1
|
Turinsky L, Rehrl S, Nguyen C, Benyahia S, Kuperwasser N, Vasseur F, Arevalo AR, Rabant M, Goudin N, Mesnard L, Canaud G, Pontoglio M, Isnard P, Terzi F. Podocyte-specific YAP activation triggers crescentic glomerulonephritis through cross-talk with parietal epithelial cells. Sci Transl Med 2025; 17:eadq7825. [PMID: 40560996 DOI: 10.1126/scitranslmed.adq7825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 12/22/2024] [Accepted: 06/04/2025] [Indexed: 06/28/2025]
Abstract
Crescentic glomerulonephritis (cGN) is a severe kidney pathology characterized by the aberrant proliferation of epithelial cells, leading to crescent formation within the Bowman's space. The molecular pathways involved in crescent formation remain poorly understood despite its clinical relevance. Given the mechanical stress experienced by podocytes, likely exacerbated in cGN, we hypothesized that activation of the mechanosensor yes-associated protein 1 (YAP), an effector of the Hippo pathway, may contribute to the development of cGN. Here, we demonstrate activation of YAP and its target genes in the nephrotoxic nephritis model, a murine model of cGN. Mechanistically, podocyte-specific hyperactivation of YAP (YAP5SA) in transgenic mice led to cell-autonomous hypertrophy of podocytes and non-cell-autonomous activation and proliferation of parietal epithelial cells (PECs), culminating in crescents. Transcriptomic profiling in a human podocyte cell line expressing the same YAP5SA isoform revealed the reactivation of developmental programs within differentiated podocytes and identified the phosphoinositide 3-kinase-protein kinase B-mechanistic target of rapamycin (PI3K-Akt-mTOR) signaling pathway as a candidate involved in YAP-induced podocyte hypertrophy. Furthermore, this analysis identified connective tissue growth factor (CTGF) and heparin-binding epidermal growth factor-like growth factor (HB-EGF) as potential mediators in the cross-talk between YAP-activated podocytes and PECs, driving PEC hyperplasia. Collectively, these findings highlight the pivotal role of YAP in the pathogenesis of cGN and indicate that targeting YAP signaling could be a promising therapeutic strategy for this severe kidney disease.
Collapse
Affiliation(s)
- Lisa Turinsky
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades (INEM), 75015 Paris, France
| | - Sonja Rehrl
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades (INEM), 75015 Paris, France
| | - Clément Nguyen
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades (INEM), 75015 Paris, France
| | - Siham Benyahia
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades (INEM), 75015 Paris, France
| | | | - Florence Vasseur
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades (INEM), 75015 Paris, France
| | - Alis Restrepo Arevalo
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades (INEM), 75015 Paris, France
| | - Marion Rabant
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades (INEM), 75015 Paris, France
- Laboratoire d'Anatomie et Cytologie Pathologiques, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants Malades, 75015 Paris, France
| | - Nicolas Goudin
- Structure Fédérative de Recherche Necker, US24-UMS3633, 75015 Paris, France
| | - Laurent Mesnard
- Sorbonne Université, INSERM UMR1155, Hôpital Tenon, 75020 Paris, France
| | - Guillaume Canaud
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades (INEM), 75015 Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Marco Pontoglio
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades (INEM), 75015 Paris, France
| | - Pierre Isnard
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades (INEM), 75015 Paris, France
- Laboratoire d'Anatomie et Cytologie Pathologiques, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants Malades, 75015 Paris, France
| | - Fabiola Terzi
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades (INEM), 75015 Paris, France
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Srivastava SP, Kopasz-Gemmen O, Kunamneni A, Thurnman A, Ozukan E, Swaroop V, Yoshida S, Hong S, Inoki K. AMPK is dispensable for physiological podocyte and glomerular functions but prevents glomerular fibrosis in experimental diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.647592. [PMID: 40291739 PMCID: PMC12026990 DOI: 10.1101/2025.04.07.647592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
AMP-activated protein kinase (AMPK) has been postulated to be crucial in regulating various renal physiology and pathophysiology processes, including energy metabolism, ion and water transport, inflammation, and hypertrophy. However, the specific roles of AMPK in the podocyte, a cell critical for maintaining glomerular filtration, have not been fully explored using genetic model animals. In this study, we generated mice lacking both AMPK α1 and α2 catalytic subunits in glomerular podocytes (pmut). Our findings revealed that, surprisingly, AMPK is dispensable for normal podocyte function. These knockout mice could live as long as their wild-type littermates without showing any pathological alterations in their glomeruli or glomerular function at two years of age. However, under type 1 diabetic conditions, the diabetic pmut mice exhibited increased lipid and collagen accumulation and an elevated expression of mesenchymal proteins in their glomeruli. They also showed more significant albuminuria compared to control diabetic mice. Under high glucose culture conditions, glomeruli isolated from pmut mice demonstrated a reduced expression of mitochondrial genes (e.g., Ndufv2) and increased leakage of mitochondrial components. Additionally, there was heightened expression of genes associated with nucleotide sensing and pro-inflammatory pathways (including mb21d2, IL-1 beta, and NF-kB). These observations suggest that while AMPK is not necessary for podocyte function in healthy kidneys, it is crucial for preventing glomerular fibrosis resulting from lipotoxicity and inflammation under diabetic conditions.
Collapse
|
5
|
Shi J, Liu X, Jiao Y, Tian J, An J, Zou G, Zhuo L. mTOR pathway: A key player in diabetic nephropathy progression and therapeutic targets. Genes Dis 2025; 12:101260. [PMID: 39717716 PMCID: PMC11665407 DOI: 10.1016/j.gendis.2024.101260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/26/2024] [Accepted: 02/21/2024] [Indexed: 12/25/2024] Open
Abstract
Diabetic nephropathy is a prevalent complication of diabetes and stands as the primary contributor to end-stage renal disease. The global prevalence of diabetic nephropathy is on the rise, however, due to its intricate pathogenesis, there is currently an absence of efficacious treatments to enhance renal prognosis in affected patients. The mammalian target of rapamycin (mTOR), a serine/threonine protease, assumes a pivotal role in cellular division, survival, apoptosis delay, and angiogenesis. It is implicated in diverse signaling pathways and has been observed to partake in the progression of diabetic nephropathy by inhibiting autophagy, promoting inflammation, and increasing oxidative stress. In this academic review, we have consolidated the understanding of the pathological mechanisms associated with four distinct resident renal cell types (podocytes, glomerular mesangial cells, renal tubular epithelial cells, and glomerular endothelial cells), as well as macrophages and T lymphocytes, within a diabetic environment. Additionally, we highlight the research progress in the treatment of diabetic nephropathy with drugs and various molecules interfering with the mTOR signaling pathway, providing a theoretical reference for the treatment and prevention of diabetic nephropathy.
Collapse
Affiliation(s)
- Jingxuan Shi
- Department of Nephrology, China-Japan Friendship Hospital, Beijing 100029, China
- China-Japan Friendship Institute of Clinical Medical Sciences, Beijing 100029, China
| | - Xinze Liu
- Beijing University of Chinese Medicine China-Japan Friendship Clinical Medical College, Beijing 100029, China
| | - Yuanyuan Jiao
- Department of Nephrology, Fuwai Hospital, Chinese Academy of Medical Science, Beijing 100037, China
| | - Jingwei Tian
- Department of Nephrology, Beijing Sixth Hospital, Beijing 100007, China
- Capital Medical University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Jiaqi An
- Department of Nephrology, China-Japan Friendship Hospital, Beijing 100029, China
- China-Japan Friendship Clinic Medical College, Peking University, Beijing 100191, China
| | - Guming Zou
- Department of Nephrology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Li Zhuo
- Department of Nephrology, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
6
|
Fogo AB, Harris RC. Crosstalk between glomeruli and tubules. Nat Rev Nephrol 2025; 21:189-199. [PMID: 39643696 DOI: 10.1038/s41581-024-00907-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2024] [Indexed: 12/09/2024]
Abstract
Models of kidney injury have classically concentrated on glomeruli as the primary site of injury leading to glomerulosclerosis or on tubules as the primary site of injury leading to tubulointerstitial fibrosis. However, current evidence on the mechanisms of progression of chronic kidney disease indicates that a complex interplay between glomeruli and tubules underlies progressive kidney injury. Primary glomerular injury can clearly lead to subsequent tubule injury. For example, damage to the glomerular filtration barrier can expose tubular cells to serum proteins, including complement and cytokines, that would not be present in physiological conditions and can promote the development of tubulointerstitial fibrosis and progressive decline in kidney function. In addition, although less well-studied, increasing evidence suggests that tubule injury, whether primary or secondary, can also promote glomerular damage. This feedback from the tubule to the glomerulus might be mediated by changes in the reabsorptive capacity of the tubule, which can affect the glomerular filtration rate, or by mediators released by injured proximal tubular cells that can induce damage in both podocytes and parietal epithelial cells. Examining the crosstalk between the various compartments of the kidney is important for understanding the mechanisms underlying kidney pathology and identifying potential therapeutic interventions.
Collapse
Affiliation(s)
- Agnes B Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Raymond C Harris
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Tennessee Department of Veterans Affairs, Nashville, TN, USA.
| |
Collapse
|
7
|
Lv J, Yu H, Du S, Xu P, Zhao Y, Qi W, Wang X. Targeting endoplasmic reticulum stress: an innovative therapeutic strategy for podocyte-related kidney diseases. J Transl Med 2025; 23:95. [PMID: 39838496 PMCID: PMC11752968 DOI: 10.1186/s12967-025-06076-3] [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: 09/29/2024] [Accepted: 01/03/2025] [Indexed: 01/23/2025] Open
Abstract
The endoplasmic reticulum (ER) is a vital organelle responsible for protein quality control, including the folding, modification, and transport of proteins. When misfolded or unfolded proteins accumulate in the ER, it triggers endoplasmic reticulum stress (ERS) and activates the unfolded protein response (UPR) to restore ER homeostasis. However, prolonged or excessive ERS can lead to apoptosis. The kidneys play a crucial role in maintaining physiological functions by excreting metabolic waste, regulating blood volume, balancing electrolytes and acid-base levels, and secreting various bioactive substances. Podocytes, epithelial cells situated outside the glomerular basement membrane, are essential for maintaining the structural integrity and permeability of the glomerular filtration barrier. Previous studies have shown that ERS in podocytes can contribute to the development of diseases such as glomerulonephritis, hereditary nephropathy, and diabetic kidney disease, potentially progressing to end-stage renal disease and causing patient mortality. As such, investigating ERS in podocytes has become a key area of focus in kidney disease research. This study examines recent advancements in understanding the effects of excessive ERS on podocytes across various kidney diseases, highlights the role of podocyte ERS in disease progression, and explores the potential therapeutic benefits of targeting the UPR to manage ERS in kidney diseases, thereby providing a scientific basis for clinical interventions.
Collapse
Affiliation(s)
- Jiao Lv
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Honghai Yu
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Sasa Du
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Pengyu Xu
- College of Acupuncture and Moxibustion, Changchun University of Traditional Chinese Medicine, Changchun, 130117, China
| | - Yunyun Zhao
- Endocrinology Department, First Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Wenxiu Qi
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Xiuge Wang
- Endocrinology Department, First Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, 130021, China.
| |
Collapse
|
8
|
Liu F, Xu Z, Chen G, Xu X, Cao H, Chen J. Evolutionary patterns and research frontiers in autophagy in podocytopathies: a bibliometric analysis. Front Med (Lausanne) 2025; 11:1445550. [PMID: 39850100 PMCID: PMC11754056 DOI: 10.3389/fmed.2024.1445550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Introduction Podocytopathies are a uniquely renal disease syndrome, in which direct or indirect podocyte injury leads to proteinuria or nephrotic syndrome. Of the many factors that contribute to podocytopathies, the abnormal regulation of autophagy, such insufficient or excessive autophagy levels, have been proposed to play a significant role in the occurrence and development of podocytopathies. However, there still has been a lack of systematic and comparative research to elucidate exact role of autophagy in podocytopathies and its current research status. This study aims to utilize bibliometric analysis to clarify the role of autophagy in the pathogenesis of podocytopathies, analyze the research focus in this area, as well as explore the future research trends. Methods We retrieved original articles and review papers with respect to autophagy in podocytopathies research published between the year 2008 and 2022 from the Web of Science Core Collection (WOSCC). Then, VOSviewer and CiteSpace software were employed to reveal the leading subjects and generate visual maps of countries/regions, organizations, authors, journals, and keyword networks in this field. Results and discussion A total of 825 publications regarding autophagy in podocytopathies published between 2008 and 2022 were included, with China contributing the most followed by the United States and Japan. Professor Koya Daisuke, Professor He Qiang, and Professor Jin Juan are the most prolific researchers in this field. Oxidative stress, the NLRP3 inflammasome, and therapeutic targets were the knowledge base for the research in this special field. Taken together, this bibliometric analysis helps us reveal the current research hotspots and guide future research directions, which provides a reference for scholars to further investigate the role of autophagy in podocytopathies as well as conduct clinical trial with autophagy regulators in podocytopathies.
