Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Pharmacogenomics J ; 10(5): 385-95, 2010 Oct.
Article in English | MEDLINE | ID: mdl-19997081

ABSTRACT

Biomarkers that can be measured in preclinical models in a high-throughput, reproducible manner offer the potential to increase the speed and efficacy of drug development. Development of therapeutic agents for many conditions is hampered by the limited number of validated preclinical biomarkers available to gauge pharmacoefficacy and disease progression, but the validation process for preclinical biomarkers has received limited attention. This report defines a five-step preclinical biomarker validation process and applies the process to a case study of diabetic retinopathy. By showing that a gene expression panel is highly reproducible, coincides with disease manifestation, accurately classifies individual animals and identifies animals treated with a known therapeutic agent, a biomarker panel can be considered validated. This particular biomarker panel consisting of 14 genes (C1inh, C1s, Carhsp1, Chi3l1, Gat3, Gbp2, Hspb1, Icam1, Jak3, Kcne2, Lama5, Lgals3, Nppa, Timp1) can be used in diabetic retinopathy pharmacotherapeutic research, and the biomarker development process outlined here is applicable to drug development efforts for other diseases.


Subject(s)
Biomarkers, Pharmacological/analysis , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Animals , Databases, Genetic , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/genetics , Endpoint Determination , Gene Expression/drug effects , Gene Expression Profiling , Genetic Markers/genetics , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/therapeutic use , Insulin/administration & dosage , Insulin/therapeutic use , Male , Rats , Rats, Sprague-Dawley , Reproducibility of Results
2.
Neuroscience ; 121(4): 883-90, 2003.
Article in English | MEDLINE | ID: mdl-14580938

ABSTRACT

For peripheral iron to reach the brain, it must transverse the blood-brain barrier. In order for the brain to obtain iron, transferrin receptors are present in the vascular endothelial cell to facilitate movement of transferrin bound iron into the brain parenchyma. However, a number of significant voids exist in our knowledge about transport of iron into the brain. These gaps in our knowledge are significant not only because iron is an essential neurotrophic factor but also because the system for delivery of iron into the brain is being viewed as an opportunity to circumvent the blood-brain barrier for delivery of neurotoxins to tumors or trophic factors in neurodegenerative diseases. In this study, we have used fluorescein-transferrin-59Fe in a bovine retinal endothelial cell culture system to determine the mechanism of transferrin-iron transport and to test the hypothesis that the iron status of the endothelial cells would influence iron transport. Our results indicated that iron is transported across endothelial cells both bound to and not bound to transferrin. The ratio of non-transferrin-bound iron to transferrin-bound iron transported is dependent upon the iron status of the cells. Blocking acidification of endosomes led to a significant decrease in transport of non-transferrin-bound iron but not transferrin-bound iron. Blocking pinocytosis had no effect on either transferrin or iron transcytosis. These results indicate that there is both transferrin-mediated and non-transferrin-mediated transcytosis of iron and that the process is influenced by the iron status of the cells. These data have considerable implications for common neurodegenerative diseases that are associated with excess brain iron accumulation and the numerous neurological complications associated with brain iron deficiency.


Subject(s)
Blood-Brain Barrier/physiology , Endothelium, Vascular/metabolism , Iron/metabolism , Transferrin/metabolism , Animals , Blood-Brain Barrier/drug effects , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/physiopathology , Cattle , Cells, Cultured , Endocytosis/drug effects , Endocytosis/physiology , Endosomes/drug effects , Endosomes/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Hydrogen-Ion Concentration/drug effects , Iron Metabolism Disorders/metabolism , Iron Metabolism Disorders/physiopathology , Microcirculation/drug effects , Microcirculation/metabolism , Models, Biological , Pinocytosis/drug effects , Pinocytosis/physiology
3.
J Biol Chem ; 276(47): 43748-55, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11560942

ABSTRACT

In addition to microvascular abnormalities, neuronal apoptosis occurs early in diabetic retinopathy, but the mechanism is unknown. Insulin may act as a neurotrophic factor in the retina via the phosphoinositide 3-kinase/Akt pathway. Excessive glucose flux through the hexosamine biosynthetic pathway (HBP) is implicated in the development of insulin resistance in peripheral tissues and diabetic complications such as nephropathy. We tested whether increased glucose flux through the HBP perturbs insulin action and induces apoptosis in retinal neuronal cells. Exposure of R28 cells, a model of retinal neurons, to 20 mm glucose for 24 h attenuated the ability of 10 nm insulin to rescue them from serum deprivation-induced apoptosis and to phosphorylate Akt compared with 5 mm glucose. Glucosamine not only impaired the neuroprotective effect of insulin but also induced apoptosis in R28 cells in a dose-dependent fashion. UDP-N-acetylhexosamines (UDP-HexNAc), end products of the HBP, were increased approximately 2- and 15-fold after a 24-h incubation in 20 mm glucose and 1.5 mm glucosamine, respectively. Azaserine, a glutamine:fructose-6-phosphate amidotransferase inhibitor, reversed the effect of 20 mm glucose, but not that of 1.5 mm glucosamine, on attenuation of the ability of insulin to promote cell survival and phosphorylate Akt as well as accumulation of UDP-HexNAc. Glucosamine also impaired insulin receptor processing in a dose-dependent manner but did not decrease ATP content. By contrast, in L6 muscle cells, glucosamine impaired insulin receptor processing but did not induce apoptosis. These results suggest that the excessive glucose flux through the HBP may direct retinal neurons to undergo apoptosis in a bimodal fashion; i.e. via perturbation of the neuroprotective effect of insulin mediated by Akt and via induction of apoptosis possibly by altered glycosylation of proteins. The HBP may be involved in retinal neurodegeneration in diabetes.


