Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Vis ; 15: 1599-610, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19693287

ABSTRACT

PURPOSE: Platelet-derived growth factor (PDGF) and insulin promote the survival of neuronal cells, including retinal ganglion cells (RGCs), via activation of phosphoinositide 3-kinase (PI 3-kinase)/Akt signaling. Of importance, recent studies have shown that imatinib inhibition of PDGF receptors induces retinal toxicity in some patients. To date, the extent of activation and the functional significance of insulin-induced PI 3-kinase/Akt signaling remain unclear in the context of dysregulated PDGF receptor signaling in retinal cells. In the present study, we tested the hypothesis that the pro-survival effect of insulin-induced PI 3-kinase/Akt signaling is compromised by imatinib inhibition of PDGF receptor signaling in RGCs. METHODS: RGC-5 cells were subjected to acute and long-term treatments with imatinib, a PDGF receptor tyrosine kinase inhibitor. Afterwards, the changes in RGC phenotype and apoptotic markers were assessed by fluorescence and phase contrast microscopy and caspase-3/poly(ADP-ribose) polymerase (PARP) cleavage, respectively. In addition, imatinib regulation of PDGF- and insulin-induced PI 3-kinase/Akt survival signaling was determined by immunoblot analyses, immunoprecipitation, and in vitro PI 3-kinase assays. RESULTS: Treatment of RGC-5 cells with imatinib for up to 48 h resulted in apoptosis, which was not rescued by insulin supplementation. The apoptotic phenotype was associated with upregulation of cleaved caspase-3 and cleaved poly(ADP-ribose) polymerase. Time dependency experiments revealed that imatinib-mediated apoptosis was preceded by early and sustained abrogation of PDGF-induced increases in PDGF receptor tyrosine phosphorylation and phosphotyrosine-associated PI 3-kinase activity. In addition, imatinib inhibited PDGF-induced downstream phosphorylation of Akt, GSK-3beta, and p70S6kinase. However, imatinib exposure did not affect insulin-induced insulin receptor substrate (IRS)-associated PI 3-kinase activity and the downstream phosphorylation of Akt, GSK-3beta, and p70S6kinase. CONCLUSIONS: Together, these data indicate that disruption of PDGF receptor signaling compromises the pro-survival effect of insulin-induced IRS-dependent PI 3-kinase/Akt signaling in RGCs, and that the maintenance of PDGF-induced PI 3-kinase/Akt signaling is critical for the survival of retinal neuronal cells.


Subject(s)
Apoptosis/drug effects , Insulin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Piperazines/pharmacology , Platelet-Derived Growth Factor/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Retinal Ganglion Cells/drug effects , Signal Transduction/drug effects , Animals , Benzamides , Caspase 3/metabolism , Cell Line , Cell Survival/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Imatinib Mesylate , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Platelet-Derived Growth Factor/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Rats , Receptor, Insulin/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/enzymology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
2.
Am J Physiol Heart Circ Physiol ; 295(4): H1657-68, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18723771

ABSTRACT

Diglycerides (DGs) are phospholipid-derived second messengers that regulate PKC-dependent signaling pathways. Distinct species of DGs are generated from inflammatory cytokines and growth factors. Growth factors increase diacyl- but not ether-linked DG species, whereas inflammatory cytokines predominately generate alkyl, acyl- and alkenyl, acyl-linked DG species in rat mesenchymal cells. These DG species have been shown to differentially regulate protein kinase C (PKC) isotypes. Ester-linked diacylglycerols activate PKC-epsilon and cellular proliferation in contrast to ether-linked DGs, which lead to growth arrest through the inactivation of PKC-epsilon. It is now hypothesized that ether-linked DGs inhibit mitogenesis through the inactivation of ERK and/or Akt signaling cascades. We demonstrate that cell-permeable ether-linked DGs reduce vascular smooth muscle cell growth by inhibiting platelet-derived growth factor-stimulated ERK in a PKC-epsilon-dependent manner. This inhibition is specific to the ERK pathway, since ether-linked DGs do not affect growth factor-induced activation of other family members of the MAPKs, including p38 MAPK and c-Jun NH(2)-terminal kinases. We also demonstrate that ether-linked DGs reduce prosurvival phosphatidylinositol 3-kinase (PI3K)/Akt signaling, independent of PKC-epsilon, by diminishing an interaction between the subunits of PI3K and not by affecting protein phosphatase 2A or lipid (phosphatase and tensin homologue deleted in chromosome 10) phosphatases. Taken together, our studies identify ether-linked DGs as potential adjuvant therapies to limit vascular smooth muscle migration and mitogenesis in atherosclerotic and restenotic models.


