1
|
Zhao Y, Zhuang Y, Shi J, Fan H, Lv Q, Guo X. Cathepsin B induces kidney diseases through different types of programmed cell death. Front Immunol 2025; 16:1535313. [PMID: 40129990 PMCID: PMC11930809 DOI: 10.3389/fimmu.2025.1535313] [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: 11/27/2024] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
Cathepsin B (CTSB), a key cysteine protease, plays essential roles in physiological and pathological processes. As research progresses, interest in how CTSB triggers different types of programmed cell death (PCD) to induce the onset and development of diseases is increasing. Several recent studies suggest that different types of PCD mediated by CTSB play key roles in kidney diseases. In this review, we outline the fundamental mechanisms by which CTSB triggers different types of PCD in several kidney diseases and discuss the function of CTSB in various segments of the kidney. Moreover, we explore the possibilities and prospects of using CTSB as a therapeutic target for kidney diseases.
Collapse
Affiliation(s)
- Yunlong Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Yong Zhuang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Jie Shi
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Qi Lv
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Xiaoqin Guo
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| |
Collapse
|
2
|
Ponticelli C. Membranous Nephropathy. J Clin Med 2025; 14:761. [PMID: 39941432 PMCID: PMC11818350 DOI: 10.3390/jcm14030761] [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: 10/30/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 02/16/2025] Open
Abstract
Membranous nephropathy is a glomerular disease that may be caused by exogenous risk factors in genetically predisposed individuals (primary MN) or may be associated with other autoimmune diseases, drug exposure, or cytotoxic agents (secondary MN). Primary membranous nephropathy (PMN) is an autoimmune disease in which antigens-mainly the phospholipase A2 receptor-are located in the podocytes and are targeted by circulating antibodies, leading to in situ formation of immune complexes that activate the complement system. Clinically, the disease is characterized by nephrotic syndrome (NS) and associated complications. The outcome of PMN can vary, but untreated patients with NS may progress to end-stage kidney disease (ESKD) in 35-40% of cases within 10 years. Treatment primarily aims to prevent NS complications and progression to ESKD. The most commonly used immunosuppressive drugs are rituximab, corticosteroids, cyclophosphamide, and calcineurin inhibitors. Most patients may experience an improvement of proteinuria, which can sometimes be followed by NS relapse. Fewer than 50% of patients with PMN achieve complete and stable remission. In addition to immunosuppressive therapy, antiproteinuric, anti-lipemic, and anticoagulant medicaments are often required.
Collapse
|
3
|
Dong C, Huoshen W, Bai Y, Liu J, Li B, Guan Y, Luo P. Uncovering the molecular networks of ferroptosis in the pathogenesis of type 2 diabetes and its complications: a multi-omics investigation. Mol Med 2024; 30:268. [PMID: 39716081 DOI: 10.1186/s10020-024-01045-w] [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: 10/25/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Diabetes is a multi-factorial disorder and related complications constitute one of the principal causes of global mortality and disability. The role of ferroptosis in diabetes and its complications is intricate and significant. This study endeavors to disclose the role of ferroptosis in the aforementioned diseases from multiple perspectives through multi-omics. METHODS We performed genetic correlation analyses via the Linkage Disequilibrium Score and High-Definition Likelihood approaches for type 2 diabetes (T2D) and its complications. The data concerning the expression of ferroptosis-related genes (FRGs) were obtained from the meta-analysis of studies on gene expression and protein abundance. Mendelian randomization analyses and cross-validation were implemented using the discovery cohort, replication cohort, and imaging genomics cohort of T2D and its complications. Moreover, we conducted colocalization analyses on T2D and tissue-specific single-cell RNA sequencing investigations on the complications to complement the results. RESULTS Genetic association analysis indicated that the selected datasets could be incorporated into a secondary analysis of T2D complications. In the primary analysis, six FRGs (CDKN1A, ENO3, FURIN, RARRES2, TYRO3, and YTHDC2) were found to be positively associated with T2D risk. Conversely, eight FRGs (ARNTL, CAMKK2, CTSB, FADS2, KDM5A, MEG3, SREBF1, and STAT3) were inversely associated with T2D risk. The 14 FRGs were included in the secondary analysis. Within the FRGs, which received full support from both the discovery and replication cohorts, and were further validated by imaging genomics, higher levels of CDKN1A were positively associated with DKD risk. Higher levels of CAMKK2 and KDM5A were associated with a decreased risk of DKD. For DCM, higher levels of CTSB were positively associated with DCM risk. And genetically predicted higher levels of ARNTL and SREBF1 were associated with a decreased risk of NAFLD. Finally, we validated the tissue-specific expression of each complication with scRNA-seq datasets. CONCLUSIONS This study identified FRGs in relation to T2D and its complications, which may enhance the understanding of the pathogenic mechanisms of their development. Meanwhile, it offers cross-validation for imaging genomics and further indicates the direction for non-invasive diagnosis.
Collapse
Affiliation(s)
- Changqing Dong
- Department of Nephrology, National Key Laboratory of Diabetes, The Second Hospital of Jilin University, No. 991 Yatai Street, Nanguan District, Changchun, Jilin, China
| | - Wuda Huoshen
- School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Yunfeng Bai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, China
| | - Jiaona Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, China
| | - Bing Li
- Department of Nephrology, National Key Laboratory of Diabetes, The Second Hospital of Jilin University, No. 991 Yatai Street, Nanguan District, Changchun, Jilin, China
| | - Yucan Guan
- Department of Nephrology, National Key Laboratory of Diabetes, The Second Hospital of Jilin University, No. 991 Yatai Street, Nanguan District, Changchun, Jilin, China
| | - Ping Luo
- Department of Nephrology, National Key Laboratory of Diabetes, The Second Hospital of Jilin University, No. 991 Yatai Street, Nanguan District, Changchun, Jilin, China.
| |
Collapse
|
4
|
Kokesh KJ, Bala N, Dogan YE, Nguyen VAL, Costa M, Alli A. Mycobacterium avium inhibits protein kinase C and MARCKS phosphorylation in human cystic fibrosis and non-cystic fibrosis cells. PLoS One 2024; 19:e0308299. [PMID: 39413095 PMCID: PMC11482691 DOI: 10.1371/journal.pone.0308299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 07/03/2024] [Indexed: 10/18/2024] Open
Abstract
In cystic fibrosis (CF), there is abnormal translocation and function of the cystic fibrosis transmembrane conductance regulator (CFTR) and an upregulation of the epithelial sodium channel (ENaC). This leads to hyperabsorption of sodium and fluid from the airway, dehydrated mucus, and an increased risk of respiratory infections. In this study, we performed a proteomic assessment of differentially regulated proteins from CF and non-CF small airway epithelial cells (SAEC) that are sensitive to Mycobacterium avium. CF SAEC and normal non-CF SAEC were infected with M. avium before the cells were harvested for protein. Protein kinase C (PKC) activity was greater in the CF cells compared to the non-CF cells, but the activity was significantly attenuated in both cell types after infection with M. avium compared to vehicle. Western blot and densitometric analysis showed a significant increase in cathepsin B protein expression in M. avium infected CF cells. Myristoylated alanine rich C-kinase substrate (MARCKS) protein was one of several differentially expressed proteins between the groups that was identified by mass spectrometry-based proteomics. Total MARCKS protein expression was greater in CF cells compared to non-CF cells. Phosphorylation of MARCKS at serine 163 was also greater in CF cells compared to non-CF cells after treating both groups of cells with M. avium. Taken together, MARCKS protein is upregulated in CF cells and there is decreased phosphorylation of the protein due to a decrease in PKC activity and presumably increased cathepsin B mediated proteolysis of the protein after M. avium infection.
Collapse
Affiliation(s)
- Kevin J. Kokesh
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Niharika Bala
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Yunus E. Dogan
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Department of Pediatrics, Erciyes University of Medicine, Kayseri, Turkey
| | - Van-Anh L. Nguyen
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Marcus Costa
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Abdel Alli
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Department of Medicine, Division of Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida College of Medicine, Gainesville, Florida, United States of America
| |
Collapse
|
5
|
Cai Z, Xu S, Liu C. Cathepsin B in cardiovascular disease: Underlying mechanisms and therapeutic strategies. J Cell Mol Med 2024; 28:e70064. [PMID: 39248527 PMCID: PMC11382359 DOI: 10.1111/jcmm.70064] [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: 05/16/2024] [Revised: 08/01/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
Cathepsin B (CTSB) is a member of the cysteine protease family, primarily responsible for degrading unnecessary organelles and proteins within the acidic milieu of lysosomes to facilitate recycling. Recent research has revealed that CTSB plays a multifaceted role beyond its function as a proteolytic enzyme in lysosomes. Importantly, recent data suggest that CTSB has significant impacts on different cardiac pathological conditions, such as atherosclerosis (AS), myocardial infarction, hypertension, heart failure and cardiomyopathy. Especially in the context of AS, preclinical models and clinical sample imaging data indicate that the cathepsin activity-based probe can reliably image CTSB activity in foam cells and atherosclerotic plaques; concurrently, it allows synchronous diagnostic and therapeutic interventions. However, our knowledge of CTSB in cardiovascular disease is still in the early stage. This paper aims to provide a comprehensive review of the significance of CTSB in cardiovascular physiology and pathology, with the objective of laying a theoretical groundwork for the development of drugs targeting CTSB.
