1
|
Zhang H, Lin J, Lu H, Zhong Y, Deng L, Kuang B, Li Q. Systemic lupus erythematosus with podocyte infolding glomerulopathy: A case report and literature review. Medicine (Baltimore) 2024; 103:e39809. [PMID: 39470480 PMCID: PMC11521074 DOI: 10.1097/md.0000000000039809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/02/2024] [Indexed: 10/30/2024] Open
Abstract
RATIONALE Podocyte infolding glomerulopathy (PIG) is a rare glomerular disease, its diagnosis mainly depends on pathological manifestations of the kidney. Few clinical cases of PIG have been reported, but it is sometimes associated with connective tissue diseases. Here we describe a case of systemic lupus erythematosus (SLE) with PIG and undertake a review of the literature. PATIENT CONCERNS A 34-year-old female patient was admitted to our hospital in August 2019 with repeated facial erythema and proteinuria for more than 10 years. The patient was previously diagnosed with SLE. DIAGNOSIS Systemic lupus erythematosus. INTERVENTIONS Renal biopsy was performed to investigate ongoing proteinuria and the results were consistent with PIG. Treatment with methylprednisolone, hydroxychloroquine sulfate, mycophenolate mofetil, and candesartan ester. OUTCOMES Improved the patient's condition and resolved the proteinuria. LESSONS This study reported a case of PIG and SLE. The patient was diagnosed according to biopsy, and the disease remain stable after immunosuppressive therapy. It is recommended to carefully study renal biopsies from patients with proteinuria and underlying autoimmune diseases to identify additional cases.
Collapse
Affiliation(s)
- Huiqing Zhang
- Guangzhou University of Chinese Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong 523000, China
| | - Jie Lin
- Guangzhou University of Chinese Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong 523000, China
| | - Hanqi Lu
- Guangzhou University of Chinese Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong 523000, China
| | - Yunliang Zhong
- Guangzhou University of Chinese Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong 523000, China
| | - Lie Deng
- Guangzhou University of Chinese Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong 523000, China
| | - Bin Kuang
- Guangzhou University of Chinese Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong 523000, China
| | - Qiang Li
- Guangzhou University of Chinese Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong 523000, China
| |
Collapse
|
2
|
Schanz M, Kimmel M, Alscher MD, Amann K, Daniel C. TIMP-2 and IGFBP7 in human kidney biopsies in renal disease. Clin Kidney J 2023; 16:1434-1446. [PMID: 37664566 PMCID: PMC10468751 DOI: 10.1093/ckj/sfad010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Indexed: 09/05/2023] Open
Abstract
Background Tissue inhibitor of matrix metalloproteinase-2 (TIMP-2) and insulin-like growth factor binding protein 7 (IGFBP7) are markers of tubular stress and urinary [TIMP-2]*[IGFBP7] is an established biomarker for risk assessment of acute kidney injury. There are no studies of expression profiles or localization of these markers in human renal tissue with confirmed renal disease. Methods We analysed 37 kidney biopsies of patients with renal disease and 10 non-diseased control biopsies for TIMP-2 and IGFBP7 expression using immunohistochemistry. Changes in glomerular morphology were evaluated by a semi-quantitative glomerulosclerosis score (GSI) and tubular interstitial changes were graded by the tubular injury score (TSI) using periodic acid-Schiff-stained paraffin sections. Interstitial fibrosis and tubular atrophy (IF/TA) were graded according to the Banff classification. Urinary [TIMP-2]*[IGFBP7] was collected at the time of biopsy. Results TIMP-2 and IGFBP7 had significantly greater expression in kidney biopsies from patients with renal disease compared with control tissue, especially in the tubular compartment. Here, IGFBP7 was detected in proximal and distal tubules while TIMP-2 was predominantly localized in the collecting ducts. Renal injury significantly correlated with staining intensity for TIMP-2 and IGFBP7: GSI weakly correlated with glomerular TIMP-2 (r = 0.36) and IGFBP7 (r = 0.35) and TSI correlated with tubular TIMP-2 (r = 0.41) and IGFBP7 (r = 0.43). Urinary [TIMP-2]*[IGFBP7] correlated weakly with the histopathological damage score but not with glomerular and tubular expression. Conclusion Our findings underline the role of TIMP-2/IGFBP7 as an unspecific marker of renal injury that is already in use for early detection of acute kidney injury.
Collapse
Affiliation(s)
- Moritz Schanz
- Department of Internal Medicine, Division of General Internal Medicine and Nephrology, Robert-Bosch Hospital Stuttgart, Germany
| | - Martin Kimmel
- Department of Internal Medicine, Division of Nephrology, Hypertension and Autoimmune Disorders, Alb-Fils Kliniken, Göppingen, Germany
| | - Mark Dominik Alscher
- Department of Internal Medicine, Division of General Internal Medicine and Nephrology, Robert-Bosch Hospital Stuttgart, Germany
| | - Kerstin Amann
- Department of Nephropathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
3
|
Juin SK, Ouseph R, Gondim DD, Jala VR, Sen U. Diabetic Nephropathy and Gaseous Modulators. Antioxidants (Basel) 2023; 12:antiox12051088. [PMID: 37237955 DOI: 10.3390/antiox12051088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Diabetic nephropathy (DN) remains the leading cause of vascular morbidity and mortality in diabetes patients. Despite the progress in understanding the diabetic disease process and advanced management of nephropathy, a number of patients still progress to end-stage renal disease (ESRD). The underlying mechanism still needs to be clarified. Gaseous signaling molecules, so-called gasotransmitters, such as nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), have been shown to play an essential role in the development, progression, and ramification of DN depending on their availability and physiological actions. Although the studies on gasotransmitter regulations of DN are still emerging, the evidence revealed an aberrant level of gasotransmitters in patients with diabetes. In studies, different gasotransmitter donors have been implicated in ameliorating diabetic renal dysfunction. In this perspective, we summarized an overview of the recent advances in the physiological relevance of the gaseous molecules and their multifaceted interaction with other potential factors, such as extracellular matrix (ECM), in the severity modulation of DN. Moreover, the perspective of the present review highlights the possible therapeutic interventions of gasotransmitters in ameliorating this dreaded disease.
Collapse
Affiliation(s)
- Subir Kumar Juin
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Microbiology & Immunology, Brown Cancer Center, Center for Microbiomics, Inflammation and Pathogenicity, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Rosemary Ouseph
- Division of Nephrology & Hypertension, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Dibson Dibe Gondim
- Department of Pathology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Venkatakrishna Rao Jala
- Department of Microbiology & Immunology, Brown Cancer Center, Center for Microbiomics, Inflammation and Pathogenicity, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
4
|
Tanaka M, Moniwa N, Nogi C, Kano T, Matsumoto M, Sakai A, Maeda T, Takizawa H, Ogawa Y, Asanuma K, Suzuki Y, Furuhashi M. Glomerular expression and urinary excretion of fatty acid-binding protein 4 in IgA nephropathy. J Nephrol 2023; 36:385-395. [PMID: 36622635 DOI: 10.1007/s40620-022-01551-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/03/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND Fatty acid-binding protein 4 (FABP4) is secreted from adipocytes and macrophages in adipose tissue and acts as an adipokine. It has recently been reported that FABP4, but not liver-type FABP (L-FABP/FABP1), is also expressed in injured glomerular endothelial cells and infiltrating macrophages in the glomerulus and that urinary FABP4 (U-FABP4) is associated with proteinuria and kidney function impairment in nephrotic patients. However, the link between glomerular FABP4 and U-FABP4 has not been fully addressed in IgA nephropathy (IgAN). METHODS We investigated the involvement of FABP4 in human and mouse IgAN. RESULTS In patients with IgAN (n = 23), the ratio of FABP4-positive area to total area within glomeruli (G-FABP4-Area) and U-FABP4 were positively correlated with proteinuria and were negatively correlated with eGFR. In 4-28-week-old male grouped ddY mice, a spontaneous IgAN-prone mouse model, FABP4 was detected in glomerular endothelial cells and macrophages, and G-FABP4-Area was positively correlated with urinary albumin-to-creatinine ratio (r = 0.957, P < 0.001). Endoplasmic reticulum stress markers were detected in glomeruli of human and mouse IgAN. In human renal glomerular endothelial cells, FABP4 was induced by treatment with vascular endothelial growth factor and was secreted from the cells. Treatment of human renal glomerular endothelial cells or mouse podocytes with palmitate-bound recombinant FABP4 significantly increased gene expression of inflammatory cytokines and endoplasmic reticulum stress markers, and the effects of FABP4 in podocytes were attenuated in the presence of an anti-FABP4 antibody. CONCLUSION FABP4 in the glomerulus contributes to proteinuria in IgAN, and U-FABP4 level is a useful surrogate biomarker for glomerular damage in IgAN.
Collapse
Affiliation(s)
- Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-Ku, Sapporo, 060-8543, Japan
| | - Norihito Moniwa
- Department of Nephrology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Chieko Nogi
- Division of Nephrology, Department of Internal Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Toshiki Kano
- Division of Nephrology, Department of Internal Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Megumi Matsumoto
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-Ku, Sapporo, 060-8543, Japan
| | - Akiko Sakai
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-Ku, Sapporo, 060-8543, Japan
| | - Takuto Maeda
- Department of Nephrology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Hideki Takizawa
- Department of Nephrology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Yayoi Ogawa
- Hokkaido Renal Pathology Center, Sapporo, Japan
| | | | - Yusuke Suzuki
- Division of Nephrology, Department of Internal Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-Ku, Sapporo, 060-8543, Japan.
| |
Collapse
|
5
|
Matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases in kidney disease. Adv Clin Chem 2021; 105:141-212. [PMID: 34809827 DOI: 10.1016/bs.acc.2021.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Matrix metalloproteinases (MMPs) are a group of zinc and calcium endopeptidases which cleave extracellular matrix (ECM) proteins. They are also involved in the degradation of cell surface components and regulate multiple cellular processes, cell to cell interactions, cell proliferation, and cell signaling pathways. MMPs function in close interaction with the endogenous tissue inhibitors of matrix metalloproteinases (TIMPs), both of which regulate cell turnover, modulate various growth factors, and participate in the progression of tissue fibrosis and apoptosis. The multiple roles of MMPs and TIMPs are continuously elucidated in kidney development and repair, as well as in a number of kidney diseases. This chapter focuses on the current findings of the significance of MMPs and TIMPs in a wide range of kidney diseases, whether they result from kidney tissue changes, hemodynamic alterations, tubular epithelial cell apoptosis, inflammation, or fibrosis. In addition, the potential use of these endopeptidases as biomarkers of renal dysfunction and as targets for therapeutic interventions to attenuate kidney disease are also explored in this review.
