1
|
Wang Y, Lan Q, Li F, Xiong J, Xie H, Gong S, Yao M, Lv L, Qin S, Xin W, Zhang A, Zhou S, Huang Y, Zhao J. Macrophage-Derived Type 1 IFN, Renal Tubular Epithelial Cell Polyploidization, and AKI-to-CKD Transition. J Am Soc Nephrol 2025; 36:766-780. [PMID: 39665291 PMCID: PMC12059107 DOI: 10.1681/asn.0000000577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/28/2024] [Indexed: 12/13/2024] Open
Abstract
Key Points Macrophage-derived IFN-β contributes to tubular epithelial cell polyploidization after AKI. IFN-β induced tubular epithelial cell polyploidization by regulating inorganic pyrophosphatase-mediated yes-associated protein (YAP) dephosphorylation. Delayed blockade of the IFN-β response attenuated persistent polyploidization and kidney fibrosis. Background AKI is recognized as a common risk factor of CKD. Renal tubular epithelial cell polyploidization after AKI is closely associated with maladaptive repair, while the regulatory and molecular mechanisms remain poorly understood. In this study, we set out to investigate the mechanism of tubular epithelial cell polyploidization and their role in AKI-to-CKD transition. Methods The change characters of polyploid tubular epithelial cells and macrophages after AKI were detected by flow cytometry and immunofluorescence. The underlying mechanism was explored by RNA-sequencing analysis, immunofluorescence, and Western blot. The role of tubular epithelial cell polyploidization in AKI-to-CKD transition was evaluated by transgenic mice and drug interventions. Results We discovered that tubular epithelial cells underwent polyploidization after AKI, and polyploid tubular epithelial cells exhibited greater fibrotic phenotypes than nonpolyploid cells. Furthermore, we revealed an upregulated IFN-β response feature within tubular epithelial cells after AKI and identified that macrophage-derived IFN-β bound to IFN-I receptor 1 of tubular epithelial cells and induced their polyploidization. Mechanistically, IFN-β , secreted by macrophages through activation of the cyclic guanosine monophosphate-AMP synthase-stimulator of IFN genes pathway, acted on tubular epithelial cells and facilitated inorganic pyrophosphatase binding to yes-associated protein (YAP), which mediated YAP dephosphorylation and subsequent nuclear translocation, culminating in p21 expression and polyploidization. Importantly, delayed blockade of the IFN-β response and pharmacological inhibition of stimulator of IFN genes or YAP activation on day 4 after AKI significantly attenuated persistent tubular epithelial cell polyploidization and AKI-induced kidney fibrosis. Conclusions Macrophage-derived IFN-β contributed to tubular epithelial cell polyploidization by regulating inorganic pyrophosphatase/YAP signaling pathway–mediated p21 expression and further promoted AKI-to-CKD transition.
Collapse
Affiliation(s)
- Yaqin Wang
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qigang Lan
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Fugang Li
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiachuan Xiong
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Hailun Xie
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuiqin Gong
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Mengying Yao
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Liangjing Lv
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shaozong Qin
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wang Xin
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Aihong Zhang
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Siyan Zhou
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yinghui Huang
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jinghong Zhao
- Department of Nephrology, Chongqing Key Laboratory of Prevention and Treatment of Kidney Disease, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
2
|
Zhang C, Guo J. Cell cycle disorders in podocytes: an emerging and increasingly recognized phenomenon. Cell Death Discov 2025; 11:182. [PMID: 40246828 PMCID: PMC12006314 DOI: 10.1038/s41420-025-02486-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
Proteinuria is observed in various kidney diseases and is frequently associated with a compromised glomerular filtration barrier. Podocytes, as a crucial component of this barrier, play an essential role in preserving the kidney's normal filtration function. Podocytes are terminally differentiated cells that typically do not proliferate. However, certain harmful stimuli can trigger podocytes to re-enter the cell cycle. Due to its unique cytoskeletal structure, podocytes are unable to maintain the structure of the foot process and complete cell division at the same time, eventually form binucleated or multinucleated podocytes. Studies have found that podocytes re-entering the cell cycle are more susceptible to injury, and are prone to detachment from the basement membrane or apoptosis, which are accompanied by the widening of foot processes. This eventually leads to podocyte mitotic catastrophe and the development of proteinuria. Podocyte cell cycle disorders have previously been found mainly in focal segmental glomerulosclerosis and IgA nephropathy. In recent years, this phenomenon has been frequently identified in diabetic kidney disease and lupus nephritis. An expanding body of research has begun to investigate the mechanisms underlying podocyte cell cycle disorders, including cell cycle re-entry, cell cycle arrest, and mitotic catastrophe. This review consolidates the existing literature on podocyte cell cycle disorders in renal diseases and summarizes the molecules that trigger podocyte re-entry into the cell cycle, thereby providing new drug targets for mitigating podocyte damage. This is essential for alleviating podocyte injury, reducing proteinuria, and delaying the progression of kidney diseases.
Collapse
Affiliation(s)
- Chaojie Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jia Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, Henan, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
3
|
Brandauer K, Lorenz A, Schobesberger S, Schuller P, Frauenlob M, Spitz S, Ertl P. Sensor-integrated gut-on-a-chip for monitoring senescence-mediated changes in the intestinal barrier. LAB ON A CHIP 2025; 25:1694-1706. [PMID: 40007323 DOI: 10.1039/d4lc00896k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
The incidence of inflammatory bowel disease among the elderly has significantly risen in recent years, posing a growing socioeconomic burden to aging societies. Moreover, non-gastrointestinal diseases, also prevalent in this demographic, have been linked to intestinal barrier dysfunction, thus highlighting the importance of investigating aged-mediated changes within the human gut. While gastrointestinal pathology often involves an impaired gut barrier, the impact of aging on the human gastrointestinal barrier function remains unclear. To explore the effect of senescence, a key hallmark of aging, on gut barrier integrity, we established and evaluated an in vitro gut-on-a-chip model tailored to investigate barrier changes by the integration of an impedance sensor. Here, a microfluidic gut-on-a-chip system containing integrated membrane-based electrode microarrays is used to non-invasively monitor epithelial barrier formation and senescence-mediated changes in barrier integrity upon treating Caco-2 cells with 0.8 μg mL-1 doxorubicin (DXR), a chemotherapeutic which induces cell cycle arrest. Results of our microfluidic human gut model reveal a DXR-mediated increase in impedance and cell hypertrophy as well as overexpression of p21, and CCL2, indicative of a senescent phenotype. Combined with the integrated electrodes, monitoring ∼57% of the cultivation area in situ and non-invasively, the developed chip-based senescent-gut model is ideally suited to study age-related malfunctions in barrier integrity.
Collapse
Affiliation(s)
- Konstanze Brandauer
- TU Wien, Faculty of Technical Chemistry, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Alexandra Lorenz
- TU Wien, Faculty of Technical Chemistry, Getreidemarkt 9, 1060 Vienna, Austria.
| | | | - Patrick Schuller
- TU Wien, Faculty of Technical Chemistry, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Martin Frauenlob
- TU Wien, Faculty of Technical Chemistry, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Sarah Spitz
- TU Wien, Faculty of Technical Chemistry, Getreidemarkt 9, 1060 Vienna, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Peter Ertl
- TU Wien, Faculty of Technical Chemistry, Getreidemarkt 9, 1060 Vienna, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
4
|
Maslyennikov Y, Bărar AA, Rusu CC, Potra AR, Tirinescu D, Ticala M, Urs A, Pralea IE, Iuga CA, Moldovan DT, Kacso IM. The Spectrum of Minimal Change Disease/Focal Segmental Glomerulosclerosis: From Pathogenesis to Proteomic Biomarker Research. Int J Mol Sci 2025; 26:2450. [PMID: 40141093 PMCID: PMC11941885 DOI: 10.3390/ijms26062450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Podocyte injury plays a central role in both focal segmental glomerulosclerosis (FSGS) and minimal change disease (MCD). Pathogenic mechanisms are diverse and incompletely understood, partially overlap between FSGS and MCD, and are not reflected by kidney biopsy. In order to optimize the current variable response to treatment, personalized management should rely on pathogenesis. One promising approach involves identifying biomarkers associated with specific pathogenic pathways. With the advancement of technology, proteomic studies could be a valuable tool to improve knowledge in this area and define valid biomarkers, as they have in other areas of glomerular disease. This work attempts to cover and discuss the main mechanisms of podocyte injury, followed by a review of the recent literature on proteomic biomarker studies in podocytopathies. Most of these studies have been conducted on biofluids, while tissue proteomic studies applied to podocytopathies remain limited. While we recognize the importance of non-invasive biofluid biomarkers, we propose a sequential approach for their development: tissue proteomics could first identify proteins with increased expression that may reflect underlying disease mechanisms; subsequently, the validation of these proteins in urine or plasma could pave the way to a diagnostic and prognostic biomarker-based approach.
Collapse
Affiliation(s)
- Yuriy Maslyennikov
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Andrada Alina Bărar
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Crina Claudia Rusu
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Alina Ramona Potra
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Dacian Tirinescu
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Maria Ticala
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Alexandra Urs
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Ioana Ecaterina Pralea
- Department of Personalized Medicine and Rare Diseases, MedFuture—Research Centre for Biomedical Research, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.E.P.); (C.A.I.)
| | - Cristina Adela Iuga
- Department of Personalized Medicine and Rare Diseases, MedFuture—Research Centre for Biomedical Research, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.E.P.); (C.A.I.)
- Department of Drug Analysis, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Diana Tania Moldovan
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| | - Ina Maria Kacso
- Department of Nephrology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (Y.M.); (A.A.B.); (C.C.R.); (A.R.P.); (D.T.); (M.T.); (A.U.); (I.M.K.)
| |
Collapse
|
5
|
Zununi Vahed S, Hosseiniyan Khatibi SM, Ardalan M. Canonical effects of cytokines on glomerulonephritis: A new outlook in nephrology. Med Res Rev 2025; 45:144-163. [PMID: 39164945 DOI: 10.1002/med.22074] [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: 11/08/2021] [Revised: 08/28/2022] [Accepted: 08/04/2024] [Indexed: 08/22/2024]
Abstract
Glomerulonephritis (GN) is an important cause of renal inflammation resulting from kidney-targeted adaptive and innate immune responses and consequent glomerular damage. Given the lack of autoantibodies, immune complexes, or the infiltrating immune cells in some forms of GN, for example, focal segmental glomerulosclerosis and minimal change disease, along with paraneoplastic syndrome and a special form of renal involvement in some viral infections, the likeliest causative scenario would be secreted factors, mainly cytokine(s). Since cytokines can modulate the inflammatory mechanisms, severity, and clinical outcomes of GN, it is rational to consider the umbrella term of cytokine GN as a new outlook to reclassify a group of previously known GN. We focus here, particularly, on cytokines that have the central "canonical effect" in the development of GN.
Collapse
|
6
|
Whittall Garcia LP, Gladman DD, Urowitz M, Bonilla D, Schneider R, Touma Z, Wither J. Interferon-α as a biomarker to predict renal outcomes in lupus nephritis. Lupus Sci Med 2024; 11:e001347. [PMID: 39613334 DOI: 10.1136/lupus-2024-001347] [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: 08/13/2024] [Accepted: 10/29/2024] [Indexed: 12/01/2024]
Abstract
OBJECTIVE To determine if serum interferon (IFN)-α levels at the time of a lupus nephritis (LN) flare are associated with renal outcomes. METHODS Patients with an LN flare who had a preflare estimated glomerular filtration rate (eGFR) ≥60 mL/min were included in the study. The following outcomes were ascertained: (1) Time to first and second LN flares during follow-up, (2) Time to a sustained decline in eGFR by 30% and 50%, and progression to end-stage renal disease (ESRD, <15 mL/min), and (3) Time to an adverse renal event (≥2 renal flares and/or at least a 30% sustained decline in eGFR during follow-up). Serum IFN-α was measured by Simoa. RESULTS 92 patients with active LN were included in the study. Elevated serum baseline levels of IFN-α predicted poor renal outcomes. Patients with higher baseline IFN-α had a greater risk of having two or more subsequent LN flares (HR: 1.31 (1.08-1.59), p=0.006), sustained 30% decline in eGFR (HR: 1.27 (1.14-1.40), p<0.001), 50% decline in eGFR (HR: 1.27 (1.12-1.33), p<0.001) and progressing to ESRD (HR: 1.29 (1.14-1.47), p<0.001). Receiver operating characteristic analysis identified an IFN-α cut-off, 0.6 pg/ml, for predicting an adverse renal event. CONCLUSIONS Elevated serum IFN-α levels measured at the time of an LN flare are associated with poor renal outcomes, including the development of ≥2 LN flares, and a clinically meaningful decline in kidney function.