Collapse
Affiliation(s)
- Zhulan Cai
- Department of Cardiology, Peking University Third Hospital, Beijing, P.R. China
| | - Shunyao Xu
- Department of Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, P.R. China
| | - Chen Liu
- Department of Geriatrics, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, P.R. China
| |
Collapse
|
6
|
Schramm WC, Bala N, Arekar T, Malik Z, Chacko KM, Lewis RL, Denslow ND, Scindia Y, Alli AA. Enrichment of Bioactive Lipids in Urinary Extracellular Vesicles and Evidence of Apoptosis in Kidneys of Hypertensive Diabetic Cathepsin B Knockout Mice after Streptozotocin Treatment. Biomedicines 2024; 12:1038. [PMID: 38791000 PMCID: PMC11117475 DOI: 10.3390/biomedicines12051038] [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: 02/01/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
Cathepsin B (CtsB) is a ubiquitously expressed cysteine protease that plays important roles in health and disease. Urinary extracellular vesicles (uEVs) are released from cells associated with urinary organs. The antibiotic streptozotocin (STZ) is known to induce pancreatic islet beta cell destruction, diabetic nephropathy, and hypertension. We hypothesized that streptozotocin-induced diabetic kidney disease and hypertension result in the release of bioactive lipids from kidney cells that induce oxidative stress and renal cell death. Lipidomics was performed on uEVs isolated from CtsB knockout mice treated with or without STZ, and their kidneys were used to investigate changes in proteins associated with cell death. Lysophosphatidylethanolamine (LPE) (18:1), lysophosphatidylserine (LPS) (22:6), and lysophosphatidylglycerol (LPG) (22:5) were among the bioactive lipids enriched in uEVs from CtsB knockout mice treated with STZ compared to untreated CtsB mice (n = 3 uEV preparations per group). Anti-oxidant programming was activated in the kidneys of the CtsB knockout mice treated with STZ, as indicated by increased expression of glutathione peroxidase 4 (GPX4) and the cystine/glutamate antiporter SLC7A11 (XCT) (n = 4 mice per group), which was supported by a higher reactivity to 4-hydroxy-2-nonenal (4-HNE), a marker for oxidative stress (n = 3 mice per group). Apoptosis but not ferroptosis was the ongoing form of cell death in these kidneys as cleaved caspase-3 levels were significantly elevated in the STZ-treated CtsB knockout mice (n = 4 mice per group). There were no appreciable differences in the pro-ferroptosis enzyme acyl-CoA synthetase long-chain family member 4 (ACSL4) or the inflammatory marker CD93 in the kidneys (n = 3 mice per group), which further supports apoptosis as the prevalent mechanism of pathology. These data suggest that STZ treatment leads to oxidative stress, inducing apoptotic injury in the kidneys during the development of diabetic kidney disease and hypertension.
Collapse
Affiliation(s)
- Whitney C. Schramm
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Niharika Bala
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Tanmay Arekar
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
| | - Zeeshan Malik
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kevin M. Chacko
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Russell L. Lewis
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32608, USA; (R.L.L.); (N.D.D.)
| | - Nancy D. Denslow
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32608, USA; (R.L.L.); (N.D.D.)
| | - Yogesh Scindia
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Abdel A. Alli
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
7
|
Nell D, Wolf R, Podgorny PM, Kuschnereit T, Kuschnereit R, Dabers T, Stracke S, Schmidt T. Complement Activation in Nephrotic Glomerular Diseases. Biomedicines 2024; 12:455. [PMID: 38398059 PMCID: PMC10886869 DOI: 10.3390/biomedicines12020455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/23/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
The nephrotic syndrome holds significant clinical importance and is characterized by a substantial protein loss in the urine. Damage to the glomerular basement membrane or podocytes frequently underlies renal protein loss. There is an increasing belief in the involvement of the complement system, a part of the innate immune system, in these conditions. Understanding the interactions between the complement system and glomerular structures continually evolves, challenging the traditional view of the blood-urine barrier as a passive filter. Clinical studies suggest that a precise inhibition of the complement system at various points may soon become feasible. However, a thorough understanding of current knowledge is imperative for planning future therapies in nephrotic glomerular diseases such as membranous glomerulopathy, membranoproliferative glomerulonephritis, lupus nephritis, focal segmental glomerulosclerosis, and minimal change disease. This review provides an overview of the complement system, its interactions with glomerular structures, and insights into specific glomerular diseases exhibiting a nephrotic course. Additionally, we explore new diagnostic tools and future therapeutic approaches.
Collapse
|
8
|
Aufy M, Hussein AM, Stojanovic T, Studenik CR, Kotob MH. Proteolytic Activation of the Epithelial Sodium Channel (ENaC): Its Mechanisms and Implications. Int J Mol Sci 2023; 24:17563. [PMID: 38139392 PMCID: PMC10743461 DOI: 10.3390/ijms242417563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Epithelial sodium channel (ENaC) are integral to maintaining salt and water homeostasis in various biological tissues, including the kidney, lung, and colon. They enable the selective reabsorption of sodium ions, which is a process critical for controlling blood pressure, electrolyte balance, and overall fluid volume. ENaC activity is finely controlled through proteolytic activation, a process wherein specific enzymes, or proteases, cleave ENaC subunits, resulting in channel activation and increased sodium reabsorption. This regulatory mechanism plays a pivotal role in adapting sodium transport to different physiological conditions. In this review article, we provide an in-depth exploration of the role of proteolytic activation in regulating ENaC activity. We elucidate the involvement of various proteases, including furin-like convertases, cysteine, and serine proteases, and detail the precise cleavage sites and regulatory mechanisms underlying ENaC activation by these proteases. We also discuss the physiological implications of proteolytic ENaC activation, focusing on its involvement in blood pressure regulation, pulmonary function, and intestinal sodium absorption. Understanding the mechanisms and consequences of ENaC proteolytic activation provides valuable insights into the pathophysiology of various diseases, including hypertension, pulmonary disorders, and various gastrointestinal conditions. Moreover, we discuss the potential therapeutic avenues that emerge from understanding these mechanisms, offering new possibilities for managing diseases associated with ENaC dysfunction. In summary, this review provides a comprehensive discussion of the intricate interplay between proteases and ENaC, emphasizing the significance of proteolytic activation in maintaining sodium and fluid balance in both health and disease.
Collapse
Affiliation(s)
- Mohammed Aufy
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
| | - Ahmed M. Hussein
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
- Department of Zoology, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt
| | - Tamara Stojanovic
- Programme for Proteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Christian R. Studenik
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
| | - Mohamed H. Kotob
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
- Department of Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut 71515, Egypt
| |
Collapse
|
9
|
Dogan YE, Bala N, Chacko KM, Tuna KM, Alli AA. Tempol treatment normalizes membrane expression of epithelial transport proteins in the kidney of salt-loaded hypertensive diabetic db/db mice. Am J Transl Res 2023; 15:6690-6700. [PMID: 38186979 PMCID: PMC10767517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/14/2023] [Indexed: 01/09/2024]
Abstract
OBJECTIVE Hypertension exacerbates the progression and severity of diabetic kidney disease. In this study, we addressed the hypothesis that tempol acts at multiple segments of the nephron to normalize the abundance of sodium coupled epithelial transport proteins in the luminal plasma membrane to mitigate high blood pressure in salt-loaded hypertensive diabetic db/db mice. METHODS Soluble and membrane fractions from freshly homogenized kidney cortex tissue samples were resolved by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and probed for specific proteins by Western blotting. Western blotting for specific urinary extracellular vesicle (uEV) markers and nanoparticle tracking analysis was performed to characterize each uEV preparation from each group. A one-way ANOVA was performed to determine statistical significance between three groups (hypertensive diabetic db/db mice treated with vehicle, hypertensive diabetic db/db mice treated with tempol, and wild-type mice). RESULTS Tempol treatment reduced systolic blood pressure in hypertensive diabetic db/db mice compared to db/db mice that received vehicle. We observed attenuated membrane protein expression of the sodium hydrogen exchanger 3 (NHE3), sodium potassium chloride co-transporter (NKCC2), sodium chloride cotransporter (NCC), and epithelial sodium channel (ENaC) in the kidney of salt-loaded hypertensive diabetic db/db mice infused with tempol by osmotic minipump for 5 days compared to hypertensive diabetic db/db mice infused with vehicle. Also, the infusion of tempol in hypertensive diabetic db/db mice reduced the augmented protein expression of protein kinase c (PKC) epsilon observed in the vehicle treated hypertensive diabetic db/db kidney when compared to the healthy wild-type kidney. The amount of uEV and their size profiles were comparable between the three groups. CONCLUSIONS This study demonstrates that tempol down-regulates epithelial transport mechanisms in each segment of the nephron and normalizes salt-induced high blood pressure in diabetic animals presumably in a PKC dependent manner.
