1
|
Zhao Q, Huang Y, Fu N, Cui C, Peng X, Kang H, Xiao J, Ke G. Podocyte senescence: from molecular mechanisms to therapeutics. Ren Fail 2024; 46:2398712. [PMID: 39248407 PMCID: PMC11385655 DOI: 10.1080/0886022x.2024.2398712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024] Open
Abstract
As an important component of the glomerular filtration membrane, the state of the podocytes is closely related to kidney function, they are also key cells involved in aging and play a central role in the damage caused by renal aging. Therefore, understanding the aging process of podocytes will allow us to understand their susceptibility to injury and identify targeted protective mechanisms. In fact, the process of physiological aging itself can induce podocyte senescence. Pathological stresses, such as oxidative stress, mitochondrial damage, secretion of senescence-associated secretory phenotype, reduced autophagy, oncogene activation, altered transcription factors, DNA damage response, and other factors, play a crucial role in inducing premature senescence and accelerating aging. Senescence-associated-β-galactosidase (SA-β-gal) is a marker of aging, and β-hydroxybutyric acid treatment can reduce SA-β-gal activity to alleviate cellular senescence and damage. In addition, CCAAT/enhancer-binding protein-α, transforming growth factor-β signaling, glycogen synthase kinase-3β, cycle-dependent kinase, programmed cell death protein 1, and plasminogen activator inhibitor-1 are closely related to aging. The absence or elevation of these factors can affect aging through different mechanisms. Podocyte injury is not an independent process, and injured podocytes interact with the surrounding epithelial cells or other kidney cells to mediate the injury or loss of podocytes. In this review, we discuss the manifestations, molecular mechanisms, biomarkers, and therapeutic drugs for podocyte senescence. We included elamipretide, lithium, calorie restriction, rapamycin; and emerging treatment strategies, such as gene and immune therapies. More importantly, we summarize how podocyte interact with other kidney cells.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongzhang Huang
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ningying Fu
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Caixia Cui
- Department of Nephrology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Xuan Peng
- Department of Nephrology, Affiliated Hospital/Clinical Medical College of Chengdu University, Chengdu, China
| | - Haiyan Kang
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jie Xiao
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guibao Ke
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
2
|
Richfield O, Cortez R, Navar LG. Modeling the interaction between tubuloglomerular feedback and myogenic mechanisms in the control of glomerular mechanics. Front Physiol 2024; 15:1410764. [PMID: 38966231 PMCID: PMC11223525 DOI: 10.3389/fphys.2024.1410764] [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: 04/01/2024] [Accepted: 05/24/2024] [Indexed: 07/06/2024] Open
Abstract
Introduction: Mechanical stresses and strains exerted on the glomerular cells have emerged as potentially influential factors in the progression of glomerular disease. Renal autoregulation, the feedback process by which the afferent arteriole changes in diameter in response to changes in blood pressure, is assumed to control glomerular mechanical stresses exerted on the glomerular capillaries. However, it is unclear how the two major mechanisms of renal autoregulation, the afferent arteriole myogenic mechanism and tubuloglomerular feedback (TGF), each contribute to the maintenance of glomerular mechanical homeostasis. Methods: In this study, we made a mathematical model of renal autoregulation and combined this model with an anatomically accurate model of glomerular blood flow and filtration, developed previously by us. We parameterized the renal autoregulation model based on data from previous literature, and we found evidence for an increased myogenic mechanism sensitivity when TGF is operant, as has been reported previously. We examined the mechanical effects of each autoregulatory mechanism (the myogenic, TGF and modified myogenic) by simulating blood flow through the glomerular capillary network with and without each mechanism operant. Results: Our model results indicate that the myogenic mechanism plays a central role in maintaining glomerular mechanical homeostasis, by providing the most protection to the glomerular capillaries. However, at higher perfusion pressures, the modulation of the myogenic mechanism sensitivity by TGF is crucial for the maintenance of glomerular mechanical homeostasis. Overall, a loss of renal autoregulation increases mechanical strain by up to twofold in the capillaries branching off the afferent arteriole. This further corroborates our previous simulation studies, that have identified glomerular capillaries nearest to the afferent arteriole as the most prone to mechanical injury in cases of disturbed glomerular hemodynamics. Discussion: Renal autoregulation is a complex process by which multiple feedback mechanisms interact to control blood flow and filtration in the glomerulus. Importantly, our study indicates that another function of renal autoregulation is control of the mechanical stresses on the glomerular cells, which indicates that loss or inhibition of renal autoregulation may have a mechanical effect that may contribute to glomerular injury in diseases such as hypertension or diabetes. This study highlights the utility of mathematical models in integrating data from previous experimental studies, estimating variables that are difficult to measure experimentally (i.e. mechanical stresses in microvascular networks) and testing hypotheses that are historically difficult or impossible to measure.
Collapse
Affiliation(s)
- Owen Richfield
- Bioinnovation PhD Program, Tulane University, New Orleans, LA, United States
| | - Ricardo Cortez
- Department of Mathematics, Tulane University, New Orleans, LA, United States
| | - L. Gabriel Navar
- Department of Physiology, Tulane School of Medicine, New Orleans, LA, United States
| |
Collapse
|
3
|
Mohandes S, Doke T, Hu H, Mukhi D, Dhillon P, Susztak K. Molecular pathways that drive diabetic kidney disease. J Clin Invest 2023; 133:165654. [PMID: 36787250 PMCID: PMC9927939 DOI: 10.1172/jci165654] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
Kidney disease is a major driver of mortality among patients with diabetes and diabetic kidney disease (DKD) is responsible for close to half of all chronic kidney disease cases. DKD usually develops in a genetically susceptible individual as a result of poor metabolic (glycemic) control. Molecular and genetic studies indicate the key role of podocytes and endothelial cells in driving albuminuria and early kidney disease in diabetes. Proximal tubule changes show a strong association with the glomerular filtration rate. Hyperglycemia represents a key cellular stress in the kidney by altering cellular metabolism in endothelial cells and podocytes and by imposing an excess workload requiring energy and oxygen for proximal tubule cells. Changes in metabolism induce early adaptive cellular hypertrophy and reorganization of the actin cytoskeleton. Later, mitochondrial defects contribute to increased oxidative stress and activation of inflammatory pathways, causing progressive kidney function decline and fibrosis. Blockade of the renin-angiotensin system or the sodium-glucose cotransporter is associated with cellular protection and slowing kidney function decline. Newly identified molecular pathways could provide the basis for the development of much-needed novel therapeutics.
Collapse
Affiliation(s)
- Samer Mohandes
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tomohito Doke
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hailong Hu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dhanunjay Mukhi
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Poonam Dhillon
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Mechanisms of podocyte injury and implications for diabetic nephropathy. Clin Sci (Lond) 2022; 136:493-520. [PMID: 35415751 PMCID: PMC9008595 DOI: 10.1042/cs20210625] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/25/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023]
Abstract
Albuminuria is the hallmark of both primary and secondary proteinuric glomerulopathies, including focal segmental glomerulosclerosis (FSGS), obesity-related nephropathy, and diabetic nephropathy (DN). Moreover, albuminuria is an important feature of all chronic kidney diseases (CKDs). Podocytes play a key role in maintaining the permselectivity of the glomerular filtration barrier (GFB) and injury of the podocyte, leading to foot process (FP) effacement and podocyte loss, the unifying underlying mechanism of proteinuric glomerulopathies. The metabolic insult of hyperglycemia is of paramount importance in the pathogenesis of DN, while insults leading to podocyte damage are poorly defined in other proteinuric glomerulopathies. However, shared mechanisms of podocyte damage have been identified. Herein, we will review the role of haemodynamic and oxidative stress, inflammation, lipotoxicity, endocannabinoid (EC) hypertone, and both mitochondrial and autophagic dysfunction in the pathogenesis of the podocyte damage, focussing particularly on their role in the pathogenesis of DN. Gaining a better insight into the mechanisms of podocyte injury may provide novel targets for treatment. Moreover, novel strategies for boosting podocyte repair may open the way to podocyte regenerative medicine.
Collapse
|
5
|
Sharma M, Singh V, Sharma R, Koul A, McCarthy ET, Savin VJ, Joshi T, Srivastava T. Glomerular Biomechanical Stress and Lipid Mediators during Cellular Changes Leading to Chronic Kidney Disease. Biomedicines 2022; 10:407. [PMID: 35203616 PMCID: PMC8962328 DOI: 10.3390/biomedicines10020407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 02/04/2023] Open
Abstract
Hyperfiltration is an important underlying cause of glomerular dysfunction associated with several systemic and intrinsic glomerular conditions leading to chronic kidney disease (CKD). These include obesity, diabetes, hypertension, focal segmental glomerulosclerosis (FSGS), congenital abnormalities and reduced renal mass (low nephron number). Hyperfiltration-associated biomechanical forces directly impact the cell membrane, generating tensile and fluid flow shear stresses in multiple segments of the nephron. Ongoing research suggests these biomechanical forces as the initial mediators of hyperfiltration-induced deterioration of podocyte structure and function leading to their detachment and irreplaceable loss from the glomerular filtration barrier. Membrane lipid-derived polyunsaturated fatty acids (PUFA) and their metabolites are potent transducers of biomechanical stress from the cell surface to intracellular compartments. Omega-6 and ω-3 long-chain PUFA from membrane phospholipids generate many versatile and autacoid oxylipins that modulate pro-inflammatory as well as anti-inflammatory autocrine and paracrine signaling. We advance the idea that lipid signaling molecules, related enzymes, metabolites and receptors are not just mediators of cellular stress but also potential targets for developing novel interventions. With the growing emphasis on lifestyle changes for wellness, dietary fatty acids are potential adjunct-therapeutics to minimize/treat hyperfiltration-induced progressive glomerular damage and CKD.
Collapse
Affiliation(s)
- Mukut Sharma
- Research and Development Service, Kansas City VA Medical Center, Kansas City, MO 64128, USA;
- Midwest Veterans’ Biomedical Research Foundation, Kansas City, MO 64128, USA; (A.K.); (V.J.S.); (T.S.)
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, MO 66160, USA;
| | - Vikas Singh
- Neurology, Kansas City VA Medical Center, Kansas City, MO 64128, USA;
| | - Ram Sharma
- Research and Development Service, Kansas City VA Medical Center, Kansas City, MO 64128, USA;
| | - Arnav Koul
- Midwest Veterans’ Biomedical Research Foundation, Kansas City, MO 64128, USA; (A.K.); (V.J.S.); (T.S.)
| | - Ellen T. McCarthy
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, MO 66160, USA;
| | - Virginia J. Savin
- Midwest Veterans’ Biomedical Research Foundation, Kansas City, MO 64128, USA; (A.K.); (V.J.S.); (T.S.)
| | - Trupti Joshi
- Department of Health Management and Informatics, University of Missouri, Columbia, MO 65201, USA;
| | - Tarak Srivastava
- Midwest Veterans’ Biomedical Research Foundation, Kansas City, MO 64128, USA; (A.K.); (V.J.S.); (T.S.)