Collapse
Affiliation(s)
| | - Dafna D Gladman
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- University of Toronto Lupus Program, Centre for Prognosis Studies in the Rheumatic Diseases, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Rheumatology, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Murray Urowitz
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- University of Toronto Lupus Program, Centre for Prognosis Studies in the Rheumatic Diseases, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Rheumatology, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Dennisse Bonilla
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- University of Toronto Lupus Program, Centre for Prognosis Studies in the Rheumatic Diseases, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Raphael Schneider
- Department of Medicine, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Zahi Touma
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- University of Toronto Lupus Program, Centre for Prognosis Studies in the Rheumatic Diseases, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Rheumatology, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Joan Wither
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Rheumatology, Schroeder Arthritis Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Yu P, Jin X, Huang W, Wang J, Zhang S, Ren L, Zhang H, Shi S. Characterization of immortalized human podocytes infected with lentivirus as an in vitro model of viral infection-associated podocytopathy. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2024; 13:204-214. [PMID: 39583339 PMCID: PMC11578807 DOI: 10.62347/bbcx1142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/25/2024] [Indexed: 11/26/2024]
Abstract
A large number of studies have shown the association of kidney disease with viral infections in the body. Viral infections cause kidney injury in two manners, the systemic inflammation (cytokine storm) and the direct infection of kidney cells. Concerning direct viral infection of podocytes, the mechanism underlying virus-induced podocyte injury remains largely unknown and requires effective experimental models to facilitate its study. Here, we performed molecular characterization of immortalized human podocyte cell line (HPC) infected with lentivirus by RNA-seq. Bioinformatics analysis revealed a strong innate immune response in the cells, including interferon production and signaling. Meanwhile, activations of ferroptosis pathway and TNF-alpha signaling were also found, consistent with an impaired viability of the cells. Lentiviral infection also upregulated expression of APOL1 as observed in patients with HIV associated nephropathy (HIVAN) and diabetic nephropathy (DN). Interestingly, when the lentiviral infected cells were treated with Adriamycin (ADR), the ADR-associated signaling pathways were not interfered and remained activated as that in the cells treated with ADR only, suggesting that the virus and ADR have distinct mechanisms in damaging podocytes. Thus, the lentivirus-infected HPC cells represent a useful in vitro model of viral infection-associated podocytopathy.
Collapse
Affiliation(s)
- Peng Yu
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Xi Jin
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Weijun Huang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Jingjing Wang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Sipang Zhang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Lu Ren
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Haitao Zhang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Shaolin Shi
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| |
Collapse
|
8
|
Lai B, Luo SF, Lai JH. Therapeutically targeting proinflammatory type I interferons in systemic lupus erythematosus: efficacy and insufficiency with a specific focus on lupus nephritis. Front Immunol 2024; 15:1489205. [PMID: 39478861 PMCID: PMC11521836 DOI: 10.3389/fimmu.2024.1489205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Type I interferons (IFN-Is) are important players in the immunopathogenesis of systemic lupus erythematosus (SLE). Pathogenic events in patients with SLE are potent triggers of IFN-I induction, yet IFN-I may induce or initiate the immunopathogenesis leading to these events. Because blocking IFN-I is effective in some clinical manifestations of SLE patients, concerns about the efficacy of anti-IFN-I therapy in patients with lupus nephritis remain. Tissues from kidney biopsies of patients with lupus nephritis revealed infiltration of various immune cells and activation of inflammatory signals; however, their correlation with renal damage is not clear, which raises serious concerns about how critical the role of IFN-I is among the potential contributors to the pathogenesis of lupus nephritis. This review addresses several issues related to the roles of IFN-I in SLE, especially in lupus nephritis, including (1) the contribution of IFN-I to the development and immunopathogenesis of SLE; (2) evidence supporting the association of IFN-I with lupus nephritis; (3) therapies targeting IFN-I and IFN-I downstream signaling molecules in SLE and lupus nephritis; (4) findings challenging the therapeutic benefits of anti-IFN-I in lupus nephritis; and (5) a perspective associated with anti-IFN-I biologics for lupus nephritis treatment. In addition to providing clear pictures of the roles of IFN-I in SLE, especially in lupus nephritis, this review addresses the lately published observations and clinical trials on this topic.
Collapse
Affiliation(s)
- Benjamin Lai
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shue-Fen Luo
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Jenn-Haung Lai
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
9
|
Suresh V, Stillman IE, Campbell KN, Meliambro K. Focal Segmental Glomerulosclerosis. ADVANCES IN KIDNEY DISEASE AND HEALTH 2024; 31:275-289. [PMID: 39084753 DOI: 10.1053/j.akdh.2024.03.009] [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: 07/22/2023] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 08/02/2024]
Abstract
Focal segmental glomerular sclerosis (FSGS) is a histological lesion characterized by sclerosis in sections (segmental) of some glomeruli (focal) in association with podocyte injury. Historically, FSGS has often been characterized as a disease, but it is a heterogeneous entity based on etiology, clinical course, and therapeutic approach. A unifying feature is podocyte injury and loss, which can be primary or the result of secondary maladaptive responses to glomerular stressors. FSGS has been demonstrated over time to carry a large health burden and remains a leading glomerular cause of ESRD globally. Recent clinical practice guidelines highlight the unmet scientific need for better understanding of disease pathogenesis, particularly for immunologic etiologies, as well as more targeted therapeutic drug development. In this review, we will discuss the current FSGS classification scheme, pathophysiologic mechanisms of injury, and treatment guidelines, along with emerging and investigational therapeutics.
Collapse
Affiliation(s)
- Varsha Suresh
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Isaac E Stillman
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kirk N Campbell
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Kristin Meliambro
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
10
|
Juliar BA, Stanaway IB, Sano F, Fu H, Smith KD, Akilesh S, Scales SJ, El Saghir J, Bhatraju PK, Liu E, Yang J, Lin J, Eddy S, Kretzler M, Zheng Y, Himmelfarb J, Harder JL, Freedman BS. Interferon-γ induces combined pyroptotic angiopathy and APOL1 expression in human kidney disease. Cell Rep 2024; 43:114310. [PMID: 38838223 PMCID: PMC11216883 DOI: 10.1016/j.celrep.2024.114310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 03/18/2024] [Accepted: 05/16/2024] [Indexed: 06/07/2024] Open
Abstract
Elevated interferon (IFN) signaling is associated with kidney diseases including COVID-19, HIV, and apolipoprotein-L1 (APOL1) nephropathy, but whether IFNs directly contribute to nephrotoxicity remains unclear. Using human kidney organoids, primary endothelial cells, and patient samples, we demonstrate that IFN-γ induces pyroptotic angiopathy in combination with APOL1 expression. Single-cell RNA sequencing, immunoblotting, and quantitative fluorescence-based assays reveal that IFN-γ-mediated expression of APOL1 is accompanied by pyroptotic endothelial network degradation in organoids. Pharmacological blockade of IFN-γ signaling inhibits APOL1 expression, prevents upregulation of pyroptosis-associated genes, and rescues vascular networks. Multiomic analyses in patients with COVID-19, proteinuric kidney disease, and collapsing glomerulopathy similarly demonstrate increased IFN signaling and pyroptosis-associated gene expression correlating with accelerated renal disease progression. Our results reveal that IFN-γ signaling simultaneously induces endothelial injury and primes renal cells for pyroptosis, suggesting a combinatorial mechanism for APOL1-mediated collapsing glomerulopathy, which can be targeted therapeutically.
Collapse
Affiliation(s)
- Benjamin A Juliar
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Ian B Stanaway
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Fumika Sano
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Hongxia Fu
- Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Division of Hematology, Department of Medicine, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington School of Medicine, Seattle, WA 98109, USA; Bloodworks Northwest Research Institute, Seattle, WA 98102, USA; Plurexa, Seattle, WA 98109, USA
| | - Kelly D Smith
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Shreeram Akilesh
- Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Suzie J Scales
- Department of Immunology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jamal El Saghir
- Division of Nephrology, Department of Internal Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Pavan K Bhatraju
- Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Esther Liu
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Johnson Yang
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jennie Lin
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Sean Eddy
- Division of Nephrology, Department of Internal Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ying Zheng
- Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Jonathan Himmelfarb
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Jennifer L Harder
- Division of Nephrology, Department of Internal Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Benjamin S Freedman
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington School of Medicine, Seattle, WA 98109, USA; Plurexa, Seattle, WA 98109, USA.
| |
Collapse
|
11
|
Horisberger A, Griffith A, Keegan J, Arazi A, Pulford J, Murzin E, Howard K, Hancock B, Fava A, Sasaki T, Ghosh T, Inamo J, Beuschel R, Cao Y, Preisinger K, Gutierrez-Arcelus M, Eisenhaure TM, Guthridge J, Hoover PJ, Dall'Era M, Wofsy D, Kamen DL, Kalunian KC, Furie R, Belmont M, Izmirly P, Clancy R, Hildeman D, Woodle ES, Apruzzese W, McMahon MA, Grossman J, Barnas JL, Payan-Schober F, Ishimori M, Weisman M, Kretzler M, Berthier CC, Hodgin JB, Demeke DS, Putterman C, Brenner MB, Anolik JH, Raychaudhuri S, Hacohen N, James JA, Davidson A, Petri MA, Buyon JP, Diamond B, Zhang F, Lederer JA, Rao DA. Blood immunophenotyping identifies distinct kidney histopathology and outcomes in patients with lupus nephritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.14.575609. [PMID: 38293222 PMCID: PMC10827101 DOI: 10.1101/2024.01.14.575609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Lupus nephritis (LN) is a frequent manifestation of systemic lupus erythematosus, and fewer than half of patients achieve complete renal response with standard immunosuppressants. Identifying non-invasive, blood-based pathologic immune alterations associated with renal injury could aid therapeutic decisions. Here, we used mass cytometry immunophenotyping of peripheral blood mononuclear cells in 145 patients with biopsy-proven LN and 40 healthy controls to evaluate the heterogeneity of immune activation in patients with LN and to identify correlates of renal parameters and treatment response. Unbiased analysis identified 3 immunologically distinct groups of patients with LN that were associated with different patterns of histopathology, renal cell infiltrates, urine proteomic profiles, and treatment response at one year. Patients with enriched circulating granzyme B+ T cells at baseline showed more severe disease and increased numbers of activated CD8 T cells in the kidney, yet they had the highest likelihood of treatment response. A second group characterized primarily by a high type I interferon signature had a lower likelihood of response to therapy, while a third group appeared immunologically inactive by immunophenotyping at enrollment but with chronic renal injuries. Main immune profiles could be distilled down to 5 simple cytometric parameters that recapitulate several of the associations, highlighting the potential for blood immune profiling to translate to clinically useful non-invasive metrics to assess immune-mediated disease in LN.
Collapse
|
12
|
Jimenez-Uribe AP, Mangos S, Hahm E. Type I IFN in Glomerular Disease: Scarring beyond the STING. Int J Mol Sci 2024; 25:2497. [PMID: 38473743 PMCID: PMC10931919 DOI: 10.3390/ijms25052497] [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: 12/31/2023] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
The field of nephrology has recently directed a considerable amount of attention towards the stimulator of interferon genes (STING) molecule since it appears to be a potent driver of chronic kidney disease (CKD). STING and its activator, the cyclic GMP-AMP synthase (cGAS), along with intracellular RIG-like receptors (RLRs) and toll-like receptors (TLRs), are potent inducers of type I interferon (IFN-I) expression. These cytokines have been long recognized as part of the mechanism used by the innate immune system to battle viral infections; however, their involvement in sterile inflammation remains unclear. Mounting evidence pointing to the involvement of the IFN-I pathway in sterile kidney inflammation provides potential insights into the complex interplay between the innate immune system and damage to the most sensitive segment of the nephron, the glomerulus. The STING pathway is often cited as one cause of renal disease not attributed to viral infections. Instead, this pathway can recognize and signal in response to host-derived nucleic acids, which are also recognized by RLRs and TLRs. It is still unclear, however, whether the development of renal diseases depends on subsequent IFN-I induction or other processes involved. This review aims to explore the main endogenous inducers of IFN-I in glomerular cells, to discuss what effects autocrine and paracrine signaling have on IFN-I induction, and to identify the pathways that are implicated in the development of glomerular damage.
