1
|
Zhu J, Xiang X, Shi L, Song Z, Dong Z. Identification of Differentially Expressed Genes in Cold Storage-associated Kidney Transplantation. Transplantation 2024; 108:2057-2071. [PMID: 38632678 PMCID: PMC11424274 DOI: 10.1097/tp.0000000000005016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
BACKGROUND Although it is acknowledged that ischemia-reperfusion injury is the primary pathology of cold storage-associated kidney transplantation, its underlying mechanism is not well elucidated. METHODS To extend the understanding of molecular events and mine hub genes posttransplantation, we performed bulk RNA sequencing at different time points (24 h, day 7, and day 14) on a murine kidney transplantation model with prolonged cold storage (10 h). RESULTS In the present study, we showed that genes related to the regulation of apoptotic process, DNA damage response, cell cycle/proliferation, and inflammatory response were steadily elevated at 24 h and day 7. The upregulated gene profiling delicately transformed to extracellular matrix organization and fibrosis at day 14. It is prominent that metabolism-associated genes persistently took the first place among downregulated genes. The gene ontology terms of particular note to enrich are fatty acid oxidation and mitochondria energy metabolism. Correspondingly, the key enzymes of the above processes were the products of hub genes as recognized. Moreover, we highlighted the proximal tubular cell-specific increased genes at 24 h by combining the data with public RNA-Seq performed on proximal tubules. We also focused on ferroptosis-related genes and fatty acid oxidation genes to show profound gene dysregulation in kidney transplantation. CONCLUSIONS The comprehensive characterization of transcriptomic analysis may help provide diagnostic biomarkers and therapeutic targets in kidney transplantation.
Collapse
Affiliation(s)
- Jiefu Zhu
- Department of Urology and Department of Transplantation, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaohong Xiang
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Lang Shi
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhixia Song
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People’s Hospital of Yichang, Yichang, Hubei 443000, China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veteran Affairs (VA) Medical Center, Augusta, GA, United States
| |
Collapse
|
2
|
Charalampous C, Dasari S, McPhail E, Theis JD, Vrana JA, Dispenzieri A, Leung N, Muchtar E, Gertz M, Ramirez-Alvarado M, Kourelis T. A proteomic atlas of kidney amyloidosis provides insights into disease pathogenesis. Kidney Int 2024; 105:484-495. [PMID: 38096952 PMCID: PMC10922603 DOI: 10.1016/j.kint.2023.11.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 10/04/2023] [Accepted: 11/05/2023] [Indexed: 01/18/2024]
Abstract
The mechanisms of tissue damage in kidney amyloidosis are not well described. To investigate this further, we used laser microdissection-mass spectrometry to identify proteins deposited in amyloid plaques (expanded proteome) and proteins overexpressed in plaques compared to controls (plaque-specific proteome). This study encompassed 2650 cases of amyloidosis due to light chain (AL), heavy chain (AH), leukocyte chemotactic factor-2-type (ALECT2), secondary (AA), fibrinogen (AFib), apo AIV (AApoAIV), apo CII (AApoCII) and 14 normal/disease controls. We found that AFib, AA, and AApoCII have the most distinct proteomes predominantly driven by increased complement pathway proteins. Clustering of cases based on the expanded proteome identified two ALECT2 and seven AL subtypes. The main differences within the AL and ALECT2 subtypes were driven by complement proteins and, for AL only, 14-3-3 family proteins (a family of structurally similar phospho-binding proteins that regulate major cellular functions) widely implicated in kidney tissue dysfunction. The kidney AL plaque-specific proteome consisted of 24 proteins, including those implicated in kidney damage (α1 antitrypsin and heat shock protein β1). Hierarchical clustering of AL cases based on their plaque-specific proteome identified four clusters, of which one was associated with improved kidney survival and was characterized by higher overall proteomic content and 14-3-3 proteins but lower levels of light chains and most signature proteins. Thus, our results suggest that there is significant heterogeneity across and within amyloid types, driven predominantly by complement proteins, and that the plaque protein burden does not correlate with amyloid toxicity.
Collapse
Affiliation(s)
| | - Surendra Dasari
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Ellen McPhail
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jason D Theis
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Julie A Vrana
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Nelson Leung
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eli Muchtar
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Morie Gertz
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | |
Collapse
|
3
|
Hadpech S, Thongboonkerd V. Epithelial-mesenchymal plasticity in kidney fibrosis. Genesis 2024; 62:e23529. [PMID: 37345818 DOI: 10.1002/dvg.23529] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 06/23/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is an important biological process contributing to kidney fibrosis and chronic kidney disease. This process is characterized by decreased epithelial phenotypes/markers and increased mesenchymal phenotypes/markers. Tubular epithelial cells (TECs) are commonly susceptible to EMT by various stimuli, for example, transforming growth factor-β (TGF-β), cellular communication network factor 2, angiotensin-II, fibroblast growth factor-2, oncostatin M, matrix metalloproteinase-2, tissue plasminogen activator (t-PA), plasmin, interleukin-1β, and reactive oxygen species. Similarly, glomerular podocytes can undergo EMT via these stimuli and by high glucose condition in diabetic kidney disease. EMT of TECs and podocytes leads to tubulointerstitial fibrosis and glomerulosclerosis, respectively. Signaling pathways involved in EMT-mediated kidney fibrosis are diverse and complex. TGF-β1/Smad and Wnt/β-catenin pathways are the major venues triggering EMT in TECs and podocytes. These two pathways thus serve as the major therapeutic targets against EMT-mediated kidney fibrosis. To date, a number of EMT inhibitors have been identified and characterized. As expected, the majority of these EMT inhibitors affect TGF-β1/Smad and Wnt/β-catenin pathways. In addition to kidney fibrosis, these EMT-targeted antifibrotic inhibitors are expected to be effective for treatment against fibrosis in other organs/tissues.
Collapse
Affiliation(s)
- Sudarat Hadpech
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
4
|
The serine protease plasmin plays detrimental roles in epithelial sodium channel activation and podocyte injury in Dahl salt-sensitive rats. Hypertens Res 2023; 46:50-62. [PMID: 36241707 DOI: 10.1038/s41440-022-01064-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/06/2022] [Accepted: 09/26/2022] [Indexed: 02/03/2023]
Abstract
Salt-sensitive hypertension is associated with poor clinical outcomes. The epithelial sodium channel (ENaC) in the kidney plays pivotal roles in sodium reabsorption and blood pressure regulation, in which its γ subunit is activated by extracellular serine proteases. In proteinuric nephropathies, plasmin filtered through injured glomeruli reportedly activates γENaC in the distal nephron and causes podocyte injury. We previously reported that Dahl salt-sensitive (DS) rats fed a high-salt (HS) diet developed hypertension and proteinuria along with γENaC activation and that a synthetic serine protease inhibitor, camostat mesilate, mitigated these changes. However, the role of plasmin in DS rats remained unclear. In this study, we evaluated the relationship between plasmin and hypertension as well as podocyte injury and the effects of plasmin inhibitors in DS rats. Five-week-old DS rats were divided into normal-salt diet, HS diet, and HS+plasmin inhibitor (either tranexamic acid [TA] or synthetic plasmin inhibitor YO-2) groups. After blood pressure measurement and 24 h urine collection over 5 weeks, rats were sacrificed for biochemical analyses. The HS group displayed severe hypertension and proteinuria together with activation of plasmin in urine and γENaC in the kidney, which was significantly attenuated by YO-2 but not TA. YO-2 inhibited the attachment of plasmin(ogen) to podocytes and alleviated podocyte injury by inhibiting apoptosis and inflammatory/profibrotic cytokines. YO-2 also suppressed upregulation of protease-activated receptor-1 and phosphorylated ERK1/2. These results indicate an important role of plasmin in the development of salt-sensitive hypertension and related podocyte injury, suggesting plasmin inhibition as a potential therapeutic strategy.
Collapse
|
5
|
PP2A Catalytic Subunit α promotes fibroblast activation and kidney fibrosis via ERK pathway. Cell Signal 2021; 90:110187. [PMID: 34780974 DOI: 10.1016/j.cellsig.2021.110187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 11/22/2022]
Abstract
Protein Phosphatase 2A (PP2A), a main serine/threonine phosphatase, plays a profibrotic role in the development of different organs. However, the role and mechanisms of PP2Acα in fibroblast activation and kidney fibrosis are not fully known. Here we found that PP2Acα expression was upregulated in kidney tissue of chronic kidney disease (CKD) patients and unilateral ureter obstructive (UUO) mice. Ablation of fibroblast PP2Acα alleviates fibroblast activation and kidney fibrosis in mouse kidneys with UUO nephropathy compared with the control littermates. In primary cultured fibroblasts, PP2Acα deletion restrains TGFβ1-induced fibroblast activation, which is accompanied by increased phosphorylation of the extracellular regulated kinase (ERK). Blocking ERK pathway activation by PD98059 could promote fibroblast activation, indicating that PP2Acα promotes TGFβ1-induced fibroblast activation via suppressing ERK pathway. Consistently, in vivo, the activation of ERK pathway was upregulated by PP2Acα ablation in kidney fibroblasts. Together, these data uncover that PP2Acα may promote fibroblast activation and kidney fibrosis via suppressing ERK pathway, suggesting that targeting PP2Acα may provide a therapeutic effect for CKD.
Collapse
|
6
|
Interplay between extracellular matrix components and cellular and molecular mechanisms in kidney fibrosis. Clin Sci (Lond) 2021; 135:1999-2029. [PMID: 34427291 DOI: 10.1042/cs20201016] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) is characterized by pathological accumulation of extracellular matrix (ECM) proteins in renal structures. Tubulointerstitial fibrosis is observed in glomerular diseases as well as in the regeneration failure of acute kidney injury (AKI). Therefore, finding antifibrotic therapies comprises an intensive research field in Nephrology. Nowadays, ECM is not only considered as a cellular scaffold, but also exerts important cellular functions. In this review, we describe the cellular and molecular mechanisms involved in kidney fibrosis, paying particular attention to ECM components, profibrotic factors and cell-matrix interactions. In response to kidney damage, activation of glomerular and/or tubular cells may induce aberrant phenotypes characterized by overproduction of proinflammatory and profibrotic factors, and thus contribute to CKD progression. Among ECM components, matricellular proteins can regulate cell-ECM interactions, as well as cellular phenotype changes. Regarding kidney fibrosis, one of the most studied matricellular proteins is cellular communication network-2 (CCN2), also called connective tissue growth factor (CTGF), currently considered as a fibrotic marker and a potential therapeutic target. Integrins connect the ECM proteins to the actin cytoskeleton and several downstream signaling pathways that enable cells to respond to external stimuli in a coordinated manner and maintain optimal tissue stiffness. In kidney fibrosis, there is an increase in ECM deposition, lower ECM degradation and ECM proteins cross-linking, leading to an alteration in the tissue mechanical properties and their responses to injurious stimuli. A better understanding of these complex cellular and molecular events could help us to improve the antifibrotic therapies for CKD.
