Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Adv Skin Wound Care ; 37(4): 180-196, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38354304

ABSTRACT

GENERAL PURPOSE: To review a practical and scientifically sound application of the wound bed preparation model for communities without ideal resources. TARGET AUDIENCE: This continuing education activity is intended for physicians, physician assistants, nurse practitioners, and registered nurses with an interest in skin and wound care. LEARNING OBJECTIVES/OUTCOMES: After participating in this educational activity, the participant will:1. Summarize issues related to wound assessment.2. Identify a class of drugs for the treatment of type II diabetes mellitus that has been shown to improve glycemia, nephroprotection, and cardiovascular outcomes.3. Synthesize strategies for wound management, including treatment in resource-limited settings.4. Specify the target time for edge advancement in chronic, healable wounds.


Chronic wound management in low-resource settings deserves special attention. Rural or underresourced settings (ie, those with limited basic needs/healthcare supplies and inconsistent availability of interprofessional team members) may not have the capacity to apply or duplicate best practices from urban or abundantly-resourced settings. The authors linked world expertise to develop a practical and scientifically sound application of the wound bed preparation model for communities without ideal resources. A group of 41 wound experts from 15 countries reached a consensus on wound bed preparation in resource-limited settings. Each statement of 10 key concepts (32 substatements) reached more than 88% consensus. The consensus statements and rationales can guide clinical practice and research for practitioners in low-resource settings. These concepts should prompt ongoing innovation to improve patient outcomes and healthcare system efficiency for all persons with foot ulcers, especially persons with diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Foot , Foot Ulcer , Humans , Delphi Technique , Diabetes Mellitus, Type 2/therapy , Diabetic Foot/diagnosis , Diabetic Foot/therapy , Resource-Limited Settings
3.
Acad Med ; 88(7): 960-6, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23702526

ABSTRACT

PURPOSE: The challenges for senior academic leadership in medicine are significant and becoming increasingly complex. Adapting to the rapidly changing environment of health care and medical education requires strong leadership and management skills. This article provides empirical evidence about the intricate needs of department chairs to provide insight into the design of support and development opportunities. METHOD: In an exploratory case study, 21 of 25 (84%) department chairs within a faculty of medicine at a large Canadian university participated in semistructured interviews from December 2009 to February 2010. The authors conducted an inductive thematic analysis and identified a coding structure through an iterative process of relating and grouping of emerging themes. RESULTS: These participants were initially often insufficiently prepared for the demands of their roles. They identified a specific set of needs. They required cultural and structural awareness to navigate their hospital and university landscapes. A comprehensive network of support was necessary for eliciting advice and exchanging information, strategy, and emotional support. They identified a critical need for infrastructure growth and development. Finally, they stressed that they needed improvement in both effective interpersonal and influence skills in order to meet their mandate. CONCLUSIONS: Given the complexities and emotional burden of their role, it is necessary for chairs to have a range of supports and capabilities to succeed in their roles. Their leadership effectiveness can be enhanced by providing transitional processes and supports, development, and mentoring as well as facilitating the development of communities of peers.


Subject(s)
Academic Medical Centers/organization & administration , Faculty, Medical , Leadership , Aged , Communication , Female , Humans , Interprofessional Relations , Male , Middle Aged , Needs Assessment , Organizational Case Studies , Workforce
4.
Am J Physiol Endocrinol Metab ; 301(4): E713-26, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21712532