- Section of Nephrology, Children’s Mercy Hospital and University of Missouri, Kansas City, MO 64108, USA
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, MO 64108, USA
| |
Collapse
|
6
|
Zhai Y, Li D, Wang Z, Shao L, Yin N, Li W. Cortex Mori Radicis attenuates streptozotocin-induced diabetic renal injury in mice via regulation of transient receptor potential canonical channel 6. Endocr Metab Immune Disord Drug Targets 2022; 22:862-873. [PMID: 35016601 DOI: 10.2174/1871530322666220110161458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/09/2021] [Accepted: 12/14/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Cortex Mori Radicis (CMR) has been reported to possess anti-pyretic, anti-convulsant, anti-allergic, anti-inflammatory, and anti-diabetic effects. In this study, we aimed to investigate the effect of CMR on streptozotocin (STZ)-induced diabetic renal injury in mice and explore the underlying mechanism. METHODS Mice were gavaged with different doses of CMR for continuous 7 days. Then, STZ (50 mg/kg) was applied to induce renal injury associated with type 1 diabetes. Firstly, blood glucose levels and metabolic parameters were evaluated, including weight, food intake, and excrement. HE and PAS staining were performed to present renal histological changes. Renal inflammation, fibrosis, and oxidative stress were assayed by real time PCR and ELISA, separately. Additionally, podocyte-related markers, such as nephrin and wilms' tumor-1 (WT-1) were detected by immunohistochemical staining and Western blot separately. Lastly, expression of transient receptor potential canonical channel 6 (TRPC6) and activation of MAPK signaling pathways were assayed. RESULTS CMR pretreatment significantly lowered the blood glucose levels, suppressed renal inflammation, fibrosis and oxidative stress, and relieved renal pathological injury, accompanying the inhibition of nephrin and WT-1 expression in STZ-induced diabetic mice. Moreover, CMR decreased the expression of TRPC6 and suppressed phosphorylation of ERK, but not P38 MAPK and JNK. Notably, the application of hyperforin, a specific activator of TRPC6, significantly abrogated the hypoglycemic effect of CMR and reversed the suppression of CMR on TRPC6 expression and ERK activation in the diabetic mice. CONCLUSION Our findings indicated that CMR attenuated early renal injury in STZ-induced diabetic mice through inhibiting ERK signaling via regulation of TRPC6, which suggests that CMR can be considered as a promising candidate for the management of diabetes-related renal complications.
Collapse
Affiliation(s)
- Yi Zhai
- Department of Cardiology, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Dan Li
- College of Acupuncture and Moxibustion, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Zhigang Wang
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Luyao Shao
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Nina Yin
- Department of Anatomy, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Weihua Li
- Department of Cardiology, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| |
Collapse
|
7
|
Richfield O, Cortez R, Navar LG. Simulations of Glomerular Shear and Hoop Stresses in Diabetes, Hypertension, and Reduced Renal Mass using a Network Model of a Rat Glomerulus. Physiol Rep 2021; 8:e14577. [PMID: 32951361 PMCID: PMC7507384 DOI: 10.14814/phy2.14577] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022] Open
Abstract
A novel anatomically accurate model of rat glomerular filtration is used to quantify shear stresses on the glomerular capillary endothelium and hoop stresses on the glomerular capillary walls. Plasma, erythrocyte volume, and plasma protein mass are distributed at network nodes using pressure differentials calculated taking into account volume loss to filtration, improving on previous models which only took into account blood apparent viscosity in calculating pressures throughout the network. Filtration is found to be heterogeneously distributed throughout the glomerular capillary network and is determined by concentration of plasma proteins and surface area of the filtering capillary segments. Hoop stress is primarily concentrated near the afferent arteriole, whereas shear stress is concentrated near the efferent arteriole. Using parameters from glomerular micropuncture studies, conditions of diabetes mellitus (DM), 5/6‐Nephrectomy (5/6‐Nx), and Angiotensin II‐induced hypertension (HTN) are simulated and compared to their own internal controls to assess the changes in mechanical stresses. Hoop stress is increased in all three conditions, while shear stress is increased in 5/6‐Nx, decreased in HTN, and maintained at control levels in DM by the hypertrophic response of the glomerular capillaries. The results indicate that these alterations in mechanical stresses and the consequent release of cytokines by or injury of the glomerular cells may play a significant role in the progression of glomerulopathy in these disease conditions.
Collapse
Affiliation(s)
- Owen Richfield
- Bioinnovation PhD Program, Tulane University, New Orleans, LA, USA.,Department of Physiology, Tulane School of Medicine, New Orleans, LA, USA
| | - Ricardo Cortez
- Department of Mathematics, Tulane University, New Orleans, LA, USA
| | - L Gabriel Navar
- Department of Physiology, Tulane School of Medicine, New Orleans, LA, USA
| |
Collapse
|
8
|
Richfield O, Cortez R, Navar LG. Simulations of increased glomerular capillary wall strain in the 5/6-nephrectomized rat. Microcirculation 2021; 28:e12721. [PMID: 34192389 PMCID: PMC9285434 DOI: 10.1111/micc.12721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/27/2021] [Accepted: 06/22/2021] [Indexed: 12/21/2022]
Abstract
Objective Chronic glomerular hypertension is associated with glomerular injury and sclerosis; however, the mechanism by which increases in pressure damage glomerular podocytes remains unclear. We tested the hypothesis that increases in glomerular pressure may deleteriously affect podocyte structural integrity by increasing the strain of the glomerular capillary walls, and that glomerular capillary wall strain may play a significant role in the perpetuation of glomerular injury in disease states that are associated with glomerular hypertension. Methods We developed an anatomically accurate mathematical model of a compliant, filtering rat glomerulus to quantify the strain of the glomerular capillary walls in a remnant glomerulus of the 5/6‐nephrectomized rat model of chronic kidney disease. In terms of estimating the mechanical stresses and strains in the glomerular capillaries, this mathematical model is a substantial improvement over previous models which do not consider pressure‐induced alterations in glomerular capillary diameters in distributing plasma and erythrocytes throughout the network. Results Using previously reported data from experiments measuring the change of glomerular volume as a function of perfusion pressure, we estimated the Young's modulus of the glomerular capillary walls in both control and 5/6‐nephrectomized conditions. We found that in 5/6‐nephrectomized conditions, the Young's modulus increased to 8.6 MPa from 7.8 MPa in control conditions, but the compliance of the capillaries increased in 5/6‐nephrectomized conditions due to a 23.3% increase in the baseline glomerular capillary diameters. We found that glomerular capillary wall strain was increased approximately threefold in 5/6‐nephrectomized conditions over control, which may deleteriously affect both mesangial cells and podocytes. The magnitudes of strain in model simulations of 5/6‐nephrectomized conditions were consistent with magnitudes of strain that elicit podocyte hypertrophy and actin cytoskeleton reorganization in vitro. Conclusions Our findings indicate that glomerular capillary wall strain may deleteriously affect podocytes directly, as well as act in concert with other mechanical changes and environmental factors inherent to the in vivo setting to potentiate glomerular injury in severe renoprival conditions.
Collapse
Affiliation(s)
- Owen Richfield
- Bioinnovation PhD Program, Tulane University, New Orleans, LA, USA.,Department of Physiology, Tulane School of Medicine, New Orleans, LA, USA
| | - Ricardo Cortez
- Department of Mathematics, Tulane University, New Orleans, LA, USA
| | - L Gabriel Navar
- Department of Physiology, Tulane School of Medicine, New Orleans, LA, USA
| |
Collapse
|
9
|
Chebotareva N, Bobkova I, Lysenko L, Moiseev S. Urinary Markers of Podocyte Dysfunction in Chronic Glomerulonephritis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1306:81-99. [PMID: 33959907 DOI: 10.1007/978-3-030-63908-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic glomerulonephritis (CGN) is a disease with a steady progressive course that involves the development of nephrosclerosis, which is especially evident in clinical courses with incidences of high proteinuria (PU). Currently, proteinuria is considered the main laboratory feature (sign) of CGN activity and progression because proteinuria is closely related to the process of tubulointerstitial fibrosis, which is correlated with the grade of renal insufficiency. The injury to podocytes, which are key components of the filtration barrier, plays a central role in proteinuria development. The detachment of podocytes from the glomerular basement membrane leading to podocytopenia is suggested to induce glomerulosclerosis and hyalinosis with obliteration of capillary loops and the progression of chronic kidney disease. Urinary markers of podocyte dysfunction could serve as useful tools while monitoring the activity and prognosis of CGN. In this chapter, the most important mechanisms of podocyte loss and urinary markers of this process are discussed.
Collapse
Affiliation(s)
- Natalia Chebotareva
- Tareev Clinic, Department of Nephrology, Sechenov First Moscow State Medical University, Moscow, Russia.
| | - Irina Bobkova
- Tareev Clinic, Department of Nephrology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Lidia Lysenko
- Tareev Clinic, Department of Nephrology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sergey Moiseev
- Tareev Clinic, Department of Nephrology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
10
|
Xu S, He L, Ding K, Zhang L, Xu X, Wang S, Qian X. Tanshinone IIA Ameliorates Streptozotocin-Induced Diabetic Nephropathy, Partly by Attenuating PERK Pathway-Induced Fibrosis. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:5773-5782. [PMID: 33408464 PMCID: PMC7780857 DOI: 10.2147/dddt.s257734] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022]
Abstract
Purpose Tanshinone IIA (Tan IIA), a compound extracted from Salvia miltiorrhiza, can improve type II diabetes, while the molecular mechanisms underlying Tan IIA-mediated protective effects in diabetic nephropathy are unclear. This study explored the protective actions of Tan IIA on renal tissues in streptozotocin (STZ)-induced diabetic nephropathy. Materials and Methods Tan IIA (2, 4, 8 mg/kg/day) was daily administered to STZ-treated rats by intraperitoneal injection for 42 days. The morphologic pathology was evaluated by hematoxylin-eosin and Masson’s trichrome staining, and transmission electron microscopy. The protein expression levels in renal tissues were evaluated by Western blotting and immunohistochemistry; the mRNA expression level was determined by quantitative real-time PCR. Results Tan IIA at 2 and 4 mg/kg attenuated the increase in the levels of uric acid and blood urea nitrogen and restored the reduction in the superoxide dismutase activity in the serum of the diabetic rats. Tan IIA at 2 and 4 mg/kg, but not 8 mg/kg, ameliorated the thickening of renal tubule in the diabetic rats; Tan IIA at 2 and 4 and 8 mg/kg attenuated the thickening of glomerular basement membrane and the collagen deposition in the renal tissues of the diabetic rats. Tan IIA treatment at 2, 4, 8 mg/kg decreased the expression levels of transforming growth factor-beta1, TSP-1, Grp78 and CHOP in the diabetic rats. Tan IIA at 2 and 4 and 8 mg/kg attenuated the increase in the protein levels of p-PERK, p-elf2α and ATF-4 from the renal tissues of diabetic rats, while the protein level of AFT-6 and the mRNA expression levels of XBP-1t, XBP-1s and p58IPK in the renal tissues were not affected by STZ or Tan IIA treatment. Conclusion Tan IIA-mediated protective effects on the STZ-induced diabetic nephropathy may be associated with the reduced endoplasmic reticulum stress via attenuating PERK signaling activities.