Collapse
Affiliation(s)
- Feng Liu
- Department of Nephrology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Ziyu Xu
- Department of Nephrology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Gaijie Chen
- Health Management Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojing Xu
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Huixia Cao
- Department of Nephrology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiefang Chen
- Department of Neurology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Lee HJ, Sun Y, Das F, Ju W, Nair V, Kevil CG, Varadarajan S, Zhang G, Ghosh Choudhury G, Singh BB, Kretzler M, Nelson RG, Sharma K, Kasinath BS. The CLCA1/TMEM16A/Cl- current axis associates with H2S deficiency in diabetic kidney injury. JCI Insight 2025; 10:e174848. [PMID: 39782685 PMCID: PMC11721299 DOI: 10.1172/jci.insight.174848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/13/2024] [Indexed: 01/30/2025] Open
Abstract
The role played by anionic channels in diabetic kidney disease (DKD) is not known. Chloride channel accessory 1 (CLCA1) facilitates the activity of TMEM16A (Anoctamin-1), a Ca2+-dependent Cl- channel. We examined if CLCA1/TMEM16A had a role in DKD. In mice with type 2 diabetes, renal cortical CLCA1 and TMEM16A content was increased. CLCA1 and TMEM16A content was associated with hydrogen sulfide (H2S) deficiency, mTOR complex 1 (mTORC1) activation, albuminuria, and matrix increase. Administering sodium hydrosulfide (NaHS), a source of H2S, mitigated these changes. In proximal tubular epithelial (MCT) cells, high glucose rapidly increased CLCA1 by recruiting the IL-6/STAT3 axis and augmented TMEM16A expression by stimulating its mRNA translation; these changes were abolished by NaHS. Patch clamp experiments showed that high glucose increased Cl- current in MCT cells that was ameliorated by NaHS and a TMEM16A chemical inhibitor. siRNA against CLCA1 or TMEM16A and TMEM16A inhibitor abolished high glucose-induced mTORC1 activation and matrix protein increase. Tubular expression of TMEM16A correlated with albuminuria in kidney biopsies from people with type 2 diabetes. We report a pathway for DKD in which H2S deficiency results in kidney injury by the recruitment of the CLCA1/TMEM16A/Cl- current system.
Collapse
Affiliation(s)
- Hak Joo Lee
- Center for Precision Medicine, Department of Medicine, and
| | - Yuyang Sun
- Department of Periodontics, University of Texas Health, San Antonio, San Antonio, Texas, USA
| | - Falguni Das
- Center for Precision Medicine, Department of Medicine, and
| | - Wenjun Ju
- Department of Internal Medicine and
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Christopher G. Kevil
- Department of Pathology, Louisiana State University Health Science Center, Shreveport, Louisiana, USA
| | - Shankara Varadarajan
- Department of Periodontics, University of Texas Health, San Antonio, San Antonio, Texas, USA
| | - Guanshi Zhang
- Center for Precision Medicine, Department of Medicine, and
| | - Goutam Ghosh Choudhury
- Center for Precision Medicine, Department of Medicine, and
- Research service and
- Geriatric Research Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Brij B. Singh
- Department of Periodontics, University of Texas Health, San Antonio, San Antonio, Texas, USA
| | - Matthias Kretzler
- Department of Internal Medicine and
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Robert G. Nelson
- National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
- Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Kumar Sharma
- Center for Precision Medicine, Department of Medicine, and
- Research service and
| | - Balakuntalam S. Kasinath
- Center for Precision Medicine, Department of Medicine, and
- Research service and
- Geriatric Research Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, USA
| |
Collapse
|
10
|
Junli W, Zhihong H, Lina W, Qiaoqun O, Jing Q, Jiaqi H, Yu S. Research hotspots and frontiers of endoplasmic reticulum in glomerular podocytes: a bibliometric and visual analysis from 2005 to 2023. Front Pharmacol 2025; 15:1488340. [PMID: 39840101 PMCID: PMC11747773 DOI: 10.3389/fphar.2024.1488340] [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: 08/29/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Background The glomerular podocyte endoplasmic reticulum is a critical component in renal function, yet its research landscape is not fully understood. This study aims to map the existing research on podocyte endoplasmic reticulum by analyzing publications in the Web of Science Core Collection (WOSCC) from the past 19 years. Methods We conducted a bibliometric analysis using Citespace, VOSviewer, the Metrology Literature Online platform, and the Bibliometrix software package to visualize and interpret the data from WOSCC. The analysis focused on publication volume, authorship, institutional contributions, and research trends. Results The analysis revealed a significant growth in publications, indicating a surge in interest in podocyte endoplasmic reticulum research. Cybulsky, Andrey V, and Papillon, Joan emerge as the most prolific authors, and the Journal of the American Society of Nephrology is the leading journal in this field. China is the top contributor in terms of publications, with McGill University being the most productive institution. The research primarily focuses on endoplasmic reticulum stress, diabetic nephropathy, and apoptosis, with emerging trends in "foot cell apoptosis," "cell signaling pathways," and "autophagy." Conclusion The findings highlight the expanding scope of podocyte endoplasmic reticulum research, with a particular emphasis on the mechanisms of endoplasmic reticulum stress and podocyte apoptosis. Future research directions may include the identification of specific therapeutic targets, detailed exploration of podocyte signaling pathways, and the role of autophagy. This study provides a comprehensive overview of the major research areas, frontiers, and trends in podocyte endoplasmic reticulum research, which are pivotal for guiding future investigations.
Collapse
Affiliation(s)
- Wang Junli
- Department of Pediatrics, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Hao Zhihong
- Department of Pediatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Wang Lina
- Department of Pediatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Ou Qiaoqun
- Department of Pediatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Qu Jing
- Department of Pediatrics, Guangzhou First People’s Hospital, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Hu Jiaqi
- Department of Pediatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Shengyou Yu
- Department of Pediatrics, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
- Department of Pediatrics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Pediatrics, Guangzhou First People’s Hospital, Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
11
|
Chatterjee A, Tumarin J, Prabhakar S. Cellular cross-talk drives mesenchymal transdifferentiation in diabetic kidney disease. Front Med (Lausanne) 2025; 11:1499473. [PMID: 39839616 PMCID: PMC11747801 DOI: 10.3389/fmed.2024.1499473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
While changes in glomerular function and structure may herald diabetic kidney disease (DKD), many studies have underscored the significance of tubule-interstitial changes in the progression of DKD. Indeed, tubule-interstitial fibrosis may be the most important determinant of progression of DKD as in many forms of chronic glomerulopathies. The mechanisms underlying the effects of tubular changes on glomerular function in DKD have intrigued many investigators, and therefore, the signaling mechanisms underlying the cross-talk between tubular cells and glomerular cells have been the focus of investigation in many recent studies. Additionally, the observations of slowing of glomerular filtration rate (GFR) decline and reduction of proteinuria by recent drugs such as SGLT-2 blockers, whose primary mechanism of action is on proximal tubules, further strengthen the concept of cross-talk between the tubular and glomerular cells. Recently, the focus of research on the pathogenesis of DKD has primarily centered around exploring the cross-talk between various signaling pathways in the diabetic kidney as well as cross-talk between tubular and glomerular endothelial cells and podocytes with special relevance to epithelial-to-mesenchymal transition (EMT) and endothelial-to-mesenchymal transition (EndoMT). The focus of this review is to provide a general description of cell-to-cell cross-talk in the diabetic kidney and to highlight these concepts with evidence in relation to the physiology and pathophysiology of DKD.
Collapse
Affiliation(s)
| | | | - Sharma Prabhakar
- Department of Internal Medicine, Texas Tech University Health Sciences Centre, Lubbock, TX, United States
| |
Collapse
|
12
|
Elliott J, Oyama MA. Sodium glucose transporter 2 inhibitors: Will these drugs benefit non-diabetic veterinary patients with cardiac and kidney diseases? J Vet Pharmacol Ther 2025; 48 Suppl 1:1-18. [PMID: 39001645 PMCID: PMC11737021 DOI: 10.1111/jvp.13472] [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: 03/24/2024] [Revised: 06/14/2024] [Accepted: 07/03/2024] [Indexed: 01/18/2025]
Abstract
Sodium glucose transporter type 2 (SGLT2) inhibitors have been introduced into human medicine where their beneficial effects go beyond the expected improvement in blood glucose control. These drugs appear to prevent progression of both cardiovascular and kidney diseases, not only in diabetic but also in non-diabetic human patients. As these drugs have received conditional approval for use in diabetic cats and are being used in other veterinary species, the intriguing question as to whether they will have similar cardioprotective and nephroprotective effects in dogs and cats is being asked. The primary mechanism(s) by which SGLT2 inhibitors are cardio- and nephroprotective remain to be fully characterized. This paper reviews these suggested mechanisms in the context of the pathophysiology of progressive cardiovascular and kidney diseases in dogs and cats with the goal of predicting which categories of non-diabetic veterinary patients these drugs might be of most benefit.
Collapse
Affiliation(s)
- Jonathan Elliott
- Department of Comparative Biomedical SciencesRoyal Veterinary College, University of LondonLondonUK
| | - Mark A. Oyama
- Department of Clinical Sciences & Advanced MedicineUniversity of Pennsylvania School of Veterinary MedicinePhiladelphiaPennsylvaniaUSA
| |
Collapse
|
13
|
Du Y, Wu M, Song S, Bian Y, Shi Y. TXNIP deficiency attenuates renal fibrosis by modulating mTORC1/TFEB-mediated autophagy in diabetic kidney disease. Ren Fail 2024; 46:2338933. [PMID: 38616177 PMCID: PMC11018024 DOI: 10.1080/0886022x.2024.2338933] [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: 04/19/2023] [Accepted: 03/30/2024] [Indexed: 04/16/2024] Open
Abstract
Thioredoxin-interacting protein (TXNIP) is an important regulatory protein for thioredoxin (TRX) that elicits the generation of reactive oxygen species (ROS) by inhibiting the redox function of TRX. Abundant evidence suggests that TXNIP is involved in the fibrotic process of diabetic kidney disease (DKD). However, the potential mechanism of TXNIP in DKD is not yet well understood. In this study, we found that TXNIP knockout suppressed renal fibrosis and activation of mammalian target of rapamycin complex 1 (mTORC1) and restored transcription factor EB (TFEB) and autophagy activation in diabetic kidneys. Simultaneously, TXNIP interference inhibited epithelial-to-mesenchymal transformation (EMT), collagen I and fibronectin expression, and mTORC1 activation, increased TFEB nuclear translocation, and promoted autophagy restoration in HK-2 cells exposed to high glucose (HG). Rapamycin, an inhibitor of mTORC1, increased TFEB nuclear translocation and autophagy in HK-2 cells under HG conditions. Moreover, the TFEB activators, curcumin analog C1 and trehalose, effectively restored HG-induced autophagy, and abrogated HG-induced EMT and collagen I and fibronectin expression in HK-2 cells. Taken together, these findings suggest that TXNIP deficiency ameliorates renal fibrosis by regulating mTORC1/TFEB-mediated autophagy in diabetic kidney diseases.
Collapse
Affiliation(s)
- Yunxia Du
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Ming Wu
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Shan Song
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Yawei Bian
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Yonghong Shi
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
14
|
Lu Q, Hu X, Hou Q, Yu L, Cao K, Ding D, Lu Y, Dai C. Rheb1 deficiency elicits mitochondrial dysfunction and accelerates podocyte senescence through promoting Atp5f1c acetylation. Cell Signal 2024; 124:111451. [PMID: 39389178 DOI: 10.1016/j.cellsig.2024.111451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
Podocyte senescence can cause persistent podocyte injury and albuminuria in diabetic kidney disease (DKD), but the mechanism remains obscure. In this study, podocyte senescence was confirmed by immunohistochemical staining in podocytes from patients and mice with DKD. Rheb1 knockout in podocytes aggravated podocyte senescence and injury in diabetic mice, but mitigated podocyte injury in mice with podocyte-specific mTORC1 activation induced by Tsc1 deletion. In cultured podocytes, Rheb1 knockdown remarkably accelerated podocyte senescence, independent of mTORC1. Mechanistically, PDH phosphorylation in podocyte was correlated with podocyte senescence in DKD patients. Rheb1 deficiency decreased ATP, mitochondrial membrane potential and partial components of respiratory chain complex, and enhanced ROS production and PDH phosphorylation, which indicates mitochondrial dysfunction, both in vitro and in vivo. Furthermore, Rheb1 interacted with Atp5f1c, and regulated its acetylation under a high-glucose condition. Together, Rheb1 deficiency elicits mitochondrial dysfunction and accelerates podocyte senescence through promoting Atp5f1c acetylation, in an mTORC1-independent manner, which provides experimental basis for the treatment of DKD.
