Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Physiol Renal Physiol ; 314(2): F306-F316, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29046300

ABSTRACT

Aquaporin-2 (AQP2) is a water channel protein expressed in principal cells (PCs) of the kidney collecting ducts (CDs) and plays a critical role in mediating water reabsorption and urine concentration. AQP2 undergoes both regulated trafficking mediated by vasopressin (VP) and constitutive recycling, which is independent of VP. For both pathways, actin cytoskeletal dynamics is a key determinant of AQP2 trafficking. We report here that manganese chloride (MnCl2) is a novel and potent regulator of AQP2 trafficking in cultured cells and in the kidney. MnCl2 treatment promoted internalization and intracellular accumulation of AQP2. The effect of MnCl2 on the intracellular accumulation of AQP2 was associated with activation of RhoA and actin polymerization without modification of AQP2 phosphorylation. Although the level of total and phosphorylated AQP2 did not change, MnCl2 treatment impeded VP-induced phosphorylation of AQP2 at its serine-256, -264, and -269 residues and dephosphorylation at serine 261. In addition, MnCl2 significantly promoted F-actin polymerization along with downregulation of RhoA activity and prevented VP-induced membrane accumulation of AQP2. Finally, MnCl2 treatment in mice resulted in significant polyuria and reduced urinary concentration, likely due to intracellular relocation of AQP2 in the PCs of kidney CDs. More importantly, the reduced urinary concentration caused by MnCl2 treatment in animals was not corrected by VP. In summary, our study identified a novel effect of MnCl2 on AQP2 trafficking through modifying RhoA activity and actin polymerization and uncovered its potent impact on water diuresis in vivo.


Subject(s)
Actin Cytoskeleton/drug effects , Actins/metabolism , Aquaporin 2/metabolism , Chlorides/toxicity , Kidney Concentrating Ability/drug effects , Kidney Tubules, Collecting/drug effects , Polyuria/chemically induced , Actin Cytoskeleton/metabolism , Animals , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/physiopathology , LLC-PK1 Cells , Male , Manganese Compounds , Mice, Inbred C57BL , Phosphorylation , Polymerization , Polyuria/metabolism , Polyuria/physiopathology , Protein Transport , Signal Transduction/drug effects , Swine , Vasopressins/pharmacology , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
2.
Sci Rep ; 7(1): 8321, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28814739

ABSTRACT

Helium ion scanning microscopy (HIM) is a novel technology that directly visualizes the cell surface ultrastructure without surface coating. Despite its very high resolution, it has not been applied extensively to study biological or pathology samples. Here we report the application of this powerful technology to examine the three-dimensional ultrastructural characteristics of proteinuric glomerulopathy in mice with CD2-associated protein (CD2AP) deficiency. HIM revealed the serial alteration of glomerular features including effacement and disorganization of the slit diaphragm, followed by foot process disappearance, flattening and fusion of major processes, and eventual transformation into a podocyte sheet as the disease progressed. The number and size of the filtration slit pores decreased. Strikingly, numerous "bleb" shaped microprojections were observed extending from podocyte processes and cell body, indicating significant membrane dynamics accompanying CD2AP deficiency. Visualizing the glomerular endothelium and podocyte-endothelium interface revealed the presence of endothelial damage, and disrupted podocyte and endothelial integrity in 6 week-old Cd2ap-KO mice. We used the HIM technology to investigate at nanometer scale resolution the ultrastructural alterations of the glomerular filtration apparatus in mice lacking the critical slit diaphragm-associated protein CD2AP, highlighting the great potential of HIM to provide new insights into the biology and (patho)physiology of glomerular diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Cytoskeletal Proteins/deficiency , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Animals , Disease Models, Animal , Endothelium/metabolism , Endothelium/pathology , Helium , Kidney Diseases/metabolism , Kidney Glomerulus/ultrastructure , Mice , Mice, Knockout , Microscopy, Confocal , Podocytes/metabolism , Podocytes/ultrastructure
3.
Am J Physiol Renal Physiol ; 313(4): F1026-F1037, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28701310