Collapse
Affiliation(s)
- Shujuan Xu
- Department of Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Lianjun He
- Precision Medicine Centre, Yijishan Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Keke Ding
- Department of Urology, Yijishan Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Lingling Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, People's Republic of China
| | - Xinhui Xu
- School of Pharmacy, Wannan Medical College, Wuhu, People's Republic of China
| | - Sheng Wang
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Xueyi Qian
- Precision Medicine Centre, Yijishan Hospital of Wannan Medical College, Wuhu, People's Republic of China
| |
Collapse
|
11
|
Bersie-Larson LM, Gyoneva L, Goodman DJ, Dorfman KD, Segal Y, Barocas VH. Glomerular filtration and podocyte tensional homeostasis: importance of the minor type IV collagen network. Biomech Model Mechanobiol 2020; 19:2433-2442. [PMID: 32462439 PMCID: PMC7606712 DOI: 10.1007/s10237-020-01347-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/13/2020] [Indexed: 03/05/2023]
Abstract
The minor type IV collagen chain, which is a significant component of the glomerular basement membrane in healthy individuals, is known to assemble into large structures (supercoils) that may contribute to the mechanical stability of the collagen network and the glomerular basement membrane as a whole. The absence of the minor chain, as in Alport syndrome, leads to glomerular capillary demise and eventually to kidney failure. An important consideration in this problem is that the glomerular capillary wall must be strong enough to withstand the filtration pressure and porous enough to permit filtration at reasonable pressures. In this work, we propose a coupled feedback loop driven by filtration demand and tensional homeostasis of the podocytes forming the outer portion of the glomerular capillary wall. Briefly, the deposition of new collagen increases the stiffness of basement membrane, helping to stress shield the podocytes, but the new collagen also decreases the permeability of the basement membrane, requiring an increase in capillary transmural pressure drop to maintain filtration; the resulting increased pressure outweighs the increased glomerular basement membrane stiffness and puts a net greater stress demand on the podocytes. This idea is explored by developing a multiscale simulation of the capillary wall, in which a macroscopic (µm scale) continuum model is connected to a set of microscopic (nm scale) fiber network models representing the collagen network and the podocyte cytoskeleton. The model considers two cases: healthy remodeling, in which the presence of the minor chain allows the collagen volume fraction to be increased by thickening fibers, and Alport syndrome remodeling, in which the absence of the minor chain allows collagen volume fraction to be increased only by adding new fibers to the network. The permeability of the network is calculated based on previous models of flow through a fiber network, and it is updated for different fiber radii and volume fractions. The analysis shows that the minor chain allows a homeostatic balance to be achieved in terms of both filtration and cell tension. Absent the minor chain, there is a fundamental change in the relation between the two effects, and the system becomes unstable. This result suggests that mechanobiological or mechanoregulatory therapies may be possible for Alport syndrome and other minor chain collagen diseases of the kidney.
Collapse
Affiliation(s)
- Lauren M Bersie-Larson
- Department of Biomedical Engineering, University of Minnesota, 7-105 Nils Hasselmo Hall, 312 Church St SE, Minneapolis, MN, 55455, USA
| | - Lazarina Gyoneva
- Department of Biomedical Engineering, University of Minnesota, 7-105 Nils Hasselmo Hall, 312 Church St SE, Minneapolis, MN, 55455, USA
| | - Daniel J Goodman
- Department of Biomedical Engineering, University of Minnesota, 7-105 Nils Hasselmo Hall, 312 Church St SE, Minneapolis, MN, 55455, USA
| | - Kevin D Dorfman
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| | - Yoav Segal
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - Victor H Barocas
- Department of Biomedical Engineering, University of Minnesota, 7-105 Nils Hasselmo Hall, 312 Church St SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
12
|
Flegeau K, Rubin S, Mucha S, Bur P, Préterre J, Siadous R, L'Azou B, Fricain JC, Combe C, Devillard R, Kalisky J, Rigothier C. Towards an in vitro model of the glomerular barrier unit with an innovative bioassembly method. Nephrol Dial Transplant 2020; 35:240-250. [PMID: 31121032 DOI: 10.1093/ndt/gfz094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 04/10/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The development of an artificial glomerular unit may be pivotal for renal pathophysiology studies at a multicellular scale. Using a tissue engineering approach, we aimed to reproduce in part the specific glomerular barrier architecture by manufacturing a glomerular microfibre (Mf). METHODS Immortalized human glomerular cell lines of endothelial cells (GEnCs) and podocytes were used. Cells and a three-dimensional (3D) matrix were characterized by immunofluorescence with confocal analysis, Western blot and polymerase chain reaction. Optical and electron microscopy were used to study Mf and cell shapes. We also analysed cell viability and cell metabolism within the 3D construct at 14 days. RESULTS Using the Mf manufacturing method, we repeatedly obtained a cellularized Mf sorting human glomerular cells in 3D. Around a central structure made of collagen I, we obtained an internal layer composed of GEnC, a newly formed glomerular basement membrane rich in α5 collagen IV and an external layer of podocytes. The cell concentration, optimal seeding time and role of physical stresses were modulated to obtain the Mf. Cell viability and expression of specific proteins (nephrin, synaptopodin, vascular endothelial growth factor receptor 2 (VEGFR2) and von Willebrandt factor (vWF)) were maintained for 19 days in the Mf system. Mf ultrastructure, observed with EM, had similarities with the human glomerular barrier. CONCLUSION In summary, with our 3D bio-engineered glomerular fibre, GEnC and podocytes produced a glomerular basement membrane. In the future, this glomerular Mf will allow us to study cell interactions in a 3D system and increase our knowledge of glomerular pathophysiology.
Collapse
Affiliation(s)
- Killian Flegeau
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France
| | - Sébastien Rubin
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France.,Service de Néphrologie Transplantation, Dialyse et Aphérèse, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Simon Mucha
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France.,Service de Néphrologie Transplantation, Dialyse et Aphérèse, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Pauline Bur
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France
| | - Julie Préterre
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France
| | - Robin Siadous
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France
| | - Béatrice L'Azou
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France
| | - Jean-Christophe Fricain
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France.,Service d'odontologie et de Santé Buccale, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Christian Combe
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France.,Service de Néphrologie Transplantation, Dialyse et Aphérèse, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Raphaël Devillard
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France.,Service d'odontologie et de Santé Buccale, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Jérôme Kalisky
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France
| | - Claire Rigothier
- Tissue Bioengineering, Université de Bordeaux, Bordeaux, France.,Tissue Bioengineering, INSERM, Bordeaux, France.,Service de Néphrologie Transplantation, Dialyse et Aphérèse, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| |
Collapse
|
13
|
Rinschen MM, Gödel M, Grahammer F, Zschiedrich S, Helmstädter M, Kretz O, Zarei M, Braun DA, Dittrich S, Pahmeyer C, Schroder P, Teetzen C, Gee H, Daouk G, Pohl M, Kuhn E, Schermer B, Küttner V, Boerries M, Busch H, Schiffer M, Bergmann C, Krüger M, Hildebrandt F, Dengjel J, Benzing T, Huber TB. A Multi-layered Quantitative In Vivo Expression Atlas of the Podocyte Unravels Kidney Disease Candidate Genes. Cell Rep 2019; 23:2495-2508. [PMID: 29791858 PMCID: PMC5986710 DOI: 10.1016/j.celrep.2018.04.059] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/07/2018] [Accepted: 04/15/2018] [Indexed: 12/31/2022] Open
Abstract
Damage to and loss of glomerular podocytes has been identified as the culprit lesion in progressive kidney diseases. Here, we combine mass spectrometry-based proteomics with mRNA sequencing, bioinformatics, and hypothesis-driven studies to provide a comprehensive and quantitative map of mammalian podocytes that identifies unanticipated signaling pathways. Comparison of the in vivo datasets with proteomics data from podocyte cell cultures showed a limited value of available cell culture models. Moreover, in vivo stable isotope labeling by amino acids uncovered surprisingly rapid synthesis of mitochondrial proteins under steady-state conditions that was perturbed under autophagy-deficient, disease-susceptible conditions. Integration of acquired omics dimensions suggested FARP1 as a candidate essential for podocyte function, which could be substantiated by genetic analysis in humans and knockdown experiments in zebrafish. This work exemplifies how the integration of multi-omics datasets can identify a framework of cell-type-specific features relevant for organ health and disease. Deep proteome and transcriptome analyses of native podocytes unravel druggable targets Static and dynamic proteomics uncover features of podocyte identity and proteostasis Candidate genes for nephrotic syndrome were predicted based on multi-omic integration FARP1 is a previously unreported candidate gene for human proteinuric kidney disease
Collapse
Affiliation(s)
- Markus M Rinschen
- Department II of Internal Medicine, University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Systems Biology of Ageing Cologne (Sybacol), University of Cologne, 50931 Cologne, Germany.
| | - Markus Gödel
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; Department of Medicine IV, Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Florian Grahammer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; Department of Medicine IV, Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Stefan Zschiedrich
- Department of Medicine IV, Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Martin Helmstädter
- Department of Medicine IV, Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Oliver Kretz
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; Department of Medicine IV, Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Mostafa Zarei
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, 79104 Freiburg, Germany; Center for Systems Biology (ZBSA), Albert Ludwigs University, 79104 Freiburg, Germany
| | - Daniela A Braun
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sebastian Dittrich
- Department II of Internal Medicine, University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Caroline Pahmeyer
- Department II of Internal Medicine, University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Patricia Schroder
- Department of Medicine/Nephrology, Hannover Medical School, 30625 Hannover, Germany; Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04609, USA
| | - Carolin Teetzen
- Department of Medicine IV, Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - HeonYung Gee
- Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04609, USA; Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Ghaleb Daouk
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Martin Pohl
- Department of General Pediatrics, Adolescent Medicine and Neonatology, Medical Center and Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Elisa Kuhn
- Center for Human Genetics, Bioscientia, 55218 Ingelheim, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine, University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Systems Biology of Ageing Cologne (Sybacol), University of Cologne, 50931 Cologne, Germany
| | - Victoria Küttner
- Department for Neuroanatomy, University of Freiburg, 79104 Freiburg, Germany; Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, 79104 Freiburg, Germany; Department of Dermatology, Medical Center - University of Freiburg, 79106 Freiburg, Germany
| | - Melanie Boerries
- Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert Ludwigs University Freiburg, 79106 Freiburg, Germany; German Cancer Consortium (DKTK), 79106 Freiburg, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hauke Busch
- Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert Ludwigs University Freiburg, 79106 Freiburg, Germany; Lübeck Institute for Experimental Dermatology (LIED), University of Lübeck, 23562 Lübeck, Germany
| | - Mario Schiffer
- Department of Medicine/Nephrology, Hannover Medical School, 30625 Hannover, Germany; Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04609, USA
| | - Carsten Bergmann
- Department of Medicine IV, Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany; Center for Human Genetics, Bioscientia, 55218 Ingelheim, Germany
| | - Marcus Krüger
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Friedhelm Hildebrandt
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joern Dengjel
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, 79104 Freiburg, Germany; Center for Systems Biology (ZBSA), Albert Ludwigs University, 79104 Freiburg, Germany; Department of Dermatology, Medical Center - University of Freiburg, 79106 Freiburg, Germany; Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland; BIOSS Centre for Biological Signaling Studies, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany
| | - Thomas Benzing
- Department II of Internal Medicine, University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Systems Biology of Ageing Cologne (Sybacol), University of Cologne, 50931 Cologne, Germany.