Collapse
|
7
|
Andersen H, Hansen MH, Buhl KB, Stæhr M, Friis UG, Enggaard C, Supramaniyam S, Lund IK, Svenningsen P, Hansen PBL, Jensen BL. Plasminogen Deficiency and Amiloride Mitigate Angiotensin II-Induced Hypertension in Type 1 Diabetic Mice Suggesting Effects Through the Epithelial Sodium Channel. J Am Heart Assoc 2020; 9:e016387. [PMID: 33215566 PMCID: PMC7763785 DOI: 10.1161/jaha.120.016387] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Diabetic nephropathy is a common diabetes mellitus complication associated with hypertension, proteinuria, and excretion of urinary plasmin that activates the epithelial sodium channel, ENaC, in vitro. Here we hypothesized that the deletion of plasminogen and amiloride treatment protect against hypertension in diabetes mellitus. Methods and Results Male plasminogen knockout (plasminogen-deficient [Plg-/-]) and wild-type mice were rendered diabetic with streptozotocin. Arterial blood pressure was recorded continuously by indwelling catheters before and during 10 days of angiotensin II infusion (ANGII; 30-60 ng/kg per minute). The effect of amiloride infusion (2 mg/kg per day, 4 days) was tested in wild-type, diabetic ANGII-treated mice. Streptozotocin increased plasma and urine glucose concentrations and 24-hour urine albumin and plasminogen excretion. Diabetic Plg-/- mice displayed larger baseline albuminuria and absence of urine plasminogen. Baseline mean arterial blood pressure did not differ between groups. Although ANGII elevated blood pressure in wild-type, diabetic wild-type, and Plg-/- control mice, ANGII did not change blood pressure in diabetic Plg-/- mice. Compared with ANGII infusion alone, wild-type ANGII-infused diabetic mice showed blood pressure reduction upon amiloride treatment. There was no difference in plasma renin, ANGII, aldosterone, tissue prorenin receptor, renal inflammation, and fibrosis between groups. Urine from wild-type mice evoked larger amiloride-sensitive current than urine from Plg-/- mice with or without diabetes mellitus. Full-length γ-ENaC and α-ENaC subunit abundances were not changed in kidney homogenates, but the 70 kDa γ-ENaC cleavage product was increased in diabetic versus nondiabetic mice. Conclusions Plasmin promotes hypertension in diabetes mellitus with albuminuria likely through the epithelial sodium channel.
Collapse
Affiliation(s)
- Henrik Andersen
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Maria Høj Hansen
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Kristian B Buhl
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Mette Stæhr
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Ulla G Friis
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Camilla Enggaard
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Shanya Supramaniyam
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Ida K Lund
- The Finsen Laboratory Copenhagen University Hospital, and Biotech Research and Innovation Center (BRIC) University of Copenhagen Denmark
| | - Per Svenningsen
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Pernille B L Hansen
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark.,Research and Early Development Cardiovascular, Renal and Metabolism BioPharmaceuticals R&D AstraZeneca Gothenburg Sweden
| | - Boye L Jensen
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| |
Collapse
|
8
|
Rayego-Mateos S, Valdivielso JM. New therapeutic targets in chronic kidney disease progression and renal fibrosis. Expert Opin Ther Targets 2020; 24:655-670. [PMID: 32338087 DOI: 10.1080/14728222.2020.1762173] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The current therapeutic armamentarium to prevent chronic kidney disease (CKD) progression is limited to the control of blood pressure and in diabetic patients, the strict control of glucose levels. Current research is primarily focused on the reduction of inflammation and fibrosis at different levels. AREAS COVERED This article examines the latest progress in this field and places an emphasis on inflammation, oxidative stress, and fibrosis. New therapeutic targets are described and evidence from experimental and clinical studies is summarized. We performed a search in Medline for articles published over the last 10 years. EXPERT OPINION The search for therapeutic targets of renal inflammation is hindered by an incomplete understanding of the pathophysiology. The determination of the specific inducers of inflammation in the kidney is an area of heightened potential. Prevention of the progression of renal fibrosis by blocking TGF-β signaling has been unsuccessful, but the investigation of signaling pathways involved in late stages of fibrosis progression could yield improved results. Preventive strategies such as the modification of microbiota-inducers of uremic toxins involved in CKD progression is a promising field because of the interaction between the gut microbiota and the renal system.
Collapse
Affiliation(s)
- Sandra Rayego-Mateos
- Red De Investigación Renal (Redinren) , Spain.,Vascular and Renal Translational Research Group, Institut De Recerca Biomèdica De Lleida IRBLleida , Lleida, Spain
| | - Jose M Valdivielso
- Red De Investigación Renal (Redinren) , Spain.,Vascular and Renal Translational Research Group, Institut De Recerca Biomèdica De Lleida IRBLleida , Lleida, Spain
| |
Collapse
|
9
|
Majumder S, Amin M, Pushpakumar S, Sen U. Collagen receptor- and metalloproteinase-dependent hypertensive stress response in mesangial and glomerular endothelial cells. Mol Cell Biochem 2020; 466:1-15. [PMID: 31912277 PMCID: PMC10809865 DOI: 10.1007/s11010-019-03680-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 12/22/2019] [Indexed: 12/25/2022]
Abstract
Progressive alteration of the extracellular matrix (ECM) is the characteristic of hypertensive nephropathy (HN). Both mesangial and endothelial cells have the ability to synthesize and degrade ECM components, including collagens through the activation of matrix metalloproteinases (MMPs) in stress conditions, such as in hypertension. On the other hand, hydrogen sulfide (H2S) has been shown to mitigate hypertensive renal matrix remodeling. Surprisingly, whether H2S ameliorates receptor-mediated (urokinase plasminogen activator receptor-associated protein, uPARAP/Endo180) collagen dysregulation in Ang-II hypertension is not clear. The purpose of this study was to determine whether Ang-II alters the expression of Endo180, tissue plasminogen activator (tPA), MMPs, and their tissue inhibitors (TIMPs) leading to the dysregulation of cellular collagen homeostasis and whether H2S mitigates the collagen turnover. Mouse mesangial cells (MCs) and glomerular endothelial cells (MGECs) were treated without or with Ang-II and H2S donor GYY (GYY4137) for 48 h. Cell lysates were analyzed by Western blot and RT-PCR, and cells were analyzed by immunocytochemistry. The results indicated that, while Ang-II differentially expressed MMP-13 and TIMP-1 in MCs and in MGECs, it predominantly decreased tPA, Endo 180, and increased plasminogen activator inhibitor-1 (PAI-1), MMP-14, and collagen IIIA and IV in both the cell types. Interestingly, H2S donor GYY treatment normalized the above changes in both the cell types. We conclude that Ang-II treatment causes ECM remodeling in MCs and MGECs through PAI-1/tPA/Endo180 and MMP/TIMP-dependent collagen remodeling, and H2S treatment mitigates remodeling, in part, by modulating these pathways.
Collapse
Affiliation(s)
- Suravi Majumder
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Matthew Amin
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Sathnur Pushpakumar
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Utpal Sen
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
10
|
Gupta S, Pepper RJ, Ashman N, Walsh SB. Nephrotic Syndrome: Oedema Formation and Its Treatment With Diuretics. Front Physiol 2019; 9:1868. [PMID: 30697163 PMCID: PMC6341062 DOI: 10.3389/fphys.2018.01868] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/11/2018] [Indexed: 01/01/2023] Open
Abstract
Oedema is a defining element of the nephrotic syndrome. Its' management varies considerably between clinicians, with no national or international clinical guidelines, and hence variable outcomes. Oedema may have serious sequelae such as immobility, skin breakdown and local or systemic infection. Treatment of nephrotic oedema is often of limited efficacy, with frequent side-effects and interactions with other pharmacotherapy. Here, we describe the current paradigms of oedema in nephrosis, including insights into emerging mechanisms such as the role of the abnormal activation of the epithelial sodium channel in the collecting duct. We then discuss the physiological basis for traditional and novel therapies for the treatment of nephrotic oedema. Despite being the cardinal symptom of nephrosis, few clinical studies guide clinicians to the rational use of therapy. This is reflected in the scarcity of publications in this field; it is time to undertake new clinical trials to direct clinical practice.
Collapse
Affiliation(s)
- Sanjana Gupta
- UCL Centre for Nephrology, University College London, London, United Kingdom.,Renal Unit, The Royal London Hospital, Bart's Health NHS Trust, London, United Kingdom
| | - Ruth J Pepper
- UCL Centre for Nephrology, University College London, London, United Kingdom
| | - Neil Ashman
- Renal Unit, The Royal London Hospital, Bart's Health NHS Trust, London, United Kingdom
| | - Stephen B Walsh
- UCL Centre for Nephrology, University College London, London, United Kingdom
| |
Collapse
|
11
|
Waasdorp M, de Rooij DM, Florquin S, Duitman J, Spek CA. Protease-activated receptor-1 contributes to renal injury and interstitial fibrosis during chronic obstructive nephropathy. J Cell Mol Med 2018; 23:1268-1279. [PMID: 30485646 PMCID: PMC6349177 DOI: 10.1111/jcmm.14028] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/05/2018] [Accepted: 10/24/2018] [Indexed: 12/17/2022] Open
Abstract
End‐stage renal disease, the final stage of all chronic kidney disorders, is associated with renal fibrosis and inevitably leads to renal failure and death. Transition of tubular epithelial cells (TECs) into mesenchymal fibroblasts constitutes a proposed mechanism underlying the progression of renal fibrosis and here we assessed whether protease‐activated receptor (PAR)‐1, which recently emerged as an inducer of epithelial‐to‐mesenchymal transition (EMT), aggravates renal fibrosis. We show that PAR‐1 activation on TECs reduces the expression of epithelial markers and simultaneously induces mesenchymal marker expression reminiscent of EMT. We next show that kidney damage was reduced in PAR‐1‐deficient mice during unilateral ureter obstruction (UUO) and that PAR‐1‐deficient mice develop a diminished fibrotic response. Importantly, however, we did hardly observe any signs of mesenchymal transition in both wild‐type and PAR‐1‐deficient mice suggesting that diminished fibrosis in PAR‐1‐deficient mice is not due to reduced EMT. Instead, the accumulation of macrophages and fibroblasts was significantly reduced in PAR‐1‐deficient animals which were accompanied by diminished production of MCP‐1 and TGF‐β. Overall, we thus show that PAR‐1 drives EMT of TECs in vitro and aggravates UUO‐induced renal fibrosis although this is likely due to PAR‐1‐dependent pro‐fibrotic cytokine production rather than EMT.
Collapse
Affiliation(s)
- Maaike Waasdorp
- Center for Experimental and Molecular Medicine, Academic Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Dennis M de Rooij
- Center for Experimental and Molecular Medicine, Academic Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Sandrine Florquin
- Pathology, Academic Medical Center Amsterdam, Amsterdam, The Netherlands
| | - JanWillem Duitman
- Center for Experimental and Molecular Medicine, Academic Medical Center Amsterdam, Amsterdam, The Netherlands.,Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation and Remodeling) and LabEx Inflamex, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Physiopathologie Et Epidémiologie Des Maladies Respiratoires, Medical School Xavier Bichat, Inserm UMR1152, Paris, France
| | - C Arnold Spek
- Center for Experimental and Molecular Medicine, Academic Medical Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Gonzalez-Calero L, Martínez PJ, Martin-Lorenzo M, Baldan-Martin M, Ruiz-Hurtado G, de la Cuesta F, Calvo E, Segura J, Lopez JA, Vázquez J, Barderas MG, Ruilope LM, Vivanco F, Alvarez-Llamas G. Urinary exosomes reveal protein signatures in hypertensive patients with albuminuria. Oncotarget 2018; 8:44217-44231. [PMID: 28562335 PMCID: PMC5546475 DOI: 10.18632/oncotarget.17787] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/20/2017] [Indexed: 01/05/2023] Open
Abstract
Albuminuria is an indicator of cardiovascular risk and renal damage in hypertensive individuals. Chronic renin-angiotensin system (RAS) suppression facilitates blood pressure control and prevents development of new-onset-albuminuria. A significant number of patients, however, develop albuminuria despite chronic RAS blockade, and the physiopathological mechanisms are underexplored. Urinary exosomes reflect pathological changes taking place in the kidney. The objective of this work was to examine exosomal protein alterations in hypertensive patients with albuminuria in the presence of chronic RAS suppression, to find novel clues underlying its development. Patients were followed-up for three years and were classified as: a) patients with persistent normoalbuminuria; b) patients developing de novo albuminuria; and c) patients with maintained albuminuria. Exosomal protein alterations between groups were identified by isobaric tag quantitation (iTRAQ). Confirmation was approached by target analysis (SRM). In total, 487 proteins were identified with high confidence. Specifically, 48 proteins showed an altered pattern in response to hypertension and/or albuminuria. Out of them, 21 proteins interact together in three main functional clusters: glycosaminoglycan degradation, coagulation and complement system, and oxidative stress. The identified proteins constitute potential targets for drug development and may help to define therapeutic strategies to evade albuminuria progression in hypertensive patients chronically treated.