Subject(s)
Cell Proliferation , Diglycerides/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glyceryl Ethers/metabolism , Muscle, Smooth, Vascular/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Becaplermin , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/enzymology , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Protein Kinase C-epsilon/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-sis , Rats , Signal Transduction/drug effects
3.
Invest Ophthalmol Vis Sci ; 49(8): 3687-98, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18421086

ABSTRACT

PURPOSE: To compare PDGF- and insulin/IGF-1-induced class I(A) PI 3-kinase/Akt survival signaling in normal retinas and retinal ganglion cells (RGCs). METHODS: Normal rat retinas and RGC-5 cells were used for (1) immunohistochemical and immunoblot studies to detect PDGF receptor (PDGFR) subtypes and (2) immunoprecipitation, immunoblot, and in vitro lipid kinase assays to determine basal and PDGF-induced class I(A) PI 3-kinase/Akt survival signaling, in comparison with insulin or IGF-1 responses. Furthermore, RGC-5 cells were exposed to broad-spectrum (LY294002) or p110 isoform-selective (PI-103) PI 3-kinase inhibitors (versus Akt inhibitor) to assess the consequent effects on Akt phosphorylation, caspase-3/PARP cleavage, apoptotic phenotype, and cell viability, as a function of serum trophic factors. RESULTS: PDGFR-alpha and -beta immunoreactivity was observed in rat retinal Müller cells and in the RGC layer and blood vessels, respectively. In addition, PDGFR-alpha and -beta protein expression was observed in RGC-5 cells. Both retinas and RGC-5 cells exhibited a similar pattern of subunit-specific basal class I(A) PI 3-kinase activity, which was stimulated in a temporal and signal-specific manner by PDGF and insulin/IGF-1. Furthermore, RGC-5 cells showed PDGFR-alpha/beta tyrosine phosphorylation that induced the p85alpha regulatory subunit to activate p110alpha/beta-associated class I(A) PI 3-kinase, which in turn enhanced Akt phosphorylation. Exposure of serum-deprived RGC-5 cells to PI 3-kinase or Akt inhibitors increased susceptibility to apoptotic phenotype as revealed by caspase-3 and PARP cleavage. CONCLUSIONS: The present findings provide direct evidence of two distinct modes of retinal class I(A) PI 3-kinase activation that occurs in response to PDGF receptor and insulin/IGF-1 receptor stimulation. PDGF-induced PI 3-kinase/PIP3/Akt axis may provide new therapeutic approaches to ameliorate cell death in diabetic retinopathy and other retinal neurodegenerations.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/pharmacology , Retina/drug effects , Retinal Ganglion Cells/drug effects , Animals , Becaplermin , Caspase 3/metabolism , Cell Line , Cell Survival , Chromones/pharmacology , Enzyme Activation , Furans/pharmacology , Immunoblotting , Immunohistochemistry , Male , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-sis , Pyridines/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Retina/metabolism , Retinal Ganglion Cells/metabolism , Signal Transduction
4.
Diabetes ; 55(9): 2401-11, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16936187

ABSTRACT

Diabetic retinopathy remains a frightening prospect to patients and frustrates physicians. Destruction of damaged retina by photocoagulation remains the primary treatment nearly 50 years after its introduction. The diabetes pandemic requires new approaches to understand the pathophysiology and improve the detection, prevention, and treatment of retinopathy. This perspective considers how the unique anatomy and physiology of the retina may predispose it to the metabolic stresses of diabetes. The roles of neural retinal alterations and impaired retinal insulin action in the pathogenesis of early retinopathy and the mechanisms of vision loss are emphasized. Potential means to overcome limitations of current animal models and diagnostic testing are also presented with the goal of accelerating therapies to manage retinopathy in the face of ongoing diabetes.


Subject(s)
Diabetic Retinopathy , Diabetic Retinopathy/complications , Diabetic Retinopathy/etiology , Glucose/adverse effects , Humans , Microcirculation/drug effects , Receptor, Insulin/physiology , Retina/anatomy & histology , Retina/physiology , Retinitis/physiopathology , Vision Disorders/etiology
5.
Diabetes ; 55(4): 1148-56, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16567541