Collapse
|
6
|
Yang X, Jiang W, Huang M, Dai Y, Li B, Wang X, Yu Y, Shen T, Wu C, Zhu Q. Intracellular complement activation in podocytes aggravates immune kidney injury in trichloroethylene-sensitized mice. J Toxicol Sci 2020; 45:681-693. [PMID: 33132242 DOI: 10.2131/jts.45.681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Trichloroethylene (TCE) as a common organic solvent in industrial production can cause occupational medicamentosa-like dermatitis (OMDT) in some exposed workers. In addition to systemic skin damage, OMDT is also accompanied by severe kidney injury. Our previous studies show that complement (C) plays an important role in immune kidney injury caused by TCE. Specifically, C3 is mainly deposited on glomeruli. Recent studies have found that intracellular complement can be activated by cathepsin L (CTSL) and exert a series of biological effects. The purpose of this study was to explore where C3 on glomeruli comes from and what role it plays. A BALB/c mouse model of skin sensitization induced by TCE in the presence or absence of CTSL inhibitor (CTSLi,10 mg/kg). In TCE sensitization-positive mice, C3 was mainly expressed on podocytes and the expression of CTSL significantly increased in podocytes. Kidney function test and related indicators showed abnormal glomerular filtration and transmission electron microscopy revealed ultrastructure damage to podocytes. These lesions were alleviated in TCE/CTSLi positive mice. These results provide the first evidence that in TCE-induced immune kidney injury, intracellular complement in podocytes can be over-activated by CTSL and aggravates podocytes injury, thereby damaging glomerular filtration function. Intracellular complement activation and cathepsin L in podocytes may be a potential target for treating immune kidney injury induced by TCE.
Collapse
Affiliation(s)
- Xiaodong Yang
- Anhui Cancer Institute, The First Affiliated Hospital of Anhui Medical University, China.,Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, China
| | - Wei Jiang
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, China
| | - Meng Huang
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, China
| | - Yuying Dai
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, China
| | - Bodong Li
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, China
| | - Xian Wang
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, China
| | - Yun Yu
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, China
| | - Tong Shen
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, China
| | - Changhao Wu
- School of Biosciences and Medicine, FHMS, University of Surrey, UK
| | - Qixing Zhu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, China.,Key Laboratory of Dermatology, Ministry of Education, China
| |
Collapse
|
7
|
Genetic Susceptibility and Protein Expression of Extracellular Matrix Turnover-Related Genes in Oral Submucous Fibrosis. Int J Mol Sci 2020; 21:ijms21218104. [PMID: 33143101 PMCID: PMC7663238 DOI: 10.3390/ijms21218104] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
Betel quid (BQ) chewing increased the risk of oral cancer and oral submucous fibrosis (OSMF), an oral premalignant disorder (OPMD) with malignant transformation potential. BQ components such as areca nut (AN), trauma by coarse AN fiber, catechin, copper, alkaloids, stimulated reactive oxygen species (ROS), inflammation and cytotoxicity are suggested to be the contributing factors. They may induce tissue inflammation, proliferation of fibroblasts and collagen deposition, myofibroblast differentiation and contraction, collagen cross-links and inhibit collagen phagocytosis, finally leading to the development of OSMF and oral cancer. These events are mediated by BQ components-induced changes of extracellular matrix (ECM) turnover via regulation of TGF-β1, plasminogen activator inhibitor-1 (PAI-1), cystatin, lysyl oxidase (LOX) and tissue inhibitors of metalloproteinases (TIMPs) and metalloproteinases (MMPs). Genetic susceptibility is also involved in these disease processes. Further understanding the molecular mechanisms of BQ-induced OSMF and oral cancer can be helpful for future disease prevention and treatment.
Collapse
|
8
|
Kwiatkowska E, Stefańska K, Zieliński M, Sakowska J, Jankowiak M, Trzonkowski P, Marek-Trzonkowska N, Kwiatkowski S. Podocytes-The Most Vulnerable Renal Cells in Preeclampsia. Int J Mol Sci 2020; 21:ijms21145051. [PMID: 32708979 PMCID: PMC7403979 DOI: 10.3390/ijms21145051] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 12/24/2022] Open
Abstract
Preeclampsia (PE) is a disorder that affects 3–5% of normal pregnancies. It was believed for a long time that the kidney, similarly to all vessels in the whole system, only sustained endothelial damage. The current knowledge gives rise to a presumption that the main role in the development of proteinuria is played by damage to the podocytes and their slit diaphragm. The podocyte damage mechanism in preeclampsia is connected to free VEGF and nitric oxide (NO) deficiency, and an increased concentration of endothelin-1 and oxidative stress. From national cohort studies, we know that women who had preeclampsia in at least one pregnancy carried five times the risk of developing end-stage renal disease (ESRD) when compared to women with physiological pregnancies. The focal segmental glomerulosclerosis (FSGS) is the dominant histopathological lesion in women with a history of PE. The kidney’s podocytes are not subject to replacement or proliferation. Podocyte depletion exceeding 20% resulted in FSGS, which is a reason for the later development of ESRD. In this review, we present the mechanism of kidney (especially podocytes) injury in preeclampsia. We try to explain how this damage affects further changes in the morphology and function of the kidneys after pregnancy.
Collapse
Affiliation(s)
- Ewa Kwiatkowska
- Clinical Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Katarzyna Stefańska
- Department of Obstetrics, Medical University of Gdańsk, 80-210 Gdańsk, Poland
- Correspondence:
| | - Maciej Zieliński
- Department of Medical Immunology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (M.Z.); (J.S.); (M.J.); (P.T.)
| | - Justyna Sakowska
- Department of Medical Immunology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (M.Z.); (J.S.); (M.J.); (P.T.)
| | - Martyna Jankowiak
- Department of Medical Immunology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (M.Z.); (J.S.); (M.J.); (P.T.)
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (M.Z.); (J.S.); (M.J.); (P.T.)
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science Cancer Immunology Group, University of Gdansk, 80-822 Gdańsk, Poland;
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Sebastian Kwiatkowski
- Department of Obstetrics and Gynecology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| |
Collapse
|
9
|
Keisuke S, Kohei M, Takuji E, Tomoki M, Yuichi M, Rina O, Tsukasa T, Mitsuru O. Role of cathepsin L in idiopathic nephrotic syndrome in children. Med Hypotheses 2020; 141:109718. [PMID: 32289645 DOI: 10.1016/j.mehy.2020.109718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 04/06/2020] [Indexed: 01/12/2023]
Abstract
Nephrotic syndrome (NS) is one of the most common glomerular diseases in children. Glomerular podocyte dysfunction can result in proteinuria, the presence of a large amount of protein in the urine. Podocytes are unique epithelial cells that divide into 3 separate structural and functional segments: a cell body, major processes, and foot processes. Since synaptopodin, dynamin, and actin are crucial components of the podocyte cytoskeleton, degradation of these proteins is associated with cytoskeleton instability, resulting in the development of proteinuria. Cathepsin L (CatL), a cysteine proteinase, plays a crucial role in various renal diseases. CatL expression is elevated in rats with puromycin aminonucleoside-induced nephropathy, which is used as a model of minimal change NS. In CatL-deficient mice, which do not develop proteinuria, dynamin is retained through the escape of CatL-mediated decomposition, resulting in no changes in the filtration barrier of podocytes. However, there is limited information on the roles of CatL in NS. Based on these data, CatL might play an important role in the development of proteinuria. Furthermore, identifying the functions of CatL may contribute to a better understanding of the pathogenesis of childhood-onset NS. We hypothesize that high levels of CatL can lead to cytoskeletal instability of podocytes, resulting in proteinuria in childhood-onset NS.
Collapse
Affiliation(s)
- Sugimoto Keisuke
- Department of Pediatrics, Kindai University, Faculty of Medicine, Osaka, Japan.
| | - Miyazaki Kohei
- Department of Pediatrics, Kindai University, Faculty of Medicine, Osaka, Japan
| | - Enya Takuji
- Department of Pediatrics, Kindai University, Faculty of Medicine, Osaka, Japan
| | - Miyazawa Tomoki
- Department of Pediatrics, Kindai University, Faculty of Medicine, Osaka, Japan
| | - Morimoto Yuichi
- Department of Pediatrics, Kindai University, Faculty of Medicine, Osaka, Japan
| | - Oshima Rina
- Department of Pediatrics, Kindai University, Faculty of Medicine, Osaka, Japan
| | - Takemura Tsukasa
- Department of Pediatrics, Kushimoto Municipality Hospital, Wakayama, Japan
| | - Okada Mitsuru
- Department of Pediatrics, Kindai University, Faculty of Medicine, Osaka, Japan
| |
Collapse
|
10
|
Fuseya S, Suzuki R, Okada R, Hagiwara K, Sato T, Narimatsu H, Yokoi H, Kasahara M, Usui T, Morito N, Yamagata K, Kudo T, Takahashi S. Mice lacking core 1-derived O-glycan in podocytes develop transient proteinuria, resulting in focal segmental glomerulosclerosis. Biochem Biophys Res Commun 2020; 523:1007-1013. [PMID: 31973821 DOI: 10.1016/j.bbrc.2020.01.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/05/2020] [Indexed: 12/25/2022]
Abstract
The glomerular filtration barrier is composed of podocytes, glomerular basement membrane, and endothelial cells. Disruption of these structures causes several glomerular injuries, such as focal segmental glomerulosclerosis (FSGS). The surface of podocyte apical membranes is coated by negatively charged sialic acids on core 1-derived mucin-type O-glycans. Here, we aimed to investigate the physiological role of core 1-derived O-glycans in the podocytes using adult mice lacking podocyte-specific core 1-derived O-glycans (iPod-Cos). iPod-Cos mice exhibited early and transient proteinuria with foot process effacements and developed typical FSGS-like disease symptoms. To identify the key molecules responsible for the FSGS-like phenotype, we focused on podocalyxin and podoplanin, which possess mucin-type O-glycans. Expression and localization of podocalyxin did not change in iPod-Cos glomeruli. Besides, western blot analysis revealed significantly lower levels of intact podocalyxin in isolated glomeruli of iPod-Cos mice, and high levels of processed forms in iPod-Cos glomeruli, as compared to that in control glomeruli. Conversely, podoplanin mRNA, and protein levels were lower in iPod-Cos mice than in control mice. These results demonstrated that core 1-derived O-glycan on podocytes is required for normal glomerular filtration and may contribute to the stable expression of podocalyxin and podoplanin.
Collapse
Affiliation(s)
- Sayaka Fuseya
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Japan; Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Japan
| | - Riku Suzuki
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Japan; Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Japan
| | - Risa Okada
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Japan
| | - Kozue Hagiwara
- Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Japan
| | - Takashi Sato
- Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Japan
| | - Hisashi Narimatsu
- Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Japan
| | - Hideki Yokoi
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Japan
| | - Masato Kasahara
- Department of Clinical Research, Nara Medical University Hospital, Japan
| | - Toshiaki Usui
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Japan; Department of Nephrology, Faculty of Medicine, University of Tsukuba, Japan
| | - Naoki Morito
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, Japan
| | - Kunihiro Yamagata
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, Japan
| | - Takashi Kudo
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Japan.
