Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Eur J Cell Biol ; 102(2): 151316, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37084657

ABSTRACT

The expression of the angiotensin-converting enzyme 2 (ACE2) is altered in multiple chronic kidney diseases like hypertension and renal fibrosis, where the signaling from the basal membrane proteins is critical for the development and progression of the various pathologies. Integrins are heterodimeric cell surface receptors that have important roles in the progression of these chronic kidney diseases by altering various cell signaling pathways in response to changes in the basement membrane proteins. It is unclear whether integrin or integrin-mediated signaling affects the ACE2 expression in the kidney. The current study tests the hypothesis that integrin ß1 regulates the expression of ACE2 in kidney epithelial cells. The role of integrin ß1 in ACE2 expression in renal epithelial cells was investigated by shRNA-mediated knockdown and pharmacological inhibition. In vivo studies were carried out using epithelial cell-specific deletion of integrin ß1 in the kidneys. Deletion of integrin ß1 from the mouse renal epithelial cells reduced the expression of ACE2 in the kidney. Furthermore, the downregulation of integrin ß1 using shRNA decreased ACE2 expression in human renal epithelial cells. ACE2 expression levels were also decreased in renal epithelial cells and cancer cells when treated with an integrin α2ß1 antagonist, BTT 3033. SARS-CoV-2 viral entry to human renal epithelial cells and cancer cells was also inhibited by BTT 3033. This study demonstrates that integrin ß1 positively regulates the expression of ACE2, which is required for the entry of SARS-CoV-2 into kidney cells.


Subject(s)
COVID-19 , Renal Insufficiency, Chronic , Humans , Animals , Mice , Integrin beta1/genetics , Integrin beta1/metabolism , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , SARS-CoV-2/metabolism , COVID-19/metabolism , COVID-19/pathology , Kidney/metabolism , Kidney/pathology , Epithelial Cells/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
2.
Kidney Int ; 103(1): 144-155, 2023 01.
Article in English | MEDLINE | ID: mdl-36273656

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) involves the development and persistent growth of fluid filled kidney cysts. In a recent study, we showed that ADPKD kidney cyst epithelial cells can stimulate the proliferation and differentiation of peri-cystic myofibroblasts. Although dense myofibroblast populations are often found surrounding kidney cysts, their role in cyst enlargement or fibrosis in ADPKD is unclear. To clarify this, we examined the effect of myofibroblast depletion in the Pkd1RC/RC (RC/RC) mouse model of ADPKD. RC/RC;αSMAtk mice that use the ganciclovir-thymidine kinase system to selectively deplete α-smooth muscle actin expressing myofibroblasts were generated. Ganciclovir treatment for four weeks depleted myofibroblasts, reduced kidney fibrosis and preserved kidney function in these mice. Importantly, myofibroblast depletion significantly reduced cyst growth and cyst epithelial cell proliferation in RC/RC;αSMAtk mouse kidneys. Similar ganciclovir treatment did not alter cyst growth or fibrosis in wild-type or RC/RC littermates. In vitro, co-culture with myofibroblasts from the kidneys of patients with ADPKD increased 3D microcyst growth of human ADPKD cyst epithelial cells. Treatment with conditioned culture media from ADPKD kidney myofibroblasts increased microcyst growth and cell proliferation of ADPKD cyst epithelial cells. Further examination of ADPKD myofibroblast conditioned media showed high levels of protease inhibitors including PAI1, TIMP1 and 2, NGAL and TFPI-2, and treatment with recombinant PAI1 and TIMP1 increased ADPKD cyst epithelial cell proliferation in vitro. Thus, our findings show that myofibroblasts directly promote cyst epithelial cell proliferation, cyst growth and fibrosis in ADPKD kidneys, and their targeting could be a novel therapeutic strategy to treat PKD.


Subject(s)
Cysts , Polycystic Kidney, Autosomal Dominant , Humans , Mice , Animals , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/pathology , Myofibroblasts , Cells, Cultured , Kidney/pathology , Cell Proliferation , Fibrosis , Cysts/drug therapy , Cysts/pathology , Epithelial Cells/pathology
3.
Int J Mol Sci ; 23(14)2022 Jul 09.
Article in English | MEDLINE | ID: mdl-35886951