ABSTRACT

Diabetic retinopathy is characterized by early onset of neuronal cell death. We previously showed that insulin mediates a prosurvival pathway in retinal neurons and that normal retina expresses a highly active basal insulin receptor/Akt signaling pathway that is stable throughout feeding and fasting. Using the streptozotocin-induced diabetic rat model, we tested the hypothesis that diabetes diminishes basal retinal insulin receptor signaling concomitantly with increased diabetes-induced retinal apoptosis. The expression, phosphorylation status, and/or kinase activity of the insulin receptor and downstream signaling proteins were investigated in retinas of age-matched control, diabetic, and insulin-treated diabetic rats. Four weeks of diabetes reduced basal insulin receptor kinase, insulin receptor substrate (IRS)-1/2-associated phosphatidylinositol 3-kinase, and Akt kinase activity without altering insulin receptor or IRS-1/2 expression or tyrosine phosphorylation. After 12 weeks of diabetes, constitutive insulin receptor autophosphorylation and IRS-2 expression were reduced, without changes in p42/p44 mitogen-activated protein kinase or IRS-1. Sustained systemic insulin treatment of diabetic rats prevented loss of insulin receptor and Akt kinase activity, and acute intravitreal insulin administration restored insulin receptor kinase activity. Insulin treatment restored insulin receptor-beta autophosphorylation in rat retinas maintained ex vivo, demonstrating functional receptors and suggesting loss of ligand as a cause for reduced retinal insulin receptor/Akt pathway activity. These results demonstrate that diabetes progressively impairs the constitutive retinal insulin receptor signaling pathway through Akt and suggests that loss of this survival pathway may contribute to the initial stages of diabetic retinopathy.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetic Retinopathy/physiopathology , Insulin/therapeutic use , Receptor, Insulin/physiology , Retina/physiopathology , Animals , Diabetic Retinopathy/drug therapy , Male , Rats , Rats, Sprague-Dawley , Receptor, Insulin/drug effects , Retina/drug effects , Retina/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
6.
Cancer Res ; 64(19): 7002-10, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15466193

ABSTRACT

Malignant melanoma is the skin cancer with the most significant impact on man, carrying the highest risk of death from metastasis. Both incidence and mortality rates continue to rise each year, with no effective long-term treatment on the horizon. In part, this reflects lack of identification of critical genes involved and specific therapies targeted to correct these defects. We report that selective activation of the Akt3 protein promotes cell survival and tumor development in 43 to 60% of nonfamilial melanomas. The predominant Akt isoform active in melanomas was identified by showing that small interfering RNA (siRNA) against only Akt3, and not Akt1 or Akt2, lowered the amount of phosphorylated (active) Akt in melanoma cells. The amount of active Akt3 increased progressively during melanoma tumor progression with highest levels present in advanced-stage metastatic melanomas. Mechanisms of Akt3 deregulation occurred through a combination of overexpression of Akt3 accompanying copy number increases of the gene and decreased PTEN protein function occurring through loss or haploinsufficiency of the PTEN gene. Targeted reduction of Akt3 activity with siRNA or by expressing active PTEN protein stimulated apoptotic signaling, which reduced cell survival by increasing apoptosis rates thereby inhibiting melanoma tumor development. Identifying Akt3 as a selective target in melanoma cells provides new therapeutic opportunities for patients in the advanced stages of this disease.


Subject(s)
Melanoma/enzymology , Melanoma/pathology , Oncogene Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Animals , Apoptosis/physiology , Disease Progression , Enzyme Activation , Female , Humans , Melanoma/genetics , Mice , Mice, Nude , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/biosynthesis , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Isoforms , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt , RNA, Small Interfering/genetics , Skin Neoplasms/genetics , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
7.
Pharmacology ; 70(1): 46-56, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14646356

ABSTRACT

Previous studies have shown that muscarinic inhibition of cardiac contractility is mediated by either activation of nitric oxide (NO)/guanosine 3',5'-cyclic monophosphate (cGMP) pathway or stimulation of inhibitory G protein (G(i)). However, it still remains controversial as to whether NO/cGMP pathway or G(i) protein or both mediate(s) the negative inotropic effect of muscarinic agonists in adult ventricular myocytes. In the present study that involves the use of adult rat ventricular myocytes, the muscarinic agonist, carbachol, inhibited beta-adrenergic (isoproterenol) stimulation of contractility (cell shortening) by 82% and increased cGMP levels by 49% within 6 min. Pretreatment of myocytes with soluble guanylyl cyclase inhibitor (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, ODQ) or NO synthase inhibitor (N(G)-monomethyl-L-arginine, L-NMMA) for 30 min blocked carbachol-induced increases in cGMP levels. However, neither ODQ nor L-NMMA pretreatment had any effect on carbachol inhibition of isoproterenol-induced contractility. In addition, carbachol did not attenuate increases in myocyte contractility induced by forskolin (a direct activator of adenylyl cyclase) or 8-(4-chlorophenylthio)-adenosine 3',5'-cyclic monophosphate (a cell-permeable cAMP analog which activates cAMP-dependent protein kinase). Pretreatment of myocytes with G(i) protein inhibitor, pertussis toxin (PTX, 1 microg/ml), for 18-20 h abolished carbachol inhibition of isoproterenol-induced contractility. Furthermore, in ventricular myocytes isolated 3 days after in vivo treatment of rats with PTX (3 microg/100 g, i.p.), there was a complete loss of the negative inotropic effect of carbachol. These data indicate that pertussis toxin-sensitive G protein but not NO/cGMP pathway is required for muscarinic inhibition of beta-adrenoceptor-mediated increases in contractility in adult rat ventricular myocytes.


