1
|
de Zoysa N, Haruhara K, Nikolic-Paterson DJ, Kerr PG, Ling J, Gazzard SE, Puelles VG, Bertram JF, Cullen-McEwen LA. Podocyte number and glomerulosclerosis indices are associated with the response to therapy for primary focal segmental glomerulosclerosis. Front Med (Lausanne) 2024; 11:1343161. [PMID: 38510448 PMCID: PMC10951056 DOI: 10.3389/fmed.2024.1343161] [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: 11/23/2023] [Accepted: 02/19/2024] [Indexed: 03/22/2024] Open
Abstract
Corticosteroid therapy, often in combination with inhibition of the renin-angiotensin system, is first-line therapy for primary focal and segmental glomerulosclerosis (FSGS) with nephrotic-range proteinuria. However, the response to treatment is variable, and therefore new approaches to indicate the response to therapy are required. Podocyte depletion is a hallmark of early FSGS, and here we investigated whether podocyte number, density and/or size in diagnostic biopsies and/or the degree of glomerulosclerosis could indicate the clinical response to first-line therapy. In this retrospective single center cohort study, 19 participants (13 responders, 6 non-responders) were included. Biopsies obtained at diagnosis were prepared for analysis of podocyte number, density and size using design-based stereology. Renal function and proteinuria were assessed 6 months after therapy commenced. Responders and non-responders had similar levels of proteinuria at the time of biopsy and similar kidney function. Patients who did not respond to treatment at 6 months had a significantly higher percentage of glomeruli with global sclerosis than responders (p < 0.05) and glomerulosclerotic index (p < 0.05). Podocyte number per glomerulus in responders was 279 (203-507; median, IQR), 50% greater than that of non-responders (186, 118-310; p < 0.05). These findings suggest that primary FSGS patients with higher podocyte number per glomerulus and less advanced glomerulosclerosis are more likely to respond to first-line therapy at 6 months. A podocyte number less than approximately 216 per glomerulus, a GSI greater than 1 and percentage global sclerosis greater than approximately 20% are associated with a lack of response to therapy. Larger, prospective studies are warranted to confirm whether these parameters may help inform therapeutic decision making at the time of diagnosis of primary FSGS.
Collapse
Affiliation(s)
- Natasha de Zoysa
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| | - Kotaro Haruhara
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Division of Nephrology and Hypertension, Jikei University School of Medicine, Tokyo, Japan
| | - David J. Nikolic-Paterson
- Department of Nephrology, Monash Medical Centre, Clayton, VIC, Australia
- Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
| | - Peter G. Kerr
- Department of Nephrology, Monash Medical Centre, Clayton, VIC, Australia
- Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
| | - Jonathan Ling
- Department of Nephrology, Monash Medical Centre, Clayton, VIC, Australia
- Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
| | - Sarah E. Gazzard
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| | - Victor G. Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - John F. Bertram
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Melbourne, VIC, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Brisbane, QLD, Australia
| | - Luise A. Cullen-McEwen
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| |
Collapse
|
2
|
Yamamoto T, Kase S, Murata M, Ishida S. Serum advanced glycation end‑products and αB‑crystallin in diabetic retinopathy patients. Biomed Rep 2022; 16:28. [DOI: 10.3892/br.2022.1511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/04/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Taku Yamamoto
- Laboratory of Ocular Cell Biology and Visual Science, Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060‑8638, Japan
| | - Satoru Kase
- Laboratory of Ocular Cell Biology and Visual Science, Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060‑8638, Japan
| | - Miyuki Murata
- Laboratory of Ocular Cell Biology and Visual Science, Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060‑8638, Japan
| | - Susumu Ishida
- Laboratory of Ocular Cell Biology and Visual Science, Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060‑8638, Japan
| |
Collapse
|
3
|
Abstract
Nuclear receptors have a broad spectrum of biological functions in normal physiology and in the pathology of various diseases, including glomerular disease. The primary therapies for many glomerular diseases are glucocorticoids, which exert their immunosuppressive and direct podocyte protective effects via the glucocorticoid receptor (GR). As glucocorticoids are associated with important adverse effects and a substantial proportion of patients show resistance to these therapies, the beneficial effects of selective GR modulators are now being explored. Peroxisome proliferator-activated receptor-γ (PPARγ) agonism using thiazolidinediones has potent podocyte cytoprotective and nephroprotective effects. Repurposing of thiazolidinediones or identification of novel PPARγ modulators are potential strategies to treat non-diabetic glomerular disease. Retinoic acid receptor-α is the key mediator of the renal protective effects of retinoic acid, and repair of the endogenous retinoic acid pathway offers another potential therapeutic strategy for glomerular disease. Vitamin D receptor, oestrogen receptor and mineralocorticoid receptor modulators regulate podocyte injury in experimental models. Further studies are needed to better understand the mechanisms of these nuclear receptors, evaluate their synergistic pathways and identify their novel modulators. Here, we focus on the role of nuclear receptors in podocyte biology and non-diabetic glomerular disease.
Collapse
|
4
|
Waller AP, Agrawal S, Wolfgang KJ, Kino J, Chanley MA, Smoyer WE, Kerlin BA. Nephrotic syndrome-associated hypercoagulopathy is alleviated by both pioglitazone and glucocorticoid which target two different nuclear receptors. Physiol Rep 2020; 8:e14515. [PMID: 32776495 PMCID: PMC7415912 DOI: 10.14814/phy2.14515] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Thrombosis is a potentially life-threatening nephrotic syndrome (NS) complication. We have previously demonstrated that hypercoagulopathy is proportional to NS severity in rat models and that pioglitazone (Pio) reduces proteinuria both independently and in combination with methylprednisolone (MP), a glucocorticoid (GC). However, the effect of these treatments on NS-associated hypercoagulopathy remains unknown. We thus sought to determine the ability of Pio and GC to alleviate NS-associated hypercoagulopathy. METHODS Puromycin aminonucleoside-induced rat NS was treated with sham, Low- or High-dose MP, Pio, or combination (Pio + Low-MP) and plasma was collected at day 11. Plasma samples were collected from children with steroid-sensitive NS (SSNS) and steroid-resistant NS (SRNS) upon presentation and after 7 weeks of GC therapy. Plasma endogenous thrombin potential (ETP), antithrombin (AT) activity, and albumin (Alb) were measured using thrombin generation, amidolytic, and colorimetric assays, respectively. RESULTS In a rat model of NS, both High-MP and Pio improved proteinuria and corrected hypoalbuminemia, ETP and AT activity (p < .05). Proteinuria (p = .005) and hypoalbuminemia (p < .001) were correlated with ETP. In childhood NS, while ETP was not different at presentation, GC therapy improved proteinuria, hypoalbuminemia, and ETP in children with SSNS (p < .001) but not SRNS (p = .330). CONCLUSIONS Both Pio and GC diminish proteinuria and significantly alleviate hypercoagulopathy. Both Pio and MP improved hypercoagulopathy in rats, and successful GC therapy (SSNS) also improved hypercoagulopathy in childhood NS. These data suggest that even a partial reduction in proteinuria may reduce NS-associated thrombotic risk.
Collapse
Affiliation(s)
- Amanda P. Waller
- Center for Clinical & Translational ResearchThe Abigail Wexner Research Institute at Nationwide Children'sColumbusOHUSA
| | - Shipra Agrawal
- Center for Clinical & Translational ResearchThe Abigail Wexner Research Institute at Nationwide Children'sColumbusOHUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOHUSA
| | - Katelyn J. Wolfgang
- Center for Clinical & Translational ResearchThe Abigail Wexner Research Institute at Nationwide Children'sColumbusOHUSA
| | - Jiro Kino
- Center for Clinical & Translational ResearchThe Abigail Wexner Research Institute at Nationwide Children'sColumbusOHUSA
| | - Melinda A. Chanley
- Center for Clinical & Translational ResearchThe Abigail Wexner Research Institute at Nationwide Children'sColumbusOHUSA
| | - William E. Smoyer
- Center for Clinical & Translational ResearchThe Abigail Wexner Research Institute at Nationwide Children'sColumbusOHUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOHUSA
| | - Bryce A. Kerlin
- Center for Clinical & Translational ResearchThe Abigail Wexner Research Institute at Nationwide Children'sColumbusOHUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOHUSA
| |
Collapse
|
5
|
Hosseiniyan Khatibi SM, Ardalan M, Abediazar S, Zununi Vahed S. The impact of steroids on the injured podocytes in nephrotic syndrome. J Steroid Biochem Mol Biol 2020; 196:105490. [PMID: 31586640 DOI: 10.1016/j.jsbmb.2019.105490] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/03/2019] [Accepted: 09/30/2019] [Indexed: 12/17/2022]
Abstract
Nephrotic syndrome (NS), a common chronic kidney disease, embraces a variety of kidney disorders. Though Glucocorticoids (GCs) are generally used in the treatment of NS, their mechanism of action is poorly understood. A plethora of evidence indicates that podocytes are considered as the main target cells for the therapeutic strategies to prevent NS. GCs regulate the transactivation and transrepression of genes in podocytes that affect their morphological and cytoskeletal features, motility, apoptosis and survival rate. Moreover, they prevent protein leakage through the glomerular barrier membrane by affecting the synthesis, trafficking and posttranslational modifications of slit diaphragms components, podocytes' intercellular junctions. The response to the treatment is variable among different ethnics and populations and resistance to the steroids is detected in almost 50% of adult patients. Not only do pharmacokinetics and pharmacogenetics of steroids play a role in GC resistance but also the genetic variations in one or more podocyte related genes are connected with the steroid resistance in cases with NS. The focus of this review is to explain the underlying cellular and molecular mechanisms of GCs in podocytes. Understanding the mechanisms by which the GCs and GCs receptors in podocytes regulate the gene expression network and crosstalk with other molecular pathways would guarantee an optimum therapeutic benefit of steroid treatment.