Collapse
Affiliation(s)
| | - Paula J Martínez
- Department of Immunology, IIS-Fundacion Jimenez Diaz, REDinREN, Madrid, Spain
| | | | | | - Gema Ruiz-Hurtado
- Hypertension Unit, Instituto de Investigación Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando de la Cuesta
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos SESCAM, Toledo, Spain
| | | | - Julian Segura
- Hypertension Unit, Instituto de Investigación Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics CNIC, Madrid, Spain
| | - Maria G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos SESCAM, Toledo, Spain
| | - Luis M Ruilope
- Hypertension Unit, Instituto de Investigación Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando Vivanco
- Department of Immunology, IIS-Fundacion Jimenez Diaz, REDinREN, Madrid, Spain.,Department of Biochemistry and Molecular Biology I, Universidad Complutense, Madrid, Spain
| | | |
Collapse
|
13
|
Tabatabaeifar M, Wlodkowski T, Simic I, Denc H, Mollet G, Weber S, Moyers JJ, Brühl B, Randles MJ, Lennon R, Antignac C, Schaefer F. An inducible mouse model of podocin-mutation-related nephrotic syndrome. PLoS One 2017; 12:e0186574. [PMID: 29049388 PMCID: PMC5648285 DOI: 10.1371/journal.pone.0186574] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/10/2017] [Indexed: 12/03/2022] Open
Abstract
Mutations in the NPHS2 gene, encoding podocin, cause hereditary nephrotic syndrome. The most common podocin mutation, R138Q, is associated with early disease onset and rapid progression to end-stage renal disease. Knock-in mice carrying a R140Q mutation, the mouse analogue of human R138Q, show developmental arrest of podocytes and lethal renal failure at neonatal age. Here we created a conditional podocin knock-in model named NPHS2R140Q/-, using a tamoxifen-inducible Cre recombinase, which permits to study the effects of the mutation in postnatal life. Within the first week of R140Q hemizygosity induction the animals developed proteinuria, which peaked after 4–5 weeks. Subsequently the animals developed progressive renal failure, with a median survival time of 12 (95% CI: 11–13) weeks. Foot process fusion was observed within one week, progressing to severe and global effacement in the course of the disease. The number of podocytes per glomerulus gradually diminished to 18% compared to healthy controls 12–16 weeks after induction. The fraction of segmentally sclerosed glomeruli was 25%, 85% and 97% at 2, 4 and 8 weeks, respectively. Severe tubulointerstitial fibrosis was present at later disease stage and was correlated quantitatively with the level of proteinuria at early disease stages. While R140Q podocin mRNA expression was elevated, protein abundance was reduced by more than 50% within one week following induction. Whereas miRNA21 expression persistently increased during the first 4 weeks, miRNA-193a expression peaked 2 weeks after induction. In conclusion, the inducible R140Q-podocin mouse model is an auspicious model of the most common genetic cause of human nephrotic syndrome, with a spontaneous disease course strongly reminiscent of the human disorder. This model constitutes a valuable tool to test the efficacy of novel pharmacological interventions aimed to improve podocyte function and viability and attenuate proteinuria, glomerulosclerosis and progressive renal failure.
Collapse
Affiliation(s)
- Mansoureh Tabatabaeifar
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Tanja Wlodkowski
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Ivana Simic
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Helga Denc
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Geraldine Mollet
- INSERM, U1163, Imagine Institute, Laboratory of Hereditary Kidney Diseases, Paris, France
- Paris Descartes-Sorbonne Paris Cité University, Paris, France
| | - Stefanie Weber
- Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Philipps-University Marburg, Marburg, Germany
| | | | - Barbara Brühl
- Institute for Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Michael Joseph Randles
- Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester, United Kingdom
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Rachel Lennon
- Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester, United Kingdom
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Corinne Antignac
- INSERM, U1163, Imagine Institute, Laboratory of Hereditary Kidney Diseases, Paris, France
- Paris Descartes-Sorbonne Paris Cité University, Paris, France
- Department of Genetics, Necker Hospital, Assistance Publique—Hôpitaux de Paris, Paris, France
| | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
14
|
Svenningsen P, Hinrichs GR, Zachar R, Ydegaard R, Jensen BL. Physiology and pathophysiology of the plasminogen system in the kidney. Pflugers Arch 2017; 469:1415-1423. [DOI: 10.1007/s00424-017-2014-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/09/2017] [Accepted: 06/12/2017] [Indexed: 12/31/2022]
|
15
|
Cheng Z, Limbu MH, Wang Z, Liu J, Liu L, Zhang X, Chen P, Liu B. MMP-2 and 9 in Chronic Kidney Disease. Int J Mol Sci 2017; 18:ijms18040776. [PMID: 28397744 PMCID: PMC5412360 DOI: 10.3390/ijms18040776] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/21/2017] [Accepted: 03/31/2017] [Indexed: 01/17/2023] Open
Abstract
Gelatinases are members of the matrix metalloproteinase (MMPs) family; they play an important role in the degradation of the extracellular matrix (ECM). This effect is also crucial in the development and progression of chronic kidney disease (CKD). Its expression, as well as its activity regulation are closely related to the cell signaling pathways, hypoxia and cell membrane structural change. Gelatinases also can affect the development and progression of CKD through the various interactions with tumor necrosis factors (TNFs), monocyte chemoattractant proteins (MCPs), growth factors (GFs), oxidative stress (OS), and so on. Currently, their non-proteolytic function is a hot topic of research, which may also be associated with the progression of CKD. Therefore, with the in-depth understanding about the function of gelatinases, we can have a more specific and accurate understanding of their role in the human body.
Collapse
Affiliation(s)
- Zhengyuan Cheng
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing 210009, China.
| | - Manoj Hang Limbu
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing 210009, China.
| | - Zhi Wang
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing 210009, China.
| | - Jing Liu
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing 210009, China.
| | - Lei Liu
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing 210009, China.
| | - Xiaoyi Zhang
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing 210009, China.
| | - Pingsheng Chen
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing 210009, China.
| | - Bicheng Liu
- Department of Nephrology, Zhongda Hospital, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing 210009, China.
| |
Collapse
|
16
|
Lin C, Borensztajn K, Spek CA. Targeting coagulation factor receptors - protease-activated receptors in idiopathic pulmonary fibrosis. J Thromb Haemost 2017; 15:597-607. [PMID: 28079978 DOI: 10.1111/jth.13623] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Indexed: 12/11/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease with a 5-year mortality rate of > 50% and unknown etiology. Treatment options remain limited and, currently, only two drugs are available, i.e. nintedanib and pirfenidone. However, both of these antifibrotic agents only slow down the progression of the disease, and do not remarkably prolong the survival of IPF patients. Hence, the discovery of new therapeutic targets for IPF is crucial. Studies exploring the mechanisms that are involved in IPF have identified several possible targets for therapeutic interventions. Among these, blood coagulation factor receptors, i.e. protease-activated receptors (PARs), are key candidates, as these receptors mediate the cellular effects of coagulation factors and play central roles in influencing inflammatory and fibrotic responses. In this review, we will focus on the controversial role of the coagulation cascade in the pathogenesis of IPF. In the light of novel data, we will attempt to reconciliate the apparently conflicting data and discuss the possibility of pharmacologic targeting of PARs for the treatment of fibroproliferative diseases.
Collapse
Affiliation(s)
- C Lin
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - K Borensztajn
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
- Inserm UMR1152, Medical School Xavier Bichat, Paris, France
- Département Hospitalo-universtaire FIRE (Fibrosis, Inflammation and Remodeling) and LabEx Inflamex, Paris, France
| | - C A Spek
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
17
|
Waasdorp M, Duitman J, Spek CA. Plasmin reduces fibronectin deposition by mesangial cells in a protease-activated receptor-1 independent manner. Biochem Biophys Rep 2017; 10:152-156. [PMID: 29114573 PMCID: PMC5637235 DOI: 10.1016/j.bbrep.2017.03.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/17/2017] [Accepted: 03/28/2017] [Indexed: 12/31/2022] Open
Abstract
Background Protease-activated receptor-1 (PAR-1) potentiates diabetic nephropathy (DN) as evident from reduced kidney injury in diabetic PAR-1 deficient mice. Although thrombin is the prototypical PAR-1 agonist, anticoagulant treatment does not limit DN in experimental animal models suggesting that thrombin is not the endogenous PAR-1 agonist driving DN. Objectives To identify the endogenous PAR-1 agonist potentiating diabetes-induced nephropathy. Methods Unbiased protease expression profiling in glomeruli from human kidneys with DN was performed using publically available microarray data. The identified prime candidate PAR-1 agonist was subsequently analysed for PAR-1-dependent induction of fibrosis in vitro. Results Of the 553 proteases expressed in the human genome, 247 qualified as potential PAR-1 agonists of which 71 were significantly expressed above background in diabetic glomeruli. The recently identified PAR-1 agonist plasmin(ogen), together with its physiological activator tissue plasminogen activator, were among the highest expressed proteases. Plasmin did however not induce mesangial proliferation and/or fibronectin deposition in vitro. In a PAR-1 independent manner, plasmin even reduced fibronectin deposition. Conclusion Expression profiling identified plasmin as potential endogenous PAR-1 agonist driving DN. Instead of inducing fibronectin expression, plasmin however reduced mesangial fibronectin deposition in vitro. Therefore we conclude that plasmin may not be the endogenous PAR-1 agonist potentiating DN. Plasmin is highly expressed in kidneys of diabetic nephropathy patients. Plasmin limits fibronectin deposition by mesangial cells. Plasmin-dependent PAR-1 activation does not drive diabetic nephropathy.
Collapse
Affiliation(s)
- Maaike Waasdorp
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam 1105 AZ, The Netherlands
| | - JanWillem Duitman
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam 1105 AZ, The Netherlands.,INSERM, UMR1152, Medical School Xavier Bichat, Paris, France.,Paris Diderot University, Sorbonne Paris Cité, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation and Remodeling), LabEx Inflamex, Paris, France
| | - C Arnold Spek
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam 1105 AZ, The Netherlands
| |
Collapse
|
18
|
Cruz-Solbes AS, Youker K. Epithelial to Mesenchymal Transition (EMT) and Endothelial to Mesenchymal Transition (EndMT): Role and Implications in Kidney Fibrosis. Results Probl Cell Differ 2017; 60:345-372. [PMID: 28409352 DOI: 10.1007/978-3-319-51436-9_13] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Tubulointerstitial injury is one of the hallmarks of renal disease. In particular, interstitial fibrosis has a prominent role in the development and progression of kidney injury. Collagen-producing fibroblasts are responsible for the ECM deposition. However, the origin of those activated fibroblasts is not clear. This chapter will discuss in detail the concept of epithelial to mesenchymal transition (EMT) and endothelial to mesenchymal transition (EndMT) in the context of fibrosis and kidney disease. In short, EMT and EndMT involve a change in cell shape, loss of polarity and increased motility associated with increased collagen production. Thus, providing a new source of fibroblasts. However, many controversies exist regarding the existence of EMT and EndMT in kidney disease, as well as its burden and role in disease development. The aim of this chapter is to provide an overview of the concepts and profibrotic pathways and to present the evidence that has been published in favor and against EMT and EndMT.