ABSTRACT

Hyperglycemia augments flux through the hexosamine biosynthetic pathway and subsequent O-linkage of single ß-N-acetyl-d-glucosamine moieties to serine and threonine residues on cytoplasmic and nuclear proteins (O-GlcNAcylation). Perturbations in this posttranslational modification have been proposed to promote glomerular matrix accumulation in diabetic nephropathy, but clear evidence and mechanism are lacking. We tested the hypothesis that O-GlcNAcylation enhances profibrotic signaling in rat mesangial cells. An adenovirus expressing shRNA directed against O-GlcNAc transferase (OGT) markedly reduced basal and high-glucose-stimulated O-GlcNAcylation. Interestingly, O-GlcNAc depletion prevented high-glucose-induced p38 mitogen-activated protein kinase (MAPK) and c-Jun NH(2)-terminal kinase phosphorylation. Downstream of p38, O-GlcNAc controlled the expression of plasminogen activator inhibitor-1, fibronectin, and transforming growth factor-ß, important factors in matrix accumulation in diabetic nephropathy. Treating mesangial cells with thiamet-G, a highly selective inhibitor of O-GlcNAc-specific hexosaminidase (O-GlcNAcase), increased O-GlcNAcylation and p38 phosphorylation. The high-glucose-stimulated kinase activity of apoptosis signal-regulating kinase 1 (ASK1), an upstream MAPK kinase kinase for p38 that is negatively regulated by Akt, was inhibited by OGT shRNA. Akt Thr(308) and Ser(473) phosphorylation were enhanced following OGT shRNA expression in high-glucose-exposed mesangial cells, but high-glucose-induced p38 phosphorylation was not attenuated by OGT shRNA in cells pretreated with the phosphatidylinositol 3-kinase inhibitor LY-294002. OGT shRNA also reduced high-glucose-stimulated reactive oxygen species (ROS) formation. In contrast, diminished O-GlcNAcylation caused elevated ERK phosphorylation and PKCδ membrane translocation. Thus, O-GlcNAcylation is coupled to profibrotic p38 MAPK signaling by high glucose in part through Akt and possibly through ROS.


Subject(s)
Acetylglucosamine/metabolism , Glucose/metabolism , Mesangial Cells/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Glucose/pharmacology , MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Kinase Kinase 5/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
5.
Exp Diabetes Res ; 2009: 910783, 2009.
Article in English | MEDLINE | ID: mdl-19609456

ABSTRACT

Peroxisome proliferator-activated receptor (PPARgamma), a ligand-dependent transcription factor, negatively modulates high glucose effects. We postulated that rosiglitazone (RSG), an activator of PPARgamma prevents the upregulation of vascular endothelial growth factor (VEGF) and collagen IV by mesangial cells exposed to high glucose. Primary cultured rat mesangial cells were growth-arrested in 5.6 mM (NG) or 25 mM D-glucose (HG) for up to 48 hours. In HG, PPARgamma mRNA and protein were reduced within 3 h, and enhanced ROS generation, expression of p22(phox), VEGF and collagen IV, and PKC-zeta membrane association were prevented by RSG. In NG, inhibition of PPARgamma caused ROS generation and VEGF expression that were unchanged by RSG. Reduced AMP-activated protein kinase (AMPK) phosphorylation in HG was unchanged with RSG, and VEGF expression was unaffected by AMPK inhibition. Hence, PPARgamma is a negative modulator of HG-induced signaling that acts through PKC-zeta but not AMPK and regulates VEGF and collagen IV expression by mesangial cells.


Subject(s)
Collagen Type IV/metabolism , Glucose/metabolism , Mesangial Cells/drug effects , PPAR gamma/agonists , PPAR gamma/metabolism , Thiazolidinediones/pharmacology , Vascular Endothelial Growth Factor A/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Chromans/pharmacology , Collagen Type IV/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Membranes/chemistry , Mesangial Cells/metabolism , Mesangial Cells/ultrastructure , NADPH Oxidases/chemistry , NADPH Oxidases/drug effects , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Protein Kinase C/metabolism , RNA, Messenger , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Response Elements/genetics , Rosiglitazone , Transfection , Troglitazone , Vascular Endothelial Growth Factor A/genetics
6.
Nephrol Dial Transplant ; 24(6): 1782-90, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19155535

ABSTRACT

BACKGROUND: Activation of protein kinase C (PKC) has been implicated in the pathogenesis of diabetic nephropathy where therapy targeting the beta isoform of this enzyme is in advanced clinical development. However, PKC-beta is also increased in various forms of human glomerulonephritis with several potentially nephrotoxic factors, other than high glucose, resulting in PKC-beta activation. Accordingly, we sought to examine the effects of PKC-beta inhibition in a non-diabetic model of progressive kidney disease. METHODS: Subtotally nephrectomized (STNx) rats were randomly assigned to receive either the selective PKC-beta inhibitor, ruboxistaurin or vehicle. In addition to functional and structural parameters, gene expression of the podocyte slit-pore diaphragm protein, nephrin, was also assessed. RESULTS: STNx animals developed hypertension, proteinuria and reduced glomerular filtration rate (GFR) in association with marked glomerulosclerosis and tubulointerstitial fibrosis. Glomerular nephrin expression was also reduced. Without affecting blood pressure, ruboxistaurin treatment attenuated the impairment in GFR and reduced the extent of both glomerulosclerosis and tubulointerstitial fibrosis in STNx rats. In contrast, neither proteinuria nor the reduction in nephrin expression was improved by ruboxistaurin. CONCLUSIONS: These findings indicate firstly that PKC-beta inhibition may provide a new therapeutic strategy in non-diabetic kidney disease and secondly that improvement in GFR is not inextricably linked to reduction in proteinuria.