Collapse
Affiliation(s)
| | | | - Sima Abediazar
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
6
|
Agrawal S, Merchant ML, Kino J, Li M, Wilkey DW, Gaweda AE, Brier ME, Chanley MA, Gooding JR, Sumner SJ, Klein JB, Smoyer WE. Predicting and Defining Steroid Resistance in Pediatric Nephrotic Syndrome Using Plasma Proteomics. Kidney Int Rep 2020; 5:66-80. [PMID: 31922062 PMCID: PMC6943770 DOI: 10.1016/j.ekir.2019.09.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/09/2019] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Nephrotic syndrome (NS) is a characterized by massive proteinuria, edema, hypoalbuminemia, and dyslipidemia. Glucocorticoids (GCs), the primary therapy for >60 years, are ineffective in approximately 50% of adults and approximately 20% of children. Unfortunately, there are no validated biomarkers able to predict steroid-resistant NS (SRNS) or to define the pathways regulating SRNS. METHODS We performed proteomic analyses on paired pediatric NS patient plasma samples obtained both at disease presentation before glucocorticoid initiation and after approximately 7 weeks of GC therapy to identify candidate biomarkers able to either predict steroid resistance before treatment or define critical molecular pathways/targets regulating steroid resistance. RESULTS Proteomic analyses of 15 paired NS patient samples identified 215 prevalent proteins, including 13 candidate biomarkers that predicted SRNS before GC treatment, and 66 candidate biomarkers that mechanistically differentiated steroid-sensitive NS (SSNS) from SRNS. Ingenuity Pathway Analyses and protein networking pathways approaches further identified proteins and pathways associated with SRNS. Validation using 37 NS patient samples (24 SSNS/13 SRNS) confirmed vitamin D binding protein (VDB) and APOL1 as strong predictive candidate biomarkers for SRNS, and VDB, hemopexin (HPX), adiponectin (ADIPOQ), sex hormone-binding globulin (SHBG), and APOL1 as strong candidate biomarkers to mechanistically distinguish SRNS from SSNS. Logistic regression analysis identified a candidate biomarker panel (VDB, ADIPOQ, and matrix metalloproteinase 2 [MMP-2]) with significant ability to predict SRNS at disease presentation (P = 0.003; area under the receiver operating characteristic curve = 0.78). CONCLUSION Plasma proteomic analyses and immunoblotting of serial samples in childhood NS identified a candidate biomarker panel able to predict SRNS at disease presentation, as well as candidate molecular targets/pathways associated with clinical steroid resistance.
Collapse
Affiliation(s)
- Shipra Agrawal
- The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Ohio State University, Columbus, Ohio, USA
| | | | - Jiro Kino
- The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Ming Li
- Kidney Disease Program, University of Louisville, Louisville, Kentucky, USA
| | - Daniel W. Wilkey
- Kidney Disease Program, University of Louisville, Louisville, Kentucky, USA
| | - Adam E. Gaweda
- Kidney Disease Program, University of Louisville, Louisville, Kentucky, USA
| | - Michael E. Brier
- Kidney Disease Program, University of Louisville, Louisville, Kentucky, USA
| | - Melinda A. Chanley
- The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Jessica R. Gooding
- National Institutes of Health Eastern Regional Comprehensive Metabolomics Resource Core at UNC, Chapel Hill, North Carolina, USA
- Discovery, Science and Technology, RTI International, Research Triangle Park, North Carolina, USA
| | - Susan J. Sumner
- National Institutes of Health Eastern Regional Comprehensive Metabolomics Resource Core at UNC, Chapel Hill, North Carolina, USA
- Nutrition Research Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jon B. Klein
- Kidney Disease Program, University of Louisville, Louisville, Kentucky, USA
- Robley Rex VA Medical Center, Louisville, Kentucky, USA
| | - William E. Smoyer
- The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
7
|
Sun M, Song H, Ye Y, Yang Q, Xu X, Zhu X, Zhang J, Shi S, Wang J, Liu Z. Differential toxicities of triptolide to immortalized podocytes and the podocytes in vivo. Biomed Pharmacother 2018; 109:2375-2386. [PMID: 30551497 DOI: 10.1016/j.biopha.2018.11.081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 12/19/2022] Open
Abstract
Triptolide (TP) has an anti-proteinuric effect and is used for the treatment of podocytopathies. TP has also been shown to act directly on immortalized podocytes in culture to protect them from injury. In the present study, we examined the effect of TP on healthy podocytes both in vitro and in vivo to better understand the action of TP on podocytes. We found that treatment of TP at 10 ng/ml, a concentration that is routinely used for podocyte protection, was sufficient to activate pro-apoptotic signaling of MAPK p38, p53 and BAX and induced apoptosis in cultured podocytes; and higher concentrations of TP exacerbated the p38, p53 and BAX activations and apoptosis. Moreover, TP severely downregulated the genes that are essential for podocyte structure and function. Interestingly, in contrast with other agents TP-induced podocyte injury was not prevented by glucocorticoids. In vivo, high-dose TP treatment for prolonged time did not cause podocyte injury, essential genes downregulation, and proteinuria in mice. TP was also not toxic to the podocytes with isolated glomeruli ex vivo. In summary, TP is toxic to immortalized podocytes in culture but not to the podocytes in animals or isolated glomeruli ex vivo. Our study suggests that immortalized podocytes might have genetically evolved to become sensitive to TP toxicity and thus caution should be taken in interpreting data from immortalized podocytes. Nevertheless, in vivo TP could be as safe as glucocorticoids in treating podocytopathies. Finally, TP may be used as a unique in vitro model for studying steroid-resistant podocytopathies.
Collapse
Affiliation(s)
- Mengjie Sun
- Department of Nephrology, Jinling Hospital, Bengbu Medical College, Bengbu, China; National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Hui Song
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yuting Ye
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Qianqian Yang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xiaodong Xu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xiaodong Zhu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jiong Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Shaolin Shi
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| | - Jinquan Wang
- Department of Nephrology, Jinling Hospital, Bengbu Medical College, Bengbu, China; National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
8
|
Zhao X, Hwang DY, Kao HY. The Role of Glucocorticoid Receptors in Podocytes and Nephrotic Syndrome. NUCLEAR RECEPTOR RESEARCH 2018; 5. [PMID: 30417008 PMCID: PMC6224173 DOI: 10.11131/2018/101323] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Glucocorticoid receptor (GC), a founding member of the nuclear hormone receptor superfamily, is a glucocorticoid-activated transcription factor that regulates gene expression and controls the development and homeostasis of human podocytes. Synthetic glucocorticoids are the standard treatment regimens for proteinuria (protein in the urine) and nephrotic syndrome (NS) caused by kidney diseases. These include minimal change disease (MCD), focal segmental glomerulosclerosis (FSGS), membranous nephropathy (MN) and immunoglobulin A nephropathy (IgAN) or subsequent complications due to diabetes mellitus or HIV infection. However, unwanted side effects and steroid-resistance remain major issues for their long-term use. Furthermore, the mechanism by which glucocorticoids elicit their renoprotective activity in podocyte and glomeruli is poorly understood. Podocytes are highly differentiated epithelial cells that contribute to the integrity of kidney glomerular filtration barrier. Injury or loss of podocytes leads to proteinuria and nephrotic syndrome. Recent studies in multiple experimental models have begun to explore the mechanism of GC action in podocytes. This review will discuss progress in our understanding of the role of glucocorticoid receptor and glucocorticoids in podocyte physiology and their renoprotective activity in nephrotic syndrome.
Collapse
Affiliation(s)
- Xuan Zhao
- Department of Biochemistry, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, USA
| | - Daw-Yang Hwang
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hung-Ying Kao
- Department of Biochemistry, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, USA
| |
Collapse
|
9
|
Multiple Targets for Novel Therapy of FSGS Associated with Circulating Permeability Factor. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6232616. [PMID: 28951873 PMCID: PMC5603123 DOI: 10.1155/2017/6232616] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 05/10/2017] [Accepted: 06/15/2017] [Indexed: 01/13/2023]
Abstract
A plasma component is responsible for altered glomerular permeability in patients with focal segmental glomerulosclerosis. Evidence includes recurrence after renal transplantation, remission after plasmapheresis, proteinuria in infants of affected mothers, transfer of proteinuria to experimental animals, and impaired glomerular permeability after exposure to patient plasma. Therapy may include decreasing synthesis of the injurious agent, removing or blocking its interaction with cells, or blocking signaling or enhancing cell defenses to restore the permeability barrier and prevent progression. Agents that may prevent the synthesis of the permeability factor include cytotoxic agents or aggressive chemotherapy. Extracorporeal therapies include plasmapheresis, immunoadsorption with protein A or anti-immunoglobulin, or lipopheresis. Oral or intravenous galactose also decreases Palb activity. Studies of glomeruli have shown that several strategies prevent the action of FSGS sera. These include blocking receptor-ligand interactions, modulating cell reactions using indomethacin or eicosanoids 20-HETE or 8,9-EET, and enhancing cytoskeleton and protein interactions using calcineurin inhibitors, glucocorticoids, or rituximab. We have identified cardiotrophin-like cytokine factor 1 (CLCF-1) as a candidate for the permeability factor. Therapies specific to CLCF-1 include potential use of cytokine receptor-like factor (CRLF-1) and inhibition of Janus kinase 2. Combined therapy using multiple modalities offers therapy to reverse proteinuria and prevent scarring.
Collapse
|
10
|
Müller-Deile J, Schiffer M. Podocytes from the diagnostic and therapeutic point of view. Pflugers Arch 2017; 469:1007-1015. [PMID: 28508947 DOI: 10.1007/s00424-017-1993-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 05/04/2017] [Indexed: 01/23/2023]
Abstract
The central role of podocytes in glomerular diseases makes this cell type an interesting diagnostic tool as well as a therapeutic target. In this review, we discuss the current literature on the use of podocytes and podocyte-specific markers as non-invasive diagnostic tools in different glomerulopathies. Furthermore, we highlight the direct effects of drugs currently used to treat primary glomerular diseases and describe their direct cellular effects on podocytes. A new therapeutic potential is seen in drugs targeting the podocytic actin cytoskeleton which is essential for podocyte foot process structure and function. Incubation of cultured human podocyte cell lines with sera from patients with active glomerular diseases is currently also used to identify novel circulating factors with pathophysiological relevance for the glomerular filtration barrier. In addition, treatment of detached urinary podocytes from patients with substances that restore their cytoskeleton might serve as a novel personalized tool to estimate their potential for podocyte recovery ex vivo.
Collapse
Affiliation(s)
- Janina Müller-Deile
- Department of Nephrology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Mario Schiffer
- Department of Nephrology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
11
|
Gamal Y, Badawy A, Swelam S, Tawfeek MSK, Gad EF. Glomerular Glucocorticoid Receptors Expression and Clinicopathological Types of Childhood Nephrotic Syndrome. Fetal Pediatr Pathol 2017; 36:16-26. [PMID: 27690709 DOI: 10.1080/15513815.2016.1225872] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Glucocorticoids are primary therapy of idiopathic nephrotic syndrome (INS). However, not all children respond to steroid therapy. We assessed glomerular glucocorticoid receptor expression in fifty-one children with INS and its relation to response to steroid therapy and to histopathological type. Clinical, laboratory and glomerular expression of glucocorticoid receptors were compared between groups with different steroid response. Glomerular glucocorticoid expression was slightly higher in controls than in minimal change early responders, which in turn was significantly higher than in minimal change late responders. There was significantly lower glomerular glucocorticoid receptor expression in steroid-resistance compared to early responders, late responders and controls. Glomerular glucocorticoid expression was significantly higher in all minimal change disease (MCD) compared to focal segmental glomerulosclerosis. In INS, response to glucocorticoid is dependent on glomerular expression of receptors and peripheral expression. Evaluation of glomerular glucocorticoid receptor expression at time of diagnosis of NS can predict response to steroid therapy.