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Japan.
| |
Collapse
|
11
|
Papakrivopoulou E, Vasilopoulou E, Lindenmeyer MT, Pacheco S, Brzóska HŁ, Price KL, Kolatsi‐Joannou M, White KE, Henderson DJ, Dean CH, Cohen CD, Salama AD, Woolf AS, Long DA. Vangl2, a planar cell polarity molecule, is implicated in irreversible and reversible kidney glomerular injury. J Pathol 2018; 246:485-496. [PMID: 30125361 PMCID: PMC6282744 DOI: 10.1002/path.5158] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 08/02/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022]
Abstract
Planar cell polarity (PCP) pathways control the orientation and alignment of epithelial cells within tissues. Van Gogh-like 2 (Vangl2) is a key PCP protein that is required for the normal differentiation of kidney glomeruli and tubules. Vangl2 has also been implicated in modifying the course of acquired glomerular disease, and here, we further explored how Vangl2 impacts on glomerular pathobiology in this context. Targeted genetic deletion of Vangl2 in mouse glomerular epithelial podocytes enhanced the severity of not only irreversible accelerated nephrotoxic nephritis but also lipopolysaccharide-induced reversible glomerular damage. In each proteinuric model, genetic deletion of Vangl2 in podocytes was associated with an increased ratio of active-MMP9 to inactive MMP9, an enzyme involved in tissue remodelling. In addition, by interrogating microarray data from two cohorts of renal patients, we report increased VANGL2 transcript levels in the glomeruli of individuals with focal segmental glomerulosclerosis, suggesting that the molecule may also be involved in certain human glomerular diseases. These observations support the conclusion that Vangl2 modulates glomerular injury, at least in part by acting as a brake on MMP9, a potentially harmful endogenous enzyme. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
MESH Headings
- Adult
- Animals
- Case-Control Studies
- Cell Polarity
- Cells, Cultured
- Disease Models, Animal
- Enzyme Activation
- Female
- Glomerulosclerosis, Focal Segmental/genetics
- Glomerulosclerosis, Focal Segmental/metabolism
- Glomerulosclerosis, Focal Segmental/pathology
- Glomerulosclerosis, Focal Segmental/physiopathology
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Kidney Glomerulus/metabolism
- Kidney Glomerulus/pathology
- Kidney Glomerulus/physiopathology
- Male
- Matrix Metalloproteinase 9/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Middle Aged
- Nephrosis, Lipoid/genetics
- Nephrosis, Lipoid/metabolism
- Nephrosis, Lipoid/pathology
- Nephrosis, Lipoid/physiopathology
- Nerve Tissue Proteins/deficiency
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Podocytes/metabolism
- Podocytes/pathology
- Signal Transduction
- Young Adult
Collapse
Affiliation(s)
- Eugenia Papakrivopoulou
- Developmental Biology and Cancer ProgrammeUCL Great Ormond Street Institute of Child HealthLondonUK
| | - Elisavet Vasilopoulou
- Developmental Biology and Cancer ProgrammeUCL Great Ormond Street Institute of Child HealthLondonUK
- Medway School of PharmacyUniversity of KentChatham MaritimeUK
| | - Maja T Lindenmeyer
- Nephrological Center, Medical Clinic and Policlinic IVUniversity of MunichMunichGermany
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Sabrina Pacheco
- Developmental Biology and Cancer ProgrammeUCL Great Ormond Street Institute of Child HealthLondonUK
| | - Hortensja Ł Brzóska
- Developmental Biology and Cancer ProgrammeUCL Great Ormond Street Institute of Child HealthLondonUK
| | - Karen L Price
- Developmental Biology and Cancer ProgrammeUCL Great Ormond Street Institute of Child HealthLondonUK
| | - Maria Kolatsi‐Joannou
- Developmental Biology and Cancer ProgrammeUCL Great Ormond Street Institute of Child HealthLondonUK
| | - Kathryn E White
- Electron Microscopy Research ServicesNewcastle UniversityNewcastle upon TyneUK
| | - Deborah J Henderson
- Cardiovascular Research CentreInstitute of Genetic Medicine, Newcastle UniversityNewcastle upon TyneUK
| | - Charlotte H Dean
- Inflammation Repair and Development SectionNational Heart and Lung Institute, Imperial College LondonLondonUK
| | - Clemens D Cohen
- Nephrological Center, Medical Clinic and Policlinic IVUniversity of MunichMunichGermany
| | - Alan D Salama
- University College London Centre for Nephrology, Royal Free HospitalLondonUK
| | - Adrian S Woolf
- Faculty of Biology Medicine and HealthSchool of Biological Sciences, University of ManchesterManchesterUK
- Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science CentreManchesterUK
| | - David A Long
- Developmental Biology and Cancer ProgrammeUCL Great Ormond Street Institute of Child HealthLondonUK
| |
Collapse
|
12
|
Kubo A, Shirato I, Hidaka T, Takagi M, Sasaki Y, Asanuma K, Ishidoh K, Suzuki Y. Expression of Cathepsin L and Its Intrinsic Inhibitors in Glomeruli of Rats With Puromycin Aminonucleoside Nephrosis. J Histochem Cytochem 2018; 66:863-877. [PMID: 30052474 DOI: 10.1369/0022155418791822] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cathepsin L, a lysosomal cysteine proteinase, may have a key role in various biological and disease processes by intracellular and extracellular degradation of proteins. We examined the levels of cathepsin L and its intrinsic inhibitors in glomeruli of rats with puromycin aminonucleoside (PAN) nephrosis. In contrast to the weak levels of cathepsin L in normal glomeruli, on days 4 and 8, strong immunostaining was detected in almost all podocytes when proteinuria and pathological changes of the podocytes developed. Cathepsin L was reduced after day 28, but remained in a focal and segmental manner. Cystatin β, an intracellular inhibitor, was not detected in podocytes. However, cystatin C, an extracellular inhibitor, was detected in podocytes after day 4, coincident with cathepsin L. Cystatin C levels were gradually reduced but sustained in many podocytes on day 28, while cystatin C was not detected in podocytes sustained cathepsin L. These results demonstrated that cathepsin L levels are not always accompanied by the levels of its inhibitors in podocytes of PAN nephrosis, suggesting a potential role of cathepsin L in podocyte injury, which is a critical process for the development and progression of tuft adhesion and sclerosis.
Collapse
Affiliation(s)
- Ayano Kubo
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | | | - Teruo Hidaka
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Miyuki Takagi
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Yu Sasaki
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Katsuhiko Asanuma
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazumi Ishidoh
- Division of Molecular Biology, Institute for Health Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
13
|
Heparanase: roles in cell survival, extracellular matrix remodelling and the development of kidney disease. Nat Rev Nephrol 2017; 13:201-212. [PMID: 28163306 DOI: 10.1038/nrneph.2017.6] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heparanase has regulatory roles in various processes, including cell communication, gene transcription and autophagy. In addition, it is the only known mammalian endoglycosidase that is capable of degrading heparan sulfate (HS). HS chains are important constituents and organizers of the extracellular matrix (ECM), and have a key role in maintaining the integrity and function of the glomerular filtration barrier. In addition, HS chains regulate the activity of numerous bioactive molecules, such as cytokines and growth factors, at the cell surface and in the ECM. Given the functional diversity of HS, its degradation by heparanase profoundly affects important pathophysiological processes, including tumour development, neovascularization and inflammation, as well as progression of kidney disease. Heparanase-mediated degradation and subsequent remodelling of HS in the ECM of the glomerulus is a key mechanism in the development of glomerular disease, as exemplified by the complete resistance of heparanase-deficient animals to diabetes and immune-mediated kidney disease. This Review summarizes the role of heparanase in the development of kidney disease, and its potential as a therapeutic target.
Collapse
|
14
|
Emlet DR, Pastor-Soler N, Marciszyn A, Wen X, Gomez H, Humphries WH, Morrisroe S, Volpe JK, Kellum JA. Insulin-like growth factor binding protein 7 and tissue inhibitor of metalloproteinases-2: differential expression and secretion in human kidney tubule cells. Am J Physiol Renal Physiol 2016; 312:F284-F296. [PMID: 28003188 DOI: 10.1152/ajprenal.00271.2016] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 11/11/2016] [Accepted: 11/28/2016] [Indexed: 01/04/2023] Open
Abstract
We have characterized the expression and secretion of the acute kidney injury (AKI) biomarkers insulin-like growth factor binding protein 7 (IGFBP7) and tissue inhibitor of metalloproteinases-2 (TIMP-2) in human kidney epithelial cells in primary cell culture and tissue. We established cell culture model systems of primary kidney cells of proximal and distal tubule origin and observed that both proteins are indeed expressed and secreted in both tubule cell types in vitro. However, TIMP-2 is both expressed and secreted preferentially by cells of distal tubule origin, while IGFBP7 is equally expressed across tubule cell types yet preferentially secreted by cells of proximal tubule origin. In human kidney tissue, strong staining of IGFBP7 was seen in the luminal brush-border region of a subset of proximal tubule cells, and TIMP-2 stained intracellularly in distal tubules. Additionally, while some tubular colocalization of both biomarkers was identified with the injury markers kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin, both biomarkers could also be seen alone, suggesting the possibility for differential mechanistic and/or temporal profiles of regulation of these early AKI biomarkers from known markers of injury. Last, an in vitro model of ischemia-reperfusion demonstrated enhancement of secretion of both markers early after reperfusion. This work provides a rationale for further investigation of these markers for their potential role in the pathogenesis of acute kidney injury.
Collapse
Affiliation(s)
- David R Emlet
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nuria Pastor-Soler
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Allison Marciszyn
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Xiaoyan Wen
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hernando Gomez
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Seth Morrisroe
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jacob K Volpe
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; .,Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW In this review, we take a combined membrane biologist's and geneticist's view of the podocyte, to examine how genetics have informed our understanding of membrane receptors, channels, and other signaling molecules affecting podocyte health and disease. RECENT FINDINGS An integral part of the kidney, the glomerulus, is responsible for the kidney's filter function. Within the glomerulus, the podocyte is a unique cell serving a critically important role: it is exposed to signals from the urinary space in Bowman's capsule, it receives and transmits signals to/from the basement membrane upon which it elaborates, and it receives signals from the vascular space with which it also communicates, thus exposed to toxins, viruses, chemicals, proteins, and cellular components or debris that flow in the blood stream. Our understanding of how podocytes perform their important role has been largely informed by human genetics, and the recent revolution afforded by exome sequencing has brought a tremendous wealth of new genetic data to light. SUMMARY Genetically defined, rare/orphan podocytopathies, as reviewed here, are critically important to study as they may reveal the next generation targets for precision medicine in nephrology.
Collapse
|
16
|
Bhattacharjee N, Barma S, Konwar N, Dewanjee S, Manna P. Mechanistic insight of diabetic nephropathy and its pharmacotherapeutic targets: An update. Eur J Pharmacol 2016; 791:8-24. [PMID: 27568833 DOI: 10.1016/j.ejphar.2016.08.022] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/03/2016] [Accepted: 08/24/2016] [Indexed: 02/09/2023]
Abstract
Diabetic nephropathy (DN), a chronic complication of diabetes, is charecterized by glomerular hypertrophy, proteinuria, decreased glomerular filtration, and renal fibrosis resulting in the loss of renal function. Although the exact cause of DN remains unclear, several mechanisms have been postulated, such as hyperglycemia-induced renal hyper filtration and renal injury, AGEs-induced increased oxidative stress, activated PKC-induced increased production of cytokines, chemokines, and different inflammatory and apoptotic signals. Among various factors, oxidative stress has been suggested to play a major role underlying the onset and propagation of DN. It triggers several signaling pathways involved in DN, like AGEs, PKC cascade, JAK/STAT signaling, MAPK, mTOR, and SMAD. Oxidative stress-induced activation of both inflammatory and apoptotic signals are two major problems in the pathogenesis of DN. The FDA approved pharmacotherapeutic agents affecting against polyol pathway principally include anti-oxidants, like α-lipoic acid, vitamin E, and vitamin C. Kremezin and benfotiamine are the FDA approved AGEs inhibitors, another therapeutic target against DN. Ruboxistaurin, telmizartan, rapamycin, fenofibrate, aliskiren, and manidipine are some FDA approved pharmacotherapeutics effective against DN via diverse mechanisms. Beside this, some therapeutic agents are still waiting for FDA approval and few drugs without FDA approval are also prescribed in some countries for the management of DN. Despite the medications available in the market to treat DN, the involvement of multiple mechanisms makes it difficult to choose an optimum therapeutic agent. Therefore, much research is required to find out new therapeutic agent/strategies for an adequate pharmacotherapy of DN.