Subject(s)
Enzyme Inhibitors/therapeutic use , Indoles/therapeutic use , Kidney Diseases/drug therapy , Kidney Diseases/enzymology , Maleimides/therapeutic use , Protein Kinase C/antagonists & inhibitors , Animals , Disease Models, Animal , Glomerular Filtration Rate/drug effects , Humans , Kidney/drug effects , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Male , Membrane Proteins/genetics , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Protein Kinase C beta , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/pharmacology
7.
Am J Physiol Renal Physiol ; 295(6): F1705-14, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18815221

ABSTRACT

Conversion of normally quiescent mesangial cells into extracellular matrix-overproducing myofibroblasts in response to high ambient glucose and transforming growth factor (TGF)-beta(1) is central to the pathogenesis of diabetic nephropathy. Previously, we reported that mesangial cells respond to high glucose by generating reactive oxygen species (ROS) from NADPH oxidase dependent on protein kinase C (PKC) -zeta activation. We investigated the role of TGF-beta(1) in this action of high glucose on primary rat mesangial cells within 1-48 h. Both high glucose and exogenous TGF-beta(1) stimulated PKC-zeta kinase activity, as measured by an immune complex kinase assay and immunofluorescence confocal cellular imaging. In high glucose, Akt Ser473 phosphorylation appeared within 1 h and Smad2/3 nuclear translocation was prevented with neutralizing TGF-beta(1) antibodies. Neutralizing TGF-beta(1) antibodies, or a TGF-beta receptor kinase inhibitor (LY364947), or a phosphatidylinositol 3,4,5-trisphosphate (PI3) kinase inhibitor (wortmannin), prevented PKC-zeta activation by high glucose. TGF-beta(1) also stimulated cellular membrane translocation of PKC-alpha, -beta(1), -delta, and -epsilon, similar to high glucose. High glucose and TGF-beta(1) enhanced ROS generation by mesangial cell NADPH oxidase, as detected by 2,7-dichlorofluorescein immunofluorescence. This response was abrogated by neutralizing TGF-beta(1) antibodies, LY364947, or a specific PKC-zeta pseudosubstrate peptide inhibitor. Expression of constitutively active PKC-zeta in normal glucose caused upregulation of p22(phox), a likely mechanism of NADPH oxidase activation. We conclude that very early responses of mesangial cells to high glucose include autocrine TGF-beta(1) stimulation of PKC isozymes including PI3 kinase activation of PKC-zeta and consequent generation of ROS by NADPH oxidase.


Subject(s)
Glomerular Mesangium/physiology , Hyperglycemia/physiopathology , NADPH Oxidases/metabolism , Protein Kinase C/metabolism , Transforming Growth Factor beta1/physiology , Animals , DNA Primers , Enzyme Activation , Glomerular Mesangium/physiopathology , Kinetics , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
8.
Nephron Exp Nephrol ; 109(2): e46-56, 2008.
Article in English | MEDLINE | ID: mdl-18600029

ABSTRACT

BACKGROUND/AIMS: We postulated that alpha-smooth muscle actin expressed in primary cultured mesangial cells is down-regulated in 3-dimensional (D) culture and up-regulated by high glucose and growth factors. METHODS: Primary rat mesangial cells were growth-arrested in 5.6 mM (NG) or 30 mM (HG) glucose for 14 days in 3-D Matrigel. Alpha-SM actin expression was analyzed by immunoblotting, real-time PCR and by alpha-SM actin promoter activity in response to 24 h stimulation with endothelin-1 (ET-1), angiotensin II (Ang II) or HG. RESULTS: Alpha-SM actin mRNA, protein and promoter activity were reduced to significantly lower levels in 3-D cells compared to cells in 2-D. Up-regulation of alpha-SM expression was stimulated by ET-1, Ang II and HG. Specific inhibitors of protein kinase C (PKC)-alpha, -beta or -zeta prevented alpha-SM upregulation in HG. In NG, PKC and ERK1/2 activation were required for alpha-SM actin accumulation in 3-D in response to ET-1 or Ang II. In HG, enhanced expression of alpha-SM actin in response to ET-1 or Ang II was unchanged during PKC or ERK1/2 inhibition. CONCLUSION: Mesangial cells in 3-D express low levels of alpha-SM actin representing a more differentiated state. Regulation of alpha-SM actin expression is dependent on specific PKC isozyme and ERK1/2 signaling.