Collapse
Affiliation(s)
- Yunus E Dogan
- Department of Physiology and Aging, College of Medicine, University of FloridaGainesville Florida 32610, USA
- Department of Pediatrics, Faculty of Medicine, Erciyes UniversityKayseri, Turkey
| | - Niharika Bala
- Department of Physiology and Aging, College of Medicine, University of FloridaGainesville Florida 32610, USA
| | - Kevin M Chacko
- Department of Physiology and Aging, College of Medicine, University of FloridaGainesville Florida 32610, USA
| | - Kubra M Tuna
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, College of Medicine, University of FloridaGainesville Florida 32610, USA
| | - Abdel A Alli
- Department of Physiology and Aging, College of Medicine, University of FloridaGainesville Florida 32610, USA
- Department of Medicine, Division of Nephrology, Hypertension and Renal Transplantation, College of Medicine, University of FloridaGainesville Florida 32610, USA
| |
Collapse
|
10
|
Claudio P, Gabriella M. Nephrotic syndrome: pathophysiology and consequences. J Nephrol 2023; 36:2179-2190. [PMID: 37466816 DOI: 10.1007/s40620-023-01697-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/30/2023] [Indexed: 07/20/2023]
Abstract
In patients with kidney disease, nephrotic syndrome can lead to several complications including progressive kidney dysfunction. Proteinuria may lead to the formation of cellular or fibrous crescents with reciprocal development of rapidly progressive glomerulonephritis or focal glomerulosclerosis. Proteinuria may also cause overload and dysfunction of tubular epithelial cells, eventually resulting in tubular atrophy and interstitial fibrosis. Hypoalbuminemia is usually associated with increased risk of mortality and kidney dysfunction. Dyslipidemia may increase the risk of atherosclerotic complications, cause podocyte dysfunction and contribute to vascular thrombosis. Urinary loss of anticoagulants and overproduction of coagulation factors may facilitate a hypercoagulable state. Edema, hypogammaglobulinemia, loss of complement factors, and immunosuppressive therapy can favor infection. Treatment of these complications may reduce their impact on the severity of NS. Nephrotic syndrome is a kidney disorder that can worsen the quality of life and increase the risk of kidney disease progression.
Collapse
Affiliation(s)
| | - Moroni Gabriella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- Nephrology and Dialysis Division, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| |
Collapse
|
11
|
van Duijvenboden S, Ramírez J, Young WJ, Olczak KJ, Ahmed F, Alhammadi MJAY, Bell CG, Morris AP, Munroe PB. Integration of genetic fine-mapping and multi-omics data reveals candidate effector genes for hypertension. Am J Hum Genet 2023; 110:1718-1734. [PMID: 37683633 PMCID: PMC10577090 DOI: 10.1016/j.ajhg.2023.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 09/10/2023] Open
Abstract
Genome-wide association studies of blood pressure (BP) have identified >1,000 loci, but the effector genes and biological pathways at these loci are mostly unknown. Using published association summary statistics, we conducted annotation-informed fine-mapping incorporating tissue-specific chromatin segmentation and colocalization to identify causal variants and candidate effector genes for systolic BP, diastolic BP, and pulse pressure. We observed 532 distinct signals associated with ≥2 BP traits and 84 with all three. For >20% of signals, a single variant accounted for >75% posterior probability, 65 were missense variants in known (SLC39A8, ADRB2, and DBH) and previously unreported BP candidate genes (NRIP1 and MMP14). In disease-relevant tissues, we colocalized >80 and >400 distinct signals for each BP trait with cis-eQTLs and regulatory regions from promoter capture Hi-C, respectively. Integrating mouse, human disorder, gene expression and tissue abundance data, and literature review, we provide consolidated evidence for 436 BP candidate genes for future functional validation and discover several potential drug targets.
Collapse
Affiliation(s)
- Stefan van Duijvenboden
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, UK; Institute of Cardiovascular Science, University College London, London, UK; Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Julia Ramírez
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, UK; Aragon Institute of Engineering Research, University of Zaragoza, Zaragoza, Spain; Centro de Investigación Biomédica en Red - Bioingeniería, Biomateriales y Nanomedicina, Zaragoza, Spain
| | - William J Young
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, UK; Barts Heart Centre, St Bartholomew's Hospital, EC1A 7BE London, UK
| | - Kaya J Olczak
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, UK
| | - Farah Ahmed
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, UK
| | | | - Christopher G Bell
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, UK
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK; National Institute of Health and Care Research, Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, UK; National Institute of Health and Care Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, EC1M 6BQ London, UK.
| |
Collapse
|
12
|
Wang J, Zheng M, Yang X, Zhou X, Zhang S. The Role of Cathepsin B in Pathophysiologies of Non-tumor and Tumor tissues: A Systematic Review. J Cancer 2023; 14:2344-2358. [PMID: 37576397 PMCID: PMC10414043 DOI: 10.7150/jca.86531] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Cathepsin B (CTSB), a lysosomal cysteine protease, plays an important role in human physiology and pathology. CTSB is associated with various human diseases, and its expression level and activity are closely related to disease progression and severity. Physiologically, CTSB is integrated into almost all lysosome-related processes, including protein turnover, degradation, and lysosome-mediated cell death. CTSB can lead to the development of various pathological processes through degradation and remodeling of the extracellular matrix. During tumor development and progression, CTSB has two opposing effects. Its pro-apoptotic properties reduce malignancy, while its proteolytic enzymatic activity promotes invasion and metastasis, thereby inducing malignancy. Here, we discuss the roles of CTSB in tumor and non-tumor disease pathophysiologies. We conclude that targeting the activity or expression of CTSB may be important for treating tumor and non-tumor diseases.
Collapse
Affiliation(s)
- Jiangping Wang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, P.R. China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300071, P.R. China
| | - Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, P.R. China
| | - Xinyue Zhou
- Graduate School, Tianjin Medical University, Tianjin, 300070, P.R. China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300071, P.R. China
| |
Collapse
|
13
|
Kawakibi T, Bala N, Liu LP, Searcy LA, Denslow ND, Alli AA. Decreased MARCKS Protein Expression in Kidney Cortex Membrane Fractions of Cathepsin B Knockout Mice Is Associated with Reduced Lysophosphatidylcholine and Protein Kinase C Activity. Biomedicines 2023; 11:1489. [PMID: 37239160 PMCID: PMC10216610 DOI: 10.3390/biomedicines11051489] [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: 02/28/2023] [Revised: 04/28/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Cathpesin B is a multi-functional protease that plays numerous roles in physiology and pathophysiology. We hypothesized that actin cytoskeleton proteins that are substrates of cathepsin B, various lipids, and kinases that are regulated by lipids would be down-regulated in the kidney of cathepsin B knockout mice. Here, we show by Western blot and densitometric analysis that the expression and proteolysis of the actin cytoskeleton proteins myristoylated alanine-rich C-kinase substrate (MARCKS) and spectrin are significantly reduced in kidney cortex membrane fractions of cathepsin B knockout mice compared to C57B6 wild-type control mice. Lipidomic results show that specific lipids are increased while other lipids, including lysophosphatidylcholine (LPC) species LPC (16:0), LPC (18:0), LPC (18:1), and LPC (18:2), are significantly decreased in membrane fractions of the kidney cortex from Cathepsin B null mice. Protein Kinase C (PKC) activity is significantly lower in the kidney cortex of cathepsin B knockout mice compared to wild-type mice, while calcium/calmodulin-dependent protein kinase II (CaMKII) activity and phospholipase D (PLD) activity are comparable between the two groups. Together, these results provide the first evidence of altered actin cytoskeleton organization, membrane lipid composition, and PKC activity in the kidneys of mice lacking cathepsin B.
Collapse
Affiliation(s)
- Tamim Kawakibi
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Niharika Bala
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Lauren P. Liu
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Louis A. Searcy
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| | - Nancy D. Denslow
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| | - Abdel A. Alli
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida College of Medicine, Gainesville, FL 32610, USA
| |
Collapse
|
14
|
Scindia YM, Gholam MF, Waleed A, Liu LP, Chacko KM, Desai D, Lopez JP, Malik Z, Schramm WC, Morales AG, Carson-Marino M, Alli AA. Metformin Alleviates Diabetes-Associated Hypertension by Attenuating the Renal Epithelial Sodium Channel. Biomedicines 2023; 11:305. [PMID: 36830842 PMCID: PMC9953274 DOI: 10.3390/biomedicines11020305] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Diabetic nephropathy is the primary cause of morbidity in type 2 diabetes mellitus (T2DM) patients. New data indicate that hypertension, a common comorbidity in T2DM, can worsen outcomes of diabetic nephropathy. While metformin is a commonly prescribed drug for treating type 2 diabetes, its blood pressure regulating ability is not well documented. The aim of this study was to investigate the effect of metformin on normalizing blood pressure in salt-loaded hypertensive diabetic db/db mice. Sixteen-week-old male and female diabetic db/db mice were individually placed in metabolic cages and then randomized to a control vehicle (saline) or metformin treatment group. We evaluated the blood pressure reducing ability of metformin in salt-induced hypertension and progression of nephropathy in db/db mice. We observed that metformin- normalized systolic blood pressure in hypertensive diabetic mice. Mechanistically, metformin treatment reduced renal cathepsin B expression. Low cathepsin B expression was associated with reduced expression and activity of the epithelial sodium channel (ENaC), sodium retention, and thus control of hypertension. In addition, we identified that urinary extracellular vesicles (EVs) from the diabetic mice are enriched in cathepsin B. Compared to treatment with urinary EVs of vehicle-treated hypertensive diabetic mice, the amiloride-sensitive transepithelial current was significantly attenuated upon exposure of renal collecting duct cells to urinary EVs isolated from metformin-treated db/db mice or cathepsin B knockout mice. Collectively, our study identifies a novel blood pressure reducing role of metformin in diabetic nephropathy by regulating the cathepsin B-ENaC axis.