ABSTRACT

The renal collecting duct (CD) contains two major cell types, intercalated (ICs) and principal cells (PCs). A previous report showed that deletion of ß1-integrin in the entire renal CD causes defective CD morphogenesis resulting in kidney dysfunction. However, subsequent deletion of ß1-integrin specifically in ICs and PCs, respectively, did not cause any morphological defects in the CDs. The discrepancy between these studies prompts us to reinvestigate the role of ß1-integrin in CD cells, specifically in the PCs. We conditionally deleted ß1-integrin in mouse CD PCs using a specific aquaporin-2 (AQP2) promoter Cre-LoxP system. The resulting mutant mice, ß-1f/fAQP2-Cre+, had lower body weight, failed to thrive, and died around 8-12 wk. Their CD tubules were dilated, and some of them contained cellular debris. Increased apoptosis and proliferation of PCs were observed in the dilated CDs. Trichrome staining and electron microscopy revealed the presence of peritubular and interstitial fibrosis that is associated with increased production of extracellular matrix proteins including collagen type IV and fibronectin, as detected by immunoblotting. Further analysis revealed a significantly increased expression of transforming growth factor-ß (TGF-ß)-induced protein, fibronectin, and TGF-ß receptor-1 mRNAs and concomitantly increased phosphorylation of SMAD-2 that indicates the activation of the TGF-ß signaling pathway. Therefore, our data reveal that normal expression of ß1-integrin in PCs is a critical determinant of CD structural and functional integrity and further support the previously reported critical role of ß1-integrin in the development and/or maintenance of the CD structure and function.


Subject(s)
Extracellular Matrix/metabolism , Gene Deletion , Integrin beta1/metabolism , Kidney Medulla/metabolism , Kidney Tubules, Collecting/metabolism , Polyuria/metabolism , Renal Insufficiency/metabolism , Age Factors , Animals , Apoptosis , Aquaporin 2/genetics , Cell Proliferation , Extracellular Matrix/ultrastructure , Failure to Thrive/genetics , Failure to Thrive/metabolism , Failure to Thrive/pathology , Fibrosis , Genetic Predisposition to Disease , Integrases/genetics , Integrin beta1/genetics , Kidney Medulla/ultrastructure , Kidney Tubules, Collecting/ultrastructure , Mice, Knockout , Phenotype , Phosphorylation , Polyuria/genetics , Polyuria/pathology , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Renal Insufficiency/genetics , Renal Insufficiency/pathology , Signal Transduction , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism
4.
Am J Physiol Renal Physiol ; 311(6): F1346-F1357, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27760768

ABSTRACT

Within the past decade tremendous efforts have been made to understand the mechanism behind aquaporin-2 (AQP2) water channel trafficking and recycling, to open a path toward effective diabetes insipidus therapeutics. A recent study has shown that integrin-linked kinase (ILK) conditional-knockdown mice developed polyuria along with decreased AQP2 expression. To understand whether ILK also regulates AQP2 trafficking in kidney tubular cells, we performed in vitro analysis using LLCPK1 cells stably expressing rat AQP2 (LLC-AQP2 cells). Upon treatment of LLC-AQP2 cells with ILK inhibitor cpd22 and ILK-siRNA, we observed increased accumulation of AQP2 in the perinuclear region, without any significant increase in the rate of endocytosis. This perinuclear accumulation did not occur in cells expressing a serine-256-aspartic acid mutation that retains AQP2 in the plasma membrane. We then examined clathrin-mediated endocytosis after ILK inhibition using rhodamine-conjugated transferrin. Despite no differences in overall transferrin endocytosis, the endocytosed transferrin also accumulated in the perinuclear region where it colocalized with AQP2. These accumulated vesicles also contained the recycling endosome marker Rab11. In parallel, the usual vasopressin-induced AQP2 membrane accumulation was prevented after ILK inhibition; however, ILK inhibition did not measurably affect AQP2 phosphorylation at serine-256 or its dephosphorylation at serine-261. Instead, we found that inhibition of ILK increased F-actin polymerization. When F-actin was depolymerized with latrunculin, the perinuclear located AQP2 dispersed. We conclude that ILK is important in orchestrating dynamic cytoskeletal architecture during recycling of AQP2, which is necessary for its subsequent entry into the exocytotic pathway.


Subject(s)
Aquaporin 2/metabolism , Cytoskeleton/metabolism , Exocytosis/physiology , Kidney/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Line , Cytoskeleton/drug effects , Endocytosis/drug effects , Endocytosis/physiology , Exocytosis/drug effects , Kidney/drug effects , Male , Mice , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics
5.
PLoS One ; 10(7): e0131719, 2015.
Article in English | MEDLINE | ID: mdl-26147297