Collapse
Affiliation(s)
- Qingmiao Lu
- Department of Endocrinology, 2(nd) Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan, Nanjing, Jiangsu, China
| | - Xiao Hu
- Department of Endocrinology, 2(nd) Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan, Nanjing, Jiangsu, China; Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, No. 666 Shengli Road, Nantong 226001, China
| | - Qing Hou
- Center for Kidney Disease, 2(nd) Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Long Yu
- Center for Kidney Disease, 2(nd) Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Kai Cao
- Center for Kidney Disease, 2(nd) Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Dafa Ding
- Department of Endocrinology, 2(nd) Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan, Nanjing, Jiangsu, China
| | - Yibing Lu
- Department of Endocrinology, 2(nd) Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan, Nanjing, Jiangsu, China.
| | - Chunsun Dai
- Center for Kidney Disease, 2(nd) Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China; Department of Clinical Genetics, 2(nd) Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China.
| |
Collapse
|
15
|
Xing J, Huang L, Ren W, Mei X. Risk factors for rapid kidney function decline in diabetes patients. Ren Fail 2024; 46:2398188. [PMID: 39258389 PMCID: PMC11391878 DOI: 10.1080/0886022x.2024.2398188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024] Open
Abstract
Diabetic nephropathy, as a severe microvascular complication of diabetes, manifests in four clinical types: classic, albuminuria regression, a rapid decline in kidney function (RDKF), and non-proteinuric or non-albuminuric DKD. Rapidly progressive diabetic nephropathy advances to end-stage renal disease more swiftly than the typical form, posing significant risks. However, a comprehensive understanding of rapidly progressive diabetic nephropathy is currently lacking. This article reviewed latest developments in genetic and clinical risk factors associated with rapidly progressive diabetic nephropathy, aiming to broad perspectives concerning the diagnosis and interventions of this condition.
Collapse
Affiliation(s)
- Jixin Xing
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Linxi Huang
- Department of Nephrology, PLA Navy No. 905 Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Weifu Ren
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xiaobin Mei
- Department of Nephrology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
16
|
Bomholt AB, Johansen CD, Galsgaard KD, Elmelund E, Winther-Sørensen M, Holst JJ, Wewer Albrechtsen NJ, Sørensen CM. Glucagon receptor activation contributes to the development of kidney injury. Am J Physiol Renal Physiol 2024; 327:F712-F724. [PMID: 39265079 PMCID: PMC11563637 DOI: 10.1152/ajprenal.00088.2024] [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: 03/18/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/14/2024] Open
Abstract
The underlying causes of diabetic kidney disease are still largely unknown. New insights into the contributing causes of diabetic nephropathy are important to prevent this complication. Hyperglycemia and hypertension are some of the risk factors for diabetic nephropathy. However, the incidence of diabetic nephropathy is increasing despite efforts to normalize blood glucose levels and blood pressure. Therefore, other factors should be investigated as causes of diabetic nephropathy. We investigated whether long-term increased plasma levels of glucagon contribute to the development of pathophysiological changes in kidney function as seen in patients with diabetic nephropathy. Using mouse models of chronic activation and inactivation of glucagon receptor signaling, we investigated whether glucagon is involved in changes in renal function, renal structure, and transcriptional changes. We found several histopathological changes in the kidney, such as thickening of the parietal layer of Bowman's capsule, glomerular mesangial cell expansion, and significant albuminuria in the mice with activated glucagon receptor signaling. Opposite effects on mesangial area expansion and the development of albuminuria were demonstrated in mice with glucagon receptor inactivation. RNA sequencing data revealed that transcription of genes related to fatty acid metabolism, podocytes, Na+-K+-ATPase, and sodium/glucose transport was significantly changed in mice with activated glucagon receptor signaling. These data implicate that glucagon receptor signaling is involved in the development of kidney injury, as seen in type 2 diabetes, and that glucagon receptor is a potential therapeutic target in the treatment of diabetes. NEW & NOTEWORTHY This study suggests that the glucagon receptor is a potential therapeutic target in the treatment of diabetic kidney disease. We show, in mice, that long-term treatment with a glucagon analog showed not only pathophysiological changes and changes in renal function but also transcriptional changes in the kidneys, whereas opposite effects were demonstrated in mice with glucagon receptor inactivation. Therefore, the use of glucagon in a treatment regimen requires investigation of possible metabolic and renal abnormalities.
Collapse
Affiliation(s)
| | - Christian Dall Johansen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Douglas Galsgaard
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emilie Elmelund
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital-Bispebjerg Hospital, Copenhagen, Denmark
| | | |
Collapse
|
17
|
Meliambro K, He JC, Campbell KN. Podocyte-targeted therapies - progress and future directions. Nat Rev Nephrol 2024; 20:643-658. [PMID: 38724717 DOI: 10.1038/s41581-024-00843-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2024] [Indexed: 09/14/2024]
Abstract
Podocytes are the key target cells for injury across the spectrum of primary and secondary proteinuric kidney disorders, which account for up to 90% of cases of kidney failure worldwide. Seminal experimental and clinical studies have established a causative link between podocyte depletion and the magnitude of proteinuria in progressive glomerular disease. However, no substantial advances have been made in glomerular disease therapies, and the standard of care for podocytopathies relies on repurposed immunosuppressive drugs. The past two decades have seen a remarkable expansion in understanding of the mechanistic basis of podocyte injury, with prospects increasing for precision-based treatment approaches. Dozens of disease-causing genes with roles in the pathogenesis of clinical podocytopathies have been identified, as well as a number of putative glomerular permeability factors. These achievements, together with the identification of novel targets of podocyte injury, the development of potential approaches to harness the endogenous podocyte regenerative potential of progenitor cell populations, ongoing clinical trials of podocyte-specific pharmacological agents and the development of podocyte-directed drug delivery systems, contribute to an optimistic outlook for the future of glomerular disease therapy.
Collapse
Affiliation(s)
- Kristin Meliambro
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John C He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kirk N Campbell
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
18
|
Swaroop V, Ozkan E, Herrmann L, Thurman A, Kopasz-Gemmen O, Kunamneni A, Inoki K. mTORC1 signaling and diabetic kidney disease. Diabetol Int 2024; 15:707-718. [PMID: 39469564 PMCID: PMC11512951 DOI: 10.1007/s13340-024-00738-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/26/2024] [Indexed: 10/30/2024]
Abstract
Diabetic kidney disease (DKD) represents the most lethal complication in both type 1 and type 2 diabetes. The disease progresses without obvious symptoms and is often refractory when apparent symptoms have emerged. Although the molecular mechanisms underlying the onset/progression of DKD have been extensively studied, only a few effective therapies are currently available. Pathogenesis of DKD involves multifaced events caused by diabetes, which include alterations of metabolisms, signals, and hemodynamics. While the considerable efficacy of sodium/glucose cotransporter-2 (SGLT2) inhibitors or angiotensin II receptor blockers (ARBs) for DKD has been recognized, the ever-increasing number of patients with diabetes and DKD warrants additional practical therapeutic approaches that prevent DKD from diabetes. One plausible but promising target is the mechanistic target of the rapamycin complex 1 (mTORC1) signaling pathway, which senses cellular nutrients to control various anabolic and catabolic processes. This review introduces the current understanding of the mTOR signaling pathway and its roles in the development of DKD and other chronic kidney diseases (CKDs), and discusses potential therapeutic approaches targeting this pathway for the future treatment of DKD.
Collapse
Affiliation(s)
- Vinamra Swaroop
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | - Eden Ozkan
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | - Lydia Herrmann
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | - Aaron Thurman
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | | | | | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, USA
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, USA
| |
Collapse
|
19
|
Zhang L, Wu M, Zhang J, Liu T, Fu S, Wang Y, Xu Z. The pivotal role of glucose transporter 1 in diabetic kidney disease. Life Sci 2024; 353:122932. [PMID: 39067659 DOI: 10.1016/j.lfs.2024.122932] [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: 04/05/2024] [Revised: 07/12/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Diabetes mellitus (DM) is a significant public health problem. Diabetic kidney disease (DKD) is the most common complication of DM, and its incidence has been increasing with the increasing prevalence of DM. Given the association between DKD and mortality in patients with DM, DKD is a significant burden on public health resources. Despite its significance in DM progression, the pathogenesis of DKD remains unclear. Aberrant glucose uptake by cells is an important pathophysiological mechanism underlying DKD renal injury. Glucose is transported across the bilayer cell membrane by a glucose transporter (GLUT) located on the cell membrane. Multiple GLUT proteins have been identified in the kidney, and GLUT1 is one of the most abundantly expressed isoforms. GLUT1 is a crucial regulator of intracellular glucose metabolism and plays a key pathological role in the phenotypic changes in DKD mesangial cells. In an attempt to understand the pathogenesis of DKD better, we here present a review of studies on the role of GLUT1 in the development and progression of DKD.
Collapse
Affiliation(s)
- Li Zhang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Meiyan Wu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jizhou Zhang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Tingting Liu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Shaojie Fu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yue Wang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zhonggao Xu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
20
|
Al-Diab O, Sünkel C, Blanc E, Catar RA, Ashraf MI, Zhao H, Wang P, Rinschen MM, Fritsche-Guenther R, Grahammer F, Bachmann S, Beule D, Kirwan JA, Rajewsky N, Huber TB, Gürgen D, Kusch A. Sex-specific molecular signature of mouse podocytes in homeostasis and in response to pharmacological challenge with rapamycin. Biol Sex Differ 2024; 15:72. [PMID: 39278930 PMCID: PMC11404044 DOI: 10.1186/s13293-024-00647-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/30/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Sex differences exist in the prevalence and progression of major glomerular diseases. Podocytes are the essential cell-type in the kidney which maintain the physiological blood-urine barrier, and pathological changes in podocyte homeostasis are critical accelerators of impairment of kidney function. However, sex-specific molecular signatures of podocytes under physiological and stress conditions remain unknown. This work aimed at identifying sexual dimorphic molecular signatures of podocytes under physiological condition and pharmacologically challenged homeostasis with mechanistic target of rapamycin (mTOR) inhibition. mTOR is a crucial regulator involved in a variety of physiological and pathological stress responses in the kidney and inhibition of this pathway may therefore serve as a general stress challenger to get fundamental insights into sex differences in podocytes. METHODS The genomic ROSAmT/mG-NPHS2 Cre mouse model was used which allows obtaining highly pure podocyte fractions for cell-specific molecular analyses, and vehicle or pharmacologic treatment with the mTOR inhibitor rapamycin was performed for 3 weeks. Subsequently, deep RNA sequencing and proteomics were performed of the isolated podocytes to identify intrinsic sex differences. Studies were supplemented with metabolomics from kidney cortex tissues. RESULTS Although kidney function and morphology remained normal in all experimental groups, RNA sequencing, proteomics and metabolomics revealed strong intrinsic sex differences in the expression levels of mitochondrial, translation and structural transcripts, protein abundances and regulation of metabolic pathways. Interestingly, rapamycin abolished prominent sex-specific clustering of podocyte gene expression and induced major changes only in male transcriptome. Several sex-biased transcription factors could be identified as possible upstream regulators of these sexually dimorphic responses. Concordant to transcriptomics, metabolomic changes were more prominent in males. Remarkably, high number of previously reported kidney disease genes showed intrinsic sexual dimorphism and/or different response patterns towards mTOR inhibition. CONCLUSIONS Our results highlight remarkable intrinsic sex-differences and sex-specific response patterns towards pharmacological challenged podocyte homeostasis which might fundamentally contribute to sex differences in kidney disease susceptibilities and progression. This work provides rationale and an in-depth database for novel targets to be tested in specific kidney disease models to advance with sex-specific treatment strategies.