Collapse
|
19
|
Sutariya B, Jhonsa D, Saraf MN. TGF-β: the connecting link between nephropathy and fibrosis. Immunopharmacol Immunotoxicol 2016; 38:39-49. [PMID: 26849902 DOI: 10.3109/08923973.2015.1127382] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Renal fibrosis is the usual outcome of an excessive accumulation of extracellular matrix (ECM) that frequently occurs in membranous and diabetic nephropathy. The result of renal fibrosis would be end-stage renal failure, which requires costly dialysis or kidney transplantation. Renal fibrosis typically results from chronic inflammation via production of several molecules, such as growth factors, angiogenic factors, fibrogenic cytokines, and proteinase. All of these factors can stimulate excessive accumulation of ECM components through epithelial to mesenchymal transition (EMT), which results in renal fibrosis. Among these, transforming growth factor-beta (TGF-β) is proposed to be the major regulator in inducing EMT. Besides ECM protein synthesis, TGF-β is involved in hypertrophy, proliferation, and apoptosis in renal cells. In particular, TGF-β is likely to be most potent and ubiquitous profibrotic factor acting through several intracellular signaling pathways including protein kinases and transcription factors. Factors that regulate TGF-β expression in renal cell include hyperglycemia, angiotensin II, advance glycation end products, complement activation (C5b-9), and oxidative stress. Over the past several years, the common understanding of the pathogenic factors that lead to renal fibrosis in nephropathy has improved considerably. This review will discuss the recent findings on the mechanisms and role of TGF-β in membranous and diabetic nephropathy.
Collapse
Affiliation(s)
- Brijesh Sutariya
- a Department of Pharmacology , Bombay College of Pharmacy , Mumbai , Maharashtra , India
| | - Dimple Jhonsa
- a Department of Pharmacology , Bombay College of Pharmacy , Mumbai , Maharashtra , India
| | - Madhusudan N Saraf
- a Department of Pharmacology , Bombay College of Pharmacy , Mumbai , Maharashtra , India
| |
Collapse
|
20
|
Kakizoe Y, Miyasato Y, Onoue T, Nakagawa T, Hayata M, Uchimura K, Morinaga J, Mizumoto T, Adachi M, Miyoshi T, Sakai Y, Tomita K, Mukoyama M, Kitamura K. A serine protease inhibitor attenuates aldosterone-induced kidney injuries via the suppression of plasmin activity. J Pharmacol Sci 2016; 132:145-153. [DOI: 10.1016/j.jphs.2016.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 09/15/2016] [Accepted: 09/19/2016] [Indexed: 11/28/2022] Open
|
21
|
Affiliation(s)
- David G. Warnock
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
22
|
Liew K, Yong PVC, Navaratnam V, Lim YM, Ho ASH. Differential proteomic analysis on the effects of 2-methoxy-1,4-naphthoquinone towards MDA-MB-231 cell line. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:517-527. [PMID: 25981917 DOI: 10.1016/j.phymed.2015.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/31/2015] [Accepted: 03/05/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND We have previously reported the anti-metastatic effects of 2-methoxy-1,4-naphthoquinone (MNQ) against MDA-MB-231 cell line. PURPOSE To investigate the molecular mechanism underlying the anti-metastatic effects of MNQ towards MDA-MB-231 cell line via the comparative proteomic approach. STUDY DESIGN/METHODS Differentially expressed proteins in MNQ-treated MDA-MB-231 cells were identified by using two-dimensional gel electrophoresis coupled with tandem mass spectrometry. Proteins and signalling pathways associated with the identified MNQ-altered proteins were studied by using Western blotting. RESULTS Significant modulation of MDA-MB-231 cell proteome was observed upon treatment with MNQ in which the expressions of 19 proteins were found to be downregulated whereas another eight were upregulated (>1.5 fold, p < 0.05). The altered proteins were mainly related to cytoskeletal functions and regulations, mRNA processing, protein modifications and oxidative stress response. Notably, two of the downregulated proteins, protein S100-A4 (S100A4) and laminin-binding protein (RPSA) are known to play key roles in driving metastasis and were verified using Western blotting. Further investigation using Western blotting also revealed that MNQ decreased the activations of pro-metastatic ERK1/2 and NF-κB signalling pathways. Moreover, MNQ was shown to stimulate the expression of the metastatic suppressor, E-cadherin. CONCLUSION This study reports a proposed mechanism by which MNQ exerts its anti-metastatic effects against MDA-MB-231 cell line. The findings from this study offer new insights on the potential of MNQ to be developed as a novel anti-metastatic agent.
Collapse
Affiliation(s)
- Kitson Liew
- School of Biosciences, Taylor's University, No.1 Jalan Taylor's, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Phelim Voon Chen Yong
- School of Biosciences, Taylor's University, No.1 Jalan Taylor's, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Visweswaran Navaratnam
- School of Biosciences, Taylor's University, No.1 Jalan Taylor's, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Yang Mooi Lim
- Department of Pre-Clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Lot PT21144, Jalan Sungai Long, Bandar Sungai Long, 43000 Kajang, Selangor Darul Ehsan, Malaysia.
| | - Anthony Siong Hock Ho
- School of Biosciences, Taylor's University, No.1 Jalan Taylor's, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
23
|
Uchida T, Oda T, Takechi H, Matsubara H, Watanabe A, Yamamoto K, Oshima N, Sakurai Y, Kono T, Shimazaki H, Tamai S, Kumagai H. Role of tubulointerstitial plasmin in the progression of IgA nephropathy. J Nephrol 2015; 29:53-62. [PMID: 25971850 DOI: 10.1007/s40620-015-0205-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 04/30/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND Plasmin has recently been reported to be associated with renal fibrosis in experimental models, but its role in human renal diseases is unclear. METHODS Fifty-seven patients with IgA nephropathy (IgAN) were evaluated retrospectively. Plasmin in their renal biopsy tissues was assessed by in situ zymography using a plasmin-sensitive synthetic peptide, and the relationships between patients' histologic or clinical parameters and their renal plasmin activity [assessed semiquantitatively by calculating the positively stained percentage of the total tubulointerstitial (TI) area] were evaluated. RESULTS Plasmin activity was observed almost exclusively in the TI space (mainly in the interstitium and partly in the tubular epithelial cells) and was significantly stronger in patients with TI lesion (tubular atrophy/interstitial fibrosis and tubulointerstitial inflammation) than in those without TI lesion. It was significantly and positively correlated with the global glomerulosclerosis rate and significantly and negatively correlated with estimated glomerular filtration rate not only at the time of renal biopsy but also at the end of the follow-up period. Double stainings for plasmin activity and inflammatory cells, cytokeratin, or α-smooth muscle actin (α-SMA) in selected patients revealed TI infiltration of inflammatory cells, attenuated tubular epithelial expression of cytokeratin, and augmented interstitial expression of α-SMA close to upregulated plasmin activity in the TI space. CONCLUSIONS These data suggest that TI plasmin is associated with TI inflammation leading to renal fibrosis, and can cause the decline in renal function seen in patients with IgAN. Reducing plasmin in situ may therefore be a promising therapeutic approach slowing renal fibrogenesis and improving renal function.
Collapse
Affiliation(s)
- Takahiro Uchida
- Department of Nephrology and Endocrinology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | - Takashi Oda
- Department of Nephrology, Tokyo Medical University Hachioji Medical Center, Hachioji, Tokyo, Japan
| | - Hanako Takechi
- Department of Nephrology and Endocrinology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Hidehito Matsubara
- Department of Nephrology and Endocrinology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Atsushi Watanabe
- Department of Nephrology and Endocrinology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Kojiro Yamamoto
- Department of Nephrology and Endocrinology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Naoki Oshima
- Department of Nephrology and Endocrinology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yutaka Sakurai
- Department of Preventive Medicine and Public Health, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Takako Kono
- Department of Laboratory Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hideyuki Shimazaki
- Department of Laboratory Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Seiichi Tamai
- Department of Laboratory Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hiroo Kumagai
- Department of Nephrology and Endocrinology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| |
Collapse
|
24
|
Darehzereshki A, Rubin N, Gamba L, Kim J, Fraser J, Huang Y, Billings J, Mohammadzadeh R, Wood J, Warburton D, Kaartinen V, Lien CL. Differential regenerative capacity of neonatal mouse hearts after cryoinjury. Dev Biol 2015; 399:91-99. [PMID: 25555840 PMCID: PMC4339535 DOI: 10.1016/j.ydbio.2014.12.018] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 12/09/2014] [Accepted: 12/10/2014] [Indexed: 01/05/2023]
Abstract
Neonatal mouse hearts fully regenerate after ventricular resection similar to adult zebrafish. We established cryoinjury models to determine if different types and varying degrees of severity in cardiac injuries trigger different responses in neonatal mouse hearts. In contrast to ventricular resection, neonatal mouse hearts fail to regenerate and show severe impairment of cardiac function post transmural cryoinjury. However, neonatal hearts fully recover after non-transmural cryoinjury. Interestingly, cardiomyocyte proliferation does not significantly increase in neonatal mouse hearts after cryoinjuries. Epicardial activation and new coronary vessel formation occur after cryoinjury. The profibrotic marker PAI-1 is highly expressed after transmural but not non-transmural cryoinjuries, which may contribute to the differential scarring. Our results suggest that regenerative medicine strategies for heart injuries should vary depending on the nature of the injury.
Collapse
Affiliation(s)
- Ali Darehzereshki
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - Nicole Rubin
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - Laurent Gamba
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - Jieun Kim
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - James Fraser
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - Ying Huang
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - Joshua Billings
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - Robabeh Mohammadzadeh
- Broad Center of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, CA
| | - John Wood
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California; Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - David Warburton
- The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California; Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Vesa Kaartinen
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan
| | - Ching-Ling Lien
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California; Department of Surgery, Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA.
| |
Collapse
|
25
|
Carmo AAF, Costa BRC, Vago JP, de Oliveira LC, Tavares LP, Nogueira CRC, Ribeiro ALC, Garcia CC, Barbosa AS, Brasil BSAF, Dusse LM, Barcelos LS, Bonjardim CA, Teixeira MM, Sousa LP. Plasmin induces in vivo monocyte recruitment through protease-activated receptor-1-, MEK/ERK-, and CCR2-mediated signaling. THE JOURNAL OF IMMUNOLOGY 2014; 193:3654-63. [PMID: 25165151 DOI: 10.4049/jimmunol.1400334] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The plasminogen (Plg)/plasmin (Pla) system is associated with a variety of biological activities beyond the classical dissolution of fibrin clots, including cell migration, tissue repair, and inflammation. Although the capacity of Plg/Pla to induce cell migration is well defined, the mechanism underlying this process in vivo is elusive. In this study, we show that Pla induces in vitro migration of murine fibroblasts and macrophages (RAW 264.7) dependent on the MEK/ERK pathway and by requiring its proteolytic activity and lysine binding sites. Plasmin injection into the pleural cavity of BALB/c mice induced a time-dependent influx of mononuclear cells that was associated with augmented ERK1/2 and IκB-α phosphorylation and increased levels of CCL2 and IL-6 in pleural exudates. The inhibition of protease activity by using a serine protease inhibitor leupeptin or two structurally different protease-activated receptor-1 antagonists (SCH79797 and RWJ56110) abolished Pla-induced mononuclear recruitment and ERK1/2 and IκB-α phosphorylation. Interestingly, inhibition of the MEK/ERK pathway abolished Pla-induced CCL2 upregulation and mononuclear cell influx. In agreement with a requirement for the CCL2/CCR2 axis to Pla-induced cell migration, the use of a CCR2 antagonist (RS504393) prevented the Plg/Pla-induced recruitment of mononuclear cells to the pleural cavity and migration of macrophages at transwell plates. Therefore, Pla-induced mononuclear cell recruitment in vivo was dependent on protease-activated receptor-1 activation of the MEK/ERK/NF-κB pathway, which led to the release of CCL2 and activation of CCR2.