Collapse
Affiliation(s)
- Yasser Gamal
- a Pathology Department , Assiut University Faculty of Medicine , Assiut , Egypt
| | - Ahlam Badawy
- b Pediatric Department , Assiut University Faculty of Medicine , Assiut , Egypt
| | - Salwa Swelam
- c Pediatric Department , Minia University Faculty of Medicine , El Minia , Egypt
| | - Mostafa S K Tawfeek
- b Pediatric Department , Assiut University Faculty of Medicine , Assiut , Egypt
| | - Eman Fathalla Gad
- b Pediatric Department , Assiut University Faculty of Medicine , Assiut , Egypt
| |
Collapse
|
12
|
Sreedharan R, Van Why SK. Heat shock proteins in the kidney. Pediatr Nephrol 2016; 31:1561-70. [PMID: 26913726 DOI: 10.1007/s00467-015-3297-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/01/2015] [Accepted: 12/07/2015] [Indexed: 12/15/2022]
Abstract
Heat shock proteins (Hsps) are essential to cell survival through their function as protein chaperones. The role they play in kidney health and disease is varied. Hsp induction may be either beneficial or detrimental to the kidney, depending on the specific Hsp, type of cell, and context. This review addresses the role of Hsps in the kidney, including during development, as osmoprotectants, and in various kidney disease models. Heat shock transcription factor, activated by a stress on renal cells, induces Hsp elaboration and separately regulates immune responses that can contribute to renal injury. Induced Hsps in the intracellular compartment are mostly beneficial in the kidney by stabilizing and restoring cell architecture and function through acting as protein chaperones. Intracellular Hsps also inhibit apoptosis and facilitate cell proliferation, preserving renal tubule viability after acute injury, but enhancing progression of cystic kidney disease and malignancy. Induced Hsps in the extracellular compartment, either circulating or located on outer cell membranes, are mainly detrimental through enhancing inflammation pathways to injury. Correctly harnessing these stress proteins promises the opportunity to alter the course of acute and chronic kidney disease.
Collapse
Affiliation(s)
- Rajasree Sreedharan
- Pediatrics, Nephrology, Medical College of Wisconsin, 999 N. 92nd St., Suite C510, Milwaukee, WI, 53226, USA
| | - Scott K Van Why
- Pediatrics, Nephrology, Medical College of Wisconsin, 999 N. 92nd St., Suite C510, Milwaukee, WI, 53226, USA.
| |
Collapse
|
13
|
Mallipattu SK, Guo Y, Revelo MP, Roa-Peña L, Miller T, Ling J, Shankland SJ, Bialkowska AB, Ly V, Estrada C, Jain MK, Lu Y, Ma'ayan A, Mehrotra A, Yacoub R, Nord EP, Woroniecki RP, Yang VW, He JC. Krüppel-Like Factor 15 Mediates Glucocorticoid-Induced Restoration of Podocyte Differentiation Markers. J Am Soc Nephrol 2016; 28:166-184. [PMID: 27288011 DOI: 10.1681/asn.2015060672] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 04/05/2016] [Indexed: 12/18/2022] Open
Abstract
Podocyte injury is the inciting event in primary glomerulopathies, such as minimal change disease and primary FSGS, and glucocorticoids remain the initial and often, the primary treatment of choice for these glomerulopathies. Because inflammation is not readily apparent in these diseases, understanding the direct effects of glucocorticoids on the podocyte, independent of the immunomodulatory effects, may lead to the identification of targets downstream of glucocorticoids that minimize toxicity without compromising efficacy. Several studies showed that treatment with glucocorticoids restores podocyte differentiation markers and normal ultrastructure and improves cell survival in murine podocytes. We previously determined that Krüppel-like factor 15 (KLF15), a kidney-enriched zinc finger transcription factor, is required for restoring podocyte differentiation markers in mice and human podocytes under cell stress. Here, we show that in vitro treatment with dexamethasone induced a rapid increase of KLF15 expression in human and murine podocytes and enhanced the affinity of glucocorticoid receptor binding to the promoter region of KLF15 In three independent proteinuric murine models, podocyte-specific loss of Klf15 abrogated dexamethasone-induced podocyte recovery. Furthermore, knockdown of KLF15 reduced cell survival and destabilized the actin cytoskeleton in differentiated human podocytes. Conversely, overexpression of KLF15 stabilized the actin cytoskeleton under cell stress in human podocytes. Finally, the level of KLF15 expression in the podocytes and glomeruli from human biopsy specimens correlated with glucocorticoid responsiveness in 35 patients with minimal change disease or primary FSGS. Thus, these studies identify the critical role of KLF15 in mediating the salutary effects of glucocorticoids in the podocyte.
Collapse
Affiliation(s)
| | - Yiqing Guo
- Division of Nephrology, Departments of Medicine and
| | - Monica P Revelo
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | | | | | - Jason Ling
- Division of Nephrology, Departments of Medicine and
| | - Stuart J Shankland
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Agnieszka B Bialkowska
- Division of Gastroenterology, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - Victoria Ly
- Division of Nephrology, Departments of Medicine and
| | | | - Mukesh K Jain
- Case Cardiovascular Institute Research Institute, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Yuan Lu
- Case Cardiovascular Institute Research Institute, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Avi Ma'ayan
- Department of Pharmacology and Systems Therapeutics and
| | - Anita Mehrotra
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; and
| | - Rabi Yacoub
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; and
| | | | | | - Vincent W Yang
- Division of Gastroenterology, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - John C He
- Department of Pharmacology and Systems Therapeutics and.,Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; and.,Renal Section, James J. Peters Veterans Affairs Medical Center, New York, New York
| |
Collapse
|
14
|
Mallipattu SK, He JC. The podocyte as a direct target for treatment of glomerular disease? Am J Physiol Renal Physiol 2016; 311:F46-51. [PMID: 27097894 DOI: 10.1152/ajprenal.00184.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/18/2016] [Indexed: 11/22/2022] Open
Abstract
The Centers for Disease Control and Prevention estimates more than 10% of adults in the United States, over 20 million Americans, have chronic kidney disease (CKD). A failure to maintain the glomerular filtration barrier directly contributes to the onset of CKD. The visceral epithelial cells, podocytes, are integral to the maintenance of this renal filtration barrier. Direct podocyte injury contributes to the onset and progression of glomerular diseases such as minimal change disease (MCD), focal segmental glomerular sclerosis (FSGS), diabetic nephropathy, and HIV-associated nephropathy (HIVAN). Since podocytes are terminally differentiated with minimal capacity to self-replicate, they are extremely sensitive to cellular injury. In the past two decades, our understanding of the mechanism(s) by which podocyte injury occurs has greatly expanded. With this newfound knowledge, therapeutic strategies have shifted to identifying targets directed specifically at the podocyte. Although the systemic effects of these agents are important, their direct effect on the podocyte proves to be essential in ameliorating glomerular disease. In this review, we highlight the mechanisms by which these agents directly target the podocyte independent of its systemic effects.
Collapse
Affiliation(s)
- Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - John C He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; and Renal Section, James J. Peters VA Medical Center, New York, New York
| |
Collapse
|
15
|
Podocyte directed therapy of nephrotic syndrome-can we bring the inside out? Pediatr Nephrol 2016; 31:393-405. [PMID: 25939817 DOI: 10.1007/s00467-015-3116-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 12/15/2022]
Abstract
Several of the drugs currently used for the treatment of glomerular diseases are prescribed for their immunotherapeutic or anti-inflammatory properties, based on the current understanding that glomerular diseases are mediated by immune responses. In recent years our understanding of podocytic signalling pathways and the crucial role of genetic predispositions in the pathology of glomerular diseases has broadened. Delineation of those signalling pathways supports the hypothesis that several of the medications and immunosuppressive agents used to treat glomerular diseases directly target glomerular podocytes. Several central downstream signalling pathways merge into regulatory pathways of the podocytic actin cytoskeleton and its connection to the slit diaphragm. The slit diaphragm and the cytoskeleton of the foot process represent a functional unit. A breakdown of the cytoskeletal backbone of the foot processes leads to internalization of slit diaphragm molecules, and internalization of slit diaphragm components in turn negatively affects cytoskeletal signalling pathways. Podocytes display a remarkable ability to recover from complete effacement and to re-form interdigitating foot processes and intact slit diaphragms after pharmacological intervention. This ability indicates an active inside-out signalling machinery which stabilizes integrin complex formations and triggers the recycling of slit diaphragm molecules from intracellular compartments to the cell surface. In this review we summarize current evidence from patient studies and model organisms on the direct impact of immunosuppressive and supportive drugs on podocyte signalling pathways. We highlight new therapeutic targets that may open novel opportunities to enhance and stabilize inside-out pathways in podocytes.
Collapse
|
16
|
Rinschen MM, Benzing T, Limbutara K, Pisitkun T. Proteomic analysis of the kidney filtration barrier--Problems and perspectives. Proteomics Clin Appl 2015; 9:1053-68. [PMID: 25907645 DOI: 10.1002/prca.201400201] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/21/2015] [Accepted: 04/20/2015] [Indexed: 12/12/2022]
Abstract
Diseases of the glomerular filter of the kidney are a leading cause of end-stage renal failure. The kidney filter is localized within the renal glomeruli, small microvascular units that are responsible for ultrafiltration of about 180 liters of primary urine every day. The renal filter consists of three layers, fenestrated endothelial cells, glomerular basement membrane, and the podocytes, terminally differentiated, arborized epithelial cells. This review demonstrates the use of proteomics to generate insights into the regulation of the renal filtration barrier at a molecular level. The advantages and disadvantages of different glomerular purification methods are examined, and the technical limitations that have been significantly improved by in silico or biochemical approaches are presented. We also comment on phosphoproteomic studies that have generated considerable molecular-level understanding of the physiological regulation of the kidney filter. Lastly, we conclude with an analysis of urinary exosomes as a potential filter-derived resource for the noninvasive discovery of glomerular disease mechanisms.
Collapse
Affiliation(s)
- Markus M Rinschen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Kavee Limbutara
- Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Trairak Pisitkun
- Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
17
|
Levamisole in steroid-sensitive nephrotic syndrome: usefulness in adult patients and laboratory insights into mechanisms of action via direct action on the kidney podocyte. Clin Sci (Lond) 2015; 128:883-93. [PMID: 25626449 DOI: 10.1042/cs20140749] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Minimal change nephropathy (MCN) is the third most common cause of primary nephrotic syndrome in adults. Most patients with MCN respond to corticosteroid therapy, but relapse is common. In children, steroid-dependent patients are often given alternative agents to spare the use of steroids and to avoid the cumulative steroid toxicity. In this respect, levamisole has shown promise due to its ability to effectively maintain remission in children with steroid-sensitive or steroid-dependent nephrotic syndrome. Despite clinical effectiveness, there is a complete lack of molecular evidence to explain its mode of action and there are no published reports on the use of this compound in adult patients. We studied the effectiveness of levamisole in a small cohort of adult patients and also tested the hypothesis that levamisole's mode of action is attributable to its direct effects on podocytes. In the clinic, we demonstrate that in our adult patients, cohort levamisole is generally well tolerated and clinically useful. Using conditionally immortalized human podocytes, we show that levamisole is able to induce expression of glucocorticoid receptor (GR) and to activate GR signalling. Furthermore, levamisole is able to protect against podocyte injury in a puromycin aminonucleoside (PAN)-treated cell model. In this model the effects of levamisole are blocked by the GR antagonist mifepristone (RU486), suggesting that GR signalling is a critical target of levamisole's action. These results indicate that levamisole is effective in nephrotic syndrome in adults, as well as in children, and point to molecular mechanisms for this drug's actions in podocyte diseases.