Collapse
Affiliation(s)
- Ola Al-Diab
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Christin Sünkel
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str 28, 10115, Berlin, Germany
| | - Eric Blanc
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Rusan Ali Catar
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Muhammad Imtiaz Ashraf
- Department of Surgery, Experimental Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Hongfan Zhao
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Pinchao Wang
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Markus M Rinschen
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Raphaela Fritsche-Guenther
- Metabolomics Platform, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Lindenberger Weg 80, 10117, Berlin, Germany
| | - Florian Grahammer
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Sebastian Bachmann
- Institute of Functional Anatomy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jennifer A Kirwan
- Metabolomics Platform, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Lindenberger Weg 80, 10117, Berlin, Germany
| | - Nikolaus Rajewsky
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str 28, 10115, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Dennis Gürgen
- Experimental Pharmacology & Oncology Berlin-Buch GmbH, 13125 Berlin-Buch, Germany
| | - Angelika Kusch
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- BIH Biomedical Innovation Academy (BIA), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
21
|
Amin NG, Rahim AA, Rohoma K, Elwafa RAA, Dabees HMF, Elrahmany S. The relation of mTOR with diabetic complications and insulin resistance in patients with type 2 diabetes mellitus. Diabetol Metab Syndr 2024; 16:222. [PMID: 39261960 PMCID: PMC11389252 DOI: 10.1186/s13098-024-01450-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Dysregulation of the mechanistic target of rapamycin (mTOR) has been related to several metabolic conditions, notably obesity and type 2 diabetes (T2DM). This study aimed to evaluate the role of mTOR in patients with T2DM, and its relationship with insulin resistance and microvascular complications. METHODS This case-control study was conducted on 90 subjects attending the Outpatient Internal Medicine Clinic in Damanhur Teaching Hospital. Subjects were divided into 3 groups, Group I: 20 healthy controls, Group II: 20 subjects with T2DM without complications, and Group III: 50 subjects with T2DM with microvascular complications. An Enzyme-linked immunosorbent assay was used to measure serum mTOR levels. T2DM and diabetic complications were defined according to the diagnostic criteria of the American Diabetes Association. RESULTS The results revealed significant positive correlations to HbA1c (r = 0.530, P < 0.001), fasting glucose (r = 0.508, P < 0.001), and HOMA- IR (r = 0.559, P < 0.001), and a significant negative correlation to eGFR (r=-0.370, P = 0.002). Multivariate analysis revealed an independent association of mTOR and HbA1c values with the presence of microvascular complications. The prediction of microvascular complications was present at a cutoff value of 8 ng/ml mTOR with a sensitivity of 100% and specificity of 95% with an AUC of 0.983 and a p-value < 0.001. CONCLUSION mTOR is a prognostic marker of diabetic microvascular and is associated with insulin resistance in patients with T2DM. TRIAL REGISTRATION The study was conducted following the Declaration of Helsinki, and approved by the Ethics Committee of Alexandria University (0201127, 19/7/2018).
Collapse
Affiliation(s)
- Noha G Amin
- Department of Internal Medicine (Diabetes, Lipidology & Metabolism), Faculty of Medicine, Alexandria University, 17, Champollion Street, El Messallah, Alexandria, Egypt.
| | - A Abdel Rahim
- Department of Internal Medicine (Diabetes, Lipidology & Metabolism), Faculty of Medicine, Alexandria University, 17, Champollion Street, El Messallah, Alexandria, Egypt
| | - Kamel Rohoma
- Department of Internal Medicine (Diabetes, Lipidology & Metabolism), Faculty of Medicine, Alexandria University, 17, Champollion Street, El Messallah, Alexandria, Egypt
| | - Reham A Abo Elwafa
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Hossam M F Dabees
- Department of Internal Medicine (Diabetes, Lipidology & Metabolism), Faculty of Medicine, Alexandria University, 17, Champollion Street, El Messallah, Alexandria, Egypt
| | - Shimaa Elrahmany
- Department of Internal Medicine (Diabetes, Lipidology & Metabolism), Faculty of Medicine, Alexandria University, 17, Champollion Street, El Messallah, Alexandria, Egypt
| |
Collapse
|
22
|
Lin J, Lin Y, Li X, He F, Gao Q, Wang Y, Huang Z, Xiong F. Uncovering the Role of Anoikis-Related Genes in Modulating Immune Infiltration and Pathogenesis of Diabetic Kidney Disease. J Inflamm Res 2024; 17:4975-4991. [PMID: 39070131 PMCID: PMC11283803 DOI: 10.2147/jir.s446752] [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: 10/26/2023] [Accepted: 05/22/2024] [Indexed: 07/30/2024] Open
Abstract
Background Diabetic kidney disease (DKD) is an intricate complication of diabetes with limited treatment options. Anoikis, a programmed cell death activated by loss of cell anchorage to the extracellular matrix, participated in various physiological and pathological processes. Our study aimed to elucidate the role of anoikis-related genes in DKD pathogenesis. Methods Differentially expressed genes (DEGs) associated with anoikis in DKD were identified. Weighted gene co-expression network analysis (WGCNA) was conducted to identify DKD-correlated modules and assess their functional implications. A diagnostic model for DKD was developed using LASSO regression and Gene set variation analysis (GSVA) was performed for enrichment analysis. Experimental validation was employed to validate the significance of selected genes in the progression of DKD. Results We identified 10 anoikis-related DEGs involved in key signaling pathways impacting DKD progression. WGCNA highlighted the yellow module's significant enrichment in immune response and regulatory pathways. Correlation analysis further revealed the association between immune infiltration and anoikis-related DEGs. Our LASSO regression-based diagnostic model demonstrated a well-predictive efficacy with seven identified genes. GSVA indicated that gene function in the high-risk group was primarily associated with immune regulation. Further experimental validation using diabetic mouse models and data analysis in the single-cell dataset confirmed the significance of PYCARD and SFN in DKD progression. High glucose stimulation in RAW264.7 and TCMK-1 cells showed significantly increased expression levels of both Pycard and Sfn. Co-expression analysis demonstrated distinct functions of PYCARD and SFN, with KEGG pathway analysis showing significant enrichment in immune regulation and cell proliferation pathway. Conclusion In conclusion, our study provides valuable insights into the molecular mechanisms involved in DKD pathogenesis, specifically highlighting the role of anoikis-related genes in modulating immune infiltration. These findings suggest that targeting these genes may hold promise for future diagnostic and therapeutic approaches in DKD management.
Collapse
Affiliation(s)
- Jiaqiong Lin
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, People’s Republic of China
- Dongguan Maternal and Child Health Care Hospital, Postdoctoral Innovation Practice Base of Southern Medical University, Dongguan, People’s Republic of China
| | - Yan Lin
- Yunkang School of Medicine and Health, Nanfang College, Guangzhou, People’s Republic of China
| | - Xiaoyong Li
- General Surgery Department; Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Fei He
- Department of Medical Genetics/Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Qinyuan Gao
- Yunkang School of Medicine and Health, Nanfang College, Guangzhou, People’s Republic of China
| | - Yuanjun Wang
- Yunkang School of Medicine and Health, Nanfang College, Guangzhou, People’s Republic of China
| | - Zena Huang
- Yunkang School of Medicine and Health, Nanfang College, Guangzhou, People’s Republic of China
| | - Fu Xiong
- Department of Medical Genetics/Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Department of Fetal Medicine and Prenatal Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
23
|
Das F, Ghosh-Choudhury N, Kasinath BS, Sharma K, Choudhury GG. High glucose-induced downregulation of PTEN-Long is sufficient for proximal tubular cell injury in diabetic kidney disease. Exp Cell Res 2024; 440:114116. [PMID: 38830568 DOI: 10.1016/j.yexcr.2024.114116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 04/24/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024]
Abstract
During the progression of diabetic kidney disease, proximal tubular epithelial cells respond to high glucose to induce hypertrophy and matrix expansion leading to renal fibrosis. Recently, a non-canonical PTEN has been shown to be translated from an upstream initiation codon CUG (leucine) to produce a longer protein called PTEN-Long (PTEN-L). Interestingly, the extended sequence present in PTEN-L contains cell secretion/penetration signal. Role of this non-canonical PTEN-L in diabetic renal tubular injury is not known. We show that high glucose decreases expression of PTEN-L. As a mechanism of its function, we find that reduced PTEN-L activates Akt-2, which phosphorylates and inactivate tuberin and PRAS40, resulting in activation of mTORC1 in tubular cells. Antibacterial agent acriflavine and antiviral agent ATA regulate translation from CUG codon. Acriflavine and ATA, respectively, decreased and increased expression of PTEN-L to altering Akt-2 and mTORC1 activation in the absence of change in expression of canonical PTEN. Consequently, acriflavine and ATA modulated high glucose-induced tubular cell hypertrophy and lamininγ1 expression. Importantly, expression of PTEN-L inhibited high glucose-stimulated Akt/mTORC1 activity to abrogate these processes. Since PTEN-L contains secretion/penetration signals, addition of conditioned medium containing PTEN-L blocked Akt-2/mTORC1 activity. Notably, in renal cortex of diabetic mice, we found reduced PTEN-L concomitant with Akt-2/mTORC1 activation, leading to renal hypertrophy and lamininγ1 expression. These results present first evidence for involvement of PTEN-L in diabetic kidney disease.
Collapse
Affiliation(s)
- Falguni Das
- VA Research, South Texas Veterans Health Care System, San Antonio, TX, USA; Department of Medicine, TX, USA
| | | | | | - Kumar Sharma
- VA Research, South Texas Veterans Health Care System, San Antonio, TX, USA; Department of Medicine, TX, USA
| | - Goutam Ghosh Choudhury
- VA Research, South Texas Veterans Health Care System, San Antonio, TX, USA; Department of Medicine, TX, USA; Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, USA.
| |
Collapse
|
24
|
Yu S, Li Y, Lu X, Han Z, Li C, Yuan X, Guo D. The regulatory role of miRNA and lncRNA on autophagy in diabetic nephropathy. Cell Signal 2024; 118:111144. [PMID: 38493883 DOI: 10.1016/j.cellsig.2024.111144] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/09/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Diabetic nephropathy (DN) is a serious complication of diabetes that causes glomerular sclerosis and end-stage renal disease, leading to ascending morbidity and mortality in diabetic patients. Excessive accumulation of aberrantly modified proteins or damaged organelles, such as advanced glycation end-products, dysfunctional mitochondria, and inflammasomes is associated with the pathogenesis of DN. As one of the main degradation pathways, autophagy recycles toxic substances to maintain cellular homeostasis and autophagy dysregulation plays a crucial role in DN progression. MicroRNA (miRNA) and long non-coding RNA (lncRNA) are non-coding RNA (ncRNA) molecules that regulate gene expression and have been implicated in both physiological and pathological conditions. Recent studies have revealed that autophagy-regulating miRNA and lncRNA have been involved in pathological processes of DN, including renal cell injury, mitochondrial dysfunction, inflammation, and renal fibrosis. This review summarizes the role of autophagy in DN and emphasizes the modulation of miRNA and lncRNA on autophagy during disease progression, for the development of promising interventions by targeting these ncRNAs in this disease.
Collapse
Affiliation(s)
- Siming Yu
- Department of Nephrology II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150036, China
| | - Yue Li
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xinxin Lu
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Zehui Han
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Chunsheng Li
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xingxing Yuan
- Heilongjiang University of Chinese Medicine, Harbin 150040, China; Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin 150006, China
| | - Dandan Guo
- Department of Cardiology, Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150001, China.
| |
Collapse
|
25
|
Njeim R, Merscher S, Fornoni A. Mechanisms and implications of podocyte autophagy in chronic kidney disease. Am J Physiol Renal Physiol 2024; 326:F877-F893. [PMID: 38601984 PMCID: PMC11386983 DOI: 10.1152/ajprenal.00415.2023] [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: 12/22/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
Autophagy is a protective mechanism through which cells degrade and recycle proteins and organelles to maintain cellular homeostasis and integrity. An accumulating body of evidence underscores the significant impact of dysregulated autophagy on podocyte injury in chronic kidney disease (CKD). In this review, we provide a comprehensive overview of the diverse types of autophagy and their regulation in cellular homeostasis, with a specific emphasis on podocytes. Furthermore, we discuss recent findings that focus on the functional role of different types of autophagy during podocyte injury in chronic kidney disease. The intricate interplay between different types of autophagy and podocyte health requires further research, which is critical for understanding the pathogenesis of CKD and developing targeted therapeutic interventions.