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; Department of Medicine IV, Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany; Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, 79104 Freiburg, Germany; Center for Systems Biology (ZBSA), Albert Ludwigs University, 79104 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
14
|
Yaribeygi H, Atkin SL, Katsiki N, Sahebkar A. Narrative review of the effects of antidiabetic drugs on albuminuria. J Cell Physiol 2018; 234:5786-5797. [PMID: 30367464 DOI: 10.1002/jcp.27503] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is the most prevalent metabolic disorder worldwide. Glycemic control is the main focus of antidiabetic therapy. However, there are data suggesting that some antidiabetic drugs may have intrinsic beneficial renal effects and protect against the development and progression of albuminuria, thus minimizing the risk of diabetic nephropathy. These pharmacological agents can suppress upstream molecular pathways involved in the pathophysiology of diabetes-induced renal dysfunction such as oxidative stress, inflammatory responses, and apoptosis. In this narrative review, the pathophysiology of albuminuria in patients with diabetic nephropathy is discussed. Furthermore, the renoprotective effects of antidiabetic drugs, focusing on albuminuria, are reviewed.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Niki Katsiki
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Chebotareva NV, Bobkova IN, Lysenko LV. The role of podocytes dysfunction in chronic glomerulonephritis progression. TERAPEVT ARKH 2018; 90:92-97. [PMID: 30701911 DOI: 10.26442/terarkh201890692-97] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the review, the mechanisms of podocytes damage underlying the development of proteinuria and progression of glomerulosclerosis in chronic glomerulonephritis are discussed in detail. The results of experimental and clinical studies are presented. Under the different immune and non-immune factors the podocytes form a stereotyped response to damage consisting in the reorganization of the actin cytoskeleton, foot process effacement, the detachment of podocytes from the glomerular basement membrane, and the appearance of specific podocyte proteins and whole cells (podocyturia) in the urine. Massive podocyturia in a limited proliferative capacity of podocytes leads to reduce their total count in the glomerulus (podocytopenia) and the development of glomerulosclerosis. The authors describe the line of markers of the podocyte injury and invasive and non-invasive methods of their assessment. In addition, the relationship of podocyturia level with proteinuria and renal dysfunction are discussed, the prospects of assessment the podocyte proteins in urine for assessing of glomerular damage severity and glomerulosclerosis risk are examined.
Collapse
Affiliation(s)
- N V Chebotareva
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia, Moscow, Russia
| | - I N Bobkova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia, Moscow, Russia
| | - L V Lysenko
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
16
|
Oh HJ, Nam BY, Wu M, Kim S, Park J, Kang S, Park JT, Yoo TH, Kang SW, Han SH. Klotho plays a protective role against glomerular hypertrophy in a cell cycle-dependent manner in diabetic nephropathy. Am J Physiol Renal Physiol 2018; 315:F791-F805. [PMID: 29638159 DOI: 10.1152/ajprenal.00462.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
There are few studies on the effect of klotho on podocytes in diabetic nephropathy. Thus, we tested whether klotho exerts a protective effect against glomerular injury in diabetes. Mouse podocytes were cultured in media containing 5.6 or 30 mM glucose(HG) with or without 200 pM of recombinant klotho (rKL). Additionally, 32 mice were injected intraperitoneally with either diluent( n = 16, C) or with streptozotocin ( n = 16, DM). Control and diabetic mice underwent sham operation and unilateral nephrectomy, respectively. Eight mice from each control and DM group were treated daily with 10 μg·kg-1·day-1 of rKL, using an osmotic minipump. Klotho was expressed in podocytes, and its expression was dependent on peroxisome proliferator-activateed receptor-γ (PPARγ). HG treatment increased the expression of cell cycle-related and apoptotic markers, and these were significantly attenuated by rKL; rKL inhibited the extracellular signal-regulated protein kinase-1/2 and p38 signaling pathways in HG-induced podocyte injury. However, siRNA against klotho gene in HG-treated podocytes failed to aggravate cell cycle arrest and apoptosis. When HG-treated podocytes were incubated in the high-klotho-conditioned medium from tubular epithelial cells, cell injury was significantly attenuated. This effect was not observed when klotho was inhibited by siRNA. In vivo, the expressions of cell cycle-related and apoptotic markers were increased in diabetic mice compared with controls, which were significantly decreased by rKL. Glomerular hypertrophy (GH) and increased profibrotic markers were significantly alleviated after rKL administration. These results showed that klotho was expressed in glomerular podocytes that and its expression was regulated by PPARγ. Additionally, administration of rKL attenuated GH via a cell cycle-dependent mechanism and decreased apoptosis.
Collapse
Affiliation(s)
- Hyung Jung Oh
- Ewha Institute of Convergence Medicine, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| | - Bo Young Nam
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Yonsei University, Seoul, Republic of Korea
| | - Meiyan Wu
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Yonsei University, Seoul, Republic of Korea
| | - Seonghun Kim
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Yonsei University, Seoul, Republic of Korea
| | - Jimin Park
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Yonsei University, Seoul, Republic of Korea
| | - Sukyung Kang
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Yonsei University, Seoul, Republic of Korea
| | - Jung Tak Park
- Divison of Nephrology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.,Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae-Hyun Yoo
- Divison of Nephrology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.,Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Shin-Wook Kang
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS, Yonsei University, Seoul, Republic of Korea.,Divison of Nephrology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.,Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Hyeok Han
- Divison of Nephrology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.,Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
17
|
Srivastava T, Thiagarajan G, Alon US, Sharma R, El-Meanawy A, McCarthy ET, Savin VJ, Sharma M. Role of biomechanical forces in hyperfiltration-mediated glomerular injury in congenital anomalies of the kidney and urinary tract. Nephrol Dial Transplant 2018; 32:759-765. [PMID: 28339567 DOI: 10.1093/ndt/gfw430] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/15/2016] [Indexed: 11/13/2022] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) including solitary kidney constitute the main cause of progressive chronic kidney disease (CKD) in children. Children born with CAKUT develop signs of CKD only during adolescence and do not respond to renin-angiotensin-aldosterone system blockers. Early cellular changes underlying CKD progression to end-stage renal disease by early adulthood are not well understood. The mechanism of maladaptive hyperfiltration that occurs from loss of functional nephrons, including solitary kidney, is not clear. We re-examine the phenomenon of hyperfiltration in the context of biomechanical forces with special reference to glomerular podocytes. Capillary stretch exerts tensile stress on podocytes through the glomerular basement membrane. The flow of ultrafiltrate over the cell surface directly causes fluid flow shear stress (FFSS) on podocytes. FFSS on the podocyte surface increases 1.5- to 2-fold in animal models of solitary kidney and its effect on podocytes is a subject of ongoing research. Podocytes (i) are mechanosensitive to tensile and shear forces, (ii) use prostaglandin E2, angiotensin-II or nitric oxide for mechanoperception and (iii) use specific signaling pathways for mechanotransduction. We discuss (i) the nature of and differences in cellular responses to biomechanical forces, (ii) methods to study biomechanical forces and (iii) effects of biomechanical forces on podocytes and glomeruli. Future studies on FFSS will likely identify novel targets for strategies for early intervention to complement and strengthen the current regimen for treating children with CAKUT.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO, USA.,Renal Research Laboratory, Research and Development, Kansas City VA Medical Center, Kansas City, MO, USA
| | - Ganesh Thiagarajan
- School of Computing and Engineering, University of Missouri at Kansas City, MO, USA
| | - Uri S Alon
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO, USA
| | - Ram Sharma
- Renal Research Laboratory, Research and Development, Kansas City VA Medical Center, Kansas City, MO, USA
| | - Ashraf El-Meanawy
- Division of Nephrology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ellen T McCarthy
- Kidney Institute, Kansas University Medical Center, Kansas City, KS, USA
| | - Virginia J Savin
- Renal Research Laboratory, Research and Development, Kansas City VA Medical Center, Kansas City, MO, USA
| | - Mukut Sharma
- Renal Research Laboratory, Research and Development, Kansas City VA Medical Center, Kansas City, MO, USA
| |
Collapse
|
18
|
Rajasekeran H, Reich HN, Hladunewich MA, Cattran D, Lovshin JA, Lytvyn Y, Bjornstad P, Lai V, Tse J, Cham L, Majumder S, Bowskill BB, Kabir MG, Advani SL, Gibson IW, Sood MM, Advani A, Cherney DZI. Dapagliflozin in focal segmental glomerulosclerosis: a combined human-rodent pilot study. Am J Physiol Renal Physiol 2017; 314:F412-F422. [PMID: 29141939 DOI: 10.1152/ajprenal.00445.2017] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is an important cause of nondiabetic chronic kidney disease (CKD). Sodium-glucose cotransporter 2 inhibition (SGLT2i) therapy attenuates the progression of diabetic nephropathy, but it remains unclear whether SGLT2i provides renoprotection in nondiabetic CKD such as FSGS. The primary aim of this pilot study was to determine the effect of 8 wk of dapagliflozin on glomerular filtration rate (GFR) in humans and in experimental FSGS. Secondary end points were related to changes in renal hemodynamic function, proteinuria, and blood pressure (BP). GFR (inulin) and renal plasma flow (para-aminohippurate), proteinuria, and BP were measured in patients with FSGS ( n = 10), and similar parameters were measured in subtotally nephrectomized (SNx) rats. In response to dapagliflozin, changes in GFR, renal plasma flow, and 24-h urine protein excretion were not statistically significant in humans or rats. Systolic BP (SBP) decreased in SNx rats (196 ± 26 vs. 165 ± 33 mmHg; P < 0.001), whereas changes were not statistically significant in humans (SBP 112.7 ± 8.5 to 112.8 ± 11.2 mmHg, diastolic BP 71.8 ± 6.5 to 69.6 ± 8.4 mmHg; P = not significant), although hematocrit increased (0.40 ± 0.05 to 0.42 ± 0.05%; P = 0.03). In archival kidney tissue from a separate patient cohort, renal parenchymal SGLT2 mRNA expression was decreased in individuals with FSGS compared with controls. Short-term treatment with the SGLT2i dapagliflozin did not modify renal hemodynamic function or attenuate proteinuria in humans or in experimental FSGS. This may be related to downregulation of renal SGLT2 expression. Studies examining the impact of SGLT2i on markers of kidney disease in patients with other causes of nondiabetic CKD are needed.
Collapse
Affiliation(s)
- Harindra Rajasekeran
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto , Toronto, Ontario , Canada.,Department of Physiology, University of Toronto , Toronto, Ontario , Canada
| | - Heather N Reich
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto , Toronto, Ontario , Canada
| | - Michelle A Hladunewich
- Department of Medicine, Division of Nephrology, Sunnybrook Health Sciences Centre, University of Toronto , Toronto, Ontario , Canada
| | - Daniel Cattran
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto , Toronto, Ontario , Canada
| | - Julie A Lovshin
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto , Toronto, Ontario , Canada
| | - Yuliya Lytvyn
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto , Toronto, Ontario , Canada
| | - Petter Bjornstad
- Department of Pediatric Endocrinology, University of Colorado School of Medicine , Aurora, Colorado.,Barbara Davis Center for Diabetes, University of Colorado Denver , Aurora, Colorado
| | - Vesta Lai
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto , Toronto, Ontario , Canada
| | - Josephine Tse
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto , Toronto, Ontario , Canada
| | - Leslie Cham
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto , Toronto, Ontario , Canada
| | - Syamantak Majumder
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital , Toronto, Ontario , Canada
| | - Bridgit B Bowskill
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital , Toronto, Ontario , Canada
| | - M Golam Kabir
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital , Toronto, Ontario , Canada
| | - Suzanne L Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital , Toronto, Ontario , Canada
| | - Ian W Gibson
- Department of Pathology, University of Manitoba , Winnipeg, Manitoba , Canada
| | - Manish M Sood
- Ottawa Hospital Research Institute, University of Ottawa , Ottawa, Ontario , Canada
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael's Hospital , Toronto, Ontario , Canada
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto , Toronto, Ontario , Canada.,Department of Physiology, University of Toronto , Toronto, Ontario , Canada
| |
Collapse
|
19
|
Bose M, Almas S, Prabhakar S. Wnt signaling and podocyte dysfunction in diabetic nephropathy. J Investig Med 2017; 65:1093-1101. [DOI: 10.1136/jim-2017-000456] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2017] [Indexed: 12/21/2022]
Abstract
Nephropathy is a major microvascular complication of diabetes mellitus and often leads to terminal renal failure in addition to contributing significantly to cardiovascular morbidity and mortality. Despites continuous advances, the pathogenesis of diabetic nephropathy remains poorly understood. Recent studies have underscored the significance of structural and functional changes in podocytes in the development and progression of diabetic nephropathy. The role of podocytes in health and diabetic nephropathy and abnormalities including podocyte hypertrophy, effacement, and apoptosis, and a detailed discussion on the role played by the Wnt-β-catenin signaling pathway in podocyte injury and dysfunction are the focus of this review. In addition, the role played by Wnt signaling in mediating the effects of known therapeutic strategies for diabetic nephropathy is also discussed.