Collapse
Affiliation(s)
- Niloy Bhattacharjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Raja S C Mullick Road, Kolkata 700032, India
| | - Sujata Barma
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Raja S C Mullick Road, Kolkata 700032, India
| | - Nandita Konwar
- Biological Science and Technology Division, CSIR-NEIST, Jorhat, Assam 785006, India
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Raja S C Mullick Road, Kolkata 700032, India.
| | - Prasenjit Manna
- Biological Science and Technology Division, CSIR-NEIST, Jorhat, Assam 785006, India.
| |
Collapse
|
17
|
Rigothier C, Daculsi R, Lepreux S, Auguste P, Villeneuve J, Dewitte A, Doudnikoff E, Saleem M, Bourget C, Combe C, Ripoche J. CD154 Induces Matrix Metalloproteinase-9 Secretion in Human Podocytes. J Cell Biochem 2016; 117:2737-2747. [DOI: 10.1002/jcb.25571] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 04/08/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Claire Rigothier
- INSERM U1026; Université de Bordeaux; F-33076 Bordeaux France
- Service de Néphrologie Transplantation Dialyse; Centre Hospitalier Universitaire de Bordeaux; F-33076 Bordeaux France
| | - Richard Daculsi
- INSERM U1026; Université de Bordeaux; F-33076 Bordeaux France
| | | | | | - Julien Villeneuve
- Cell and Developmental Biology Programme; Centre for Genomic Regulation; 08003 Barcelona Spain
- Department of Molecular and Cell Biology; Howard Hughes Medical Institute; University of California; Berkeley California 94720-3200
| | - Antoine Dewitte
- INSERM U1026; Université de Bordeaux; F-33076 Bordeaux France
- Service d'Anesthésie-Réanimation II; Centre Hospitalier Universitaire de Bordeaux; F-33600 Pessac France
| | - Evelyne Doudnikoff
- CNRS UMR 5293; Institut des Maladies Neurodégénératives; F-33076 Bordeaux France
| | - Moin Saleem
- Children's Renal Unit and Academic Renal Unit; University of Bristol; Bristol United Kingdom
| | - Chantal Bourget
- INSERM U1026; Université de Bordeaux; F-33076 Bordeaux France
| | - Christian Combe
- INSERM U1026; Université de Bordeaux; F-33076 Bordeaux France
- Service de Néphrologie Transplantation Dialyse; Centre Hospitalier Universitaire de Bordeaux; F-33076 Bordeaux France
| | - Jean Ripoche
- INSERM U1026; Université de Bordeaux; F-33076 Bordeaux France
| |
Collapse
|
18
|
Sudhan DR, Rabaglino MB, Wood CE, Siemann DW. Cathepsin L in tumor angiogenesis and its therapeutic intervention by the small molecule inhibitor KGP94. Clin Exp Metastasis 2016; 33:461-73. [PMID: 27055649 DOI: 10.1007/s10585-016-9790-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/01/2016] [Indexed: 12/22/2022]
Abstract
A significant proportion of breast cancer patients harbor clinically undetectable micrometastases at the time of diagnosis. If left untreated, these micro-metastases may lead to disease relapse and possibly death. Hence, there is significant interest in the development of novel anti-metastatic agents that could also curb the growth of pre-established micrometastases. Like primary tumor, the growth of metastases also is driven by angiogenesis. Although the role of cysteine protease Cathepsin L (CTSL) in metastasis associated tumor cell functions such as migration and invasion is well recognized, its role in tumor angiogenesis remains less explored. The present study examines the contribution of CTSL to breast cancer angiogenesis and evaluates the anti-angiogenic efficacy of CTSL inhibitor KGP94. CTSL semi-quantitative RT-PCR analysis on breast tissue panels revealed significant upregulation of CTSL in breast cancer patients which strongly correlated with increased relapse and metastatic incidence and poor overall survival. Preclinically, CTSL ablation using shRNA or KGP94 treatment led to a significant reduction in MDA-MB-231 tumor cell induced angiogenesis in vivo. In-vitro assessments demonstrated a significant decrease in various angiogenic properties such as endothelial cell sprouting, migration, invasion, tube formation and proliferation in the presence of KGP94. Microarray analyses revealed a significant upregulation of cell cycle related genes by CTSL. Western blot analyses further confirmed upregulation of members of the cyclin family by CTSL. Collectively, these data indicate that CTSL is an important contributor to tumor angiogenesis and that the CTSL inhibition may have therapeutic utility in the treatment of breast cancer patients.
Collapse
Affiliation(s)
- Dhivya R Sudhan
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, USA. .,Cancer and Genetics Research Complex, University of Florida Health Cancer Center, Room 485E, Gainesville, FL, 32610, USA.
| | - Maria B Rabaglino
- CEPROCOR, National Scientific and Technical Research Council (CONICET), Córdoba, Argentina
| | - Charles E Wood
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, USA
| | - Dietmar W Siemann
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, USA.,Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, USA
| |
Collapse
|
19
|
Sugimoto K, Miyazawa T, Enya T, Miyazaki K, Okada M, Takemura T. Cyclosporine A induced histological changes of Cathepsin L and CD2AP expression in renal glomeruli and tubules. Clin Exp Nephrol 2016; 21:83-91. [PMID: 26975192 DOI: 10.1007/s10157-016-1257-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/03/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Cyclosporine A (CsA) is used globally as an immunosuppressant for the treatment of immune-mediated nephrotic syndrome (NS). However, its long-term use causes nephrotoxicity characterized by tubulointerstitial injury and glomerulosclerosis. The present study aimed to investigate the associations between histomorphological findings and immunohistological expression of Cathepsin L (CatL) and CD2-associated protein (CD2AP) in patients with NS mediated with CsA. METHODS A total of 18 patients with child-onset NS were divided into two groups after treatment with CsA for 2 years (group A; n = 10) and more than 4 years (group B; n = 8), respectively. Analyses of relationships between tubulointerstitial disorders and expression of CatL and CD2AP proteins were performed using immunohistochemistry of paired renal specimens. RESULTS Glomeruli with arteriole hyalinization were significantly increased in both groups depending on dosage periods, although degrees of tubule and interstitial injury did not differ between groups. CD2AP expression was significantly greater in podocytes (P = 0.046) and was significantly less in proximal tubule cells (P = 0.014) in patients of group B compared with those of group A. Moreover, CD2AP expression was significantly increased in lateral tubule cells in both groups (group A, P = 0.02; group B, P = 0.001), and CatL expression in glomeruli and tubule cells did not change with the duration of CsA treatment in either patient group. CONCLUSIONS CD2AP expression in renal tubules may histologically associate with tissue hypoxia and reflected recovery from CsA-mediated renal injury in patients, even with mild histological features of tubulointerstitial disorder.
Collapse
Affiliation(s)
- Keisuke Sugimoto
- Department of Pediatrics, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka, 589-8511, Japan.
| | - Tomoki Miyazawa
- Department of Pediatrics, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka, 589-8511, Japan
| | - Takuji Enya
- Department of Pediatrics, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka, 589-8511, Japan
| | - Kouhei Miyazaki
- Department of Pediatrics, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka, 589-8511, Japan
| | - Mitsuru Okada
- Department of Pediatrics, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka, 589-8511, Japan
| | - Tsukasa Takemura
- Department of Pediatrics, Kinki University Faculty of Medicine, 377-2 Ohno-higashi, Osaka-sayama, Osaka, 589-8511, Japan
| |
Collapse
|
20
|
Sudhan DR, Siemann DW. Cathepsin L targeting in cancer treatment. Pharmacol Ther 2015; 155:105-16. [PMID: 26299995 DOI: 10.1016/j.pharmthera.2015.08.007] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/17/2015] [Indexed: 12/14/2022]
Abstract
Proteolytic enzymes may serve as promising targets for novel therapeutic treatment strategies seeking to impede cancer progression and metastasis. One such enzyme is cathepsin L (CTSL), a lysosomal cysteine protease. CTSL upregulation, a common occurrence in a variety of human cancers, has been widely correlated with metastatic aggressiveness and poor patient prognosis. In addition, CTSL has been implicated to contribute to cancer-associated osteolysis, a debilitating morbidity affecting both life expectancy and the quality of life. In this review, we highlight the mechanisms by which CTSL contributes to tumor progression and dissemination and discuss the therapeutic utility of CTSL intervention strategies aimed at impeding metastatic progression and bone resorption.
Collapse
Affiliation(s)
- Dhivya R Sudhan
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Dietmar W Siemann
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA; Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
21
|
Kim YS, Lee WH, Choi EJ, Choi JP, Heo YJ, Gho YS, Jee YK, Oh YM, Kim YK. Extracellular vesicles derived from Gram-negative bacteria, such as Escherichia coli, induce emphysema mainly via IL-17A-mediated neutrophilic inflammation. THE JOURNAL OF IMMUNOLOGY 2015; 194:3361-8. [PMID: 25716999 DOI: 10.4049/jimmunol.1402268] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Recent evidence indicates that Gram-negative bacteria-derived extracellular vesicles (EVs) in indoor dust can evoke neutrophilic pulmonary inflammation, which is a key pathology of chronic obstructive pulmonary disease (COPD). Escherichia coli is a ubiquitous bacterium present in indoor dust and secretes nanometer-sized vesicles into the extracellular milieu. In the current study, we evaluated the role of E. coli-derived EVs on the development of COPD, such as emphysema. E. coli EVs were prepared by sequential ultrafiltration and ultracentrifugation. COPD phenotypes and immune responses were evaluated in C57BL/6 wild-type (WT), IFN-γ-deficient, or IL-17A-deficient mice after airway exposure to E. coli EVs. The present study showed that indoor dust from a bed mattress harbors E. coli EVs. Airway exposure to E. coli EVs increased the production of proinflammatory cytokines, such as TNF-α and IL-6. In addition, the repeated inhalation of E. coli EVs for 4 wk induced neutrophilic inflammation and emphysema, which are associated with enhanced elastase activity. Emphysema and elastase activity enhanced by E. coli EVs were reversed by the absence of IFN-γ or IL-17A genes. In addition, during the early period, lung inflammation is dependent on IL-17A and TNF-α, but not on IFN-γ, and also on TLR4. Moreover, the production of IFN-γ is eliminated by the absence of IL-17A, whereas IL-17A production is not abolished by IFN-γ absence. Taken together, the present data suggest that E. coli-derived EVs induce IL-17A-dependent neutrophilic inflammation and thereby emphysema, possibly via upregulation of elastase activity.