Collapse
Affiliation(s)
- Rachel Njeim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, United States
| |
Collapse
|
26
|
Cybulsky AV, Papillon J, Guillemette J, Navarro-Betancourt JR, Chung CF, Iwawaki T, Fantus IG. Deletion of IRE1α in podocytes exacerbates diabetic nephropathy in mice. Sci Rep 2024; 14:11718. [PMID: 38778209 PMCID: PMC11111796 DOI: 10.1038/s41598-024-62599-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024] Open
Abstract
Protein misfolding in the endoplasmic reticulum (ER) of podocytes contributes to the pathogenesis of glomerular diseases. Protein misfolding activates the unfolded protein response (UPR), a compensatory signaling network. We address the role of the UPR and the UPR transducer, inositol-requiring enzyme 1α (IRE1α), in streptozotocin-induced diabetic nephropathy in mice. Diabetes caused progressive albuminuria in control mice that was exacerbated in podocyte-specific IRE1α knockout (KO) mice. Compared to diabetic controls, diabetic IRE1α KO mice showed reductions in podocyte number and synaptopodin. Glomerular ultrastructure was altered only in diabetic IRE1α KO mice; the major changes included widening of podocyte foot processes and glomerular basement membrane. Activation of the UPR and autophagy was evident in diabetic control, but not diabetic IRE1α KO mice. Analysis of human glomerular gene expression in the JuCKD-Glom database demonstrated induction of genes associated with the ER, UPR and autophagy in diabetic nephropathy. Thus, mice with podocyte-specific deletion of IRE1α demonstrate more severe diabetic nephropathy and attenuation of the glomerular UPR and autophagy, implying a protective effect of IRE1α. These results are consistent with data in human diabetic nephropathy and highlight the potential for therapeutically targeting these pathways.
Collapse
Affiliation(s)
- Andrey V Cybulsky
- Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, QC, Canada.
| | - Joan Papillon
- Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, QC, Canada
| | - Julie Guillemette
- Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, QC, Canada
| | - José R Navarro-Betancourt
- Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, QC, Canada
| | - Chen-Fang Chung
- Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, QC, Canada
| | - Takao Iwawaki
- Department of Life Science, Kanazawa Medical University, Uchinada, Japan
| | - I George Fantus
- Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
27
|
von Rauchhaupt E, Klaus M, Ribeiro A, Honarpisheh M, Li C, Liu M, Köhler P, Adamowicz K, Schmaderer C, Lindenmeyer M, Steiger S, Anders HJ, Lech M. GDF-15 Suppresses Puromycin Aminonucleoside-Induced Podocyte Injury by Reducing Endoplasmic Reticulum Stress and Glomerular Inflammation. Cells 2024; 13:637. [PMID: 38607075 PMCID: PMC11011265 DOI: 10.3390/cells13070637] [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: 02/12/2024] [Revised: 03/22/2024] [Accepted: 03/30/2024] [Indexed: 04/13/2024] Open
Abstract
GDF15, also known as MIC1, is a member of the TGF-beta superfamily. Previous studies reported elevated serum levels of GDF15 in patients with kidney disorder, and its association with kidney disease progression, while other studies identified GDF15 to have protective effects. To investigate the potential protective role of GDF15 on podocytes, we first performed in vitro studies using a Gdf15-deficient podocyte cell line. The lack of GDF15 intensified puromycin aminonucleoside (PAN)-triggered endoplasmic reticulum stress and induced cell death in cultivated podocytes. This was evidenced by elevated expressions of Xbp1 and ER-associated chaperones, alongside AnnexinV/PI staining and LDH release. Additionally, we subjected mice to nephrotoxic PAN treatment. Our observations revealed a noteworthy increase in both GDF15 expression and secretion subsequent to PAN administration. Gdf15 knockout mice displayed a moderate loss of WT1+ cells (podocytes) in the glomeruli compared to wild-type controls. However, this finding could not be substantiated through digital evaluation. The parameters of kidney function, including serum BUN, creatinine, and albumin-creatinine ratio (ACR), were increased in Gdf15 knockout mice as compared to wild-type mice upon PAN treatment. This was associated with an increase in the number of glomerular macrophages, neutrophils, inflammatory cytokines, and chemokines in Gdf15-deficient mice. In summary, our findings unveil a novel renoprotective effect of GDF15 during kidney injury and inflammation by promoting podocyte survival and regulating endoplasmic reticulum stress in podocytes, and, subsequently, the infiltration of inflammatory cells via paracrine effects on surrounding glomerular cells.
Collapse
Affiliation(s)
- Ekaterina von Rauchhaupt
- Department of Medicine IV, Renal Division, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (E.v.R.); (M.K.); (A.R.); (M.H.); (C.L.); (M.L.); (P.K.); (S.S.); (H.-J.A.)
| | - Martin Klaus
- Department of Medicine IV, Renal Division, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (E.v.R.); (M.K.); (A.R.); (M.H.); (C.L.); (M.L.); (P.K.); (S.S.); (H.-J.A.)
| | - Andrea Ribeiro
- Department of Medicine IV, Renal Division, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (E.v.R.); (M.K.); (A.R.); (M.H.); (C.L.); (M.L.); (P.K.); (S.S.); (H.-J.A.)
- Klinikum Rechts der Isar, Department of Nephrology, Technical University Munich, 81675 Munich, Germany;
| | - Mohsen Honarpisheh
- Department of Medicine IV, Renal Division, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (E.v.R.); (M.K.); (A.R.); (M.H.); (C.L.); (M.L.); (P.K.); (S.S.); (H.-J.A.)
| | - Chenyu Li
- Department of Medicine IV, Renal Division, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (E.v.R.); (M.K.); (A.R.); (M.H.); (C.L.); (M.L.); (P.K.); (S.S.); (H.-J.A.)
| | - Min Liu
- Department of Medicine IV, Renal Division, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (E.v.R.); (M.K.); (A.R.); (M.H.); (C.L.); (M.L.); (P.K.); (S.S.); (H.-J.A.)
| | - Paulina Köhler
- Department of Medicine IV, Renal Division, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (E.v.R.); (M.K.); (A.R.); (M.H.); (C.L.); (M.L.); (P.K.); (S.S.); (H.-J.A.)
| | - Karina Adamowicz
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology of Jagiellonian University, 30-387 Krakow, Poland;
| | - Christoph Schmaderer
- Klinikum Rechts der Isar, Department of Nephrology, Technical University Munich, 81675 Munich, Germany;
| | - Maja Lindenmeyer
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
| | - Stefanie Steiger
- Department of Medicine IV, Renal Division, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (E.v.R.); (M.K.); (A.R.); (M.H.); (C.L.); (M.L.); (P.K.); (S.S.); (H.-J.A.)
| | - Hans-Joachim Anders
- Department of Medicine IV, Renal Division, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (E.v.R.); (M.K.); (A.R.); (M.H.); (C.L.); (M.L.); (P.K.); (S.S.); (H.-J.A.)
| | - Maciej Lech
- Department of Medicine IV, Renal Division, Ludwig-Maximilians-University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany; (E.v.R.); (M.K.); (A.R.); (M.H.); (C.L.); (M.L.); (P.K.); (S.S.); (H.-J.A.)
| |
Collapse
|
28
|
Tanaka R, Imamura R, Matsumura S, Fukae S, Taniguchi A, Nakazawa S, Yamanaka K, Namba-Hamano T, Kakuta Y, Takao T, Fushimi H, Nonomura N. Remarkable Improvement of Diabetic Nephropathy in Transplanted Allograft after Kidney Transplantation. Nephron Clin Pract 2024; 148:468-473. [PMID: 38452745 DOI: 10.1159/000535877] [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: 08/23/2023] [Accepted: 12/07/2023] [Indexed: 03/09/2024] Open
Abstract
Although glomerular damage caused by diabetic nephropathy was thought to be irreversible, in recent years, there have been reports on improvement in glomerular damage with strict glycemic control. However, few reports are available on the pathologic course after renal transplantation of donor-derived grafts with findings of diabetic nephropathy. A 53-year-old woman underwent an ABO blood-type compatible living-donor renal transplant. The recipient had no history of diabetes, and fasting blood glucose and hemoglobin A1c levels were both normal. The donor was a 57-year-old male who had received treatment for type 2 diabetes mellitus for 10 years. Transplant renal biopsy performed 1 h after revascularization showed mesangial matrix expansion and arterial hyalinosis due to diabetic nephropathy. The blood glucose level was within the normal range after transplantation. Mesangial matrix expansion and arterial hyalinosis disappeared in allograft biopsy samples 7 years after transplantation. We observed significant improvement in the pathological findings of donor-derived diabetic nephropathy after renal transplantation in the subsequent follow-ups.
Collapse
Affiliation(s)
- Ryo Tanaka
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan,
| | - Ryoichi Imamura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Urology, Nagasaki University Graduate School of Biomedical Science, Nagasaki, Japan
| | - Soichi Matsumura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shota Fukae
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ayumu Taniguchi
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Urology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Shigeaki Nakazawa
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuaki Yamanaka
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoko Namba-Hamano
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoichi Kakuta
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuya Takao
- Department of Urology, Osaka General Medical Center, Osaka, Japan
| | - Hiroaki Fushimi
- Department of Pathology, Osaka General Medical Center, Osaka, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
29
|
Hu S, Hang X, Wei Y, Wang H, Zhang L, Zhao L. Crosstalk among podocytes, glomerular endothelial cells and mesangial cells in diabetic kidney disease: an updated review. Cell Commun Signal 2024; 22:136. [PMID: 38374141 PMCID: PMC10875896 DOI: 10.1186/s12964-024-01502-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 01/28/2024] [Indexed: 02/21/2024] Open
Abstract
Diabetic kidney disease (DKD) is a long-term and serious complication of diabetes that affects millions of people worldwide. It is characterized by proteinuria, glomerular damage, and renal fibrosis, leading to end-stage renal disease, and the pathogenesis is complex and involves multiple cellular and molecular mechanisms. Among three kinds of intraglomerular cells including podocytes, glomerular endothelial cells (GECs) and mesangial cells (MCs), the alterations in one cell type can produce changes in the others. The cell-to-cell crosstalk plays a crucial role in maintaining the glomerular filtration barrier (GFB) and homeostasis. In this review, we summarized the recent advances in understanding the pathological changes and interactions of these three types of cells in DKD and then focused on the signaling pathways and factors that mediate the crosstalk, such as angiopoietins, vascular endothelial growth factors, transforming growth factor-β, Krüppel-like factors, retinoic acid receptor response protein 1 and exosomes, etc. Furthermore, we also simply introduce the application of the latest technologies in studying cell interactions within glomerular cells and new promising mediators for cell crosstalk in DKD. In conclusion, this review provides a comprehensive and updated overview of the glomerular crosstalk in DKD and highlights its importance for the development of novel intervention approaches.
Collapse
Affiliation(s)
- Shiwan Hu
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xing Hang
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu Wei
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Han Wang
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Lili Zhang
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
30
|
Yamahara K, Yasuda-Yamahara M, Kume S. A novel therapeutic target for kidney diseases: Lessons learned from starvation response. Pharmacol Ther 2024; 254:108590. [PMID: 38286162 DOI: 10.1016/j.pharmthera.2024.108590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/16/2023] [Accepted: 01/05/2024] [Indexed: 01/31/2024]
Abstract
The prevalence of chronic kidney disease (CKD) is increasing worldwide, making the disease an urgent clinical challenge. Caloric restriction has various anti-aging and organ-protective effects, and unraveling its molecular mechanisms may provide insight into the pathophysiology of CKD. In response to changes in nutritional status, intracellular nutrient signaling pathways show adaptive changes. When nutrients are abundant, signals such as mechanistic target of rapamycin complex 1 (mTORC1) are activated, driving cell proliferation and other processes. Conversely, others, such as sirtuins and AMP-activated protein kinase, are activated during energy scarcity, in an attempt to compensate. Autophagy, a cellular self-maintenance mechanism that is regulated by such signals, has also been reported to contribute to the progression of various kidney diseases. Furthermore, in recent years, ketone bodies, which have long been considered to be detrimental, have been reported to play a role as starvation signals, and thereby to have renoprotective effects, via the inhibition of mTORC1. Therefore, in this review, we discuss the role of mTORC1, which is one of the most extensively studied nutrient-related signals associated with kidney diseases, autophagy, and ketone body metabolism; and kidney energy metabolism as a novel therapeutic target for CKD.