Collapse
Affiliation(s)
- Yogesh M. Scindia
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Pathology, University of Florida, Gainesville, FL 32610, USA
| | - Mohammed F. Gholam
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah 21423, Saudi Arabia
| | - Alina Waleed
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Lauren P. Liu
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kevin M. Chacko
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Dhruv Desai
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Juliana Pena Lopez
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Zeeshan Malik
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Whitney C. Schramm
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Angelica G. Morales
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Morgan Carson-Marino
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Abdel A. Alli
- Division of Nephrology, Hypertension and Renal Transplantation, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
15
|
Lugo CI, Liu LP, Bala N, Morales AG, Gholam MF, Abchee JC, Elmoujahid N, Elshikha AS, Avdiaj R, Searcy LA, Denslow ND, Song S, Alli AA. Human Alpha-1 Antitrypsin Attenuates ENaC and MARCKS and Lowers Blood Pressure in Hypertensive Diabetic db/db Mice. Biomolecules 2022; 13:66. [PMID: 36671451 PMCID: PMC9856210 DOI: 10.3390/biom13010066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/17/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Hypertension may develop before or after the onset of diabetes and it is known to increase the risk of developing diabetic nephropathy. Alpha-1 antitrypsin (AAT) is a multi-functional protein with beneficial effects in various diseases but its role in reducing blood pressure in the diabetic kidney has not been thoroughly studied. Like blood pressure, epithelial sodium channels (ENaC) and its adaptor protein myristoylated alanine-rich C-kinase substrate (MARCKS) are regulated by circadian rhythms. Our hypothesis is that administration of human AAT (hAAT) reduces blood pressure in hypertensive diabetic mice by attenuating membrane expression of ENaC and its association with the actin cytoskeleton. First, we show hAAT administration results in reduced blood pressure in diabetic db/db mice compared to vehicle treatment in both the inactive and active cycles. Western blotting and immunohistochemistry analyses showed a reduction of ENaC and the actin cytoskeleton protein, MARCKS in the kidneys of diabetic db/db mice treated with hAAT compared to vehicle. hAAT treatment resulted in elevated amounts of extracellular vesicles present in the urine of diabetic db/db mice compared to vehicle treatment both in the inactive and active cycles. Multiple hexosylceramides, among other lipid classes increased in urinary EVs released from hAAT treated hypertensive diabetic mice compared to vehicle treated mice. Taken together, these data suggest hAAT treatment could normalize blood pressure in the diabetic kidney in a mechanism involving attenuation of renal ENaC and MARCKS protein expression and possibly ceramide metabolism to hexosylceramide in kidney cells.
Collapse
Affiliation(s)
- Carlos I. Lugo
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Lauren P. Liu
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Niharika Bala
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Angelica G. Morales
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Mohammed F. Gholam
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah 21423, Saudi Arabia
| | - Julia C. Abchee
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Nasseem Elmoujahid
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Ahmed Samir Elshikha
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL 32610, USA
| | - Rigena Avdiaj
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL 32610, USA
| | - Louis A. Searcy
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| | - Nancy D. Denslow
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida College of Veterinary Medicine, Gainesville, FL 32610, USA
| | - Sihong Song
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL 32610, USA
| | - Abdel A. Alli
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
- Department of Medicine Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida College of Medicine, Gainesville, FL 32610, USA
| |
Collapse
|
16
|
Yue Q, Al-Khalili O, Moseley A, Yoshigi M, Wynne BM, Ma H, Eaton DC. PIP 2 Interacts Electrostatically with MARCKS-like Protein-1 and ENaC in Renal Epithelial Cells. BIOLOGY 2022; 11:biology11121694. [PMID: 36552204 PMCID: PMC9774185 DOI: 10.3390/biology11121694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/30/2022] [Accepted: 10/17/2022] [Indexed: 11/25/2022]
Abstract
We examined the interaction of a membrane-associated protein, MARCKS-like Protein-1 (MLP-1), and an ion channel, Epithelial Sodium Channel (ENaC), with the anionic lipid, phosphatidylinositol 4, 5-bisphosphate (PIP2). We found that PIP2 strongly activates ENaC in excised, inside-out patches with a half-activating concentration of 21 ± 1.17 µM. We have identified 2 PIP2 binding sites in the N-terminus of ENaC β and γ with a high concentration of basic residues. Normal channel activity requires MLP-1's strongly positively charged effector domain to electrostatically sequester most of the membrane PIP2 and increase the local concentration of PIP2. Our previous data showed that ENaC covalently binds MLP-1 so PIP2 bound to MLP-1 would be near PIP2 binding sites on the cytosolic N terminal regions of ENaC. We have modified the charge structure of the PIP2 -binding domains of MLP-1 and ENaC and showed that the changes affect membrane localization and ENaC activity in a way consistent with electrostatic theory.
Collapse
Affiliation(s)
- Qiang Yue
- Division of Nephrology, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Otor Al-Khalili
- Division of Nephrology, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Auriel Moseley
- Division of Nephrology, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Masaaki Yoshigi
- Division of Nephrology & Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Brandi Michele Wynne
- Division of Nephrology & Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Heping Ma
- Department of Physiology, Emory University, Atlanta, GA 30322, USA
| | - Douglas C. Eaton
- Division of Nephrology, Department of Medicine, Emory University, Atlanta, GA 30322, USA
- Correspondence: ; Tel.: +1-404-727-4533; Fax: +1-404-727-3425
| |
Collapse
|
17
|
Liu J, Li X, Xu N, Han H, Li X. Role of ion channels in the mechanism of proteinuria (Review). Exp Ther Med 2022; 25:27. [PMID: 36561615 PMCID: PMC9748662 DOI: 10.3892/etm.2022.11726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Proteinuria is a common clinical manifestation of kidney diseases, such as glomerulonephritis, nephrotic syndrome, immunoglobulin A nephropathy and diabetic nephropathy. Therefore, proteinuria is considered to be a risk factor for renal dysfunction. Furthermore, proteinuria is also significantly associated with the progression of kidney diseases and increased mortality. Its occurrence is closely associated with damage to the structure of the glomerular filtration membrane. An impaired glomerular filtration membrane can affect the selective filtration function of the kidneys; therefore, several macromolecular substances, such as proteins, may pass through the filtration membrane and promote the manifestation of proteinuria. It has been reported that ion channels play a significant role in the mechanisms underlying proteinuria. Ion channel mutations or other dysfunctions have been implicated in several diseases, therefore ion channels could be used as major therapeutic targets. The mechanisms underlying the action of ion channels and ion transporters in proteinuria have been overlooked in the literature, despite their importance in identifying novel targets for treating proteinuria and delaying the progression of kidney diseases. The current review article focused on the four key ion channel groups, namely Na+, Ca2+, Cl- and K+ ion channels and the associated ion transporters.
Collapse
Affiliation(s)
- Jie Liu
- Department of Nephrology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Xuewei Li
- Department of Rheumatology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Ning Xu
- Department of Nephrology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Huirong Han
- Department of Anesthesiology, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Xiangling Li
- Department of Nephrology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China,Correspondence to: Professor Xiangling Li, Department of Nephrology, Affiliated Hospital of Weifang Medical University, 2428 Yu He Road, Weifang, Shandong 261000, P.R. China
| |
Collapse
|
18
|
Mannan-binding lectin serine protease-2 (MASP-2) in human kidney and its relevance for proteolytic activation of the epithelial sodium channel. Sci Rep 2022; 12:15955. [PMID: 36153401 PMCID: PMC9509361 DOI: 10.1038/s41598-022-20213-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/09/2022] [Indexed: 11/08/2022] Open
Abstract
AbstractProteolytic activation of the renal epithelial sodium channel (ENaC) is increased by aldosterone. The aldosterone-sensitive protease remains unidentified. In humans, elevated circulating aldosterone is associated with increased urinary extracellular vesicle (uEVs) excretion of mannan-binding lectin associated serine protease-2 (MASP-2). We hypothesized that MASP-2 is a physiologically relevant ENaC-activating protease. It was confirmed that MASP2 mRNA is abundantly present in liver but not in human and mouse kidneys. Aldosterone-stimulation of murine cortical colleting duct (mCCD) cells did not induce MASP-2 mRNA. In human kidney collecting duct, MASP-2 protein was detected in AQP2-negative/ATP6VB1-positive intercalated cells suggestive of MASP2 protein uptake. Plasma concentration of full-length MASP-2 and the short splice variant MAp19 were not changed in a cross-over intervention study in healthy humans with low (70 mmol/day) versus high (250 mmol/day) Na+ intake despite changes in aldosterone. The ratio of MAp19/MASP-2 in plasma was significantly increased with a high Na+ diet and the ratio correlated with changes in aldosterone and fractional Na+ excretion. MASP-2 was not detected in crude urine or in uEVs. MASP2 activated an amiloride-sensitive current when co-expressed with ENaC in Xenopus oocytes, but not when added to the bath solution. In monolayers of collecting duct M1 cells, MASP2 expression did not increase amiloride-sensitive current and in HEK293 cells, MASP-2 did not affect γENaC cleavage. MASP-2 is neither expressed nor co-localized and co-regulated with ENaC in the human kidney or in urine after low Na+ intake. MASP-2 does not mediate physiological ENaC cleavage in low salt/high aldosterone settings.
