Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 109(17): 6680-5, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22493269

ABSTRACT

The epithelial cell tight junction separates apical and basolateral domains and is essential for barrier function. Disruption of the tight junction is a hallmark of epithelial cell damage and can lead to end organ damage including renal failure. Herein, we identify Gα12 activation by H(2)O(2) leading to tight junction disruption and demonstrate a critical role for Gα12 activation during bilateral renal ischemia/reperfusion injury. Madin-Darby canine kidney (MDCK) cells with inducible Gα12 (Gα12-MDCK) and silenced Gα12 (shGα12-MDCK) were subjected to ATP depletion/repletion and H(2)O(2)/catalase as models of tight junction disruption and recovery by monitoring transepithelial resistance. In ATP depleted cells, barrier disruption and recovery was not affected by Gα12, but reassembly was accelerated by Gα12 depletion. In contrast, silencing of Gα12 completely protected cells from H(2)O(2)-stimulated barrier disruption, a response that rapidly occurred in control cells. H(2)O(2) activated Src and Rho, and Src inhibition (by PP2), but not Rho (by Y27632), protected cells from H(2)O(2)-mediated barrier disruption. Immunofluorescent and biochemical analysis showed that H(2)O(2) led to increased tyrosine phosphorylation of numerous proteins and altered membrane localization of tight junction proteins through Gα12/Src signaling pathway. Gα12 and Src were activated in vivo during ischemia/reperfusion injury, and transgenic mice with renal tubular QLα12 (activated mutant) expression were delayed in recovery and showed more extensive injury. Conversely, Gα12 knockout mice were nearly completely protected from ischemia/reperfusion injury. Taken together, these studies reveal that ROS stimulates Gα12 to activate injury pathways and identifies a therapeutic target for ameliorating ROS mediated injury.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Hydrogen Peroxide/pharmacology , Intercellular Junctions/drug effects , Reperfusion Injury/metabolism , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Dogs , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Gene Silencing , Intercellular Junctions/metabolism , Mice , Mice, Transgenic , Phosphorylation
2.
Lab Invest ; 92(5): 662-75, 2012 May.
Article in English | MEDLINE | ID: mdl-22249312

ABSTRACT

Glomerulosclerosis is a common pathological finding that often progresses to renal failure. The mechanisms of chronic kidney disease progression are not well defined, but may include activation of numerous vasoactive and inflammatory pathways. We hypothesized that podocytes are susceptible to filtered plasma components, including hormones and growth factors that stimulate signaling pathways leading to glomerulosclerosis. Gα12 couples to numerous G-protein-coupled receptors (GPCRs) and regulates multiple epithelial responses, including proliferation, apoptosis, permeability and the actin cytoskeleton. Herein, we report that genetic activation of Gα12 in podocytes leads to time-dependent increases in proteinuria and glomerulosclerosis. To mimic activation of Gα12 pathways, constitutively active Gα12 (QL) was conditionally expressed in podocytes using Nphs2-Cre and LacZ/floxed QLα12 transgenic mice. Some QLα12(LacZ+/Cre+) mice developed proteinuria at 4-6 months, and most were proteinuric by 12 months. Proteinuria increased with age, and by 12-14 months, many demonstrated glomerulosclerosis with ultrastructural changes, including foot process fusion and both mesangial and subendothelial deposits. QLα12(LacZ+/Cre+) mice showed no changes in podocyte number, apoptosis, proliferation or Rho/Src activation. Real-time PCR revealed no significant changes in Nphs1, Nphs2, Cd2ap or Trpc6 expression, but Col4a2 message was increased in younger and older mice, while Col4a5 was decreased in older mice. Confocal microscopy revealed disordered collagen IVα1/2 staining in older mice and loss of α5 without changes in other collagen IV subunits. Taken together, these studies suggest that Gα12 activation promotes glomerular injury without podocyte depletion through a novel mechanism regulating collagen (α)IV expression, and supports the notion that glomerular damage may accrue through persistent GPCR activation in podocytes.