Collapse
Affiliation(s)
- Kosuke Yamahara
- Department of Medicine, Shiga University of Medical Science, Shiga, Japan
| | | | - Shinji Kume
- Department of Medicine, Shiga University of Medical Science, Shiga, Japan.
| |
Collapse
|
31
|
Farooqui Z, Banday AA. Angiotensin 1-7 exerts antioxidant effects, suppresses Mammalian Target of Rapamycin (mTOR) signaling, and inhibits apoptosis in renal proximal tubular cells. Peptides 2024; 172:171136. [PMID: 38104660 DOI: 10.1016/j.peptides.2023.171136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/09/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
Oxidative stress is one of the crucial pathogenic factors involved in the progression of renal injury. Angiotensin (ANG) 1-7, a bioactive heptapeptide of the renin-angiotensin-aldosterone system is known to exert antioxidant and nephroprotective effects. However, the cellular mechanism involved in the beneficial effect of ANG 1-7 is not clear. Here, we assessed ANG 1-7's effect on H2O2-mediated oxidative damage in the human proximal tubular (HK2) cells and the underlying mechanisms. HK2 cells were incubated with H2O2 (500 µM, 4 h) pre-treated with and without ANG 1-7 (100 nM, 24 h), and reactive oxygen species (ROS) generation, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, apoptosis and mammalian target of rapamycin (mTOR) signaling were determined H2O2 induced an increase in oxidative and ER stress together with loss of mitochondrial membrane potential, decreased ATP levels, and induced apoptosis in HK2 cells. Moreover, H2O2 treatment resulted in the activation of mTOR complexes (mTORC1 and mTORC2) in these cells. ANG 1-7 significantly attenuated H2O2-induced ROS generation, ER stress and apoptosis, and also improved mitochondrial function. Additionally, pre-treatment of ANG 1-7 inhibited the H2O2-mediated mTOR activation. These effects of ANG 1-7 were blocked by co-treatment with the Mas receptor (MasR) inhibitor, A779. Furthermore, transfection of HK2 cells with Mas receptor siRNA also abolished the inhibitory effect of ANG 1-7 on mTOR activities. In conclusion, ANG 1-7 via MasR mitigates oxidative stress, suppresses mTOR signaling, and protects HK2 cells from ER stress, mitochondrial dysfunction, and apoptosis, suggesting ANG 1-7-MasR renoprotective effects.
Collapse
Affiliation(s)
- Zeba Farooqui
- Heart and Kidney Institute, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Anees Ahmad Banday
- Heart and Kidney Institute, College of Pharmacy, University of Houston, Houston, TX 77204, USA.
| |
Collapse
|
32
|
Yamahara K, Yasuda-Yamahara M, Kuwagata S, Chin-Kanasaki M, Kume S. Ketone Body Metabolism in Diabetic Kidney Disease. KIDNEY360 2024; 5:320-326. [PMID: 38227425 PMCID: PMC10914200 DOI: 10.34067/kid.0000000000000359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/10/2024] [Indexed: 01/17/2024]
Abstract
Ketone bodies have a negative image because of ketoacidosis, one of the acute and serious complications in diabetes. The negative image persists despite the fact that ketone bodies are physiologically produced in the liver and serve as an indispensable energy source in extrahepatic organs, particularly during long-term fasting. However, accumulating experimental evidence suggests that ketone bodies exert various health benefits. Particularly in the field of aging research, there is growing interest in the potential organoprotective effects of ketone bodies. In addition, ketone bodies have a potential role in preventing kidney diseases, including diabetic kidney disease (DKD), a diabetic complication caused by prolonged hyperglycemia that leads to a decline in kidney function. Ketone bodies may help alleviate the renal burden from hyperglycemia by being used as an alternative energy source in patients with diabetes. Furthermore, ketone body production may reduce inflammation and delay the progression of several kidney diseases in addition to DKD. Although there is still insufficient research on the use of ketone bodies as a treatment and their effects, their renoprotective effects are being gradually proven. This review outlines the ketone body-mediated renoprotective effects in DKD and other kidney diseases.
Collapse
Affiliation(s)
- Kosuke Yamahara
- Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | | | | | | | | |
Collapse
|
33
|
Safaie N, Masoumi S, Alizadeh S, Mirzajanzadeh P, Nejabati HR, Hajiabbasi M, Alivirdiloo V, Basmenji NC, Derakhshi Radvar A, Majidi Z, Faridvand Y. SGLT2 inhibitors and AMPK: The road to cellular housekeeping? Cell Biochem Funct 2024; 42:e3922. [PMID: 38269506 DOI: 10.1002/cbf.3922] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 01/26/2024]
Abstract
Sodium-glucose co-transporter-2 (SGLT2) inhibitors, known as Gliflozins, are a class of Glucose-lowering drugs in adults with type 2 diabetes (T2D) that induce glucosuria by blocking SGLT2 co-transporters in the proximal tubules. Several lines of evidence suggest that SGLT2 inhibitors regulate multiple mechanisms associated with the regulation of varying cellular pathways. The 5'-adenosine monophosphate-activated protein kinase (AMPK) pathway plays an important role in metabolic homeostasis by influencing cellular processes. Recently, it has been shown that SGLT2 inhibitors can affect the AMPK pathway in differing physiological and pathological ways, resulting in kidney, intestinal, cardiovascular, and liver protective effects. Additionally, they have therapeutic effects on nonalcoholic fatty liver disease and diabetes mellitus-associated complications. In this review, we summarize the results of studies of AMPK-associated therapeutic effects of SGLT2 inhibitors in different organelle functions.
Collapse
Affiliation(s)
- Nasser Safaie
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahab Masoumi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Cardiovascular Fellowship, Vanderbilt University of Medical center, Nashville, Tennessee, USA
| | - Shaban Alizadeh
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | - Vahid Alivirdiloo
- Ramsar Campus, Mazandaran University of Medical Sciences, Ramasr, Iran
| | | | | | - Ziba Majidi
- Department of Medical Laboratory Science, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Faridvand
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
Tseng CH, Shah KM, Chiu IJ, Hsiao LL. The Role of Autophagy in Type 2 Diabetic Kidney Disease Management. Cells 2023; 12:2691. [PMID: 38067119 PMCID: PMC10705810 DOI: 10.3390/cells12232691] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Diabetic kidney disease (DKD), or diabetic nephropathy (DN), is one of the most prevalent complications of type 2 diabetes mellitus (T2DM) and causes severe burden on the general welfare of T2DM patients around the world. While several new agents have shown promise in treating this condition and potentially halting the progression of the disease, more work is needed to understand the complex regulatory network involved in the disorder. Recent studies have provided new insights into the connection between autophagy, a physiological metabolic process known to maintain cellular homeostasis, and the pathophysiological pathways of DKD. Typically, autophagic activity plays a role in DKD progression mainly by promoting an inflammatory response to tissue damage, while both overactivated and downregulated autophagy worsen disease outcomes in different stages of DKD. This correlation demonstrates the potential of autophagy as a novel therapeutic target for the disease, and also highlights new possibilities for utilizing already available DN-related medications. In this review, we summarize findings on the relationship between autophagy and DKD, and the impact of these results on clinical management strategies.
Collapse
Affiliation(s)
- Che-Hao Tseng
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (C.-H.T.); (K.M.S.)
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Kavya M. Shah
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (C.-H.T.); (K.M.S.)
| | - I-Jen Chiu
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (C.-H.T.); (K.M.S.)
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU-Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei 11031, Taiwan
| | - Li-Li Hsiao
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (C.-H.T.); (K.M.S.)
| |
Collapse
|
35
|
Chevalier RL. Why is chronic kidney disease progressive? Evolutionary adaptations and maladaptations. Am J Physiol Renal Physiol 2023; 325:F595-F617. [PMID: 37675460 DOI: 10.1152/ajprenal.00134.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/08/2023] [Accepted: 08/27/2023] [Indexed: 09/08/2023] Open
Abstract
Despite significant advances in renal physiology, the global prevalence of chronic kidney disease (CKD) continues to increase. The emergence of multicellular organisms gave rise to increasing complexity of life resulting in trade-offs reflecting ancestral adaptations to changing environments. Three evolutionary traits shape CKD over the lifespan: 1) variation in nephron number at birth, 2) progressive nephron loss with aging, and 3) adaptive kidney growth in response to decreased nephron number. Although providing plasticity in adaptation to changing environments, the cell cycle must function within constraints dictated by available energy. Prioritized allocation of energy available through the placenta can restrict fetal nephrogenesis, a risk factor for CKD. Moreover, nephron loss with aging is a consequence of cell senescence, a pathway accelerated by adaptive nephron hypertrophy that maintains metabolic homeostasis at the expense of increased vulnerability to stressors. Driven by reproductive fitness, natural selection operates in early life but diminishes thereafter, leading to an exponential increase in CKD with aging, a product of antagonistic pleiotropy. A deeper understanding of the evolutionary constraints on the cell cycle may lead to manipulation of the balance between progenitor cell renewal and differentiation, regulation of cell senescence, and modulation of the balance between cell proliferation and hypertrophy. Application of an evolutionary perspective may enhance understanding of adaptation and maladaptation by nephrons in the progression of CKD, leading to new therapeutic advances.
Collapse
Affiliation(s)
- Robert L Chevalier
- Department of Pediatrics, The University of Virginia, Charlottesville, Virginia, United States
| |
Collapse
|
36
|
Salemkour Y, Yildiz D, Dionet L, ‘t Hart DC, Verheijden KA, Saito R, Mahtal N, Delbet JD, Letavernier E, Rabant M, Karras A, van der Vlag J, Nijenhuis T, Tharaux PL, Lenoir O. Podocyte Injury in Diabetic Kidney Disease in Mouse Models Involves TRPC6-mediated Calpain Activation Impairing Autophagy. J Am Soc Nephrol 2023; 34:1823-1842. [PMID: 37678257 PMCID: PMC10631601 DOI: 10.1681/asn.0000000000000212] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/28/2023] [Indexed: 09/09/2023] Open
Abstract
SIGNIFICANCE STATEMENT Autophagy protects podocytes from injury in diabetic kidney disease (DKD). Restoring glomerular autophagy is a promising approach to limit DKD. This study demonstrates a novel regulatory mechanism of autophagy that blocks this critical protection of the glomerular filtration barrier. We demonstrated that TRPC6 induced in podocytes in mouse models of diabetes mediates calpain activation, thereby impairing podocyte autophagy, causing injury and accelerating DKD. Furthermore, this study provides proof of principle for druggable targets for DKD because restoration of podocyte autophagy by calpain inhibitors effectively limits glomerular destruction. BACKGROUND Diabetic kidney disease is associated with impaired podocyte autophagy and subsequent podocyte injury. The regulation of podocyte autophagy is unique because it minimally uses the mTOR and AMPK pathways. Thus, the molecular mechanisms underlying the impaired autophagy in podocytes in diabetic kidney disease remain largely elusive. METHODS This study investigated how the calcium channel TRPC6 and the cysteine protease calpains deleteriously affect podocyte autophagy in diabetic kidney disease in mice. We demonstrated that TRPC6 knockdown in podocytes increased the autophagic flux because of decreased cysteine protease calpain activity. Diabetic kidney disease was induced in vivo using streptozotocin with unilateral nephrectomy and the BTBR ob/ob mouse models. RESULTS Diabetes increased TRPC6 expression in podocytes in vivo with decreased podocyte autophagic flux. Transgenic overexpression of the endogenous calpain inhibitor calpastatin, as well as pharmacologic inhibition of calpain activity, normalized podocyte autophagic flux, reduced nephrin loss, and prevented the development of albuminuria in diabetic mice. In kidney biopsies from patients with diabetes, we further confirmed that TRPC6 overexpression in podocytes correlates with decreased calpastatin expression, autophagy blockade, and podocyte injury. CONCLUSIONS Overall, we discovered a new mechanism that connects TRPC6 and calpain activity to impaired podocyte autophagy, increased podocyte injury, and development of proteinuria in the context of diabetic kidney disease. Therefore, targeting TRPC6 and/or calpain to restore podocyte autophagy might be a promising therapeutic strategy for diabetic kidney disease.
Collapse
Affiliation(s)
| | - Dilemin Yildiz
- Department of Nephrology, Research Institute of Medical Innovations, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Léa Dionet
- Université Paris Cité, Inserm, PARCC, Paris, France
| | - Daan C. ‘t Hart
- Department of Nephrology, Research Institute of Medical Innovations, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Kim A.T. Verheijden
- Department of Nephrology, Research Institute of Medical Innovations, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Ryuta Saito
- Discovery Technology Laboratories, Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | | | - Jean-Daniel Delbet
- Université Paris Cité, Inserm, PARCC, Paris, France
- Pediatric Nephrology Department, Armand Trousseau Hospital, DMU Origyne, APHP, Paris and French Reference Center for Rare Diseases MARHEA, Paris, France
| | - Emmanuel Letavernier
- Sorbonne Université, Hôpital Tenon, Paris, France
- INSERM UMR S 1155, Hôpital Tenon, Paris, France
- Explorations Fonctionnelles Multidisciplinaires, AP-HP, Hôpital Tenon, Paris, France
| | - Marion Rabant
- Pathology Department, Necker-Enfants Malades Hospital - Paris, Paris, France
| | - Alexandre Karras
- Université Paris Cité, Inserm, PARCC, Paris, France
- Nephrology Unit, Georges Pompidou European Hospital - Paris, Paris, France
| | - Johan van der Vlag
- Department of Nephrology, Research Institute of Medical Innovations, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Tom Nijenhuis
- Department of Nephrology, Research Institute of Medical Innovations, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | | |
Collapse
|
37
|
Mancini MC, Noland RC, Collier JJ, Burke SJ, Stadler K, Heden TD. Lysosomal glucose sensing and glycophagy in metabolism. Trends Endocrinol Metab 2023; 34:764-777. [PMID: 37633800 PMCID: PMC10592240 DOI: 10.1016/j.tem.2023.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/28/2023]
Abstract
Lysosomes are cellular organelles that function to catabolize both extra- and intracellular cargo, act as a platform for nutrient sensing, and represent a core signaling node integrating bioenergetic cues to changes in cellular metabolism. Although lysosomal amino acid and lipid sensing in metabolism has been well characterized, lysosomal glucose sensing and the role of lysosomes in glucose metabolism is unrefined. This review will highlight the role of the lysosome in glucose metabolism with a focus on lysosomal glucose and glycogen sensing, glycophagy, and lysosomal glucose transport and how these processes impact autophagy and energy metabolism. Additionally, the role of lysosomal glucose metabolism in genetic and metabolic diseases will be briefly discussed.