Collapse
|
20
|
Affiliation(s)
- Dae Ryong Cha
- Division of Nephrology, Department of Internal Medicine, Korea University Ansan Hospital, 516 Kojan-dong, Ansan City, Gyeonggi-do, 425-020, Korea
| |
Collapse
|
21
|
Secondary Focal Segmental Glomerulosclerosis: From Podocyte Injury to Glomerulosclerosis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1630365. [PMID: 27088082 PMCID: PMC4819087 DOI: 10.1155/2016/1630365] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 03/10/2016] [Indexed: 11/18/2022]
Abstract
Focal segmental glomerulosclerosis (FSGS) is a common cause of proteinuria and nephrotic syndrome leading to end stage renal disease (ESRD). There are two types of FSGS, primary (idiopathic) and secondary forms. Secondary FSGS shows less severe clinical features compared to those of the primary one. However, secondary FSGS has an important clinical significance because a variety of renal diseases progress to ESRD thorough the form of secondary FSGS. The defining feature of FSGS is proteinuria. The key event of FSGS is podocyte injury which is caused by multiple factors. Unanswered questions about how these factors act on podocytes to cause secondary FSGS are various and ill-defined. In this review, we provide brief overview and new insights into FSGS, podocyte injury, and their potential linkage suggesting clues to answer for treatment of the disease.
Collapse
|
22
|
Drummond BE, Wingert RA. Insights into kidney stem cell development and regeneration using zebrafish. World J Stem Cells 2016; 8:22-31. [PMID: 26981168 PMCID: PMC4766248 DOI: 10.4252/wjsc.v8.i2.22] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 11/28/2015] [Accepted: 01/11/2016] [Indexed: 02/06/2023] Open
Abstract
Kidney disease is an escalating global health problem, for which the formulation of therapeutic approaches using stem cells has received increasing research attention. The complexity of kidney anatomy and function, which includes the diversity of renal cell types, poses formidable challenges in the identification of methods to generate replacement structures. Recent work using the zebrafish has revealed their high capacity to regenerate the integral working units of the kidney, known as nephrons, following acute injury. Here, we discuss these findings and explore the ways that zebrafish can be further utilized to gain a deeper molecular appreciation of renal stem cell biology, which may uncover important clues for regenerative medicine.
Collapse
|
23
|
Abstract
Podocyte hypertrophy and apoptosis are two hallmarks of diabetic glomeruli, but the sequence in which these processes occur remains a matter of debate. Here we investigated the effects of inhibiting hypertrophy on apoptosis, and vice versa, in both podocytes and glomeruli, under diabetic conditions. Hypertrophy and apoptosis were inhibited using an epidermal growth factor receptor inhibitor (PKI 166) and a pan-caspase inhibitor (zAsp-DCB), respectively. We observed significant increases in the protein expression of p27, p21, phospho-eukaryotic elongation factor 4E-binding protein 1, and phospho-p70 S6 ribosomal protein kinase, in both cultured podocytes exposed to high-glucose (HG) medium, and streptozotocin-induced diabetes mellitus (DM) rat glomeruli. These increases were significantly inhibited by PKI 166, but not by zAsp-DCB. In addition, the amount of protein per cell, the relative cell size, and the glomerular volume were all significantly increased under diabetic conditions, and these changes were also blocked by treatment with PKI 166, but not zAsp-DCB. Increased protein expression of cleaved caspase-3 and cleaved poly (ADP-ribose) polymerase, together with increased Bax/Bcl-2 ratios, were also observed in HG-stimulated podocytes and DM glomeruli. Treatment with either zAsp-DCB or PKI 166 resulted in a significant attenuation of these effects. Both PKI 166 and zAsp-DCB also inhibited the increase in number of apoptotic cells, as assessed by Hoechst 33342 staining and TUNEL assay. Under diabetic conditions, inhibition of podocyte hypertrophy results in attenuated apoptosis, whereas blocking apoptosis has no effect on podocyte hypertrophy, suggesting that podocyte hypertrophy precedes apoptosis.
Collapse
|
24
|
Redox Signaling in Diabetic Nephropathy: Hypertrophy versus Death Choices in Mesangial Cells and Podocytes. Mediators Inflamm 2015; 2015:604208. [PMID: 26491232 PMCID: PMC4600552 DOI: 10.1155/2015/604208] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 01/18/2015] [Indexed: 02/06/2023] Open
Abstract
This review emphasizes the role of oxidative stress in diabetic nephropathy, acting as trigger, modulator, and linker within the complex network of pathologic events. It highlights key molecular pathways and new hypothesis in diabetic nephropathy, related to the interferences of metabolic, oxidative, and inflammatory stresses. Main topics this review is addressing are biomarkers of oxidative stress in diabetic nephropathy, the sources of reactive oxygen species (mitochondria, NADPH-oxidases, hyperglycemia, and inflammation), and the redox-sensitive signaling networks (protein kinases, transcription factors, and epigenetic regulators). Molecular switches deciding on the renal cells fate in diabetic nephropathy are presented, such as hypertrophy versus death choices in mesangial cells and podocytes. Finally, the antioxidant response of renal cells in diabetic nephropathy is tackled, with emphasis on targeted therapy. An integrative approach is needed for identifying key molecular networks which control cellular responses triggered by the array of stressors in diabetic nephropathy. This will foster the discovery of reliable biomarkers for early diagnosis and prognosis, and will guide the discovery of new therapeutic approaches for personalized medicine in diabetic nephropathy.
Collapse
|
25
|
Subathra M, Korrapati M, Howell LA, Arthur JM, Shayman JA, Schnellmann RG, Siskind LJ. Kidney glycosphingolipids are elevated early in diabetic nephropathy and mediate hypertrophy of mesangial cells. Am J Physiol Renal Physiol 2015; 309:F204-15. [PMID: 26041445 DOI: 10.1152/ajprenal.00150.2015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/27/2015] [Indexed: 12/11/2022] Open
Abstract
Glycosphingolipids (GSLs) play a role in insulin resistance and diabetes, but their role in diabetic nephropathy (DN) has received limited attention. We used 9- and 17-wk-old nondiabetic db/m and diabetic db/db mice to examine the role of GSLs in DN. Cerebrosides or monoglycosylated GSLs [hexosylceramides (HexCers); glucosyl- and galactosylceramides] and lactosylceramide (LacCers) were elevated in db/db mouse kidney cortices, specifically in glomeruli, and also in urine. In our recent paper (25), we observed that the kidneys exhibited glomerular hypertrophy and proximal tubular vacuolization and increased fibrosis markers at these time points. Mesangial cells contribute to hyperglycemia-induced glomerular hypertrophy in DN. Hyperglycemic culture conditions, similar to that present in diabetes, were sufficient to elevate mesangial cell HexCers and increase markers of fibrosis, extracellular matrix proteins, and cellular hypertrophy. Inhibition of glucosylceramide synthase or lowering glucose levels decreased markers of fibrosis and extracellular matrix proteins and reversed mesangial cell hypertrophy. Hyperglycemia increased phosphorylated (p)SMAD3 and pAkt levels and reduced phosphatase and tensin homolog levels, which were reversed with glucosylceramide synthase inhibition. These data suggest that inhibition of glucosylceramide synthase reversed mesangial cell hypertrophy through decreased pAkt and pSmad3 and increased pathways responsible for protein degradation. Importantly, urinary GSL levels were higher in patients with DN compared with healthy control subjects, implicating a role for these lipids in human DN. Thus, hyperglycemia in type II diabetes leads to renal dysfunction at least in part by inducing accumulation of HexCers and LacCers in mesangial cells, resulting in fibrosis, extracellular matrix production, and hypertrophy.
Collapse
Affiliation(s)
- Marimuthu Subathra
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Midhun Korrapati
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Lauren A Howell
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida
| | - John M Arthur
- University of Arkansas for Medical Sciences, Little Rock, Arkansas; Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| | - James A Shayman
- Nephrology Division, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan; and
| | - Rick G Schnellmann
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina; Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky;
| |
Collapse
|
26
|
Neal CR. Podocytes … What's Under Yours? (Podocytes and Foot Processes and How They Change in Nephropathy). Front Endocrinol (Lausanne) 2015; 6:9. [PMID: 25755650 PMCID: PMC4337384 DOI: 10.3389/fendo.2015.00009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 01/15/2015] [Indexed: 12/25/2022] Open
Abstract
Most of the described structures of podocytes in health and disease have been inferred from light and electron microscopic studies of rodent models. The variation in filtration barrier features is measured on micrographs, the aim being statistical significance. This is the technical campaign waged against kidney disease but this approach can be misleading. The signaling cascades and connectivity of the podocyte and foot processes (FPs) are inferred from in vitro studies that at best blurr the reality of the in vivo state. This review will outline actin signaling connectivity and the key differences in the structural and functional domains squeezed into the FPs and the relationship of these domains to other parts of the podocyte. It covers the changes in podocytes during nephropathy concentrating on FP and finally proposes an alternative interpretation of FP ultrastructure derived from articles published over the last 60 years.
Collapse
Affiliation(s)
- Chris R. Neal
- Bristol Renal, University of Bristol, Bristol, UK
- *Correspondence: Chris R. Neal, Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK e-mail:
| |
Collapse
|
27
|
|
28
|
BARGMAN JOANNEM, AVILA-CASADO CARMEN. Resolution of Proteinuria in Lupus Nephritis: Hurry Up and Wait. J Rheumatol 2014; 41:622-5. [DOI: 10.3899/jrheum.140157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
29
|
Lohmann F, Sachs M, Meyer TN, Sievert H, Lindenmeyer MT, Wiech T, Cohen CD, Balabanov S, Stahl RAK, Meyer-Schwesinger C. UCH-L1 induces podocyte hypertrophy in membranous nephropathy by protein accumulation. Biochim Biophys Acta Mol Basis Dis 2014; 1842:945-58. [PMID: 24583340 DOI: 10.1016/j.bbadis.2014.02.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 02/18/2014] [Accepted: 02/21/2014] [Indexed: 11/18/2022]
Abstract
Podocytes are terminally differentiated cells of the glomerular filtration barrier that react with hypertrophy in the course of injury such as in membranous nephropathy (MGN). The neuronal deubiquitinase ubiquitin C-terminal hydrolase L1 (UCH-L1) is expressed and activated in podocytes of human and rodent MGN. UCH-L1 regulates the mono-ubiquitin pool and induces accumulation of poly-ubiquitinated proteins in affected podocytes. Here, we investigated the role of UCH-L1 in podocyte hypertrophy and in the homeostasis of the hypertrophy associated "model protein" p27(Kip1). A better understanding of the basic mechanisms leading to podocyte hypertrophy is crucial for the development of specific therapies in MGN. In human and rat MGN, hypertrophic podocytes exhibited a simultaneous up-regulation of UCH-L1 and of cytoplasmic p27(Kip1) content. Functionally, inhibition of UCH-L1 activity and knockdown or inhibition of UCH-L1 attenuated podocyte hypertrophy by decreasing the total protein content in isolated glomeruli and in cultured podocytes. In contrast, UCH-L1 levels and activity increased podocyte hypertrophy and total protein content in culture, specifically of cytoplasmic p27(Kip1). UCH-L1 enhanced cytoplasmic p27(Kip1) levels by nuclear export and decreased poly-ubiquitination and proteasomal degradation of p27(Kip1). In parallel, UCH-L1 increased podocyte turnover, migration and cytoskeletal rearrangement, which are associated with known oncogenic functions of cytoplasmic p27(Kip1) in cancer. We propose that UCH-L1 induces podocyte hypertrophy in MGN by increasing the total protein content through altered degradation and accumulation of proteins such as p27(Kip1) in the cytoplasm of podocytes. Modification of both UCH-L1 activity and levels could be a new therapeutic avenue to podocyte hypertrophy in MGN.