Subject(s)
Actins/metabolism , Extracellular Matrix/metabolism , Glucose/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/physiology , Mesangial Cells/physiology , Protein Kinase C/metabolism , Animals , Cell Culture Techniques/methods , Cells, Cultured , Extracellular Matrix/chemistry , Gene Expression Regulation/physiology , Mesangial Cells/cytology , Rats , Rats, Sprague-Dawley
10.
Proteins ; 72(1): 447-60, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18214957

ABSTRACT

Protein kinase C (PKC) isozymes are an important class of enzymes in cell signaling and as drug targets. They are involved in specific pathways and have selectivity towards certain ligands, despite their high sequence similarities. Ruboxistaurin is a specific inhibitor of PKC-beta. To understand the molecular determinants for the selectivity of ruboxistaurin, we derived the three-dimensional structures of the kinase domains of PKC-alpha, -betaI, and -zeta using homology modeling. Several binding orientations of ruboxistaurin in the binding sites of these PKC catalytic domains were analyzed, and a putative alternative binding site for PKC-zeta was identified in its kinase domain. The calculated free energy of binding correlates well with the IC(50) of the inhibitor against each PKC isozyme. A residue-based energy decomposition analysis attributed the binding free energy to several key residues in the catalytic sites of these enzymes, revealing potential protein-ligand interactions responsible for ligand binding. The contiguous binding site revealed in the catalytic domain of PKC-zeta provides avenues for selective drug design. The details of structural nuances for specific inhibition of PKC isozymes are presented in the context of the three-dimensional structures of this important class of enzymes.


Subject(s)
Indoles/metabolism , Maleimides/metabolism , Protein Kinase C/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Binding Sites , Hydrogen Bonding , Indoles/chemistry , Isoenzymes/chemistry , Isoenzymes/metabolism , Maleimides/chemistry , Models, Molecular , Molecular Sequence Data , Protein Kinase C/chemistry , Protein Structure, Tertiary , Sequence Alignment , Structural Homology, Protein , Substrate Specificity , Thermodynamics
11.
Am J Physiol Endocrinol Metab ; 293(5): E1280-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17711990

ABSTRACT

Vascular endothelial growth factor (VEGF) is implicated in the development of proteinuria in diabetic nephropathy. High ambient glucose present in diabetes stimulates VEGF expression in several cell types, but the molecular mechanisms are incompletely understood. Here primary cultured rat mesangial cells served as a model to investigate the signal transduction pathways involved in high-glucose-induced VEGF expression. Exposure to high glucose (25 mM) significantly increased VEGF mRNA evaluated by real-time PCR by 3 h, VEGF cellular protein content assessed by immunoblotting or immunofluorescence within 24 h, and VEGF secretion by 24 h. High-glucose-induced VEGF expression was blocked by an antioxidant, Tempol, and antisense oligonucleotides directed against p22(phox), a NADPH oxidase subunit. Inhibition of protein kinase C (PKC)-beta(1) with the specific pharmacological inhibitor LY-333531 or inhibition of PKC-zeta with a cell permeable specific pseudosubstrate peptide also prevented enhanced VEGF expression in high glucose. Enhanced VEGF secretion in high glucose was prevented by Tempol, PKC-beta(1), or PKC-zeta inhibition. In normal glucose (5.6 mM), overexpression of p22(phox) or constitutively active PKC-zeta enhanced VEGF expression. Hypoxia inducible factor-1alpha protein was significantly increased in high glucose only by 24 h, suggesting a possible contribution to high-glucose-stimulated VEGF expression at later time points. Thus reactive oxygen species generated by NADPH oxidase, and both PKC-beta(1) and -zeta, play important roles in high-glucose-stimulated VEGF expression and secretion by mesangial cells.