ABSTRACT

Vasopressin type-2 receptor (V2R) is ectopically expressed and plays a pathogenic role in clear cell renal cell carcinoma (ccRCC) tumor cells. Here we examined how V2R signaling within human ccRCC tumor cells (Caki1 cells) stimulates stromal cancer-associated fibroblasts (CAFs). We found that cell culture conditioned media from Caki1 cells increased activation, migration, and proliferation of fibroblasts in vitro, which was inhibited by V2R gene silencing in Caki1 cells. Analysis of the conditioned media and mRNA of the V2R gene silenced and control Caki1 cells showed that V2R regulates the production of CAF-activating factors. Some of these factors were also found to be regulated by YAP in these Caki1 cells. YAP expression colocalized and correlated with V2R expression in ccRCC tumor tissue. V2R gene silencing or V2R antagonist significantly reduced YAP in Caki1 cells. Moreover, the V2R antagonist reduced YAP expression and myofibroblasts in mouse xenograft tumors. These results suggest that V2R plays an important role in secreting pro-fibrotic factors that stimulate fibroblast activation by a YAP-dependent mechanism in ccRCC tumors. Our results demonstrate a novel role for the V2R-YAP axis in the regulation of myofibroblasts in ccRCC and a potential therapeutic target.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Renal Cell , Kidney Neoplasms , Receptors, Vasopressin , Animals , Antidiuretic Hormone Receptor Antagonists/pharmacology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Culture Media, Conditioned , Fibroblasts/metabolism , Humans , Kidney/drug effects , Kidney/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology , Vasopressins/genetics , Vasopressins/metabolism , Xenograft Model Antitumor Assays
4.
Cell Death Dis ; 12(10): 947, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34650051

ABSTRACT

Autosomal-dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease and is characterized by progressive growth of fluid-filled cysts. Growth factors binding to receptor tyrosine kinases (RTKs) stimulate cell proliferation and cyst growth in PKD. Nintedanib, a triple RTK inhibitor, targets the vascular endothelial growth-factor receptor (VEGFR), platelet-derived growth-factor receptor (PDGFR), and fibroblast growth-factor receptor (FGFR), and is an approved drug for the treatment of non-small-cell lung carcinoma and idiopathic lung fibrosis. To determine if RTK inhibition using nintedanib can slow ADPKD progression, we tested its effect on human ADPKD renal cyst epithelial cells and myofibroblasts in vitro, and on Pkd1f/fPkhd1Cre and Pkd1RC/RC, orthologous mouse models of ADPKD. Nintedanib significantly inhibited cell proliferation and in vitro cyst growth of human ADPKD renal cyst epithelial cells, and cell viability and migration of human ADPKD renal myofibroblasts. Consistently, nintedanib treatment significantly reduced kidney-to-body-weight ratio, renal cystic index, cystic epithelial cell proliferation, and blood-urea nitrogen levels in both the Pkd1f/fPkhd1Cre and Pkd1RC/RC mice. There was a corresponding reduction in ERK, AKT, STAT3, and mTOR activity and expression of proproliferative factors, including Yes-associated protein (YAP), c-Myc, and Cyclin D1. Nintedanib treatment significantly reduced fibrosis in Pkd1RC/RC mice, but did not affect renal fibrosis in Pkd1f/fPkhd1Cre mice. Overall, these results suggest that nintedanib may be repurposed to effectively slow cyst growth in ADPKD.


Subject(s)
Indoles/therapeutic use , Polycystic Kidney, Autosomal Dominant/drug therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Indoles/pharmacology , Kidney/drug effects , Kidney/pathology , Mice, Knockout , Myofibroblasts/drug effects , Myofibroblasts/pathology , Protein Kinase Inhibitors/pharmacology , Receptors, Cell Surface/metabolism , Signal Transduction/drug effects
5.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638957

ABSTRACT

The tumor microenvironment plays a critical role in defining the growth and malignancy of solid tumors. Extracellular matrix (ECM) proteins such as collagen, vitronectin, and fibronectin are major components of the tumor microenvironment. Tumor growth-promoting reciprocal interaction between ECM and cytoplasmic proteins is regulated by the cell surface receptors called integrins. This study investigated the mechanism by which integrin ß1 promotes pancreatic tumor growth. In MIA PaCa-2 pancreatic cancer cell line, the loss of integrin ß1 protein reduced the ability of cells to proliferate in a 3D matrix and compromised the ability to form a focal adhesion complex. Decreased expression of integrin α5 was observed in KO cells, which resulted in impaired cell spreading and adhesion on vitronectin and fibronectin. Reduced expression of the integrin-associated protein, kindlin-2 was also recorded. The downregulation of kindlin-2 decreased the phosphorylation of Smad2/3 by reducing the expression of TGF-ß receptor 2. These results unravel a new mechanism of integrin ß1 in tumor growth by modifying the expression of kindlin-2 and TGF-ß receptor 2 signaling.