Collapse
Affiliation(s)
- Melina C Mancini
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Robert C Noland
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - J Jason Collier
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Susan J Burke
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | | | - Timothy D Heden
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| |
Collapse
|
38
|
Aquilani R, Verri M. Nutrition for Podocyte Repair in Nephrotic Syndrome? Nutrients 2023; 15:4615. [PMID: 37960268 PMCID: PMC10650452 DOI: 10.3390/nu15214615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 10/10/2023] [Indexed: 11/15/2023] Open
Abstract
Nephrotic syndrome (NS) poses a number of nutritional and metabolic problems due to glomerulus injured podocytes, which are responsible for the loss of barrier function, causing proteinuria, altered fluid and electrolyte balances, and hypoalbuminemia [...].
Collapse
Affiliation(s)
| | - Manuela Verri
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
39
|
Lu J, Li XQ, Chen PP, Zhang JX, Liu L, Wang GH, Liu XQ, Jiang TT, Wang MY, Liu WT, Ruan XZ, Ma KL. Activation of acetyl-CoA synthetase 2 mediates kidney injury in diabetic nephropathy. JCI Insight 2023; 8:e165817. [PMID: 37870960 PMCID: PMC10619493 DOI: 10.1172/jci.insight.165817] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 09/05/2023] [Indexed: 10/25/2023] Open
Abstract
Albuminuria and podocyte injury are the key cellular events in the progression of diabetic nephropathy (DN). Acetyl-CoA synthetase 2 (ACSS2) is a nucleocytosolic enzyme responsible for the regulation of metabolic homeostasis in mammalian cells. This study aimed to investigate the possible roles of ACSS2 in kidney injury in DN. We constructed an ACSS2-deleted mouse model to investigate the role of ACSS2 in podocyte dysfunction and kidney injury in diabetic mouse models. In vitro, podocytes were chosen and transfected with ACSS2 siRNA and ACSS2 inhibitor and treated with high glucose. We found that ACSS2 expression was significantly elevated in the podocytes of patients with DN and diabetic mice. ACSS2 upregulation promoted phenotype transformation and inflammatory cytokine expression while inhibiting podocytes' autophagy. Conversely, ACSS2 inhibition improved autophagy and alleviated podocyte injury. Furthermore, ACSS2 epigenetically activated raptor expression by histone H3K9 acetylation, promoting activation of the mammalian target of rapamycin complex 1 (mTORC1) pathway. Pharmacological inhibition or genetic depletion of ACSS2 in the streptozotocin-induced diabetic mouse model greatly ameliorated kidney injury and podocyte dysfunction. To conclude, ACSS2 activation promoted podocyte injury in DN by raptor/mTORC1-mediated autophagy inhibition.
Collapse
Affiliation(s)
- Jian Lu
- Department of Nephrology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xue Qi Li
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Pei Pei Chen
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jia Xiu Zhang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Liang Liu
- Department of Intensive Care Unit, People’s Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Gui Hua Wang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiao Qi Liu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ting Ting Jiang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Meng Ying Wang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Wen Tao Liu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiong Zhong Ruan
- John Moorhead Research Laboratory, Department of Renal Medicine, University College London Medical School, Royal Free Campus, London, United Kingdom
| | - Kun Ling Ma
- Department of Nephrology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
40
|
Hejazian SM, Ardalan M, Hosseiniyan Khatibi SM, Rahbar Saadat Y, Barzegari A, Gueguen V, Meddahi-Pellé A, Anagnostou F, Zununi Vahed S, Pavon-Djavid G. Biofactors regulating mitochondrial function and dynamics in podocytes and podocytopathies. J Cell Physiol 2023; 238:2206-2227. [PMID: 37659096 DOI: 10.1002/jcp.31110] [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: 03/30/2023] [Revised: 06/25/2023] [Accepted: 08/14/2023] [Indexed: 09/04/2023]
Abstract
Podocytes are terminally differentiated kidney cells acting as the main gatekeepers of the glomerular filtration barrier; hence, inhibiting proteinuria. Podocytopathies are classified as kidney diseases caused by podocyte damage. Different genetic and environmental risk factors can cause podocyte damage and death. Recent evidence shows that mitochondrial dysfunction also contributes to podocyte damage. Understanding alterations in mitochondrial metabolism and function in podocytopathies and whether altered mitochondrial homeostasis/dynamics is a cause or effect of podocyte damage are issues that need in-depth studies. This review highlights the roles of mitochondria and their bioenergetics in podocytes. Then, factors/signalings that regulate mitochondria in podocytes are discussed. After that, the role of mitochondrial dysfunction is reviewed in podocyte injury and the development of different podocytopathies. Finally, the mitochondrial therapeutic targets are considered.
Collapse
Affiliation(s)
| | | | | | | | - Abolfazl Barzegari
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| | - Virginie Gueguen
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| | - Anne Meddahi-Pellé
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| | - Fani Anagnostou
- Université de Paris, CNRS UMR 7052 INSERM U1271, B3OA, Paris, France
| | | | - Graciela Pavon-Djavid
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| |
Collapse
|
41
|
Guo W, Li H, Li Y, Kong W. Renal intrinsic cells remodeling in diabetic kidney disease and the regulatory effects of SGLT2 Inhibitors. Biomed Pharmacother 2023; 165:115025. [PMID: 37385209 DOI: 10.1016/j.biopha.2023.115025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 07/01/2023] Open
Abstract
Diabetic kidney disease (DKD) is a prevalent complication of diabetes and a major secondary factor leading to end-stage renal disease. The kidney, a vital organ, is composed of a heterogeneous group of intrinsic cells, including glomerular endothelial cells, podocytes, mesangial cells, tubular epithelial cells, and interstitial fibroblasts. In the context of DKD, hyperglycemia elicits direct or indirect injury to these intrinsic cells, leading to their structural and functional changes, such as cell proliferation, apoptosis, and transdifferentiation. The dynamic remodeling of intrinsic cells represents an adaptive response to stimulus during the pathogenesis of diabetic kidney disease. However, the persistent stimulus may trigger an irreversible remodeling, leading to fibrosis and functional deterioration of the kidney. Sodium-glucose cotransporter 2 (SGLT2) inhibitors, a new class of hypoglycemic drugs, exhibit efficacy in reducing blood glucose levels by curtailing renal tubular glucose reabsorption. Furthermore, SGLT2 inhibitors have been shown to modulate intrinsic cell remodeling in the kidney, ameliorate kidney structure and function, and decelerate DKD progression. This review will elaborate on the intrinsic cell remodeling in DKD and the underlying mechanism of SGLT2 inhibitors in modulating it from the perspective of the renal intrinsic cell, providing insights into the pathogenesis of DKD and the renal protective action of SGLT2 inhibitors.
Collapse
Affiliation(s)
- Wenwen Guo
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, Hubei 430022, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Wuhan, Hubei 430022, China
| | - Han Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, Hubei 430022, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Wuhan, Hubei 430022, China
| | - Yixuan Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, Hubei 430022, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Wuhan, Hubei 430022, China
| | - Wen Kong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, Hubei 430022, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Wuhan, Hubei 430022, China.
| |
Collapse
|
42
|
Kleibert M, Zygmunciak P, Łakomska K, Mila K, Zgliczyński W, Mrozikiewicz-Rakowska B. Insight into the Molecular Mechanism of Diabetic Kidney Disease and the Role of Metformin in Its Pathogenesis. Int J Mol Sci 2023; 24:13038. [PMID: 37685845 PMCID: PMC10487922 DOI: 10.3390/ijms241713038] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 09/10/2023] Open
Abstract
Diabetic kidney disease (DKD) is one of the leading causes of death among patients diagnosed with diabetes mellitus. Despite the growing knowledge about the pathogenesis of DKD, we still do not have effective direct pharmacotherapy. Accurate blood sugar control is essential in slowing down DKD. It seems that metformin has a positive impact on kidneys and this effect is not only mediated by its hypoglycemic action, but also by direct molecular regulation of pathways involved in DKD. The molecular mechanism of DKD is complex and we can distinguish polyol, hexosamine, PKC, and AGE pathways which play key roles in the development and progression of this disease. Each of these pathways is overactivated in a hyperglycemic environment and it seems that most of them may be regulated by metformin. In this article, we summarize the knowledge about DKD pathogenesis and the potential mechanism of the nephroprotective effect of metformin. Additionally, we describe the impact of metformin on glomerular endothelial cells and podocytes, which are harmed in DKD.
Collapse
Affiliation(s)
- Marcin Kleibert
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Przemysław Zygmunciak
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland; (P.Z.); (K.M.)
| | - Klaudia Łakomska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Klaudia Mila
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland; (P.Z.); (K.M.)
| | - Wojciech Zgliczyński
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, 01-809 Warsaw, Poland;
| | - Beata Mrozikiewicz-Rakowska
- Department of Endocrinology, Centre of Postgraduate Medical Education, Bielanski Hospital, 01-809 Warsaw, Poland;
| |
Collapse
|
43
|
Liu T, Jin Q, Yang L, Mao H, Ma F, Wang Y, Li P, Zhan Y. Regulation of autophagy by natural polyphenols in the treatment of diabetic kidney disease: therapeutic potential and mechanism. Front Endocrinol (Lausanne) 2023; 14:1142276. [PMID: 37635982 PMCID: PMC10448531 DOI: 10.3389/fendo.2023.1142276] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Diabetic kidney disease (DKD) is a major microvascular complication of diabetes and a leading cause of end-stage renal disease worldwide. Autophagy plays an important role in maintaining cellular homeostasis in renal physiology. In DKD, the accumulation of advanced glycation end products induces decreased renal autophagy-related protein expression and transcription factor EB (TFEB) nuclear transfer, leading to impaired autophagy and lysosomal function and blockage of autophagic flux. This accelerates renal resident cell injury and apoptosis, mediates macrophage infiltration and phenotypic changes, ultimately leading to aggravated proteinuria and fibrosis in DKD. Natural polyphenols show promise in treating DKD by regulating autophagy and promoting nuclear transfer of TFEB and lysosomal repair. This review summarizes the characteristics of autophagy in DKD, and the potential application and mechanisms of some known natural polyphenols as autophagy regulators in DKD, with the goal of contributing to a deeper understanding of natural polyphenol mechanisms in the treatment of DKD and promoting the development of their applications. Finally, we point out the limitations of polyphenols in current DKD research and provide an outlook for their future research.
Collapse
Affiliation(s)
- Tongtong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Jin
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fang Ma
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyang Wang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Yongli Zhan
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
44
|
Chhuon C, Herrera-Marcos LV, Zhang SY, Charrière-Bertrand C, Jung V, Lipecka J, Savas B, Nasser N, Pawlak A, Boulmerka H, Audard V, Sahali D, Guerrera IC, Ollero M. Proteomics of Plasma and Plasma-Treated Podocytes: Application to Focal and Segmental Glomerulosclerosis. Int J Mol Sci 2023; 24:12124. [PMID: 37569500 PMCID: PMC10418338 DOI: 10.3390/ijms241512124] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Focal and segmental glomerulosclerosis (FSGS) is a severe form of idiopathic nephrotic syndrome (INS), a glomerulopathy of presumably immune origin that is attributed to extrarenal pathogenic circulating factors. The recurrence of FSGS (rFSGS) after transplant occurs in 30% to 50% of cases. The direct analysis of patient plasma proteome has scarcely been addressed to date, mainly due to the methodological difficulties associated with plasma complexity and dynamic range. In this study, first, we compared different methods of plasma preparation, second, we compared the plasma proteomes of rFSGS and controls using two preparation methods, and third, we analyzed the early proximal signaling events in podocytes subjected to patient plasma, through a combination of phosphoproteomics and lipid-raft proteomics (raftomics). By combining immunodepletion and high pH fractionation, we performed a differential proteomic analysis of soluble plasma proteins and of extracellular vesicles (EV) obtained from healthy controls, non-INS patient controls, and rFSGS patients (n = 4). In both the soluble- and the EV-protein sets from the rFSGS patients, we found a statistically significant increase in a cluster of proteins involved in neutrophil degranulation. A group of lipid-binding proteins, generally associated with lipoproteins, was found to be decreased in the soluble set from the rFSGS patients. In addition, three amino acid transporters involved in mTORC1 activation were found to be significantly increased in the EV from the rFSGS. Next, we incubated human podocytes for 30 min with 10% plasma from both groups of patients. The phosphoproteomics and raftomics of the podocytes revealed profound differences in the proteins involved in the mTOR pathway, in autophagy, and in cytoskeleton organization. We analyzed the correlation between the abundance of plasma and plasma-regulated podocyte proteins. The observed changes highlight some of the mechanisms involved in FSGS recurrence and could be used as specific early markers of circulating-factor activity in podocytes.