Collapse
Affiliation(s)
- Frithjof Lohmann
- Department of Internal Medicine, Nephrology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marlies Sachs
- Department of Internal Medicine, Nephrology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias N Meyer
- Department of Internal Medicine, Nephrology, Asklepios Klinikum Barmbek, Hamburg, Germany
| | - Henning Sievert
- Department of Oncology, Haematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maja T Lindenmeyer
- Institute of Physiology and Division of Nephrology, University of Zurich, Switzerland
| | - Thorsten Wiech
- Department of Pathology, Division of Renal Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clemens D Cohen
- Institute of Physiology and Division of Nephrology, University of Zurich, Switzerland
| | | | - R A K Stahl
- Department of Internal Medicine, Nephrology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|
30
|
Abstract
Diabetes mellitus contributes greatly to morbidity, mortality, and overall health care costs. In major part, these outcomes derive from the high incidence of progressive kidney dysfunction in patients with diabetes making diabetic nephropathy a leading cause of end-stage renal disease. A better understanding of the molecular mechanism involved and of the early dysfunctions observed in the diabetic kidney may permit the development of new strategies to prevent diabetic nephropathy. Here we review the pathophysiological changes that occur in the kidney in response to hyperglycemia, including the cellular responses to high glucose and the responses in vascular, glomerular, podocyte, and tubular function. The molecular basis, characteristics, and consequences of the unique growth phenotypes observed in the diabetic kidney, including glomerular structures and tubular segments, are outlined. We delineate mechanisms of early diabetic glomerular hyperfiltration including primary vascular events as well as the primary role of tubular growth, hyperreabsorption, and tubuloglomerular communication as part of a "tubulocentric" concept of early diabetic kidney function. The latter also explains the "salt paradox" of the early diabetic kidney, that is, a unique and inverse relationship between glomerular filtration rate and dietary salt intake. The mechanisms and consequences of the intrarenal activation of the renin-angiotensin system and of diabetes-induced tubular glycogen accumulation are discussed. Moreover, we aim to link the changes that occur early in the diabetic kidney including the growth phenotype, oxidative stress, hypoxia, and formation of advanced glycation end products to mechanisms involved in progressive kidney disease.
Collapse
Affiliation(s)
- Volker Vallon
- Department of Medicine, University of California San Diego & VA San Diego Healthcare System, San Diego, California, USA.
| | | |
Collapse
|
31
|
Babelova A, Jansen F, Sander K, Löhn M, Schäfer L, Fork C, Ruetten H, Plettenburg O, Stark H, Daniel C, Amann K, Pavenstädt H, Jung O, Brandes RP. Activation of Rac-1 and RhoA contributes to podocyte injury in chronic kidney disease. PLoS One 2013; 8:e80328. [PMID: 24244677 PMCID: PMC3820652 DOI: 10.1371/journal.pone.0080328] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 10/02/2013] [Indexed: 12/12/2022] Open
Abstract
Rho-family GTPases like RhoA and Rac-1 are potent regulators of cellular signaling that control gene expression, migration and inflammation. Activation of Rho-GTPases has been linked to podocyte dysfunction, a feature of chronic kidney diseases (CKD). We investigated the effect of Rac-1 and Rho kinase (ROCK) inhibition on progressive renal failure in mice and studied the underlying mechanisms in podocytes. SV129 mice were subjected to 5/6-nephrectomy which resulted in arterial hypertension and albuminuria. Subgroups of animals were treated with the Rac-1 inhibitor EHT1846, the ROCK inhibitor SAR407899 and the ACE inhibitor Ramipril. Only Ramipril reduced hypertension. In contrast, all inhibitors markedly attenuated albumin excretion as well as glomerular and tubulo-interstitial damage. The combination of SAR407899 and Ramipril was more effective in preventing albuminuria than Ramipril alone. To study the involved mechanisms, podocytes were cultured from SV129 mice and exposed to static stretch in the Flexcell device. This activated RhoA and Rac-1 and led via TGFβ to apoptosis and a switch of the cells into a more mesenchymal phenotype, as evident from loss of WT-1 and nephrin and induction of α-SMA and fibronectin expression. Rac-1 and ROCK inhibition as well as blockade of TGFβ dramatically attenuated all these responses. This suggests that Rac-1 and RhoA are mediators of podocyte dysfunction in CKD. Inhibition of Rho-GTPases may be a novel approach for the treatment of CKD.
Collapse
Affiliation(s)
| | - Felix Jansen
- Physiology I, Goethe-University, Frankfurt am Main, Germany
| | - Kerstin Sander
- Institute for Pharmaceutical Chemistry, Goethe-University, Frankfurt am Main, Germany
| | | | - Liliana Schäfer
- General Pharmacology and Toxicology, Goethe-University, Frankfurt am Main, Germany
| | - Christian Fork
- Physiology I, Goethe-University, Frankfurt am Main, Germany
| | | | | | - Holger Stark
- Institute for Pharmaceutical Chemistry, Goethe-University, Frankfurt am Main, Germany
| | - Christoph Daniel
- Department of Pathology, Nephropathology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Kerstin Amann
- Department of Pathology, Nephropathology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Hermann Pavenstädt
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Oliver Jung
- Physiology I, Goethe-University, Frankfurt am Main, Germany
- Internal Medicine/Nephrology, Goethe-University, Frankfurt am Main, Germany
| | - Ralf P. Brandes
- Physiology I, Goethe-University, Frankfurt am Main, Germany
- * E-mail:
| |
Collapse
|
32
|
Srivastava T, Celsi GE, Sharma M, Dai H, McCarthy ET, Ruiz M, Cudmore PA, Alon US, Sharma R, Savin VA. Fluid flow shear stress over podocytes is increased in the solitary kidney. Nephrol Dial Transplant 2013; 29:65-72. [PMID: 24166460 DOI: 10.1093/ndt/gft387] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Glomerular hyperfiltration is emerging as the key risk factor for progression of chronic kidney disease (CKD). Podocytes are exposed to fluid flow shear stress (FFSS) caused by the flow of ultrafiltrate within Bowman's space. The mechanism of hyperfiltration-induced podocyte injury is not clear. We postulated that glomerular hyperfiltration in solitary kidney increases FFSS over podocytes. METHODS Infant Sprague-Dawley rats at 5 days of age and C57BL/6J 14-week-old adult mice underwent unilateral nephrectomy. Micropuncture and morphological studies were then performed on 20- and 60-day-old rats. FFSS over podocytes in uninephrectomized rats and mice was calculated using the recently published equation by Friedrich et al. which includes the variables-single nephron glomerular filtration rate (SNGFR), filtration fraction (f), glomerular tuft diameter (2RT) and width of Bowman's space (s). RESULTS Glomerular hypertrophy was observed in uninephrectomized rats and mice. Uninephrectomized rats on Day 20 showed a 2.0-fold increase in SNGFR, 1.0-fold increase in 2RT and 2.1-fold increase in FFSS, and on Day 60 showed a 1.9-fold increase in SNGFR, 1.3-fold increase in 2RT and 1.5-fold increase in FFSS, at all values of modeled 's'. Similarly, uninephrectomized mice showed a 2- to 3-fold increase in FFSS at all values of modeled SNGFR. CONCLUSIONS FFSS over podocytes is increased in solitary kidneys in both infant rats and adult mice. This increase is a consequence of increased SNGFR. We speculate that increased FFSS caused by reduced nephron number contributes to podocyte injury and promotes the progression of CKD.
Collapse
Affiliation(s)
- Tarak Srivastava
- Section of Nephrology, Children's Mercy Hospital and University of Missouri at Kansas City, Kansas City, MO, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
The Expression of Intermediate Filament Protein Nestin and Its Association With Cyclin-dependent Kinase 5 in the Glomeruli of Rats With Diabetic Nephropathy. Am J Med Sci 2013; 345:470-7. [DOI: 10.1097/maj.0b013e3182648459] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
34
|
Li Y, Wingert RA. Regenerative medicine for the kidney: stem cell prospects & challenges. Clin Transl Med 2013; 2:11. [PMID: 23688352 PMCID: PMC3665577 DOI: 10.1186/2001-1326-2-11] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/14/2013] [Indexed: 12/22/2022] Open
Abstract
The kidney has key roles in maintaining human health. There is an escalating medical crisis in nephrology as growing numbers of patients suffer from kidney diseases that culminate in organ failure. While dialysis and transplantation provide life-saving treatments, these therapies are rife with limitations and place significant burdens on patients and healthcare systems. It has become imperative to find alternative ways to treat existing kidney conditions and preemptive means to stave off renal dysfunction. The creation of innovative medical approaches that utilize stem cells has received growing research attention. In this review, we discuss the regenerative and maladaptive cellular responses that occur during acute and chronic kidney disease, the emerging evidence about renal stem cells, and some of the issues that lie ahead in bridging the gap between basic stem cell biology and regenerative medicine for the kidney.
Collapse
Affiliation(s)
- Yue Li
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rebecca A Wingert
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
35
|
Barocas VH, Dorfman KD, Segal Y. A model of strain-dependent glomerular basement membrane maintenance and its potential ramifications in health and disease. J Biomech Eng 2013; 134:081006. [PMID: 22938359 DOI: 10.1115/1.4007098] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A model is developed and analyzed for type IV collagen turnover in the kidney glomerular basement membrane (GBM), which is the primary structural element in the glomerular capillary wall. The model incorporates strain dependence in both deposition and removal of the GBM, leading to an equilibrium tissue strain at which deposition and removal are balanced. The GBM thickening decreases tissue strain per unit of transcapillary pressure drop according to the law of Laplace, but increases the transcapillary pressure drop required to maintain glomerular filtration. The model results are in agreement with the observed GBM alterations in Alport syndrome and thin basement membrane disease, and the model-predicted linear relation between the inverse capillary radius and inverse capillary thickness at equilibrium is consistent with published data on different mammals. In addition, the model predicts a minimum achievable strain in the GBM based on the geometry, properties, and mechanical environment; that is, an infinitely thick GBM would still experience a finite strain. Although the model assumptions would be invalid for an extremely thick GBM, the minimum achievable strain could be significant in diseases, such as Alport syndrome, characterized by focal GBM thickening. Finally, an examination of reasonable values for the model parameters suggests that the oncotic pressure drop-the osmotic pressure difference between the plasma and the filtrate due to large molecules-plays an important role in setting the GBM strain and, thus, leakage of protein into the urine may be protective against some GBM damage.
