 DOI: 10.1016/j.kint.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/04/2024] [Indexed: 11/20/2024]
Affiliation(s)
- Vivek Kasinath
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| | - George C Tsokos
- Division of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
| |
Collapse
|
5
|
Bhargava R, Li H, Maeda K, Tsokos MG, Tsokos GC. Calcium calmodulin kinase IV deficiency in podocytes prevents the development of lupus nephritis. Clin Immunol 2025; 271:110427. [PMID: 39793862 DOI: 10.1016/j.clim.2025.110427] [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: 11/08/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by widespread organ involvement including the kidney. Calcium/calmodulin-dependent protein kinase IV (CaMK4) has been shown to conrol immune cell nad podocyte function. To address the effect of genetic podocyte-specific CaMK4 deficiency on systemic autoimmunity and kidney pathology in lupus-prone mice we generated B6.lpr.Camk4flox..podocincre mice. Although podocyte-specific CaMK4 deletion in the lupus-prone Br.lpr mice did not affect systemic autoimmune response parameters, it led to significant improvement of kidney pathology and clinical outcomes. Specifically, B6.lpr.Camk4flox..podocincre mice exhibited reduced glomerular pathology, characterized by less mesangial cell proliferation and diminished immune complex deposition, accompanied by decreased levels of albuminuria and improved creatinine levels. CaMK4 deficiency in podocytes averted the deposition of immune complexes in the kidney. Interestingly, we found increased deposition of immune complexes in the liver. We conclude that CaMK4 expression in podocytes is central to the development of LN and its targeted deletion in podocytes prevents its development without affecting systemic autoimmunity while immune complexes appear to be re-directed from the kidney to the liver.
Collapse
Affiliation(s)
- Rhea Bhargava
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America; Division of Nephrology, Department of Medicine, Tulane University, New Orleans, LA, United States of America.
| | - Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Kayaho Maeda
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| |
Collapse
|
6
|
Yu MC, Huang XD, Kuo CW, Zhang KF, Liang PC, Jeng US, Huang PY, Tam FWK, Lee YC. Developing a Label-Free Infrared Spectroscopic Analysis with Chemometrics and Computational Enhancement for Assessing Lupus Nephritis Activity. BIOSENSORS 2025; 15:39. [PMID: 39852090 PMCID: PMC11763532 DOI: 10.3390/bios15010039] [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: 11/11/2024] [Revised: 12/22/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025]
Abstract
Patterns of disease and therapeutic responses vary widely among patients with autoimmune glomerulonephritis. This study introduces groundbreaking personalized infrared (IR)-based diagnostics for real-time monitoring of disease status and treatment responses in lupus nephritis (LN). We have established a relative absorption difference (RAD) equation to assess characteristic spectral indices based on the temporal peak heights (PHs) of two characteristic serum absorption bands: ν1 as the target signal and ν2 as the PH reference for the ν1 absorption band, measured at each dehydration time (t) during dehydration. The RAD gap (Ψ), defined as the difference in the RAD values between the initial and final stages of serum dehydration, enables the measurement of serum levels of IgG glycosylation (ν1 (1030 cm-1), ν2 (1171 cm-1)), serum lactate (ν1 (1021 cm-1), ν2 (1171 cm-1)), serum hydrophobicity (ν1 (2930 cm-1), ν2 (2960 cm-1)), serum hydrophilicity (ν1 (1550 cm-1), ν2 (1650 cm-1)), and albumin (ν1 (1400 cm-1), ν2 (1450 cm-1)). Furthermore, this IR-based assay incorporates an innovative algorithm and our proprietary iPath software (ver. 1.0), which calculates the prognosis prediction function (PPF, Φ) from the RAD gaps of five spectral markers and correlates these with conventional clinical renal biomarkers. We propose that this algorithm-assisted, IR-based approach can augment the patient-centric care of LN patients, particularly by focusing on changes in serum IgG glycosylation.
Collapse
Affiliation(s)
- Mei-Ching Yu
- Division of Pediatric Nephrology, Department of Pediatrics, Lin-Kou Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City 243303, Taiwan
| | - Xiang-Di Huang
- Division of Pediatric Nephrology, Department of Pediatrics, Lin-Kou Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan;
| | - Chin-Wei Kuo
- National Synchrotron Radiation Research Center, Hsinchu 300092, Taiwan; (C.-W.K.); (K.-F.Z.); (P.-C.L.); (U.-S.J.); (P.-Y.H.)
| | - Kai-Fu Zhang
- National Synchrotron Radiation Research Center, Hsinchu 300092, Taiwan; (C.-W.K.); (K.-F.Z.); (P.-C.L.); (U.-S.J.); (P.-Y.H.)
| | - Ping-Chung Liang
- National Synchrotron Radiation Research Center, Hsinchu 300092, Taiwan; (C.-W.K.); (K.-F.Z.); (P.-C.L.); (U.-S.J.); (P.-Y.H.)
| | - U-Ser Jeng
- National Synchrotron Radiation Research Center, Hsinchu 300092, Taiwan; (C.-W.K.); (K.-F.Z.); (P.-C.L.); (U.-S.J.); (P.-Y.H.)
| | - Pei-Yu Huang
- National Synchrotron Radiation Research Center, Hsinchu 300092, Taiwan; (C.-W.K.); (K.-F.Z.); (P.-C.L.); (U.-S.J.); (P.-Y.H.)
| | - Frederick Wai Keung Tam
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK;
| | - Yao-Chang Lee
- National Synchrotron Radiation Research Center, Hsinchu 300092, Taiwan; (C.-W.K.); (K.-F.Z.); (P.-C.L.); (U.-S.J.); (P.-Y.H.)
- Department of Optics and Photonics, National Central University, Chung-Li 320317, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu 30044, Taiwan
| |
Collapse
|
7
|
Liu Q, Liu Y, Feng H, Zhao L, Wan T. Exploring genetic associations in systemic lupus erythematosus through Mendelian randomization: implications for novel biomarkers and therapeutic targets. Clin Rheumatol 2025; 44:193-205. [PMID: 39126578 DOI: 10.1007/s10067-024-07094-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/01/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by a significant health burden. There is an essential need for novel biomarkers and therapeutic targets to improve diagnosis and management. Mendelian randomization (MR) was applied to explore causal links between SLE and various biomarkers like immune cells, metabolites, and inflammatory cytokines using multiple databases. Initially, biomarkers significantly associated with SLE were identified. Bidirectional MR helped clarify these relationships, and a two-step mediation MR examined their effects on SLE risk. Intersection analysis was used to identify biomarkers with consistent effects across datasets. Four biomarkers were identified as having significant associations with SLE risk: 1-palmitoyl-2-arachidonoyl-GPI levels [odds ratio (OR), 1.379; 95% confidence interval (CI), 1.180 to 1.613; FDR, 0.046], IL-17A levels (OR, 2.197; 95% CI, 1.412 to 3.418; FDR, 0.044), N-acetyl-aspartyl-glutamate (NAAG) levels (OR, 0.882; 95% CI, 0.831 to 0.936; FDR, 0.030), and ribitol levels (OR, 0.743; 95% CI, 0.644 to 0.857; FDR, 0.012). Bidirectional MR showed an inverse effect of NAAG on IL-17A levels (OR, 0.978; 95% CI, 0.962 to 0.994; p = 0.006). Mediation analysis indicated that NAAG influenced SLE risk both directly (beta = - 0.108) and indirectly through IL-17A (beta = - 0.018), highlighting the potential mediating role of IL-17A. After expanding the significance criteria to p < 0.05, intersection analysis across multiple datasets revealed 29 biomarkers with consistent beta directions, including 19 potential risk factors (beta > 0) and 10 protective factors (beta < 0) for SLE. This research has revealed significant genetic associations with SLE and demonstrated that IL-17A mediates the relationship between NAAG levels and SLE risk, highlighting potential new targets for personalized therapeutic interventions. Key Points • This study employs MR to identify significant genetic associations between various biomarkers and SLE, providing novel insights into potential biomarkers and therapeutic targets. • Four key biomarkers were identified as significantly associated with SLE risk: 1-palmitoyl-2-arachidonoyl-GPI, IL-17A, N-acetyl-aspartyl-glutamate (NAAG), and ribitol. • The findings suggest that NAAG levels have a protective effect against SLE, partly mediated through IL-17A, indicating a complex interplay between these biomarkers in the pathogenesis of SLE. • Intersectional analysis across multiple datasets revealed 29 biomarkers with consistent effects on SLE risk, highlighting new directions for future research and potential personalized therapeutic strategies.