Subject(s)
Carcinogenesis/metabolism , Cell Proliferation/genetics , Integrin beta1/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction/genetics , Carcinogenesis/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Integrin beta1/genetics , Pancreatic Neoplasms/pathology , Transfection
6.
FASEB J ; 34(10): 13597-13608, 2020 10.
Article in English | MEDLINE | ID: mdl-32813289

ABSTRACT

Acute kidney injury (AKI) is characterized by injury to the tubular epithelium that leads to the sudden loss of renal function. Proper tubular regeneration is essential to prevent progression to chronic kidney disease. In this study, we examined the role of FoxM1, a forkhead box family member transcription factor in tubular repair after AKI. Renal FoxM1 expression increased after renal ischemia/reperfusion (I/R)-induced AKI in mouse kidneys. Treatment with thiostrepton, a FoxM1 inhibitor, reduced FoxM1 regulated pro-proliferative factors and cell proliferation in vitro, and tubular regeneration in mouse kidneys after AKI. Glycogen synthase kinase-3 (GSK3) was found to be an upstream regulator of FoxM1 because GSK3 inhibition or renal tubular GSK3ß gene deletion significantly increased FoxM1 expression, and improved tubular repair and renal function. GSK3 inactivation increased ß-catenin, Cyclin D1, and c-Myc, and reduced cell cycle inhibitors p21 and p27. Importantly, thiostrepton treatment abolished the improved tubular repair in GSK3ß knockout mice following AKI. These results demonstrate that FoxM1 is important for renal tubular regeneration following AKI and that GSK3ß suppresses tubular repair by inhibiting FoxM1.


Subject(s)
Acute Kidney Injury/metabolism , Forkhead Box Protein M1/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Reperfusion Injury/metabolism , Animals , Cell Line , Cell Proliferation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Kidney Tubules/pathology , Kidney Tubules/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Regeneration
7.
Nephron ; 144(12): 609-612, 2020.
Article in English | MEDLINE | ID: mdl-32726778

ABSTRACT

Acute kidney injury (AKI) is a common clinical syndrome that involves renal tubular epithelial cell death and leads to acute decline in renal function. Improper tubular regeneration following AKI often leads to CKD. We discuss the role of a serine/threonine protein kinase called glycogen synthase kinase-3 (GSK3) in renal tubular injury and renal fibrosis. We also highlight the importance of GSK3 as a potential drug target in AKI patients and molecular mechanisms promoting tissue regeneration.


Subject(s)
Acute Kidney Injury/enzymology , Glycogen Synthase Kinase 3/metabolism , Signal Transduction , Animals , Apoptosis , Epithelial Cells/pathology , Humans , Kidney Tubules/pathology
8.
J Am Soc Nephrol ; 31(8): 1697-1710, 2020 08.
Article in English | MEDLINE | ID: mdl-32554753

ABSTRACT

BACKGROUND: Fibrosis is a major cause of loss of renal function in autosomal dominant polycystic kidney disease (ADPKD). In this study, we examined whether vasopressin type-2 receptor (V2R) activity in cystic epithelial cells can stimulate interstitial myofibroblasts and fibrosis in ADPKD kidneys. METHODS: We treated Pkd1 gene knockout (Pkd1KO) mice with dDAVP, a V2R agonist, for 3 days and evaluated the effect on myofibroblast deposition of extracellular matrix (ECM). We also analyzed the effects of conditioned media from primary cultures of human ADPKD cystic epithelial cells on myofibroblast activation. Because secretion of the profibrotic connective tissue growth factor (CCN2) increased significantly in dDAVP-treated Pkd1KO mouse kidneys, we examined its role in V2R-dependent fibrosis in ADPKD as well as that of yes-associated protein (YAP). RESULTS: V2R stimulation using dDAVP increased the renal interstitial myofibroblast population and ECM deposition. Similarly, conditioned media from human ADPKD cystic epithelial cells increased myofibroblast activation in vitro, suggesting a paracrine mechanism. Renal collecting duct-specific gene deletion of CCN2 significantly reduced cyst growth and myofibroblasts in Pkd1KO mouse kidneys. We found that YAP regulates CCN2, and YAP inhibition or gene deletion reduces renal fibrosis in Pkd1KO mouse kidneys. Importantly, YAP inactivation blocks the dDAVP-induced increase in myofibroblasts in Pkd1KO kidneys. Further in vitro studies showed that V2R regulates YAP by an ERK1/2-dependent mechanism in human ADPKD cystic epithelial cells. CONCLUSIONS: Our results demonstrate a novel mechanism by which cystic epithelial cells stimulate myofibroblasts in the pericystic microenvironment, leading to fibrosis in ADPKD. The V2R-YAP-CCN2 cell signaling pathway may present a potential therapeutic target for fibrosis in ADPKD.