Collapse
|
19
|
Xiao M, Bohnert BN, Grahammer F, Artunc F. Rodent models to study sodium retention in experimental nephrotic syndrome. Acta Physiol (Oxf) 2022; 235:e13844. [PMID: 35569011 DOI: 10.1111/apha.13844] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 12/12/2022]
Abstract
Sodium retention and edema are hallmarks of nephrotic syndrome (NS). Different experimental rodent models have been established for simulating NS, however, not all of them feature sodium retention which requires proteinuria to exceed a certain threshold. In rats, puromycin aminonucleoside nephrosis (PAN) is a classic NS model introduced in 1955 that was adopted as doxorubicin-induced nephropathy (DIN) in 129S1/SvImJ mice. In recent years, mice with inducible podocin deletion (Nphs2Δipod ) or podocyte apoptosis (POD-ATTAC) have been developed. In these models, sodium retention is thought to be caused by activation of the epithelial sodium channel (ENaC) in the distal nephron through aberrantly filtered serine proteases or proteasuria. Strikingly, rodent NS models follow an identical chronological time course after the development of proteinuria featuring sodium retention within days and spontaneous reversal thereafter. In DIN and Nphs2Δipod mice, inhibition of ENaC by amiloride or urinary serine protease activity by aprotinin prevents sodium retention, opening up new and promising therapeutic approaches that could be translated into the treatment of nephrotic patients. However, the essential serine protease(s) responsible for ENaC activation is (are) still unknown. With the use of nephrotic rodent models, there is the possibility that this (these) will be identified in the future. This review summarizes the various rodent models used to study experimental nephrotic syndrome and the insights gained from these models with regard to the pathophysiology of sodium retention.
Collapse
Affiliation(s)
- Mengyun Xiao
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine University Hospital Tübingen Tübingen Germany
| | - Bernhard N. Bohnert
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine University Hospital Tübingen Tübingen Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University Tübingen Tübingen Germany
- German Center for Diabetes Research (DZD) at the University Tübingen Tübingen Germany
| | - Florian Grahammer
- III. Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Ferruh Artunc
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine University Hospital Tübingen Tübingen Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University Tübingen Tübingen Germany
- German Center for Diabetes Research (DZD) at the University Tübingen Tübingen Germany
| |
Collapse
|
20
|
Anand D, Hummler E, Rickman OJ. ENaC activation by proteases. Acta Physiol (Oxf) 2022; 235:e13811. [PMID: 35276025 PMCID: PMC9540061 DOI: 10.1111/apha.13811] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022]
Abstract
Proteases are fundamental for a plethora of biological processes, including signalling and tissue remodelling, and dysregulated proteolytic activity can result in pathogenesis. In this review, we focus on a subclass of membrane‐bound and soluble proteases that are defined as channel‐activating proteases (CAPs), since they induce Na+ ion transport through an autocrine mechanism when co‐expressed with the highly amiloride‐sensitive epithelial sodium channel (ENaC) in Xenopus oocytes. These experiments first identified CAP1 (channel‐activating protease 1, prostasin) followed by CAP2 (channel‐activating protease 2, TMPRSS4) and CAP3 (channel‐activating protease 3, matriptase) as in vitro mediators of ENaC current. Since then, more serine‐, cysteine‐ and metalloproteases were confirmed as in vitro CAPs that potentially cleave and regulate ENaC, and thus this nomenclature was not further followed, but is accepted as functional term or alias. The precise mechanism of ENaC modulation by proteases has not been fully elucidated. Studies in organ‐specific protease knockout models revealed evidence for their role in increasing ENaC activity, although the proteases responsible for ENaC activation are yet to be identified. We summarize recent findings in animal models of these CAPs with respect to their implication in ENaC activation. We discuss the consequences of dysregulated CAPs underlying epithelial phenotypes in pathophysiological conditions, and the role of selected protease inhibitors. We believe that these proteases may present interesting therapeutic targets for diseases with aberrant sodium homoeostasis.
Collapse
Affiliation(s)
- Deepika Anand
- Department of Biomedical Sciences University of Lausanne Lausanne Switzerland
- National Center of Competence in Research, Kidney.CH Lausanne Switzerland
| | - Edith Hummler
- Department of Biomedical Sciences University of Lausanne Lausanne Switzerland
- National Center of Competence in Research, Kidney.CH Lausanne Switzerland
| | - Olivia J. Rickman
- Department of Biomedical Sciences University of Lausanne Lausanne Switzerland
- National Center of Competence in Research, Kidney.CH Lausanne Switzerland
| |
Collapse
|
21
|
Saudenova M, Promnitz J, Ohrenschall G, Himmerkus N, Böttner M, Kunke M, Bleich M, Theilig F. Behind every smile there's teeth: Cathepsin B's function in health and disease with a kidney view. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119190. [PMID: 34968578 DOI: 10.1016/j.bbamcr.2021.119190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
Cathepsin B (CatB) is a very abundant lysosomal protease with endo- and carboxydipeptidase activities and even ligase features. In this review, we will provide a general characterization of CatB and describe structure, structure-derived properties and location-dependent proteolytic actions. We depict CatB action within lysosome and its important roles in lysosomal biogenesis, lysosomal homeostasis and autophagy rendering this protease a key player in orchestrating lysosomal functions. Lysosomal leakage and subsequent escape of CatB into the cytosol lead to harmful actions, e.g. the role in activating the NLPR3 inflammasome, affecting immune responses and cell death. The second focus of this review addresses CatB functions in the kidney, i.e. the glomerulus, the proximal tubule and collecting duct with strong emphasis of its role in pathology of the respective segment. Finally, observations regarding CatB functions that need to be considered in cell culture will be discussed. In conclusion, CatB a physiologically important molecule may, upon aberrant expression in different cellular context, become a harmful player effectively showing its teeth behind its smile.
Collapse
Affiliation(s)
- Makhabbat Saudenova
- Institute of Anatomy, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Jessica Promnitz
- Institute of Anatomy, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Gerrit Ohrenschall
- Institute of Anatomy, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Nina Himmerkus
- Institute of Physiology, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Martina Böttner
- Institute of Anatomy, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Madlen Kunke
- Institute of Anatomy, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Markus Bleich
- Institute of Physiology, Department of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Franziska Theilig
- Institute of Anatomy, Department of Medicine, Christian-Albrechts-University Kiel, Germany.
| |
Collapse
|
22
|
Liu LP, Gholam MF, Elshikha AS, Kawakibi T, Elmoujahid N, Moussa HH, Song S, Alli AA. Transgenic Mice Overexpressing Human Alpha-1 Antitrypsin Exhibit Low Blood Pressure and Altered Epithelial Transport Mechanisms in the Inactive and Active Cycles. Front Physiol 2021; 12:710313. [PMID: 34630137 PMCID: PMC8493122 DOI: 10.3389/fphys.2021.710313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/20/2021] [Indexed: 11/13/2022] Open
Abstract
Human alpha-1 antitrypsin (hAAT) is a versatile protease inhibitor, but little is known about its targets in the aldosterone-sensitive distal nephron and its role in electrolyte balance and blood pressure control. We analyzed urinary electrolytes, osmolality, and blood pressure from hAAT transgenic (hAAT-Tg) mice and C57B/6 wild-type control mice maintained on either a normal salt or high salt diet. Urinary sodium, potassium, and chloride concentrations as well as urinary osmolality were lower in hAAT-Tg mice maintained on a high salt diet during both the active and inactive cycles. hAAT-Tg mice showed a lower systolic blood pressure compared to C57B6 mice when maintained on a normal salt diet but this was not observed when they were maintained on a high salt diet. Cathepsin B protein activity was less in hAAT-Tg mice compared to wild-type controls. Protein expression of the alpha subunit of the sodium epithelial channel (ENaC) alpha was also reduced in the hAAT-Tg mice. Natriuretic peptide receptor C (NPRC) protein expression in membrane fractions of the kidney cortex was reduced while circulating levels of atrial natriuretic peptide (ANP) were greater in hAAT-Tg mice compared to wild-type controls. This study characterizes the electrolyte and blood pressure phenotype of hAAT-Tg mice during the inactive and active cycles and investigates the mechanism by which ENaC activation is inhibited in part by a mechanism involving decreased cathepsin B activity and increased ANP levels in the systemic circulation.