Collapse
Affiliation(s)
| | | | - Eunsil Hahm
- Department of Internal Medicine, Division of Nephrology, Rush University Medical Center, Chicago, IL 60612, USA; (A.P.J.-U.); (S.M.)
| |
Collapse
|
13
|
Manoharan J, Rana R, Kuenze G, Gupta D, Elwakiel A, Ambreen S, Wang H, Banerjee K, Zimmermann S, Singh K, Gupta A, Fatima S, Kretschmer S, Schaefer L, Zeng-Brouwers J, Schwab C, Al-Dabet MM, Gadi I, Altmann H, Koch T, Poitz DM, Baber R, Kohli S, Shahzad K, Geffers R, Lee-Kirsch MA, Kalinke U, Meiler J, Mackman N, Isermann B. Tissue factor binds to and inhibits interferon-α receptor 1 signaling. Immunity 2024; 57:68-85.e11. [PMID: 38141610 DOI: 10.1016/j.immuni.2023.11.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 08/02/2023] [Accepted: 11/28/2023] [Indexed: 12/25/2023]
Abstract
Tissue factor (TF), which is a member of the cytokine receptor family, promotes coagulation and coagulation-dependent inflammation. TF also exerts protective effects through unknown mechanisms. Here, we showed that TF bound to interferon-α receptor 1 (IFNAR1) and antagonized its signaling, preventing spontaneous sterile inflammation and maintaining immune homeostasis. Structural modeling and direct binding studies revealed binding of the TF C-terminal fibronectin III domain to IFNAR1, which restricted the expression of interferon-stimulated genes (ISGs). Podocyte-specific loss of TF in mice (PodΔF3) resulted in sterile renal inflammation, characterized by JAK/STAT signaling, proinflammatory cytokine expression, disrupted immune homeostasis, and glomerulopathy. Inhibiting IFNAR1 signaling or loss of Ifnar1 expression in podocytes attenuated these effects in PodΔF3 mice. As a heteromer, TF and IFNAR1 were both inactive, while dissociation of the TF-IFNAR1 heteromer promoted TF activity and IFNAR1 signaling. These data suggest that the TF-IFNAR1 heteromer is a molecular switch that controls thrombo-inflammation.
Collapse
Affiliation(s)
- Jayakumar Manoharan
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Rajiv Rana
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Georg Kuenze
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - Dheerendra Gupta
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Ahmed Elwakiel
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Saira Ambreen
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Hongjie Wang
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kuheli Banerjee
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Silke Zimmermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Kunal Singh
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Anubhuti Gupta
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Sameen Fatima
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Stefanie Kretschmer
- Department of Pediatrics, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt am Main, Germany
| | - Jinyang Zeng-Brouwers
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt am Main, Germany
| | - Constantin Schwab
- Tissue Bank of the National Center for Tumor Diseases, Heidelberg, Germany
| | - Moh'd Mohanad Al-Dabet
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Ihsan Gadi
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Heidi Altmann
- Dresden Integrated Liquid Biobank, Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Thea Koch
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - David M Poitz
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ronny Baber
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany; Leipzig Medical Biobank, Leipzig University, Leipzig, Germany
| | - Shrey Kohli
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Khurrum Shahzad
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| | - Robert Geffers
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Min Ae Lee-Kirsch
- Department of Pediatrics, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, 30625 Hannover, Germany
| | - Jens Meiler
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany.
| |
Collapse
|
14
|
Liu J, Chen L, Li G, Tian Y, Zeng T, Xu W, Xu Q, Lu L, Gu T. Immunopromoter improves liver apoptosis and immune response in Shaoxing ducklings. Anim Biotechnol 2023; 34:4667-4674. [PMID: 36861935 DOI: 10.1080/10495398.2023.2183215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Antibiotics as feed additives, play a vital role in animal husbandry. However, overused antibiotics could cause endogenous infections in animals, and even endanger human health through the food chain. And immunopotentiators can make the low immune function improve and accelerate the induction of immune response. The aim of this study was to investigate the effects of five different immunopotentiators on the expression of liver apoptosis and immune factor related genes in Shaoxing ducklings (Anas Platyrhynchos). A total of 150 one-day-old Shaoxing ducklings were randomly divided into six groups including saline, chlorogenic acid, β-D-glucan, astragalus flavone, CpG DNA and chicken IgG, which were injected subcutaneously into the neck, respectively. At 18 days old, the liver tissues were collected to detecte the mRNA and protein expression levels of inflammatory and apoptosis-related genes. The results showed that compared with the control group, the mRNA and protein levels of liver Bcl2 with chlorogenic acid, β-D-glucan, astragalus flavone, CpG-DNA and chicken IgG were significantly decreased (p < 0.05), while the expression level of Caspase3 was up-regulated in some different degrees. In addition,The expression levels of liver iNOS and COX2 were significantly increased after the injection of five immunopotentiators (p < 0.05), and the mRNA levels of IFN-α, IFN-β, IL-1β, RIG-I, TLR3 and TLR7 genes were also significantly up-regulated compared with the control group (p < 0.05). In conclusion, chlorogenic acid, β-D-glucan, astragalus flavone, CpG-DNA and chicken IgG can be used as immunopotentiators to regulate duck innate immunity. This study provides a new way to prevent important infectious diseases of ducks, and also provides a certain reference for the application of antibiotic substitutes in animal production.
Collapse
Affiliation(s)
- Jinyu Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Li Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Guoqin Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Tao Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Wenwu Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Qi Xu
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, China
| | - Lizhi Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Tiantian Gu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| |
Collapse
|
15
|
Shankland SJ, Jefferson JA, Wessely O. Repurposing drugs for diseases associated with podocyte dysfunction. Kidney Int 2023; 104:455-462. [PMID: 37290603 PMCID: PMC11088848 DOI: 10.1016/j.kint.2023.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/02/2023] [Accepted: 05/11/2023] [Indexed: 06/10/2023]
Abstract
The majority of podocyte disorders are progressive in nature leading to chronic kidney disease and often kidney failure. The scope of current therapies is typically nonspecific immunosuppressant medications, which are accompanied by unwanted and serious side effects. However, many exciting clinical trials are underway to reduce the burden of podocyte diseases in our patients. Major advances and discoveries have recently been made experimentally in our understanding of the molecular and cellular mechanisms underlying podocyte injury in disease. This begs the question of how best to take advantage of these impressive strides. One approach to consider is the repurposing of therapeutics that have already been approved by the Food and Drug Administration, European Medicines Agency, and other regulatory agencies for indications beyond the kidney. The advantages of therapy repurposing include known safety profiles, drug development that has already been completed, and overall reduced costs for studying alternative indications for selected therapies. The purpose of this mini review is to examine the experimental literature of podocyte damage and determine if there are mechanistic targets in which prior approved therapies can be considered for repurposing to podocyte disorders.
Collapse
Affiliation(s)
- Stuart J Shankland
- Division of Nephrology, University of Washington, Seattle, Washington, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington, USA.
| | - J Ashley Jefferson
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Oliver Wessely
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA.
| |
Collapse
|
16
|
Li H, Li M, Liu C, He P, Dong A, Dong S, Zhang M. Causal effects of systemic inflammatory regulators on chronic kidney diseases and renal function: a bidirectional Mendelian randomization study. Front Immunol 2023; 14:1229636. [PMID: 37711613 PMCID: PMC10498994 DOI: 10.3389/fimmu.2023.1229636] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Background While targeted systemic inflammatory modulators show promise in preventing chronic kidney disease (CKD) progression, the causal link between specific inflammatory factors and CKD remains uncertain. Methods Using a genome-wide association study of 41 serum cytokines from 8,293 Finnish individuals, we conducted a bidirectional two-sample Mendelian randomization (MR) analysis. In addition, we genetically predicted causal associations between inflammatory factors and 5 phenotypes, including CKD, estimated glomerular filtration rate (eGFR), dialysis, rapid progression of CKD, and rapid decline in eGFR. Inverse variance weighting (IVW) served as the primary MR method, while MR-Egger, weighted median, and MR-pleiotropy residual sum and outlier (MR-PRESSO) were utilized for sensitivity analysis. Cochrane's Q test for heterogeneity. Leave-one-out method ensured stability of MR results, and Bonferroni correction assessed causal relationship strength. Results Seventeen cytokines were associated with diverse renal outcomes. Among them, after Bonferroni correction test, higher tumor necrosis factor alpha levels were associated with a rapid decrease in eGFR (OR = 1.064, 95% CI 1.028 - 1.103, P = 0.001), higher interleukin-4 levels were associated with an increase in eGFR (β = 0.003, 95% CI 0.001 - 0.005, P = 0.002), and higher growth regulated oncogene alpha (GROα) levels were associated with an increased risk of CKD (OR=1.035, 95% CI 1.012 - 1.058, P = 0.003). In contrast, genetic susceptibility to CKD was associated with an increase in GROa, and a decrease in eGFR may lead to an increase in stem cell factor. We did not find the presence of horizontal pleiotropy during the analysis. Conclusion We discovered causally related inflammatory factors that contribute to the initiation and progression of CKD at the genetic prediction level.
Collapse
Affiliation(s)
- Hongdian Li
- Department of Nephrology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingxuan Li
- Department of Cardiology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Cong Liu
- Department of Nephrology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Pengfei He
- Department of Nephrology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ao Dong
- Department of Nephrology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shaoning Dong
- Department of Nephrology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Mianzhi Zhang
- Department of Nephrology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
- Department of Nephrology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| |
Collapse
|
17
|
Anders HJ, Kitching AR, Leung N, Romagnani P. Glomerulonephritis: immunopathogenesis and immunotherapy. Nat Rev Immunol 2023; 23:453-471. [PMID: 36635359 PMCID: PMC9838307 DOI: 10.1038/s41577-022-00816-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2022] [Indexed: 01/14/2023]
Abstract
'Glomerulonephritis' (GN) is a term used to describe a group of heterogeneous immune-mediated disorders characterized by inflammation of the filtration units of the kidney (the glomeruli). These disorders are currently classified largely on the basis of histopathological lesion patterns, but these patterns do not align well with their diverse pathological mechanisms and hence do not inform optimal therapy. Instead, we propose grouping GN disorders into five categories according to their immunopathogenesis: infection-related GN, autoimmune GN, alloimmune GN, autoinflammatory GN and monoclonal gammopathy-related GN. This categorization can inform the appropriate treatment; for example, infection control for infection-related GN, suppression of adaptive immunity for autoimmune GN and alloimmune GN, inhibition of single cytokines or complement factors for autoinflammatory GN arising from inborn errors in innate immunity, and plasma cell clone-directed or B cell clone-directed therapy for monoclonal gammopathies. Here we present the immunopathogenesis of GN and immunotherapies in use and in development and discuss how an immunopathogenesis-based GN classification can focus research, and improve patient management and teaching.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilian University Munich, Munich, Germany.
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
- Department of Paediatric Nephrology, Monash Health, Clayton, VIC, Australia
| | - Nelson Leung
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Paola Romagnani
- Department of Experimental and Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| |
Collapse
|
18
|
La Bella S, Di Ludovico A, Di Donato G, Scorrano G, Chiarelli F, Vivarelli M, Breda L. Renal involvement in monogenic autoinflammatory diseases: A narrative review. Nephrology (Carlton) 2023. [PMID: 37142240 DOI: 10.1111/nep.14166] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/06/2023]
Abstract
Autoinflammatory diseases (AIDs) are mostly caused by dysfunctions in single genes encoding for proteins with a prominent role in the regulation of innate immunity, such as complement factors, inflammasome components, tumour necrosis factor (TNF)-α, and proteins belonging to type I-interferon (IFN) signalling pathways. Due to the deposition of amyloid A (AA) fibrils in the glomeruli, unprovoked inflammation in AIDs frequently affects renal health. In fact, secondary AA amyloidosis is the most common form of amyloidosis in children. It is caused by the extracellular deposition of fibrillar low-molecular weight protein subunits resulting from the degradation and accumulation of serum amyloid A (SAA) in numerous tissues and organs, primarily the kidneys. The molecular mechanisms underlying AA amyloidosis in AIDs are the elevated levels of SAA, produced by the liver in response to pro-inflammatory cytokines, and a genetic predisposition due to specific SAA isoforms. Despite the prevalence of amyloid kidney disease, non-amyloid kidney diseases may also be responsible for chronic renal damage in children with AIDs, albeit with distinct characteristics. Glomerular damage can result in various forms of glomerulonephritis with distinct histologic characteristics and a different underlying pathophysiology. This review aims to describe the potential renal implications in patients with inflammasomopathies, type-I interferonopathies, and other rare AIDs in an effort to improve the clinical course and quality of life in paediatric patients with renal involvement.