Subject(s)
Cell Cycle Proteins/physiology , Connective Tissue Growth Factor/physiology , Kidney/pathology , Myofibroblasts/physiology , Polycystic Kidney, Autosomal Dominant/pathology , Receptors, Vasopressin/physiology , Transcription Factors/physiology , Animals , Deamino Arginine Vasopressin/pharmacology , Extracellular Matrix/metabolism , Fibrosis , Humans , Mice , TRPP Cation Channels/physiology
9.
Oncogene ; 39(6): 1231-1245, 2020 02.
Article in English | MEDLINE | ID: mdl-31616061

ABSTRACT

Arginine vasopressin (AVP) and its type-2 receptor (V2R) play an essential role in the regulation of salt and water homeostasis by the kidneys. V2R activation also stimulates proliferation of renal cell carcinoma (RCC) cell lines in vitro. The current studies investigated V2R expression and activity in human RCC tumors, and its role in RCC tumor growth. Examination of the cancer genome atlas (TCGA) database, and analysis of human RCC tumor tissue microarrays, cDNA arrays and tumor biopsy samples demonstrated V2R expression and activity in clear cell RCC (ccRCC). In vitro, V2R antagonists OPC31260 and Tolvaptan, or V2R gene silencing reduced wound closure and cell viability of 786-O and Caki-1 human ccRCC cell lines. Similarly in mouse xenograft models, Tolvaptan and OPC31260 decreased RCC tumor growth by reducing cell proliferation and angiogenesis, while increasing apoptosis. In contrast, the V2R agonist dDAVP significantly increased tumor growth. High intracellular cAMP levels and ERK1/2 activation were observed in human ccRCC tumors. In mouse tumors and Caki-1 cells, V2R agonists reduced cAMP and ERK1/2 activation, while dDAVP treatment had the reverse effect. V2R gene silencing in Caki-1 cells also reduced cAMP and ERK1/2 activation. These results provide novel evidence for a pathogenic role of V2R signaling in ccRCC, and suggest that inhibitors of the AVP-V2R pathway, including the FDA-approved drug Tolvaptan, could be utilized as novel ccRCC therapeutics.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Kidney Neoplasms/drug therapy , Receptors, Vasopressin/chemistry , Tolvaptan/pharmacology , Animals , Antidiuretic Hormone Receptor Antagonists/pharmacology , Apoptosis , Biomarkers, Tumor , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Case-Control Studies , Cell Cycle , Cell Proliferation , Female , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Nude , Prognosis , Receptors, Vasopressin/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Am J Physiol Renal Physiol ; 317(2): F343-F360, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31091126

ABSTRACT

Polycystic kidney disease (PKD) is characterized by slowly expanding renal cysts that damage the kidney, typically resulting in renal failure by the fifth decade. The most common cause of death in these patients, however, is cardiovascular disease. Expanding cysts in PKD induce chronic kidney injury that is accompanied by immune cell infiltration, including macrophages, which we and others have shown can promote disease progression in PKD mouse models. Here, we show that monocyte chemoattractant protein-1 [MCP-1/chemokine (C-C motif) ligand 2 (CCL2)] is responsible for the majority of monocyte chemoattractant activity produced by renal PKD cells from both mice and humans. To test whether the absence of MCP-1 lowers renal macrophage concentration and slows disease progression, we generated genetic knockout (KO) of MCP-1 in a mouse model of PKD [congenital polycystic kidney (cpk) mice]. Cpk mice are born with rapidly expanding renal cysts, accompanied by a decline in kidney function and death by postnatal day 21. Here, we report that KO of MCP-1 in these mice increased survival, with some mice living past 3 mo. Surprisingly, however, there was no significant difference in renal macrophage concentration, nor was there improvement in cystic disease or kidney function. Examination of mice revealed cardiac hypertrophy in cpk mice, and measurement of cardiac electrical activity via ECG revealed repolarization abnormalities. MCP-1 KO did not affect the number of cardiac macrophages, nor did it alleviate the cardiac aberrancies. However, MCP-1 KO did prevent the development of pulmonary edema, which occurred in cpk mice, and promoted decreased resting heart rate and increased heart rate variability in both cpk and noncystic mice. These data suggest that in this mouse model of PKD, MCP-1 altered cardiac/pulmonary function and promoted death outside of its role as a macrophage chemoattractant.