Subject(s)
Carbachol/pharmacology , GTP-Binding Proteins/drug effects , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Pertussis Toxin/pharmacokinetics , Adenylyl Cyclases , Animals , Carbachol/antagonists & inhibitors , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases , Cyclic GMP/biosynthesis , Electric Stimulation , GTP-Binding Proteins/physiology , Injections, Intraperitoneal , Isoproterenol/pharmacology , Male , Myocytes, Cardiac/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/physiology , Oxadiazoles/pharmacology , Pertussis Toxin/administration & dosage , Quinoxalines/pharmacology , Rats , Time Factors , omega-N-Methylarginine/pharmacology
8.
Am J Physiol Endocrinol Metab ; 285(4): E763-74, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12799319

ABSTRACT

Insulin receptor (IR) signaling cascades have been studied in many tissues, but retinal insulin action has received little attention. Retinal IR signaling and activity were investigated in vivo in rats that were freely fed, fasted, or injected with insulin by phosphotyrosine immunoblotting and by measuring kinase activity. A retina explant system was utilized to investigate the IR signaling cascade, and immunohistochemistry was used to determine which retinal cell layers respond to insulin. Basal IR activity in the retina was equivalent to that in brain and significantly greater than that of liver, and it remained constant between freely fed and fasted rats. Furthermore, IR signaling increased in the retina after portal vein administration of supraphysiological doses of insulin. Ex vivo retinas responded to 10 nM insulin with IR beta-subunit (IRbeta) and IR substrate-2 (IRS-2) tyrosine phosphorylation and AktSer473 phosphorylation. The retina expresses mRNA for all three Akt isoforms as determined by in situ hybridization, and insulin specifically increases Akt-1 kinase activity. Phospho-AktSer473 immunoreactivity increases in retinal nuclear cell layers with insulin treatment. These results demonstrate that the retinal IR signaling cascade to Akt-1 possesses constitutive activity, and that exogenous insulin further stimulates this prosurvival pathway. These findings may have implications in understanding normal and dysfunctional retinal physiology.


Subject(s)
Insulin/pharmacology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Receptor, Insulin/metabolism , Retina/cytology , Retina/metabolism , Animals , Enzyme Activation , Male , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Retina/drug effects , Signal Transduction/physiology
9.
J Biol Chem ; 277(5): 3286-92, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11723139

ABSTRACT

We recently demonstrated that ceramide-coated balloon catheters limit vascular smooth muscle cell (VSMC) growth after stretch injury in vivo. In that study, inhibition of VSMC growth was correlated with a decrease in phosphorylation of the cell survival kinase Akt (protein kinase B). Utilizing cultured A7r5 VSMCs, we have now examined the mechanism by which ceramide inhibits Akt phosphorylation/activation. Our initial studies showed that ceramide-induced inhibition of Akt phosphorylation was not mediated through diminution in phosphoinositide 3-kinase activity. As we have previously demonstrated that protein kinase Czeta (PKCzeta) is a target of ceramide, we proposed an alternative signaling mechanism by which ceramide induces inhibition of Akt through activation of PKCzeta. We demonstrate that C(6)-ceramide (but not the inactive analog dihydro-C(6)-ceramide) induced PKCzeta activity and also caused a selective increase in the association between Akt and PKCzeta, without affecting PKCepsilon, in A7r5 cells. In addition, the ability of ceramide to significantly decrease platelet-derived growth factor-induced Akt phosphorylation or cell proliferation was abrogated in A7r5 cells overexpressing a dominant-negative mutant of PKCzeta. Taken together, these data suggest that ceramide-mediated activation of PKCzeta leads to diminished Akt activation and consequent growth arrest in VSMCs. The therapeutic potential for ceramide to limit dysregulated VSMC growth has direct applicability to vascular diseases such as restenosis and atherosclerosis.


Subject(s)
Cell Division/physiology , Muscle, Smooth, Vascular/cytology , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Animals , Aorta, Thoracic , Becaplermin , Cell Division/drug effects , Cells, Cultured , Ceramides/pharmacology , Embryo, Mammalian , Humans , Insulin-Like Growth Factor I/pharmacology , Kinetics , Muscle, Smooth, Vascular/drug effects , Platelet-Derived Growth Factor/pharmacology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-sis , Rats , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...