Collapse
Affiliation(s)
- Qi Liu
- Department of Hepatobiliary Surgery, 8th Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
- Medical School of Chinese People's Liberation Army, Beijing, China
| | - Yuyang Liu
- Department of Neurosurgery, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Hui Feng
- Department of Zhantansi Outpatient, Jingzhong Medical District of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lin Zhao
- Department of Endocrinology, 8th Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Tao Wan
- Department of Hepatobiliary Surgery, 8th Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China.
| |
Collapse
|
8
|
Maeda K, Abdi R, Tsokos GC. The Role of Podocytes in Lupus Pathology. Curr Rheumatol Rep 2024; 27:10. [PMID: 39731699 DOI: 10.1007/s11926-024-01175-4] [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] [Accepted: 12/17/2024] [Indexed: 12/30/2024]
Abstract
PURPOSE OF REVIEW Kidney injury due to lupus nephritis (LN) is a severe and sometimes life-threatening sequela of systemic lupus erythematosus. Autoimmune injury to podocytes has been increasingly demonstrated to be a key driver of LN-related kidney injury because these cells play key roles in glomerular filtration barrier homeostasis. Irreparable podocyte injury impairs these processes and can lead to proteinuria, which is an indicator of poor prognosis in LN. This review highlights recent advances in our understanding of the involvement of podocytes in the pathogenesis of LN and discusses new podocyte-targeted therapeutic strategies. RECENT FINDINGS Podocytes play a key role in glomerular filtration barrier homeostasis, both by helping to secrete and organize the glomerular basement membrane and by the formation of a glomerular slit diaphragm between adjacent cells. Recent studies revealed the involvement of abnormal calcium signaling, dysregulation of actin-related proteins, and mitotic catastrophe in LN progression. In addition, podocytes express many molecules related to the innate and adaptive immune responses. IgG from patients with LN induces direct injury of podocytes, inflammasome, and interactions with immune cells which have been shown to promote the development of LN. Our understanding of the role of podocytes in the pathogenesis of LN has been improved. Recent studies have shed light on potential therapeutic strategies targeting podocytes to control kidney injury.
Collapse
Affiliation(s)
- Kayaho Maeda
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Reza Abdi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS-937, Boston, MA, 02215, USA.
| |
Collapse
|
9
|
Beyze A, Larroque C, Le Quintrec M. The role of antibody glycosylation in autoimmune and alloimmune kidney diseases. Nat Rev Nephrol 2024; 20:672-689. [PMID: 38961307 DOI: 10.1038/s41581-024-00850-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 07/05/2024]
Abstract
Immunoglobulin glycosylation is a pivotal mechanism that drives the diversification of antibody functions. The composition of the IgG glycome is influenced by environmental factors, genetic traits and inflammatory contexts. Differential IgG glycosylation has been shown to intricately modulate IgG effector functions and has a role in the initiation and progression of various diseases. Analysis of IgG glycosylation is therefore a promising tool for predicting disease severity. Several autoimmune and alloimmune disorders, including critical and potentially life-threatening conditions such as systemic lupus erythematosus, anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis and antibody-mediated kidney graft rejection, are driven by immunoglobulin. In certain IgG-driven kidney diseases, including primary membranous nephropathy, IgA nephropathy and lupus nephritis, particular glycome characteristics can enhance in situ complement activation and the recruitment of innate immune cells, resulting in more severe kidney damage. Hypofucosylation, hypogalactosylation and hyposialylation are the most common IgG glycosylation traits identified in these diseases. Modulating IgG glycosylation could therefore be a promising therapeutic strategy for regulating the immune mechanisms that underlie IgG-driven kidney diseases and potentially reduce the burden of immunosuppressive drugs in affected patients.
Collapse
Affiliation(s)
- Anaïs Beyze
- Institute of Regenerative Medicine and Biotherapy, IRMB U1183, Montpellier, France.
- Department of Nephrology, Dialysis and Transplantation, Montpellier University Hospital, Montpellier, France.
- University of Montpellier, Montpellier, France.
| | - Christian Larroque
- Institute of Regenerative Medicine and Biotherapy, IRMB U1183, Montpellier, France
- Department of Nephrology, Dialysis and Transplantation, Montpellier University Hospital, Montpellier, France
- University of Montpellier, Montpellier, France
| | - Moglie Le Quintrec
- Institute of Regenerative Medicine and Biotherapy, IRMB U1183, Montpellier, France.
- Department of Nephrology, Dialysis and Transplantation, Montpellier University Hospital, Montpellier, France.
- University of Montpellier, Montpellier, France.
| |
Collapse
|
10
|
Fu R, Wang W, Huo Y, Li L, Chen R, Lin Z, Tao Y, Peng X, Huang W, Guo C. The mechanosensitive ion channel Piezo1 contributes to podocyte cytoskeleton remodeling and development of proteinuria in lupus nephritis. Kidney Int 2024; 106:625-639. [PMID: 39084260 DOI: 10.1016/j.kint.2024.06.025] [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: 09/12/2023] [Revised: 06/10/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024]
Abstract
Piezo1 functions as a special transducer of mechanostress into electrochemical signals and is implicated in the pathogenesis of various diseases across different disciplines. However, whether Piezo1 contributes to the pathogenesis of lupus nephritis (LN) remains elusive. To study this, we applied an agonist and antagonist of Piezo1 to treat lupus-prone MRL/lpr mice. Additionally, a podocyte-specific Piezo1 knockout mouse model was also generated to substantiate the role of Piezo1 in podocyte injury induced by pristane, a murine model of LN. A marked upregulation of Piezo1 was found in podocytes in both human and murine LN. The Piezo1 antagonist, GsMTx4, significantly alleviated glomerulonephritis and tubulointerstitial damage, improved kidney function, decreased proteinuria, and mitigated podocyte foot process effacement in MRL/lpr mice. Moreover, podocyte-specific Piezo1 deletion showed protective effects on the progression of proteinuria and podocyte foot process effacement in the murine LN model. Mechanistically, Piezo1 expression was upregulated by inflammatory cytokines (IL-6, TNF-α and IFN-γ), soluble urokinase Plasminogen Activator Receptor and its own activation. Activation of Piezo1 elicited calcium influx, which subsequently enhanced Rac1 activity and increased active paxillin, thereby promoting cytoskeleton remodeling and decreasing podocyte motility. Thus, our work demonstrated that Piezo1 contributed to podocyte injury and proteinuria progression in LN. Hence, targeted therapy aimed at decreasing or inhibiting Piezo1 could represent a novel strategy to treat LN.
Collapse
Affiliation(s)
- Rong Fu
- Department of Rheumatology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wenqian Wang
- Department of Hematology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongbao Huo
- Department of Rheumatology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Liu Li
- Department of Rheumatology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ruilin Chen
- Department of Rheumatology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zeying Lin
- Department of Rheumatology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yi Tao
- Department of Rheumatology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xuan Peng
- Department of Nephrology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Wenhui Huang
- Department of Rheumatology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Chaohuan Guo
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
11
|
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
|
12
|
Han Z, Gong L, Xue Y, Wang R, Liu J, Wang X, Zhao W, Liao H, Li R. Effects of Inonotus obliquus on ameliorating podocyte injury in ORG mice through TNF pathway and prediction of active compounds. Front Pharmacol 2024; 15:1426917. [PMID: 39234117 PMCID: PMC11371614 DOI: 10.3389/fphar.2024.1426917] [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: 05/02/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024] Open
Abstract
Background Podocyte injury is a common pathologic mechanism in diabetic kidney disease (DKD) and obesity-related glomerulopathy (ORG). Our previous study confirmed that Inonotus obliquus (IO) improved podocyte injury on DKD rats. The current study explored the pharmacological effects, related mechanisms and possible active components of IO on ORG mice. Methods Firstly, by combining ultra-high performance liquid chromatography tandem mass spectrometry analysis (UPLC-Q-TOF-MS) with network pharmacology to construct the human protein-protein interaction mechanism and enrich the pathway, which led to discover the crucial mechanism of IO against ORG. Then, ORG mice were established by high-fat diet and biochemical assays, histopathology, and Western blot were used to explore the effects of IO on obesity and podocyte injury. Finally, network pharmacology-based findings were confirmed by immunohistochemistry. The compositions of IO absorbed in mice plasma were analyzed by UPLC-Q-TOF-MS and molecular docking was used to predict the possible active compounds. Results The network pharmacology result suggested that IO alleviated the inflammatory response of ORG by modulating TNF signal. The 20-week in vivo experiment confirmed that IO improved glomerular hypertrophy, podocyte injury under electron microscopy, renal nephrin, synaptopodin, TNF-α and IL-6 expressions with Western blotting and immunohistochemical staining. Other indicators of ORG such as body weight, kidney weight, serum total cholesterol, liver triglyceride also improved by IO intervention. The components analysis showed that triterpenoids, including inoterpene F and trametenolic acid, might be the pharmacodynamic basis. Conclusion The research based on UPLC-Q-TOF-MS analysis, network pharmacology and in vivo experiment suggested that the amelioration of IO on podocyte injury in ORG mice via its modulation on TNF signal. Triterpenoids were predicated as acting components.