Subject(s)
Diabetic Nephropathies/metabolism , Glucose/administration & dosage , Mesangial Cells/metabolism , Molecular Chaperones/metabolism , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Cyclic N-Oxides/pharmacology , Diabetic Nephropathies/enzymology , Fluorescent Antibody Technique , Glucose/metabolism , Indoles/pharmacology , Maleimides/pharmacology , Molecular Chaperones/antagonists & inhibitors , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , NADPH Oxidases/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C beta , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Spin Labels , Vascular Endothelial Growth Factor A/genetics
13.
Endocrinology ; 148(3): 1089-98, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17110421

ABSTRACT

Long-chain, monounsaturated fatty acids (FAs) stimulate secretion of the incretin hormone, glucagon-like peptide-1 (GLP-1) from the intestinal L cell. Because the atypical protein kinase C (PKC), PKCzeta, is involved in FA signaling in many cells, the role of PKCzeta in FA-induced GLP-1 secretion was investigated, using the murine GLUTag L cell line and primary rat intestinal L cells. GLUTag cells expressed mRNA for several PKC isoforms, including PKCzeta, and PKCzeta protein was localized throughout the cytoplasm in GLUTag and primary L cells as well as normal mouse and rat L cells. Treatment with oleic acid (150-1000 microm) for 2 h increased GLP-1 secretion (P < 0.001), and this was abrogated by the PKCzeta inhibitor ZI (P < 0.05) and PKCzeta small interfering RNA transfection (P < 0.05) but not inhibition of classical/novel PKC isoforms. Although most PKCzeta was localized in the particulate compartment of GLUTag cells, oleate treatment did not alter PKCzeta levels or activity in this cell fraction. GLUTag cells expressed mRNA for the Gq-coupled FA receptor GPR120; however, oleic acid did not induce any changes in Akt, MAPK, or calcium, and pretreatment with LY294002 and PD98059 to inhibit phosphatidylinositol 3-kinase and MAPK, respectively, did not prevent the effects of oleic acid. Finally, GLUTag cells also released GLP-1 in response to arachidonic acid (P < 0.001) but were not affected by other long-chain FAs. These findings demonstrate that PKCzeta is required for oleic acid-induced GLP-1 secretion. This enzyme may therefore serve as a therapeutic target to enhance GLP-1 release in type 2 diabetes.


Subject(s)
Enteroendocrine Cells/drug effects , Glucagon-Like Peptide 1/metabolism , Oleic Acid/pharmacology , Protein Kinase C/physiology , Animals , Cells, Cultured , Embryo, Mammalian , Enteroendocrine Cells/metabolism , Mice , Protein Kinase C/metabolism , Rats , Rats, Wistar , Tissue Distribution
14.
Endocrinology ; 147(1): 222-31, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16365142

ABSTRACT

Metabolic flux through the hexosamine biosynthetic pathway (HBP) is increased in the presence of high glucose (HG) and potentially stimulates the expression of genes associated with the development of diabetic nephropathy. A number of synthetic processes are coupled to the HBP, including enzymatic intracellular O-glycosylation (O-GlcNAcylation), the addition of single O-linked N-acetylglucosamine monosaccharides to serine or threonine residues. Despite much data linking flow through the HBP and gene expression, the exact contribution of O-GlcNAcylation to HG-stimulated gene expression remains unclear. In glomerular mesangial cells, HG-stimulated plasminogen activator inhibitor-1 (PAI-1) gene expression requires the HBP and the transcription factor, Sp1. In this study, the specific role of O-GlcNAcylation in HG-induced PAI-1 expression was tested by limiting this modification with a dominant-negative O-linked N-acetylglucosamine transferase, by overexpression of neutral beta-N-acetylglucosaminidase, and by knockdown of O-linked beta-N-acetylglucosamine transferase expression by RNA interference. Decreasing O-GlcNAcylation by these means inhibited the ability of HG to increase endogenous PAI-1 mRNA and protein levels, the activity of a PAI-1 promoter-luciferase reporter gene, and Sp1 transcriptional activation. Conversely, treatment with the beta-N-acetylglucosaminidase inhibitor, O-(2-acetamido-2-deoxy-d-glucopyranosylidene)amino-N-phenylcarbamate, in the presence of normal glucose increased Sp1 O-GlcNAcylation and PAI-1 mRNA and protein levels. These findings demonstrate for the first time that among the pathways served by the HBP, O-GlcNAcylation, is obligatory for HG-induced PAI-1 gene expression and Sp1 transcriptional activation in mesangial cells.