Collapse
Affiliation(s)
- Lauren P Liu
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Mohammed F Gholam
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Ahmed Samir Elshikha
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Tamim Kawakibi
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Nasseem Elmoujahid
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Hassan H Moussa
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Sihong Song
- Department of Pharmaceutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Abdel A Alli
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States.,Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
23
|
Bohnert BN, Essigke D, Janessa A, Schneider JC, Wörn M, Kalo MZ, Xiao M, Kong L, Omage K, Hennenlotter J, Amend B, Birkenfeld AL, Artunc F. Experimental nephrotic syndrome leads to proteolytic activation of the epithelial Na + channel in the mouse kidney. Am J Physiol Renal Physiol 2021; 321:F480-F493. [PMID: 34423678 DOI: 10.1152/ajprenal.00199.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Proteolytic activation of the renal epithelial Na+ channel (ENaC) involves cleavage events in its α- and γ-subunits and is thought to mediate Na+ retention in nephrotic syndrome (NS). However, the detection of proteolytically processed ENaC in kidney tissue from nephrotic mice has been elusive so far. We used a refined Western blot technique to reliably discriminate full-length α-ENaC and γ-ENaC and their cleavage products after proteolysis at their proximal and distal cleavage sites (designated from the NH2-terminus), respectively. Proteolytic ENaC activation was investigated in kidneys from mice with experimental NS induced by doxorubicin or inducible podocin deficiency with or without treatment with the serine protease inhibitor aprotinin. Nephrotic mice developed Na+ retention and increased expression of fragments of α-ENaC and γ-ENaC cleaved at both the proximal cleavage site and, more prominently, the distal cleavage site, respectively. Treatment with aprotinin but not with the mineralocorticoid receptor antagonist canrenoate prevented Na+ retention and upregulation of the cleavage products in nephrotic mice. Increased expression of cleavage products of α-ENaC and γ-ENaC was similarly found in healthy mice treated with a low-salt diet, sensitive to mineralocorticoid receptor blockade. In human nephrectomy specimens, γ-ENaC was found in the full-length form and predominantly cleaved at its distal cleavage site. In conclusion, murine experimental NS leads to aprotinin-sensitive proteolytic activation of ENaC at both proximal and, more prominently, distal cleavage sites of its α- and γ-subunit, most likely by urinary serine protease activity or proteasuria.NEW & NOTEWORTHY This study demonstrates that murine experimental nephrotic syndrome leads to aprotinin-sensitive proteolytic activation of the epithelial Na+ channel at both the α- and γ-subunit, most likely by urinary serine protease activity or proteasuria.
Collapse
Affiliation(s)
- Bernhard N Bohnert
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany.,Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University Tübingen, Tübingen, Germany.,German Center for Diabetes Research, University Tübingen, Tübingen, Germany
| | - Daniel Essigke
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Andrea Janessa
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Jonas C Schneider
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Matthias Wörn
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - M Zaher Kalo
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Mengyun Xiao
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Lingsi Kong
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Kingsley Omage
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Jörg Hennenlotter
- Department of Urology, University Hospital Tübingen, Tübingen, Germany
| | - Bastian Amend
- Department of Urology, University Hospital Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany.,Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University Tübingen, Tübingen, Germany.,German Center for Diabetes Research, University Tübingen, Tübingen, Germany
| | - Ferruh Artunc
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany.,Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University Tübingen, Tübingen, Germany.,German Center for Diabetes Research, University Tübingen, Tübingen, Germany
| |
Collapse
|
24
|
Nouri MZ, Yu L, Liu L, Chacko KM, Denslow ND, LaDisa JF, Alli AA. Increased endothelial sodium channel activity by extracellular vesicles in human aortic endothelial cells: Putative role of MLP1 and bioactive lipids. Am J Physiol Cell Physiol 2021; 321:C535-C548. [PMID: 34288724 DOI: 10.1152/ajpcell.00092.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) contain biological molecules and are secreted by cells into the extracellular milieu. The endothelial sodium channel (EnNaC) plays an important role in modulating endothelial cell stiffness. We hypothesized EVs secreted from human aortic endothelial cells (hAoEC) positively regulate EnNaC in an autocrine dependent manner. A comprehensive lipidomic analysis using targeted mass spectrometry was performed on multiple preparations of EVs isolated from the conditioned media of hAoEC or complete growth media of these cells. Cultured hAoEC challenged with EVs isolated from the conditioned media of these cells resulted in an increase in EnNaC activity when compared to the same concentration of media derived EVs or vehicle alone. EVs isolated from the conditioned media of hAoEC but not human fibroblast cells were enriched in MARCKS Like Protein 1 (MLP1). The pharmacological inhibition of the negative regulator of MLP1, protein kinase C, in cultured hAoEC resulted in an increase in EV size and release compared to vehicle or pharmacological inhibition of protein kinase D. The MLP1 enriched EVs increased the density of actin filaments in cultured hAoEC compared to EVs isolated from human fibroblast cells lacking MLP1. We quantified 141 lipids from glycerolipids, glycerophospholipids, and sphingolipids in conditioned media EVs that represented twice the number found in control media EVs. The concentrations of sphingomyelin, lysophosphatidylcholine and phosphatidylethanolamine were higher in conditioned media EVs. These results provide the first evidence for EnNaC regulation in hAoEC by EVs and provide insight into a possible mechanism involving MLP1, unsaturated lipids, and bioactive lipids.
Collapse
Affiliation(s)
- Mohammad-Zaman Nouri
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, United States
| | - Ling Yu
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States
| | - Lauren Liu
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States
| | - Kevin M Chacko
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States
| | - Nancy D Denslow
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL, United States
| | - John F LaDisa
- Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Abdel A Alli
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States.,Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL, United States
| |
Collapse
|
25
|
McKelvey MC, Brown R, Ryan S, Mall MA, Weldon S, Taggart CC. Proteases, Mucus, and Mucosal Immunity in Chronic Lung Disease. Int J Mol Sci 2021; 22:5018. [PMID: 34065111 PMCID: PMC8125985 DOI: 10.3390/ijms22095018] [Citation(s) in RCA: 10] [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: 04/13/2021] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
Dysregulated protease activity has long been implicated in the pathogenesis of chronic lung diseases and especially in conditions that display mucus obstruction, such as chronic obstructive pulmonary disease, cystic fibrosis, and non-cystic fibrosis bronchiectasis. However, our appreciation of the roles of proteases in various aspects of such diseases continues to grow. Patients with muco-obstructive lung disease experience progressive spirals of inflammation, mucostasis, airway infection and lung function decline. Some therapies exist for the treatment of these symptoms, but they are unable to halt disease progression and patients may benefit from novel adjunct therapies. In this review, we highlight how proteases act as multifunctional enzymes that are vital for normal airway homeostasis but, when their activity becomes immoderate, also directly contribute to airway dysfunction, and impair the processes that could resolve disease. We focus on how proteases regulate the state of mucus at the airway surface, impair mucociliary clearance and ultimately, promote mucostasis. We discuss how, in parallel, proteases are able to promote an inflammatory environment in the airways by mediating proinflammatory signalling, compromising host defence mechanisms and perpetuating their own proteolytic activity causing structural lung damage. Finally, we discuss some possible reasons for the clinical inefficacy of protease inhibitors to date and propose that, especially in a combination therapy approach, proteases represent attractive therapeutic targets for muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Michael C. McKelvey
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Ryan Brown
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Sinéad Ryan
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany;
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- German Center for Lung Research (DZL), 35392 Gießen, Germany
| | - Sinéad Weldon
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK; (M.C.M.); (R.B.); (S.R.); (S.W.)
| |
Collapse
|
26
|
Xiao M, Bohnert BN, Aypek H, Kretz O, Grahammer F, Aukschun U, Wörn M, Janessa A, Essigke D, Daniel C, Amann K, Huber TB, Plow EF, Birkenfeld AL, Artunc F. Plasminogen deficiency does not prevent sodium retention in a genetic mouse model of experimental nephrotic syndrome. Acta Physiol (Oxf) 2021; 231:e13512. [PMID: 32455507 DOI: 10.1111/apha.13512] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/19/2022]
Abstract
AIM Sodium retention is the hallmark of nephrotic syndrome (NS) and mediated by the proteolytic activation of the epithelial sodium channel (ENaC) by aberrantly filtered serine proteases. Plasmin is highly abundant in nephrotic urine and has been proposed to be the principal serine protease responsible for ENaC activation in NS. However, a proof of the essential role of plasmin in experimental NS is lacking. METHODS We used a genetic mouse model of NS based on an inducible podocin knockout (Bl6-Nphs2tm3.1Antc *Tg(Nphs1-rtTA*3G)8Jhm *Tg(tetO-cre)1Jaw or nphs2Δipod ). These mice were crossed with plasminogen deficient mice (Bl6-Plgtm1Jld or plg-/- ) to generate double knockout mice (nphs2Δipod *plg-/- ). NS was induced after oral doxycycline treatment for 14 days and mice were followed for subsequent 14 days. RESULTS Uninduced nphs2Δipod *plg-/- mice had normal kidney function and sodium handling. After induction, proteinuria increased similarly in both nphs2Δipod *plg+/+ and nphs2Δipod *plg-/- mice. Western blot revealed the urinary excretion of plasminogen and plasmin in nphs2Δipod *plg+/+ mice which were absent in nphs2Δipod *plg-/- mice. After the onset of proteinuria, amiloride-sensitive natriuresis was increased compared to the uninduced state in both genotypes. Subsequently, urinary sodium excretion dropped in both genotypes leading to an increase in body weight and development of ascites. Treatment with the serine protease inhibitor aprotinin prevented sodium retention in both genotypes. CONCLUSIONS This study shows that mice lacking urinary plasminogen are not protected from ENaC-mediated sodium retention in experimental NS. This points to an essential role of other urinary serine proteases in the absence of plasminogen.