Subject(s)
Arrhythmias, Cardiac/metabolism , Cardiomegaly/metabolism , Chemokine CCL2/metabolism , Kidney/metabolism , Lung/metabolism , Myocardium/metabolism , Polycystic Kidney Diseases/metabolism , Pulmonary Edema/metabolism , Animals , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cells, Cultured , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Disease Models, Animal , Disease Progression , Fibrosis , Humans , Inflammation Mediators/metabolism , Kidney/pathology , Kidney/physiopathology , Lung/pathology , Lung/physiopathology , Macrophages/metabolism , Macrophages/pathology , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Polycystic Kidney Diseases/pathology , Polycystic Kidney Diseases/physiopathology , Pulmonary Edema/pathology , Pulmonary Edema/physiopathology , Pulmonary Edema/prevention & control , Time Factors
11.
J Mol Cell Biol ; 8(6): 464-476, 2016 12.
Article in English | MEDLINE | ID: mdl-27190311

ABSTRACT

Glycogen synthase kinase 3ß (GSK3ß), a serine/threonine protein kinase, is commonly known to be regulated at the level of its activity. However, in some diseases including polycystic kidney disease (PKD), GSK3ß expression is increased and plays a pathophysiological role. The current studies aimed to determine the mechanism for the increased GSK3ß expression in PKD and its significance to disease progression. In mouse models of PKD, increases in renal GSK3ß corresponded with increases in renal cAMP levels and disease progression. In vivo and in vitro studies revealed that GSK3ß is a cAMP-responsive gene, and elevated cAMP levels, as seen in PKD, can increase GSK3ß expression. In normal mice, vasopressin signaling induced by water deprivation increased GSK3ß expression, which decreased following rehydration. Examination of the GSK3ß promoter revealed five potential binding sites for the transcription factor, cAMP response element binding protein (CREB). CREB was found to bind to GSK3ß promoter and essential for cAMP-mediated regulation of GSK3ß. Importantly, this regulation was demonstrated to be part of a feed-forward loop in which cAMP through CREB regulates GSK3ß expression, and GSK3ß in turn positively regulates cAMP generation. GSK3ß or CREB inhibition reduced transepithelial fluid secretion and cyst expansion in vitro Thus, disruption at any point of this destructive cycle may be therapeutically useful to reduce cyst expansion and preserve renal function in PKD.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Polycystic Kidney Diseases/metabolism , Animals , Body Fluids/metabolism , Cyclic AMP , Dogs , Gene Knockout Techniques , Glycogen Synthase Kinase 3 beta/genetics , Humans , Kidney/enzymology , Kidney/pathology , Madin Darby Canine Kidney Cells , Mice, Inbred C57BL , Polycystic Kidney Diseases/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , TRPP Cation Channels , Vasopressins/metabolism
12.
J Am Soc Nephrol ; 27(6): 1604-7, 2016 06.
Article in English | MEDLINE | ID: mdl-27026364

ABSTRACT

The Kidney Tutored Research and Education for Kidney Students (TREKS) Program is a product of the American Society of Nephrology (ASN) Workforce Committee that seeks to connect medical and graduate students to nephrology. This program starts with a weeklong camp-like course introducing participants to renal physiology through classic and modern experiments. Next, each student is matched with a nephrology mentor at his or her home institution to foster a better understanding of a nephrology career. Lastly, the students are encouraged to participate in scholarly activities and attend the ASN Kidney Week. Now in its third year, with a total of 84 participants, survey data suggest early success of the program, with a self-reported 40% increased interest in nephrology fellowship and/or research careers. In addition, students give high ratings to the course components and mentorship pairings. Continued student tracking will be necessary to determine the long-term program effect.


Subject(s)
Career Choice , Nephrology/education , Education, Medical, Graduate , Female , Humans , Male , Mentors , Societies, Medical , United States
13.
Dis Model Mech ; 8(8): 931-40, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26092126

ABSTRACT

Glycogen synthase kinase-3ß (GSK3ß) is a serine/threonine protein kinase that plays an important role in renal tubular injury and regeneration in acute kidney injury. However, its role in the development of renal fibrosis, often a long-term consequence of acute kidney injury, is unknown. Using a mouse model of renal fibrosis induced by ischemia-reperfusion injury, we demonstrate increased GSK3ß expression and activity in fibrotic kidneys, and its presence in myofibroblasts in addition to tubular epithelial cells. Pharmacological inhibition of GSK3 using TDZD-8 starting before or after ischemia-reperfusion significantly suppressed renal fibrosis by reducing the myofibroblast population, collagen-1 and fibronectin deposition, inflammatory cytokines, and macrophage infiltration. GSK3 inhibition in vivo reduced TGF-ß1, SMAD3 activation and plasminogen activator inhibitor-1 levels. Consistently in vitro, TGF-ß1 treatment increased GSK3ß expression and GSK3 inhibition abolished TGF-ß1-induced SMAD3 activation and α-smooth muscle actin (α-SMA) expression in cultured renal fibroblasts. Importantly, overexpression of constitutively active GSK3ß stimulated α-SMA expression even in the absence of TGF-ß1 treatment. These results suggest that TGF-ß regulates GSK3ß, which in turn is important for TGF-ß-SMAD3 signaling and fibroblast-to-myofibroblast differentiation. Overall, these studies demonstrate that GSK3 could promote renal fibrosis by activation of TGF-ß signaling and the use of GSK3 inhibitors might represent a novel therapeutic approach for progressive renal fibrosis that develops as a consequence of acute kidney injury.