Subject(s)
Glucose/metabolism , Mesangial Cells/physiology , Plasminogen Activator Inhibitor 1/genetics , Protein Processing, Post-Translational , Sp1 Transcription Factor/metabolism , Acetylglucosamine/metabolism , Animals , Base Sequence , Gene Expression Regulation , Glycosylation , Plasmids , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Transcriptional Activation
15.
Kidney Int ; 68(6): 2526-41, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16316329

ABSTRACT

BACKGROUND: We postulated that in mesangial cells exposed to high glucose, protein kinase C-zeta (PKC-zeta) is necessary for the generation of reactive oxygen species (ROS) by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and that the requirement of PKC-zeta for filamentous (F)-actin disassembly may involve ROS. To identify signaling mechanisms relevant to PKC-zeta activation and ROS generation, including phosphoinositide 3 kinase (PI3 kinase), we examined mesangial cell stimulation with platelet-derived growth factor (PDGF). METHODS: In primary rat mesangial cells cultured in 5.6 mmol/L or 30 mmol/L d-glucose, PKC-zeta expression was identified with immunoblotting and activity was analyzed in cell membrane immunoprecipitates and by confocal immunofluorescence imaging. ROS generation was measured by dichlorofluorescein fluorescence using confocal microscopy and was inhibited by transfection of antisense against NADPH subunits p22(phox) or p47(phox) or with Tempol. F-actin disassembly was observed by dual-channel confocal fluorescence imaging. PI3 kinase activity was detected by immunoblotting of phosphorylated Akt. RESULTS: In high glucose, generation of NADPH oxidase-dependent ROS was dependent on PKC-zeta. Conversely, sustained PKC-zeta activity was dependent on ROS generation, suggesting a positive feedback. PKC-zeta-dependent F-actin disassembly in high glucose required ROS generation. PDGF stimulated NADPH oxidase generation of ROS through a PKC-zeta mechanism that was independent of Akt phosphorylation and remained unchanged in high glucose. CONCLUSION: In high glucose, mesangial cell PKC-zeta is required for ROS generation from NADPH oxidase similar to PDGF stimulation of PKC-zeta-dependent ROS generation through a pathway independent of PI3 kinase. F-actin disassembly in high glucose also requires ROS. A positive feedback loop occurs between ROS and the activation of PKC-zeta in high glucose.


Subject(s)
Glucose/pharmacology , Mesangial Cells/enzymology , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , Actins/metabolism , Animals , Cells, Cultured , Male , Membrane Transport Proteins/genetics , Mesangial Cells/cytology , Mesangial Cells/drug effects , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Oligonucleotides, Antisense , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/genetics , Platelet-Derived Growth Factor/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
16.
Am J Physiol Renal Physiol ; 289(5): F1078-87, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15998840

ABSTRACT

In high glucose, glomerular mesangial cells (MCs) demonstrate impaired Ca(2+) signaling in response to seven-transmembrane receptor stimulation. To identify the mechanism, we first postulated decreased release from intracellular stores. Intracellular Ca(2+) was measured in fluo-3-loaded primary cultured rat MCs using confocal fluorescence microscopy. In high glucose (HG) 30 mM for 48 h, the 25 nM ionomycin-stimulated intracellular Ca(2+) response was reduced to 82% of that observed in normal glucose (NG). In NG 5.6 mM, Ca(2+) responses to endothelin (ET)-1 and platelet-derived growth factor (PDGF) were unchanged in cells cultured in 50 nM Ca(2+) vs. 1.8 mM Ca(2+). Depletion of intracellular Ca(2+) stores with thapsigargin eliminated ET-1-stimulated Ca(2+) responses. Incubation in 30 mM glucose (HG) for 48 h or stimulation with phorbol myristate acetate (PMA) for 10 min eliminated the Ca(2+) response to ET-1 but had no effect on the PDGF response. Downregulation of protein kinase C (PKC) with 24-h PMA or inhibition with Gö6976 in HG normalized the Ca(2+) response to ET-1. Because ET-1 and PDGF stimulate Ca(2+) signaling through different phospholipase C pathways, we hypothesized that, in HG, PKC selectively phosphorylates and inhibits PLC-beta(3). Using confocal immunofluorescence imaging, in NG, a 1.6- to 1.7-fold increase in PLC-beta(3) Ser(1105) phosphorylation was observed following PMA or ET-1 stimulation for 10 min. In HG, immunofluorescent imaging and immunoblotting showed increased PLC-beta(3) phosphorylation, without change in total PLC-beta(3), which was reversed with 24-h PMA or Gö6976. We conclude that reduced Ca(2+) signaling in HG cannot be explained by reduced Ca(2+) stores but is due to conventional PKC-dependent phosphorylation and inactivation of PLC-beta(3).