Collapse
Affiliation(s)
- Aline A F Carmo
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Programa de Pós-Graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Bruno R C Costa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Juliana P Vago
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Programa de Pós-Graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Leonardo C de Oliveira
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; and
| | - Luciana P Tavares
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Camila R C Nogueira
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Ana Luíza C Ribeiro
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Cristiana C Garcia
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Alan S Barbosa
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | | | - Luci M Dusse
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Lucíola S Barcelos
- Programa de Pós-Graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Cláudio A Bonjardim
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; and
| | - Mauro M Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil;
| | - Lirlândia P Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Programa de Pós-Graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil;
| |
Collapse
|
26
|
Joshi N, Kopec AK, Towery K, Williams KJ, Luyendyk JP. The antifibrinolytic drug tranexamic acid reduces liver injury and fibrosis in a mouse model of chronic bile duct injury. J Pharmacol Exp Ther 2014; 349:383-92. [PMID: 24633426 PMCID: PMC4019324 DOI: 10.1124/jpet.113.210880] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 03/13/2014] [Indexed: 01/24/2023] Open
Abstract
Hepatic fibrin deposition has been shown to inhibit hepatocellular injury in mice exposed to the bile duct toxicant α-naphthylisothiocyanate (ANIT). Degradation of fibrin clots by fibrinolysis controls the duration and extent of tissue fibrin deposition. Thus, we sought to determine the effect of treatment with the antifibrinolytic drug tranexamic acid (TA) and plasminogen activator inhibitor-1 (PAI-1) deficiency on ANIT-induced liver injury and fibrosis in mice. Plasmin-dependent lysis of fibrin clots was impaired in plasma from mice treated with TA (1200 mg/kg i.p., administered twice daily). Prophylactic TA administration reduced hepatic inflammation and hepatocellular necrosis in mice fed a diet containing 0.025% ANIT for 2 weeks. Hepatic type 1 collagen mRNA expression and deposition increased markedly in livers of mice fed ANIT diet for 4 weeks. To determine whether TA treatment could inhibit this progression of liver fibrosis, mice were fed ANIT diet for 4 weeks and treated with TA for the last 2 weeks. Interestingly, TA treatment largely prevented increased deposition of type 1 collagen in livers of mice fed ANIT diet for 4 weeks. In contrast, biliary hyperplasia/inflammation and liver fibrosis were significantly increased in PAI-1(-/-) mice fed ANIT diet for 4 weeks. Overall, the results indicate that fibrinolytic activity contributes to ANIT diet-induced liver injury and fibrosis in mice. In addition, these proof-of-principle studies suggest the possibility that therapeutic intervention with an antifibrinolytic drug could form a novel strategy to prevent or reduce liver injury and fibrosis in patients with liver disease.
Collapse
Affiliation(s)
- Nikita Joshi
- Department of Pathobiology & Diagnostic Investigation (A.K.K., K.T., K.J.W., J.P.L.), Department of Pharmacology & Toxicology (N.J.), and Center for Integrative Toxicology (N.J., A.K.K., J.P.L.), Michigan State University, East Lansing, Michigan
| | | | | | | | | |
Collapse
|
27
|
Kitching AR. Dendritic cells in progressive renal disease: some answers, many questions. Nephrol Dial Transplant 2014; 29:2185-93. [DOI: 10.1093/ndt/gfu076] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
28
|
Lin C, Duitman J, Daalhuisen J, ten Brink M, von der Thüsen J, van der Poll T, Borensztajn K, Spek CA. Targeting protease activated receptor-1 with P1pal-12 limits bleomycin-induced pulmonary fibrosis. Thorax 2013; 69:152-60. [DOI: 10.1136/thoraxjnl-2013-203877] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
29
|
Zhong YC, Zhang T, Di W, Li WP. Thrombin promotes epithelial ovarian cancer cell invasion by inducing epithelial-mesenchymal transition. J Gynecol Oncol 2013; 24:265-72. [PMID: 23875077 PMCID: PMC3714465 DOI: 10.3802/jgo.2013.24.3.265] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 05/02/2013] [Accepted: 05/12/2013] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Over-expression of thrombin in ovarian cancer cells is associated with poor prognosis. In this study, we investigated the role of thrombin in inducing epithelial-mesenchymal transition (EMT) in SKOV3 epithelial ovarian cancer cells. METHODS After thrombin treatment SKOV3 cells were subjected to western blots, reverse-transcription PCR, and enzyme-linked immunosorbent assay to quantify EMT-related proteins, mRNA expression of SMAD2, DKK1, and sFRP1, and the secretion of matrix metalloproteinases (MMPs) and cytokines. Meanwhile, invasion ability was evaluated using transwell assays. RESULTS The results indicated a dose- and time-dependent down-regulation of E-cadherin and upregulation of N-cadherin and vimentin in thrombin-treated SKOV3 cells, compared with the thrombin-free control group (p<0.05). There was a dose- and time-dependent increase in the levels of SMAD2 and DKK1 mRNAs and a decrease in the levels of sFRP1 mRNA in thrombin-treated SKOV3 cells compared to control cells (p<0.05). Thrombin-treated SKOV3 cells exhibited increased secretion of MMP-9, MMP-2, interleukin (IL)-8, and IL-6 and increased invasion compared to untreated cells (p<0.05). Thrombin altered the morphology of SKOV3 cells to a spindle-like phenotype. Addition of hirudin to thrombin-treated cells reversed the effects of thrombin. CONCLUSION Thrombin induced EMT and promoted the invasion of SKOV3 cells, possibly via distinct signaling pathways. Hirudin inhibited the effects of thrombin, suggesting that anticoagulant therapy could be a novel therapeutic strategy for ovarian carcinoma.
Collapse
Affiliation(s)
- Yi-Cun Zhong
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China. ; Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | | | | | | |
Collapse
|
30
|
Wang Z, Famulski K, Lee J, Das SK, Wang X, Halloran P, Oudit GY, Kassiri Z. TIMP2 and TIMP3 have divergent roles in early renal tubulointerstitial injury. Kidney Int 2013; 85:82-93. [PMID: 23760282 DOI: 10.1038/ki.2013.225] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 03/26/2013] [Accepted: 04/25/2013] [Indexed: 01/13/2023]
Abstract
Tissue inhibitors of metalloproteinases (TIMPs) are endogenous inhibitors of matrix metalloproteinases (MMPs). While TIMP2 and TIMP3 inhibit MMPs, TIMP3 also inhibits activation of pro-MMP2, whereas TIMP2 promotes it. Here we assessed the differential role of TIMP2 and TIMP3 in renal injury using the unilateral ureteral obstruction model. Gene microarray assay showed that post obstruction, the lack of TIMP3 had a greater impact on gene expression of intermediate, late injury- and repair-induced transcripts, kidney selective transcripts, and solute carriers. Renal injury in TIMP3(-/-), but not in TIMP2(-/-), mice increased the expression of collagen type I/III, connective tissue growth factor, transforming growth factor-β, and the downstream Smad2/3 pathway. Interestingly, ureteral obstruction markedly increased MMP2 activation in the kidneys of TIMP3(-/-) mice, which was completely blocked in the kidneys of TIMP2(-/-) mice. These changes are consistent with enhanced renal tubulointerstitial fibrosis in TIMP3(-/-) and its reduction in TIMP2(-/-) mice. The activities of tumor necrosis factor-α-converting enzyme, caspase-3, and mitogen-activated kinases were elevated in the kidneys of TIMP3(-/-) mice but not TIMP2(-/-) mice, suggesting enhanced activation of apoptotic and pathological signaling pathways only in the obstructed kidney of TIMP3(-/-) mice. Thus, TIMP2 and TIMP3 play differential and contrasting roles in renal injury: TIMP3 protects from damage, whereas TIMP2 promotes injury through MMP2 activation.
Collapse
Affiliation(s)
- Zuocheng Wang
- 1] Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada [2] Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Konrad Famulski
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Jiwon Lee
- 1] Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada [2] Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Subhash K Das
- 1] Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada [2] Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Xiuhua Wang
- 1] Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada [2] Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Philip Halloran
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- 1] Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada [2] Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada [3] Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- 1] Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada [2] Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
31
|
Thymosin β4 and its degradation product, Ac-SDKP, are novel reparative factors in renal fibrosis. Kidney Int 2013; 84:1166-75. [PMID: 23739235 PMCID: PMC3830708 DOI: 10.1038/ki.2013.209] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 02/14/2013] [Accepted: 03/14/2013] [Indexed: 02/07/2023]
Abstract
Previously we found thymosin β4 (Tβ4) is up-regulated in glomerulosclerosis and required for angiotensin II-induced expression of plasminogen activator inhibitor-1 (PAI-1) in glomerular endothelial cells. Tβ4 has beneficial effects in dermal and corneal wound healing and heart disease yet its effects in kidney disease are unknown. Here we studied renal fibrosis in wild type and PAI-1 knockout mice following unilateral ureteral obstruction to explore the impact of Tβ4 and its prolyl oligopeptidase tetrapeptide degradation product, Ac-SDKP, in renal fibrosis. Additionally, we explored interactions of Tβ4 with PAI-1. Treatment with Ac-SDKP significantly decreased fibrosis in both wild type and PAI-1 knockout mice, as observed by decreased collagen and fibronectin deposition, fewer myofibroblasts and macrophages, and suppressed pro-fibrotic factors. In contrast, Tβ4 plus a prolyl oligopeptidase inhibitor significantly increased fibrosis in wild type mice. Tβ4 alone also promoted repair and reduced late fibrosis in wild type mice. Importantly, both pro-fibrotic effects of Tβ4 plus the prolyl oligopeptidase inhibitor, and late reparative effects of Tβ4 alone, were absent in PAI-1 knockout mice. Thus, Tβ4 combined with prolyl oligopeptidase inhibition, is consistently pro-fibrotic, but by itself, has anti-fibrotic effects in late stage fibrosis, while Ac-SDKP has consistent anti-fibrotic effects in both early and late stages of kidney injury. These effects of Tβ4 are dependent on PAI-1.