Subject(s)
Fibroblasts/enzymology , Fibroblasts/pathology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Kidney/pathology , Protein Kinase Inhibitors/pharmacology , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Animals , Cell Differentiation/drug effects , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Fibrosis , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Inflammation Mediators/metabolism , Kidney/drug effects , Kidney/enzymology , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Protein Kinase Inhibitors/therapeutic use , Reperfusion Injury/drug therapy , Signal Transduction/drug effects , Thiadiazoles/pharmacology , Thiadiazoles/therapeutic use , Transforming Growth Factor beta/metabolism , beta Catenin/metabolism
14.
Kidney Int ; 87(6): 1164-75, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25629553

ABSTRACT

Polycystic kidney diseases (PKDs) are inherited disorders characterized by the formation of fluid filled renal cysts. Elevated cAMP levels in PKDs stimulate progressive cyst enlargement involving cell proliferation and transepithelial fluid secretion often leading to end-stage renal disease. The glycogen synthase kinase-3 (GSK3) family of protein kinases consists of GSK3α and GSK3ß isoforms and has a crucial role in multiple cellular signaling pathways. We previously found that GSK3ß, a regulator of cell proliferation, is also crucial for cAMP generation and vasopressin-mediated urine concentration by the kidneys. However, the role of GSK3ß in the pathogenesis of PKDs is not known. Here we found that GSK3ß expression and activity were markedly upregulated and associated with cyst-lining epithelia in the kidneys of mice and humans with PKD. Renal collecting duct-specific gene knockout of GSK3ß or pharmacological inhibition of GSK3 effectively slowed down the progression of PKD in mouse models of autosomal recessive or autosomal dominant PKD. GSK3 inactivation inhibited cAMP generation and cell proliferation resulting in reduced cyst expansion, improved renal function, and extended life span. GSK3ß inhibition also reduced pERK, c-Myc, and cyclin-D1, known mitogens in proliferation of cystic epithelial cells. Thus, GSK3ß has a novel functional role in PKD pathophysiology, and its inhibition may be therapeutically useful to slow down cyst expansion and progression of PKD.


Subject(s)
Cyclic AMP/metabolism , Cysts/metabolism , Cysts/pathology , Glycogen Synthase Kinase 3/metabolism , Polycystic Kidney Diseases/enzymology , Animals , Cell Proliferation/drug effects , Cyclin D1/metabolism , Enzyme Inhibitors/pharmacology , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Kidney/enzymology , Kidney Tubules, Collecting/enzymology , Mice , Mice, Knockout , Organ Size/drug effects , Polycystic Kidney Diseases/pathology , Polycystic Kidney Diseases/physiopathology , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Thiadiazoles/pharmacology
15.
Am J Physiol Renal Physiol ; 308(6): F650-60, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25608967

ABSTRACT

In mammals, glycogen synthase kinase (GSK)3 comprises GSK3α and GSK3ß isoforms. GSK3ß has been shown to play a role in the ability of kidneys to concentrate urine by regulating vasopressin-mediated water permeability of collecting ducts, whereas the role of GSK3α has yet to be discerned. To investigate the role of GSK3α in urine concentration, we compared GSK3α knockout (GSK3αKO) mice with wild-type (WT) littermates. Under normal conditions, GSK3αKO mice had higher water intake and urine output. GSK3αKO mice also showed reduced urine osmolality and aquaporin-2 levels but higher urinary vasopressin. When water deprived, they failed to concentrate their urine to the same level as WT littermates. The addition of 1-desamino-8-d-arginine vasopressin to isolated inner medullary collecting ducts increased the cAMP response in WT mice, but this response was reduced in GSK3αKO mice, suggesting reduced responsiveness to vasopressin. Gene silencing of GSK3α in mpkCCD cells also reduced forskolin-induced aquaporin-2 expression. When treated with LiCl, an isoform nonselective inhibitor of GSK3 and known inducer of polyuria, WT mice developed significant polyuria within 6 days. However, in GSK3αKO mice, the polyuric response was markedly reduced. This study demonstrates, for the first time, that GSK3α could play a crucial role in renal urine concentration and suggest that GSK3α might be one of the initial targets of Li(+) in LiCl-induced nephrogenic diabetes insipidus.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Kidney Tubules, Collecting/enzymology , Urine/physiology , Animals , Aquaporin 2/metabolism , Gene Silencing , Glycogen Synthase Kinase 3/genetics , Lithium Chloride , Mice, Knockout , Polyuria/genetics
16.
J Biol Chem ; 288(7): 5223-31, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23283969