Collapse
Affiliation(s)
- Zhaodi Han
- Drug Clinical Trial Institution, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Le Gong
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Yani Xue
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Rui Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Jing Liu
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Xinyu Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Wenyan Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Hui Liao
- Drug Clinical Trial Institution, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Rongshan Li
- Department of Nephrology, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
13
|
Tsokos GC. The immunology of systemic lupus erythematosus. Nat Immunol 2024; 25:1332-1343. [PMID: 39009839 DOI: 10.1038/s41590-024-01898-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024]
Abstract
Understanding the pathogenesis and clinical manifestations of systemic lupus erythematosus (SLE) has been a great challenge. Reductionist approaches to understand the nature of the disease have identified many pathogenetic contributors that parallel clinical heterogeneity. This Review outlines the immunological control of SLE and looks to experimental tools and approaches that are improving our understanding of the complex contribution of interacting genetics, environment, sex and immunoregulatory factors and their interface with processes inherent to tissue parenchymal cells. Efforts to advance precision medicine in the care of patients with SLE along with treatment strategies to correct the immune system hold hope and are also examined.
Collapse
Affiliation(s)
- George C Tsokos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Xu H, Yong L, Gao X, Chen Y, Wang Y, Wang F, Hou X. CaMK4: Structure, physiological functions, and therapeutic potential. Biochem Pharmacol 2024; 224:116204. [PMID: 38615920 DOI: 10.1016/j.bcp.2024.116204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/22/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Calcium/calmodulin-dependent protein kinase IV (CaMK4) is a versatile serine/threonine kinase involved in various cellular functions. It regulates T-cell differentiation, podocyte function, tumor cell proliferation/apoptosis, β cell mass, and insulin sensitivity. However, the underlying molecular mechanisms are complex and remain incompletely understood. The aims of this review are to highlight the latest advances in the regulatory mechanisms of CaMK4 underlying T-cell imbalance and parenchymal cell mass in multiple diseases. The structural motifs and activation of CaMK4, as well as the potential role of CaMK4 as a novel therapeutic target are also discussed.
Collapse
Affiliation(s)
- Hao Xu
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Liang Yong
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, PR China
| | - Xianxian Gao
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Yandong Chen
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Yixuan Wang
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Fuyan Wang
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China; Clinical Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, Shandong 266033, PR China
| | - Xin Hou
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, PR China.
| |
Collapse
|
15
|
Rosetti F, Madera-Salcedo IK, Crispín JC. Relevance of acquired T cell molecular defects in the immunopathogenesis of SLE. Clin Immunol 2024; 263:110225. [PMID: 38642784 DOI: 10.1016/j.clim.2024.110225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/07/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
Systemic lupus erythematosus (SLE) and other autoimmune diseases are thought to develop in genetically predisposed individuals when triggered by environmental factors. This paradigm does not fully explain disease development, as it fails to consider the delay between birth and disease expression. In this review, we discuss observations described in T cells from patients with SLE that are not related to hereditary factors and have therefore been considered secondary to the disease process itself. Here, we contextualize some of those observations and argue that they may represent a pathogenic layer between genetic factors and disease development. Acquired changes in T cell phenotype and function in the setting of SLE may affect the immune system, creating a predisposition towards a more inflammatory and pathogenic system that amplifies autoimmunity and facilitates disease development.
Collapse
Affiliation(s)
- Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Mexico City 14080, Mexico
| | - Iris K Madera-Salcedo
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Mexico City 14080, Mexico
| | - José C Crispín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Mexico City 14080, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Eugenio Garza Sada 2501, Monterrey, N.L. 64849, Mexico.
| |
Collapse
|
16
|
Qi YY. Response to: Correspondence on 'SGLT2 inhibitors alleviated podocyte damage in lupus nephritis by decreasing inflammation and enhancing autophagy' by Zhao et al. Ann Rheum Dis 2024; 83:e14. [PMID: 38171601 DOI: 10.1136/ard-2023-225255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Affiliation(s)
- Yuan-Yuan Qi
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
- Zhengzhou University, Zhengzhou, Henan, People's Republic of China
- Laboratory of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
17
|
Ichinose K. The role of podocytes in lupus nephritis: Insights and implications. Clin Immunol 2024; 262:110180. [PMID: 38462157 DOI: 10.1016/j.clim.2024.110180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/09/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024]
Abstract
Lupus nephritis (LN) is a severe complication of systemic lupus erythematosus, with high mortality rates despite medical advancements. The complexity of its pathogenesis, including the pivotal role of podocytes - kidney-localized cells - remains a challenge, lacking effective treatments and biomarkers. Recent studies highlight the significant contribution of these cells to LN's development, particularly through their immune-related functions and interaction with other kidney cells. This new understanding opens possibilities for targeted therapies aimed at these cellular mechanisms. This review aims to summarize these recent developments, shedding light on the intricate involvement of podocytes in LN and potential avenues for innovative treatments.
Collapse
Affiliation(s)
- Kunihiro Ichinose
- Department of Rheumatology, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo 693-8501, Japan; Integrated Kidney Research and Advance, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo 693-8501, Japan.
| |
Collapse
|
18
|
Tsokos GC, Boulougoura A, Kasinath V, Endo Y, Abdi R, Li H. The immunoregulatory roles of non-haematopoietic cells in the kidney. Nat Rev Nephrol 2024; 20:206-217. [PMID: 37985868 PMCID: PMC11005998 DOI: 10.1038/s41581-023-00786-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2023] [Indexed: 11/22/2023]
Abstract
The deposition of immune complexes, activation of complement and infiltration of the kidney by cells of the adaptive and innate immune systems have long been considered responsible for the induction of kidney damage in autoimmune, alloimmune and other inflammatory kidney diseases. However, emerging findings have highlighted the contribution of resident immune cells and of immune molecules expressed by kidney-resident parenchymal cells to disease processes. Several types of kidney parenchymal cells seem to express a variety of immune molecules with a distinct topographic distribution, which may reflect the exposure of these cells to different pathogenic threats or microenvironments. A growing body of literature suggests that these cells can stimulate the infiltration of immune cells that provide protection against infections or contribute to inflammation - a process that is also regulated by draining kidney lymph nodes. Moreover, components of the immune system, such as autoantibodies, cytokines and immune cells, can influence the metabolic profile of kidney parenchymal cells in the kidney, highlighting the importance of crosstalk in pathogenic processes. The development of targeted nanomedicine approaches that modulate the immune response or control inflammation and damage directly within the kidney has the potential to eliminate the need for systemically acting drugs.
Collapse
Affiliation(s)
- George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | | | - Vivek Kasinath
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yushiro Endo
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Reza Abdi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
19
|
Zhang Z, Hu H, Luo Q, Yang K, Zou Z, Shi M, Liang W. Dihydroxyacetone phosphate accumulation leads to podocyte pyroptosis in diabetic kidney disease. J Cell Mol Med 2024; 28:e18073. [PMID: 38063077 PMCID: PMC10844688 DOI: 10.1111/jcmm.18073] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/10/2023] [Accepted: 11/25/2023] [Indexed: 02/08/2024] Open
Abstract
Diabetic kidney disease (DKD) can lead to accumulation of glucose upstream metabolites due to dysfunctional glycolysis. But the effects of accumulated glycolysis metabolites on podocytes in DKD remain unknown. The present study examined the effect of dihydroxyacetone phosphate (DHAP) on high glucose induced podocyte pyroptosis. By metabolomics, levels of DHAP, GAP, glucose-6-phosphate and fructose 1, 6-bisphosphate were significantly increased in glomeruli of db/db mice. Furthermore, the expression of LDHA and PKM2 were decreased. mRNA sequencing showed upregulation of pyroptosis-related genes (Nlrp3, Casp1, etc.). Targeted metabolomics demonstrated higher level of DHAP in HG-treated podocytes. In vitro, ALDOB expression in HG-treated podocytes was significantly increased. siALDOB-transfected podocytes showed less DHAP level, mTORC1 activation, reactive oxygen species (ROS) production, and pyroptosis, while overexpression of ALDOB had opposite effects. Furthermore, GAP had no effect on mTORC1 activation, and mTORC1 inhibitor rapamycin alleviated ROS production and pyroptosis in HG-stimulated podocytes. Our findings demonstrate that DHAP represents a critical metabolic product for pyroptosis in HG-stimulated podocytes through regulation of mTORC1 pathway. In addition, the results provide evidence that podocyte injury in DKD may be treated by reducing DHAP.