Collapse
Affiliation(s)
- Saverio La Bella
- Department of Pediatrics, University of Chieti-Pescara "G. d'Annunzio", Chieti, Italy
| | - Armando Di Ludovico
- Department of Pediatrics, University of Chieti-Pescara "G. d'Annunzio", Chieti, Italy
| | - Giulia Di Donato
- Department of Pediatrics, University of Chieti-Pescara "G. d'Annunzio", Chieti, Italy
| | - Giovanna Scorrano
- Department of Pediatrics, University of Chieti-Pescara "G. d'Annunzio", Chieti, Italy
| | - Francesco Chiarelli
- Department of Pediatrics, University of Chieti-Pescara "G. d'Annunzio", Chieti, Italy
| | - Marina Vivarelli
- Division of Nephrology, Laboratory of Nephrology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Luciana Breda
- Department of Pediatrics, University of Chieti-Pescara "G. d'Annunzio", Chieti, Italy
| |
Collapse
|
19
|
Smith KD, Akilesh S. Collapsing glomerulopathy: unraveling varied pathogeneses. Curr Opin Nephrol Hypertens 2023; 32:213-222. [PMID: 36811644 DOI: 10.1097/mnh.0000000000000873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
PURPOSE OF REVIEW Collapsing glomerulopathy presents clinically with nephrotic syndrome and rapid progressive loss of kidney function. Animal models and patient studies have uncovered numerous clinical and genetic conditions associated with collapsing glomerulopathy, as well as putative mechanisms, which will be reviewed here. RECENT FINDINGS Collapsing glomerulopathy is classified pathologically as a variant of focal and segmental glomerulosclerosis (FSGS). As such, most research efforts have focused on the causative role of podocyte injury in driving the disease. However, studies have also shown that injury to the glomerular endothelium or interruption of the podocyte-glomerular endothelial cell signaling axis can also cause collapsing glomerulopathy. Furthermore, emerging technologies are now enabling exploration of diverse molecular pathways that can precipitate collapsing glomerulopathy using biopsies from patients with the disease. SUMMARY Since its original description in the 1980s, collapsing glomerulopathy has been the subject of intense study, and these efforts have uncovered numerous insights into potential disease mechanisms. Newer technologies will enable profiling of the intra-patient and inter-patient variability in collapsing glomerulopathy mechanisms directly in patient biopsies, which will improve the diagnosis and classification of collapsing glomerulopathy.
Collapse
Affiliation(s)
- Kelly D Smith
- Department of Laboratory Medicine and Pathology, University of Washington
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington
- Kidney Research Institute, Seattle, Washington, USA
| |
Collapse
|
20
|
Meng X, Gao X, Shi K, Zhao J, Zhang X, Zhou X, Liu X, Yu J. Interferon-α2b-Induced RARRES3 Upregulation Inhibits Hypertrophic Scar Fibroblasts' Proliferation and Migration Through Wnt/β-Catenin Pathway Suppression. J Interferon Cytokine Res 2023; 43:23-34. [PMID: 36520614 DOI: 10.1089/jir.2022.0183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Hypertrophic scar (HS) is a severe skin fibrotic disorder with unclear pathogenesis. Interferon-α2b (IFN-α2b) exerts inhibitory effects on HS in vivo and in vitro; however, the exact mechanism remains unclear. In this study, we aimed to evaluate the inhibitory effects of IFN-α2b on hypertrophic scar fibroblasts' (HSFs) proliferation and migration, and to further investigate the associated molecular mechanism. Cell Counting Kit-8 and CyQUANT assays were used to assess HSFs' proliferation; wound healing and Transwell assays were used to assess HSFs' migration; real-time quantitative polymerase chain reaction and Western blotting were used to detect messenger RNA and protein levels, respectively, of related genes; bioinformatics analysis was performed to predict the downstream target of IFN-α2b. Our findings are as follows: (1) IFN-α2b inhibited HSFs' proliferation and migration in a dose-dependent manner. (2) IFN-α2b inhibited HSFs' proliferation and migration by suppressing the Wnt/β-catenin pathway. (3) Retinoic-acid receptor responder 3 (RARRES3) was predicted as a functional downstream molecule of IFN-α2b, which was low in HSFs. (4) IFN-α2b inhibited HSF phenotypes and the Wnt/β-catenin pathway by upregulating RARRES3 expression. (5) RARRES3 restrained HSFs' proliferation and migration by repressing the Wnt/β-catenin pathway. In conclusion, IFN-α2b-induced RARRES3 upregulation inhibited HSFs' proliferation and migration through Wnt/β-catenin pathway suppression.
Collapse
Affiliation(s)
- Xianglong Meng
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, China
- Jilin Provincial Skin Repair and Regeneration Engineering Research Center, Jilin University, Changchun, China
| | - Xinxin Gao
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, China
- Jilin Provincial Skin Repair and Regeneration Engineering Research Center, Jilin University, Changchun, China
| | - Kai Shi
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, China
- Jilin Provincial Skin Repair and Regeneration Engineering Research Center, Jilin University, Changchun, China
| | - Jingchun Zhao
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, China
- Jilin Provincial Skin Repair and Regeneration Engineering Research Center, Jilin University, Changchun, China
| | - Xiuhang Zhang
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, China
- Jilin Provincial Skin Repair and Regeneration Engineering Research Center, Jilin University, Changchun, China
| | - Xin Zhou
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, China
- Jilin Provincial Skin Repair and Regeneration Engineering Research Center, Jilin University, Changchun, China
| | - Xianjun Liu
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Jiaao Yu
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, China
- Jilin Provincial Skin Repair and Regeneration Engineering Research Center, Jilin University, Changchun, China
| |
Collapse
|
21
|
Zheng X, Higdon L, Gaudet A, Shah M, Balistieri A, Li C, Nadai P, Palaniappan L, Yang X, Santo B, Ginley B, Wang XX, Myakala K, Nallagatla P, Levi M, Sarder P, Rosenberg A, Maltzman JS, de Freitas Caires N, Bhalla V. Endothelial Cell-Specific Molecule-1 Inhibits Albuminuria in Diabetic Mice. KIDNEY360 2022; 3:2059-2076. [PMID: 36591362 PMCID: PMC9802554 DOI: 10.34067/kid.0001712022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/19/2022] [Indexed: 01/13/2023]
Abstract
Background Diabetic kidney disease (DKD) is the most common cause of kidney failure in the world, and novel predictive biomarkers and molecular mechanisms of disease are needed. Endothelial cell-specific molecule-1 (Esm-1) is a secreted proteoglycan that attenuates inflammation. We previously identified that a glomerular deficiency of Esm-1 associates with more pronounced albuminuria and glomerular inflammation in DKD-susceptible relative to DKD-resistant mice, but its contribution to DKD remains unexplored. Methods Using hydrodynamic tail-vein injection, we overexpress Esm-1 in DKD-susceptible DBA/2 mice and delete Esm-1 in DKD-resistant C57BL/6 mice to study the contribution of Esm-1 to DKD. We analyze clinical indices of DKD, leukocyte infiltration, podocytopenia, and extracellular matrix production. We also study transcriptomic changes to assess potential mechanisms of Esm-1 in glomeruli. Results In DKD-susceptible mice, Esm-1 inversely correlates with albuminuria and glomerular leukocyte infiltration. We show that overexpression of Esm-1 reduces albuminuria and diabetes-induced podocyte injury, independent of changes in leukocyte infiltration. Using a complementary approach, we find that constitutive deletion of Esm-1 in DKD-resistant mice modestly increases the degree of diabetes-induced albuminuria versus wild-type controls. By glomerular RNAseq, we identify that Esm-1 attenuates expression of kidney disease-promoting and interferon (IFN)-related genes, including Ackr2 and Cxcl11. Conclusions We demonstrate that, in DKD-susceptible mice, Esm-1 protects against diabetes-induced albuminuria and podocytopathy, possibly through select IFN signaling. Companion studies in patients with diabetes suggest a role of Esm-1 in human DKD.
Collapse
Affiliation(s)
- Xiaoyi Zheng
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Lauren Higdon
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California
- Veterans Affairs Palo Alto Heath Care System, Palo Alto, California
| | - Alexandre Gaudet
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1019-UMR9017-Center for Infection & Immunity of Lille, Pasteur Institute of Lille, University of Lille, Lille, France
| | - Manav Shah
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Angela Balistieri
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Catherine Li
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Patricia Nadai
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1019-UMR9017-Center for Infection & Immunity of Lille, Pasteur Institute of Lille, University of Lille, Lille, France
| | - Latha Palaniappan
- Division of Primary Care and Population Health, Stanford University School of Medicine, Stanford, California
| | - Xiaoping Yang
- Division of Kidney-Urologic Pathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Briana Santo
- Department of Pathology and Anatomical Sciences, University at Buffalo–The State University of New York, Buffalo, New York
| | - Brandon Ginley
- Department of Pathology and Anatomical Sciences, University at Buffalo–The State University of New York, Buffalo, New York
| | - Xiaoxin X. Wang
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC
| | - Komuraiah Myakala
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC
| | | | - Moshe Levi
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC
| | - Pinaki Sarder
- Department of Pathology and Anatomical Sciences, University at Buffalo–The State University of New York, Buffalo, New York
| | - Avi Rosenberg
- Division of Kidney-Urologic Pathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jonathan S. Maltzman
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California
- Veterans Affairs Palo Alto Heath Care System, Palo Alto, California
| | - Nathalie de Freitas Caires
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1019-UMR9017-Center for Infection & Immunity of Lille, Pasteur Institute of Lille, University of Lille, Lille, France
- Biothelis, Lille, France
| | - Vivek Bhalla
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
22
|
Infante B, Mercuri S, Dello Strologo A, Franzin R, Catalano V, Troise D, Cataldo E, Pontrelli P, Alfieri C, Binda V, Frontini G, Netti GS, Ranieri E, Gesualdo L, Castellano G, Stallone G. Unraveling the Link between Interferon-α and Systemic Lupus Erythematosus: From the Molecular Mechanisms to Target Therapies. Int J Mol Sci 2022; 23:ijms232415998. [PMID: 36555640 PMCID: PMC9783870 DOI: 10.3390/ijms232415998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/05/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic, systemic autoimmune disease with a wide range of clinical expressions. The kidney is often affected, usually within 5 years of the onset of SLE, and lupus nephropathy (LN) carries a high risk for increased morbidity. The clinical heterogeneity of the disease is accompanied by complex disturbances affecting the immune system with inflammation and tissue damage due to loss of tolerance to nuclear antigens and the deposition of immune complexes in tissues. Several studies have reported that in human SLE, there is an important role of the Type-I-interferons (INF) system suggested by the upregulation of INF-inducible genes observed in serial gene expression microarray studies. This review aims to describe the transduction pathways of Type-I-interferons, in particular INFα, and its immune-regulatory function in the pathogenesis of SLE and, in particular, in LN. In addition, recent novelties concerning biologic therapy in LN will be discussed.
Collapse
Affiliation(s)
- Barbara Infante
- Unit of Nephology, Dialysis and Transplantation, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Silvia Mercuri
- Unit of Nephology, Dialysis and Transplantation, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Andrea Dello Strologo
- Unit of Nephology, Dialysis and Transplantation, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Valeria Catalano
- Unit of Clinical Pathology, Center for Molecular Medicine, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy
| | - Dario Troise
- Unit of Nephology, Dialysis and Transplantation, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Emanuela Cataldo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Paola Pontrelli
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Carlo Alfieri
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Valentina Binda
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, 20122 Milan, Italy
| | - Giulia Frontini
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, 20122 Milan, Italy
| | - Giuseppe Stefano Netti
- Unit of Clinical Pathology, Center for Molecular Medicine, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy
| | - Elena Ranieri
- Unit of Clinical Pathology, Center for Molecular Medicine, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Giuseppe Castellano
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-0255034551; Fax: +39-0255034550
| | - Giovanni Stallone
- Unit of Nephology, Dialysis and Transplantation, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
23
|
Córdoba-David G, García-Giménez J, Cardoso Castelo-Branco R, Carrasco S, Cannata P, Ortiz A, Ramos AM. Crosstalk between TBK1/IKKε and the type I interferon pathway contributes to tubulointerstitial inflammation and kidney tubular injury. Front Pharmacol 2022; 13:987979. [PMID: 36386242 PMCID: PMC9647636 DOI: 10.3389/fphar.2022.987979] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/22/2022] [Indexed: 09/01/2023] Open
Abstract
The type I interferon (TI-IFN) pathway regulates innate immunity, inflammation, and apoptosis during infection. However, the contribution of the TI-IFN pathway or upstream signaling pathways to tubular injury in kidney disease is poorly understood. Upon observing evidence of activation of upstream regulators of the TI-IFN pathway in a transcriptomics analysis of murine kidney tubulointerstitial injury, we have now addressed the impact of the TI-IFN and upstream signaling pathways on kidney tubulointerstitial injury. In cultured tubular cells and kidney tissue, IFNα/β binding to IFNAR activated the TI-IFN pathway and recruited antiviral interferon-stimulated genes (ISG) and NF-κB-associated proinflammatory responses. TWEAK and lipopolysaccharide (LPS) signaled through TBK1/IKKε and IRF3 to activate both ISGs and NF-κB. In addition, TWEAK recruited TLR4 to stimulate TBK1/IKKε-dependent ISG and inflammatory responses. Dual pharmacological inhibition of TBK1/IKKε with amlexanox decreased TWEAK- or LPS-induced ISG and cytokine responses, as well as cell death induced by a complex inflammatory milieu that included TWEAK. TBK1 or IRF3 siRNA prevented the TWEAK-induced ISG and inflammatory gene expression while IKKε siRNA did not. In vivo, kidney IFNAR and IFNβ were increased in murine LPS and folic acid nephrotoxicity while IFNAR was increased in human kidney biopsies with tubulointerstitial damage. Inhibition of TBK1/IKKε with amlexanox or IFNAR neutralization decreased TI-IFN pathway activation and protected from kidney injury induced by folic acid or LPS. In conclusion, TI-IFNs, TWEAK, and LPS engage interrelated proinflammatory and antiviral responses in tubular cells. Moreover, inhibition of TBK1/IKKε with amlexanox, and IFNAR targeting, may protect from tubulointerstitial kidney injury.