ABSTRACT

The epithelial sodium channel (ENaC) participates in the regulation of plasma sodium and volume, and gain of function mutations in the human channel cause salt-sensitive hypertension. Roles for the arachidonic acid epoxygenase metabolites, the epoxyeicosatrienoic acids (EETs), in ENaC activity have been identified; however, their mechanisms of action remain unknown. In polarized M1 cells, 14,15-EET inhibited amiloride-sensitive apical to basolateral sodium transport as effectively as epidermal growth factor (EGF). The EET effects were associated with increased threonine phosphorylation of the ENaC ß and γ subunits and abolished by inhibitors of (a) mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/extracellular signal regulated kinases 1 and 2 (MEK/ERK1/2) and (b) EGF receptor signaling. CYP2C44 epoxygenase knockdown blunted the sodium transport effects of EGF, and its 14,15-EET metabolite rescued the knockdown phenotype. The relevance of these findings is indicated by (a) the hypertension that results in mice administered cetuximab, an inhibitor of EGF receptor binding, and (b) immunological data showing an association between the pressure effects of cetuximab and reductions in ENaCγ phosphorylation. These studies (a) identify an ERK1/2-dependent mechanism for ENaC inhibition by 14,15-EET, (b) point to ENaC as a proximal target for EET-activated ERK1/2 mitogenic kinases, (c) characterize a mechanistic commonality between EGF and epoxygenase metabolites as ENaC inhibitors, and (d) suggest a CYP2C epoxygenase-mediated pathway for the regulation of distal sodium transport.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Epithelial Sodium Channels/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , 8,11,14-Eicosatrienoic Acid/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antihypertensive Agents/pharmacology , Cetuximab , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P450 Family 2 , Epidermal Growth Factor/metabolism , Humans , Hypertension , Kidney/metabolism , Male , Mice , Models, Biological , Phosphorylation
17.
Kidney Int ; 82(9): 1000-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22785175

ABSTRACT

Renal proximal tubular damage and repair are hallmarks of acute kidney injury. As glycogen synthase kinase-3ß (GSK3ß) is an important cellular regulator of survival and proliferation, we determined its role during injury and recovery of proximal tubules in a mercuric chloride-induced nephrotoxic model of acute kidney injury. Renal proximal tubule-specific GSK3ß knockout mice exposed to mercuric chloride had improved survival and renal function compared to wild-type mice. Apoptosis, measured by TUNEL staining, Bax activation, and caspase 3 cleavage, was reduced in the knockout mice. The restoration of renal structure, function, and cell proliferation was also accelerated in the GSK3ß knockout mice. This enhanced repair, evidenced by increased Ki-67 and BRDU staining, along with increased cyclin D1 and c-myc levels, was recapitulated by treatment of wild-type mice with the small-molecule GSK3 inhibitor TDZD-8 following injury. This confirmed that hastened repair in the knockout mice was not merely due to lower initial injury levels. Thus, inhibition of GSK3ß prior to nephrotoxic insult protects from renal injury. Such treatment after acute kidney injury may accelerate repair and regeneration.


Subject(s)
Acute Kidney Injury/physiopathology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Kidney Tubules, Proximal/physiology , Acute Disease , Acute Kidney Injury/chemically induced , Acute Kidney Injury/mortality , Acute Kidney Injury/pathology , Animals , Anti-Infective Agents, Local/toxicity , Apoptosis/physiology , Cell Proliferation , Disease Models, Animal , Female , Gene Deletion , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Kaplan-Meier Estimate , Kidney Tubules, Proximal/pathology , Mercuric Chloride/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Thiadiazoles/pharmacology
18.
Curr Opin Nephrol Hypertens ; 21(5): 541-6, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22691876