Collapse
Affiliation(s)
- Zongwei Zhang
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina
- Nephrology and Urology Research Institute of Wuhan UniversityWuhanChina
| | - Hongtu Hu
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina
- Nephrology and Urology Research Institute of Wuhan UniversityWuhanChina
| | - Qiang Luo
- Department of NephrologyThe Central Hospital of WuhanWuhanChina
| | - Keju Yang
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina
- Nephrology and Urology Research Institute of Wuhan UniversityWuhanChina
| | - Zhengping Zou
- Qianjiang Hospital Affiliated to Renmin Hospital of Wuhan UniversityQianjiangChina
- Qianjiang Clinical Medical CollegeHealth Science CenterYangtze UniversityQianjiangChina
| | - Ming Shi
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina
- Nephrology and Urology Research Institute of Wuhan UniversityWuhanChina
| | - Wei Liang
- Division of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina
- Nephrology and Urology Research Institute of Wuhan UniversityWuhanChina
| |
Collapse
|
20
|
Scherlinger M, Li H, Pan W, Li W, Karino K, Vichos T, Boulougoura A, Yoshida N, Tsokos MG, Tsokos GC. CaMK4 controls follicular helper T cell expansion and function during normal and autoimmune T-dependent B cell responses. Nat Commun 2024; 15:840. [PMID: 38287012 PMCID: PMC10825135 DOI: 10.1038/s41467-024-45080-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/11/2024] [Indexed: 01/31/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by dysregulated B cell compartment responsible for the production of autoantibodies. Here, we show that T cell-specific expression of calcium/calmodulin-dependent protein kinase IV (CaMK4) leads to T follicular helper (Tfh) cells expansion in models of T-dependent immunization and autoimmunity. Mechanistically, CaMK4 controls the Tfh-specific transcription factor B cell lymphoma 6 (Bcl6) at the transcriptional level through the cAMP responsive element modulator α (CREMα). In the absence of CaMK4 in T cells, germinal center formation and humoral immunity is impaired in immunized mice, resulting in reduced anti-dsDNA titres, as well as IgG and complement kidney deposition in the lupus-prone B6.lpr mouse. In human Tfh cells, CaMK4 inhibition reduced BCL6 expression and IL-21 secretion ex vivo, resulting in impaired plasmablast formation and IgG production. In patients with SLE, CAMK4 mRNA levels in Tfh cells correlated with those of BCL6. In conclusion, we identify CaMK4/CREMα as a driver of T cell-dependent B cell dysregulation in autoimmunity.
Collapse
Affiliation(s)
- Marc Scherlinger
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Rheumatology department, Strasbourg University Hospital of Hautepierre, Strasbourg, France.
- Laboratoire d'ImmunoRhumatologie Moléculaire, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1109, Strasbourg, France.
| | - Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Wenliang Pan
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Wei Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kohei Karino
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Theodoros Vichos
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Nobuya Yoshida
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
21
|
Guo Z, Guo Q, Li X, Gao X, Zhang L, Xu K. Urinary biomarkers associated with podocyte injury in lupus nephritis. Front Pharmacol 2024; 15:1324540. [PMID: 38313309 PMCID: PMC10834635 DOI: 10.3389/fphar.2024.1324540] [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: 11/30/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
The most prevalent and devastating form of organ damage in systemic lupus erythematosus (SLE) is lupus nephritis (LN). LN is characterized by glomerular injury, inflammation, cell proliferation, and necrosis, leading to podocyte injury and tubular epithelial cell damage. Assays for urine biomarkers have demonstrated significant promise in the early detection of LN, evaluation of disease activity, and tracking of reaction to therapy. This is because they are non-invasive, allow for frequent monitoring and easy self-collection, transport and storage. Podocyte injury is believed to be a essential factor in LN. The extent and type of podocyte injury could be connected to the severity of proteinuria, making podocyte-derived cellular debris and injury-related urinary proteins potential markers for the diagnosis and monitoring of LN. This article focuses on studies examining urinary biomarkers associated with podocyte injury in LN, offering fresh perspectives on the application of biomarkers in the early detection and management of LN.
Collapse
Affiliation(s)
| | | | | | | | | | - Ke Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
22
|
Alduraibi FK, Tsokos GC. Lupus Nephritis Biomarkers: A Critical Review. Int J Mol Sci 2024; 25:805. [PMID: 38255879 PMCID: PMC10815779 DOI: 10.3390/ijms25020805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Lupus nephritis (LN), a major complication in individuals diagnosed with systemic lupus erythematosus, substantially increases morbidity and mortality. Despite marked improvements in the survival of patients with severe LN over the past 50 years, complete clinical remission after immunosuppressive therapy is achieved in only half of the patients. Therefore, timely detection of LN is vital for initiating prompt therapeutic interventions and improving patient outcomes. Biomarkers have emerged as valuable tools for LN detection and monitoring; however, the complex role of these biomarkers in LN pathogenesis remains unclear. Renal biopsy remains the gold standard for the identification of the histological phenotypes of LN and guides disease management. However, the molecular pathophysiology of specific renal lesions remains poorly understood. In this review, we provide a critical, up-to-date overview of the latest developments in the field of LN biomarkers.
Collapse
Affiliation(s)
- Fatima K. Alduraibi
- Department of Medicine, Division of Clinical Immunology and Rheumatology, Beth Israel Deaconess Medical Center, Harvard Teaching Hospital, Boston, MA 02215, USA
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Medicine, Division of Clinical Immunology and Rheumatology, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - George C. Tsokos
- Department of Medicine, Division of Clinical Immunology and Rheumatology, Beth Israel Deaconess Medical Center, Harvard Teaching Hospital, Boston, MA 02215, USA
| |
Collapse
|
23
|
Zeng H, Li D, Dong J, Zhou X, Ou M, Xue W, Zhang R, Zou Y, Tang D, Yin L, Dai Y. Qualitative Proteome-wide Lysine Crotonylation Profiling Reveals Protein Modification Alteration in the Leukocyte Extravasation Pathway in Systemic Lupus Erythematosus. ACS OMEGA 2023; 8:44905-44919. [PMID: 38046296 PMCID: PMC10688171 DOI: 10.1021/acsomega.3c06293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a severe systemic autoimmune disease with multiple manifestations. Lysine crotonylation (Kcr) is a newly discovered posttranslational modification epigenetic pattern that may affect gene expression and is linked to diseases causally. METHODS We collected blood samples from 11 SLE individuals and 36 healthy subjects. Then, we used highly sensitive liquid chromatography-mass spectrometry technology to carry out proteomics and quantitative crotonylome analysis of SLE peripheral blood mononuclear cells in this investigation, which indicated the unique etiology of SLE. Finally, we verified the expression of critical protein in the leukocyte extravasation pathway by online database analysis and Western blot. RESULTS There were 618 differentially expressed proteins (DEPs), and 612 crotonylated lysine sites for 272 differentially modified proteins (DMPs) found. These DEPs and DMPs are primarily enriched in the leukocyte extravasation signaling pathway, such as MMP8, MMP9, and ITGAM. CONCLUSIONS This is the first study of crotonylated modification proteomics in SLE. The leukocyte extravasation signaling pathway had a considerable concentration of DEPs and DMPs, indicating that this pathway may be involved in the pathogenic development of SLE.