Collapse
Affiliation(s)
- Gina Córdoba-David
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jorge García-Giménez
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Susana Carrasco
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
- RICORS 2040, Madrid, Spain
| | - Pablo Cannata
- Department of Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
- RICORS 2040, Madrid, Spain
- Department of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Adrián M. Ramos
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
- RICORS 2040, Madrid, Spain
| |
Collapse
|
24
|
Typiak M, Audzeyenka I, Dubaniewicz A. Presence and possible impact of Fcγ receptors on resident kidney cells in health and disease. Immunol Cell Biol 2022; 100:591-604. [DOI: 10.1111/imcb.12570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/13/2022] [Accepted: 06/28/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Marlena Typiak
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute Polish Academy of Sciences Gdansk Poland
- Department of General and Medical Biochemistry, Faculty of Biology University of Gdansk Gdansk Poland
| | - Irena Audzeyenka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute Polish Academy of Sciences Gdansk Poland
- Department of Molecular Biotechnology, Faculty of Chemistry University of Gdansk Gdansk Poland
| | - Anna Dubaniewicz
- Department of Pulmonology Medical University of Gdansk Gdansk Poland
| |
Collapse
|
25
|
Lin L, Tian E, Ren J, Wu Z, Deng J, Yang J. Traditional Chinese Medicine in Treating Primary Podocytosis: From Fundamental Science to Clinical Research. Front Pharmacol 2022; 13:932739. [PMID: 36003509 PMCID: PMC9393213 DOI: 10.3389/fphar.2022.932739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022] Open
Abstract
Podocytes form a key component of the glomerular filtration barrier. Damage to podocytes is referred to as “podocyte disease.” There are many causes of podocyte injury, including primary injury, secondary injury, and gene mutations. Primary podocytosis mostly manifests as nephrotic syndrome. At present, first-line treatment is based on glucocorticoid administration combined with immunosuppressive therapy, but some patients still progress to end-stage renal disease. In Asia, especially in China, traditional Chinese medicine (TCM) still plays an important role in the treatment of kidney diseases. This study summarizes the potential mechanism of TCM and its active components in protecting podocytes, such as repairing podocyte injury, inhibiting podocyte proliferation, reducing podocyte apoptosis and excretion, maintaining podocyte skeleton structure, and upregulating podocyte-related protein expression. At the same time, the clinical efficacy of TCM in the treatment of primary podocytosis (including idiopathic membranous nephropathy, minimal change disease, and focal segmental glomerulosclerosis) is summarized to support the development of new treatment strategies for primary podocytosis.
Collapse
Affiliation(s)
- Lirong Lin
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (General Hospital), Chongqing, China
| | - En Tian
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (General Hospital), Chongqing, China
| | - Jiangwen Ren
- Department of Nephrology, Rheumatism and Immunology, Jiulongpo District People’s Hospital of Chongqing, Chongqing, China
| | - Zhifeng Wu
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (General Hospital), Chongqing, China
| | | | - Jurong Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University (General Hospital), Chongqing, China
- *Correspondence: Jurong Yang,
| |
Collapse
|
26
|
Affiliation(s)
- Stefanie Steiger
- Renal Division, Department of Medicine IV, University Hospital of the Ludwig Maximilian University, Munich, Germany
| | - Hans-Joachim Anders
- Renal Division, Department of Medicine IV, University Hospital of the Ludwig Maximilian University, Munich, Germany.
| |
Collapse
|
27
|
Eskandarian Boroujeni M, Sekrecka A, Antonczyk A, Hassani S, Sekrecki M, Nowicka H, Lopacinska N, Olya A, Kluzek K, Wesoly J, Bluyssen HAR. Dysregulated Interferon Response and Immune Hyperactivation in Severe COVID-19: Targeting STATs as a Novel Therapeutic Strategy. Front Immunol 2022; 13:888897. [PMID: 35663932 PMCID: PMC9156796 DOI: 10.3389/fimmu.2022.888897] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/13/2022] [Indexed: 01/08/2023] Open
Abstract
A disease outbreak in December 2019, caused by a novel coronavirus SARS-CoV-2, was named COVID-19. SARS-CoV-2 infects cells from the upper and lower respiratory tract system and is transmitted by inhalation or contact with infected droplets. Common clinical symptoms include fatigue, fever, and cough, but also shortness of breath and lung abnormalities. Still, some 5% of SARS-CoV-2 infections progress to severe pneumonia and acute respiratory distress syndrome (ARDS), with pulmonary edema, acute kidney injury, and/or multiple organ failure as important consequences, which can lead to death. The innate immune system recognizes viral RNAs and triggers the expression of interferons (IFN). IFNs activate anti-viral effectors and components of the adaptive immune system by activating members of the STAT and IRF families that induce the expression of IFN-stimulated genes (ISG)s. Among other coronaviruses, such as Middle East respiratory syndrome coronavirus (MERS-CoV) and SARS-CoV, common strategies have been identified to antagonize IFN signaling. This typically coincides with hyperactive inflammatory host responses known as the “cytokine storm” that mediate severe lung damage. Likewise, SARS-CoV-2 infection combines a dysregulated IFN response with excessive production of inflammatory cytokines in the lungs. This excessive inflammatory response in the lungs is associated with the local recruitment of immune cells that create a pathogenic inflammatory loop. Together, it causes severe lung pathology, including ARDS, as well as damage to other vulnerable organs, like the heart, spleen, lymph nodes, and kidney, as well as the brain. This can rapidly progress to multiple organ exhaustion and correlates with a poor prognosis in COVID-19 patients. In this review, we focus on the crucial role of different types of IFN that underlies the progression of SARS-CoV-2 infection and leads to immune cell hyper-activation in the lungs, exuberant systemic inflammation, and multiple organ damage. Consequently, to protect from systemic inflammation, it will be critical to interfere with signaling cascades activated by IFNs and other inflammatory cytokines. Targeting members of the STAT family could therefore be proposed as a novel therapeutic strategy in patients with severe COVID-19.
Collapse
Affiliation(s)
- Mahdi Eskandarian Boroujeni
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Agata Sekrecka
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Aleksandra Antonczyk
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Sanaz Hassani
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Michal Sekrecki
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Hanna Nowicka
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Natalia Lopacinska
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Arta Olya
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Katarzyna Kluzek
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Joanna Wesoly
- Laboratory of High Throughput Technologies, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Hans A R Bluyssen
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| |
Collapse
|
28
|
Lodi L, Mastrolia MV, Bello F, Rossi GM, Angelotti ML, Crow YJ, Romagnani P, Vaglio A. Type I interferon-related kidney disorders. Kidney Int 2022; 101:1142-1159. [PMID: 35339535 DOI: 10.1016/j.kint.2022.02.031] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/04/2022] [Accepted: 02/22/2022] [Indexed: 12/15/2022]
Abstract
Type I interferon (IFN-I) mediates tissue damage in a wide range of kidney disorders, directly affecting the biology and function of several renal cell types including podocytes, mesangial, endothelial and parietal epithelial cells (PECs).Enhanced IFN-I signalling is observed in the context of viral infections, autoimmunity (e.g., systemic lupus erythematosus, SLE), and the type 1 interferonopathies (T1Is), rare monogenic disorders characterised by constitutive activation of the IFN-I pathway. All of these IFN I-related disorders can cause renal dysfunction, and share pathogenic and histopathological features. Collapsing glomerulopathy, a histopathological lesion characterised by podocyte loss, collapse of the vascular tuft and PEC proliferation, is commonly associated with viral infections, has been described in T1Is such as Aicardi-Goutières syndrome and STING-associated vasculopathy with onset in infancy (SAVI), and can also be induced by recombinant IFN-therapy. In all of these conditions, podocytes and PECs seem to be the primary target of IFN I-mediated damage. Additionally, immune-mediated glomerular injury is common to viral infections, SLE, and T1Is such as COPA syndrome and DNASE1L3 deficiency, diseases in which IFN-I apparently promotes immune-mediated kidney injury. Finally, kidney pathology primarily characterised by vascular lesions (e.g., thrombotic microangiopathy, vasculitis) is a hallmark of the T1I ADA2 deficiency as well as of SLE, viral infections and IFN-therapy.Defining the nosology, pathogenic mechanisms and histopathological patterns of IFN I-related kidney disorders has diagnostic and therapeutic implications, especially considering the likely near-term availability of novel drugs targeting the IFN-I pathway.
Collapse
Affiliation(s)
- Lorenzo Lodi
- Department of Health Sciences, University of Firenze; Immunology Unit, Department of Pediatrics, Meyer Children's Hospital, Firenze, Italy
| | - Maria V Mastrolia
- Rheumatology Unit, Department of Pediatrics, Meyer Children's Hospital, Firenze, Italy
| | - Federica Bello
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | | | - Maria L Angelotti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Firenze, Firenze, Italy
| | - Yanick J Crow
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK; Laboratory of Neurogenetics and Neuroinflammation, Institut Imagine, Université de Paris, Paris, France
| | - Paola Romagnani
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Firenze, Firenze, Italy; Nephrology and Dialysis Unit, Department of Pediatrics, Meyer Children's Hospital, Firenze, Italy
| | - Augusto Vaglio
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Firenze, Firenze, Italy; Nephrology and Dialysis Unit, Department of Pediatrics, Meyer Children's Hospital, Firenze, Italy.
| |
Collapse
|
29
|
Picerno A, Castellano G, Curci C, Kopaczka K, Stasi A, Pertosa GB, Sabbà C, Gesualdo L, Gramignoli R, Sallustio F. The Icarus Flight of Perinatal Stem and Renal Progenitor Cells Within Immune System. Front Immunol 2022; 13:840146. [PMID: 35355984 PMCID: PMC8959820 DOI: 10.3389/fimmu.2022.840146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/11/2022] [Indexed: 12/02/2022] Open
Abstract
Our immune system actively fights bacteria and viruses, and it must strike a delicate balance between over- and under-reaction, just like Daedalus and Icarus in Greek mythology, who could not escape their imprisonment by flying too high or too low. Both human amniotic epithelial and mesenchymal stromal cells and the conditioned medium generated from their culture exert multiple immunosuppressive activities. They have strong immunomodulatory properties that are influenced by the types and intensity of inflammatory stimuli present in the microenvironment. Notably, very recently, the immunomodulatory activity of human adult renal stem/progenitor cells (ARPCs) has been discovered. ARPCs cause a decrease in Tregs and CD3+ CD4- CD8- (DN) T cells in the early stages of inflammation, encouraging inflammation, and an increase in the late stages of inflammation, favoring inflammation quenching. If the inflammatory trigger continues, however, ARPCs cause a further increase in DN T cells to avoid the development of a harmful inflammatory state. As in the flight of Daedalus and Icarus, who could not fly too high or too low to not destroy their wings by the heat of the sun or the humidity of the sea, in response to an inflammatory environment, stem cells seem to behave by paying attention to regulating T cells in the balance between immune tolerance and autoimmunity. Recognizing the existence of both suppressive and stimulatory properties, and the mechanisms that underpin the duality of immune reaction, will aid in the development of active immunotherapeutic approaches that manipulate the immune system to achieve therapeutic benefit.