Collapse
|
18
|
A case of nephrotic syndrome, showing evidence of response to saquinavir. Case Rep Nephrol 2015; 2015:512549. [PMID: 25802775 PMCID: PMC4329738 DOI: 10.1155/2015/512549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 01/06/2015] [Accepted: 01/11/2015] [Indexed: 11/17/2022] Open
Abstract
The treatment of primary nephrotic syndrome such as minimal change nephropathy, membranous nephropathy, and focal segmental glomerulosclerosis nephropathy remains challenging. Whilst most cases of idiopathic nephrotic syndrome respond to steroid therapy and experience a limited number of relapses prior to complete remission, some cases suffer from frequent relapses and become steroid dependent or are primarily steroid resistant. Treatment options are limited to immunosuppressive drugs with significant side effect profiles. New modalities targeting novel pathways in the pathogenesis of nephrotic syndrome are actively sought. Here we report the case of a patient with steroid dependent focal segmental glomerulosclerosis (FSGS) nephrotic syndrome with a favourable response to a novel proteasome inhibitor saquinavir.
Collapse
|
19
|
Nagayama Y, Braun GS, Jakobs CM, Maruta Y, van Roeyen CR, Klinkhammer BM, Boor P, Villa L, Raffetseder U, Trautwein C, Görtz D, Müller-Newen G, Ostendorf T, Floege J. Gp130-dependent signaling in the podocyte. Am J Physiol Renal Physiol 2014; 307:F346-55. [PMID: 24899055 DOI: 10.1152/ajprenal.00620.2013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Renal inflammation, in particular glomerular, is often characterized by increased IL-6 levels. The in vivo relevance of IL-6 signaling in glomerular podocytes, which play central roles in most glomerular diseases, is unknown. Here, we show that in normal mice, podocytes express gp130, the common signal-transducing receptor subunit of the IL-6 family of cytokines. Following systemic IL-6 or LPS injection in mice, podocyte IL-6 signaling was evidenced by downstream STAT3 phosphorylation. Next, we generated mice deficient for gp130 in podocytes. Expectedly, these mice exhibited abrogated IL-6 downstream signaling in podocytes. At the age of 40 wk, they did not show spontaneous renal pathology or abnormal renal function. The mice were then challenged using two LPS injury models as well as nephrotoxic serum to induce crescentic nephritis. Under all conditions, circulating IL-6 levels increased markedly and the mice developed the pathological hallmarks of the corresponding injury models such as proteinuria and development of glomerular crescents, respectively. However, despite the capacity of normal podocytes to transduce IL-6 family signals downstream, there were no significant differences between mice bearing the podocyte-specific gp130 deletion and their control littermates in any of these models. In conclusion, under the different conditions tested, gp130 signaling was not a critical component of the (patho-)biology of the podocyte in vivo.
Collapse
Affiliation(s)
- Yoshikuni Nagayama
- Division of Nephrology and Immunology, RWTH Aachen University, Aachen Germany; Division of Nephrology, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Gerald S Braun
- Division of Nephrology and Immunology, RWTH Aachen University, Aachen Germany; Institute of Biochemistry and Molecular Biology, RWTH Aachen University, Aachen, Germany;
| | - Christina M Jakobs
- Division of Nephrology and Immunology, RWTH Aachen University, Aachen Germany
| | - Yuichi Maruta
- Division of Nephrology and Immunology, RWTH Aachen University, Aachen Germany; Division of Nephrology, Showa University Fujigaoka Hospital, Yokohama, Japan
| | | | | | - Peter Boor
- Division of Nephrology and Immunology, RWTH Aachen University, Aachen Germany; Institute of Pathology, RWTH Aachen University, Aachen, Germany; Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia; and
| | - Luigi Villa
- Division of Nephrology and Immunology, RWTH Aachen University, Aachen Germany
| | - Ute Raffetseder
- Division of Nephrology and Immunology, RWTH Aachen University, Aachen Germany
| | - Christian Trautwein
- Division of Gastroenterology, Metabolic Diseases, and Intensive Care, RWTH Aachen University, Aachen, Germany
| | - Dieter Görtz
- Institute of Biochemistry and Molecular Biology, RWTH Aachen University, Aachen, Germany
| | - Gerhard Müller-Newen
- Institute of Biochemistry and Molecular Biology, RWTH Aachen University, Aachen, Germany
| | - Tammo Ostendorf
- Division of Nephrology and Immunology, RWTH Aachen University, Aachen Germany
| | - Jürgen Floege
- Division of Nephrology and Immunology, RWTH Aachen University, Aachen Germany
| |
Collapse
|
20
|
Altintas MM, Moriwaki K, Wei C, Möller CC, Flesche J, Li J, Yaddanapudi S, Faridi MH, Gödel M, Huber TB, Preston RA, Jiang JX, Kerjaschki D, Sever S, Reiser J. Reduction of proteinuria through podocyte alkalinization. J Biol Chem 2014; 289:17454-67. [PMID: 24817115 DOI: 10.1074/jbc.m114.568998] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Podocytes are highly differentiated cells and critical elements for the filtration barrier of the kidney. Loss of their foot process (FP) architecture (FP effacement) results in urinary protein loss. Here we show a novel role for the neutral amino acid glutamine in structural and functional regulation of the kidney filtration barrier. Metabolic flux analysis of cultured podocytes using genetic, toxic, and immunologic injury models identified increased glutamine utilization pathways. We show that glutamine uptake is increased in diseased podocytes to couple nutrient support to increased demand during the disease state of FP effacement. This feature can be utilized to transport increased amounts of glutamine into damaged podocytes. The availability of glutamine determines the regulation of podocyte intracellular pH (pHi). Podocyte alkalinization reduces cytosolic cathepsin L protease activity and protects the podocyte cytoskeleton. Podocyte glutamine supplementation reduces proteinuria in LPS-treated mice, whereas acidification increases glomerular injury. In summary, our data provide a metabolic opportunity to combat urinary protein loss through modulation of podocyte amino acid utilization and pHi.
Collapse
Affiliation(s)
- Mehmet M Altintas
- From the Department of Medicine, Rush University Medical Center, Chicago, Illinois 60035
| | - Kumiko Moriwaki
- the Department of Medicine, Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Changli Wei
- From the Department of Medicine, Rush University Medical Center, Chicago, Illinois 60035
| | - Clemens C Möller
- the Division of Nephrology and Program in Glomerular Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02129
| | - Jan Flesche
- the Division of Nephrology and Program in Glomerular Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02129
| | - Jing Li
- From the Department of Medicine, Rush University Medical Center, Chicago, Illinois 60035
| | - Suma Yaddanapudi
- the Division of Nephrology and Program in Glomerular Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02129
| | - Mohd Hafeez Faridi
- From the Department of Medicine, Rush University Medical Center, Chicago, Illinois 60035
| | - Markus Gödel
- the Renal Division, University Hospital Freiburg, 79106 Freiburg, Germany
| | - Tobias B Huber
- the Renal Division, University Hospital Freiburg, 79106 Freiburg, Germany, the BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs University, 79106 Freiburg, Germany
| | - Richard A Preston
- the Department of Medicine, Division of Clinical Pharmacology, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Jean X Jiang
- the Department of Biochemistry, University of Texas Health Science Center, San Antonio, Texas 78229, and
| | - Dontscho Kerjaschki
- the Clinical Institute of Pathology, Medical University of Vienna, 1090 Vienna, Austria
| | - Sanja Sever
- the Division of Nephrology and Program in Glomerular Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02129
| | - Jochen Reiser
- From the Department of Medicine, Rush University Medical Center, Chicago, Illinois 60035,
| |
Collapse
|
21
|
Cheng X, Zhao X, Khurana S, Bruggeman LA, Kao HY. Microarray analyses of glucocorticoid and vitamin D3 target genes in differentiating cultured human podocytes. PLoS One 2013; 8:e60213. [PMID: 23593176 PMCID: PMC3617172 DOI: 10.1371/journal.pone.0060213] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/22/2013] [Indexed: 12/26/2022] Open
Abstract
Glomerular podocytes are highly differentiated epithelial cells that are key components of the kidney filtration units. Podocyte damage or loss is the hallmark of nephritic diseases characterized by severe proteinuria. Recent studies implicate that hormones including glucocorticoids (ligand for glucocorticoid receptor) and vitamin D3 (ligand for vitamin D receptor) protect or promote repair of podocytes from injury. In order to elucidate the mechanisms underlying hormone-mediated podocyte-protecting activity from injury, we carried out microarray gene expression studies to identify the target genes and corresponding pathways in response to these hormones during podocyte differentiation. We used immortalized human cultured podocytes (HPCs) as a model system and carried out in vitro differentiation assays followed by dexamethasone (Dex) or vitamin D3 (VD3) treatment. Upon the induction of differentiation, multiple functional categories including cell cycle, organelle dynamics, mitochondrion, apoptosis and cytoskeleton organization were among the most significantly affected. Interestingly, while Dex and VD3 are capable of protecting podocytes from injury, they only share limited target genes and affected pathways. Compared to VD3 treatment, Dex had a broader and greater impact on gene expression profiles. In-depth analyses of Dex altered genes indicate that Dex crosstalks with a broad spectrum of signaling pathways, of which inflammatory responses, cell migration, angiogenesis, NF-κB and TGFβ pathways are predominantly altered. Together, our study provides new information and identifies several new avenues for future investigation of hormone signaling in podocytes.
Collapse
Affiliation(s)
- Xiwen Cheng
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU) and the Comprehensive Cancer Center of CWRU, Cleveland, Ohio, United States of America
| | - Xuan Zhao
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU) and the Comprehensive Cancer Center of CWRU, Cleveland, Ohio, United States of America
| | - Simran Khurana
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU) and the Comprehensive Cancer Center of CWRU, Cleveland, Ohio, United States of America
| | - Leslie A. Bruggeman
- Rammelkamp Center for Education and Research and Department of Medicine, MetroHealth Medical Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Hung-Ying Kao
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU) and the Comprehensive Cancer Center of CWRU, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
22
|
Yoshida Y, Miyamoto M, Taguchi I, Xu B, Zhang Y, Yaoita E, Fujinaka H, Yamamoto T. Human kidney glomerulus proteome and biomarker discovery of kidney diseases. Proteomics Clin Appl 2012; 2:420-7. [PMID: 21136843 DOI: 10.1002/prca.200780016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The kidney glomerulus is the site of plasma filtration and production of primary urine in the kidney. The structure not only plays a pivotal role in ultrafiltration of plasma into urine but also is the locus of kidney diseases progressing to chronic renal failure. Patients afflicted with these glomerular diseases frequently progress to irreversible loss of renal function and inevitably require replacement therapies. The diagnosis and treatment of glomerular diseases are now based on clinical manifestations, urinary protein excretion level, and renal pathology of needle biopsy specimens. The molecular mechanisms underlying the progression of glomerular diseases are still obscure despite a great number of clinical and experimental studies. Proteomics is a particularly promising approach for the discovery of proteins relevant to physiological and pathophysiological processes, and has been recently employed in nephrology. Although until now most efforts of proteomic analysis have been conducted with urine, the biological fluid that is easily collected without invasive procedures, proteomic analysis of the glomerulus, the tissue most proximal to the disease loci, is the most straightforward approach. In this review, we attempt to outline the current status of clinical proteomics of the glomerulus and provide a perspective of protein biomarker discovery of glomerular diseases.