Collapse
Affiliation(s)
- Victor H Barocas
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
36
|
Abstract
Glomerular hypertension (ie, increased glomerular capillary pressure), has been shown to cause podocyte damage progressing to glomerulosclerosis in animal models. Increased glomerular capillary pressure results in an increase in wall tension that acts primarily as circumferential tensile stress on the capillary wall. The elastic properties of the glomerular basement membrane (GBM) and the elastic as well as contractile properties of the cytoskeleton of the endothelium and of podocyte foot processes resist circumferential tensile stress. Whether the contractile forces generated by podocytes are able to equal circumferential tensile stress to effectively counteract wall tension is an open question. Mechanical stress is transmitted from the GBM to the actin cytoskeleton of podocyte foot processes via cell-matrix contacts that contain mainly integrin α3β1 and a variety of linker, scaffolding, and signaling proteins, which are not well characterized in podocytes. We know from in vitro studies that podocytes are sensitive to stretch, however, the crucial mechanosensor in podocytes remains unclear. On the other hand, in vitro studies have shown that in stretched podocytes specific signaling cascades are activated, the synthesis and secretion of various hormones and their receptors are increased, cell-cycle arrest is reinforced, cell adhesion is altered through secretion of matricellular proteins and changes in integrin expression, and the actin cytoskeleton is reorganized in a way that stress fibers are lost. In summary, current evidence suggests that in glomerular hypertension podocytes primarily aim to maintain the delicate architecture of interdigitating foot processes in the face of an expanding GBM area.
Collapse
Affiliation(s)
- Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | | |
Collapse
|
37
|
Abstract
The kidney is widely regarded as an organ without regenerative abilities. However, in recent years this dogma has been challenged on the basis of observations of kidney recovery following acute injury, and the identification of renal populations that demonstrate stem cell characteristics in various species. It is currently speculated that the human kidney can regenerate in some contexts, but the mechanisms of renal regeneration remain poorly understood. Numerous controversies surround the potency, behaviour and origins of the cell types that are proposed to perform kidney regeneration. The present review explores the current understanding of renal stem cells and kidney regeneration events, and examines the future challenges in using these insights to create new clinical treatments for kidney disease.
Collapse
|
38
|
Kim DK, Nam BY, Li JJ, Park JT, Lee SH, Kim DH, Kim JY, Kang HY, Han SH, Yoo TH, Han DS, Kang SW. Translationally controlled tumour protein is associated with podocyte hypertrophy in a mouse model of type 1 diabetes. Diabetologia 2012; 55:1205-17. [PMID: 22311416 DOI: 10.1007/s00125-012-2467-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 12/31/2011] [Indexed: 02/06/2023]
Abstract
AIMS/HYPOTHESIS Translationally controlled tumour protein (TCTP) is thought to be involved in cell growth by regulating mTOR complex 1 (mTORC1) signalling. As diabetes characteristically induces podocyte hypertrophy and mTORC1 has been implicated in this process, TCTP may have a role in the pathogenesis of diabetes-induced podocyte hypertrophy. METHODS We investigated the effects and molecular mechanisms of TCTP in diabetic mice and in high glucose-stimulated cultured podocytes. To characterise the role of TCTP, we conducted lentivirus-mediated gene silencing of TCTP both in vivo and in vitro. RESULTS Glomerular production of TCTP was significantly higher in streptozotocin induced-diabetic DBA/2J mice than in control animals. Double-immunofluorescence staining for TCTP and synaptopodin revealed that podocyte was the principal cell responsible for this increase. TCTP knockdown attenuated the activation of mTORC1 downstream effectors and the overproduction of cyclin-dependent kinase inhibitors (CKIs) in diabetic glomeruli, along with a reduction in proteinuria and a decrease in the sizes of podocytes as well as glomeruli. In addition, knockdown of TCTP in db/db mice prevented the development of diabetic nephropathy, as indicated by the amelioration of proteinuria, mesangial expansion, podocytopenia and glomerulosclerosis. In accordance with the in vivo data, TCTP inhibition abrogated high glucose-induced hypertrophy in cultured podocytes, which was accompanied by the downregulation of mTORC1 effectors and CKIs. CONCLUSIONS/INTERPRETATION These findings suggest that TCTP might play an important role in the process of podocyte hypertrophy under diabetic conditions via the regulation of mTORC1 activity and the induction of cell-cycle arrest.
Collapse
Affiliation(s)
- D K Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Huang C, Bruggeman LA, Hydo LM, Miller RT. Shear stress induces cell apoptosis via a c-Src-phospholipase D-mTOR signaling pathway in cultured podocytes. Exp Cell Res 2012; 318:1075-85. [PMID: 22472346 DOI: 10.1016/j.yexcr.2012.03.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 03/15/2012] [Accepted: 03/18/2012] [Indexed: 02/06/2023]
Abstract
The glomerular capillary wall, composed of endothelial cells, the glomerular basement membrane and the podocytes, is continually subjected to hemodynamic force arising from tractional stress due to blood pressure and shear stress due to blood flow. Exposure of glomeruli to abnormal hemodynamic force such as hyperfiltration is associated with glomerular injury and progressive renal disease, and the conversion of mechanical stimuli to chemical signals in the regulation of the process is poorly understood in podocytes. By examining DNA fragmentation, apoptotic nuclear changes and cytochrome c release, we found that shear stress induced cell apoptosis in cultured podocytes. Meanwhile, podocytes exposed to shear stress also stimulated c-Src phosphorylation, phospholipase D (PLD) activation and mammalian target of rapamycin (mTOR) signaling. Using the antibodies against c-Src, PLD(1), and PLD(2) to perform reciprocal co-immunoprecipitations and in vitro PLD activity assay, our data indicated that c-Src interacted with and activated PLD(1) but not PLD(2). The inhibition of shear stress-induced c-Src phosphorylation by PP(2) (a specific inhibitor of c-Src kinase) resulted in reduced PLD activity. Phosphatidic acid, produced by shear stress-induced PLD activation, stimulated mTOR signaling, and caused podocyte hypertrophy and apoptosis.
Collapse
Affiliation(s)
- Chunfa Huang
- Louis Stokes Cleveland Veteran Affairs Medical Center, Case Western Reserve University, USA.
| | | | | | | |
Collapse
|
40
|
Darouich S, Goucha R, Jaafoura MH, Zekri S, Ben Maiz H, Kheder A. Clinicopathological characteristics of obesity-associated focal segmental glomerulosclerosis. Ultrastruct Pathol 2011; 35:176-82. [PMID: 21657818 DOI: 10.3109/01913123.2011.584657] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Obesity-related glomerulopathy (ORG) is a secondary form of focal segmental glomerulosclerosis (FSGS) occurring in obese patients with a body-mass index higher than 30 kg/m(2). It is typically manifested by nephrotic-range proteinuria without full nephrotic syndrome, and progressive renal insufficiency. Characteristic morphologic features include the consistent presence of glomerulomegaly, predominance of perihilar variant of FSGS, and the relatively mild fusion of visceral epithelial cell foot processes. The concept of podocyte depletion as a driver of the glomerular scarring in obesity-associated FSGS is well documented. The underlying mechanisms are likely to be related in part to the oxidative stress and the impairment of the integrity of the slit diaphragm and cell adhesion resulting mainly from angiotensin II and transforming growth factor-β. These proapoptotic cytokines are upregulated in obesity in response to insulin resistance, compensatory hyperinsulinemia and glomerular hyperfiltration-hypertension mediated mechanical stress. This review is designed to discuss the clinicopathologic features of obesity-associated FSGS, with a focus on the podocyte injury, which is involved in the onset and progression of the glomerulosclerotic process. Ultrastructural glomerular lesions are documented.
Collapse
Affiliation(s)
- Sihem Darouich
- Electron Microscopy Laboratory, Faculty of Medicine of Tunis, Tunis, Tunisia
| | | | | | | | | | | |
Collapse
|
41
|
Klein J, Gonzalez J, Miravete M, Caubet C, Chaaya R, Decramer S, Bandin F, Bascands JL, Buffin-Meyer B, Schanstra JP. Congenital ureteropelvic junction obstruction: human disease and animal models. Int J Exp Pathol 2011; 92:168-92. [PMID: 20681980 PMCID: PMC3101490 DOI: 10.1111/j.1365-2613.2010.00727.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 06/03/2010] [Indexed: 02/06/2023] Open
Abstract
Ureteropelvic junction (UPJ) obstruction is the most frequently observed cause of obstructive nephropathy in children. Neonatal and foetal animal models have been developed that mimic closely what is observed in human disease. The purpose of this review is to discuss how obstructive nephropathy alters kidney histology and function and describe the molecular mechanisms involved in the progression of the lesions, including inflammation, proliferation/apoptosis, renin-angiotensin system activation and fibrosis, based on both human and animal data. Also we propose that during obstructive nephropathy, hydrodynamic modifications are early inducers of the tubular lesions, which are potentially at the origin of the pathology. Finally, an important observation in animal models is that relief of obstruction during kidney development has important effects on renal function later in adult life. A major short-coming is the absence of data on the impact of UPJ obstruction on long-term adult renal function to elucidate whether these animal data are also valid in humans.
Collapse
Affiliation(s)
- Julie Klein
- Institut National de la Santé et de la Recherche Médicale (INSERM)Toulouse, France
- Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de RangueilToulouse, France
| | - Julien Gonzalez
- Institut National de la Santé et de la Recherche Médicale (INSERM)Toulouse, France
- Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de RangueilToulouse, France
| | - Mathieu Miravete
- Institut National de la Santé et de la Recherche Médicale (INSERM)Toulouse, France
- Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de RangueilToulouse, France
| | - Cécile Caubet
- Institut National de la Santé et de la Recherche Médicale (INSERM)Toulouse, France
- Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de RangueilToulouse, France
| | - Rana Chaaya
- Institut National de la Santé et de la Recherche Médicale (INSERM)Toulouse, France
- Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de RangueilToulouse, France
| | - Stéphane Decramer
- Institut National de la Santé et de la Recherche Médicale (INSERM)Toulouse, France
- Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de RangueilToulouse, France
- Department of Pediatric Nephrology, Hôpital des Enfants, Centre de Référence du Sud Ouest des Maladies Rénales RaresToulouse, France
| | - Flavio Bandin
- Department of Pediatric Nephrology, Hôpital des Enfants, Centre de Référence du Sud Ouest des Maladies Rénales RaresToulouse, France
| | - Jean-Loup Bascands
- Institut National de la Santé et de la Recherche Médicale (INSERM)Toulouse, France
- Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de RangueilToulouse, France
| | - Bénédicte Buffin-Meyer
- Institut National de la Santé et de la Recherche Médicale (INSERM)Toulouse, France
- Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de RangueilToulouse, France
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM)Toulouse, France
- Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de RangueilToulouse, France
| |
Collapse
|
42
|
Effects of angiotensin receptor blocker on phenotypic alterations of podocytes in early diabetic nephropathy. Am J Med Sci 2011; 341:207-14. [PMID: 21326079 DOI: 10.1097/maj.0b013e3182010da9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Emerging evidence suggests that podocyte injury is a crucial event in the stage of diabetic nephropathy (DN), a process in which angiotensin II is implicated. In this study, the authors investigated the influence of irbesartan, an angiotensin receptor blocker, on the phenotypic alterations of podocytes in experimental DN. METHODS DN was induced by combination of high-sucrose, high-fat diet and intraperitoneal injection of low dose of streptozotocin (35 mg/kg) in spontaneously hypertensive rats. Diabetic rats were treated with irbesartan (50 mg/kg/d) by gavage for 8 weeks. Nondiabetic normotensive Wistar-Kyoto rats, which have the same genetic background as spontaneously hypertensive rat, were used as controls. The renal histological changes were investigated by light and electron microscopy. The epithelial marker of nephrin and mesenchymal marker of desmin were detected by real-time reverse transcriptase-polymerase chain reaction and Western blotting. RESULTS Compared with controls, diabetic rats were associated with mesangial matrix deposition, thickening of glomerular basement membrane, albuminuria, loss of podocytes and effacement of foot processes. Furthermore, the expression of nephrin was significantly reduced whereas desmin was increased. Irbesartan treatment not only lowered blood pressure and albuminuria but also attenuated podocyte loss, maintenance of nephrin expression and inhibition of desmin expression. CONCLUSIONS This study demonstrates that early irbesartan intervention attenuates the podocyte damage and ameliorates phenotypic alterations of podocytes, which provides a novel insight for the early application of angiotensin receptor blocker to prevent the development of DN.