Subject(s)
Calcium Signaling/physiology , Hyperglycemia/complications , Isoenzymes/metabolism , Protein Kinase C/metabolism , Type C Phospholipases/metabolism , Animals , Down-Regulation , Endothelin-1/physiology , Gene Expression Profiling , Glomerular Mesangium/cytology , Glomerular Mesangium/physiology , Glucose/physiology , Isoenzymes/antagonists & inhibitors , Male , Phospholipase C beta , Phosphorylation , Rats , Rats, Sprague-Dawley , Type C Phospholipases/antagonists & inhibitors
18.
Exp Eye Res ; 80(5): 651-62, 2005 May.
Article in English | MEDLINE | ID: mdl-15862172

ABSTRACT

Retinal pigment epithelial (RPE) cells express vascular endothelial growth factor (VEGF) in response to high glucose or hypoxia. We hypothesised that VEGF expression and secretion by RPE cells in high glucose and hypoxia are regulated by protein kinase C (PKC). Primary cultured RPE cells from Sprague-Dawley rats were growth-arrested for 48 hr in 0.5% FBS in 5.6 or 30 mm D-glucose. Cells were exposed to hypoxic conditions (<1% O(2), 5% CO(2)) for the last 15-18 hr of growth-arrest. PKC -alpha, -beta(1), -delta, -epsilon, and -zeta were expressed by RPE cells and exposure to high glucose for 48 hr had no effect on expression as demonstrated by Western immunoblotting. High glucose, hypoxia or VEGF stimulated translocation of a number of the PKC isozymes to the membrane or particulate fractions implying activation. In response to high glucose or acute phorbol myristate acetate (PMA) stimulation, VEGF mRNA analysed by RT-PCR was increased. Intracellular VEGF protein identified by immunoblotting and confocal immunofluorescence imaging was significantly increased by high glucose, hypoxia or acute PMA stimulation. Calphostin C or a specific inhibitor of PKC-zeta prevented high glucose-stimulated VEGF expression in high glucose. VEGF secretion, as measured by ELISA in the culture medium, was enhanced in hypoxia but not in high glucose. Following exposure of RPE cells to PMA for 24 hr, PKC-delta was significantly down regulated, whereas PKC-alpha, -beta, -epsilon and -zeta remained unchanged. Secretion of VEGF in normal or high glucose, or hypoxia was significantly reduced following treatment with PMA for 24 hr but not with the PKC-zeta inhibitor. We conclude that in high glucose and hypoxia PKC isozymes are activated and are necessary for VEGF expression. Secretion of VEGF is enhanced in hypoxia and appears to be regulated by PKC-delta. RPE cells may contribute to the pathogenesis of retinopathy caused by high glucose and hypoxia through the expression and secretion of VEGF that are regulated by PKC isozymes.


Subject(s)
Glucose/pharmacology , Isoenzymes/metabolism , Pigment Epithelium of Eye/chemistry , Protein Kinase C/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Blotting, Western/methods , Cell Hypoxia , Cells, Cultured , Enzyme Activation , Enzyme-Linked Immunosorbent Assay/methods , Isoenzymes/antagonists & inhibitors , Male , Microscopy, Confocal , Naphthalenes/pharmacology , Pigment Epithelium of Eye/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-delta , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tetradecanoylphorbol Acetate/pharmacology , Vascular Endothelial Growth Factor A/metabolism
19.
Curr Hypertens Rep ; 7(2): 148-54, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15748541

ABSTRACT

High glucose activates a myriad of signaling and gene expression pathways in non-insulin-dependent target cells causing diabetes complications. One of the earliest responses to high glucose by vascular cells is the generation of reactive oxygen species (ROS) that act directly on intracellular proteins and DNA, or indirectly as second messengers, transforming these cells into disease phenotypes. ROS are produced by mitochondria and/or NADPH oxidase in all target cells exposed to high glucose studied to date. Reports using cell cultures and diabetic animal models indicate that inhibition of ROS generation prevents the amplification of signaling and gene expression that are implicated in vascular complications. These models convincingly demonstrate that maneuvers preventing ROS production attenuate or completely abrogate early micro- and macrovascular end-organ damage of diabetes, including nephropathy, retinopathy, and large-vessel atherosclerosis. Attention now turns to the development of more effective antioxidants that could be used in clinical trials in the prevention and treatment of diabetes complications.