Collapse
Affiliation(s)
- Yutaka Yoshida
- Department of Structural Pathology, Institute of Nephrology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Moysiadis DK, Perysinaki GS, Bertsias G, Stratakis S, Kyriacou K, Nakopoulou L, Boumpas DT, Daphnis E. Early treatment with glucocorticoids or cyclophosphamide retains the slit diaphragm proteins nephrin and podocin in experimental lupus nephritis. Lupus 2012; 21:1196-207. [DOI: 10.1177/0961203312451784] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Renal podocytes and their slit diaphragms ensure the integrity of renal basement membrane and prevent urinary protein loss. We have previously reported that decreases of the podocyte slit diaphragm proteins nephrin and podocin represent early events in the podocytopathy of lupus nephritis (LN). We asked whether immunosuppressive agents such as glucocorticoids and cyclophosphamide may have direct effects on podocytes. We assessed in New Zealand Black/New Zealand White (NZB/W) F1 LN mice glomerular nephrin and podocin expression and localization by the use of Western blot and immunofluorescence; mRNA levels were measured by real-time polymerase chain reaction (PCR) and renal histology by light and electron microscopy. Early treatment with glucocorticoids and cyclophosphamide halted the histologic alterations associated with LN, preserving podocyte foot processes. Nephrin and podocin protein expression significantly increased in both glucocorticoid and cyclophosphamide groups as early as after three months of therapy. Real-time PCR revealed similar enhancement in nephrin and podocin mRNA levels after three to six months of treatment. This study documents that early treatment in experimental LN with glucocorticoids or cyclophosphamide preserves slit diaphragm proteins in podocytes and halts histological changes of the glomeruli, thus raising the possibility of a direct protective effect of these drugs on podocytes.
Collapse
Affiliation(s)
- DK Moysiadis
- Laboratory of Nephrology, Medical Department, University of Crete, Greece
| | - GS Perysinaki
- Laboratory of Nephrology, Medical Department, University of Crete, Greece
| | - G Bertsias
- Rheumatology, Clinical Immunology and Allergy, Medical Department, University of Crete, Greece
| | - S Stratakis
- Laboratory of Nephrology, Medical Department, University of Crete, Greece
| | - K Kyriacou
- Department of Electron Microscopy/Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Cyprus
| | - L Nakopoulou
- Pathology Department, Medical School, National Kapodistrian University of Athens, Greece
| | - DT Boumpas
- Rheumatology, Clinical Immunology and Allergy, Medical Department, University of Crete, Greece
| | - E Daphnis
- Laboratory of Nephrology, Medical Department, University of Crete, Greece
| |
Collapse
|
24
|
Gordon J, Kopp JB. Off the beaten renin-angiotensin-aldosterone system pathway: new perspectives on antiproteinuric therapy. Adv Chronic Kidney Dis 2011; 18:300-11. [PMID: 21782136 DOI: 10.1053/j.ackd.2011.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 05/31/2011] [Accepted: 06/01/2011] [Indexed: 01/23/2023]
Abstract
CKD is a major public health problem in the developed and the developing world. The degree of proteinuria associated with renal failure is a generally well accepted marker of disease severity. Agents with direct antiproteinuric effects are highly desirable therapeutic strategies for slowing, or even halting, progressive loss of kidney function. We review progress on therapies acting further downstream of the renin-angiotensin-aldosterone system pathway (e.g., transforming growth factor-beta antagonism, endothelin antagonism) and on those acting independent of the renin-angiotensin-aldosterone system pathway. In all, we discuss 26 therapeutic targets or compounds and 2 lifestyle changes (dietary modification and weight loss) that have been used clinically for diabetic or nondiabetic kidney disease. These therapies include endogenous molecules (estrogens, isotretinoin), biologic antagonists (monoclonal antibodies, soluble receptors), and small molecules. Where mechanistic data are available, these therapies have been shown to exert favorable effects on glomerular cell phenotype. In some cases, recent work has indicated surprising new molecular pathways for some therapies, such as direct effects on the podocyte by glucocorticoids, rituximab, and erythropoietin. It is hoped that recent advances in the basic science of kidney injury will prompt development of more effective pharmaceutical and biologic therapies for proteinuria.
Collapse
|
25
|
Lindquist S, Hassinger S, Lindquist JA, Sailer M. The balance of pro-inflammatory and trophic factors in multiple sclerosis patients: effects of acute relapse and immunomodulatory treatment. Mult Scler 2011; 17:851-66. [DOI: 10.1177/1352458511399797] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background: In multiple sclerosis inflammation is primarily injurious to the central nervous system, but its therapeutic suppression might inhibit repair-promoting factors. Objectives: We aimed at better describing the complexity of biological effects during an acute relapse and analysed the effects of intervention with high-dose i.v. glucocorticoids and immunomodulatory treatment with interferon-beta (IFNβ). Methods: We studied the intracellular expression levels of the pro-inflammatory mediators tumour necrosis factor alpha (TNFα) and inducible nitric oxide synthase (iNOS) together with the neurotrophins ciliary neurotrophic factor (CNTF) and brain-derived neurotrophic factor (BDNF) in freshly isolated peripheral blood mononuclear cells of multiple sclerosis patients during an acute relapse, after intervention with i.v. methylprednisolone and at baseline, using a highly quantitative flow-cytometric approach. Results: We demonstrated the expression of CNTF in human leucocytes. We showed that CNTF levels differed in acutely relapsing multiple sclerosis patients compared with controls and increased after corticosteroid treatment. CNTF can counteract the toxicity of TNFα towards oligodendrocytes and we found TNFα increased during acute relapses. Following corticosteroids, neither TNFα nor iNOS expression was reduced. Levels of BDNF were not affected by glucocorticoids, but increased during IFNβ therapy. However, IFNβ also increased the expression of iNOS and major histocompatibility complex class I (MHC-I), underlining its immunomodulatory potential. Conclusions: Multiple sclerosis patients might benefit from reparative, and not solely from anti-inflammatory, effects of glucocorticoids. Interactive effects of glucocorticoid- and IFNβ-treatment need to be considered to improve neuroprotection and remyelination resulting from immunomodulatory treatment.
Collapse
Affiliation(s)
- Sabine Lindquist
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Leibniz-Institute for Neurobiology, Magdeburg, Germany
| | - Sarah Hassinger
- University Clinic for Neurology, Otto-von-Guericke-University, Magdeburg, Germany
| | - Jonathan A Lindquist
- Institute for Molecular and Clinical Immunology, Otto-von-Guericke-University, Magdeburg, Germany
- Co-senior authors
| | - Michael Sailer
- University Clinic for Neurology, Otto-von-Guericke-University, Magdeburg, Germany
- Centre for Neurological Rehabilitation, Magdeburg, Germany
- Co-senior authors
| |
Collapse
|
26
|
Blutke A, Block C, Berendt F, Herbach N, Kemter E, Amann K, Fröhlich T, Arnold GJ, Wanke R. Differential glomerular proteome analysis of two murine nephropathy models at onset of albuminuria. Proteomics Clin Appl 2011; 5:375-81. [DOI: 10.1002/prca.201000103] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 12/08/2010] [Accepted: 12/15/2010] [Indexed: 11/07/2022]
|
27
|
Abstract
Organs are complex structures that consist of multiple tissues with different levels of gene expression. To achieve comprehensive coverage and accurate quantitation data, organs ideally should be separated into morphologic and/or functional substructures before gene or protein expression analysis. However, because of complex morphology and elaborate isolation protocols, to date this often has been difficult to achieve. Kidneys are organs in which functional and morphologic subdivision is especially important. Each subunit of the kidney, the nephron, consists of more than 10 subsegments with distinct morphologic and functional characteristics. For a full understanding of kidney physiology, global gene and protein expression analyses have to be performed at the level of the nephron subsegments; however, such studies have been extremely rare to date. Here we describe the latest approaches in quantitative high-accuracy mass spectrometry-based proteomics and their application to quantitative proteomics studies of the whole kidney and nephron subsegments, both in human beings and in animal models. We compare these studies with similar studies performed on other organ substructures. We argue that the newest technologies used for preparation, processing, and measurement of small amounts of starting material are finally enabling global and subsegment-specific quantitative measurement of protein levels in the kidney and other organs. These new technologies and approaches are making a decisive impact on our understanding of the (patho)physiological processes at the molecular level.
Collapse
|
28
|
Marshall CB, Krofft RD, Blonski MJ, Kowalewska J, Logar CM, Pippin JW, Kim F, Feil R, Alpers CE, Shankland SJ. Role of smooth muscle protein SM22α in glomerular epithelial cell injury. Am J Physiol Renal Physiol 2011; 300:F1026-42. [PMID: 21289056 DOI: 10.1152/ajprenal.00187.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Podocytes are considered terminally differentiated cells in the mature kidney under normal conditions. In the face of injury, podocytes may proceed along several possible pathways, including dedifferentiation and proliferation, persistent cell cycle arrest, hypertrophy, apoptosis, or necrosis. There is mounting evidence that transdifferentiation into a dysregulated phenotype may also be a potential cell fate. We have previously reported that the transcript of SM22α, an actin-binding protein considered one of the earliest markers of smooth muscle differentiation, is upregulated nearly 70-fold in glomeruli of rats with passive Heymann nephritis (PHN). In contrast, the SM22α transcript is absent in normal adult rat glomeruli. The purpose of this study was to define SM22α's expression during kidney development and its role in glomerular diseases characterized by podocyte injury and proteinuria. During glomerulogenesis and podocyte differentiation, SM22α was expressed in glomeruli. This expression disappeared with glomerular maturation. Along with SM22α induction in PHN, confirmed at both mRNA and protein levels, SM22α was also induced across a broad range of proteinuric diseases, including experimental animal models (puromycin aminonucleoside nephropathy, adriamycin nephropathy, passive nephrotoxic nephritis, and diet-induced obesity) and human diseases (collapsing glomerulopathy, diabetic nephropathy, classic focal segmental glomerulosclerosis, IgA nephropathy, minimal-change disease, membranous nephropathy, and membranoproliferative glomerulonephritis). Crescentic glomerulonephritis was induced in SM22α +/+ and SM22α -/- mice by intraperitoneal injection of sheep anti-rabbit glomeruli antibody 12.5 mg/20 g body wt × 2 doses (n = 12-15/group), with mice euthanized at 7 and 14 days. Compared with SM22α -/- mice, SM22α +/+ mice demonstrated worse disease by histopathological parameters. In addition, there was greater apoptosis (cleaved caspase-3 immunostaining), fewer podocytes (Wilms' tumor-1 immunostaining), and less proliferation (Ki-67 immunostaining) in diseased SM22α +/+ mice. Furthermore, there was decreased activation of Erk1/2 in diseased SM22α +/+ mice. We conclude that the de novo expression of SM22α in glomerular epithelial cells affects the course of crescentic glomerulonephritis.