Collapse
|
32
|
Xiong M, Gong J, Liu Y, Xiang R, Tan X. Loss of vitamin D receptor in chronic kidney disease: a potential mechanism linking inflammation to epithelial-to-mesenchymal transition. Am J Physiol Renal Physiol 2012; 303:F1107-F1115. [PMID: 22791341 DOI: 10.1152/ajprenal.00151.2012] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Both peritubular inflammation and tubular epithelial-to-mesenchymal transition (EMT) are critical events during the pathogenesis of renal fibrosis. However, the relationship between these two processes is unclear. Here, we investigated the potential role of the vitamin D receptor (VDR) in coupling peritubular inflammation and EMT. In a mouse model of unilateral ureteral obstruction (UUO), loss of VDR was observed as early as 1 day after surgery. In cultured proximal tubular epithelial HK-2 cells, proinflammatory TNF-α inhibited the expression of VDR in a dose- and time-dependant manner. Treatment with TNF-α sensitized HK-2 cells to EMT stimulated by transforming growth factor (TGF)-β1. Ectopic expression of VDR counteracted the synergistic effect of TNF-α and TGF-β1 on EMT. Furthermore, knockdown of VDR using a small interfering RNA strategy mimicked the effect of TNF-α on facilitating EMT. Either TNF-α treatment or a loss of VDR induced β-catenin activation and its nuclear translocation. The VDR ligand calcitriol reversed the VDR loss and inhibited EMT in the mouse UUO model, and late administration of active vitamin D was effective in restoring VDR expression as well, and reduced collagen accumulation and deposition compared with the vehicle control. β-Catenin expression induced by UUO was also significantly inhibited after the late administration of vitamin D. These results indicate that the early loss of VDR in chronic kidney diseases was likely mediated by proinflammatory TNF-α, which renders tubular cells susceptible to EMT. Our data suggest that loss of VDR couples peritubular inflammation and EMT, two key events in renal fibrogenesis.
Collapse
Affiliation(s)
- Min Xiong
- Dept. of Pathology, Nankai Univ., Medical School, R116 Medical School Bldg., 94 Weijin Rd., Nankai District, Tianjin, 300071 China.
| | | | | | | | | |
Collapse
|
33
|
Snead AN, Insel PA. Defining the cellular repertoire of GPCRs identifies a profibrotic role for the most highly expressed receptor, protease-activated receptor 1, in cardiac fibroblasts. FASEB J 2012; 26:4540-7. [PMID: 22859370 DOI: 10.1096/fj.12-213496] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
G-protein-coupled receptors (GPCRs) have many roles in cell regulation and are commonly used as drug targets, but the repertoire of GPCRs expressed by individual cell types has not been defined. Here we use an unbiased approach, GPCR RT-PCR array, to define the expression of nonchemosensory GPCRs by cardiac fibroblasts (CFs) isolated from Rattus norvegicus. CFs were selected because of their importance for cardiac structure and function and their contribution to cardiac fibrosis, which occurs with advanced age, after acute injury (e.g., myocardial infarction), and in disease states (e.g., diabetes mellitus, hypertension). We discovered that adult rat CFs express 190 GPCRs and that activation of protease-activated receptor 1 (PAR1), the most highly expressed receptor, raises the expression of profibrotic markers in rat CFs, resulting in a 60% increase in collagen synthesis and conversion to a profibrogenic myofibroblast phenotype. We use siRNA knockdown of PAR1 (90% decrease in mRNA) to show that the profibrotic effects of thrombin are PAR1-dependent. These findings, which define the expression of GPCRs in CFs, provide a proof of principle of an approach to discover previously unappreciated, functionally relevant GPCRs and reveal a potential role for thrombin and PAR1 in wound repair and pathophysiology of the adult heart.
Collapse
Affiliation(s)
- Aaron N Snead
- Department of Pharmacology, University of California at San Diego, La Jolla, California 92093, USA
| | | |
Collapse
|
34
|
Iwaki T, Urano T, Umemura K. PAI-1, progress in understanding the clinical problem and its aetiology. Br J Haematol 2012; 157:291-8. [PMID: 22360729 DOI: 10.1111/j.1365-2141.2012.09074.x] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/24/2012] [Indexed: 01/07/2023]
Abstract
Plasminogen activator inhibitor-1 (PAI-1, also known as SERPINE1) is a member of the serine protease inhibitor (SERPIN) superfamily and is the primary physiological regulator of urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA) activity. Although the principal function of PAI-1 is the inhibition of fibrinolysis, PAI-1 possesses pleiotropic functions besides haemostasis. In the quarter century since its discovery, a number of studies have focused on improving our understanding of PAI-1 functions in vivo and in vitro. The use of Serpine1-deficient mice has particularly enhanced our understanding of the functions of PAI-1 in various physiological and pathophysiological conditions. In this review, the results of recent studies on PAI-1 and its role in clinical conditions are discussed.
Collapse
Affiliation(s)
- Takayuki Iwaki
- Department of Pharmacology, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | | | | |
Collapse
|
35
|
Giron-Michel J, Azzi S, Khawam K, Mortier E, Caignard A, Devocelle A, Ferrini S, Croce M, François H, Lecru L, Charpentier B, Chouaib S, Azzarone B, Eid P. Interleukin-15 plays a central role in human kidney physiology and cancer through the γc signaling pathway. PLoS One 2012; 7:e31624. [PMID: 22363690 PMCID: PMC3283658 DOI: 10.1371/journal.pone.0031624] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 01/16/2012] [Indexed: 12/16/2022] Open
Abstract
The ability of Interleukin-15 (IL-15) to activate many immune antitumor mechanisms renders the cytokine a good candidate for the therapy of solid tumors, particularly renal cell carcinoma. Although IL-15 is being currently used in clinical trials, the function of the cytokine on kidney's components has not been extensively studied; we thus investigated the role of IL-15 on normal and tumor renal epithelial cells. Herein, we analyzed the expression and the biological functions of IL-15 in normal renal proximal tubuli (RPTEC) and in their neoplastic counterparts, the renal clear cell carcinomas (RCC). This study shows that RPTEC express a functional heterotrimeric IL-15Rαβγc complex whose stimulation with physiologic concentrations of rhIL-15 is sufficient to inhibit epithelial mesenchymal transition (EMT) commitment preserving E-cadherin expression. Indeed, IL-15 is not only a survival factor for epithelial cells, but it can also preserve the renal epithelial phenotype through the γc-signaling pathway, demonstrating that the cytokine possess a wide range of action in epithelial homeostasis. In contrast, in RCC in vitro and in vivo studies reveal a defect in the expression of γc-receptor and JAK3 associated kinase, which strongly impacts IL-15 signaling. Indeed, in the absence of the γc/JAK3 couple we demonstrate the assembly of an unprecedented functional high affinity IL-15Rαβ heterodimer, that in response to physiologic concentrations of IL-15, triggers an unbalanced signal causing the down-regulation of the tumor suppressor gene E-cadherin, favoring RCC EMT process. Remarkably, the rescue of IL-15/γc-dependent signaling (STAT5), by co-transfecting γc and JAK3 in RCC, inhibits EMT reversion. In conclusion, these data highlight the central role of IL-15 and γc-receptor signaling in renal homeostasis through the control of E-cadherin expression and preservation of epithelial phenotype both in RPTEC (up-regulation) and RCC (down-regulation).
Collapse
Affiliation(s)
- Julien Giron-Michel
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Sandy Azzi
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Krystel Khawam
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Erwan Mortier
- INSERM UMRS 892, Institut de Recherche Thérapeutique de l'Université de Nantes (IRT UN), Nantes, France
| | - Anne Caignard
- Institut Cochin, Université Paris Descartes, INSERM U1016, Paris, France
| | - Aurore Devocelle
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Silvano Ferrini
- Laboratory of Immunotherapy, Instituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Michela Croce
- Laboratory of Immunotherapy, Instituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Hélène François
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Lola Lecru
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Bernard Charpentier
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
| | - Salem Chouaib
- INSERM UMR 753, Université de Paris-Sud, Institut Gustave Roussy (IGR), Villejuif, France
| | - Bruno Azzarone
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
- * E-mail: (BA); (PE)
| | - Pierre Eid
- INSERM UMR 1014, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Sud P11, Paris, France
- * E-mail: (BA); (PE)
| |
Collapse
|
36
|
Cuccioloni M, Bonfili L, Mozzicafreddo M, Cecarini V, Eleuteri AM, Angeletti M. Sanguisorba minor extract suppresses plasmin-mediated mechanisms of cancer cell migration. Biochim Biophys Acta Gen Subj 2012; 1820:1027-34. [PMID: 22348918 DOI: 10.1016/j.bbagen.2012.02.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 01/13/2012] [Accepted: 02/03/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND Sanguisorba minor, as well as several other edible herbs and vegetables, has been used extensively in traditional medicine. The observed beneficial effects can be attributed at least in part to the direct modulation of several enzymatic activities by its polyphenolic constituents. METHODS The ethanol extract of Sanguisorba minor was characterized by reversed-phase liquid chromatography, and most relevant analytes were identified by multiple stage mass spectrometry. The whole extract and the most relevant isolated constituents were tested for their ability to modulate the activity of human plasmin both toward a synthetic substrate and in human breast cancer cell culture models. Kinetic and equilibrium parameters were obtained by a concerted spectrophotometric and biosensor-based approach. RESULTS Quercetin-3-glucuronide was recognized as the compound mainly responsible for the in vitro plasmin inhibition by S. minor extract, with an inhibition constant in the high nanomolar range; in detail, our approach based on bioinformatic, enzymatic and binding analyses classified the inhibition as competitive. Most interestingly, cell-based assays showed that this flavonoid was effective in suppressing plasmin-induced loss of cancer cell adhesion. GENERAL SIGNIFICANCE Our results show that the extract from Sanguisorba minor limits plasmin-mediated tumor cell motility in vitro, mostly due to quercetin-3-glucuronide. This glucuronated flavonoid is a promising template for rational designing of anticancer drugs to be used in the treatment of pathological states involving the unregulated activity of plasmin.
Collapse
|
37
|
Abstract
Fibrosis is defined as a fibroproliferative or abnormal fibroblast activation-related disease. Deregulation of wound healing leads to hyperactivation of fibroblasts and excessive accumulation of extracellular matrix (ECM) proteins in the wound area, the pathological manifestation of fibrosis. The accumulation of excessive levels of collagen in the ECM depends on two factors: an increased rate of collagen synthesis and or decreased rate of collagen degradation by cellular proteolytic activities. The urokinase/tissue type plasminogen activator (uPA/tPA) and plasmin play significant roles in the cellular proteolytic degradation of ECM proteins and the maintenance of tissue homeostasis. The activities of uPA/tPA/plasmin and plasmin-dependent MMPs rely mostly on the activity of a potent inhibitor of uPA/tPA, plasminogen activator inhibitor-1 (PAI-1). Under normal physiologic conditions, PAI-1 controls the activities of uPA/tPA/plasmin/MMP proteolytic activities and thus maintains the tissue homeostasis. During wound healing, elevated levels of PAI-1 inhibit uPA/tPA/plasmin and plasmin-dependent MMP activities, and, thus, help expedite wound healing. In contrast to this scenario, under pathologic conditions, excessive PAI-1 contributes to excessive accumulation of collagen and other ECM protein in the wound area, and thus preserves scarring. While the level of PAI-1 is significantly elevated in fibrotic tissues, lack of PAI-1 protects different organs from fibrosis in response to injury-related profibrotic signals. Thus, PAI-1 is implicated in the pathology of fibrosis in different organs including the heart, lung, kidney, liver, and skin. Paradoxically, PAI-1 deficiency promotes spontaneous cardiac-selective fibrosis. In this review, we discuss the significance of PAI-1 in the pathogenesis of fibrosis in multiple organs.