ABSTRACT

PURPOSE OF REVIEW: Glycogen synthase kinase-3 (GSK3) is an enzyme that is gaining prominence as a critical signaling molecule in the epithelial cells of renal tubules. This review will focus on recent findings exploring the role of GSK3 in renal collecting ducts, especially its role in urine concentration involving vasopressin signaling. RECENT FINDINGS: Recent studies using inhibition or tissue-specific gene deletion of GSK3 revealed the mechanism by which GSK3 regulates aquaporin 2 water channels via adenylate cyclase or the prostaglandin-E2 pathway. In other studies, postnatal treatment with lithium, an inhibitor of GSK3, increased cell proliferation and led to microcyst formation in rat kidneys. These studies suggest that loss of GSK3 activity could interfere with renal water transport at two levels. In the short term, it could disrupt vasopressin signaling in collecting duct cells and in the long term it could alter the structure of the collecting ducts, making them less responsive to the hydro-osmotic effects of vasopressin. SUMMARY: Ongoing studies reveal the crucial role played by GSK3 in the regulation of vasopressin action in the renal collecting ducts and suggest a possible use of GSK3 inhibitors in disease conditions associated with disrupted vasopressin signaling.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Kidney Tubules, Collecting/enzymology , Animals , Aquaporin 2/metabolism , Biological Transport , Humans , Signal Transduction , Vasopressins/metabolism
19.
J Am Soc Nephrol ; 21(3): 428-37, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20056751

ABSTRACT

Glycogen synthase kinase 3beta (GSK3beta), a serine/threonine protein kinase, is a key target of drug discovery in several diseases, including diabetes and Alzheimer disease. Because lithium, a potent inhibitor of GSK3beta, causes nephrogenic diabetes insipidus, GSK3beta may play a crucial role in regulating water homeostasis. We developed renal collecting duct-specific GSK3beta knockout mice to determine whether deletion of GSK3beta affects arginine vasopressin-dependent renal water reabsorption. Although only mildly polyuric under normal conditions, knockout mice exhibited an impaired urinary concentrating ability in response to water deprivation or treatment with a vasopressin analogue. The knockout mice had reduced levels of mRNA, protein, and membrane localization of the vasopressin-responsive water channel aquaporin 2 compared with wild-type mice. The knockout mice also expressed lower levels of pS256-AQP2, a phosphorylated form crucial for membrane trafficking. Levels of cAMP, a major regulator of aquaporin 2 expression and trafficking, were also lower in the knockout mice. Both GSK3beta gene deletion and pharmacologic inhibition of GSK3beta reduced adenylate cyclase activity. In summary, GSK3beta inactivation or deletion reduces aquaporin 2 expression by modulating adenylate cyclase activity and cAMP generation, thereby impairing responses to vasopressin in the renal collecting duct.


Subject(s)
Adenylyl Cyclases/metabolism , Antidiuretic Agents/pharmacology , Deamino Arginine Vasopressin/pharmacology , Glycogen Synthase Kinase 3/metabolism , Kidney Concentrating Ability/physiology , Kidney Tubules, Collecting/enzymology , Adenylyl Cyclases/genetics , Animals , Aquaporin 2/genetics , Aquaporin 2/metabolism , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Female , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Kidney Concentrating Ability/drug effects , Kidney Tubules, Collecting/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Water Deprivation/physiology , Water-Electrolyte Balance/drug effects , Water-Electrolyte Balance/physiology
20.
J Biol Chem ; 282(23): 16959-68, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17401137

ABSTRACT

Prostaglandin E2 (PGE(2)), a major product of cyclooxygenase, exerts its functions by binding to four G protein-coupled receptors (EP1-4) and has been implicated in modulating angiogenesis. The present study examined the role of the EP4 receptor in regulating endothelial cell proliferation, migration, and tubulogenesis. Primary pulmonary microvascular endothelial cells were isolated from EP4(flox/flox) mice and were rendered null for the EP4 receptor with adenoCre virus. Whereas treatment with PGE(2) or the EP4 selective agonists PGE(1)-OH and ONO-AE1-329 induced migration, tubulogenesis, ERK activation and cAMP production in control adenovirus-transduced endothelial EP4(flox/flox) cells, no effects were seen in adenoCre-transduced EP4(flox/flox) cells. The EP4 agonist-induced endothelial cell migration was inhibited by ERK, but not PKA inhibitors, defining a functional link between PGE(2)-induced endothelial cell migration and EP4-mediated ERK signaling. Finally, PGE(2), as well as PGE(1)-OH and ONO-AE1-329, also promoted angiogenesis in an in vivo sponge assay providing evidence that the EP4 receptor mediates de novo vascularization in vivo.


Subject(s)
Cell Movement/physiology , Dinoprostone/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Neovascularization, Physiologic , Receptors, Prostaglandin E/physiology , Animals , Base Sequence , Blotting, Western , Cell Proliferation , Cells, Cultured , DNA Primers , Enzyme Activation , Mice , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP4 Subtype , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...