Collapse
Affiliation(s)
- Cerina Chhuon
- Proteomic Platform Necker, Université Paris Cité Structure Fédérative de Recherche SFR Necker US24, 75015 Paris, France; (C.C.); (V.J.); (J.L.)
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Luis Vicente Herrera-Marcos
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Shao-Yu Zhang
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Cécile Charrière-Bertrand
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Vincent Jung
- Proteomic Platform Necker, Université Paris Cité Structure Fédérative de Recherche SFR Necker US24, 75015 Paris, France; (C.C.); (V.J.); (J.L.)
| | - Joanna Lipecka
- Proteomic Platform Necker, Université Paris Cité Structure Fédérative de Recherche SFR Necker US24, 75015 Paris, France; (C.C.); (V.J.); (J.L.)
| | - Berkan Savas
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Nour Nasser
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - André Pawlak
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Hocine Boulmerka
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| | - Vincent Audard
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
- AP-HP, Hôpitaux Universitaires Henri Mondor, Service de Néphrologie, F-94010 Creteil, France
| | - Dil Sahali
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
- AP-HP, Hôpitaux Universitaires Henri Mondor, Service de Néphrologie, F-94010 Creteil, France
| | - Ida Chiara Guerrera
- Proteomic Platform Necker, Université Paris Cité Structure Fédérative de Recherche SFR Necker US24, 75015 Paris, France; (C.C.); (V.J.); (J.L.)
| | - Mario Ollero
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; (L.V.H.-M.); (S.-Y.Z.); (C.C.-B.); (B.S.); (N.N.); (A.P.); (H.B.); (V.A.); (D.S.)
| |
Collapse
|
45
|
Peres RAS, Peruchetti DB, Silva-Aguiar RP, Teixeira DE, Gomes CP, Takiya CM, Pinheiro AAS, Caruso-Neves C. Rapamycin treatment induces tubular proteinuria: role of megalin-mediated protein reabsorption. Front Pharmacol 2023; 14:1194816. [PMID: 37484026 PMCID: PMC10359992 DOI: 10.3389/fphar.2023.1194816] [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: 03/27/2023] [Accepted: 06/27/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction: Rapamycin is an immunosuppressor that acts by inhibiting the serine/threonine kinase mechanistic target of rapamycin complex 1. Therapeutic use of rapamycin is limited by its adverse effects. Proteinuria is an important marker of kidney damage and a risk factor for kidney diseases progression and has been reported in patients and animal models treated with rapamycin. However, the mechanism underlying proteinuria induced by rapamycin is still an open matter. In this work, we investigated the effects of rapamycin on parameters of renal function and structure and on protein handling by proximal tubule epithelial cells (PTECs). Methods: Healthy BALB/c mice were treated with 1.5 mg/kg rapamycin by oral gavage for 1, 3, or 7 days. At the end of each treatment, the animals were kept in metabolic cages and renal function and structural parameters were analyzed. LLC-PK1 cell line was used as a model of PTECs to test specific effect of rapamycin. Results: Rapamycin treatment did not change parameters of glomerular structure and function. Conversely, there was a transient increase in 24-h proteinuria, urinary protein to creatinine ratio (UPCr), and albuminuria in the groups treated with rapamycin. In accordance with these findings, rapamycin treatment decreased albumin-fluorescein isothiocyanate uptake in the renal cortex. This effect was associated with reduced brush border expression and impaired subcellular distribution of megalin in PTECs. The effect of rapamycin seems to be specific for albumin endocytosis machinery because it did not modify renal sodium handling or (Na++K+)ATPase activity in BALB/c mice and in the LLC-PK1 cell line. A positive Pearson correlation was found between megalin expression and albumin uptake while an inverse correlation was shown between albumin uptake and UPCr or 24-h proteinuria. Despite its effect on albumin handling in PTECs, rapamycin treatment did not induce tubular injury measured by interstitial space and collagen deposition. Conclusion: These findings suggest that proteinuria induced by rapamycin could have a tubular rather than a glomerular origin. This effect involves a specific change in protein endocytosis machinery. Our results open new perspectives on understanding the undesired effect of proteinuria generated by rapamycin.
Collapse
Affiliation(s)
- Rodrigo A. S. Peres
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo B. Peruchetti
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo P. Silva-Aguiar
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas E. Teixeira
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos P. Gomes
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- School of Medicine and Surgery, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Christina M. Takiya
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Acacia S. Pinheiro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| |
Collapse
|
46
|
Pietrobon A, Stanford WL. Tuberous Sclerosis Complex Kidney Lesion Pathogenesis: A Developmental Perspective. J Am Soc Nephrol 2023; 34:1135-1149. [PMID: 37060140 PMCID: PMC10356159 DOI: 10.1681/asn.0000000000000146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/27/2023] [Indexed: 04/16/2023] Open
Abstract
The phenotypic diversity of tuberous sclerosis complex (TSC) kidney pathology is enigmatic. Despite a well-established monogenic etiology, an incomplete understanding of lesion pathogenesis persists. In this review, we explore the question: How do TSC kidney lesions arise? We appraise literature findings in the context of mutational timing and cell-of-origin. Through a developmental lens, we integrate the critical results from clinical studies, human specimens, and genetic animal models. We also review novel insights gleaned from emerging organoid and single-cell sequencing technologies. We present a new model of pathogenesis which posits a phenotypic continuum, whereby lesions arise by mutagenesis during development from variably timed second-hit events. This model can serve as a conceptual framework for testing hypotheses of TSC lesion pathogenesis, both in the kidney and in other affected tissues.
Collapse
Affiliation(s)
- Adam Pietrobon
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| | - William L. Stanford
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| |
Collapse
|
47
|
Vidigal AC, de Lucena DD, Beyerstedt S, Rangel ÉB. A comprehensive update of the metabolic and toxicological considerations for immunosuppressive drugs used during pancreas transplantation. Expert Opin Drug Metab Toxicol 2023; 19:405-427. [PMID: 37542452 DOI: 10.1080/17425255.2023.2243808] [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: 03/26/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/07/2023]
Abstract
INTRODUCTION Despite significant advancements in immunosuppressive regimens and surgical techniques, the prevalence of adverse events related to immunosuppression remains a major challenge affecting the long-term survival rates of pancreas and kidney allografts. AREAS COVERED This article presents a comprehensive review of the literature and knowledge (Jan/2012-Feb/2023) concerning glucose metabolism disorders and nephrotoxicity associated with tacrolimus and mammalian target of rapamycin inhibitors (mTORi). Novel signaling pathways potentially implicated in these adverse events are discussed. Furthermore, we extensively examine the findings from clinical trials evaluating the efficacy and safety of tacrolimus, mTORi, and steroid minimization. EXPERT OPINION Tacrolimus-based regimens continue to be the standard treatment following pancreas transplants. However, prolonged use of tacrolimus and mTORi may lead to hyperglycemia and nephrotoxicity. Understanding and interpreting experimental data, particularly concerning novel signaling pathways beyond calcineurin-NFAT and mTOR pathways, can offer valuable insights for therapeutic interventions to mitigate hyperglycemia and nephrotoxicity. Additionally, critically analyzing clinical trial results can identify opportunities for personalized safety-based approaches to minimize side effects. It is imperative to conduct randomized-controlled studies to assess the impact of mTORi use and steroid-free protocols on pancreatic allograft survival. Such studies will aid in tailoring treatment strategies for improved transplant outcomes.
Collapse
Affiliation(s)
- Ana Cláudia Vidigal
- Nephrology Division, Department of Medicine, Federal University of São Paulo, SP, Brazil
| | - Débora D de Lucena
- Nephrology Division, Department of Medicine, Federal University of São Paulo, SP, Brazil
| | - Stephany Beyerstedt
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, SP, São Paulo, Brazil
| | - Érika B Rangel
- Nephrology Division, Department of Medicine, Federal University of São Paulo, SP, Brazil
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, SP, São Paulo, Brazil
| |
Collapse
|
48
|
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: 17] [Impact Index Per Article: 8.5] [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
|
49
|
Tabei A, Sakairi T, Hamatani H, Ohishi Y, Watanabe M, Nakasatomi M, Ikeuchi H, Kaneko Y, Kopp JB, Hiromura K. The miR-143/145 cluster induced by TGF-β1 suppresses Wilms' tumor 1 expression in cultured human podocytes. Am J Physiol Renal Physiol 2023; 325:F121-F133. [PMID: 37167274 PMCID: PMC10511167 DOI: 10.1152/ajprenal.00313.2022] [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: 12/23/2022] [Revised: 05/01/2023] [Accepted: 05/01/2023] [Indexed: 05/13/2023] Open
Abstract
Transforming growth factor (TGF)-β1 contributes to podocyte injury in various glomerular diseases, including diabetic kidney disease, probably at least in part by attenuating the expression of Wilms' tumor 1 (WT1). However, the precise mechanisms remain to be defined. We performed miRNA microarray analysis in a human podocyte cell line cultured with TGF-β1 to examine the roles of miRNAs in podocyte damage. The microarray analysis identified miR-143-3p as the miRNA with the greatest increase following exposure to TGF-β1. Quantitative RT-PCR confirmed a significant increase in the miR-143-3p/145-5p cluster in TGF-β1-supplemented cultured podocytes and demonstrated upregulation of miR-143-3p in the glomeruli of mice with type 2 diabetes. Ectopic expression of miR-143-3p and miR-145-5p suppressed WT1 expression in cultured podocytes. Furthermore, inhibition of Smad or mammalian target of rapamycin signaling each partially reversed the TGF-β1-induced increase in miR-143-3p/145-5p and decrease in WT1. In conclusion, TGF-β1 induces expression of miR-143-3p/145-5p in part through Smad and mammalian target of rapamycin pathways, and miR-143-3p/145-5p reduces expression of WT1 in cultured human podocytes. miR-143-3p/145-5p may contribute to TGF-β1-induced podocyte injury.NEW & NOTEWORTHY This study by miRNA microarray analysis demonstrated that miR-143-3p expression was upregulated in cultured human podocytes following exposure to transforming growth factor (TGF)-β1. Furthermore, we report that the miR-143/145 cluster contributes to decreased expression of Wilms' tumor 1, which represents a possible mechanism for podocyte injury induced by TGF-β1. This study is important because it presents a novel mechanism for TGF-β-associated glomerular diseases, including diabetic kidney disease (DKD), and suggests potential therapeutic strategies targeting miR-143-3p/145-5p.
Collapse
Affiliation(s)
- Akifumi Tabei
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Toru Sakairi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hiroko Hamatani
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yuko Ohishi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Mitsuharu Watanabe
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Masao Nakasatomi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hidekazu Ikeuchi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yoriaki Kaneko
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Keiju Hiromura
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| |
Collapse
|
50
|
Huang CW, Lo SH. Tensins in Kidney Function and Diseases. Life (Basel) 2023; 13:1244. [PMID: 37374025 PMCID: PMC10305691 DOI: 10.3390/life13061244] [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: 03/21/2023] [Revised: 05/11/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Tensins are focal adhesion proteins that regulate various biological processes, such as mechanical sensing, cell adhesion, migration, invasion, and proliferation, through their multiple binding activities that transduce critical signals across the plasma membrane. When these molecular interactions and/or mediated signaling are disrupted, cellular activities and tissue functions are compromised, leading to disease development. Here, we focus on the significance of the tensin family in renal function and diseases. The expression pattern of each tensin in the kidney, their roles in chronic kidney diseases, renal cell carcinoma, and their potentials as prognostic markers and/or therapeutic targets are discussed in this review.
Collapse
Affiliation(s)
- Chien-Wei Huang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Su Hao Lo
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA
| |
Collapse
|