Collapse
Affiliation(s)
- Mengyun Xiao
- Department of Internal Medicine Division of Endocrinology, Diabetology and Nephrology University Hospital Tübingen Tübingen Germany
| | - Bernhard N. Bohnert
- Department of Internal Medicine Division of Endocrinology, Diabetology and Nephrology University Hospital Tübingen Tübingen Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University Tübingen Tübingen Germany
- German Center for Diabetes Research (DZD) at the University Tübingen Tübingen Germany
| | - Hande Aypek
- III. Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Oliver Kretz
- III. Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Florian Grahammer
- III. Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Ute Aukschun
- IV. Department of Medicine, Faculty and University Medical Center Freiburg Freiburg Germany
| | - Matthias Wörn
- Department of Internal Medicine Division of Endocrinology, Diabetology and Nephrology University Hospital Tübingen Tübingen Germany
| | - Andrea Janessa
- Department of Internal Medicine Division of Endocrinology, Diabetology and Nephrology University Hospital Tübingen Tübingen Germany
| | - Daniel Essigke
- Department of Internal Medicine Division of Endocrinology, Diabetology and Nephrology University Hospital Tübingen Tübingen Germany
| | - Christoph Daniel
- Institute of Nephropathology Friedrich‐Alexander University Erlangen‐Nürnberg (FAU) Erlangen Germany
| | - Kerstin Amann
- Institute of Nephropathology Friedrich‐Alexander University Erlangen‐Nürnberg (FAU) Erlangen Germany
| | - Tobias B. Huber
- III. Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Edward F. Plow
- Lerner Research InstituteCleveland Clinic Cleveland OH USA
| | - Andreas L. Birkenfeld
- Department of Internal Medicine Division of Endocrinology, Diabetology and Nephrology University Hospital Tübingen Tübingen Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University Tübingen Tübingen Germany
- German Center for Diabetes Research (DZD) at the University Tübingen Tübingen Germany
| | - Ferruh Artunc
- Department of Internal Medicine Division of Endocrinology, Diabetology and Nephrology University Hospital Tübingen Tübingen Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University Tübingen Tübingen Germany
- German Center for Diabetes Research (DZD) at the University Tübingen Tübingen Germany
| |
Collapse
|
27
|
Proteasuria in nephrotic syndrome-quantification and proteomic profiling. J Proteomics 2020; 230:103981. [PMID: 32927112 DOI: 10.1016/j.jprot.2020.103981] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/24/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023]
Abstract
Nephrotic syndrome is characterized by urinary excretion of plasma proteases or proteasuria. There is a lack of data on the quantity, activity status and identity of these aberrantly filtered proteases. We established a fluorescence-based substrate assay to quantify protease activity in urine samples from healthy and nephrotic humans and mice. Protease class activity was determined after addition of specific inhibitors. Individual proteases were identified by tandem mass spectrometry (MS/MS). In spot urine samples from 10 patients with acute nephrotic syndrome of various etiology, urinary protease activity was significantly increased compared to that of healthy persons (753 ± 178 vs. 244 ± 65 relative units, p < 0.05). The corresponding proteases were highly sensitive to inhibition by the serine protease inhibitors AEBSF (reduction by 85 ± 6% and 72 ± 8%, respectively) and aprotinin (83 ± 9% vs. 25 ± 6%, p < 0.05). MS/MS of all urinary proteins or after AEBSF purification showed that most of them were active serine proteases from the coagulation and complement cascade. These findings were recapitulated in mice, pointing to a similar pathophysiology. In conclusion, nephrotic syndrome leads to increased urinary excretion of active plasma proteases which can be termed proteasuria. Serine proteases account for the vast majority of urinary protease activity in health and nephrotic syndrome. SIGNIFICANCE STATEMENT: In this study, we found that nephrotic urine samples of humans and mice have a significantly increased protease activity compared to healthy urine samples, using a universal pentapeptide substrate library. This was driven by increased excretion of aprotinin-sensitive serine proteases. With tandem mass spectrometry, we provide a comprehensive and systematic overview of all urinary proteases or the "urine proteasome". We identified renally expressed proteases in health and addition of proteases from the coagulation and complement cascade in the nephrotic state. These results set the basis to study the role of urinary proteases at both health and nephrotic syndrome to find diagnostic markers of renal disease as well as possible therapeutic targets.
Collapse
|
28
|
Vizovišek M, Vidak E, Javoršek U, Mikhaylov G, Bratovš A, Turk B. Cysteine cathepsins as therapeutic targets in inflammatory diseases. Expert Opin Ther Targets 2020; 24:573-588. [PMID: 32228244 DOI: 10.1080/14728222.2020.1746765] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Cysteine cathepsins are involved in the development and progression of numerous inflammation-associated diseases such as cancer, arthritis, bone and immune disorders. Consequently, there is a drive to progress research efforts focused on cathepsin use in diagnostics and as therapeutic targets in disease.Areas covered: This review discusses the potential of cysteine cathepsins as therapeutic targets in inflammation-associated diseases and recent advances in preclinical and clinical research. We describe direct targeting of cathepsins for treatment purposes and their indirect use in diagnostics.Expert opinion: The targeting of cysteine cathepsins has not translated into the clinic; this failure is attributed to off- and on-target side effects and/or the lack of companion biomarkers. This field now embraces developments in diagnostic imaging, the activation of prodrugs and antibody-drug conjugates for targeted drug delivery. The future lies in improved molecular tools and therapeutic concepts that will find a wide spectrum of uses in diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Matej Vizovišek
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Ljubljana, Slovenia.,Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Eva Vidak
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Ljubljana, Slovenia.,Jozef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Urban Javoršek
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Ljubljana, Slovenia.,Jozef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Georgy Mikhaylov
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Andreja Bratovš
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Ljubljana, Slovenia.,Jozef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Ljubljana, Slovenia.,Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
29
|
Van JAD, Clotet-Freixas S, Zhou J, Batruch I, Sun C, Glogauer M, Rampoldi L, Elia Y, Mahmud FH, Sochett E, Diamandis EP, Scholey JW, Konvalinka A. Peptidomic Analysis of Urine from Youths with Early Type 1 Diabetes Reveals Novel Bioactivity of Uromodulin Peptides In Vitro. Mol Cell Proteomics 2020; 19:501-517. [PMID: 31879271 PMCID: PMC7050109 DOI: 10.1074/mcp.ra119.001858] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/20/2019] [Indexed: 12/17/2022] Open
Abstract
Chronic hyperglycemia is known to disrupt the proteolytic milieu, initiating compensatory and maladaptive pathways in the diabetic kidney. Such changes in intrarenal proteolysis are captured by the urinary peptidome. To elucidate the early kidney response to chronic hyperglycemia, we conducted a peptidomic investigation into urines from otherwise healthy youths with type 1 diabetes and their non-diabetic peers using unbiased and targeted mass spectrometry-based techniques. This cross-sectional study included two separate cohorts for the discovery (n = 30) and internal validation (n = 30) of differential peptide excretion. Peptide bioactivity was predicted using PeptideRanker and subsequently verified in vitro Proteasix and the Nephroseq database were used to identify putative proteases responsible for peptide generation and examine their expression in diabetic nephropathy. A total of 6550 urinary peptides were identified in the discovery analysis. We further examined the subset of 162 peptides, which were quantified across all thirty samples. Of the 15 differentially excreted peptides (p < 0.05), seven derived from a C-terminal region (589SGSVIDQSRVLNLGPITRK607) of uromodulin, a kidney-specific protein. Increased excretion of five uromodulin peptides was replicated in the validation cohort using parallel reaction monitoring (p < 0.05). One of the validated peptides (SGSVIDQSRVLNLGPI) activated NFκB and AP-1 signaling, stimulated cytokine release, and enhanced neutrophil migration in vitro. In silico analyses highlighted several potential proteases such as hepsin, meprin A, and cathepsin B to be responsible for generating these peptides. In summary, we identified a urinary signature of uromodulin peptides associated with early type 1 diabetes before clinical manifestations of kidney disease and discovered novel bioactivity of uromodulin peptides in vitro Our present findings lay the groundwork for future studies to validate peptide excretion in larger and broader populations, to investigate the role of bioactive uromodulin peptides in high glucose conditions, and to examine proteases that cleave uromodulin.