Collapse
Affiliation(s)
- You-Sun Kim
- Asan Institute for Life Science, Seoul 138-736, Republic of Korea; Ulsan University College of Medicine, Seoul 138-736, Republic of Korea
| | - Won-Hee Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Eun-Jeong Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Jun-Pyo Choi
- Institute of Convergence Medicine, Ewha Womans University School of Medicine, Seoul 158-710, Republic of Korea; and
| | - Young Joo Heo
- Asan Institute for Life Science, Seoul 138-736, Republic of Korea
| | - Yong Song Gho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Young-Koo Jee
- Department of Internal Medicine, Dankook University College of Medicine, Cheonan 330-714, Republic of Korea
| | - Yeon-Mok Oh
- Asan Institute for Life Science, Seoul 138-736, Republic of Korea; Ulsan University College of Medicine, Seoul 138-736, Republic of Korea;
| | - Yoon-Keun Kim
- Institute of Convergence Medicine, Ewha Womans University School of Medicine, Seoul 158-710, Republic of Korea; and
| |
Collapse
|
22
|
Zhong Y, Zhang X, Cai X, Wang K, Chen Y, Deng Y. Puerarin attenuated early diabetic kidney injury through down-regulation of matrix metalloproteinase 9 in streptozotocin-induced diabetic rats. PLoS One 2014; 9:e85690. [PMID: 24454919 PMCID: PMC3893265 DOI: 10.1371/journal.pone.0085690] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 12/05/2013] [Indexed: 01/13/2023] Open
Abstract
Radix puerariae, a traditional Chinese herbal medication, has been used successfully to treat patients with early stage of diabetic nephropathy. However, the underlined mechanism of this renal protective effect has not been determined. In the current study, we investigated the effects and the mechanism of puerarin in Streptozotocin (STZ)-induced diabetic rats. We treated STZ-rats with either puerarin or losartan, an angiotensin II receptor blocker, as compared to those treated with vehicle. We found that both puerarin and losartan attenuated kidney hypertrophy, mesangial expansion, proteinuria, and podocyte foot process effacement in STZ rats. In addition, both puerarin and losartan increased expression of podocyte slit diaphragm proteins such as nephrin and podocin. Interestingly, we found that puerarin treatment induced a more pronounced suppression of oxidative stress production and S-nitrosylation of proteins in the diabetic kidneys as compared to losartan treatment. Furthermore, we found that matrix metalloproteinase-9 (MMP-9), which is known to be activated by oxidative stress and S-nitrosylation of proteins, was also suppressed more extensively by puerarin than losartan. In conclusion, these data provide for the first time the potential mechanism to support the use of puerarin in the treatment of early diabetic nephropathy.
Collapse
Affiliation(s)
- Yifei Zhong
- Division of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xianwen Zhang
- Division of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xianfan Cai
- Division of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ke Wang
- National Engineering Research Center for Biochip at Shanghai, Shanghai, China
| | - Yiping Chen
- Division of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yueyi Deng
- Division of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
23
|
Prunotto M, Farina A, Lane L, Pernin A, Schifferli J, Hochstrasser DF, Lescuyer P, Moll S. Proteomic analysis of podocyte exosome-enriched fraction from normal human urine. J Proteomics 2013; 82:193-229. [PMID: 23376485 DOI: 10.1016/j.jprot.2013.01.012] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 12/17/2012] [Accepted: 01/03/2013] [Indexed: 12/14/2022]
Abstract
Urine results from a coordinated activity of glomerular and tubular compartments of the kidney. As a footprint of these cellular functional processes, urinary exosomes, and 40-80 nm membrane vesicles released after fusion with the plasma membrane into the extracellular environment by renal epithelial cells, are a source for identification of proteins and investigation of their role in the kidney. The aim of the present study was the identification of podocyte exosome proteins based on urine immunoabsorption using podocyte-specific CR1-immunocoated beads followed by proteomic analysis using LC MS/MS techniques. This methodology allowed the identification of 1195 proteins. By using a bioinformatic approach, 27 brain-expressed proteins were identified, in which 14 out of them were newly demonstrated to be expressed in the kidney at a mRNA level, and, one of them, the COMT protein, was demonstrated to be expressed in podocytes at a protein level. These results, attesting the reliability of the methodology to identify podocyte proteins, need now to be completed by further experiments to analyze more precisely their biological function(s) in the podocytes.
Collapse
Affiliation(s)
- Marco Prunotto
- Institute of Clinical Pathology, Geneva University Hospitals, Geneva CH-1211, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Dubern B, Lafarge JC, Fellahi S, Naour N, Copin N, Bastard JP, Simon C, Clément K, Bonnet D, Tounian P, Guerre-Millo M. Relevance of increased serum cystatin C to vascular alterations in obese children. Pediatr Obes 2012; 7:374-81. [PMID: 22653843 DOI: 10.1111/j.2047-6310.2012.00053.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 01/15/2012] [Accepted: 02/08/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE Epidemiological studies report a positive relationship between serum cystatin C and cardiovascular outcomes in adults. Here, we tested the relevance of cystatin C as a biomarker for early vascular alterations in severely obese children. METHODS Two hundred nineteen obese (140 girls; age = 11.7 ± 2.7 years, BMI Z-score = 4.7 ± 1.2 SD) and 262 non-obese children (129 girls; age = 11.6 ± 0.6 years, body mass index [BMI] Z-score = 0.1 ± 1.0 SD). Serum cystatin C was measured by immunonephelometry. Intima media thickness (IMT), incremental elastic modulus, and flow-mediated and glyceryl-trinitrate-mediated dilations were determined at the common carotid artery and the brachial artery in obese children. RESULTS Obese children had significantly higher serum cystatin C than normal weight controls (0.86 ± 0.01 vs. 0.80 ± 0.01, P < 0.0001). In obese children, serum cystatin C correlates positively with BMI and the homeostasis model assessment index and negatively with the quantitative insulin sensitivity check index and adiponectin. A positive relationship was found between serum cystatin C and carotid IMT (r = 0.23, P = 0.0005), which remained significant in multivariate models adjusted for BMI (P = 0.01) and adiponectin with a trend towards significance (P = 0.05). CONCLUSION This study positions cystatin C and adiponectin as covariables associated with arterial wall thickness in obese children. Although the underlying pathophysiology linking cystatin C to early vascular disease remains to be deciphered, cystatin C may represent a novel adipose tissue-derived biomarker implicated in obesity-related comorbidities early in life.
Collapse
Affiliation(s)
- B Dubern
- Institute of Cardiometabolism and Nutrition, Paris, France; INSERM, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Severe intraglomerular detachment of podocytes in a Gitelman syndrome patient. Clin Exp Nephrol 2012; 16:495-500. [PMID: 22484642 DOI: 10.1007/s10157-012-0624-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 03/08/2012] [Indexed: 12/13/2022]
Abstract
We report the case of a 38-year-old woman diagnosed with Gitelman syndrome. A kidney biopsy showed abundant floating cells in the Bowman's space of the mildly cystic glomeruli, moderate tubulointerstitial changes and apparent intimal thickening of small arteries. These floating cells were immunohistologically identified as podocytes, by the expression of podocalyxin, vimentin, Wilms' tumor 1, synaptopodin and nephrin with positivities of 100%, 88.4%, 80.4%, 74.7% and 22.6%, respectively. In these phenotypes, nephrin expression was notably decreased in both detached and capillary-attached podocytes in comparison with normal control podocytes. Immunostaining of both detached and capillary-attached podocytes for Bax, Bcl-2, desmin, fibroblast-specific protein-1, α-smooth muscle actin and Ki-67 was negative, as were TUNEL assays. These results suggest that apoptosis and epithelial-mesenchymal transition were not the main cause of podocyte detachment in this patient. In addition, levels of urinary podocalyxin were not elevated, suggesting the detached podocytes were not excreted in the urine. To the best of our knowledge, this is the first report of severe intraglomerular non-apoptotic detachment of podocytes in Gitelman syndrome. This podocyte detachment may be associated with nephron obstruction and reduced nephrin expression.
Collapse
|
26
|
Abstract
As an integral member of the filtration barrier in the kidney glomerulus, the podocyte is in a unique geographical position: It is exposed to chemical signals from the urinary space (Bowman's capsule), it receives and transmits chemical and mechanical signals to/from the glomerular basement membrane upon which it elaborates, and it receives chemical and mechanical signals from the vascular space with which it also communicates. As with every cell, the ability of the podocyte to receive signals from the surrounding environment and to translate them to the intracellular milieu is dependent largely on molecules residing on the cell membrane. These molecules are the first-line soldiers in the ongoing battle to sense the environment, to respond to friendly signals, and to defend against injurious foes. In this review, we take a membrane biologist's view of the podocyte, examining the many membrane receptors, channels, and other signaling molecules that have been implicated in podocyte biology. Although we attempt to be comprehensive, our goal is not to capture every membrane-mediated pathway but rather to emphasize that this approach may be fruitful in understanding the podocyte and its unique properties.
Collapse
Affiliation(s)
- Anna Greka
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA.
| | | |
Collapse
|
27
|
|
28
|
Magee TR, Tafti SA, Desai M, Liu Q, Ross MG, Nast CC. Maternal undernourished fetal kidneys exhibit differential regulation of nephrogenic genes including downregulation of the Notch signaling pathway. Reprod Sci 2011; 18:563-76. [PMID: 21273641 DOI: 10.1177/1933719110393025] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Maternal undernutrition results in offspring nephron number reduction and hypertension that are hypothesized to begin as compensatory changes in fetal gene expression during gestation. To evaluate mechanisms of dysregulated nephrogenesis, pregnant Sprague Dawley rats were 50% food restricted from embryonic day (E) 10 to E20. At E20, fetal male kidneys were examined by microarray analysis. A total of 476 differentially expressed transcripts were detected including those regulating development and differentiation, mitosis and cell cycle, chromatin assembly, and steroid hormone regulation. Differentially regulated genes were detected in MAPK/ERK, Wnt, and Notch signaling pathways. Validation of the microarray results was performed for the Notch signaling pathway, an important pathway in nephron formation. Protein expression of Notch pathway factors by Western blotting showed significantly decreased Notch2 and downstream effector Hey1 protein expression, while Ctbp1 co-repressor was increased. These data together show that maternal undernutrition results in developmental disruption in fetal nephrogenesis gene expression signaling.