Collapse
|
43
|
Glomerular podocytes: A study of mechanical properties and mechano-chemical signaling. Biochem Biophys Res Commun 2011; 406:229-33. [DOI: 10.1016/j.bbrc.2011.02.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 02/04/2011] [Indexed: 11/22/2022]
|
44
|
Campbell KN, Raij L, Mundel P. Role of angiotensin II in the development of nephropathy and podocytopathy of diabetes. Curr Diabetes Rev 2011; 7:3-7. [PMID: 21067505 PMCID: PMC3690294 DOI: 10.2174/157339911794273973] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 09/10/2010] [Indexed: 01/03/2023]
Abstract
Diabetic kidney disease is the leading cause of end-stage renal disease worldwide. Podocytes are highly differentiated, pericyte-like cells that are essential for normal function of the kidney filter. Loss of podocytes is a hallmark of progressive kidney diseases including diabetic nephropathy. Podocytes are a direct target for angiotensin II - mediated injury by altered expression and distribution of podocyte proteins. Additionally, angiotensin II promotes podocyte injury indirectly by increasing calcium influx and production of reactive oxygen species. Notwithstanding the convincing rationale for angiotensin II blockade as a treatment modality, the incidence of diabetes-related end stage renal disease has increased steadily despite widespread use of angiotensin converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs). Recently published clinical trials have rekindled a debate on the safety and efficacy of dual blockade of the renin-angiotensin system (RAS). This review summarizes the rationale for blockade of angiotensin II as a therapeutic target in treating diabetic kidney disease, including the critical role played by podocytes. Recent relevant clinical trials on the role of RAS blockade in the treatment of diabetic kidney disease are discussed.
Collapse
Affiliation(s)
- Kirk N Campbell
- Department of Medicine, Leonard Miller School of Medicine, University of Miami, 1580 NW 10th Avenue, Miami, FL 33136, USA.
| | | | | |
Collapse
|
45
|
Schordan S, Schordan E, Endlich K, Endlich N. AlphaV-integrins mediate the mechanoprotective action of osteopontin in podocytes. Am J Physiol Renal Physiol 2010; 300:F119-32. [PMID: 21048023 DOI: 10.1152/ajprenal.00143.2010] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Increased mechanical load in podocytes due to glomerular hypertension is one of the important factors leading to podocyte damage and chronic kidney disease. In previous studies, we have shown that mechanical stretch increases osteopontin (OPN) expression in podocytes and that exogenous OPN is mechanoprotective via facilitating cytoskeletal reorganization of podocytes. In the present study, we asked whether the mechanoprotective effect of OPN in podocytes is mediated through specific integrins and whether endogenous OPN of podocytes is required for mechanoprotection. Conditionally immortalized mouse podocytes and primary podocytes (PP) from OPN-/- and OPN+/+ mice were used. Cyclic biaxial mechanical stretch (0.5 Hz, 7% linear strain) was applied for up to 3 days. Stretch-induced cell loss was ∼30% higher in OPN-/- PP compared with OPN+/+ PP. Increased cell loss of OPN-/- PP was rescued by OPN coating. Analysis of integrin expression by RT-PCR, application of RGD and SLAYGLR peptides and anti-integrin antibodies, small-interfering RNA knockdown of integrins, and application of kinase inhibitors identified αV-integrins (αVβ1, αVβ3, and αVβ5) to mediate the mechano-protective effect of OPN in podocytes involving focal adhesion kinase, Src, phosphatidylinositol 3-kinase, and mitogen-activated protein kinase. Our results demonstrate that endogenous OPN of podocytes plays a nonredundant role in podocyte adaptation to mechanical stretch, and that OPN signaling via α(V)-integrins may represent a relevant therapeutical target in podocytes.
Collapse
Affiliation(s)
- Sandra Schordan
- Department of Anatomy and Cell Biology, University of Greifswald, Greifswald, Germany
| | | | | | | |
Collapse
|
46
|
López-Novoa JM, Martínez-Salgado C, Rodríguez-Peña AB, Hernández FJL. Common pathophysiological mechanisms of chronic kidney disease: Therapeutic perspectives. Pharmacol Ther 2010; 128:61-81. [DOI: 10.1016/j.pharmthera.2010.05.006] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 05/25/2010] [Indexed: 12/17/2022]
|
47
|
Sakoda M, Ichihara A, Kurauchi-Mito A, Narita T, Kinouchi K, Murohashi-Bokuda K, Saleem MA, Nishiyama A, Suzuki F, Itoh H. Aliskiren inhibits intracellular angiotensin II levels without affecting (pro)renin receptor signals in human podocytes. Am J Hypertens 2010; 23:575-80. [PMID: 20075844 DOI: 10.1038/ajh.2009.273] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND A direct renin inhibitor (DRI) had a benefit in decreasing albuminuria in type 2 diabetic patients having already been treated with angiotensin (Ang) II type 1 receptor blocker (ARB), suggesting that aliskiren may have another effect other than blockade of the traditional renin-angiotensin system (RAS). Recently, prorenin bound to (pro)renin receptor ((P)RR) was found and shown to evoke two pathways; the generation of Ang peptides and the receptor-dependent activation of extracellular signal-related protein kinase (ERK). Because (P)RR is present in the podocytes, a central component of the glomerular filtration barrier, we hypothesized that aliskiren influences the (P)RR-induced two pathways in human podocytes. METHODS Human podocytes were treated with 2 nmol/l prorenin in the presence and absence of an angiotensin-converting enzyme inhibitor (ACEi) imidaprilat, an ARB candesartan, a DRI aliskiren, or the siRNA knocking down the (P)RR mRNA and the intracellular AngII levels and the phosphorylation of ERK were determined. RESULTS The expression of (P)RR mRNA of human podocytes was unaffected by the treatment with RAS inhibitors, but decreased by 69% with the siRNA treatment. The basal levels of intracellular AngII and the prorenin-induced increase in intracellular AngII were significantly reduced by aliskiren and siRNA treatment, compared with imidaprilat and candesartan. The prorenin-induced ERK activation was reduced to control level by the siRNA treatment, but it was unaffected by imidaprilat, candesartan, or aliskiren. CONCLUSIONS Aliskiren is the most potent inhibitor of intracellular AngII levels of human podocytes among RAS inhibitors, although it is incapable of inhibiting the (P)RR-dependent ERK phosphorylation.
Collapse
|
48
|
Faour WH, Thibodeau JF, Kennedy CRJ. Mechanical stretch and prostaglandin E2 modulate critical signaling pathways in mouse podocytes. Cell Signal 2010; 22:1222-30. [PMID: 20362052 DOI: 10.1016/j.cellsig.2010.03.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 03/23/2010] [Accepted: 03/24/2010] [Indexed: 11/27/2022]
Abstract
Elevated glomerular capillary pressure (Pgc) and hyperglycemia contribute to glomerular filtration barrier injury observed in diabetic nephropathy (DN). Previous studies showed that hypertensive conditions alone or in combination with a diabetic milieu impact podocyte cellular function which results in podocyte death, detachment or hypertrophy. The present study was aimed at uncovering the initial signaling profile activated by Pgc (mimicked by in vitro mechanical stretch), hyperglycemia (high glucose (HG), 25mM d-glucose) and prostaglandin E(2) (PGE(2)) in conditionally-immortalized mouse podocytes. PGE(2) significantly reduced the active form of AKT by selectively blunting its phosphorylation on S473, but not on T308. AKT inhibition by PGE(2) was reversed following either siRNA-mediated EP(4) knockdown, PKA inhibition (H89), or phosphatase inhibition (orthovanadate). Podocytes treated for 20min with H(2)O(2) (10(-4)M), which mimics reactive oxygen species generation by cells challenged by hyperglycemic or enhanced Pgc conditions, significantly increased the levels of active p38 MAPK, AKT, JNK and ERK1/2. Interestingly, stretch and PGE(2) each significantly reduced H(2)O(2)-mediated AKT phosphorylation and was reversed by pretreatment with orthovanadate while stretch alone reduced GSK-3beta inhibitory phosphorylation at ser-9. Finally, mechanical stretch alone or in combination with HG, induced ERK1/2 and JNK activation, via the EGF receptor since AG1478, a specific EGF receptor kinase inhibitor, blocked this activation. These results show that cellular signaling in podocytes is significantly altered under diabetic conditions (i.e., hyperglycemia and increased Pgc). These changes in MAPKs and AKT activities might impact cellular integrity required for a functional glomerular filtration barrier thereby contributing to the onset of proteinuria in DN.
Collapse
Affiliation(s)
- Wissam H Faour
- School of Medicine, Lebanese American University, Byblos, Lebanon.
| | | | | |
Collapse
|
49
|
Romero M, Ortega A, Izquierdo A, López-Luna P, Bosch RJ. Parathyroid hormone-related protein induces hypertrophy in podocytes via TGF-beta(1) and p27(Kip1): implications for diabetic nephropathy. Nephrol Dial Transplant 2010; 25:2447-57. [PMID: 20200004 DOI: 10.1093/ndt/gfq104] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Hypertrophy of podocytes is characteristic in diabetic nephropathy (DN). Previously, we observed the upregulation of parathyroid hormone-related protein (PTHrP) and its receptor PTH1R, in experimental DN, associated with renal hypertrophy. Herein, we test the hypothesis that PTHrP participates in the mechanism of high glucose (HG)-induced podocyte hypertrophy. METHODS On mouse podocytes, hypertrophy was assessed by protein content/cell and [H(3)]leucine incorporation. Podocytes were stimulated with HG (25 mM), PTHrP(1-36) (100 nM), angiotensin II (AngII) (100 nM) or TGF-beta(1) (5 ng/mL) in the presence or absence of PTHrP-neutralizing antibodies (alpha-PTHrP), the PTH1R antagonist JB4250 (10 microM), PTHrP silencer RNA (siRNA) or TGF-beta(1) siRNA. Protein expression was analysed by western blot and immunohistochemistry. RESULTS HG-induced hypertrophy was abolished in the presence of either alpha-PTHrP or PTHrP siRNA. This effect was associated with an inhibition of the upregulation of TGF-beta(1) and p27(Kip1). JB4250 also inhibited HG-induced p27(Kip1) upregulation. Interestingly, whilst HG and AngII were unable to stimulate the expression of p27(Kip1) on PTHrP siRNA-transfected podocytes, TGF-beta(1) was still able to upregulate p27(Kip1) in these cells. Moreover, HG and PTHrP-induced hypertrophy as well as p27(Kip1) upregulation were abolished on TGF-beta(1) siRNA-transfected podocytes. Furthermore, the glomeruli of transgenic PTHrP-overexpressing mice showed a constitutive overexpression of TGF-beta(1) and p27(Kip1) to a degree similar to that of diabetic animals. CONCLUSIONS PTHrP seems to participate in the hypertrophic signalling triggered by HG. In this condition, AngII induces the upregulation of PTHrP, which might induce the expression of TGF-beta(1) and p27(Kip1). These findings provide new insights into the protective effects of AngII antagonists in DN, opening new paths for intervention.
Collapse
Affiliation(s)
- Montserrat Romero
- Laboratory of Renal Physiology and Experimental Nephrology, Department of Physiology, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | | | | | | | | |
Collapse
|
50
|
Lewko B, Stepinski J. Hyperglycemia and mechanical stress: Targeting the renal podocyte. J Cell Physiol 2009; 221:288-95. [DOI: 10.1002/jcp.21856] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|