Collapse
Affiliation(s)
- Huiyi Zeng
- Institute
of Nephrology and Blood Purification, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
- The First
Affiliated Hospital, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, China
| | - Dandan Li
- Department
of Clinical Medical Research Center, The
Second Clinical Medical College of Jinan University, Shenzhen People’s
Hospital, Shenzhen, Guangdong 518020, China
- Experimental
Center, Shenzhen Pingle Orthopedic Hospital
(Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangdong 518118, China
- The First
Affiliated Hospital, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, China
| | - Jingjing Dong
- Institute
of Nephrology and Blood Purification, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
- Department
of Clinical Medical Research Center, The
Second Clinical Medical College of Jinan University, Shenzhen People’s
Hospital, Shenzhen, Guangdong 518020, China
| | - Xianqing Zhou
- Guangxi
Key Laboratory of Metabolic Diseases Research, No. 924 Hospital of PLA Joint Logistic Support
Force, Guilin, Guangxi 541002, China
| | - Minglin Ou
- Guangxi
Key Laboratory of Metabolic Diseases Research, No. 924 Hospital of PLA Joint Logistic Support
Force, Guilin, Guangxi 541002, China
| | - Wen Xue
- Guangxi
Key Laboratory of Metabolic Diseases Research, No. 924 Hospital of PLA Joint Logistic Support
Force, Guilin, Guangxi 541002, China
| | - Ruohan Zhang
- Guangxi
Key Laboratory of Metabolic Diseases Research, No. 924 Hospital of PLA Joint Logistic Support
Force, Guilin, Guangxi 541002, China
| | - Yaoshuang Zou
- Guangxi
Key Laboratory of Metabolic Diseases Research, No. 924 Hospital of PLA Joint Logistic Support
Force, Guilin, Guangxi 541002, China
| | - Donge Tang
- Department
of Clinical Medical Research Center, The
Second Clinical Medical College of Jinan University, Shenzhen People’s
Hospital, Shenzhen, Guangdong 518020, China
| | - Lianghong Yin
- Institute
of Nephrology and Blood Purification, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
- Huangpu
Institute of Materials, Guangzhou, Guangdong 510663, China
| | - Yong Dai
- Department
of Clinical Medical Research Center, The
Second Clinical Medical College of Jinan University, Shenzhen People’s
Hospital, Shenzhen, Guangdong 518020, China
- Guangxi
Key Laboratory of Metabolic Diseases Research, No. 924 Hospital of PLA Joint Logistic Support
Force, Guilin, Guangxi 541002, China
- The First
Affiliated Hospital, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, China
| |
Collapse
|
24
|
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
|
25
|
Lu X, Wang L, Wang M, Li Y, Zhao Q, Shi Y, Zhang Y, Wang Y, Wang W, Ji L, Hou H, Li D. Association between immunoglobulin G N-glycosylation and lupus nephritis in female patients with systemic lupus erythematosus: a case-control study. Front Immunol 2023; 14:1257906. [PMID: 37809087 PMCID: PMC10552529 DOI: 10.3389/fimmu.2023.1257906] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
Background Lupus nephritis (LN) is a crucial complication of systemic lupus erythematosus (SLE) and has important clinical implications in guiding treatment. N-glycosylation of immunoglobulin G (IgG) plays a key role in the development of SLE by affecting the balance of anti-inflammatory and proinflammatory responses. This study aimed to evaluate the performance of IgG N-glycosylation for diagnosing LN in a sample of female SLE patients. Methods This case-control study recruited 188 women with SLE, including 94 patients with LN and 94 age-matched patients without LN. The profiles of plasma IgG N-glycans were detected by hydrophilic interaction chromatography with ultra-performance liquid chromatography (HILIC-UPLC). A multivariate logistic regression model was used to explore the associations between IgG N-glycans and LN. A diagnostic model was developed using the significant glycans as well as demographic factors. The performance of IgG N-glycans in the diagnosis of LN was evaluated by receiver operating characteristic (ROC) curve analysis, and the area under the curve (AUC) and its 95% confidence interval (CI) were calculated. Results There were significant differences in 9 initial glycans (GP2, GP4, GP6, GP8, GP10, GP14, GP16, GP18 and GP23) between women with SLE with and without LN (P < 0.05). The levels of sialylated, galactosylated and fucosylated glycans were significantly lower in the LN patients than in the control group, while bisected N-acetylglucosamine (GlcNAc) glycans were increased in LN patients (P < 0.05). GP8, GP10, GP18, and anemia were included in our diagnostic model, which performed well in differentiating female SLE patients with LN from those without LN (AUC = 0.792, 95% CI: 0.727 to 0.858). Conclusion Our findings indicate that decreased sialylation, galactosylation, and core fucosylation and increased bisecting GlcNAc might play a role in the development of LN by upregulating the proinflammatory response of IgG. IgG N-glycans can serve as potential biomarkers to differentiate individuals with LN among SLE patients.
Collapse
Affiliation(s)
- Xinxia Lu
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Liangao Wang
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Meng Wang
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Jinshan District Center for Disease Control and Prevention, Shanghai, China
| | - Yuejin Li
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, China
| | - Qinqin Zhao
- Department of Geriatric Cognitive Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Yanjun Shi
- Department of Rheumatology and Immunology, Liaocheng People’s Hospital, Liao’cheng, China
| | - Yujing Zhang
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yingjie Wang
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wei Wang
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Long Ji
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, China
| | - Haifeng Hou
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Dong Li
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Clinical Research Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| |
Collapse
|
26
|
Li L, Li L, Shao Y, Du R, Li L, Shi X, Bai Y. Calcium/calmodulin dependent protein kinase IV in trophoblast cells under insulin resistance: functional and metabolomic analyses. Mol Med 2023; 29:82. [PMID: 37386367 DOI: 10.1186/s10020-023-00669-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 05/24/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Insulin resistance (IR) is an important determinant of glucose metabolic disturbance and placental dysplasia in gestational diabetes mellitus (GDM). Calcium/calmodulin dependent protein kinase IV (CAMK4) improves insulin IR induced by a high-fat diet (HFD). The current study sought to elucidate the role and potential mechanism of CAMK4 in GDM. METHODS A GDM model was established in female C57BL/6J mice via HFD feeding for one week before mating and throughout gestation. The IR was elicited by 10-6 M insulin treatment for 48 h in HTR-8/SVneo cells and mouse primary trophoblast cells. The function of CAMK4 was investigated by transfection of overexpression plasmid in HTR-8/SVneo cells and infection of lentivirus loaded with CAMK4 encoding sequence in primary trophoblast cells. Real-time PCR, western blot, cell counting kit-8, transwell, wound healing, dual-luciferase reporter assay, and liquid chromatography/mass spectrometry-based untargeted metabolomics were performed to confirm the effects of CAMK4 on trophoblast cells. RESULTS Decreased CAMK4 expression was found in the placenta of GDM mice. CAMK4 overexpression ameliorated IR-induced viability impairment, migratory and invasive capacity inhibition, autophagy blocking, insulin signaling inactivation and glucose uptake disorder in trophoblast cells. CAMK4 also transcriptionally activated orphan nuclear receptor NUR77, and the effects of CAMK4 were abrogated by silencing of NUR77. Metabolomics analysis revealed that CAMK4 overexpression caused alterations of amino acid, lipid and carbohydrate metabolism, which were important in GDM. CONCLUSION Our results indicated that CAMK4/NUR77 axis may provide novel potential targets in GDM treatment.
Collapse
Affiliation(s)
- Ling Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning, People's Republic of China
| | - Li Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning, People's Republic of China
| | - Ying Shao
- Department of Endocrinology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning, People's Republic of China
| | - Runyu Du
- Department of Endocrinology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning, People's Republic of China
| | - Ling Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning, People's Republic of China
| | - Xiaoguang Shi
- Department of Endocrinology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning, People's Republic of China.
| | - Yu Bai
- Department of Endocrinology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning, People's Republic of China.
| |
Collapse
|
27
|
Diamond B. Not Dead Yet. Annu Rev Immunol 2023; 41:1-15. [PMID: 37126416 DOI: 10.1146/annurev-immunol-101721-065214] [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: 05/02/2023]
Abstract
I have been a scientific grasshopper throughout my career, moving from question to question within the domain of lupus. This has proven to be immensely gratifying. Scientific exploration is endlessly fascinating, and succeeding in studies you care about with colleagues and trainees leads to strong and lasting bonds. Science isn't easy; being a woman in science presents challenges, but the drive to understand a disease remains strong.