Collapse
Affiliation(s)
- Thomas R Magee
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
29
|
Cathepsins B and L in peripheral blood mononuclear cells of pediatric acute myeloid leukemia: potential poor prognostic markers. Ann Hematol 2010; 89:1223-32. [PMID: 20567828 DOI: 10.1007/s00277-010-1012-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 06/08/2010] [Indexed: 10/19/2022]
Abstract
The diagnostic and prognostic significance of cathepsin B (CTSB) and L (CTSL) is well documented for solid tumors. However, their significance in acute leukemias is lacking. This study was planned to investigate expression and significance of these proteases in peripheral blood mononuclear cells (PBMCs) of patients with pediatric acute myeloid leukemia (AML). CTSL and CTSB activities were assayed in PBMCs of 24 children with AML and ten healthy controls by spectrofluorimetry. The mRNA levels of these proteases and their specific endogenous inhibitor cystatin C and transcriptional upregulator vascular endothelial growth factor (VEGF) were quantitated by real-time PCR. Correlation analysis of CTSL and CTSB activities/expression with their inhibitor/upregulator and event-free survival (EFS) was done using appropriate statistical tools. CTSL and CTSB protease activity and their mRNA expression were significantly higher in AML patients compared to controls (p ≤ 0.001). A strong positive correlation was observed between VEGF expression and CTSL (r = 0.812; p ≤ 0.001). Similarly, VEGF exhibited a strong positive correlation with CTSB (r = 0.501; p = 0.013). Cystatin expression though significantly high (p ≤ 0.001) in AML was negatively correlated with CTSL (r = -0.920; p ≤ 0.001) and CTSB (r = -0.580, p ≤ 0.001) expression. AML patients with higher CTSL and CTSB activity exhibited an inferior EFS (CTSL: p = 0.045; CTSB: p = 0.002) and overall survival (OS; CTSL: p = 0.05; CTSB: p = 0.004) compared to patients with lower levels of these proteases. This is the first report demonstrating increased expression of CTSL and CTSB in AML, mechanism of their increased expression in relation to VEGF, and their association with poor EFS and OS. These results suggest a potential utility of these proteases as prognostic markers for this malignancy.
Collapse
|
30
|
Kistler AD, Peev V, Forst AL, El Hindi S, Altintas MM, Reiser J. Enzymatic disease of the podocyte. Pediatr Nephrol 2010; 25:1017-23. [PMID: 20130922 PMCID: PMC4109305 DOI: 10.1007/s00467-009-1425-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 11/11/2009] [Accepted: 11/17/2009] [Indexed: 01/08/2023]
Abstract
Proteinuria is an early sign of kidney disease and has gained increasing attention over the past decade because of its close association with cardio-vascular and renal morbidity and mortality. Podocytes have emerged as the cell type that is critical in maintaining proper functioning of the kidney filter. A few genes have been identified that explain genetic glomerular failure and recent insights shed light on the pathogenesis of acquired proteinuric diseases. This review highlights the unique role of the cysteine protease cathepsin L as a regulatory rather than a digestive protease and its action on podocyte structure and function. We provide arguments why many glomerular diseases can be regarded as podocyte enzymatic disorders.
Collapse
Affiliation(s)
- Andreas D. Kistler
- Department of Nephrology, University Hospital, Zürich, Switzerland,Miami Institute of Renal Medicine, Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, 1580 NW 10th Street, Batchelor Bldg. #633A, Miami, FL 33136, USA
| | - Vasil Peev
- Miami Institute of Renal Medicine, Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, 1580 NW 10th Street, Batchelor Bldg. #633A, Miami, FL 33136, USA
| | - Anna-Lena Forst
- Miami Institute of Renal Medicine, Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, 1580 NW 10th Street, Batchelor Bldg. #633A, Miami, FL 33136, USA
| | - Shafic El Hindi
- Miami Institute of Renal Medicine, Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, 1580 NW 10th Street, Batchelor Bldg. #633A, Miami, FL 33136, USA
| | - Mehmet M. Altintas
- Miami Institute of Renal Medicine, Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, 1580 NW 10th Street, Batchelor Bldg. #633A, Miami, FL 33136, USA
| | - Jochen Reiser
- Miami Institute of Renal Medicine, Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, 1580 NW 10th Street, Batchelor Bldg. #633A, Miami, FL 33136, USA
| |
Collapse
|
31
|
Abstract
Proteinuria is a major health-care problem that affects several hundred million people worldwide. Proteinuria is a cardinal sign and a prognostic marker of kidney disease, and also an independent risk factor for cardiovascular morbidity and mortality. Microalbuminuria is the earliest cue of renal complications of diabetes, obesity, and the metabolic syndrome. It can often progress to overt proteinuria that in 10-50% of patients is associated with the development of chronic kidney disease, ultimately requiring dialysis or transplantation. Therefore, reduction or prevention of proteinuria is highly desirable. Here we review recent novel insights into the pathogenesis and treatment of proteinuria, with a special emphasis on the emerging concept that proteinuria can result from enzymatic cleavage of essential regulators of podocyte actin dynamics by cytosolic cathepsin L (CatL), resulting in a motile podocyte phenotype. Finally, we describe signaling pathways controlling the podocyte actin cytoskeleton and motility and how these pathways can be manipulated for therapeutic benefit.
Collapse
Affiliation(s)
- Peter Mundel
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA.
| | | |
Collapse
|
32
|
Tsai CH, Yang SF, Huang FM, Chang YC. The upregulation of cystatin C in human gingival fibroblasts stimulated with cyclosporine A. J Periodontal Res 2009; 44:459-64. [DOI: 10.1111/j.1600-0765.2008.01147.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
33
|
Park SJ, Lee BH, Kim DJ. Identification of proteins that interact with podocin using the yeast 2-hybrid system. Yonsei Med J 2009; 50:273-9. [PMID: 19430563 PMCID: PMC2678704 DOI: 10.3349/ymj.2009.50.2.273] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 08/28/2008] [Accepted: 08/28/2008] [Indexed: 11/27/2022] Open
Abstract
PURPOSE As a membrane protein at the insertion site of the slit diaphragm (SD) complex in podocyte foot processes, podocin has been reported to act as a scaffolding protein required to maintain or regulate the structural integrity of the SD. In order to identify proteins that associate or interact with podocin, we screened a mouse kidney complementary DNA (cDNA) library using a yeast 2-hybrid system. MATERIALS AND METHODS 1) The full-length cDNA of podocin from the mouse kidney was amplified by Polymerase Chain Reaction (PCR), 2) The PCR product was cloned into a pGBKT7 vector, pGBKT7-podocin, 3) After the pGBKT7-podocin was transformed into AH109, the AH109/pGBKT7-podocin product was obtained, 4) The mouse kidney cDNA library was transformed into the AH109/pGBKT7-podocin and screened by selection steps, 5) Next, twelve clones were cultured and isolated, 6) The yeast-purified plasmids were transformed into Escherichia coli (E. coli) by heat shock, and 7) To identify the activation domain (AD)/library inserts, we digested them with Him III, and the fragments were then sequenced. RESULTS 12 positive clones that interacted with podocin were obtained by screening a mouse kidney cDNA library using pGBKT7-podocin. Among them, only 4 clones were found to function at the podocyte where podocin is present. CONCLUSION Additional studies are needed to clarify the role and interaction with podocin and candidates.
Collapse
Affiliation(s)
- Soo Jin Park
- Clinical Research Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Byung Ha Lee
- Clinical Research Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dae Joong Kim
- Division of Nephrology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
34
|
Thrailkill KM, Clay Bunn R, Fowlkes JL. Matrix metalloproteinases: their potential role in the pathogenesis of diabetic nephropathy. Endocrine 2009; 35:1-10. [PMID: 18972226 PMCID: PMC2629499 DOI: 10.1007/s12020-008-9114-6] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Revised: 07/21/2008] [Accepted: 09/03/2008] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinases (MMPs), a family of proteinases including collagenases, gelatinases, stromelysins, matrilysins, and membrane-type MMPs, affect the breakdown and turnover of extracellular matrix (ECM). Moreover, they are major physiologic determinants of ECM degradation and turnover in the glomerulus. Renal hypertrophy and abnormal ECM deposition are hallmarks of diabetic nephropathy (DN), suggesting that altered MMP expression or activation contributes to renal injury in DN. Herein, we review and summarize recent information supporting a role for MMPs in the pathogenesis of DN. Specifically, studies describing dysregulated activity of MMPs and/or their tissue inhibitors in various experimental models of diabetes, including animal models of type 1 or type 2 diabetes, clinical investigations of human type 1 or type 2 diabetes, and kidney cell culture studies are reviewed.
Collapse
Affiliation(s)
- Kathryn M Thrailkill
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA.
| | | | | |
Collapse
|
35
|
Oneda B, Lods N, Lottaz D, Becker-Pauly C, Stöcker W, Pippin J, Huguenin M, Ambort D, Marti HP, Sterchi EE. Metalloprotease meprin beta in rat kidney: glomerular localization and differential expression in glomerulonephritis. PLoS One 2008; 3:e2278. [PMID: 18509531 PMCID: PMC2386549 DOI: 10.1371/journal.pone.0002278] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 04/17/2008] [Indexed: 01/06/2023] Open
Abstract
Meprin (EC 3.4.24.18) is an oligomeric metalloendopeptidase found in microvillar membranes of kidney proximal tubular epithelial cells. Here, we present the first report on the expression of meprin beta in rat glomerular epithelial cells and suggest a potential involvement in experimental glomerular disease. We detected meprin beta in glomeruli of immunostained rat kidney sections on the protein level and by quantitative RT-PCR of laser-capture microdissected glomeruli on the mRNA level. Using immuno-gold staining we identified the membrane of podocyte foot processes as the main site of meprin beta expression. The glomerular meprin beta expression pattern was altered in anti-Thy 1.1 and passive Heymann nephritis (PHN). In addition, the meprin beta staining pattern in the latter was reminiscent of immunostaining with the sheep anti-Fx1A antiserum, commonly used in PHN induction. Using Western blot and immunoprecipitation assays we demonstrated that meprin beta is recognized by Fx1A antiserum and may therefore represent an auto-antigen in PHN. In anti-Thy 1.1 glomerulonephritis we observed a striking redistribution of meprin beta in tubular epithelial cells from the apical to the basolateral side and the cytosol. This might point to an involvement of meprin beta in this form of glomerulonephritis.
Collapse
Affiliation(s)
- Beatrice Oneda
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Nadège Lods
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Daniel Lottaz
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | | | - Walter Stöcker
- Institute of Zoology, Johannes Gutenberg University, Mainz, Germany
| | - Jeffrey Pippin
- Division of Nephrology, University of Washington, Seattle, Washington, United States of America
| | - Maya Huguenin
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Daniel Ambort
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Hans-Peter Marti
- Division of Nephrology/Hypertension, Inselspital, University of Bern, Bern, Switzerland
| | - Erwin E. Sterchi
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
36
|
Chang HR, Kuo WH, Hsieh YS, Yang SF, Lin CC, Lee ML, Lian JD, Chu SC. Circulating matrix metalloproteinase-2 is associated with cystatin C level, posttransplant duration, and diabetes mellitus in kidney transplant recipients. Transl Res 2008; 151:217-23. [PMID: 18355769 DOI: 10.1016/j.trsl.2007.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Revised: 12/17/2007] [Accepted: 12/18/2007] [Indexed: 12/30/2022]
Abstract
Studies have indicated that matrix metalloproteinase-2 (MMP-2) is vital for the patient's condition after renal transplantation. Although the allograft survival rate has been improved, the relationships between various clinical parameters in stable graft function and serum MMP-2 still need to be clarified. In this study, gelatin zymography and enzyme-linked immunosorbent assay were employed to measure MMP-2 level in the plasma of 152 kidney transplant recipients, 41 chronic kidney disease patients, and 50 healthy control subjects. The creatinine and the MMP-2 levels in the transplant recipients were significantly greater (P < 0.001) than those of control subjects. Univariate and stepwise regression analysis demonstrated the MMP-2 level was associated with cystatin C level (P < 0.001), creatinine level (P = 0.036), proteinuria (P = 0.043), posttransplant days (P = 0.025), and posttransplant diabetes mellitus (P = 0.03). We conclude that circulating MMP-2 is associated with cystatin C, posttransplant duration, and diabetes mellitus in kidney transplant recipients and suggest that MMP-2 may be critical for graft survival.