Collapse
Affiliation(s)
- Asish K. Ghosh
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Douglas E. Vaughan
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
38
|
Abstract
BACKGROUND The plasminogen-plasmin system affects tissue fibrosis, presumably by interacting with metalloproteinases (MMPs) and macrophage recruitment. This study tests the influence of plasminogen activator inhibitor-1 (PAI-1) and tissue-type plasminogen activator (tPa) on angiotensin II-mediated hypertensive kidney and heart injury. METHOD Hypertension was induced by continuous angiotensin II (Ang II) infusion via osmotic mini-pumps over 4 weeks. The effects of Ang II infusion were determined in mice lacking PAI-1 (PAI-1), mice lacking tPa (tPa), and wild-type mice. Normotensive mice of the respective genotype served as controls. Blood pressure was recorded by continuous radiotelemetric intra-arterial measurements. RESULTS Ang II infusion significantly enhanced arterial blood pressure in all groups. However, the increase in blood pressure was more pronounced in the tPa group. Albuminuria was highest in hypertensive wild-type compared to the other Ang II-infused groups. Hypertensive PAI-1 mice exhibited less glomerulosclerosis, higher nephrin immunostaining, and lower renal interstitial collagen I deposition. Gelatin zymography revealed higher activity of MMP-2 in hypertensive PAI-1, whereas no differences were observed in macrophage infiltration. tPa deficiency did not alter kidney fibrosis, although hypertensive tPa revealed less renal expression of fibrotic genes, less macrophage infiltration, and reduced MMP-2 activity. On the other hand, hypertension-induced fibrosis as well as macrophage infiltration in the heart was profoundly enhanced in PAI-1 mice. Fibrin staining revealed perivascular exudations in the myocardium of hypertensive PAI-1 suggesting vascular leakage. CONCLUSION These findings underscore the unexpectedly complex role of plasminogen activation for hypertensive target organ damage.
Collapse
|
39
|
Carlson KE, McMurry TJ, Hunt SW. Pepducins: lipopeptide allosteric modulators of GPCR signaling. DRUG DISCOVERY TODAY. TECHNOLOGIES 2012; 9:e1-e70. [PMID: 24064242 DOI: 10.1016/j.ddtec.2011.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
|
40
|
Jung GS, Kim MK, Jung YA, Kim HS, Park IS, Min BH, Lee KU, Kim JG, Park KG, Lee IK. Clusterin attenuates the development of renal fibrosis. J Am Soc Nephrol 2012; 23:73-85. [PMID: 22052058 PMCID: PMC3269926 DOI: 10.1681/asn.2011010048] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 08/12/2011] [Indexed: 11/03/2022] Open
Abstract
Upregulation of clusterin occurs in several renal diseases and models of nephrotoxicity, but whether this promotes injury or is a protective reaction to injury is unknown. Here, in the mouse unilateral ureteral obstruction model, obstruction markedly increased the expression of clusterin, plasminogen activator inhibitor-1 (PAI-1), type I collagen, and fibronectin. Compared with wild-type mice, clusterin-deficient mice exhibited higher levels of PAI-1, type I collagen, and fibronectin and accelerated renal fibrosis in response to obstruction. In cultured rat tubular epithelium-like cells, adenovirus-mediated overexpression of clusterin inhibited the expression of TGF-β-stimulated PAI-1, type I collagen, and fibronectin. Clusterin inhibited TGF-β-stimulated Smad3 activity via inhibition of Smad3 phosphorylation and its nuclear translocation. Moreover, intrarenal delivery of adenovirus-expressing clusterin upregulated expression of clusterin in tubular epithelium-like cells and attenuated obstruction-induced renal fibrosis. In conclusion, clusterin attenuates renal fibrosis in obstructive nephropathy. These results suggest that upregulation of clusterin during renal injury is a protective response against the development of renal fibrosis.
Collapse
Affiliation(s)
- Gwon-Soo Jung
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea
| | - Mi-Kyung Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea
| | - Yun-A Jung
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea
| | - Hye-Soon Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea
| | - In-Sun Park
- Department of Anatomy and Center for Advanced Medical Education by BK21 project, College of Medicine, Inha University, Incheon, South Korea
| | - Bon-Hong Min
- Department of Pharmacology and BK21 Program for Medical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Ki-Up Lee
- Department of Internal Medicine, University of Ulsan College of Medicine, Seoul, South Korea; and
| | - Jung-Guk Kim
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, South Korea
| | - Keun-Gyu Park
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, South Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, South Korea
| |
Collapse
|
41
|
López-Guisa JM, Cai X, Collins SJ, Yamaguchi I, Okamura DM, Bugge TH, Isacke CM, Emson CL, Turner SM, Shankland SJ, Eddy AA. Mannose receptor 2 attenuates renal fibrosis. J Am Soc Nephrol 2011; 23:236-51. [PMID: 22095946 DOI: 10.1681/asn.2011030310] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mannose receptor 2 (Mrc2) expresses an extracellular fibronectin type II domain that binds to and internalizes collagen, suggesting that it may play a role in modulating renal fibrosis. Here, we found that Mrc2 levels were very low in normal kidneys but subsets of interstitial myofibroblasts and macrophages upregulated Mrc2 after unilateral ureteral obstruction (UUO). Renal fibrosis and renal parenchymal damage were significantly worse in Mrc2-deficient mice. Similarly, Mrc2-deficient Col4α3(-/-) mice with hereditary nephritis had significantly higher levels of total kidney collagen, serum BUN, and urinary protein than Mrc2-sufficient Col4α3(-/-) mice. The more severe phenotype seemed to be the result of reduced collagen turnover, because procollagen III (α1) mRNA levels and fractional collagen synthesis in the wild-type and Mrc2-deficient kidneys were similar after UUO. Although Mrc2 associates with the urokinase receptor, differences in renal urokinase activity did not account for the increased fibrosis in the Mrc2-deficient mice. Treating wild-type mice with a cathepsin inhibitor, which blocks proteases implicated in Mrc2-mediated collagen degradation, worsened UUO-induced renal fibrosis. Cathepsin mRNA profiles were similar in Mrc2-positive fibroblasts and macrophages, and Mrc2 genotype did not alter relative cathepsin mRNA levels. Taken together, these data establish an important fibrosis-attenuating role for Mrc2-expressing renal interstitial cells and suggest the involvement of a lysosomal collagen turnover pathway.
Collapse
Affiliation(s)
- Jesús M López-Guisa
- Seattle Children's Research Institute and Department of Pediatrics, University of Washington, Seattle, WA 98101-1309, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Fragiadaki M, Mason RM. Epithelial-mesenchymal transition in renal fibrosis - evidence for and against. Int J Exp Pathol 2011. [PMID: 21554437 DOI: 10.1111/j.1365-2613.2011.00775.x.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a well established biological process in metazoan embryological development. Over the past 15 years, investigators have sought to establish whether EMT also occurs in renal epithelial cells, following kidney injury, and to show that the mesenchymal cells formed could give rise to myofibroblasts which populate the renal interstitium, causing fibrosis within it. There is no doubt that proximal tubular epithelial cells (PTECs) can undergo EMT in vitro in response to TGFβ-1 and other inflammatory stimuli. Moreover, the results of experiments with animal models of renal fibrosis and examination of biopsies from patients with chronic kidney disease have lent support to the hypothesis that EMT occurs in vivo. This review discusses some of the key evidence underlying that idea and summarises recent advances in understanding the molecular mechanism underlying the process. Early experiments using mice which were genetically engineered to mark PTECs with the LacZ gene to trace their fate following kidney injury provided evidence supporting the occurrence of EMT. Recently, however, cell lineage tracking experiments using the red fluorescent protein (RFP) as a high-resolution marker for cells of renal epithelial origin did not replicate this result; the interstitial space following kidney injury was devoid of RFP expressing cells, leading the investigators to reject the renal EMT hypothesis.
Collapse
Affiliation(s)
- Maria Fragiadaki
- Imperial College Kidney and Transplant Institute, Imperial College London, Hammersmith Hospital, London, UK
| | | |
Collapse
|
43
|
Fragiadaki M, Mason RM. Epithelial-mesenchymal transition in renal fibrosis - evidence for and against. Int J Exp Pathol 2011; 92:143-50. [PMID: 21554437 DOI: 10.1111/j.1365-2613.2011.00775.x] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a well established biological process in metazoan embryological development. Over the past 15 years, investigators have sought to establish whether EMT also occurs in renal epithelial cells, following kidney injury, and to show that the mesenchymal cells formed could give rise to myofibroblasts which populate the renal interstitium, causing fibrosis within it. There is no doubt that proximal tubular epithelial cells (PTECs) can undergo EMT in vitro in response to TGFβ-1 and other inflammatory stimuli. Moreover, the results of experiments with animal models of renal fibrosis and examination of biopsies from patients with chronic kidney disease have lent support to the hypothesis that EMT occurs in vivo. This review discusses some of the key evidence underlying that idea and summarises recent advances in understanding the molecular mechanism underlying the process. Early experiments using mice which were genetically engineered to mark PTECs with the LacZ gene to trace their fate following kidney injury provided evidence supporting the occurrence of EMT. Recently, however, cell lineage tracking experiments using the red fluorescent protein (RFP) as a high-resolution marker for cells of renal epithelial origin did not replicate this result; the interstitial space following kidney injury was devoid of RFP expressing cells, leading the investigators to reject the renal EMT hypothesis.
Collapse
Affiliation(s)
- Maria Fragiadaki
- Imperial College Kidney and Transplant Institute, Imperial College London, Hammersmith Hospital, London, UK
| | | |
Collapse
|
44
|
Quaggin SE, Kapus A. Scar wars: mapping the fate of epithelial-mesenchymal-myofibroblast transition. Kidney Int 2011; 80:41-50. [PMID: 21430641 DOI: 10.1038/ki.2011.77] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The hypothesis that epithelial-mesenchymal transition (EMT) might be a contributor to the accumulation of fibroblasts and myofibroblasts (MFs) in the kidney during fibrogenesis was postulated 15 years ago. This paradigm offered an elegant explanation of how the loss of epithelial functions is coupled to the gain of deleterious mesenchymal functions; for example, excessive matrix deposition. Moreover, it interpreted chronic kidney disease in a developmental context: because the tubular epithelium originates from the metanephric mesenchyme, EMT can be viewed as a dedifferentiation process in response to injury, which might serve healing or--if dysregulated--might facilitate fibrosis. Several observations support the role of EMT in renal fibrosis: (1) Tubular cells can transform to fibroblasts and MFs in vitro. (2) Histological 'snapshots' reveal the coexistence of epithelial and mesenchymal markers in transitioning tubular cells in fibrosis models and human kidney diseases. (3) Early lineage-tracing experiments detected mesenchymal markers in the genetically tagged epithelium. However, the paradigm has been recently challenged; new fate-mapping studies found no evidence for the expression of (myo)fibroblast markers in the epithelium during fibrogenesis. This review summarizes the key findings and caveats, aiming at a balanced view, which neither overestimates the role of the epithelium in MF generation nor denies the importance of epithelial plasticity in fibrogenesis.