Collapse
Affiliation(s)
- Caroline B Marshall
- Div. of Nephrology, Department of Medicine, Univ. of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Schönenberger E, Ehrich JH, Haller H, Schiffer M. The podocyte as a direct target of immunosuppressive agents. Nephrol Dial Transplant 2010; 26:18-24. [PMID: 20937691 DOI: 10.1093/ndt/gfq617] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Podocytes play a key role in maintaining the blood-urine barrier for high-molecular-weight proteins. They are considered to be terminally differentiated, and podocyte loss cannot be compensated by regenerative proliferation. Various diseases leading to podocyte damage and loss result in proteinuria and cause nephrotic syndrome. Therefore, direct therapeutical strategies to protect podocytes in disease situations are a logical concept to prevent disease or to delay disease progression. Acquired podocytopathies like idiopathic focal segmental glomerulosclerosis and minimal change disease are historically considered as immunological diseases. Therefore, immunosuppressive agents such as steroids and calcineurin inhibitors are the commonly used treatment strategies. However, the causative disease mechanisms behind these treatment strategies remain elusive. Recent evidence shows that immunosuppressive agents, in addition to the effect on the immune system, directly influence the unique structure and function of podocytes. In this context, the actin cytoskeleton of the podocyte and cytokines such as vascular endothelial growth factor play a pivotal role. In this review, we summarize the direct effects on podocytes obtained in vivo and in vitro after treatment with calcineurin inhibitors, mTOR inhibitors and glucocorticoids. These direct effects could play a key role in the treatment concepts of podocytopathies with an important impact on the long-term renal function in patients with pharmacological immunosuppression.
Collapse
|
30
|
Abstract
Podocyte injury is a feature of glomerulopathies associated with proteinuria, which in turn has been used as a clinical prognostic factor for glomerular diseases. The goal of this study is to investigate the relationship between podocyte injury found in biopsied renal tissue and change of proteinuria in IgA nephropathy (IgAN). In all, 35 patients with biopsy-proven IgAN and proteinuria (>1.0 g per 24 h) were enrolled in the IgAN group, while 8 patients with excision of renal harmatoma or carcinoma served as kidney controls (Control). Immunohistochemistry was applied to detect the expression of nestin, cell-cycle regulatory protein p27, as well as complement C5b-9 and complement receptor 1 (CR1). Podocyte foot process width (FPW) and podocyte population in renal biopsied samples were measured by morphometric analysis. On the basis of the podocyte density (Nv), the IgAN patients were divided into podocytopenic group (n=17, Nv<57.10 /microm(3) x 10(6)) and normopodocytic group (n=18, Nv> or =57.10 /microm(3) x 10(6)). Changes of proteinuria were followed for 18 months after biopsy. Compared with the Control, IgAN glomeruli had reduced podocyte expression of p27 and nestin along with decreased podocyte number. IgAN glomeruli also showed activation of C5b-9 in mesangial and subepithelial areas with decreased CR1 expression in podocytes. The C5b-9 positivity was inversely correlated with the number of WT-1-positive podocytes. Although the magnitude of proteinuria at biopsy correlated with podocyte FPW (P<0.05), the change in the amount of proteinuria expressed as proteinuria progression rate significantly correlated with the podocyte density. Thus, the normopodocytic group showed significantly lower proteinuria progression rate than the podocytopenic group regardless the comparable clinical features at biopsy and treatment regimen between the two groups. The results of this study indicate that, in IgAN, podocyte injury is involved in development of proteinuria and loss of podocytes predicts progression of the proteinuria. Complement activation may contribute to podocyte damage in IgAN.
Collapse
|
31
|
Gbadegesin R, Lavin P, Janssens L, Bartkowiak B, Homstad A, Wu G, Bowling B, Eckel J, Potocky C, Abbott D, Conlon P, Scott WK, Howell D, Hauser E, Winn MP. A new locus for familial FSGS on chromosome 2p. J Am Soc Nephrol 2010; 21:1390-7. [PMID: 20616172 PMCID: PMC2938593 DOI: 10.1681/asn.2009101046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Accepted: 04/13/2010] [Indexed: 11/03/2022] Open
Abstract
FSGS is a clinicopathologic entity characterized by nephrotic syndrome and progression to ESRD. Although the pathogenesis is unknown, the podocyte seems to play a central role in this disorder. Here, we present six kindreds with hereditary FSGS that did not associate with mutations in known causal genes, and we report a new locus for the disease on chromosome 2p15 in one kindred. We performed genome-wide linkage analysis and refined the linkage area with microsatellite markers and haplotype analysis to define the minimal candidate region. Genome-wide linkage analysis yielded a maximum two-point logarithm of odds (LOD) score of 3.6 for the six families on chromosome 2p. One family contributed the largest proportion of the additive score (LOD 2.02) at this locus. Multipoint parametric LOD score calculation in this family yielded a significant LOD score of 3.1 at markers D2S393 and D2S337, and fine mapping of this region with microsatellite markers defined a minimal candidate region of 0.9 Mb with observed recombinations at markers D2S2332 and RS1919481. We excluded the remaining five families from linkage to this region by haplotype analysis. These data support a new gene locus for familial FSGS on chromosome 2p15. Identification of the mutated gene at this locus may provide further insight into the disease mechanisms of FSGS.
Collapse
Affiliation(s)
- Rasheed Gbadegesin
- Departments of *Pediatrics
- Center for Human Genetics, Duke University Medical Center, Durham, North Carolina
| | | | - Louis Janssens
- Department of Nephrology, AZ Groeninge, Kortrijk, Belgium
| | - Bartlomiej Bartkowiak
- Departments of *Pediatrics
- Center for Human Genetics, Duke University Medical Center, Durham, North Carolina
| | - Alison Homstad
- Departments of *Pediatrics
- Center for Human Genetics, Duke University Medical Center, Durham, North Carolina
| | | | | | | | | | | | - Peter Conlon
- Department of Nephrology, Beaumont Hospital, Dublin, Ireland; and
| | - William K. Scott
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute of Human Genomics, Miller School of Medicine, University of Miami, Miami, Florida
| | | | | | | |
Collapse
|
32
|
Guess A, Agrawal S, Wei CC, Ransom RF, Benndorf R, Smoyer WE. Dose- and time-dependent glucocorticoid receptor signaling in podocytes. Am J Physiol Renal Physiol 2010; 299:F845-53. [PMID: 20630936 DOI: 10.1152/ajprenal.00161.2010] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Glucocorticoids (GC) are the primary therapy for idiopathic nephrotic syndrome (NS). Recent evidence has identified glomerular podocytes as a potential site of GC action in this disease. The objectives of this study were to determine the presence of key components of the glucocorticoid receptor (GR) complex and the functionality of this signaling pathway in podocytes and to explore potential opportunities for manipulation of GC responsiveness. Here, we show that cultured murine podocytes express key components of the GR complex, including the GR, heat shock protein 90, and the immunophilins FKBP51 and FKBP52. The functionality of GR-mediated signaling was verified by measuring several GC (dexamethasone)-induced responses, including 1) increases in mRNA and protein levels of selected GC-regulated genes (FKBP51, phenol sulfotransferase 1, αB-crystallin); 2) downregulation of the GR protein; 3) increased phosphorylation of the GR; and 4) translocation of the GR into the nuclear fraction. Dexamethasone-induced phosphorylation and downregulation of GR protein were also demonstrated in isolated rat glomeruli. Podocyte gene expression in response to dexamethasone was regulated at both the transcriptional and posttranscriptional levels, the latter also including protein degradation. Short-term, high-dose GC treatment resulted in similar changes in gene expression and GR phosphorylation to that of long-term, low-dose GC treatment, thus providing a molecular rationale for the known efficacy of pulse GC therapy in NS. Induction of FKBP51 and downregulation of the GR represent negative feedback mechanisms that can potentially be exploited to improve clinical GC efficacy. Collectively, these findings demonstrate the presence of key molecular components of the GR signaling pathway and its functionality in podocytes and identify novel opportunities for improving clinical GC efficacy in the treatment of NS.
Collapse
Affiliation(s)
- Adam Guess
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, 700 Children's Dr., Columbus, OH 43205, USA
| | | | | | | | | | | |
Collapse
|
33
|
Matsumoto T, Hess S, Kajiyama H, Sakairi T, Saleem MA, Mathieson PW, Nojima Y, Kopp JB. Proteomic analysis identifies insulin-like growth factor-binding protein-related protein-1 as a podocyte product. Am J Physiol Renal Physiol 2010; 299:F776-84. [PMID: 20630940 DOI: 10.1152/ajprenal.00597.2009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The podocyte secretory proteome may influence the phenotype of adjacent podocytes, endothelial cells, parietal epithelial cells, and tubular epithelial cells but has not been systematically characterized. We have initiated studies to characterize this proteome, with the goal of further understanding the podocyte cell biology. We cultured differentiated conditionally immortalized human podocytes and subjected the proteins in conditioned medium to mass spectrometry. At a false discovery rate of <3%, we identified 111 candidates from conditioned medium, including 44 proteins that have signal peptides or are described as secreted proteins in the UniProt database. As validation, we confirmed that one of these proteins, insulin-like growth factor-binding protein-related protein-1 (IGFBP-rP1), was expressed in mRNA and protein of cultured podocytes. In addition, transforming growth factor-β1 stimulation increased IGFBP-rP1 in conditioned medium. We analyzed IGFBP-rP1 glomerular expression in a mouse model of human immunodeficiency virus-associated nephropathy. IGFBP-rP1 was absent from podocytes of normal mice and was expressed in podocytes and pseudocrescents of transgenic mice, where it was coexpressed with desmin, a podocyte injury marker. We conclude that IGFBP-rP1 may be a product of injured podocytes. Further analysis of the podocyte secretory proteome may identify biomarkers of podocyte injury.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Förster A, Emmler T, Schwalm S, Ebadi M, Heringdorf DMZ, Nieuwenhuis B, Kleuser B, Huwiler A, Pfeilschifter J. Glucocorticoids protect renal mesangial cells from apoptosis by increasing cellular sphingosine-1-phosphate. Kidney Int 2010; 77:870-9. [PMID: 20375982 DOI: 10.1038/ki.2010.62] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neutral ceramidase (NCDase) and sphingosine kinases (SphKs) are key enzymes regulating cellular sphingosine-1-phosphate (S1P) levels. In this study we found that stress factor-induced apoptosis of rat renal mesangial cells was significantly reduced by dexamethasone treatment. Concomitantly, dexamethasone increased cellular S1P levels, suggesting an activation of sphingolipid-metabolizing enzymes. The cell-protective effect of glucocorticoids was reversed by a SphK inhibitor, was completely absent in SphK1-deficient cells, and was associated with upregulated mRNA and protein expression of NCDase and SphK1. Additionally, in vivo experiments in mice showed that dexamethasone also upregulated SphK1 mRNA and activity, and NCDase protein expression in the kidney. Fragments (2285, 1724, and 1126 bp) of the rat NCDase promoter linked to a luciferase reporter were transfected into rat kidney fibroblasts and mesangial cells. There was enhanced NCDase promoter activity upon glucocorticoids treatment that was abolished by the glucocorticoid receptor antagonist RU-486. Single and double mutations of the two putative glucocorticoid response element sites within the promoter reduced the dexamethasone effect, suggesting that both glucocorticoid response elements are functionally active and required for induction. Our study shows that glucocorticoids exert a protective effect on stress-induced mesangial cell apoptosis in vitro and in vivo by upregulating NCDase and SphK1 expression and activity, resulting in enhanced levels of the protective lipid second messenger S1P.