Collapse
Affiliation(s)
- Horng-Rong Chang
- Division of Nephrology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Floege J, Eitner F, Alpers CE. A New Look at Platelet-Derived Growth Factor in Renal Disease. J Am Soc Nephrol 2007; 19:12-23. [DOI: 10.1681/asn.2007050532] [Citation(s) in RCA: 235] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
38
|
Sever S, Altintas MM, Nankoe SR, Möller CC, Ko D, Wei C, Henderson J, del Re EC, Hsing L, Erickson A, Cohen CD, Kretzler M, Kerjaschki D, Rudensky A, Nikolic B, Reiser J. Proteolytic processing of dynamin by cytoplasmic cathepsin L is a mechanism for proteinuric kidney disease. J Clin Invest 2007. [PMID: 17671649 DOI: 10.1172/jci32022.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Kidney podocytes and their foot processes maintain the ultrafiltration barrier and prevent urinary protein loss (proteinuria). Here we show that the GTPase dynamin is essential for podocyte function. During proteinuric kidney disease, induction of cytoplasmic cathepsin L leads to cleavage of dynamin at an evolutionary conserved site, resulting in reorganization of the podocyte actin cytoskeleton and proteinuria. Dynamin mutants that lack the cathepsin L site, or render the cathepsin L site inaccessible through dynamin self-assembly, are resistant to cathepsin L cleavage. When delivered into mice, these mutants restored podocyte function and resolve proteinuria. Our study identifies dynamin as a critical regulator of renal permselectivity that is specifically targeted by proteolysis under pathological conditions.
Collapse
Affiliation(s)
- Sanja Sever
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts 02129, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sever S, Altintas MM, Nankoe SR, Möller CC, Ko D, Wei C, Henderson J, del Re EC, Hsing L, Erickson A, Cohen CD, Kretzler M, Kerjaschki D, Rudensky A, Nikolic B, Reiser J. Proteolytic processing of dynamin by cytoplasmic cathepsin L is a mechanism for proteinuric kidney disease. J Clin Invest 2007; 117:2095-104. [PMID: 17671649 PMCID: PMC1934589 DOI: 10.1172/jci32022] [Citation(s) in RCA: 168] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Accepted: 05/09/2007] [Indexed: 12/11/2022] Open
Abstract
Kidney podocytes and their foot processes maintain the ultrafiltration barrier and prevent urinary protein loss (proteinuria). Here we show that the GTPase dynamin is essential for podocyte function. During proteinuric kidney disease, induction of cytoplasmic cathepsin L leads to cleavage of dynamin at an evolutionary conserved site, resulting in reorganization of the podocyte actin cytoskeleton and proteinuria. Dynamin mutants that lack the cathepsin L site, or render the cathepsin L site inaccessible through dynamin self-assembly, are resistant to cathepsin L cleavage. When delivered into mice, these mutants restored podocyte function and resolve proteinuria. Our study identifies dynamin as a critical regulator of renal permselectivity that is specifically targeted by proteolysis under pathological conditions.
Collapse
Affiliation(s)
- Sanja Sever
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Mehmet M. Altintas
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Sharif R. Nankoe
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Clemens C. Möller
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - David Ko
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Changli Wei
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Joel Henderson
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Elizabetta C. del Re
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Lianne Hsing
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Ann Erickson
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Clemens D. Cohen
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Matthias Kretzler
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Dontscho Kerjaschki
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Alexander Rudensky
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Boris Nikolic
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Jochen Reiser
- Department of Medicine, Nephrology Division and Program in Glomerular Disease, Massachusetts General Hospital (MGH) and Harvard Medical School, Boston, Massachusetts, USA.
Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
Department of Immunology, University of Washington, Seattle, Washington, USA.
Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA.
Medizinische Poliklinik, University of Munich, Munich, Germany.
Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
Clinical Institute of Pathology, Vienna Medical University, Vienna, Austria.
Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
40
|
Thrailkill KM, Bunn RC, Moreau CS, Cockrell GE, Simpson PM, Coleman HN, Frindik JP, Kemp SF, Fowlkes JL. Matrix metalloproteinase-2 dysregulation in type 1 diabetes. Diabetes Care 2007; 30:2321-6. [PMID: 17563344 PMCID: PMC2239013 DOI: 10.2337/dc07-0162] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Dysregulation of matrix metalloproteinase (MMP)-2 may contribute pathologically to the development of diabetes complications, including diabetic retinopathy and coronary and peripheral arterial disease. Our objective was to explore whether systemic MMP-2 dysregulation could be demonstrated in type 1 diabetes and to determine how MMP-2 concentration relates to disease status. RESEARCH DESIGN AND METHODS In this cross-sectional study, MMP-2 concentrations and MMP-2 activity were measured in plasma and timed urine samples from 93 type 1 diabetic and 50 healthy control subjects, aged 14-40 years. Relationships between MMP-2 concentrations in these biological fluids and subject characteristics (sex, age, and duration of type 1 diabetes), indexes of glycemic control (A1C, fasting plasma glucose, and continuous glucose monitoring system average daily glucose), and measurements of renal function (urinary albumin excretion and glomerular filtration rate) were examined. RESULTS Urine and plasma MMP-2 concentrations and plasma MMP-2 activity were all significantly elevated in type 1 diabetic subjects compared with those in control subjects. Urine MMP-2 concentrations, in particular, were correlated with several clinical parameters that infer increased risk for diabetic comorbidity and specifically for diabetic nephropathy, including higher A1C, longer duration of disease, evidence of renal hyperfiltration, and the presence of microalbuminuria. CONCLUSIONS Urine and plasma MMP-2 concentrations are dysregulated in type 1 diabetes; urinary excretion of MMP-2, in particular, might provide a unique biomarker of diabetes-induced intrarenal pathologic processes.
Collapse
Affiliation(s)
- Kathryn M Thrailkill
- Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital Research Institute, Little Rock, Arkansas 72202, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Keerthivasan S, Keerthivasan G, Mittal S, Chauhan SS. Transcriptional upregulation of human cathepsin L by VEGF in glioblastoma cells. Gene 2007; 399:129-36. [PMID: 17574778 DOI: 10.1016/j.gene.2007.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Revised: 04/21/2007] [Accepted: 05/05/2007] [Indexed: 11/27/2022]
Abstract
The role of vascular endothelial growth factor (VEGF) on cathepsin L expression was investigated in human glioblastoma cells (U87MG). Our results demonstrate the transcriptional upregulation of cathepsin L expression by VEGF. Transient transfection of U87MG cells with VEGF expression vector significantly increased cathepsin L activity. These results were further corroborated by a parallel increase in the mRNA levels and promoter activity of cathepsin L by VEGF. By deletion analysis, we identified a 47 base pair VEGF response element (VRE) in human cathepsin L promoter. Site directed mutagenesis studies demonstrated that both SP-1 and AP-4 motifs present in this region contribute to VEGF responsiveness. These results prove for the first time that over-expression of VEGF in human glioblastoma cells induces cathepsin L expression at the transcriptional level. This mechanism could be involved in the enhanced tumorogenic potential of these cells.
Collapse
Affiliation(s)
- S Keerthivasan
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi-110029, India.
| | | | | | | |
Collapse
|
42
|
Chung-Hung T, Shun-Fa Y, Yu-Chao C. The upregulation of cystatin C in oral submucous fibrosis. Oral Oncol 2006; 43:680-5. [PMID: 17070095 DOI: 10.1016/j.oraloncology.2006.08.009] [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] [Received: 07/04/2006] [Accepted: 08/11/2006] [Indexed: 10/24/2022]
Abstract
Cystatin C is a 13kDa non-glycosylated basic protein belonging to cystatin family. It is consistently and dramatically upregulated in a variety of fibrotic diseases. The aim of this study was to compare cystatin C expression in normal human buccal mucosa and oral submucous fibrosis (OSF) specimens and further explore the potential mechanism that may lead to induction of cystatin C expression. Twenty-five OSF specimens and six of normal buccal mucosa were examined by immunohistochemistry. The activity of cystatin C from fibroblasts cultured from OSF and normal buccal mucosa were evaluated by using reverse-transcriptase polymerase chain reaction and enzyme-linked immunosorbent assay. Furthermore, the effect of arecoline, the major areca nut alkaloid, was explored. Cystatin C expression was significantly higher in OSF specimens (p<0.05) and expressed mainly by fibroblasts, endothelial cells, and inflammatory cells. OSF demonstrated significantly higher cystatin C expression than normal buccal mucosa fibroblasts both in mRNA and protein levels (p<0.05). In addition, arecoline was also found to elevate cystatin C mRNA and protein expression in a dose-dependent manner (p<0.05). Taken together, the data demonstrate that cystatin C expression is significantly upregulated in OSF from areca quid chewers and arecoline may be responsible for the enhanced cystatin C expression in vivo.
Collapse
Affiliation(s)
- Tsai Chung-Hung
- Department of Oral Pathology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | | | | |
Collapse
|
43
|
McDevitt TM, Gonzales LW, Savani RC, Ballard PL. Role of endogenous TGF-beta in glucocorticoid-induced lung type II cell differentiation. Am J Physiol Lung Cell Mol Physiol 2006; 292:L249-57. [PMID: 16997883 DOI: 10.1152/ajplung.00088.2006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the fetal lung, endogenous transforming growth factor (TGF)-beta inhibits early morphogenesis and blocks hormone-induced type II cell differentiation. We hypothesized that endogenous TGF-beta inhibits type II cell differentiation and that the stimulatory effects of glucocorticoids result in part from suppression of TGF-beta. Epithelial cells were isolated from human fetal lung and cultured under defined conditions with and without dexamethasone plus cAMP to promote type II cell differentiation. Control cells produced TGF-beta, which was activated in part by alpha(V)beta(6)-integrin. Treatment with dexamethasone, but not cAMP, reduced TGF-beta1 and -beta2 transcripts and TGF-beta bioactivity in culture medium. To examine the effects of decreased TGF-beta in the absence of glucocorticoid, cells were treated with antibodies to TGF-beta and its receptors. By real-time RT-PCR, antibody blockade of TGF-beta reduced serpine1, a TGF-beta-inducible gene, and increased gene expression for sftpa, sftpb, sftpc, and titf1, mimicking the response to hormone treatment. By microarray analysis, 29 additional genes were induced by both TGF-beta antibody and hormone treatment, and 20 other genes were repressed by both treatments. For some genes, the fold response was comparable for antibody and hormone treatment. We conclude that endogenous TGF-beta suppresses expression of surfactant proteins and selected other type II cell genes in fetal lung, in part secondary to increased expression of titf1, and we propose that the mechanism of glucocorticoid-induced type II cell differentiation includes antagonism of TGF-beta gene suppression. Surfactant production during fetal development is likely influenced by relative levels of TGF-beta and glucocorticoids.