Collapse
Affiliation(s)
- Susan E Quaggin
- Division of Nephrology, St Michael's Hospital, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
45
|
López-Guisa JM, Rassa AC, Cai X, Collins SJ, Eddy AA. Vitronectin accumulates in the interstitium but minimally impacts fibrogenesis in experimental chronic kidney disease. Am J Physiol Renal Physiol 2011; 300:F1244-54. [PMID: 21270094 DOI: 10.1152/ajprenal.00701.2010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Vitronectin (Vtn) is a glycoprotein found in normal serum and pathological extracellular matrix. Given its known interactions with plasminogen activator inhibitor-1 (PAI-1) and Vtn cellular receptors, especially αvβ3 integrin and the urokinase receptor (uPAR), this study was designed to investigate its role in renal fibrogenesis in the mouse model of unilateral ureteral obstruction (UUO). Kidney Vtn mRNA levels were increased ×1.8-5.1 and Vtn protein levels ×1.9-3 on days 7, 14, and 21 after UUO compared with sham kidney levels. Groups of age-matched C57BL/6 wild-type (Vtn+/+) and Vtn-/- mice (n = 10-11/group) were killed 7, 14, or 21 days after UUO. Absence of Vtn resulted in the following significant differences, but only on day 14: fewer αSMA+ interstitial myofibroblasts (×0.53), lower procollagen III mRNA levels (×0.41), lower PAI-1 protein (×0.23), higher uPA activity (×1.1), and lower αv protein (×0.32). The number of CD68+ macrophages did not differ between the genotypes. Despite these transient differences on day 14, the absence of Vtn had no effect on fibrosis severity based on both picrosirius red-positive interstitial area and total kidney collagen measured by the hydroxyproline assay. These findings suggest that despite significant interstitial Vtn deposition in the UUO model of chronic kidney disease, its fibrogenic role is either nonessential or redundant. These data are remarkable given Vtn's strong affinity for the potent fibrogenic molecule PAI-1.
Collapse
Affiliation(s)
- Jesús M López-Guisa
- Seattle Children’s Research Institute, Department of Pediatrics, University of Washington, USA
| | | | | | | | | |
Collapse
|
46
|
Pharmacology, biodistribution, and efficacy of GPCR-based pepducins in disease models. Methods Mol Biol 2011; 683:259-75. [PMID: 21053136 DOI: 10.1007/978-1-60761-919-2_19] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
G protein-coupled receptors (GPCR) are a superfamily of receptors that are vital in a wide array of physiological processes. Modulation of GPCR signaling has been an intensive area of therapeutic study, mainly due to the diverse pathophysiological significance of GPCRs. Pepducins are cell-penetrating lipidated peptides designed to target the intracellular loops of the GPCR of interest. Pepducins can function as agonists or antagonists of their cognate receptor, making them highly useful compounds for the study of GPCR signaling. Pepducins have been used to control platelet-dependent hemostasis and thrombosis, tumor growth, invasion, and angiogenesis, as well as to improve sepsis outcomes in mice. Pepducins have been successfully designed against a wide variety of GPCRs including the protease-activated receptors (PAR1, 2, 4), the chemokine receptors (CXCR1, 2, 4), the sphingosine-1-phosphate receptor (S1P3), the adrenergic receptor (ADRA1B), and have the potential to help reveal the functions of intractable GPCRs. Pharmacokinetic, pharmacodynamic, and biodistribution studies have showed that pepducins are widely distributed throughout the body except the brain and possess appropriate drug-like properties for use in vivo. Here, we discuss the delivery, pharmacology, and biodistribution of pepducins, as well as the effects of pepducins in models of inflammation, cardiovascular disease, cancer, and angiogenesis.
Collapse
|
47
|
Liu X, Lü L, Tao BB, Zhou AL, Zhu YC. Amelioration of glomerulosclerosis with all-trans retinoic acid is linked to decreased plasminogen activator inhibitor-1 and α-smooth muscle actin. Acta Pharmacol Sin 2011; 32:70-8. [PMID: 21206504 PMCID: PMC4003321 DOI: 10.1038/aps.2010.200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Accepted: 09/01/2010] [Indexed: 12/17/2022]
Abstract
AIM To examine the effects of all-trans retinoic acid (atRA) on renal morphology and function as well as on renal plasminogen activator inhibitor-1 (PAI-1) expression and plasmin activity in rats with 5/6 nephrectomy. METHODS Adult male Sprague Dawley rats were given 5/6 nephrectomy or sham operation. Renal function was measured 2 weeks later. The nephrectomized rats were assigned to groups matched for proteinuria and treated with vehicle or atRA (5 or 10 mg/kg by gastric gavage once daily) for the next 12 weeks. Rats with sham operation were treated with vehicle. At the end of the treatments, kidneys were collected for histological examination, Western blot analysis, and enzymatic activity measurements. RESULTS The 5/6 nephrectomy promoted hypertension, renal dysfunction, and glomerulosclerosis. These changes were significantly reduced in the atRA-treated group. The expressions of PAI-1 and α-smooth muscle actin (α-SMA) were significantly increased in the vehicle-treated nephrectomized rats. Treatment with atRA significantly reduced the expressions of PAI-1 and α-SMA. However, plasmin activity remained unchanged following atRA treatment. CONCLUSION Treatment with atRA ameliorates glomerulosclerosis and improves renal function in rats with 5/6 nephrectomy. This is associated with a decrease in PAI-1 and α-SMA, but not with a change in plasmin activity.
Collapse
Affiliation(s)
- Xia Liu
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai 200032, China
- Department of Pathophysiology, Nantong University Nantong Medical College, Nantong 226001, China
| | - Lei Lü
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Bei-bei Tao
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Ai-ling Zhou
- Department of Pathophysiology, Nantong University Nantong Medical College, Nantong 226001, China
| | - Yi-chun Zhu
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai 200032, China
| |
Collapse
|
48
|
Zhang G, Thomas AL, Marshall AL, Kernan KA, Su Y, Zheng Y, Takano J, Saido TC, Eddy AA. Nicotinic acetylcholine receptor α1 promotes calpain-1 activation and macrophage inflammation in hypercholesterolemic nephropathy. J Transl Med 2011; 91:106-23. [PMID: 20661225 PMCID: PMC3188436 DOI: 10.1038/labinvest.2010.135] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The nicotinic acetylcholine receptor α1 (nAChRα1) was investigated as a potential proinflammatory molecule in the kidney, given a recent report that it is an alternative urokinase plasminogen activator (uPA) receptor, in addition to the classical receptor uPAR. Two animal models and in vitro monocyte studies were involved: (1) In an ApoE(-/-) mouse model of chronic kidney disease, glomerular-resident cells and monocytes/macrophages were identified as the primary cell types that express nAChRα1 during hypercholesterolemia/uninephrectomy-induced nephropathy. Silencing of the nAChRα1 gene for 4 months (6 months on Western diet) prevented the increases in renal monocyte chemoattractant protein-1 and osteopontin expression levels and F4/80+ macrophage infiltration compared with the nonsilenced mice. These changes were associated with significantly reduced transforming growth factor-β1 mRNA (50% decrease) and α smooth muscle actin-positive (αSMA+) myofibroblasts (90% decrease), better glomerular and tubular basement membranes (GBM/TBM) preservation (threefold less disintegration), and better renal function preservation (serum creatinine 40% lower) in the nAChRα1-silenced mice. The nAChRα1 silencing was also associated with significantly reduced renal tissue calcium deposition (78% decrease) and calpain-1 (but not calpain-2) activation (70% decrease). (2) The nAChRα1 was expressed in vitro by mouse monocyte cell line WEHI-274.1. The silencing of nAChRα1 significantly reduced both calpain-1 and -2 activities, and reduced the degradation of the calpain substrate talin. (3) To further explore the role of calpain-1 activity in hypercholesterolemic nephropathy, disease severities were compared in CAST(-/-)ApoE(-/-) (calpain overactive) mice and ApoE(-/-) mice fed with Western diet for 10 months (n=12). Macrophages were the main cell type of renal calpain-1 production in the model. The number of renal F4/80+ macrophages was 10-fold higher in the CAST(-/-)ApoE(-/-) mice (P<0.05), and was associated with a significantly higher level of αSMA+ cells, increased GBM/TBM destruction, and higher serum creatinine levels. Our studies suggest that the receptor nAChRα1 is an important regulator of calpain-1 activation and inflammation in the chronic hypercholesterolemic nephropathy. This new proinflammatory pathway may also be relevant to other disorders beyond hyperlipidemic nephropathy.
Collapse
Affiliation(s)
- Guoqiang Zhang
- Division of Nephrology, Seattle Children's Hospital Research Institute, Department of Pediatrics, University of Washington, Seattle, WA 98101, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Steensgaard M, Svenningsen P, Tinning AR, Nielsen TD, Jørgensen F, Kjaersgaard G, Madsen K, Jensen BL. Apical serine protease activity is necessary for assembly of a high-resistance renal collecting duct epithelium. Acta Physiol (Oxf) 2010; 200:347-59. [PMID: 20645929 DOI: 10.1111/j.1748-1716.2010.02170.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
AIM We hypothesized that the serine protease prostasin is necessary for differentiation of a high-resistance renal collecting duct epithelium governed by glucocorticoid. METHODS Postnatal rat kidney and adult human kidney was used to study the expression and localization of prostasin. The murine collecting duct cell line (M-1) was cultured in Snapwell inserts to investigate the significance of prostasin for the development of transepithelial electrical resistance (TER). RESULTS In the cortex and medulla of rat kidney, prostasin mRNA and protein increased significantly between birth and weaning (day 21) and was detected in collecting ducts. Immunoreactive prostasin was associated with collecting ducts and loops of Henle in human kidney. In rat, adrenalectomy at day 10 had no effect on prostasin mRNA level in kidney at day 20. Cultured M-1 cells exhibited parallel increases in prostasin mRNA, protein and TER 5 days after seeding. Apical addition of the serine protease inhibitor aprotinin to M-1 cell cultures inhibited development of TER and led to aberrant localization of E-cadherin. This effect was mimicked by the protease inhibitor nafamostat. Apical addition of phospholipase C to cleave glycosylphosphatidylinositol (GPI) anchors released prostasin to the medium and attenuated development of TER with time of culture. Disruption of lipid rafts by methyl-β-cyclodextrin attenuated development of TER in M-1 cells. Omission of dexamethasone impaired development of TER in M-1 cells, while prostasin protein abundance and E-cadherin distribution did not change. CONCLUSION Apical, GPI-anchored, lipid raft-associated serine protease activity, compatible with prostasin, is necessary for the development of a high-resistance collecting duct epithelium.
Collapse
Affiliation(s)
- M Steensgaard
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Wang X, Zhou Y, Tan R, Xiong M, He W, Fang L, Wen P, Jiang L, Yang J. Mice lacking the matrix metalloproteinase-9 gene reduce renal interstitial fibrosis in obstructive nephropathy. Am J Physiol Renal Physiol 2010; 299:F973-82. [PMID: 20844022 DOI: 10.1152/ajprenal.00216.2010] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinase-9 (MMP-9) is one of the major components of the matrix proteolytic network, and its role in the pathogenesis of renal interstitial fibrosis remains largely unknown. Here, we demonstrate that ablation of MMP-9 attenuated renal interstitial fibrotic lesions in obstructive nephropathy. Mice lacking MMP-9 were less likely to develop morphological injury, which was characterized by a reduced disruption of tubular basement membrane (TBM) and expression of fibronectin as well as deposition of total tissue collagen in the kidneys after sustained ureteral obstruction compared with their wild-type counterparts. Deficiency of MMP-9 blocked tubular epithelial-to-myofibroblast transition (EMT) but did not alter the induction of transforming growth factor (TGF)-β1 axis expression in the obstructed kidneys. In vitro, TBM, which was digested by MMP-9 instead of MMP-9 itself, induces EMT and enhances migration of transformed cells. Thus increased MMP-9 is detrimental in renal interstitial fibrogenesis through a cascade of events that leads to TBM destruction and in turn to promotion of EMT. Our findings establish a crucial and definite importance of MMP-9 in the pathogenesis of renal interstitial fibrosis at the whole-animal level.
Collapse
Affiliation(s)
- Xiaohua Wang
- Center of Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yang Zhou
- Center of Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ruoyun Tan
- Center of Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Mingxia Xiong
- Center of Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Weichun He
- Center of Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li Fang
- Center of Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ping Wen
- Center of Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lei Jiang
- Center of Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Junwei Yang
- Center of Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|