Collapse
Affiliation(s)
- Angela Picerno
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Bari, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Renal Transplant Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Claudia Curci
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Katarzyna Kopaczka
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Alessandra Stasi
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Battista Pertosa
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Carlo Sabbà
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Bari, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari Aldo Moro, Bari, Italy
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Fabio Sallustio
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Bari, Italy
| |
Collapse
|
30
|
Modes of podocyte death in diabetic kidney disease: an update. J Nephrol 2022; 35:1571-1584. [PMID: 35201595 DOI: 10.1007/s40620-022-01269-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/01/2022] [Indexed: 02/06/2023]
Abstract
Diabetic kidney disease (DKD) accounts for a large proportion of end-stage renal diseases that require renal replacement therapies including dialysis and transplantation. Therefore, it is critical to understand the occurrence and development of DKD. Podocytes are mainly injured during the development of DKD, ultimately leading to their extensive death and loss. In turn, the injury and death of glomerular podocytes are also the main culprits of DKD. This review introduces the characteristics of podocytes and summarizes the modes of their death in DKD, including apoptosis, autophagy, mitotic catastrophe (MC), anoikis, necroptosis, and pyroptosis. Apoptosis is characterized by nuclear condensation and the formation of apoptotic bodies, and it exerts a different effect from autophagy in mediating DKD-induced podocyte loss. MC mediates a faulty mitotic process while anoikis separates podocytes from the basement membrane. Moreover, pyroptosis activates inflammatory factors to aggravate podocyte injuries whilst necroptosis drives signaling cascades, such as receptor-interacting protein kinases 1 and 3 and mixed lineage kinase domain-like, ultimately promoting the death of podocytes. In conclusion, a thorough knowledge of the modes of podocyte death in DKD can help us understand the development of DKD and lay the foundation for strategies in DKD disease therapy.
Collapse
|
31
|
Ravaglia F, Melica ME, Angelotti ML, De Chiara L, Romagnani P, Lasagni L. The Pathology Lesion Patterns of Podocytopathies: How and why? Front Cell Dev Biol 2022; 10:838272. [PMID: 35281116 PMCID: PMC8907833 DOI: 10.3389/fcell.2022.838272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Podocytopathies are a group of proteinuric glomerular disorders driven by primary podocyte injury that are associated with a set of lesion patterns observed on kidney biopsy, i.e., minimal changes, focal segmental glomerulosclerosis, diffuse mesangial sclerosis and collapsing glomerulopathy. These unspecific lesion patterns have long been considered as independent disease entities. By contrast, recent evidence from genetics and experimental studies demonstrated that they represent signs of repeated injury and repair attempts. These ongoing processes depend on the type, length, and severity of podocyte injury, as well as on the ability of parietal epithelial cells to drive repair. In this review, we discuss the main pathology patterns of podocytopathies with a focus on the cellular and molecular response of podocytes and parietal epithelial cells.
Collapse
Affiliation(s)
| | - Maria Elena Melica
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Maria Lucia Angelotti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Letizia De Chiara
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Paola Romagnani
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
- Nephrology Unit, Meyer Children’s Hospital, Florence, Italy
| | - Laura Lasagni
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| |
Collapse
|
32
|
Legrand M, Bell S, Forni L, Joannidis M, Koyner JL, Liu K, Cantaluppi V. Pathophysiology of COVID-19-associated acute kidney injury. Nat Rev Nephrol 2021; 17:751-764. [PMID: 34226718 PMCID: PMC8256398 DOI: 10.1038/s41581-021-00452-0] [Citation(s) in RCA: 291] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 02/06/2023]
Abstract
Although respiratory failure and hypoxaemia are the main manifestations of COVID-19, kidney involvement is also common. Available evidence supports a number of potential pathophysiological pathways through which acute kidney injury (AKI) can develop in the context of SARS-CoV-2 infection. Histopathological findings have highlighted both similarities and differences between AKI in patients with COVID-19 and in those with AKI in non-COVID-related sepsis. Acute tubular injury is common, although it is often mild, despite markedly reduced kidney function. Systemic haemodynamic instability very likely contributes to tubular injury. Despite descriptions of COVID-19 as a cytokine storm syndrome, levels of circulating cytokines are often lower in patients with COVID-19 than in patients with acute respiratory distress syndrome with causes other than COVID-19. Tissue inflammation and local immune cell infiltration have been repeatedly observed and might have a critical role in kidney injury, as might endothelial injury and microvascular thrombi. Findings of high viral load in patients who have died with AKI suggest a contribution of viral invasion in the kidneys, although the issue of renal tropism remains controversial. An impaired type I interferon response has also been reported in patients with severe COVID-19. In light of these observations, the potential pathophysiological mechanisms of COVID-19-associated AKI may provide insights into therapeutic strategies.
Collapse
Affiliation(s)
- Matthieu Legrand
- Department of Anesthesia and Perioperative Care, Division of Critical Care Medicine, University of California, San Francisco, CA, USA.
- Investigation Network Initiative-Cardiovascular and Renal Clinical Trialists network, Nancy, France.
| | - Samira Bell
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - Lui Forni
- Intensive Care Unit, Royal Surrey Hospital NHS Foundation Trust, Surrey, UK
- Department of Clinical and Experimental Medicine, Faculty of Health Sciences, University of Surrey, Surrey, UK
| | - Michael Joannidis
- Division of Intensive Care and Emergency Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Jay L Koyner
- Divisions of Nephrology, Departments of Medicine, University of Chicago, Chicago, IL, USA
| | - Kathleen Liu
- Divisions of Nephrology and Critical Care Medicine, Departments of Medicine and Anesthesia, University of San Francisco, San Francisco, CA, USA
| | - Vincenzo Cantaluppi
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
33
|
Ni L, Yuan C, Wu X. The recruitment mechanisms and potential therapeutic targets of podocytes from parietal epithelial cells. J Transl Med 2021; 19:441. [PMID: 34674704 PMCID: PMC8529729 DOI: 10.1186/s12967-021-03101-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/01/2021] [Indexed: 01/02/2023] Open
Abstract
Podocytes are differentiated postmitotic cells which cannot be replaced after podocyte injury. The mechanism of podocyte repopulation after injury has aroused wide concern. Parietal epithelial cells (PECs) are heterogeneous and only a specific subpopulation of PECs has the capacity to replace podocytes. Major progress has been achieved in recent years regarding the role and function of a subset of PECs which could transdifferentiate toward podocytes. Additionally, several factors, such as Notch, Wnt/ß-catenin, Wilms’ tumor-1, miR-193a and growth arrest-specific protein 1, have been shown to be involved in these processes. Finally, PECs serve as a potential therapeutic target in the conditions of podocyte loss. In this review, we discuss the latest observations and concepts about the recruitment of podocytes from PECs in glomerular diseases as well as newly identified mechanisms and the most recent treatments for this process.
Collapse
Affiliation(s)
- Lihua Ni
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Cheng Yuan
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China.
| | - Xiaoyan Wu
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China.
| |
Collapse
|
34
|
Lee J, Park Y, Jang SG, Hong SM, Song YS, Kim MJ, Baek S, Park SH, Kwok SK. Baricitinib Attenuates Autoimmune Phenotype and Podocyte Injury in a Murine Model of Systemic Lupus Erythematosus. Front Immunol 2021; 12:704526. [PMID: 34497607 PMCID: PMC8419414 DOI: 10.3389/fimmu.2021.704526] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/04/2021] [Indexed: 11/29/2022] Open
Abstract
Objective Baricitinib, a selective inhibitor for janus kinase (JAK) 1 and JAK2, is approved for use in rheumatoid arthritis. Systemic lupus erythematosus (SLE) is recently regarded as a potential candidate targeted by JAK inhibitors because of the relationship between its pathogenesis and JAK/signal transducer and activator of transcription (STAT) pathway-mediated cytokines such as type I interferons. The objective of this study was to determine whether baricitinib could effectively ameliorate SLE using a murine model Methods To investigate effects of baricitinib on various autoimmune features, especially renal involvements in SLE, eight-week-old MRL/Mp-Faslpr (MRL/lpr) mice were used as a lupus-prone animal model and treated with baricitinib for eight weeks. Immortalized podocytes and primary podocytes and B cells isolated from C57BL/6 mice were used to determine the in vitro efficacy of baricitinib. Results Baricitinib remarkably suppressed lupus-like phenotypes of MRL/lpr mice, such as splenomegaly, lymphadenopathy, proteinuria, and systemic autoimmunity including circulating autoantibodies and pro-inflammatory cytokines. It also modulated immune cell populations and effectively ameliorated renal inflammation, leading to the recovery of the expression of structural proteins in podocytes. According to in vitro experiments, baricitinib treatment could mitigate B cell differentiation and restore disrupted cytoskeletal structures of podocytes under inflammatory stimulation by blocking the JAK/STAT pathway. Conclusions The present study demonstrated that baricitinib could effectively attenuate autoimmune features including renal inflammation of lupus-prone mice by suppressing aberrant B cell activation and podocyte abnormalities. Thus, baricitinib as a selective JAK inhibitor could be a promising therapeutic candidate in the treatment of SLE.
Collapse
Affiliation(s)
- Jaeseon Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Youngjae Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Se Gwang Jang
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seung-Min Hong
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Young-Seok Song
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Min-Jun Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - SeungYe Baek
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seung-Ki Kwok
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
35
|
Abstract
Skewing of type I interferon (IFN) production and responses is a hallmark of systemic lupus erythematosus (SLE). Genetic and environmental contributions to IFN production lead to aberrant innate and adaptive immune activation even before clinical development of disease. Basic and translational research in this arena continues to identify contributions of IFNs to disease pathogenesis, and several promising therapeutic options for targeting of type I IFNs and their signaling pathways are in development for treatment of SLE patients.
Collapse
Affiliation(s)
- Sirisha Sirobhushanam
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, 5568 MSRB 2, 1150 West Medical Center Drive, Ann Arbor, MI 49109, USA
| | - Stephanie Lazar
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, 5568 MSRB 2, 1150 West Medical Center Drive, Ann Arbor, MI 49109, USA
| | - J Michelle Kahlenberg
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, 5570A MSRB 2, 1150 West Medical Center Drive, Ann Arbor, MI 49109, USA; Department of Dermatology, University of Michigan, 5570A MSRB 2, 1150 West Medical Center Drive, Ann Arbor, MI 49109, USA.
| |
Collapse
|
36
|
Xia X, Wang X, Wang H, Lin Z, Shao K, Xu J, Zhao Y. Ameliorative effect of white tea from 50-year-old tree of Camellia sinensis L. (Theaceae) on kidney damage in diabetic mice via SIRT1/AMPK pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 272:113919. [PMID: 33577915 DOI: 10.1016/j.jep.2021.113919] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 01/26/2021] [Accepted: 02/06/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Diabetic kidney damage (DKD) is one of the most common complications of diabetes, which is known as a chronic inflammatory kidney disease caused by persistent hyperglycemia. White tea was originally used as a folk medicine to treat measles in ancient China. What arouses our interest is that there is a traditional method to treat diabetes with white tea taken from over 30-year-old tree of Camellia sinensis L. However, there are few reports on the renal protection of white tea. AIM OF THE STUDY This present study was designed to study the potential protective effects of white tea (WT) and old tree white tea (OTWT) on high-fat-diet (HFD) combined with streptozotocin (STZ)-induced type 2 diabetic mice to explore the possible mechanism of WT/OTWT against DKD. MATERIALS AND METHODS C57BL/6 mice were randomly divided into four groups: NC, T2D, WT (400 mg/kg·b.w, p.o.), OTWT (400 mg/kg·b.w, p.o.). Diabetes was established in all groups except NC group, by six weeks of HFD feeding combined with STZ (50 mg/kg, i.p.) for 3 times, treatments were administered for six weeks and then all the animals were decapitated; kidney tissues and blood samples were collected for the further analysis, including: levels of insulin, lipid metabolism (TG, TC, HDL, LDL, FFA), antioxidative enzymes (catalase (CAT), super oxide dismutase (SOD), glutathione peroxidase (GPx)), blood urea nitrogen (BUN) and creatine, inflammatory cytokines (TNF-α, IL-1β, COX-2, iNOS, MCP-1), advanced glycation end products (AGE), receptor of AGE (RAGE), Nrf2, AMPK, SIRT1, and PGC-1α. H&E, PAS and Masson staining were performed to examine the histopathological alterations of the kidneys. RESULTS Our data showed that WT and OTWT reversed the abnormal serum lipids (TG, TC, HDL, LDL, FFA) in T2D mice, upregulated antioxidative enzymes levels (CAT, SOD, GPx) and inhibit the excessive production of proinflammatory mediators (including MCP-1, TNF-α, IL1β, COX-2 and iNOS) by varying degrees, and OTWT was more effective. In histopathology, OTWT could significantly alleviate the accumulation of renal AGE in T2D mice, thereby improving the structural changes of the kidneys, such as glomerular hypertrophy, glomerular basement membrane thickening and kidney FIbrosis. CONCLUSIONS Both WT and OTWT could alleviate the diabetic changes in T2D mice via hypoglycemic, hypolipidemic, anti-oxidative and anti-inflammatory effects, while OTWT was more evident. OTWT could prominently alleviate the accumulation of AGE in the kidneys of T2D mice, thereby ameliorating the renal oxidative stress and inflammatory damage, which was associated with the activation of SIRT1/AMPK pathway.
Collapse
Affiliation(s)
- Xiaoyan Xia
- School of Traditional Chinese Medicine, Shanxi Datong University, Datong, 037009, China; School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Xude Wang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Hua Wang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Zhenchuan Lin
- Pinpin Tea Industry Co., Ltd., Fujian, 355200, China.
| | - Keping Shao
- Pinpin Tea Industry Co., Ltd., Fujian, 355200, China.
| | - Jing Xu
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Yuqing Zhao
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, China; Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
37
|
Iwakura T, Marschner JA, Zhao ZB, Świderska MK, Anders HJ. Electric cell-substrate impedance sensing in kidney research. Nephrol Dial Transplant 2021; 36:216-223. [PMID: 31598727 DOI: 10.1093/ndt/gfz191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/18/2019] [Indexed: 12/20/2022] Open
Abstract
Electric cell-substrate impedance sensing (ECIS) is a quantitative, label-free, non-invasive analytical method allowing continuous monitoring of the behaviour of adherent cells by online recording of transcellular impedance. ECIS offers a wide range of practical applications to study cell proliferation, migration, differentiation, toxicity and monolayer barrier integrity. All of these applications are relevant for basic kidney research, e.g. on endothelial cells, tubular and glomerular epithelial cells. This review gives an overview on the fundamental principles of the ECIS technology. We name strengths and remaining hurdles for practical applications, present an ECIS array reuse protocol, and review its past, present and potential future contributions to preclinical kidney research.
Collapse
Affiliation(s)
- Takamasa Iwakura
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany.,Internal Medicine I, Division of Nephrology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Julian A Marschner
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| | - Zhi Bo Zhao
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| | - Monika Katarzyna Świderska
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| | - Hans-Joachim Anders
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| |
Collapse
|
38
|
Ding X, Ren Y, He X. IFN-I Mediates Lupus Nephritis From the Beginning to Renal Fibrosis. Front Immunol 2021; 12:676082. [PMID: 33959133 PMCID: PMC8093624 DOI: 10.3389/fimmu.2021.676082] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Lupus nephritis (LN) is a common complication of systemic lupus erythematosus (SLE) and a major risk factor for morbidity and mortality. The abundant cell-free nucleic (DNA/RNA) in SLE patients, especially dsDNA, is a key substance in the pathogenesis of SLE and LN. The deposition of DNA/RNA-immune complexes (DNA/RNA-ICs) in the glomerulus causes a series of inflammatory reactions that lead to resident renal cell disturbance and eventually renal fibrosis. Cell-free DNA/RNA is the most effective inducer of type I interferons (IFN-I). Resident renal cells (rather than infiltrating immune cells) are the main source of IFN-I in the kidney. IFN-I in turn damages resident renal cells. Not only are resident renal cells victims, but also participants in this immunity war. However, the mechanism for generation of IFN-I in resident renal cells and the pathological mechanism of IFN-I promoting renal fibrosis have not been fully elucidated. This paper reviews the latest epidemiology of LN and its development process, discusses the mechanism for generation of IFN-I in resident renal cells and the role of IFN-I in the pathogenesis of LN, and may open a new perspective for the treatment of LN.
Collapse
Affiliation(s)
- Xuewei Ding
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Ren
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Pediatric Internal Medicine Department, Haikou Maternal and Child Health Hospital, Haikou, China
| | - Xiaojie He
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
39
|
Fenaroli P, Rossi GM, Angelotti ML, Antonelli G, Volpi S, Grossi A, Delsante M, Lodi L, Landini S, Romagnani P, Vaglio A. Collapsing Glomerulopathy as a Complication of Type I Interferon-Mediated Glomerulopathy in a Patient With RNASEH2B-Related Aicardi-Goutières Syndrome. Am J Kidney Dis 2021; 78:750-754. [PMID: 33872687 DOI: 10.1053/j.ajkd.2021.02.330] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/03/2021] [Indexed: 11/11/2022]
Abstract
Aicardi-Goutières syndrome (AGS) is a well-characterized monogenic type I interferonopathy presenting with prominent neurologic manifestations. Among extraneurologic features, renal involvement has been described in only 1 patient with an IFIH1 mutation in whom membranous nephropathy developed. The pathogenic role of augmented interferon (IFN) signaling in tissues other than the central nervous system remains to be elucidated. We report a case of collapsing glomerulopathy in a 15-year-old girl affected by AGS with RNASEH2B mutation (an alanine-to-threonine change at amino acid 177), which led to kidney failure. The patient had no lupus-like features and lacked the APOL1 G1 and G2 risk alleles. Kidney biopsy showed findings consistent with collapsing glomerulopathy. MxA, a protein involved in antiviral immunity and induced by type I IFNs, was selectively expressed in CD133-positive parietal epithelial cells (PECs) but not in podocytes that stained for synaptopodin or in other glomerular cells. MxA also colocalized within pseudocrescents with CD44, a marker of PEC activation involved in cellular proliferation, differentiation, and migration and in glomerular scarring. Our findings suggest that collapsing glomerulopathy can be a complication of the type I interferonopathy AGS and that a constitutively enhanced type I IFN response in CD133-positive PECs can drive collapsing glomerulopathy.
Collapse
Affiliation(s)
| | | | - Maria Lucia Angelotti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence
| | - Giulia Antonelli
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence
| | - Stefano Volpi
- Rheumatology Unit, G. Gaslini Institute, Genoa, Italy
| | - Alice Grossi
- Genetics and Genomics Laboratory for Rare Diseases, G. Gaslini Institute, Genoa, Italy
| | | | - Lorenzo Lodi
- Section of Pediatrics, Department of Health Sciences, Meyer Children's Hospital, University of Firenze, Florence
| | | | - Paola Romagnani
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence; Medical Genetics Unit, Meyer Children's Hospital, Florence; Nephrology and Dialysis Unit, Meyer Children's Hospital, Florence
| | - Augusto Vaglio
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence; Medical Genetics Unit, Meyer Children's Hospital, Florence; Nephrology and Dialysis Unit, Meyer Children's Hospital, Florence.
| |
Collapse
|
40
|
Petreski T, Piko N, Ekart R, Hojs R, Bevc S. Review on Inflammation Markers in Chronic Kidney Disease. Biomedicines 2021; 9:182. [PMID: 33670423 PMCID: PMC7917900 DOI: 10.3390/biomedicines9020182] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) is one of the major health problems of the modern age. It represents an important public health challenge with an ever-lasting rising prevalence, which reached almost 700 million by the year 2017. Therefore, it is very important to identify patients at risk for CKD development and discover risk factors that cause the progression of the disease. Several studies have tackled this conundrum in recent years, novel markers have been identified, and new insights into the pathogenesis of CKD have been gained. This review summarizes the evidence on markers of inflammation and their role in the development and progression of CKD. It will focus primarily on cytokines, chemokines, and cell adhesion molecules. Nevertheless, further large, multicenter studies are needed to establish the role of these markers and confirm possible treatment options in everyday clinical practice.
Collapse
Affiliation(s)
- Tadej Petreski
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia; (T.P.); (N.P.); (R.H.)
- Department of Internal Medicine and Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| | - Nejc Piko
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia; (T.P.); (N.P.); (R.H.)
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia;
| | - Robert Ekart
- Department of Internal Medicine and Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia;
| | - Radovan Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia; (T.P.); (N.P.); (R.H.)
- Department of Internal Medicine and Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia; (T.P.); (N.P.); (R.H.)
- Department of Internal Medicine and Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| |
Collapse
|
41
|
Kawaguchi T, Hasegawa K, Yasuda I, Muraoka H, Umino H, Tokuyama H, Hashiguchi A, Wakino S, Itoh H. Diabetic condition induces hypertrophy and vacuolization in glomerular parietal epithelial cells. Sci Rep 2021; 11:1515. [PMID: 33452384 PMCID: PMC7810998 DOI: 10.1038/s41598-021-81027-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 12/09/2020] [Indexed: 01/29/2023] Open
Abstract
Diabetic nephropathy (DN) is accompanied by characteristic changes in the glomerulus, but little is known about the effect of diabetes on parietal epithelial cells (PECs). In this study, a descriptive analysis of PECs was undertaken in diabetic db/db mice and in diabetic patients. PEC hypertrophy was significantly more prominent in diabetic mice than in nondiabetic mice, and this was evident even at the early stage. Additionally, the number of vacuoles in PECs was markedly increased in diabetic mice, suggesting the presence of cellular injury in PECs in DN. Although rare, binuclear cells were observed in mice with early diabetes. In cultured PECs, a high glucose condition, compared with normal glucose condition, induced cellular hypertrophy and apoptosis. Flow cytometry showed that some PECs in the G0 phase reentered the cell cycle but got arrested in the S phase. Finally, in human diabetic subjects, hypertrophy and vacuolization were observed in the PECs. Our data showed that PECs undergo substantial changes in DN and may participate in rearrangement for differentiation into podocytes.
Collapse
Affiliation(s)
- Takahisa Kawaguchi
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Kazuhiro Hasegawa
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Itaru Yasuda
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Hirokazu Muraoka
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Hiroyuki Umino
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Hirobumi Tokuyama
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Akinori Hashiguchi
- grid.26091.3c0000 0004 1936 9959Department of Pathology, School of Medicine, Keio University, Tokyo, 160-8582 Japan
| | - Shu Wakino
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Hiroshi Itoh
- grid.26091.3c0000 0004 1936 9959Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| |
Collapse
|
42
|
Molecular Mechanisms of Renal Progenitor Regulation: How Many Pieces in the Puzzle? Cells 2021; 10:cells10010059. [PMID: 33401654 PMCID: PMC7823786 DOI: 10.3390/cells10010059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/26/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
Kidneys of mice, rats and humans possess progenitors that maintain daily homeostasis and take part in endogenous regenerative processes following injury, owing to their capacity to proliferate and differentiate. In the glomerular and tubular compartments of the nephron, consistent studies demonstrated that well-characterized, distinct populations of progenitor cells, localized in the parietal epithelium of Bowman capsule and scattered in the proximal and distal tubules, could generate segment-specific cells in physiological conditions and following tissue injury. However, defective or abnormal regenerative responses of these progenitors can contribute to pathologic conditions. The molecular characteristics of renal progenitors have been extensively studied, revealing that numerous classical and evolutionarily conserved pathways, such as Notch or Wnt/β-catenin, play a major role in cell regulation. Others, such as retinoic acid, renin-angiotensin-aldosterone system, TLR2 (Toll-like receptor 2) and leptin, are also important in this process. In this review, we summarize the plethora of molecular mechanisms directing renal progenitor responses during homeostasis and following kidney injury. Finally, we will explore how single-cell RNA sequencing could bring the characterization of renal progenitors to the next level, while knowing their molecular signature is gaining relevance in the clinic.
Collapse
|
43
|
Goyal R, Singhal PC. APOL1 risk variants and the development of HIV-associated nephropathy. FEBS J 2020; 288:5586-5597. [PMID: 33340240 DOI: 10.1111/febs.15677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/08/2020] [Accepted: 12/16/2020] [Indexed: 01/03/2023]
Abstract
HIV-associated nephropathy (HIVAN) remains a concern among untreated HIV patients, notably of African descent, as patients can reach end-stage renal disease within 3 years. Two variants (G1 and G2) of the APOL1 gene, common in African populations to protect against African sleeping sickness, have been associated with an increased risk of several glomerular disorders including HIVAN, hypertension-attributed chronic kidney disease, and idiopathic focal segmental glomerulosclerosis and are accordingly named renal risk variants (RRVs). This review examines the mechanisms by which APOL1 RRVs drive glomerular injury in the setting of HIV infection and their potential application to patient management. Innate antiviral mechanisms activated by chronic HIV infection, especially those involving type 1 interferons, are of particular interest as they have been shown to upregulate APOL1 expression. Additionally, the downregulation of miRNA 193a (a repressor of APOL1) is also associated with the upregulation of APOL1. Interestingly, glomerular damage affected by APOL1 RRVs is caused by both loss- and gain-of-function changes in the protein, explicitly characterizing these effects. Their intracellular localization offers a further understanding of the nuances of APOL1 variant effects in promoting renal disease. Finally, although APOL1 variants have been recognized as a critical genetic player in mediating kidney disease, there are significant gaps in their application to patient management for screening, diagnosis, and treatment.