Collapse
Affiliation(s)
- Betty Diamond
- Center of Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA;
| |
Collapse
|
28
|
Bhargava R, Li H, Tsokos GC. Pathogenesis of lupus nephritis: the contribution of immune and kidney resident cells. Curr Opin Rheumatol 2023; 35:107-116. [PMID: 35797522 DOI: 10.1097/bor.0000000000000887] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Lupus nephritis is associated with significant mortality and morbidity. We lack effective therapeutics and biomarkers mostly because of our limited understanding of its complex pathogenesis. We aim to present an overview of the recent advances in the field to gain a deeper understanding of the underlying cellular and molecular mechanisms involved in lupus nephritis pathogenesis. RECENT FINDINGS Recent studies have identified distinct roles for each resident kidney cell in the pathogenesis of lupus nephritis. Podocytes share many elements of innate and adaptive immune cells and they can present antigens and participate in the formation of crescents in coordination with parietal epithelial cells. Mesangial cells produce pro-inflammatory cytokines and secrete extracellular matrix contributing to glomerular fibrosis. Tubular epithelial cells modulate the milieu of the interstitium to promote T cell infiltration and formation of tertiary lymphoid organs. Modulation of specific genes in kidney resident cells can ward off the effectors of the autoimmune response including autoantibodies, cytokines and immune cells. SUMMARY The development of lupus nephritis is multifactorial involving genetic susceptibility, environmental triggers and systemic inflammation. However, the role of resident kidney cells in the development of lupus nephritis is becoming more defined and distinct. More recent studies point to the restoration of kidney resident cell function using cell targeted approaches to prevent and treat lupus nephritis.
Collapse
Affiliation(s)
- Rhea Bhargava
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard, Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
29
|
Yang Y, Yan C, Yu L, Zhang X, Shang J, Fan J, Zhang R, Ren J, Duan X. The star target in SLE: IL-17. Inflamm Res 2023; 72:313-328. [PMID: 36538077 DOI: 10.1007/s00011-022-01674-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/30/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE The purpose of this review is to discuss the significance of IL-17 in SLE and the potential of IL-17-targeted therapy. BACKGROUND Systemic lupus erythematosus (SLE) is an autoimmune disease that can affect many organs and tissues throughout the body. It is characterized by overactive B and T cells and loss of immune tolerance to autoantigens. Interleukin-17 (IL-17) is a cytokine that promotes inflammation and has been implicated in the pathogenesis of several autoimmune diseases as well as inflammatory diseases. In in vitro cellular experiments in lupus susceptible mice or SLE patients, there is substantial evidence that IL-17 is a highly promising therapeutic target. METHODS We searched papers from PubMed database using the search terms, such as interleukin-17, systemic lupus erythematosus, treatment targets, T cells, lupus nephritis, and other relevant terms. RESULTS We discuss in this paper the molecular mechanisms of IL-17 expression, Th17 cell proliferation, and the relationship between IL-17 and Th17. The significance of IL-17 in SLE and the potential of IL-17-targeted therapy are further discussed in detail. CONCLUSION IL-17 has a very high potential for the development as a star target in SLE.
Collapse
Affiliation(s)
- Yi Yang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chen Yan
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Le Yu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiuling Zhang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jingjing Shang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jie Fan
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rongwei Zhang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jie Ren
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xinwang Duan
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
30
|
Ramos-Martínez I, Ramos-Martínez E, Cerbón M, Pérez-Torres A, Pérez-Campos Mayoral L, Hernández-Huerta MT, Martínez-Cruz M, Pérez-Santiago AD, Sánchez-Medina MA, García-Montalvo IA, Zenteno E, Matias-Cervantes CA, Ojeda-Meixueiro V, Pérez-Campos E. The Role of B Cell and T Cell Glycosylation in Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:ijms24010863. [PMID: 36614306 PMCID: PMC9820943 DOI: 10.3390/ijms24010863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 01/05/2023] Open
Abstract
Glycosylation is a post-translational modification that affects the stability, structure, antigenicity and charge of proteins. In the immune system, glycosylation is involved in the regulation of ligand-receptor interactions, such as in B-cell and T-cell activating receptors. Alterations in glycosylation have been described in several autoimmune diseases, such as systemic lupus erythematosus (SLE), in which alterations have been found mainly in the glycosylation of B lymphocytes, T lymphocytes and immunoglobulins. In immunoglobulin G of lupus patients, a decrease in galactosylation, sialylation, and nucleotide fucose, as well as an increase in the N-acetylglucosamine bisector, are observed. These changes in glycoisolation affect the interactions of immunoglobulins with Fc receptors and are associated with pericarditis, proteinuria, nephritis, and the presence of antinuclear antibodies. In T cells, alterations have been described in the glycosylation of receptors involved in activation, such as the T cell receptor; these changes affect the affinity with their ligands and modulate the binding to endogenous lectins such as galectins. In T cells from lupus patients, a decrease in galectin 1 binding is observed, which could favor activation and reduce apoptosis. Furthermore, these alterations in glycosylation correlate with disease activity and clinical manifestations, and thus have potential use as biomarkers. In this review, we summarize findings on glycosylation alterations in SLE and how they relate to immune system defects and their clinical manifestations.
Collapse
Affiliation(s)
- Ivan Ramos-Martínez
- Departamento de Medicina y Zootecnia de Cerdos, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Edgar Ramos-Martínez
- Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
- Escuela de Ciencias, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología “Isidro Espinosa de los Reyes”—Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Armando Pérez-Torres
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | | | - María Teresa Hernández-Huerta
- CONACyT, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca (UABJO), Oaxaca 68020, Mexico
| | | | | | | | | | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | | | | | | |
Collapse
|
31
|
B7-1 mediates podocyte injury and glomerulosclerosis through communication with Hsp90ab1-LRP5-β-catenin pathway. Cell Death Differ 2022; 29:2399-2416. [PMID: 35710882 PMCID: PMC9750974 DOI: 10.1038/s41418-022-01026-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 02/08/2023] Open
Abstract
Podocyte injury is a hallmark of glomerular diseases; however, the underlying mechanisms remain unclear. B7-1 is increased in injured podocytes, but its intrinsic role is controversial. The clinical data here revealed the intimate correlation of urinary B7-1 with severity of glomerular injury. Through transcriptomic and biological assays in B7-1 transgenic and adriamycin nephropathy models, we identified B7-1 is a key mediator in podocyte injury and glomerulosclerosis through a series of signal transmission to β-catenin. Using LC-MS/MS, Hsp90ab1, a conserved molecular chaperone, was distinguished to be an anchor for transmitting signals from B7-1 to β-catenin. Molecular docking and subsequent mutant analysis further identified the residue K69 in the N terminal domain of Hsp90ab1 was the key binding site for B7-1 to activate LRP5/β-catenin pathway. The interaction and biological functions of B7-1-Hsp90ab1-LRP5 complex were further demonstrated in vitro and in vivo. We also found B7-1 is a novel downstream target of β-catenin. Our results indicate an intercrossed network of B7-1, which collectively induces podocyte injury and glomerulosclerosis. Our study provides an important clue to improve the therapeutic strategies to target B7-1.
Collapse
|
32
|
Liu T, Yang L, Mao H, Ma F, Wang Y, Li S, Li P, Zhan Y. Sirtuins as novel pharmacological targets in podocyte injury and related glomerular diseases. Biomed Pharmacother 2022; 155:113620. [PMID: 36122519 DOI: 10.1016/j.biopha.2022.113620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/10/2022] [Accepted: 08/27/2022] [Indexed: 11/29/2022] Open
Abstract
Podocyte injury is a major cause of proteinuria in kidney diseases, and persistent loss of podocytes leads to rapid irreversible progression of kidney disease. Sirtuins, a class of nicotinamide adenine dinucleotide-dependent deacetylases, can promote DNA repair, modify transcription factors, and regulate the cell cycle. Additionally, sirtuins play a critical role in renoprotection, particularly against podocyte injury. They also have pleiotropic protective effects on podocyte injury-related glomerular diseases, such as improving the immune inflammatory status and oxidative stress levels, maintaining mitochondrial homeostasis, enhancing autophagy, and regulating lipid metabolism. Sirtuins deficiency causes podocyte injury in different glomerular diseases. Studies using podocyte sirtuin-specific knockout and transgenic models corroborate this conclusion. Of note, sirtuin activators have protective effects in different podocyte injury-related glomerular diseases, including diabetic kidney disease, focal segmental glomerulosclerosis, membranous nephropathy, IgA nephropathy, and lupus nephritis. These findings suggest that sirtuins are promising therapeutic targets for preventing podocyte injury. This review provides an overview of recent advances in the role of sirtuins in kidney diseases, especially their role in podocyte injury, and summarizes the possible rationale for sirtuins as targets for pharmacological intervention in podocyte injury-related glomerular diseases.
Collapse
Affiliation(s)
- Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyang Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shen Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China.