Collapse
Affiliation(s)
- Ankathrin Förster
- Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Schordan S, Schordan E, Endlich N, Lindenmeyer MT, Meyer-Schwesinger C, Meyer TN, Giebel J, Cohen CD, Endlich K, Maurer MH. Alterations of the podocyte proteome in response to high glucose concentrations. Proteomics 2009; 9:4519-28. [PMID: 19688724 DOI: 10.1002/pmic.200800214] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Diabetic nephropathy is one of the most common complications of diabetes mellitus and the leading cause of end-stage renal disease. A reduction in podocyte number has been documented in the kidneys of these patients. To identify the molecular changes in podocytes that are primarily caused by high glucose (HG) concentrations and not by secondary alterations (e.g. glomerular hypertension), we investigated the protein expression profiles in a podocyte cell line under long-term HG exposure (30 versus 10 mM for 2 wk). Proteins were separated by 2-DE, and we identified 39 different proteins in 48 spots that were differentially regulated by more than twofold in response to HG concentrations using MALDI-TOF MS and MASCOT software. These proteins belong to several protein classes, including cytoskeletal proteins and specific annexins (annexins III and VI). Downregulation of annexins III and VI by HG concentrations was confirmed by qRT-PCR, Western blot, and immunostaining, and was also observed in glomeruli of kidney biopsies from patients with diabetic nephropathy. Our data demonstrate that HG concentrations per se are sufficient to strongly modify the protein expression profile of podocytes, the analysis of which contributes to the identification of novel targets involved in diabetic nephropathy.
Collapse
Affiliation(s)
- Sandra Schordan
- Department of Anatomy and Cell Biology, Ernst Moritz Arndt University, D-17487 Greifswald, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Vilasi A, Cutillas PR, Unwin RJ. Application of proteomic techniques to the study of urine and renal tissue. Proteomics Clin Appl 2008; 2:1564-74. [DOI: 10.1002/prca.200800035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Indexed: 01/28/2023]
|
37
|
Tesař V, Zima T. Recent Progress in the Pathogenesis of Nephrotic Proteinuria. Crit Rev Clin Lab Sci 2008; 45:139-220. [DOI: 10.1080/10408360801934865] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
38
|
Lescuyer P, Pernin A, Hainard A, Bigeire C, Burgess JA, Zimmermann-Ivol C, Sanchez JC, Schifferli JA, Hochstrasser DF, Moll S. Proteomic analysis of a podocyte vesicle-enriched fraction from human normal and pathological urine samples. Proteomics Clin Appl 2008; 2:1008-18. [DOI: 10.1002/prca.200800033] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Indexed: 12/15/2022]
|
39
|
NPHS3: new clues for understanding idiopathic nephrotic syndrome. Pediatr Nephrol 2008; 23:847-50. [PMID: 18270750 DOI: 10.1007/s00467-008-0747-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2007] [Revised: 11/26/2007] [Accepted: 12/14/2007] [Indexed: 01/15/2023]
Abstract
Hereditary forms of childhood nephrotic syndrome (H-CHNS) have long been counted as rare variants of steroid-resistant nephrotic syndrome (SRNS). This concept must be specified by two new findings: First, a study on nephrotic syndrome manifesting in the first year of life documents that H-CHNS are actually the predominant cause of nephrotic syndrome in infants. Second, the recent identification of autosomal recessive nephrotic syndrome type 3 (NPHS3) caused by mutations in the phospholipase PLCE1 gene has, for the first time, shown steroid responsiveness in H-CHNS. NPHS3 is a severe form of isolated nephrotic syndrome with rapid progression to terminal renal failure. NPHS3 is caused by a developmental rather than structural podocyte dysfunction and is a major cause of diffuse mesangial sclerosis. Therapy response in NPHS3 is documented and could open insights into direct genomic and nongenomic effects of glucocorticoids on podocytes. The findings on NPHS3 support the idea that both clinical course and histology in H-CHNS are subject to genotypic variability and that mutational analysis is the most reliable diagnostic tool. Future studies are needed to determine the clinical implications of NPHS3. Identification of further variants of H-CHNS can be anticipated and may include steroid-responsive hereditary diseases.
Collapse
|
40
|
Raaijmakers R, Pluk W, Schröder CH, Gloerich J, Cornelissen EAM, Wessels HJCT, Willems JL, Monnens LAH, van den Heuvel LPWJ. Proteomic profiling and identification in peritoneal fluid of children treated by peritoneal dialysis. Nephrol Dial Transplant 2008; 23:2402-5. [PMID: 18424818 DOI: 10.1093/ndt/gfn212] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Proteomic technologies offer a high-throughput analysis of the expression of proteins in biological samples. The global analysis of the proteins in peritoneal dialysis (PD) fluid will provide a better understanding of the biological processes of the peritoneal membrane. METHODS The dialysate of nine paediatric PD patients was collected from peritoneal equilibrium tests with 3.86% glucose. Proteins were separated on a 10% SDS-PAGE gel and in-gel digested with trypsin. Peptide mixtures were analysed using nanoLC-MS/MS and results were searched against the NCBI database. RESULTS A total number of 189 proteins were identified in the PD fluid of nine patients, with 88 proteins shared by all patients. These 88 proteins accounted for 47% of the identified proteins and >90% of the total protein content in the analysed samples. Proteins were subdivided into eight different classes according to function. CONCLUSIONS This study gives a representative overview of the proteins present in PD fluid. The proteins in PD fluid reflect plasma proteins as well as local peritoneal processes. Potentially interesting proteins are revealed.
Collapse
Affiliation(s)
- Renske Raaijmakers
- Department of Paediatric Nephrology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Viney RL, Morrison AA, van den Heuvel LP, Ni L, Mathieson PW, Saleem MA, Ladomery MR. A proteomic investigation of glomerular podocytes from a Denys-Drash Syndrome patient with a mutation in the Wilms tumour suppressor geneWT1. Proteomics 2007; 7:804-15. [PMID: 17295355 DOI: 10.1002/pmic.200600666] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Glomerular podocytes are essential for blood filtration in the kidney underpinned by their unique cytoskeletal morphology. An increasing number of kidney diseases are being associated with key podocyte abnormalities. The Wilms tumour suppressor gene (WT1) encodes a zinc finger protein with a crucial role in normal kidney development; and in the adult, WT1 is required for normal podocyte function. Denys-Drash Syndrome (DDS) results from mutations affecting the zinc finger domain of WT1. The aim of this study was to undertake, for the first time, a proteomic analysis of cultured human podocytes; and to analyse the molecular changes in DDS podocytes. The morphology of DDS podocytes was highly irregular, reminiscent of a fibroblastic appearance. A reference 2-D gel was generated, and 75 proteins were identified of which 43% involved in cytoskeletal architecture. The DDS and wild-type proteomes were compared by 2-D DIGE. The level of 95.6% of proteins was unaltered; but 4.4% were altered more than two-fold. A sample of proteins involved in cytoskeletal architecture appeared to be misexpressed in DDS podocytes. Consistent with this finding, overall levels of filamentous actin also appeared reduced in DDS podocytes. We conclude that one of WT1 functions in podocytes is to regulate the expression of key components and regulators of the cytoskeleton.
Collapse
Affiliation(s)
- Rebecca L Viney
- Bristol Genomics Research Institute, Centre for Research in Biomedicine, Faculty of Applied Sciences, University of the West of England, Bristol, UK
| | | | | | | | | | | | | |
Collapse
|
42
|
Demasi MAA, Montor WR, Ferreira GB, Pimenta DC, Labriola L, Sogayar MC. Differential proteomic analysis of the anti-proliferative effect of glucocorticoid hormones in ST1 rat glioma cells. J Steroid Biochem Mol Biol 2007; 103:137-48. [PMID: 17127050 DOI: 10.1016/j.jsbmb.2006.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2005] [Accepted: 08/17/2006] [Indexed: 02/02/2023]
Abstract
Glucocorticoid hormones (GCs) exert a potent anti-proliferative activity on several cell types. The classic molecular mechanism of GCs involves modulation of the activity of the glucocorticoids receptor, a transcriptional regulator. However, the anti-proliferative effect of GCs may also involve modulation of processes such as translation, subcellular localization and post-translational modifications, which are not reflected at the mRNA level. To investigate these potential effects of GCs, we employed the proteomic approach (two-dimensional electrophoresis and mass spectrometry) and the ST1 cells, obtained from the C6 rat glioma cell line, as a model. GC treatment leads ST1 cells to a complete transformed-to-normal phenotypic reversion and loss of their tumorigenic potential. By comparing sets of 2D nuclear protein profiles of ST1 cells treated (or not) with hydrocortisone (Hy), 13 polypeptides displaying >or=two-fold difference in abundance upon Hy treatment were found. Five of these polypeptides were identified by peptide mass fingerprinting, including Annexin 2 (ANX2), hnRNP A3 and Ubiquitin. Evidence obtained by Western blot analysis indicates that ANX2 is present in the nucleus and has its subcellular localization modulated by GC-treatment of ST1 cells. Our findings indicate complementary mechanisms contributing to the regulation of gene expression associated with ST1 cells' response to GCs.