Subject(s)
Blood Vessels/pathology , Diabetic Angiopathies/drug therapy , Indoles/therapeutic use , Maleimides/therapeutic use , Protein Kinase C/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Animals , Blood Vessels/metabolism , Cells, Cultured , DNA/genetics , Diabetic Angiopathies/genetics , Diabetic Angiopathies/metabolism , Gene Expression/drug effects , Gene Expression/physiology , Humans , Intracellular Space/metabolism , Intracellular Space/ultrastructure , Protein Kinase C beta , Reactive Oxygen Species/antagonists & inhibitors , Treatment Outcome
20.
J Biol Chem ; 278(36): 33951-62, 2003 Sep 05.
Article in English | MEDLINE | ID: mdl-12821678

ABSTRACT

High glucose (HG) is the underlying factor contributing to long term complications of diabetes mellitus. The molecular mechanisms transforming the glomerular mesangial cell phenotype to cause nephropathy including diacylglycerol-sensitive protein kinase C (PKC) are still being defined. Reactive oxygen species (ROS) have been postulated as a unifying mechanism for HG-induced complications. We hypothesized that in HG an interaction between ROS generation, from NADPH oxidase, and PKC suppresses mesangial Ca2+ signaling in response to endothelin-1 (ET-1). In primary rat mesangial cells, growth-arrested (48 h) in 5.6 mM (NG) or 30 mm (HG) glucose, the total cell peak [Ca2+]i response to ET-1 (50 nM) was 630 +/- 102 nM in NG and was reduced to 159 +/- 15 nM in HG, measured by confocal imaging. Inhibition of PKC with phorbol ester down-regulation in HG normalized the ET-1-stimulated [Ca2+]i response to 541 +/- 74 nM. Conversely, an inhibitory peptide specific for PKC-zeta did not alter Ca2+ signaling in HG. Furthermore, overexpression of conventional PKC-beta or novel PKC-delta in NG diminished the [Ca2+]i response to ET-1, reflecting the condition observed in HG. Likewise, catalase or p47phox antisense oligonucleotide normalized the [Ca2+]i response to ET-1 in HG to 521 +/- 58 nM and 514 +/- 48 nM, respectively. Pretreatment with carbonyl cyanide m-chlorophenylhydrazone or rotenone did not restore Ca2+ signaling in HG. Detection of increased intracellular ROS in HG by dichlorofluorescein was inhibited by catalase, diphenyleneiodonium, or p47phox antisense oligonucleotide. HG increased p47phox mRNA by 1.7 +/- 0.1-fold as measured by reverse transcriptase-PCR. In NG, H2O2 increased membrane-enriched PKC-beta and -delta, suggesting activation of these isozymes. HG-enhanced immunoreactivity of PKC-delta visualized by confocal imaging was attenuated by diphenyleneiodium chloride. Thus, mesangial cell [Ca2+]i signaling in response to ET-1 in HG is attenuated through an interaction mechanism between NADPH oxidase ROS production and diacylglycerol-sensitive PKC.


Subject(s)
Calcium/metabolism , Diglycerides/pharmacology , Endothelin-1/metabolism , Glucose/metabolism , NADPH Oxidases/metabolism , Protein Kinase C/chemistry , Actins/metabolism , Animals , Catalase/metabolism , Cell Membrane/metabolism , Cells, Cultured , Diglycerides/metabolism , Down-Regulation , Electron Transport , Green Fluorescent Proteins , Hydrogen Peroxide/pharmacology , Ionomycin/pharmacology , Ionophores/pharmacology , Kidney Glomerulus/cytology , Luminescent Proteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Mitochondria/metabolism , Oligonucleotides/metabolism , Oligonucleotides, Antisense/pharmacology , Onium Compounds/chemistry , Peptides/chemistry , Phosphoproteins/metabolism , Protein Isoforms , Protein Kinase C/metabolism , Protein Kinase C beta , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL