1
|
Zhang J, Cheng L, Jiang S, Zhu D. Machine learning based identification of anoikis related gene classification patterns and immunoinfiltration characteristics in diabetic nephropathy. Sci Rep 2025; 15:15271. [PMID: 40312440 PMCID: PMC12046048 DOI: 10.1038/s41598-025-99395-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 04/21/2025] [Indexed: 05/03/2025] Open
Abstract
Anoikis and immune cell infiltration are pivotal factors in the pathophysiological mechanism of diabetic nephropathy (DN), yet a comprehensive understanding of the mechanism is lacking. This work aimed to pinpoint distinctive anoikis-related genes (ARGs) in DN and delve into their impact on the immune landscape. Three datasets (GSE30528, GSE47184, and GSE96804) were downloaded from the gene expression omnibus (GEO) dataset. Differentially expressed genes (DEGs) were identified using the "limma" package, while ARGs were obtained from GSEA, GeneCard, and Harmonizome datasets. The intersection of DEGs and ARGs was analyzed for Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. The CIBERSORT algorithm was employed to estimate the infiltration percentage of 22 immune cell types in DN renal tissue. Subsequently, the least absolute shrinkage and selection operator (LASSO), support vector machine recursive feature elimination (SVM-RFE), and random forest (RF) algorithms were adopted to screen key ARGs related to DN. After that, receiver operating characteristic (ROC) analysis was employed to assess the diagnostic accuracy of each gene and the real-time quantitative polymerase chain reaction (RT-qPCR) was adopted to quantitatively detect the expression of biomarkers in DN cell models. Finally, correlations between key genes and immune cell infiltration were analyzed, and a competitive endogenous ribonucleic acid (RNA) (ceRNA) network based on key genes was constructed. A total of 59 DEARGs were identified. GO functional annotation enrichment analysis revealed their involvement in kidney development, extracellular matrix (ECM), cytoplasmic vesicle cavity, immunoinflammatory response, and cytokine effect. KEGG pathway analysis indicated that MAPK, PI3K -Akt, IL -17, TNF, and HIF- 1 signaling pathways are critical for DN. In addition, seven key genes, including PDK4, S100A8, HTRA1, CHI3L1, WT1, CDKN1B, and EGF, were screened by machine learning algorithm. Most of these genes exhibited low expression in renal tissue of DN patients and positive correlation with neutrophils, and their expressions were verified in an external dataset cell model. The ceRNA analysis suggested potential regulatory pathways (H19/miR-15b-5p/PDK4 and KCNQ1T1/miR-1207-3p/WT1) influencing early DN progression. This work provided a comprehensive analysis of the role of DEARGs in DN for the first time, offering valuable insights for further understanding the disease mechanism and guiding clinical diagnosis, treatment, and research of DN.
Collapse
Affiliation(s)
- Jing Zhang
- The Third Department of Jiaozhou City Traditional Chinese Medicine Hospital, Jiaozhou, 266300, Shandong, China
| | - Lulu Cheng
- Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, China
| | - Shan Jiang
- Traditional Chinese Medicine Department of Linyi People's Hospital, Linyi, 276000, Shandong, China
| | - Duosheng Zhu
- The Third Department of Jiaozhou City Traditional Chinese Medicine Hospital, Jiaozhou, 266300, Shandong, China.
| |
Collapse
|
2
|
Gupta AK, Minocha E, Koss KM, Naved BA, Safar-Boueri L, Wertheim JA, Gallon L. A kidney organoid-based readout to assess disease activity in primary and recurrent focal segmental glomerulosclerosis. Kidney Int 2025; 107:888-902. [PMID: 39914654 DOI: 10.1016/j.kint.2025.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 02/23/2025]
Abstract
Primary focal segmental glomerulosclerosis (pFSGS) is an acquired kidney disorder that frequently leads to kidney failure and confers an elevated risk of recurrence after kidney transplantation, termed recurrent pFSGS. Unfortunately, there is no diagnostic method to foresee recurrence of pFSGS after kidney transplantation. Progress in developing assays to test disease activity is hampered by few preclinical models to replicate disease and inability of in vitro cultured primary podocytes to remain terminally differentiated. In recent years, advancements in kidney organoid biology have led to the development of kidney tissues with glomeruli and major nephron segments including podocytes. To develop a pFSGS model, we studied the effect of plasma from patients diagnosed with pFSGS on kidney organoids differentiated from human pluripotent stem cells. The pFSGS plasma treatment induced podocytopathy, extracellular matrix protein deposition, fibrosis and apoptosis within organoids, whereas non-recurrent plasma did not affect organoid structure. pFSGS plasma also led to loss of normal expression patterns of podocyte specific proteins, nephrin and podocin within podocytes. Further, cytokine array profiling revealed that pFSGS plasma induced secretion of cytokines associated with inflammation and angiogenesis. Additionally, kidney organoids treated with plasma obtained after therapeutic plasma exchange for recurrent pFSGS led to lower cell death in organoids after sequential exchanges with the final exchange showing the least apoptotic cells without morphological abnormality. Overall, our results demonstrate the potential of kidney organoids in advancing kidney disease modeling. These insights could be applied in clinical settings to assist in gauging FSGS recurrence risk prior to kidney transplantation.
Collapse
Affiliation(s)
- Ashwani Kumar Gupta
- Department of Surgery, University of Arizona College of Medicine, Tucson, Arizona, USA; Bio5 Institute, University of Arizona, Tucson, Arizona, USA; Surgery Service, Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona, USA; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ekta Minocha
- Department of Surgery, University of Arizona College of Medicine, Tucson, Arizona, USA; Bio5 Institute, University of Arizona, Tucson, Arizona, USA; Surgery Service, Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona, USA
| | - Kyle M Koss
- Department of Surgery, University of Arizona College of Medicine, Tucson, Arizona, USA; Bio5 Institute, University of Arizona, Tucson, Arizona, USA; Surgery Service, Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona, USA; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Neurobiology, University of Texas Medical Branch, Galveston, Texas, USA; Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, Texas, USA
| | - Bilal A Naved
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA
| | - Luisa Safar-Boueri
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jason A Wertheim
- Department of Surgery, University of Arizona College of Medicine, Tucson, Arizona, USA; Bio5 Institute, University of Arizona, Tucson, Arizona, USA; Surgery Service, Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona, USA; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA; Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA.
| | - Lorenzo Gallon
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Medicine, Abdominal Organ Transplant Program, University of Illinois Chicago, Chicago, Illinois, USA.
| |
Collapse
|
3
|
Laudon A, Wang Z, Zou A, Sharma R, Ji J, Tan W, Kim C, Qian Y, Ye Q, Chen H, Henderson JM, Zhang C, Kolachalama VB, Lu W. Digital pathology assessment of kidney glomerular filtration barrier ultrastructure in an animal model of podocytopathy. Biol Methods Protoc 2025; 10:bpaf024. [PMID: 40223818 PMCID: PMC11992336 DOI: 10.1093/biomethods/bpaf024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/16/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
Transmission electron microscopy (TEM) images can visualize kidney glomerular filtration barrier ultrastructure, including the glomerular basement membrane (GBM) and podocyte foot processes (PFP). Podocytopathy is associated with glomerular filtration barrier morphological changes observed experimentally and clinically by measuring GBM or PFP width. However, these measurements are currently performed manually. This limits research on podocytopathy disease mechanisms and therapeutics due to labor intensiveness and inter-operator variability. We developed a deep learning-based digital pathology computational method to measure GBM and PFP width in TEM images from the kidneys of Integrin-Linked Kinase (ILK) podocyte-specific conditional knockout (cKO) mouse, an animal model of podocytopathy, compared to wild-type (WT) control mouse. We obtained TEM images from WT and ILK cKO littermate mice at 4 weeks old. Our automated method was composed of two stages: a U-Net model for GBM segmentation, followed by an image processing algorithm for GBM and PFP width measurement. We evaluated its performance with a 4-fold cross-validation study on WT and ILK cKO mouse kidney pairs. Mean [95% confidence interval (CI)] GBM segmentation accuracy, calculated as Jaccard index, was 0.73 (0.70-0.76) for WT and 0.85 (0.83-0.87) for ILK cKO TEM images. Automated and manual GBM width measurements were similar for both WT (P = .49) and ILK cKO (P = .06) specimens. While automated and manual PFP width measurements were similar for WT (P = .89), they differed for ILK cKO (P < .05) specimens. WT and ILK cKO specimens were morphologically distinguishable by manual GBM (P < .05) and PFP (P < .05) width measurements. This phenotypic difference was reflected in the automated GBM (P < .05) more than PFP (P = .06) widths. Our deep learning-based digital pathology tool automated measurements in a mouse model of podocytopathy. This proposed method provides high-throughput, objective morphological analysis and could facilitate podocytopathy research.
Collapse
Affiliation(s)
- Aksel Laudon
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
- Nephrology Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Zhaoze Wang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Anqi Zou
- Computational Biomedicine Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Richa Sharma
- Nephrology Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Jiayi Ji
- Nephrology Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Winston Tan
- Nephrology Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Connor Kim
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Yingzhe Qian
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Qin Ye
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Hui Chen
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Joel M Henderson
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Chao Zhang
- Computational Biomedicine Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| | - Vijaya B Kolachalama
- Computational Biomedicine Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, United States
- Department of Computer Science and Faculty of Computing & Data Sciences, Boston University, Boston, MA 02215, United States
| | - Weining Lu
- Nephrology Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, United States
| |
Collapse
|
4
|
Köhler P, Ribeiro A, Honarpisheh M, von Rauchhaupt E, Lorenz G, Li C, Martin L, Steiger S, Lindenmeyer M, Schmaderer C, Anders HJ, Thomasova D, Lech M. Podocyte A20/TNFAIP3 Controls Glomerulonephritis Severity via the Regulation of Inflammatory Responses and Effects on the Cytoskeleton. Cells 2025; 14:381. [PMID: 40072109 PMCID: PMC11898495 DOI: 10.3390/cells14050381] [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: 12/11/2024] [Revised: 01/22/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
A20/Tnfaip3, an early NF-κB response gene and key negative regulator of NF-κB signaling, suppresses proinflammatory responses. Its ubiquitinase and deubiquitinase activities mediate proteasomal degradation within the NF-κB pathway. This study investigated the involvement of A20 signaling alterations in podocytes in the development of kidney injury. The phenotypes of A20Δpodocyte (podocyte-specific knockout of A20) mice were compared with those of control mice at 6 months of age to identify spontaneous changes in kidney function. A20Δpodocyte mice presented elevated serum urea nitrogen and creatinine levels, along with increased accumulation of inflammatory cells-neutrophils and macrophages-within the glomeruli. Additionally, A20Δpodocyte mice displayed significant podocyte loss. Ultrastructural analysis of A20 podocyte-knockout mouse glomeruli revealed hypocellularity of the glomerular tuft, expansion of the extracellular matrix, podocytopenia associated with foot process effacement, karyopyknosis, micronuclei, and podocyte detachment. In addition to podocyte death, we also observed damage to intracapillary endothelial cells with vacuolation of the cytoplasm and condensation of nuclear chromatin. A20 expression downregulation and CRISPR-Cas9 genome editing targeting A20 in a podocyte cell line confirmed these findings in vitro, highlighting the significant contribution of A20 activity in podocytes to glomerular injury pathogenesis. Finally, we analyzed TNFAIP3 transcription levels alongside genes involved in apoptosis, anoikis, NF-κB regulation, and cell attachment in glomerular and tubular compartments of kidney biopsies of patients with various renal diseases.
Collapse
Affiliation(s)
- Paulina Köhler
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Andrea Ribeiro
- Klinikum Rechts der Isar, Department of Nephrology, Technical University Munich (TUM), 80333 München, Germany
| | - Mohsen Honarpisheh
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Ekaterina von Rauchhaupt
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Georg Lorenz
- Klinikum Rechts der Isar, Department of Nephrology, Technical University Munich (TUM), 80333 München, Germany
| | - Chenyu Li
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Lucas Martin
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Stefanie Steiger
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Maja Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Christoph Schmaderer
- Klinikum Rechts der Isar, Department of Nephrology, Technical University Munich (TUM), 80333 München, Germany
| | - Hans-Joachim Anders
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Dana Thomasova
- Institute of Biology and Medical Genetics, 2nd Faculty of Medicine, Charles University Prague and University Hospital Motol, 15006 Prague, Czech Republic
| | - Maciej Lech
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| |
Collapse
|
5
|
Zago CCFB, Oliveira BDF, Uehara G, da Silva ALC, Rocha LP, Custódio FB, Araújo LS, da Silva CA, Reis MA, Machado JR. Influence of Podocyte Injury on the Development of Class IV Lupus Nephritis. Int J Nephrol Renovasc Dis 2024; 17:215-225. [PMID: 39381781 PMCID: PMC11460275 DOI: 10.2147/ijnrd.s473616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/27/2024] [Indexed: 10/10/2024] Open
Abstract
Purpose In the kidneys, Systemic Lupus Erythematosus leads to Lupus Nephritis (LN), a form of glomerulonephritis. There is evidence that patients with LN may present activation of specific pathways for podocyte injury. This injury can occur through different mechanisms such as loss of podocyte adhesion to the glomerular basement membrane, cell death or dedifferentiation. Podocyturia with consequent podocytopenia has been described in some nephropathies such as LN, highlighting the importance of studying podocyte injuries in this condition. Evaluating in situ morphological characteristics of podocytes becomes relevant for a better understanding of the processes involved in their pathogenesis. This study investigated podocytes in different classes of LN in renal biopsies performed by the Kidney Research Center at the Federal University of Triângulo Mineiro. Patients and Methods Twenty control cases and 29 biopsy cases diagnosed with LN were selected, divided according to the histopathological classes of the disease. Podocyte density was assessed through immunohistochemistry for Wilms tumor 1 protein and the evaluation of foot process effacement was performed by transmission electron microscopy. Results Podocyte density was lower in the LN and this reduction was observed in all analyzed classes when compared to the control group. More foot process effacement was observed in the LN group, with more effacement in classes I/II and class IV compared to the control group. The class IV group showed more foot process effacement than the class III group and presented higher proteinuria levels compared to the classes I/II group. A strong, positive, and significant correlation was observed between the activity index and foot process effacement in the class IV group. Conclusion Podocytes play an important role in the development of LN, and possibly, injuries to these cells are more closely related to the inflammatory/diffuse proliferative cellular process developed in class IV LN.
Collapse
Affiliation(s)
- Cyntia Camilo Franco Borges Zago
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Bruna de Freitas Oliveira
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Giovanna Uehara
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Ana Laura Carvalho da Silva
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Laura Penna Rocha
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | | | - Liliane Silvano Araújo
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Crislaine Aparecida da Silva
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Marlene Antônia Reis
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Juliana Reis Machado
- Kidney Research Center-Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| |
Collapse
|
6
|
Kajio Y, Suzuki T, Kobayashi K, Kanazawa N, Iyoda M, Honda H, Honda K. Activation of the inflammasome and pyroptosis cascade in podocytes of patients with minimal change disease. Clin Kidney J 2024; 17:sfae216. [PMID: 39114498 PMCID: PMC11304592 DOI: 10.1093/ckj/sfae216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Indexed: 08/10/2024] Open
Abstract
Background In contrast to childhood minimal change disease (MCD), adult-onset MCD frequently recurs and requires prolonged immunosuppressive therapy. Accordingly, an investigation of the pathogenesis of adult MCD is required. MCD is usually accompanied by severe dyslipidaemia. Oxidized low-density lipoprotein (ox-LDL) is known to function in a damage-associated molecular pattern (DAMP) through CD36, triggering the NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome and programmed cell death called pyroptosis. However, the relationship between MCD pathogenesis and NLRP3 inflammasome/pyroptosis activation via CD36 is not fully understood. Methods We conducted comprehensive histological and clinical evaluations by analysing renal biopsy (RBx) specimens and urine samples obtained from 26 patients with MCD. These samples were compared with control kidneys from 15 transplant donors and urine samples from 15 healthy volunteers. Results The number of podocytes was lower in the MCD group than in the control group. Urinary ox-LDL levels were higher in the MCD group than in the control group. Immunofluorescence staining revealed that NLRP3 and CD36 were upregulated in MCD podocytes. Urinary interleukin (IL)-18 levels increased in patients with MCD. Steroid therapy performed before RBx appeared to maintain the podocyte number and reduce urinary ox-LDL and IL-18 levels. Conclusion In MCD, the NLRP3 inflammasome and pyroptosis cascade seem to be activated via upregulation of CD36 in podocytes, associated with increased urinary ox-LDL. Elevated urinary IL-18 levels suggest that pyroptosis may occur in MCD. Further research is required to confirm the significance of the podocyte NLRP3 inflammasome/pyroptosis in MCD.
Collapse
Affiliation(s)
- Yuki Kajio
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Taihei Suzuki
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Kazuki Kobayashi
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Nobuhiro Kanazawa
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Masayuki Iyoda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
| | - Hirokazu Honda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Kazuho Honda
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
7
|
Laudon A, Wang Z, Zou A, Sharma R, Ji J, Kim C, Qian Y, Ye Q, Chen H, Henderson JM, Zhang C, Kolachalama VB, Lu W. Digital pathology assessment of kidney glomerular filtration barrier ultrastructure in an animal model of podocytopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.599097. [PMID: 38948787 PMCID: PMC11212870 DOI: 10.1101/2024.06.14.599097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Transmission electron microscopy (TEM) images can visualize kidney glomerular filtration barrier ultrastructure, including the glomerular basement membrane (GBM) and podocyte foot processes (PFP). Podocytopathy is associated with glomerular filtration barrier morphological changes observed experimentally and clinically by measuring GBM or PFP width. However, these measurements are currently performed manually. This limits research on podocytopathy disease mechanisms and therapeutics due to labor intensiveness and inter-operator variability. Methods We developed a deep learning-based digital pathology computational method to measure GBM and PFP width in TEM images from the kidneys of Integrin-Linked Kinase (ILK) podocyte-specific conditional knockout (cKO) mouse, an animal model of podocytopathy, compared to wild-type (WT) control mouse. We obtained TEM images from WT and ILK cKO littermate mice at 4 weeks old. Our automated method was composed of two stages: a U-Net model for GBM segmentation, followed by an image processing algorithm for GBM and PFP width measurement. We evaluated its performance with a 4-fold cross-validation study on WT and ILK cKO mouse kidney pairs. Results Mean (95% confidence interval) GBM segmentation accuracy, calculated as Jaccard index, was 0.73 (0.70-0.76) for WT and 0.85 (0.83-0.87) for ILK cKO TEM images. Automated and manual GBM width measurements were similar for both WT (p=0.49) and ILK cKO (p=0.06) specimens. While automated and manual PFP width measurements were similar for WT (p=0.89), they differed for ILK cKO (p<0.05) specimens. WT and ILK cKO specimens were morphologically distinguishable by manual GBM (p<0.05) and PFP (p<0.05) width measurements. This phenotypic difference was reflected in the automated GBM (p<0.05) more than PFP (p=0.06) widths. Conclusions These results suggest that certain automated measurements enabled via deep learning-based digital pathology tools could distinguish healthy kidneys from those with podocytopathy. Our proposed method provides high-throughput, objective morphological analysis and could facilitate podocytopathy research and translate into clinical diagnosis.
Collapse
Affiliation(s)
- Aksel Laudon
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Nephrology Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Zhaoze Wang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Anqi Zou
- Computational Biomedicine Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Richa Sharma
- Nephrology Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Jiayi Ji
- Nephrology Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Connor Kim
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Yingzhe Qian
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Qin Ye
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Hui Chen
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Joel M Henderson
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Chao Zhang
- Computational Biomedicine Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Vijaya B Kolachalama
- Computational Biomedicine Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
- Department of Computer Science and Faculty of Computing & Data Sciences, Boston University, Boston, MA, USA
| | - Weining Lu
- Nephrology Section, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| |
Collapse
|
8
|
Zhang Y, Yu C, Li X. Kidney Aging and Chronic Kidney Disease. Int J Mol Sci 2024; 25:6585. [PMID: 38928291 PMCID: PMC11204319 DOI: 10.3390/ijms25126585] [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: 04/27/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
The process of aging inevitably leads to an increase in age-related comorbidities, including chronic kidney disease (CKD). In many aspects, CKD can be considered a state of accelerated and premature aging. Aging kidney and CKD have numerous common characteristic features, ranging from pathological presentation and clinical manifestation to underlying mechanisms. The shared mechanisms underlying the process of kidney aging and the development of CKD include the increase in cellular senescence, the decrease in autophagy, mitochondrial dysfunction, and the alterations of epigenetic regulation, suggesting the existence of potential therapeutic targets that are applicable to both conditions. In this review, we provide a comprehensive overview of the common characteristics between aging kidney and CKD, encompassing morphological changes, functional alterations, and recent advancements in understanding the underlying mechanisms. Moreover, we discuss potential therapeutic strategies for targeting senescent cells in both the aging process and CKD.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Chen Yu
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China;
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
9
|
Zhao M, Yin Y, Yang B, Chang M, Ma S, Shi X, Li Q, Li P, Zhang Y. Ameliorative effects of Modified Huangqi Chifeng decoction on podocyte injury via autophagy mediated by PI3K/AKT/mTOR and AMPK/mTOR pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117520. [PMID: 38042389 DOI: 10.1016/j.jep.2023.117520] [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: 10/22/2023] [Revised: 11/18/2023] [Accepted: 11/26/2023] [Indexed: 12/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Proteinuria is recognized as a risk factor for the exacerbation of chronic kidney disease. Modified Huangqi Chifeng decoction (MHCD) has distinct advantages in reducing proteinuria. Our previous experimental results have shown that MHCD can inhibit excessive autophagy. However, the specific mechanism by which MHCD regulates autophagy needs to be further explored. AIM OF THE STUDY In this study, in vivo and in vitro experiments were conducted to further clarify the protective mechanism of MHCD on the kidney and podocytes by regulating autophagy based on phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) and adenosine monophosphate-activated protein kinase (AMPK)/mTOR signaling pathways. MATERIALS AND METHODS By a single injection via the tail vein, Sprague-Dawley rats received Adriamycin (5 mg/kg) to establish a model of proteinuria nephropathy. They were divided into control, model, MHCD, 3-methyladenine (3 MA), 3 MA + MHCD, and telmisartan groups and were administered continuously for 6 weeks. The MHCD-containing serum was prepared, and a model of podocyte injury induced by Adriamycin (0.2 μg/mL) was established. RESULTS MHCD reduced the 24-h urine protein levels and relieved pathological kidney damage. During autophagy in the kidneys of rats with Adriamycin-induced nephropathy, the PI3K/AKT/mTOR signaling pathway is inhibited, while the AMPK/mTOR signaling pathway is activated. MHCD antagonized these effects, thereby inhibiting excessive autophagy. MHCD alleviated Adriamycin-induced podocyte autophagy, as demonstrated using Pik3r1 siRNA and an overexpression plasmid for Prkaa1/Prkaa2. Furthermore, MHCD could activate the PI3K/AKT/mTOR signaling pathway while suppressing the AMPK/mTOR signaling pathway. CONCLUSIONS This study demonstrated that MHCD can activate the interaction between the PI3K/AKT/mTOR and the AMPK/mTOR signaling pathways to maintain autophagy balance, inhibit excessive autophagy, and play a role in protecting the kidneys and podocytes.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Yundong Yin
- Postdoctoral Research Station, China Academy of Chinese Medical Sciences, Beijing, 100700, China; Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Bin Yang
- Department of Pathology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Meiying Chang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Sijia Ma
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Xiujie Shi
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Qi Li
- Department of Clinical Laboratory, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Peng Li
- Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Key Laboratory of Pharmacology of Chinese Materia Medica of Beijing, Beijing, 100091, China.
| | - Yu Zhang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China; Xin-Huangpu Joint Innovation Institute of Chinese Medicine, Guangzhou, 510000, China.
| |
Collapse
|
10
|
Bhatia D, Choi ME. Autophagy and mitophagy: physiological implications in kidney inflammation and diseases. Am J Physiol Renal Physiol 2023; 325:F1-F21. [PMID: 37167272 PMCID: PMC10292977 DOI: 10.1152/ajprenal.00012.2023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/25/2023] [Accepted: 05/09/2023] [Indexed: 05/13/2023] Open
Abstract
Autophagy is a ubiquitous intracellular cytoprotective quality control program that maintains cellular homeostasis by recycling superfluous cytoplasmic components (lipid droplets, protein, or glycogen aggregates) and invading pathogens. Mitophagy is a selective form of autophagy that by recycling damaged mitochondrial material, which can extracellularly act as damage-associated molecular patterns, prevents their release. Autophagy and mitophagy are indispensable for the maintenance of kidney homeostasis and exert crucial functions during both physiological and disease conditions. Impaired autophagy and mitophagy can negatively impact the pathophysiological state and promote its progression. Autophagy helps in maintaining structural integrity of the kidney. Mitophagy-mediated mitochondrial quality control is explicitly critical for regulating cellular homeostasis in the kidney. Both autophagy and mitophagy attenuate inflammatory responses in the kidney. An accumulating body of evidence highlights that persistent kidney injury-induced oxidative stress can contribute to dysregulated autophagic and mitophagic responses and cell death. Autophagy and mitophagy also communicate with programmed cell death pathways (apoptosis and necroptosis) and play important roles in cell survival by preventing nutrient deprivation and regulating oxidative stress. Autophagy and mitophagy are activated in the kidney after acute injury. However, their aberrant hyperactivation can be deleterious and cause tissue damage. The findings on the functions of autophagy and mitophagy in various models of chronic kidney disease are heterogeneous and cell type- and context-specific dependent. In this review, we discuss the roles of autophagy and mitophagy in the kidney in regulating inflammatory responses and during various pathological manifestations.
Collapse
Affiliation(s)
- Divya Bhatia
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, United States
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, United States
| |
Collapse
|
11
|
Ma L, Zhang Y, Zhang L, Shao C, Yu N. Development and validation of a simple-to-use nomogram for predicting minimal change disease based on quantification of color Doppler sonography data from a region of interest. Abdom Radiol (NY) 2023; 48:1020-1032. [PMID: 36627405 DOI: 10.1007/s00261-022-03780-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023]
Abstract
OBJECTIVES To establish a simple-to-use nomogram based on quantification of color Doppler sonography data from a region of interest (ROI) to diagnose minimal change disease (MCD) promptly and non-invasively, and to evaluate the prediction capability of the nomogram. METHODS We recruited 564 patients with pathology-proven renal disease who were admitted to our hospital from July 2020 to July 2021 (388 patients in the training dataset and 176 patients in the validation dataset), and their color Doppler sonography data were acquired from a ROI and underwent ipsilateral renal biopsy. The collected clinical features and ultrasonic features were imported into Rstuido and statistically significant features were selected by stepwise regression using the forward-backward method. Multivariate Logistic regression analysis was combined with clinical analysis to obtain the final modeling features. General and dynamic nomogram models were constructed with the selected features, depending on whether they were MCD or not. Bootstrapping and internal validation were used for internal and external validation of the nomogram, respectively. The performance of the nomogram was assessed by C-index, calibration curve, and receiver operating characteristic (ROC) curve. RESULTS Age and VI were independent factors in predicting MCD. The value of Age (Best cut-off value: 33.5 years) combined with VI (Best cut-off value: 40.50 points) in the diagnosis of MCD was significantly higher than that of single diagnosis (AUC 0.901, 95% CI 0.863-0.938). The C-index of the nomogram constructed with age and VI in the training and validation datasets was 0.915 [95% confidence interval (CI) 0.874-0.956 and 0.875 95% CI 0.783-0.967], respectively. Calibration curves were fitted well. The sensitivity, specificity, and accuracy were 76.1%, 95.6%, and 78.3%, respectively, in the training dataset, and 74.1%, 94.4%, and 76.1% in the validation dataset, respectively. CONCLUSION The nomogram constructed with age and VI showed a satisfactory degree of differentiation and accuracy, which is of great significance for early, non-invasively, and individually analysis of the risk of MCD.
Collapse
Affiliation(s)
- Leiyuan Ma
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, No.16, Jiangsu Road, Shinan District, Qingdao, 266003, China
| | - Yuhan Zhang
- University of Southern California, Los Angeles, USA
| | - Liang Zhang
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, No.16, Jiangsu Road, Shinan District, Qingdao, 266003, China
| | - Changjie Shao
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, No.16, Jiangsu Road, Shinan District, Qingdao, 266003, China
| | - Ning Yu
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, No.16, Jiangsu Road, Shinan District, Qingdao, 266003, China.
| |
Collapse
|
12
|
Martins ALMDS, Bernardes AB, Ferreira VA, Wanderley DC, Araújo SDA, do Carmo Neto JR, da Silva CA, Lira RCP, Araújo LS, Dos Reis MA, Machado JR. In situ assessment of Mindin as a biomarker of podocyte lesions in diabetic nephropathy. PLoS One 2023; 18:e0284789. [PMID: 37130106 PMCID: PMC10153717 DOI: 10.1371/journal.pone.0284789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 04/07/2023] [Indexed: 05/03/2023] Open
Abstract
Diabetic nephropathy (DN) is the leading cause of chronic kidney disease and end-stage renal failure worldwide. Several mechanisms are involved in the pathogenesis of this disease, which culminate in morphological changes such as podocyte injury. Despite the complex diagnosis and pathogenesis, limited attempts have been made to establish new biomarkers for DN. The higher concentration of Mindin protein in the urine of patients with type 2 diabetes mellitus suggests that it plays a role in DN. Therefore, this study investigated whether in situ protein expression of Mindin can be considered a potential DN biomarker. Fifty renal biopsies from patients diagnosed with DN, 57 with nondiabetic glomerular diseases, including 17 with focal segmental glomerulosclerosis (FSGS), 14 with minimal lesion disease (MLD) and 27 with immunoglobulin A nephropathy (IgAN), and 23 adult kidney samples from autopsies (control group) were evaluated for Mindin expression by immunohistochemistry. Podocyte density was inferred by Wilms' tumor 1 (WT1) immunostaining, while foot process effacement was assessed by transmission electron microscopy. Receiver operative characteristic (ROC) analysis was performed to determine the biomarker sensitivity/specificity. Low podocyte density and increased Mindin expression were observed in all cases of DN, regardless of their class. In the DN group, Mindin expression was significantly higher than that in the FSGS, MCD, IgAN and control groups. Higher Mindin expression was significantly positively correlated with foot process effacement only in class III DN cases. Furthermore, Mindin protein presented high specificity in the biopsies of patients with DN (p < 0.0001). Our data suggest that Mindin may play a role in DN pathogenesis and is a promising biomarker of podocyte lesions.
Collapse
Affiliation(s)
- Ana Luisa Monteiro Dos Santos Martins
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences of Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Alexia Borges Bernardes
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences of Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Verônica Aparecida Ferreira
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences of Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - David Campos Wanderley
- Institute of Nephropathology, Center for Electron Microscopy, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Stanley de Almeida Araújo
- Institute of Nephropathology, Center for Electron Microscopy, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - José Rodrigues do Carmo Neto
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiania, Goiás, Brazil
| | - Crislaine Aparecida da Silva
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences of Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Régia Caroline Peixoto Lira
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences of Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Liliane Silvano Araújo
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences of Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Marlene Antônia Dos Reis
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences of Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Juliana Reis Machado
- Department of Pathology, Genetics and Evolution, Discipline of General Pathology, Institute of Biological and Natural Sciences of Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| |
Collapse
|
13
|
Kaneko S, Yanai K, Ishii H, Aomatsu A, Hirai K, Ookawara S, Ishibashi K, Morishita Y. miR-122-5p Regulates Renal Fibrosis In Vivo. Int J Mol Sci 2022; 23:ijms232315423. [PMID: 36499744 PMCID: PMC9736395 DOI: 10.3390/ijms232315423] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/01/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
The role of exogenous microRNAs (miRNAs) in renal fibrosis is poorly understood. Here, the effect of exogenous miRNAs on renal fibrosis was investigated using a renal fibrosis mouse model generated by unilateral ureteral obstruction (UUO). miRNA microarray analysis and quantitative reverse-transcription polymerase chain reaction showed that miR-122-5p was the most downregulated (0.28-fold) miRNA in the kidneys of UUO mice. The injection of an miR-122-5p mimic promoted renal fibrosis and upregulated COL1A2 and FN1, whereas an miR-122-5p inhibitor suppressed renal fibrosis and downregulated COL1A2 and FN1. The expression levels of fibrosis-related mRNAs, which were predicted targets of miR-122-5p, were evaluated. The expression level of TGFBR2, a pro-fibrotic mRNA, was upregulated by the miR-122-5p mimic, and the expression level of FOXO3, an anti-fibrotic mRNA, was upregulated by the miR-122-5p inhibitor. The protein expressions of TGFBR2 and FOXO3 were confirmed by immunohistochemistry. Additionally, the expression levels of LC3, downstream anti-fibrotic mRNAs of FOXO3, were upregulated by the miR-122-5p inhibitor. These results suggest that miR-122-5p has critical roles in renal fibrosis.
Collapse
Affiliation(s)
- Shohei Kaneko
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Katsunori Yanai
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Hiroki Ishii
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Akinori Aomatsu
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
- Division of Intensive Care Unit, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Keiji Hirai
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Susumu Ookawara
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Kenichi Ishibashi
- Department of Medical Physiology, Meiji Pharmaceutical University, Tokyo 204-8588, Japan
| | - Yoshiyuki Morishita
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
- Correspondence:
| |
Collapse
|
14
|
Chen Y, Yang A, Hou Y, Liu L, Lin J, Huang X, Li J, Liu X, Lu F, Lin Q, Yang H, Yue S, Jiang S, Wang L, Zou C. Comparison between outcomes of IgA nephropathy with nephrotic-range proteinuria and nephrotic syndrome: do podocytes play a role? Ren Fail 2022; 44:1443-1453. [PMID: 36017686 PMCID: PMC9423834 DOI: 10.1080/0886022x.2022.2113796] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background Nephrotic syndrome (NS) and nephrotic-range proteinuria (NRP) are uncommon in IgA nephropathy (IgAN), and their clinicopathology and prognosis have not been discussed. Podocytes may play an important role in both clinical phenotypes. Methods We investigated 119 biopsy-proven IgAN patients with proteinuria over 2 g/d. The patients were divided into three groups according to proteinuria level: the overt proteinuria (OP) group, NS group, and NRP group. In addition, according to the severity of foot process effacement (FPE), the patients were divided into three groups: the segmental FPE (SFPE) group, moderate FPE (MFPE) group, and diffuse FPE (DFPE) group. The outcome was survival from a combined event defined by a doubling of the baseline serum creatinine and a 50% reduction in eGFR or ESRD. Results Compared with the NRP group, patients in the NS group had more severe microscopic hematuria, presented with more severe endocapillary hypercellularity and had a higher percentage of DFPE. The Kaplan–Meier curve showed that MFPE patients had a better outcome in the NRP group <50% of tubular atrophy/interstitial fibrosis. In the multivariate model, the NRP group (HR = 17.098, 95% CI = 3.835–76.224) was associated with an increased risk of the combined event, while MFPE (HR = 0.260, 95% CI = 0.078–0.864; p = 0.028) was associated with a reduced risk of the combined event. After the addition of renin-angiotensin system inhibitors (RASi), the incidence of the combined event in the MFPE group (HR = 0.179, 95% CI = 0.047–0.689; p = 0.012) was further reduced. Conclusions NS presented more active lesions and more severe FPE in IgAN. NRP was an independent risk factor for progression to the renal endpoint, while MFPE indicated a better prognosis in NRP without obvious chronic renal lesions, which may benefit from RASi.
Collapse
Affiliation(s)
- Yizhen Chen
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Aicheng Yang
- Department of Nephrology, The Affiliated Jiangmen TCM Hospital of Jinan University, Jiangmen, China
| | - Yuansheng Hou
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Longhui Liu
- Department of Nephrology, The Affiliated Jiangmen TCM Hospital of Jinan University, Jiangmen, China
| | - Jiehua Lin
- Department of Nephrology, The Affiliated Jiangmen TCM Hospital of Jinan University, Jiangmen, China
| | - Xiaodan Huang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jundu Li
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xusheng Liu
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fuhua Lu
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qizhan Lin
- Department of Hemodialysis, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haifeng Yang
- Department of Pathology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuling Yue
- Guangzhou Kingmed Diagnostic Laboratory Ltd, Guangzhou, China
| | - Shujun Jiang
- Department of Hemodialysis, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lixin Wang
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuan Zou
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
15
|
Packer M. Critical Reanalysis of the Mechanisms Underlying the Cardiorenal Benefits of SGLT2 Inhibitors and Reaffirmation of the Nutrient Deprivation Signaling/Autophagy Hypothesis. Circulation 2022; 146:1383-1405. [PMID: 36315602 PMCID: PMC9624240 DOI: 10.1161/circulationaha.122.061732] [Citation(s) in RCA: 231] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/10/2022] [Indexed: 02/06/2023]
Abstract
SGLT2 (sodium-glucose cotransporter 2) inhibitors produce a distinctive pattern of benefits on the evolution and progression of cardiomyopathy and nephropathy, which is characterized by a reduction in oxidative and endoplasmic reticulum stress, restoration of mitochondrial health and enhanced mitochondrial biogenesis, a decrease in proinflammatory and profibrotic pathways, and preservation of cellular and organ integrity and viability. A substantial body of evidence indicates that this characteristic pattern of responses can be explained by the action of SGLT2 inhibitors to promote cellular housekeeping by enhancing autophagic flux, an effect that may be related to the action of these drugs to produce simultaneous upregulation of nutrient deprivation signaling and downregulation of nutrient surplus signaling, as manifested by an increase in the expression and activity of AMPK (adenosine monophosphate-activated protein kinase), SIRT1 (sirtuin 1), SIRT3 (sirtuin 3), SIRT6 (sirtuin 6), and PGC1-α (peroxisome proliferator-activated receptor γ coactivator 1-α) and decreased activation of mTOR (mammalian target of rapamycin). The distinctive pattern of cardioprotective and renoprotective effects of SGLT2 inhibitors is abolished by specific inhibition or knockdown of autophagy, AMPK, and sirtuins. In the clinical setting, the pattern of differentially increased proteins identified in proteomics analyses of blood collected in randomized trials is consistent with these findings. Clinical studies have also shown that SGLT2 inhibitors promote gluconeogenesis, ketogenesis, and erythrocytosis and reduce uricemia, the hallmarks of nutrient deprivation signaling and the principal statistical mediators of the ability of SGLT2 inhibitors to reduce the risk of heart failure and serious renal events. The action of SGLT2 inhibitors to augment autophagic flux is seen in isolated cells and tissues that do not express SGLT2 and are not exposed to changes in environmental glucose or ketones and may be related to an ability of these drugs to bind directly to sirtuins or mTOR. Changes in renal or cardiovascular physiology or metabolism cannot explain the benefits of SGLT2 inhibitors either experimentally or clinically. The direct molecular effects of SGLT2 inhibitors in isolated cells are consistent with the concept that SGLT2 acts as a nutrient surplus sensor, and thus, its inhibition causes enhanced nutrient deprivation signaling and its attendant cytoprotective effects, which can be abolished by specific inhibition or knockdown of AMPK, sirtuins, and autophagic flux.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Dallas, TX. Imperial College, London, United Kingdom
| |
Collapse
|
16
|
Navarro-Betancourt JR, Cybulsky AV. The IRE1α pathway in glomerular diseases: The unfolded protein response and beyond. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:971247. [PMID: 39086958 PMCID: PMC11285563 DOI: 10.3389/fmmed.2022.971247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/07/2022] [Indexed: 08/02/2024]
Abstract
Endoplasmic reticulum (ER) function is vital for protein homeostasis ("proteostasis"). Protein misfolding in the ER of podocytes (glomerular visceral epithelial cells) is an important contributor to the pathogenesis of human glomerular diseases. ER protein misfolding causes ER stress and activates a compensatory signaling network called the unfolded protein response (UPR). Disruption of the UPR, in particular deletion of the UPR transducer, inositol-requiring enzyme 1α (IRE1α) in mouse podocytes leads to podocyte injury and albuminuria in aging, and exacerbates injury in glomerulonephritis. The UPR may interact in a coordinated manner with autophagy to relieve protein misfolding and its consequences. Recent studies have identified novel downstream targets of IRE1α, which provide new mechanistic insights into proteostatic pathways. Novel pathways of IRE1α signaling involve reticulophagy, mitochondria, metabolism, vesicular trafficking, microRNAs, and others. Mechanism-based therapies for glomerulopathies are limited, and development of non-invasive ER stress biomarkers, as well as targeting ER stress with pharmacological compounds may represent a therapeutic opportunity for preventing or attenuating progression of chronic kidney disease.
Collapse
Affiliation(s)
| | - Andrey V. Cybulsky
- Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
17
|
Tang L, Cai Z, Wang SX, Zhao WJ. Transition from minimal change disease to focal segmental glomerulosclerosis related to occupational exposure: A case report. World J Clin Cases 2022; 10:5861-5868. [PMID: 35979127 PMCID: PMC9258360 DOI: 10.12998/wjcc.v10.i17.5861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/21/2022] [Accepted: 04/24/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Although minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS) have been described as two separate forms of nephrotic syndrome (NS), they are not completely independent. We report a case of a patient transitioning from MCD to FSGS, review the literature, and explore the relationship between the two diseases.
CASE SUMMARY A 42-year-old male welder, presenting with lower extremity edema and elevated serum creatinine, was diagnosed with NS and end-stage kidney disease (ESKD) based on laboratory test results. The patient had undergone a kidney biopsy for NS 20 years previously, which indicated MCD, and a second recent kidney biopsy suggested FSGS. The patient was an electric welder with excessive levels of cadmium and lead in his blood. Consequently, we suspect that his aggravated pathology and occurrence of ESKD were related to metal nephrotoxicity. The patient eventually received kidney replacement therapy and quit his job which involved long-term exposure to metals. During the 1-year follow-up period, the patient was negative for metal elements in the blood and urine and recovered partial kidney function.
CONCLUSION MCD and FSGS may be different stages of the same disease. The transition from MCD to FSGS in this case indicates disease progression, which may be related to excessive metal contaminants caused by the patient’s occupation.
Collapse
Affiliation(s)
- Long Tang
- Department of Nephrology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Zhen Cai
- Department of Nephrology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Su-Xia Wang
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Peking University Institute of Nephrology, Beijing 100034, China
| | - Wen-Jing Zhao
- Department of Nephrology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| |
Collapse
|
18
|
Tomita N, Hotta Y, Naiki-Ito A, Sanagawa A, Kataoka T, Furukawa-Hibi Y, Takahashi S, Kimura K. Protective effects of tadalafil on damaged podocytes in an adriamycin-induced nephrotic syndrome model. J Pharmacol Sci 2022; 149:53-59. [DOI: 10.1016/j.jphs.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/21/2022] [Accepted: 03/18/2022] [Indexed: 10/18/2022] Open
|
19
|
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
|
20
|
Zhang S, Fan Q, Moktefi A, Ory V, Audard V, Pawlak A, Ollero M, Sahali D, Henique C. CMIP interacts with WT1 and targets it on the proteasome degradation pathway. Clin Transl Med 2021; 11:e460. [PMID: 34323419 PMCID: PMC8299046 DOI: 10.1002/ctm2.460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/29/2021] [Accepted: 05/25/2021] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND The Wilms tumor 1 suppressor gene, WT1, is expressed throughout life in podocytes and is essential for their function. Downregulation of WT1 has been reported in podocyte diseases but the underlying mechanisms remain unclear. Podocyte injury is the hallmark of idiopathic nephrotic syndrome (INS), the most frequent glomerular disease in children and young adults. An increase in the abundance of Cmaf-inducing protein (CMIP) has been found to alter podocyte function, but it is not known whether CMIP affects WT1 expression. METHODS Transcriptional and post-transcriptional regulation of WT1in the presence of CMIP was studied using transient transfection, mouse models, and siRNA handling. RESULTS We showed that overproduction of CMIP in the podocyte was consistently associated with a downregulation of WT1 according to two mechanisms. We found that CMIP prevented the NF-kB-mediated transcriptional activation of WT1. We demonstrated that CMIP interacts directly with WT1 through its leucine-rich repeat domain. Overexpression of CMIP in the M15 cell line induced a downregulation of WT1, which was prevented by lactacystin, a potent proteasome inhibitor. We showed that CMIP exhibits an E3 ligase activity and targets WT1 to proteasome degradation. Intravenous injection of Cmip-siRNA specifically prevented the repression of Wt1 in lipopolysaccharides-induced proteinuria in mice. CONCLUSIONS These data suggest that CMIP is a repressor of WT1 and might be a critical player in the pathophysiology of some podocyte diseases. Because WT1 is required for podocyte integrity, CMIP could be considered a therapeutic target in podocyte diseases.
Collapse
Affiliation(s)
- Shao‐Yu Zhang
- INSERMCreteilFrance
- Faculté de santéUniversité Paris Est CreteilCreteilFrance
| | - Qingfeng Fan
- INSERMCreteilFrance
- Faculté de santéUniversité Paris Est CreteilCreteilFrance
| | - Anissa Moktefi
- INSERMCreteilFrance
- Faculté de santéUniversité Paris Est CreteilCreteilFrance
- AP‐HPGroupe hospitalier Henri Mondor‐Albert ChenevierDépartement de pathologieCreteilFrance
| | - Virginie Ory
- INSERMCreteilFrance
- Faculté de santéUniversité Paris Est CreteilCreteilFrance
| | - Vincent Audard
- INSERMCreteilFrance
- Faculté de santéUniversité Paris Est CreteilCreteilFrance
- AP‐HPGroupe Henri‐Mondor Albert‐ChenevierService de NéphrologieCreteilFrance
| | - Andre Pawlak
- INSERMCreteilFrance
- Faculté de santéUniversité Paris Est CreteilCreteilFrance
| | - Mario Ollero
- INSERMCreteilFrance
- Faculté de santéUniversité Paris Est CreteilCreteilFrance
| | - Dil Sahali
- INSERMCreteilFrance
- Faculté de santéUniversité Paris Est CreteilCreteilFrance
- AP‐HPGroupe Henri‐Mondor Albert‐ChenevierService de NéphrologieCreteilFrance
| | - Carole Henique
- INSERMCreteilFrance
- Faculté de santéUniversité Paris Est CreteilCreteilFrance
| |
Collapse
|