Collapse
Affiliation(s)
- Marcos A A Demasi
- Instituto de Química, Universidade de São Paulo, 05508-900 SP, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
Proteomic technologies are used with increasing frequency in the renal community. In this review, we highlight the use in renal research of a number of available techniques including two-dimensional gel electrophoresis, liquid chromatography/mass spectrometry, surface-enhanced laser desorption/ionization, capillary electrophoresis/mass spectrometry, and antibody and tissue arrays. These techniques have been used to identify proteins or changes in proteins specific to regions of the kidney or associated with renal diseases or toxicity. They have also been used to examine protein expression changes and posttranslational modifications of proteins during signaling. A number of studies have used proteomic methodologies to look for diagnostic biomarkers in body fluids. The rapid rate of development of the technologies along with the combination of classic physiological and biochemical techniques with proteomics will enable new discoveries.
Collapse
Affiliation(s)
- Michael G Janech
- Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29425-2220, USA
| | | | | |
Collapse
|
44
|
Xing CY, Saleem MA, Coward RJ, Ni L, Witherden IR, Mathieson PW. Direct effects of dexamethasone on human podocytes. Kidney Int 2006; 70:1038-45. [PMID: 16837924 DOI: 10.1038/sj.ki.5001655] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Glucocorticoids are widely used in the treatment of human glomerular diseases, but their mode of action is poorly understood particularly in steroid-sensitive nephrotic syndrome, which is most common in childhood and is characterized by a lack of inflammation in the kidney. The podocyte is a key cell in the glomerulus in health and disease: until recently, human podocytes have been difficult to study in vitro. We have developed a conditionally immortalized human podocyte cell line transfected with a temperature-sensitive simian virus 40 transgene: when the transgene is inactivated in vitro, these cells adopt the phenotype of differentiated podocytes. We have used these cells to evaluate, using immunocytochemistry, reverse transcriptase-polymerase chain reaction, and Western blotting, direct effects of the glucocorticoid dexamethasone at concentrations designed to mimic in vivo therapeutic corticosteroid levels. Dexamethasone upregulated expression of nephrin and tubulin-alpha, and downregulated vascular endothelial growth factor. Effects on cell cycle were complex with downregulation of cyclin kinase inhibitor p21 and augmentation of podocyte survival, without any effect on apoptosis. We report cytokine production by human podocytes, especially interleukin (IL)-6 and -8; IL-6 expression was suppressed by dexamethasone. These potent direct effects on podocytes illustrate a novel mode of action of glucocorticoids and suggest potential new therapeutic strategies for glomerular disease.
Collapse
Affiliation(s)
- C-Y Xing
- Academic Renal Unit, University of Bristol, Southmead Hospital, Bristol, UK
| | | | | | | | | | | |
Collapse
|
45
|
Yamauchi K, Takano Y, Kasai A, Hayakawa K, Hiramatsu N, Enomoto N, Yao J, Kitamura M. Screening and identification of substances that regulate nephrin gene expression using engineered reporter podocytes. Kidney Int 2006; 70:892-900. [PMID: 16820792 DOI: 10.1038/sj.ki.5001625] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Downregulation of nephrin in podocytes leads to development of proteinuria in human and experimental kidney diseases. However, little is understood about pathophysiologic substances that regulate nephrin expression. In this report, we established conditionally immortalized reporter podocytes REPON for sensitive, continuous monitoring of nephrin gene expression. A murine podocyte cell line harboring a temperature-sensitive simian virus 40 large T antigen was stably transfected with a gene encoding secreted alkaline phosphatase (SEAP) under the control of the 5.4 or 8.3 kb nephrin gene promoter. The established reporter cells REPON5.4 and REPON8.3 were exposed to various pathophysiologic substances, and culture media were subjected to SEAP assay to identify regulators of nephrin gene expression. Among the bioactive substances tested, three physiological ligands of nuclear receptors including all-trans-retinoic acid, 1,25-dihydroxyvitamin D3, and dexamethasone significantly activated the nephrin gene promoter in a dose-dependent manner. These effects were observed in both REPON5.4 and REPON8.3 and were associated with upregulation of nephrin mRNA. The effects of these substances were synergistic, and the maximum effect was observed by combination of three agents. In contrast, inflammatory cytokines interleukin-1beta (IL-1beta) and tumor necrosis factor-alpha as well as phorbol ester significantly downregulated the activity of the nephrin promoter as well as nephrin gene expression. These results elucidated the bidirectional regulation of nephrin by distinct pathophysiologic substances and may provide molecular bases for explaining how proteinuria is induced under pathologic situations and why some ligands for nuclear receptors have the anti-proteinuric potential.
Collapse
Affiliation(s)
- K Yamauchi
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Xing Y, Ding J, Fan Q, Guan N. Diversities of podocyte molecular changes induced by different antiproteinuria drugs. Exp Biol Med (Maywood) 2006; 231:585-93. [PMID: 16636307 DOI: 10.1177/153537020623100513] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Nephrin, podocin, CD2AP, and alpha-actinin-4 are important podocyte proteins that help maintain the integrity of the slit diaphragm and prevent proteinuria. Studies have shown that angiotensin-converting enzyme inhibitors, glucocorticoids, and all-trans retinoic acid (ATRA) have antiproteinuric effects. However, it is still unclear whether these drugs, with different pharmacological mechanisms, lead to a reduction in proteinuria by changing the expression and distribution of these important podocyte proteins. In this study, changes in the expression and distribution of nephrin, podocin, CD2AP, and alpha-actinin-4 were dynamically detected in Adriamycin-induced nephrotic (ADR) rats treated with three different drugs: lisinopril, prednisone, and ATRA. Nephropathy was induced by an intravenous injection of Adriamycin. After Adriamycin injection, rats received lisinopril, prednisone, and ATRA treatment, respectively. Renal tissues were collected at Days 3, 7, 14, and 28. The distribution and the expression of messenger RNA and protein of nephrin, podocin, CD2AP, and alpha-actinin-4 were detected by indirect immunofluorescence, real-time polymerase chain reaction, and Western blotting, respectively. With the intervention of lisinopril, prednisone, and ATRA, changes in the expression of nephrin, podocin, and CD2AP were diverse, which was different from that detected in ADR rats. After lisinopril and prednisone intervention, podocin exhibited prominent earlier changes compared with those of nephrin and CD2AP, whereas CD2AP showed more prominent changes after ATRA intervention. There was no change in the expression of alpha-actinin-4 molecule. In summary, we conclude that the antiproteinuric effects of lisinopril, prednisone, and ATRA were achieved by changes in the expression and distribution of the important podocyte molecules nephrin, podocin, CD2AP, and alpha-actinin-4. The pattern in the change of podocyte molecules after lisinopril and prednisone intervention was similar, but the pattern in the change of podocyte molecules after ATRA intervention was different from that of lisinopril or prednisone intervention.
Collapse
Affiliation(s)
- Yan Xing
- Department of Pediatrics, Peking University First Hospital, Beijing, People's Republic of China
| | | | | | | |
Collapse
|
47
|
Abstract
The terminally differentiated podocyte, also called glomerular visceral epithelial cell, are highly specialized cells. They function as a critical size and charge barrier to prevent proteinuria. Podocytes are injured in diabetic and non-diabetic renal diseases. The clinical signature of podocyte injury is proteinuria, with or without loss of renal function owing to glomerulosclerosis. There is an exciting and expanding literature showing that hereditary, congenital, or acquired abnormalities in the molecular anatomy of podocytes leads to proteinuria, and at times, glomerulosclerosis. The change in podocyte shape, called effacement, is not simply a passive process following injury, but is owing to a complex interplay of proteins that comprise the molecular anatomy of the different protein domains of podocytes. These will be discussed in this review. Recent studies have also highlighted that a reduction in podocyte number directly causes proteinuria and glomerulosclerosis. This is owing to several factors, including the relative inability for these cells to proliferate, detachment, and apoptosis. The mechanisms of these events are being elucidated, and are discussed in this review. It is the hope that by delineating the events following injury to podocytes, therapies might be developed to reduce the burden of proteinuric renal diseases.
Collapse
Affiliation(s)
- S J Shankland
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, Washington 98195, USA.
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Podocyte injury is a central event in the development of glomerulosclerosis. This review highlights contributions from the past year to our understanding of mechanisms of podocyte injury and implications for potential treatment strategies of glomerular disease. RECENT FINDINGS Rearrangement of the actin cytoskeleton, the backbone linking the slit diaphragm, apical domain and sole plate, serves as a common denominator during foot process effacement. Reports on the role of synaptopodin and CDK5 on actin dynamics as well as cathepsin L and B7.1 in subsequent cell migration have expanded our understanding of the podocyte response to injury. Mounting evidence supports an expanding role of the slit diaphragm in signal transduction to mediate downstream cellular responses, including prosurvival effects of the integral proteins nephrin and CD2AP. The discovery that TRPC6 localizes to the slit diaphragm and identification of specific mutations of the transport channel in kindreds of familial focal segmental glomerulosclerosis implicate a causal role for aberrant calcium signaling in podocyte injury. Disruption of the dystroglycan complex, which anchors the podocyte to the underlying basement membrane, in states of foot process effacement may have implications for the recent finding of viable podocytes in the urine in glomerular disease. SUMMARY The resurgence of research in podocyte biology over the past decade underscores the importance of this unique cell in preserving glomerular structure and function. A greater understanding of the complex signaling mechanisms governing podocyte biology in health and disease will ultimately lead to novel therapeutic avenues for treating disorders of the podocyte.
Collapse
Affiliation(s)
- Raghu V Durvasula
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Box 356521, Seattle, WA 98195, USA.
| | | |
Collapse
|
49
|
Endlich N, Endlich K. Stretch, tension and adhesion – Adaptive mechanisms of the actin cytoskeleton in podocytes. Eur J Cell Biol 2006; 85:229-34. [PMID: 16546566 DOI: 10.1016/j.ejcb.2005.09.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Podocytes form an epithelial layer on the outer aspect of the basement membrane of glomerular capillaries. The interdigitating pattern of podocyte foot processes (PFPs) generates a unique and extremely long cell-cell contact area - the filtration slit. Thus, the interdigitating PFPs are the morphological basis for the high hydraulic conductivity of the glomerular capillaries. Any disturbance in this interdigitating pattern results in a drop of glomerular filtration rate impairing renal function. PFPs are based on the actin cytoskeleton, consisting of a subplasmalemmal network and a central core of filament bundles. Besides giving PFPs their morphology, the actin cytoskeleton anchors cell-cell contact and cell-matrix proteins in podocytes. Several human genetic diseases as well as transgenic mouse models provide evidence for the crucial role of the actin cytoskeleton in podocytes. Varying flow rates of the filtrate, increased glomerular capillary pressure in glomerular hypertension, and varying activation states of contractile proteins in PFPs impose a mechanical load on the actin cytoskeleton, challenging the intricate arrangement of PFPs and podocyte adhesion. Here we review data about the actin cytoskeleton of podocytes and the response of podocytes to mechanical load. From these data possible mechanisms are emerging how the actin cytoskeleton may allow podocytes to adapt to states of increased mechanical load.
Collapse
Affiliation(s)
- Nicole Endlich
- Department of Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|
50
|
Current Awareness on Comparative and Functional Genomics. Comp Funct Genomics 2005. [PMCID: PMC2447491 DOI: 10.1002/cfg.425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|