Collapse
Affiliation(s)
- Rohan Goyal
- SUNY Downstate Health Sciences University, New York, NY, USA
| | - Pravin C Singhal
- Institute of Molecular Medicine, Feinstein Institute for Medical Research and Zucker School of Medicine at Hofstra-Northwell, Manhasset, NY, USA
| |
Collapse
|
44
|
Janus kinases inhibitors for treating patients with rhupus. Joint Bone Spine 2020; 87:673-674. [PMID: 32534202 DOI: 10.1016/j.jbspin.2020.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/21/2020] [Indexed: 12/24/2022]
|
45
|
Aslam R, Hussain A, Cheng K, Kumar V, Malhotra A, Gupta S, Singhal PC. Transplantation of mesenchymal stem cells preserves podocyte homeostasis through modulation of parietal epithelial cell activation in adriamycin-induced mouse kidney injury model. Histol Histopathol 2020; 35:1483-1492. [PMID: 33124682 DOI: 10.14670/hh-18-276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
To determine the role of the transplantation of bone marrow-derived mesenchymal stem cells (MSCs) in podocyte renewal, we studied BALB/C mice with or without adriamycin-induced acute kidney injury. MSCs were transplanted ectopically under the capsule of the left kidney or into the peritoneal cavity after the onset of kidney injury to test testing their local or systemic paracrine effects, respectively. Adriamycin produced increases in urine protein: creatinine ratios, blood urea nitrogen, and blood pressure, which improved after both renal subcapsular and intraperitoneal MSCs transplants. The histological changes of adriamycin kidney changes regressed in both kidneys and in only the ipsilateral kidney after intraperitoneal or renal subcapsular transplants indicating that the benefits of transplanted MSCs were related to the extent of paracrine factor distribution. Analysis of kidney tissues for p57-positive parietal epithelial cells (PECs) showed that MSC transplants restored adriamycin-induced decreases in the abundance of these cells to normal levels, although after renal subcapsular transplants these changes did not extend to contralateral kidneys. Moreover, adriamycin caused inflammatory activation of PECs with coexpression of CD44 and phospho-ERK, which was normalized in both or only ipsilateral kidneys depending on whether MSCs were transplanted in the peritoneal cavity or subcapsular space, respectively.
Collapse
Affiliation(s)
- Rukhsana Aslam
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA
| | - Ali Hussain
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA
| | - Kang Cheng
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA
| | - Vinod Kumar
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA
| | - Ashwani Malhotra
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA
| | - Sanjeev Gupta
- Department of Medicine, Department of Pathology, Marion Bessin Liver Research Center, Diabetes Center, The Irwin S. and Sylvia Chanin Institute for Cancer Research, and Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, USA
| | - Pravin C Singhal
- Department of Medicine, Hofstra Northwell School of Medicine, Hempstead, New York, USA.
| |
Collapse
|
46
|
Ramiah Rajasekaran P, Chapin AA, Quan DN, Herberholz J, Bentley WE, Ghodssi R. 3D-Printed electrochemical sensor-integrated transwell systems. MICROSYSTEMS & NANOENGINEERING 2020; 6:100. [PMID: 34567709 PMCID: PMC8433167 DOI: 10.1038/s41378-020-00208-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/03/2020] [Accepted: 08/16/2020] [Indexed: 05/05/2023]
Abstract
This work presents a 3D-printed, modular, electrochemical sensor-integrated transwell system for monitoring cellular and molecular events in situ without sample extraction or microfluidics-assisted downstream omics. Simple additive manufacturing techniques such as 3D printing, shadow masking, and molding are used to fabricate this modular system, which is autoclavable, biocompatible, and designed to operate following standard operating protocols (SOPs) of cellular biology. Integral to the platform is a flexible porous membrane, which is used as a cell culture substrate similarly to a commercial transwell insert. Multimodal electrochemical sensors fabricated on the membrane allow direct access to cells and their products. A pair of gold electrodes on the top side of the membrane measures impedance over the course of cell attachment and growth, characterized by an exponential decrease (~160% at 10 Hz) due to an increase in the double layer capacitance from secreted extracellular matrix (ECM) proteins. Cyclic voltammetry (CV) sensor electrodes, fabricated on the bottom side of the membrane, enable sensing of molecular release at the site of cell culture without the need for downstream fluidics. Real-time detection of ferrocene dimethanol injection across the membrane showed a three order-of-magnitude higher signal at the membrane than in the bulk media after reaching equilibrium. This modular sensor-integrated transwell system allows unprecedented direct, real-time, and noninvasive access to physical and biochemical information, which cannot be obtained in a conventional transwell system.
Collapse
Affiliation(s)
| | - Ashley Augustiny Chapin
- Institute for Systems Research, University of Maryland, College Park, MD USA
- Fischell Department of Bioengineering, University of Maryland, College Park, MD USA
| | - David N. Quan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD USA
| | - Jens Herberholz
- Department of Psychology and Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD USA
| | - William E. Bentley
- Fischell Department of Bioengineering, University of Maryland, College Park, MD USA
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD USA
| | - Reza Ghodssi
- Institute for Systems Research, University of Maryland, College Park, MD USA
- Fischell Department of Bioengineering, University of Maryland, College Park, MD USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD USA
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD USA
| |
Collapse
|
47
|
Abstract
Podocytopathies are kidney diseases in which direct or indirect podocyte injury drives proteinuria or nephrotic syndrome. In children and young adults, genetic variants in >50 podocyte-expressed genes, syndromal non-podocyte-specific genes and phenocopies with other underlying genetic abnormalities cause podocytopathies associated with steroid-resistant nephrotic syndrome or severe proteinuria. A variety of genetic variants likely contribute to disease development. Among genes with non-Mendelian inheritance, variants in APOL1 have the largest effect size. In addition to genetic variants, environmental triggers such as immune-related, infection-related, toxic and haemodynamic factors and obesity are also important causes of podocyte injury and frequently combine to cause various degrees of proteinuria in children and adults. Typical manifestations on kidney biopsy are minimal change lesions and focal segmental glomerulosclerosis lesions. Standard treatment for primary podocytopathies manifesting with focal segmental glomerulosclerosis lesions includes glucocorticoids and other immunosuppressive drugs; individuals not responding with a resolution of proteinuria have a poor renal prognosis. Renin-angiotensin system antagonists help to control proteinuria and slow the progression of fibrosis. Symptomatic management may include the use of diuretics, statins, infection prophylaxis and anticoagulation. This Primer discusses a shift in paradigm from patient stratification based on kidney biopsy findings towards personalized management based on clinical, morphological and genetic data as well as pathophysiological understanding.
Collapse
|
48
|
Zaidan M, Burtin M, Zhang JD, Blanc T, Barre P, Garbay S, Nguyen C, Vasseur F, Yammine L, Germano S, Badi L, Gubler MC, Gallazzini M, Friedlander G, Pontoglio M, Terzi F. Signaling pathways predisposing to chronic kidney disease progression. JCI Insight 2020; 5:126183. [PMID: 32376805 DOI: 10.1172/jci.insight.126183] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/08/2020] [Indexed: 12/18/2022] Open
Abstract
The loss of functional nephrons after kidney injury triggers the compensatory growth of the remaining ones to allow functional adaptation. However, in some cases, these compensatory events activate signaling pathways that lead to pathological alterations and chronic kidney disease. Little is known about the identity of these pathways and how they lead to the development of renal lesions. Here, we combined mouse strains that differently react to nephron reduction with molecular and temporal genome-wide transcriptome studies to elucidate the molecular mechanisms involved in these events. We demonstrated that nephron reduction led to 2 waves of cell proliferation: the first one occurred during the compensatory growth regardless of the genetic background, whereas the second one occurred, after a quiescent phase, exclusively in the sensitive strain and accompanied the development of renal lesions. Similarly, clustering by coinertia analysis revealed the existence of 2 waves of gene expression. Interestingly, we identified type I interferon (IFN) response as an early (first-wave) and specific signature of the sensitive (FVB/N) mice. Activation of type I IFN response was associated with G1/S cell cycle arrest, which correlated with p21 nuclear translocation. Remarkably, the transient induction of type I IFN response by poly(I:C) injections during the compensatory growth resulted in renal lesions in otherwise-resistant C57BL6 mice. Collectively, these results suggest that the early molecular and cellular events occurring after nephron reduction determine the risk of developing late renal lesions and point to type I IFN response as a crucial event of the deterioration process.
Collapse
Affiliation(s)
- Mohamad Zaidan
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France.,Service de Néphrologie-Transplantation, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris (AP-HP), Le Kremlin-Bicêtre, France
| | - Martine Burtin
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France
| | - Jitao David Zhang
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Thomas Blanc
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France.,Service de Chirurgie Viscérale et Urologie Pédiatrique, Hôpital Necker Enfants Malades, AP-HP, Paris, France
| | - Pauline Barre
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France
| | - Serge Garbay
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France
| | - Clément Nguyen
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France
| | - Florence Vasseur
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France
| | - Lucie Yammine
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France
| | - Serena Germano
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France
| | - Laura Badi
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | | | - Morgan Gallazzini
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France
| | - Gérard Friedlander
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France.,Service d'Explorations Fonctionnelles, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Marco Pontoglio
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France
| | - Fabiola Terzi
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Department of Growth and Signaling, Université de Paris, Paris, France
| |
Collapse
|
49
|
The Use of High-Throughput Transcriptomics to Identify Pathways with Therapeutic Significance in Podocytes. Int J Mol Sci 2019; 21:ijms21010274. [PMID: 31906131 PMCID: PMC6981397 DOI: 10.3390/ijms21010274] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/24/2019] [Accepted: 12/26/2019] [Indexed: 12/25/2022] Open
Abstract
Podocytes have a unique structure that supports glomerular filtration function, and many glomerular diseases result in loss of this structure, leading to podocyte dysfunction and ESRD (end stage renal disease). These structural and functional changes involve a complex set of molecular and cellular mechanisms that remain poorly understood. To understand the molecular signature of podocyte injury, we performed transcriptome analysis of cultured human podocytes injured either with PAN (puromycin aminonucleoside) or doxorubicin/adriamycin (ADR). The pathway analysis through DE (differential expression) and gene-enrichment analysis of the injured podocytes showed Tumor protein p53 (P53) as one of the major signaling pathways that was significantly upregulated upon podocyte injury. Accordingly, P53 expression was also up-regulated in the glomeruli of nephrotoxic serum (NTS) and ADR-injured mice. To further confirm these observations, cultured podocytes were treated with the P53 inhibitor pifithrin-α, which showed significant protection from ADR-induced actin cytoskeleton damage. In conclusion, signaling pathways that are involved in podocyte pathogenesis and can be therapeutically targeted were identified by high-throughput transcriptomic analysis of injured podocytes.
Collapse
|
50
|
Andrianova NV, Buyan MI, Zorova LD, Pevzner IB, Popkov VA, Babenko VA, Silachev DN, Plotnikov EY, Zorov DB. Kidney Cells Regeneration: Dedifferentiation of Tubular Epithelium, Resident Stem Cells and Possible Niches for Renal Progenitors. Int J Mol Sci 2019; 20:ijms20246326. [PMID: 31847447 PMCID: PMC6941132 DOI: 10.3390/ijms20246326] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/10/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022] Open
Abstract
A kidney is an organ with relatively low basal cellular regenerative potential. However, renal cells have a pronounced ability to proliferate after injury, which undermines that the kidney cells are able to regenerate under induced conditions. The majority of studies explain yielded regeneration either by the dedifferentiation of the mature tubular epithelium or by the presence of a resident pool of progenitor cells in the kidney tissue. Whether cells responsible for the regeneration of the kidney initially have progenitor properties or if they obtain a “progenitor phenotype” during dedifferentiation after an injury, still stays the open question. The major stumbling block in resolving the issue is the lack of specific methods for distinguishing between dedifferentiated cells and resident progenitor cells. Transgenic animals, single-cell transcriptomics, and other recent approaches could be powerful tools to solve this problem. This review examines the main mechanisms of kidney regeneration: dedifferentiation of epithelial cells and activation of progenitor cells with special attention to potential niches of kidney progenitor cells. We attempted to give a detailed description of the most controversial topics in this field and ways to resolve these issues.
Collapse
Affiliation(s)
- Nadezda V. Andrianova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Marina I. Buyan
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Ljubava D. Zorova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Irina B. Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Vasily A. Popkov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Valentina A. Babenko
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Denis N. Silachev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, 119991 Moscow, Russia
- Correspondence: (E.Y.P.); (D.B.Z.); Tel.: +7-495-939-5944 (E.Y.P.)
| | - Dmitry B. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
- Correspondence: (E.Y.P.); (D.B.Z.); Tel.: +7-495-939-5944 (E.Y.P.)
| |
Collapse
|