Collapse
Affiliation(s)
- Julie A D Van
- Institute of Medical Science, University of Toronto, Toronto, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.
| | - Sergi Clotet-Freixas
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Joyce Zhou
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Ihor Batruch
- Department of Laboratory Medicine and Pathobiology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada
| | - Chunxiang Sun
- Faculty of Dentistry, University of Toronto, Toronto, Canada
| | | | - Luca Rampoldi
- Molecular Genetics of Renal Disorders Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | | | | | | | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada; Department of Clinical Biochemistry, University Health Network, University of Toronto, Toronto, Canada
| | - James W Scholey
- Institute of Medical Science, University of Toronto, Toronto, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Department of Medicine, Division of Nephrology, University Health Network, Toronto, Canada
| | - Ana Konvalinka
- Institute of Medical Science, University of Toronto, Toronto, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Department of Medicine, Division of Nephrology, University Health Network, Toronto, Canada
| |
Collapse
|
30
|
|
31
|
Kleyman TR, Eaton DC. Regulating ENaC's gate. Am J Physiol Cell Physiol 2020; 318:C150-C162. [PMID: 31721612 PMCID: PMC6985836 DOI: 10.1152/ajpcell.00418.2019] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/24/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023]
Abstract
Epithelial Na+ channels (ENaCs) are members of a family of cation channels that function as sensors of the extracellular environment. ENaCs are activated by specific proteases in the biosynthetic pathway and at the cell surface and remove embedded inhibitory tracts, which allows channels to transition to higher open-probability states. Resolved structures of ENaC and an acid-sensing ion channel revealed highly organized extracellular regions. Within the periphery of ENaC subunits are unique domains formed by antiparallel β-strands containing the inhibitory tracts and protease cleavage sites. ENaCs are inhibited by Na+ binding to specific extracellular site(s), which promotes channel transition to a lower open-probability state. Specific inositol phospholipids and channel modification by Cys-palmitoylation enhance channel open probability. How these regulatory factors interact in a concerted manner to influence channel open probability is an important question that has not been resolved. These various factors are reviewed, and the impact of specific factors on human disorders is discussed.
Collapse
Affiliation(s)
- Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, and Departments of Cell Biology and of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Douglas C Eaton
- Division of Nephrology, Department of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
32
|
Hinrichs GR, Weyer K, Friis UG, Svenningsen P, Lund IK, Nielsen R, Mollet G, Antignac C, Bistrup C, Jensen BL, Birn H. Urokinase-type plasminogen activator contributes to amiloride-sensitive sodium retention in nephrotic range glomerular proteinuria in mice. Acta Physiol (Oxf) 2019; 227:e13362. [PMID: 31423748 DOI: 10.1111/apha.13362] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/12/2019] [Accepted: 08/15/2019] [Indexed: 12/12/2022]
Abstract
AIM Activation of sodium reabsorption by urinary proteases has been implicated in sodium retention associated with nephrotic syndrome. The study was designed to test the hypothesis that nephrotic proteinuria in mice after conditional deletion of podocin leads to urokinase-dependent, amiloride-sensitive plasmin-mediated sodium and water retention. METHODS Ten days after podocin knockout, urine and faeces were collected for 10 days in metabolic cages and analysed for electrolytes, plasminogen, protease activity and ability to activate γENaC by patch clamp and western blot. Mice were treated with amiloride (2.5 mg kg-1 for 2 days and 10 mg kg-1 for 2 days) or an anti-urokinase-type plasminogen activator (uPA) targeting antibody (120 mg kg-1 /24 h) and compared to controls. RESULTS Twelve days after deletion, podocin-deficient mice developed significant protein and albuminuria associated with increased body wt, ascites, sodium accumulation and suppressed plasma renin. This was associated with increased urinary excretion of plasmin and plasminogen that correlated with albumin excretion, urine protease activity co-migrating with active plasmin, and the ability of urine to induce an amiloride-sensitive inward current in M1 cells in vitro. Amiloride treatment in podocin-deficient mice resulted in weight loss, increased sodium excretion, normalization of sodium balance and prevention of the activation of plasminogen to plasmin in urine in a reversible way. Administration of uPA targeting antibody abolished urine activation of plasminogen, attenuated sodium accumulation and prevented cleavage of γENaC. CONCLUSIONS Nephrotic range glomerular proteinuria leads to urokinase-dependent intratubular plasminogen activation and γENaC cleavage which contribute to sodium accumulation.
Collapse
Affiliation(s)
- Gitte R. Hinrichs
- Department of Molecular Medicine, Cardiovascular and Renal Research University of Southern Denmark Odense Denmark
| | - Kathrin Weyer
- Department of Biomedicine Aarhus University Aarhus Denmark
| | - Ulla G. Friis
- Department of Molecular Medicine, Cardiovascular and Renal Research University of Southern Denmark Odense Denmark
| | - Per Svenningsen
- Department of Molecular Medicine, Cardiovascular and Renal Research University of Southern Denmark Odense Denmark
| | - Ida K. Lund
- The Finsen Laboratory Rigshospitalet Copenhagen Denmark
- Biotech Research & Innovation Centre (BRIC) University of Copenhagen Copenhagen Denmark
| | - Rikke Nielsen
- Department of Biomedicine Aarhus University Aarhus Denmark
| | - Géraldine Mollet
- Laboratory of Hereditary Kidney Diseases Imagine Institute Inserm, U1163 Paris Descartes‐Sorbonne Paris Cité University Paris France
| | - Corinne Antignac
- Laboratory of Hereditary Kidney Diseases Imagine Institute Inserm, U1163 Paris Descartes‐Sorbonne Paris Cité University Paris France
- Department of Genetics Necker Hospital Assistance Publique‐Hôpitaux de Paris Paris France
| | - Claus Bistrup
- Department of Nephrology Odense University Hospital Odense Denmark
- Department of Clinical Research University of Southern Denmark Odense Denmark
| | - Boye L. Jensen
- Department of Molecular Medicine, Cardiovascular and Renal Research University of Southern Denmark Odense Denmark
| | - Henrik Birn
- Department of Biomedicine Aarhus University Aarhus Denmark
- Department of Renal Medicine Aarhus University Hospital Aarhus Denmark
| |
Collapse
|
33
|
Larionov A, Dahlke E, Kunke M, Zanon Rodriguez L, Schiessl IM, Magnin JL, Kern U, Alli AA, Mollet G, Schilling O, Castrop H, Theilig F. Cathepsin B increases ENaC activity leading to hypertension early in nephrotic syndrome. J Cell Mol Med 2019; 23:6543-6553. [PMID: 31368174 PMCID: PMC6787568 DOI: 10.1111/jcmm.14387] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/04/2019] [Accepted: 05/06/2019] [Indexed: 11/28/2022] Open
Abstract
The NPHS2 gene, encoding the slit diaphragm protein podocin, accounts for genetic and sporadic forms of nephrotic syndrome (NS). Patients with NS often present symptoms of volume retention, such as oedema formation or hypertension. The primary dysregulation in sodium handling involves an inappropriate activation of the epithelial sodium channel, ENaC. Plasma proteases in a proteinuria‐dependent fashion have been made responsible; however, referring to the timeline of symptoms occurring and underlying mechanisms, contradictory results have been published. Characterizing the mouse model of podocyte inactivation of NPHS2 (Nphs2∆pod) with respect to volume handling and proteinuria revealed that sodium retention, hypertension and gross proteinuria appeared sequentially in a chronological order. Detailed analysis of Nphs2∆pod during early sodium retention, revealed increased expression of full‐length ENaC subunits and αENaC cleavage product with concomitant increase in ENaC activity as tested by amiloride application, and augmented collecting duct Na+/K+‐ATPase expression. Urinary proteolytic activity was increased and several proteases were identified by mass spectrometry including cathepsin B, which was found to process αENaC. Renal expression levels of precursor and active cathepsin B were increased and could be localized to glomeruli and intercalated cells. Inhibition of cathepsin B prevented hypertension. With the appearance of gross proteinuria, plasmin occurs in the urine and additional cleavage of γENaC is encountered. In conclusion, characterizing the volume handling of Nphs2∆pod revealed early sodium retention occurring independent to aberrantly filtered plasma proteases. As an underlying mechanism cathepsin B induced αENaC processing leading to augmented channel activity and hypertension was identified.
Collapse
Affiliation(s)
- Alexey Larionov
- Institute of Anatomy, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Eileen Dahlke
- Institute of Anatomy, Christian Albrechts-University Kiel, Kiel, Germany
| | - Madlen Kunke
- Institute of Anatomy, Christian Albrechts-University Kiel, Kiel, Germany
| | | | - Ina M Schiessl
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | | | - Ursula Kern
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Abdel A Alli
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Geraldine Mollet
- Laboratory of Hereditary Kidney Diseases, INSERM UMR 1163, Université Paris Descartes-Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Oliver Schilling
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,BIOSS Center for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Franziska Theilig
- Institute of Anatomy, Department of Medicine, University of Fribourg, Fribourg, Switzerland.,Institute of Anatomy, Christian Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|