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
33
|
Ren W, Bian Q, Cai Y. Mass spectrometry-based N-glycosylation analysis in kidney disease. Front Mol Biosci 2022; 9:976298. [PMID: 36072428 PMCID: PMC9442644 DOI: 10.3389/fmolb.2022.976298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/18/2022] [Indexed: 11/14/2022] Open
Abstract
Kidney disease is a global health concern with an enormous expense. It is estimated that more than 10% of the population worldwide is affected by kidney disease and millions of patients would progress to death prematurely and unnecessarily. Although creatinine detection and renal biopsy are well-established tools for kidney disease diagnosis, they are limited by several inevitable defects. Therefore, diagnostic tools need to be upgraded, especially for the early stage of the disease and possible progression. As one of the most common post-translational modifications of proteins, N-glycosylation plays a vital role in renal structure and function. Deepening research on N-glycosylation in kidney disease provides new insights into the pathophysiology and paves the way for clinical application. In this study, we reviewed recent N-glycosylation studies on several kidney diseases. We also summarized the development of mass spectrometric methods in the field of N-glycoproteomics and N-glycomics.
Collapse
Affiliation(s)
- Weifu Ren
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Nephrology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qi Bian
- Department of Nephrology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yan Cai
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Gaun A, Preciado López M, Olsson N, Wang JCK, Chan LJG, O'Brien J, Li W, Zavala‐Solorio J, Zhang C, Eaton D, McAllister FE. Triple‐threat quantitative multiplexed plasma proteomics analysis on immune complex disease MRL‐lpr mice. Proteomics 2022; 22:e2100242. [DOI: 10.1002/pmic.202100242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/30/2022] [Accepted: 07/22/2022] [Indexed: 11/07/2022]
|
35
|
Liou LB, Chen CC, Chiang WY, Chen MH. De-sialylated and sialylated IgG anti-dsDNA antibodies respectively worsen and mitigate experimental mouse lupus proteinuria and possible mechanisms. Int Immunopharmacol 2022; 109:108837. [DOI: 10.1016/j.intimp.2022.108837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 01/10/2023]
|
36
|
Intertwined pathways of complement activation command the pathogenesis of lupus nephritis. Transl Res 2022; 245:18-29. [PMID: 35296451 PMCID: PMC9167748 DOI: 10.1016/j.trsl.2022.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/26/2022]
Abstract
The complement system is involved in the origin of autoimmunity and systemic lupus erythematosus. Both genetic deficiency of complement components and excessive activation are involved in primary and secondary renal diseases, including lupus nephritis. Among the pathways, the classical pathway has long been accepted as the main pathway of complement activation in systemic lupus erythematosus. However, more recent studies have shown the contribution of factors B and D which implies the involvement of the alternative pathway. While there is evidence on the role of the lectin pathway in systemic lupus erythematosus, it is yet to be demonstrated whether this pathway is protective or harmful in lupus nephritis. Complement is being explored for the development of disease biomarkers and therapeutic targeting. In the current review we discuss the involvement of complement in lupus nephritis.
Collapse
|
37
|
Flevaris K, Kontoravdi C. Immunoglobulin G N-glycan Biomarkers for Autoimmune Diseases: Current State and a Glycoinformatics Perspective. Int J Mol Sci 2022; 23:5180. [PMID: 35563570 PMCID: PMC9100869 DOI: 10.3390/ijms23095180] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
The effective treatment of autoimmune disorders can greatly benefit from disease-specific biomarkers that are functionally involved in immune system regulation and can be collected through minimally invasive procedures. In this regard, human serum IgG N-glycans are promising for uncovering disease predisposition and monitoring progression, and for the identification of specific molecular targets for advanced therapies. In particular, the IgG N-glycome in diseased tissues is considered to be disease-dependent; thus, specific glycan structures may be involved in the pathophysiology of autoimmune diseases. This study provides a critical overview of the literature on human IgG N-glycomics, with a focus on the identification of disease-specific glycan alterations. In order to expedite the establishment of clinically-relevant N-glycan biomarkers, the employment of advanced computational tools for the interpretation of clinical data and their relationship with the underlying molecular mechanisms may be critical. Glycoinformatics tools, including artificial intelligence and systems glycobiology approaches, are reviewed for their potential to provide insight into patient stratification and disease etiology. Challenges in the integration of such glycoinformatics approaches in N-glycan biomarker research are critically discussed.
Collapse
Affiliation(s)
| | - Cleo Kontoravdi
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
38
|
Alves I, Fernandes Â, Santos-Pereira B, Azevedo CM, Pinho SS. Glycans as a key factor in self and non-self discrimination: Impact on the breach of immune tolerance. FEBS Lett 2022; 596:1485-1502. [PMID: 35383918 DOI: 10.1002/1873-3468.14347] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/17/2022] [Accepted: 03/29/2022] [Indexed: 11/09/2022]
Abstract
Glycans are carbohydrates that are made by all organisms and covalently conjugated to other biomolecules. Glycans cover the surface of both human cells and pathogens and are fundamental to defining the identity of a cell or an organism, thereby contributing to discriminating self from non-self. As such, glycans are a class of "Self-Associated Molecular Patterns" that can fine-tune host inflammatory processes. In fact, glycans can be sensed and recognized by a variety of glycan-binding proteins (GBP) expressed by immune cells, such as galectins, siglecs and C-type lectins, which recognize changes in the cellular glycosylation, instructing both pro-inflammatory or anti-inflammatory responses. In this review, we introduce glycans as cell-identification structures, discussing how glycans modulate host-pathogen interactions and how they can fine-tune inflammatory processes associated with infection, inflammation and autoimmunity. Finally, from the clinical standpoint, we discuss how glycoscience research can benefit life sciences and clinical medicine by providing a source of valuable biomarkers and therapeutic targets for immunity.
Collapse
Affiliation(s)
- Inês Alves
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ângela Fernandes
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Beatriz Santos-Pereira
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Catarina M Azevedo
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal
| | - Salomé S Pinho
- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar, University of Porto, Portugal
| |
Collapse
|
39
|
Vicente MM, Alves I, Gaifem J, Rodrigues CS, Fernandes Â, Dias AM, Štambuk J, Petrović T, Oliveira P, Ferreira-da-Silva F, Soares A, Seixas N, Teixeira T, Malheiro L, Abreu MM, Lauc G, Sarmento E Castro R, Pinho SS. Altered IgG glycosylation at COVID-19 diagnosis predicts disease severity. Eur J Immunol 2022; 52:946-957. [PMID: 35307819 PMCID: PMC9087392 DOI: 10.1002/eji.202149491] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 02/04/2022] [Accepted: 03/15/2022] [Indexed: 01/08/2023]
Abstract
The nature of the immune responses associated with COVID‐19 pathogenesis and disease severity, as well as the breadth of vaccine coverage and duration of immunity, is still unclear. Given the unpredictability for developing a severe/complicated disease, there is an urgent need in the field for predictive biomarkers of COVID‐19. We have analyzed IgG Fc N‐glycan traits of 82 SARS‐CoV‐2+ unvaccinated patients, at diagnosis, by nano‐LC‐ESI‐MS. We determined the impact of IgG Fc glyco‐variations in the induction of NK cells activation, further evaluating the association between IgG Fc N‐glycans and disease severity/prognosis. We found that SARS‐CoV‐2+ individuals display, at diagnosis, variations in the glycans composition of circulating IgGs. Importantly, levels of galactose and sialic acid structures on IgGs are able to predict the development of a poor COVID‐19 disease. Mechanistically, we demonstrated that a deficiency on galactose structures on IgG Fc in COVID‐19 patients appears to induce NK cells activation associated with increased release of IFN‐γ and TNF‐α, which indicates the presence of pro‐inflammatory immunoglobulins and higher immune activation, associated with a poor disease course. This study brings to light a novel blood biomarker based on IgG Fc glycome composition with capacity to stratify patients at diagnosis.