ABSTRACT

In renal collecting duct (CD) principal cells (PCs), vasopressin (VP) acts through its receptor, V2R, to increase intracellular cAMP leading to phosphorylation and apical membrane accumulation of the water channel aquaporin 2 (AQP2). The trafficking and function of basolaterally located AQP2 is, however, poorly understood. Here we report the successful application of a 3-dimensional Madin-Darby canine kidney (MDCK) epithelial model to study polarized AQP2 trafficking. This model recapitulates the luminal architecture of the CD and bi-polarized distribution of AQP2 as seen in kidney. Without stimulation, AQP2 is located in the subapical and basolateral regions. Treatment with VP, forskolin (FK), or 8-(4-Chlorophenylthio)-2'-O-methyladenosine 3',5'-cyclic monophosphate monosodium hydrate (CPT-cAMP) leads to translocation of cytosolic AQP2 to the apical membrane, but not to the basolateral membrane. Treating cells with methyl-ß-cyclodextrin (mßCD) to acutely block endocytosis causes accumulation of AQP2 on the basolateral membrane, but not on the apical membrane. Our data suggest that AQP2 may traffic differently at the apical and basolateral domains in this 3D epithelial model. In addition, application of a panel of phosphorylation specific AQP2 antibodies reveals the polarized, subcellular localization of differentially phosphorylated AQP2 at S256, S261, S264 and S269 in the 3D culture model, which is consistent with observations made in the CDs of VP treated animals, suggesting the preservation of phosphorylation dependent regulatory mechanism of AQP2 trafficking in this model. Therefore we have established a 3D culture model for the study of trafficking and regulation of both the apical and basolaterally targeted AQP2. The new model will enable further characterization of the complex mechanism regulating bi-polarized trafficking of AQP2 in vitro.


Subject(s)
Aquaporin 2/metabolism , Animals , Cells, Cultured , Dogs , Epithelial Cells/metabolism , Madin Darby Canine Kidney Cells , Phosphorylation , Protein Transport
6.
J Cell Mol Med ; 18(4): 656-70, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24495224

ABSTRACT

Posterior capsular opacification (PCO) is the major complication arising after cataract treatment. PCO occurs when the lens epithelial cells remaining following surgery (LCs) undergo a wound healing response producing a mixture of α-smooth muscle actin (α-SMA)-expressing myofibroblasts and lens fibre cells, which impair vision. Prior investigations have proposed that integrins play a central role in PCO and we found that, in a mouse fibre cell removal model of cataract surgery, expression of αV integrin and its interacting ß-subunits ß1, ß5, ß6, ß8 are up-regulated concomitant with α-SMA in LCs following surgery. To test the hypothesis that αV integrins are functionally important in PCO pathogenesis, we created mice lacking the αV integrin subunit in all lens cells. Adult lenses lacking αV integrins are transparent and show no apparent morphological abnormalities when compared with control lenses. However, following surgical fibre cell removal, the LCs in control eyes increased cell proliferation, and up-regulated the expression of α-SMA, ß1-integrin, fibronectin, tenascin-C and transforming growth factor beta (TGF-ß)-induced protein within 48 hrs, while LCs lacking αV integrins exhibited much less cell proliferation and little to no up-regulation of any of the fibrotic markers tested. This effect appears to result from the known roles of αV integrins in latent TGF-ß activation as αV integrin null lenses do not exhibit detectable SMAD-3 phosphorylation after surgery, while this occurs robustly in control lenses, consistent with the known roles for TGF-ß in fibrotic PCO. These data suggest that therapeutics antagonizing αV integrin function could be used to prevent fibrotic PCO following cataract surgery.


Subject(s)
Capsule Opacification/metabolism , Capsule Opacification/pathology , Cataract Extraction/adverse effects , Integrin alphaV/metabolism , Transforming Growth Factor beta/metabolism , Actins/biosynthesis , Animals , Capsule Opacification/etiology , Cell Proliferation , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Lens Capsule, Crystalline/metabolism , Lens Capsule, Crystalline/pathology , Mice , Myofibroblasts/metabolism , Myofibroblasts/pathology , Smad3 Protein/biosynthesis , Wound Healing
7.
J Cell Mol Med ; 16(3): 445-55, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21883891

ABSTRACT

Transforming growth factor-ß (TGF-ß) has roles in embryonic development, the prevention of inappropriate inflammation and tumour suppression. However, TGF-ß signalling also regulates pathological epithelial-to-mesenchymal transition (EMT), inducing or progressing a number of diseases ranging from inflammatory disorders, to fibrosis and cancer. However, TGF-ß signalling does not proceed linearly but rather induces a complex network of cascades that mutually influence each other and cross-talk with other pathways to successfully induce EMT. Particularly, there is substantial evidence for cross-talk between αV integrins and TGF-ß during EMT, and anti-integrin therapeutics are under development as treatments for TGF-ß-related disorders. However, TGF-ß's complex signalling network makes the development of therapeutics to block TGF-ß-mediated pathology challenging. Moreover, despite our current understanding of integrins and TGF-ß function during EMT, the precise mechanism of their role during physiological versus pathological EMT is not fully understood. This review focuses on the circle of regulation between αV integrin and TGF-ß signalling during TGF-ß induced EMT, which pose as a significant driver to many known TGF-ß-mediated disorders.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Integrin alphaV/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Adhesion , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Fibrosis , Gene Expression Regulation , Genes, Regulator , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Integrin alphaV/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction , Transforming Growth Factor beta/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...