Subject(s)
Collagen Type IV/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Glomerulosclerosis, Focal Segmental/metabolism , Podocytes/metabolism , Proteinuria/metabolism , Receptors, G-Protein-Coupled/metabolism , Acute-Phase Proteins/metabolism , Age Factors , Animals , Cells, Cultured , Female , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Genes, src/physiology , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron , Podocytes/pathology , Proteinuria/pathology
3.
Am J Pathol ; 178(3): 1092-105, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21356361

ABSTRACT

The release of nucleotides after injury activates purinergic receptors, leading to phosphorylation of site-specific residues on epidermal growth factor receptor (EGFR). To elucidate the differences between the injury-induced response and that induced by exogenous EGF, we examined recruitment of docking proteins, internalization of EGFR, and migration after injury. Injury induced by scratch wounds or stimulation by addition of UTP caused a brief internalization of EGFR, which paralleled the lesser association with growth factor receptor-bound protein 2 (Grb2) and phosphorylation of EGFR. The internalization caused by EGF was sustained and detected for longer than 60 minutes and correlated with phosphorylation of the receptor. The EGF caused recruitment of Grb2, phospholipase C-γ-1 (PLCγ1), Shc, and Src to EGFR. Glutathione S-transferase pull downs were performed, and glutathione S-transferase-PLCγ1 showed binding of Grb2 when stimulated with EGF but not with UTP or injury. Furthermore, UTP did not induce PLCγ1 phosphorylation, and the phosphorylation induced by EGF was attenuated by costimulation with UTP. The response to heparin-binding EGF was equivalent to that of EGF. Site-directed mutagenesis showed that phosphorylation of Y1068 and Y1086 of EGFR is required for repair. Together, our results show that injury and activation of purinergic receptors and direct activation of EGFR via EGF induce distinct downstream pathways.


Subject(s)
Epithelial Cells/enzymology , Epithelial Cells/pathology , ErbB Receptors/metabolism , Uridine Triphosphate/pharmacology , Wound Healing/drug effects , Animals , Cell Movement/drug effects , Cytoplasm/drug effects , Cytoplasm/metabolism , Endocytosis/drug effects , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , GRB2 Adaptor Protein/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Mutant Proteins/metabolism , Phospholipase C gamma/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Rabbits , Sus scrofa
4.
Am J Physiol Cell Physiol ; 299(2): C411-21, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20427708

ABSTRACT

Injury to epithelial cells results in the release of ATP and stimulation of purinergic receptors and is thought to alter cell migration and wound repair. Medium from the injured cells triggers Ca(2+) mobilization and phosphorylation of ERK, both of which are inhibited if the medium is pretreated with apyrase. To understand the wound repair mechanism that occurs with injury, our goal was to determine which purinergic receptor(s) was the critical player in the wound response. We hypothesize that the P2Y(2) receptor is the key player in the response of corneal epithelial cells to cell damage and subsequent repair events. Cells transfected with short interfering RNA to either P2Y(2) or P2Y(4) were stimulated either by injury or addition of UTP and imaged using fluo 3-AM to monitor changes in fluorescence. When cells with downregulated P2Y(2) receptors were injured or stimulated with UTP, the intensity of the Ca(2+) release was reduced significantly. However, when cells with downregulated P2Y(4) receptors were stimulated, only the UTP-induced Ca(2+) response was reduced significantly. In addition, downregulation of the P2Y(2) receptor inhibited wound closure compared with unstimulated cells or cells transfected with nontargeting sequence. This downregulation resulted also in an attenuation in phosphorylation of Src and ERK. Together, these data indicate that the P2Y(2) receptor plays a major biological role in the corneal injury response and repair mechanisms.