Collapse
Affiliation(s)
- Manuel M Vicente
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Graduate Program in Areas of Basic and Applied Biology (GABBA), ICBAS, University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of University of Porto (IPATIMUP), Porto, Portugal
| | - Inês Alves
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of University of Porto (IPATIMUP), Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Joana Gaifem
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of University of Porto (IPATIMUP), Porto, Portugal
| | - Cláudia S Rodrigues
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of University of Porto (IPATIMUP), Porto, Portugal
| | - Ângela Fernandes
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of University of Porto (IPATIMUP), Porto, Portugal
| | - Ana M Dias
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of University of Porto (IPATIMUP), Porto, Portugal
| | - Jerko Štambuk
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
| | - Tea Petrović
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
| | - Pedro Oliveira
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Frederico Ferreira-da-Silva
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Adriana Soares
- Internal Medicine Department, Hospital Beatriz Ângelo, Loures, Portugal
| | - Nair Seixas
- Department of Infectious Diseases, Centro Hospitalar Vila Nova de Gaia/Espinho, Gaia, Portugal
| | - Tiago Teixeira
- Department of Infectious Diseases, Centro Hospitalar Vila Nova de Gaia/Espinho, Gaia, Portugal
| | - Luis Malheiro
- Department of Clinical Pathology, Centro Hospitalar Vila Nova de Gaia/Espinho, Gaia, Portugal
| | - Miguel M Abreu
- Department of Infectious Diseases, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Gordan Lauc
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Rui Sarmento E Castro
- Department of Infectious Diseases, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Salomé S Pinho
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of University of Porto (IPATIMUP), Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
40
|
Jorge AM, Lao T, Kim R, Licciardi S, El Khoury J, Luster AD, Means TK, Ramirez-Ortiz ZG. SCARF1-Induced Efferocytosis Plays an Immunomodulatory Role in Humans, and Autoantibodies Targeting SCARF1 Are Produced in Patients with Systemic Lupus Erythematosus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:955-967. [PMID: 35082161 PMCID: PMC8852219 DOI: 10.4049/jimmunol.2100532] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 12/02/2021] [Indexed: 12/20/2022]
Abstract
Deficiency in the clearance of cellular debris is a major pathogenic factor in the emergence of autoimmune diseases. We previously demonstrated that mice deficient for scavenger receptor class F member 1 (SCARF1) develop a lupus-like autoimmune disease with symptoms similar to human systemic lupus erythematosus (SLE), including a pronounced accumulation of apoptotic cells (ACs). Therefore, we hypothesized that SCARF1 will be important for clearance of ACs and maintenance of self-tolerance in humans, and that dysregulation of this process could contribute to SLE. In this article, we show that SCARF1 is highly expressed on phagocytic cells, where it functions as an efferocytosis receptor. In healthy individuals, we discovered that engagement of SCARF1 by ACs on BDCA1+ dendritic cells initiates an IL-10 anti-inflammatory response mediated by the phosphorylation of STAT1 and STAT3. Unexpectedly, there was no significant difference in SCARF1 expression in samples of patients with SLE compared with healthy donor samples. However, we detected anti-SCARF1 autoantibodies in 26% of patients with SLE, which was associated with dsDNA Ab positivity. Furthermore, our data show a direct correlation of the levels of anti-SCARF1 in the serum and defects in the removal of ACs. Depletion of Ig restores efferocytosis in SLE serum, suggesting that defects in the removal of ACs are partially mediated by SCARF1 pathogenic autoantibodies. Our data demonstrate that human SCARF1 is an AC receptor in dendritic cells and plays a role in maintaining tolerance and homeostasis.
Collapse
Affiliation(s)
- April M Jorge
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA
| | - Taotao Lao
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA
| | - Rachel Kim
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA
| | - Samantha Licciardi
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA; and
| | - Joseph El Khoury
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA
| | - Terry K Means
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA
- Autoimmunity Cluster, Immunology & Inflammation Research Therapeutic Area, Sanofi, Cambridge, MA
| | - Zaida G Ramirez-Ortiz
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA;
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA; and
| |
Collapse
|
41
|
Guo A, Sun Y, Xu X, Xing Q. MicroRNA-30a Targets Notch1 to Alleviate Podocyte Injury in Lupus Nephritis. Immunol Invest 2022; 51:1694-1706. [PMID: 35023444 DOI: 10.1080/08820139.2022.2027440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The microRNA miR-30a has been reported to mitigate podocyte damage and resist injurious factors in lupus nephritis (LN), but the precise molecular mechanisms underlying these effects remain elusive. We hypothesized that miR-30a can ameliorate podocyte injury by downregulating the Notch1 signaling pathway and investigated the role of miR-30a in the pathogenesis of podocyte-treated with Immunoglobulin G from patients with LN (IgG-LN). The study enrolled 30 patients from new-onset systemic lupus erythematosus and 28 healthy individuals, then evaluated the levels of their serum miR-30a using RT-qPCR. Additionally, MPC5 cells were transfected with NICD-vector to overexpress Notch1, then with miR-30a mimics or inhibitors to determine miR-30a effects on Notch1. Analysis of function and regulatory mechanisms were performed with RT-qPCR, Western blotting, and CCK8 assays. Furthermore, we verified the candidate sequence targeted by miR-30a using a luciferase reporter gene assay. We observed a significant decrease in the serum miR-30a levels in patients with LN. Also, in IgG-LN-treated podocytes, miR-30a decreased and Notch1 expression was elevated. Bioinformatic analysis and transfection experiments revealed that Notch1 is a direct target of miR-30a. Further supporting this finding, miR-30a upregulation appeared to alleviate IgG-LN-treated podocyte injury, and Notch1 overexpression reversed this effect. To conclude, miR-30a can ameliorate podocyte injury via suppression of the Notch1 signaling pathway.
Collapse
Affiliation(s)
- Aoyang Guo
- Department of Immune Rheumatology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
| | - Yadi Sun
- Department of Immune Rheumatology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
| | - Xiaona Xu
- Department of Immune Rheumatology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
| | - Qian Xing
- Department of Immune Rheumatology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
42
|
Koga T, Ichinose K, Tsokos GC. Tissue resident cell processes determine organ damage in systemic lupus erythematosus. Clin Immunol 2022; 234:108919. [PMID: 34974170 DOI: 10.1016/j.clim.2021.108919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 12/25/2021] [Indexed: 11/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that affects almost any organ. Multiple immunological abnormalities involving every domain of the immune system contribute to the expression of the disease. It is now recognized that elements of the immune system instigate processes in tissue resident cells which execute organ damage. Although correction of ongoing immune aberrations is important in the control of disease activity, targeting tissue specific injurious processes may prove desirable in limiting organ damage.
Collapse
Affiliation(s)
- Tomohiro Koga
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - Kunihiro Ichinose
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - George C Tsokos
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
43
|
Glycosylation influences IgG effects in LN. Nat Rev Rheumatol 2021; 17:310. [PMID: 33941914 DOI: 10.1038/s41584-021-00626-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
44
|
Clinical Mass Spectrometry Discovered Human IgG Sialylation as a Potential Biosignature for Kidney Function. J Pers Med 2021; 11:jpm11080761. [PMID: 34442405 PMCID: PMC8401842 DOI: 10.3390/jpm11080761] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023] Open
Abstract
Immunoglobulin G (IgG) N-glycosylation was discovered to have an association with inflammation status, which has the potential to be a novel biomarker for kidney diseases. In this study, we applied an ultra-high performance liquid chromatography–tandem mass spectrometry (UHPLC-MS/MS) method to plasma and urine samples from 57 individuals with different levels of kidney function. Natural abundances of total IgG, IgG1, IgG2, and IgG3 subclasses in plasma showed positive correlations to the estimated glomerular filtration rates (eGFRs). Eighteen IgG glycopeptides also showed positive correlations. In contrast, higher IgG amounts were found in urine samples from participants with lower eGFR values. After normalizing IgG glycopeptides from plasma to their respective protein amounts, H4N4F1S1-IgG1 (r = 0.37, p = 0.0047, significant) and H5N4F1S1-IgG1 (r = 0.25, p = 0.063, marginally significant) were the two glycopeptides that still had positive correlations with eGFRs. The results showed that the UHPLC-MS/MS method is capable of investigating IgG profiles, and monitoring IgG and glycosylation patterns is worthy of further clinical application for kidney disease.
Collapse
|
45
|
Abstract
Changes in immunoglobulin G (IgG) glycosylation pattern have been observed in a vast array of auto- and alloimmune, infectious, cardiometabolic, malignant, and other diseases. This chapter contains an updated catalog of over 140 studies within which IgG glycosylation analysis was performed in a disease setting. Since the composition of IgG glycans is known to modulate its effector functions, it is suggested that a changed IgG glycosylation pattern in patients might be involved in disease development and progression, representing a predisposition and/or a functional effector in disease pathology. In contrast to the glycopattern of bulk serum IgG, which likely relates to the systemic inflammatory background, the glycosylation profile of antigen-specific IgG probably plays a direct role in disease pathology in several infectious and allo- and autoimmune antibody-dependent diseases. Depending on the specifics of any given disease, IgG glycosylation read-out might therefore in the future be developed into a useful clinical biomarker or a supplementary to currently used biomarkers.
Collapse
Affiliation(s)
- Marija Pezer
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia.
| |
Collapse
|