Collapse
Affiliation(s)
- Theresa M McDevitt
- Division of Neonatology, Department of Pediatrics, University of California-San Francisco, 3333 California St., Suite 150, San Francisco, CA 94118-1981, USA
| | | | | | | |
Collapse
|
44
|
Rao VH, Meehan DT, Delimont D, Nakajima M, Wada T, Gratton MA, Cosgrove D. Role for macrophage metalloelastase in glomerular basement membrane damage associated with alport syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:32-46. [PMID: 16816359 PMCID: PMC1698763 DOI: 10.2353/ajpath.2006.050896] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Alport syndrome is a glomerular basement membrane (GBM) disease caused by mutations in type IV collagen genes. A unique irregular thickening and thinning of the GBM characterizes the progressive glomerular pathology. The metabolic imbalances responsible for these GBM irregularities are not known. Here we show that macrophage metalloelastase (MMP-12) expression is >40-fold induced in glomeruli from Alport mice and is markedly induced in glomeruli of both humans and dogs with Alport syndrome. Treatment of Alport mice with MMI270 (CGS27023A), a broad spectrum MMP inhibitor that blocks MMP-12 activity, results in largely restored GBM ultrastructure and function. Treatment with BAY-129566, a broad spectrum MMP inhibitor that does not inhibit MMP-12, had no effect. We show that inhibition of CC chemokine receptor 2 (CCR2) receptor signaling with propagermanium blocks induction of MMP-12 mRNA and prevents GBM damage. CCR2 receptor is expressed in glomerular podocytes of Alport mice, suggesting MCP-1 activation of CCR2 on podocytes may underlie induction of MMP-12. These data indicate that the irregular GBM that characterizes Alport syndrome may be mediated, in part, by focal degradation of the GBM due to MMP dysregulation, in particular, MMP-12. Thus, MMP-12/CCR2 inhibitors may provide a novel and effective therapeutic stra-tegy for Alport glomerular disease.
Collapse
Affiliation(s)
- Velidi H Rao
- Boys Town National Research Hospital, 555 No. 30th St., Omaha, NE 68131, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
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
|
46
|
Liu S, Li Y, Zhao H, Chen D, Huang Q, Wang S, Zou W, Zhang Y, Li X, Huang H. Increase in extracellular cross-linking by tissue transglutaminase and reduction in expression of MMP-9 contribute differentially to focal segmental glomerulosclerosis in rats. Mol Cell Biochem 2006; 284:9-17. [PMID: 16477388 DOI: 10.1007/s11010-005-9005-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Accepted: 09/13/2005] [Indexed: 12/23/2022]
Abstract
Tissue transglutaminase (tTG) is a Ca(2+)-dependent enzyme which stabilizes the extracellular matrix (ECM) through post-translational modification, and may play an important role in the pathogenesis of focal and segmental glomerulosclerosis (FSGS). Here, we have investigated whether tTG contributes to the glomerular ECM expansion in the puromycin aminonucleoside (PAN)-injection-induced experimental rat model of FSGS. The localization and expression of tTG, MMP-9 gelatinase, and the ECM component fibronectin (FN) in kidneys was determined by immunohistochemistry and measured by semi-quantitative analysis. Protein levels of tTG and MMP-9 were also analyzed by Western blotting.In situtransglutaminase activity was assayed by measurement of incorporated substrate and the immunofluorescence staining for the cross-linking product, epsilon-(gamma-glutamyl) lysine. Prominent proteinuria, a typical pathological feature of FSGS, was observed in PAN injection group rats. tTG immunoreactivity was located markedly in glomeruli and the levels of this protein in whole-kidney homogenates of PAN injection group rats were significantly increased (361+/- 106% control, P< 0.05). Similarly, transglutaminase activity and epsilon-(gamma-glutamyl) lysine were also predominately located within glomeruli and were much more intense in the PAN-injected group than that in control animals. MMP-9 was also located primarily within glomeruli. In PAN-injected kidneys, protein levels of active MMP-9 were significantly reduced (59+/- 27% control, P< 0.01), while pro-MMP-9 levels increased (148+/- 42% control, P< 0.05). Remarkable expression of glomerular fibronectin (FN) was found in PAN injection group rats. Semi-quantitative analysis demonstrated this increased intensity of FN staining in the PAN-injected rats was 149+/- 23% of the control values (P< 0.05). Enhanced cross-linking of ECM by tissue transglutaminase and decreased degradation due to reduced active MMP-9 expression may be at least partially responsible for the deposition of FN within injured glomeruli in experimental FSGS.
Collapse
Affiliation(s)
- Senyan Liu
- Division of Nephrology, Peking University First Hospital, Beijing, 100034, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Liu S, Liang Y, Huang H, Wang L, Li Y, Li J, Li X, Wang H. ERK-dependent signaling pathway and transcriptional factor Ets-1 regulate matrix metalloproteinase-9 production in transforming growth factor-beta1 stimulated glomerular podocytes. Cell Physiol Biochem 2006; 16:207-16. [PMID: 16301820 DOI: 10.1159/000089846] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2005] [Indexed: 12/31/2022] Open
Abstract
The unregulated synthesis of glomerular basement membrane (GBM) components, extracelluar matrix (ECM) proteins, or the secretion of ECM-degradation enzymes, matrix metalloproteinases (MMPs), by podocytes under pathological conditions might be major factors in GBM damage. The present study examined the effects and the underlying molecular mechanism of transforming growth factor beta1 (TGFbeta1) on the production of gelatinase in cultured murine podocytes. Our results showed that TGFbeta1 is the most potent inducer of MMP-9 secretion in both a dose- and time-dependent manner, but has very little effect on MMP-2 secretion. TGFbeta1 upregulated MMP-9 mRNA levels, but did not affect the expression of matrix mettaloproteinases TIMP-1 mRNA. TGFbeta1 induced activation of both Smad2 and extracellular signal-regulated kinases (ERK1/2). However, blockade of Smad2 signaling pathway by Staurosporine did not affect the TGFbeta1-stimulated secretion of MMP-9, whereas inhibition of activation of ERK1/2 by PD98059 abolished TGFbeta1-stimulated secretion of MMP-9 and expression of MMP-9 mRNA. Protein levels of the transcriptional factor Ets-1 increased and were sustained for 12 h by TGFbeta1-stimulation. Our data also showed that blockage of ERK1/2 activation by PD98059 led to a reduction in the level of Ets-1 protein and to a consequent decrease in MMP-9 mRNA levels. These results demonstrate that TGFbeta1 can induce the production of MMP-9 in podocytes through the ERK1/2 MAPK pathway, and suggested that an increase in MMP-9 enzymatic activities may be involved in the damage of the GBM in response to inflammatory factors, ultimately leading to glomerulosclerosis.
Collapse
Affiliation(s)
- Senyan Liu
- Division of Nephrology, Peking University First Hospital, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Bai Y, Wang L, Li Y, Liu S, Li J, Wang H, Huang H. High Ambient Glucose Levels Modulates the Production of MMP-9 and a5(IV) Collagen by Cultured Podocytes. Cell Physiol Biochem 2006; 17:57-68. [PMID: 16543722 DOI: 10.1159/000091464] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Recent evidences have demonstrated an important role for glomerular visceral epithelial cell (podocyte) in the development and progression of diabetic nephropathy. We investigated the high-glucose (HG)-triggered signaling pathway and its role in matrix metalloproteinase (MMP) production in murine podocytes. The activity of 92-kDa (MMP-9) gelatinase, but not of 72 kDa (MMP-2), in an HG medium significantly increased during incubation of 2 to 3 days and decreased during incubation of more than 5 days revealed by Gelatin zymography. Opposite to the increases in MMP-9 activity, HG medium produced significant decreases in the protein levels of alpha5(IV) collagen. Changes in MMP-9 activity were associated with the same pattern as MMP-9 mRNA levels in podocytes exposed to HG media. HG medium rapidly activated ERK1/2 MAPK in podocytes. Moreover, ERK1/2 activation was required for HG-induced enhancement of MMP-9 activity and a decrease in the level of alpha5(IV) collagen. HG incubation rapidly induced an increase in the nuclear accumulation of Ets-1 protein. Blocking the ERK pathway suppressed HG-induced expression and nuclear accumulation of transcriptional factor Ets-1, and MMP-9 mRNA expression. We suggest that short- or long-term exposure to HG concentrations increases or decreases MMP-9 production and alpha5(IV) collagen expression in podocytes, this may contribute to the GBM abnormality caused by an imbalance in extracellular matrix (ECM) synthesis and degradation, and may play a critical role in the pathogenesis of proteinuria in diabetic nephropathy.
Collapse
Affiliation(s)
- Yaling Bai
- Division of Nephrology, Peking University First Hospital, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Hayden MR, Whaley-Connell A, Sowers JR. Renal redox stress and remodeling in metabolic syndrome, type 2 diabetes mellitus, and diabetic nephropathy: paying homage to the podocyte. Am J Nephrol 2005; 25:553-69. [PMID: 16210838 DOI: 10.1159/000088810] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Accepted: 08/24/2005] [Indexed: 12/20/2022]
Abstract
Type 2 diabetes mellitus has reached epidemic proportions and diabetic nephropathy is the leading cause of end-stage renal disease. The metabolic syndrome constitutes a milieu conducive to tissue redox stress. This loss of redox homeostasis contributes to renal remodeling and parallels the concurrent increased vascular redox stress associated with the cardiometabolic syndrome. The multiple metabolic toxicities, redox stress and endothelial dysfunction combine to weave the complicated mosaic fabric of diabetic glomerulosclerosis and diabetic nephropathy. A better understanding may provide both the clinician and researcher tools to unravel this complicated disease process. Cellular remodeling of podocyte foot processes in the Ren-2 transgenic rat model of tissue angiotensin II overexpression (TG(mREN-2)27) and the Zucker diabetic fatty model of type 2 diabetes mellitus have been observed in preliminary studies. Importantly, angiotensin II receptor blockers have been shown to abrogate these ultrastructural changes in the foot processes of the podocyte in preliminary studies. An integrated, global risk reduction, approach in therapy addressing the multiple metabolic abnormalities combined with attempts to reach therapeutic goals at an earlier stage could have a profound effect on the development and progressive nature to end-stage renal disease and ultimately renal replacement therapy.
Collapse
Affiliation(s)
- Melvin R Hayden
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Diabetes and Cardiovascular Disease Center, University of Missouri School of Medicine, Columbia, 65212, USA.
| | | | | |
Collapse
|
50
|
Vattimo MDFF, Santos OFP. Functional interface between cathepsins and growth factors in the kidney development. Ren Fail 2005; 27:615-22. [PMID: 16153003 DOI: 10.1080/08860220500200486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
During kidney development many proteases are involved with the remodeling process of the extracellular matrix (ECM) during nephrogenesis. This study used embryonic kidneys culture, tridimensional cell culture, and reverse transcriptase-polymerase chain reaction (RT-PCR) techniques in order to investigate the expression of cathepsins S (CS) and cathepsin H (CH) during metanephrogenesis and their functional interface with hepatic growth factor (HGF) and nerve growth factor (NGF). Results have shown that cathepsin S has been expressed early than the cathepsin H in the nephrogenesis. NGF antibody in the embryonic kidney cultures, in a dose-dependent mechanism inhibited the CS but not CH genic expression by RT-PCR. The tridimensional cells culture with MDCK and IMCD cells confirmed the interface between HGF and CS and CH once their inhibitors added to the culture, reduced the fancy branching formation induced by this growth factor. In summary, this study suggests that CS and CH are differently expressed during nephrogenesis and also that they are involved with the tubulogenesis probably mediating specific growth factors such as NGF and HGF.
Collapse
|