Subject(s)
Cell Movement/physiology , Cornea/physiology , Corneal Injuries , Epithelial Cells/physiology , Receptors, Purinergic P2/physiology , Wound Healing/physiology , Animals , Cattle , Cell Line, Transformed , Cell Migration Inhibition/genetics , Cornea/cytology , Down-Regulation/genetics , Gene Knockdown Techniques , Humans , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y2 , Wound Healing/genetics
5.
Mol Biol Cell ; 20(21): 4596-610, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19776354

ABSTRACT

Regulation of epithelial cell attachment and migration are essential for normal development and maintenance of numerous tissues. G proteins and integrins are critical signaling proteins regulating these processes, yet in polarized cells little is known about the interaction of these pathways. Herein, we demonstrate that G alpha 12 inhibits interaction of MDCK cells with collagen-I, the major ligand for alpha2 beta1 integrin. Activating G alpha 12 (QL point mutation or stimulating endogenous G alpha 12 with thrombin) inhibited focal adhesions and lamellipodia formation and led to impaired cell migration. Consistent with G alpha 12-regulated attachment to collagen-I, G alpha 12-silenced MDCK cells revealed a more adherent phenotype. Inhibiting Rho kinase completely restored normal attachment in G alpha 12-activated cells, and there was partial recovery with inhibition of Src and protein phosphatase pathways. G alpha 12 activation led to decreased phosphorylation of focal adhesion kinase and paxillin with displacement of alpha2 integrin from the focal adhesion protein complex. Using the MDCK cell 3D-tubulogenesis assay, activated G alpha 12 inhibited tubulogenesis and led to the formation of cyst-like structures. Furthermore, G alpha 12-silenced MDCK cells were resistant to thrombin-stimulated cyst development. Taken together, these studies provide direct evidence for G alpha 12-integrin regulation of epithelial cell spreading and migration necessary for normal tubulogenesis.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Collagen Type I/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Integrin alpha2beta1/metabolism , Kidney Tubules/growth & development , Animals , Cell Line , Collagen Type I/genetics , Dogs , Enzyme Activation , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Humans , Integrin alpha2beta1/genetics , Kidney Tubules/cytology , Kidney Tubules/metabolism , Paxillin/metabolism , Pseudopodia/metabolism , Signal Transduction/physiology , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
6.
Exp Eye Res ; 85(1): 130-41, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17490650

ABSTRACT

The early events that occur rapidly after injury trigger signal cascades that are essential for proper wound closure of corneal epithelial cells. We hypothesize that injury releases ATP, which stimulates purinergic receptors and elicits the phosphorylation of epidermal growth factor receptor (EGFR) tyrosine residues and subsequent cell migration by a MMP and HB-EGF dependent pathway. We demonstrated that the inhibition of purinergic receptors with the antagonist, Reactive Blue 2, abrogated the phosphorylation of EGFR and ERK. Pre-incubation of cells with the EGFR kinase inhibitor, AG1478, and subsequent stimulation by injury or ATP resulted in a decrease in phosphorylation of EGFR and migration. Furthermore, downregulation of EGFR by siRNA, inhibited the EGF-induced intracellular Ca(2+) wave. However, the response to injury and ATP was retained indicating the presence of two signaling pathways. Inhibition with either CRM197 or TIMP-3 decreased injury and nucleotide-induced phosphorylation of both EGFR and ERK. Incubation in the presence of a functional blocking antibody to HB-EGF also resulted in a decrease in the phosphorylation of EGFR. In addition, cell migration was inhibited by CRM197 and rescued when cells were incubated with HB-EGF. We showed that injury-induced phosphorylation of specific tyrosine residues and found that a similar pattern of phosphorylation was induced by trinucleotides. These studies indicate that injury-induced purinergic receptor activation leads to phosphorylation of EGFR, ERK and migration.


Subject(s)
Corneal Injuries , ErbB Receptors/metabolism , Nucleotides/metabolism , Adenosine Triphosphate/metabolism , Cell Line , Cell Movement/physiology , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor , Epithelial Cells/metabolism , Epithelium, Corneal/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , RNA, Small Interfering/metabolism , Receptors, Purinergic/metabolism , Signal Transduction/physiology , Triazines/pharmacology , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...