Subject(s)
Apoptosis/drug effects , Hexosamines/pharmacology , Insulin Antagonists/pharmacology , Insulin/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Retina/drug effects , Adenosine Triphosphate/metabolism , Animals , Azaserine/pharmacology , Cell Line , Glucosamine/pharmacology , Glucose/pharmacology , Neurons/cytology , Neurons/metabolism , Phosphorylation , Protein Processing, Post-Translational/drug effects , Receptor, Insulin/metabolism , Retina/cytology , Retina/metabolism
4.
J Biol Chem ; 276(35): 32814-21, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11443130

ABSTRACT

The ability of insulin to protect neurons from apoptosis was examined in differentiated R28 cells, a neural cell line derived from the neonatal rat retina. Apoptosis was induced by serum deprivation, and the number of pyknotic cells was counted. p53 and Akt were examined by immunoblotting after serum deprivation and insulin treatment, and caspase-3 activation was examined by immunocytochemistry. Serum deprivation for 24 h caused approximately 20% of R28 cells to undergo apoptosis, detected by both pyknosis and activation of caspase-3. 10 nm insulin maximally reduced the amount of apoptosis with a similar potency as 1.3 nm (10 ng/ml) insulin-like growth factor 1, which acted as a positive control. Insulin induced serine phosphorylation of Akt, through the phosphatidylinositol (PI) 3-kinase pathway. Inhibition of PI 3-kinase with wortmannin or LY294002 blocked the ability of insulin to rescue the cells from apoptosis. SN50, a peptide inhibitor of NF-kappaB nuclear translocation, blocked the rescue effect of insulin, but neither insulin or serum deprivation induced phosphorylation of IkappaB. These results suggest that insulin is a survival factor for retinal neurons by activating the PI 3-kinase/Akt pathway and by reducing caspase-3 activation. The rescue effect of insulin does not appear to be mediated by NF-kappaB or p53. These data suggest that insulin provides trophic support for retinal neurons through a PI 3-kinase/Akt-dependent pathway.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Insulin/pharmacology , Neurons/cytology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Retina/cytology , Androstadienes/pharmacology , Animals , Caspase 3 , Cattle , Cell Line , Chromones/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Insulin-Like Growth Factor I/pharmacology , Morpholines/pharmacology , Neurons/drug effects , Neurons/physiology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt , Rats , Receptor, IGF Type 1/drug effects , Receptor, IGF Type 1/physiology , Receptor, Insulin/drug effects , Receptor, Insulin/physiology , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Transfection , Wortmannin
5.
Am J Physiol Heart Circ Physiol ; 281(1): H105-13, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11406474

ABSTRACT

Previous studies determined that shear stress imposed on bovine aortic endothelial cell (BAEC) monolayers increased the hydraulic conductivity (L(P)); however, the mechanism by which shear stress increases L(P) remains unknown. This study tested the hypothesis that shear stress regulates paracellular transport by altering the expression and phosphorylation state of the tight junction protein occludin. The effect of shear stress on occludin content was examined by Western blot analysis. Ten dyn/cm(2) significantly reduced occludin content in a time-dependent manner such that after a 3 h exposure to shear, occludin content decreased to 44% of control. Twenty dyn/cm(2) decreased occludin content to 50% of control and increased L(P) by 4.7-fold after 3 h. Occludin expression and L(P) depend on tyrosine kinase activity because erbstatin A (10 microM) attenuated both the shear-induced decrease in occludin content and increase in L(P). Shear stress increased occludin phosphorylation after 5 min, 15 min, and 3 h exposures. The shear-induced increase in occludin phosphorylation was attenuated with dibutyryl (DB) cAMP (1 mM), a reagent previously shown to reverse the shear-induced increase in L(P). We conclude that shear stress rapidly (< or = 5 min) increases occludin phosphorylation and significantly decreases the expression of occludin over 1-4 h. Alterations in the occludin phosphorylation state and occludin total content are potential mechanisms by which shear stress increases L(P).


Subject(s)
Aorta/metabolism , Endothelium, Vascular/metabolism , Membrane Proteins/metabolism , Animals , Aorta/cytology , Bucladesine/pharmacology , Cattle , Cells, Cultured , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Hydroquinones/pharmacology , Occludin , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Stress, Mechanical , Tissue Distribution , Zonula Occludens-1 Protein
6.
Invest Ophthalmol Vis Sci ; 41(13): 4256-61, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11095623

ABSTRACT

PURPOSE: Vascular endothelial growth factor (VEGF) increases microvascular permeability in vivo and has been hypothesized to play a role in plasma leakage in diabetic retinopathy. Few controlled studies have been conducted to determine the mechanism underlying the effect of VEGF on transport properties (e.g., hydraulic conductivity [Lp]). This study was conducted to determine the effect of VEGF on bovine retinal microvascular endothelial LP and the role of nitric oxide (NO) and the guanylate cyclase/guanosine 3', 5'-cyclic monophosphate/protein kinase G (GC/cGMP/PKG) pathway downstream of NO in mediating the VEGF response. METHODS: Bovine retinal microvascular endothelial cells (BRECs) were grown on porous polycarbonate filters, and water flux across BREC monolayers in response to a pressure differential was measured to determine endothelial LP RESULTS: VEGF (100 ng/ml) increased endothelial LP: within 30 minutes of addition and by 13.8-fold at the end of 3 hours of exposure. VEGF stimulated endothelial monolayers to release NO and incubation of the BRECs with the nitric oxide synthase inhibitor N(G)-monomethyl-L-arginine (L-NMMA; 100 microM) significantly attenuated the VEGF-induced LP increase. It was observed that incubation of the monolayers with the GC inhibitor LY-83583 (10 microM) did not alter the VEGF-mediated LP: response. Addition of the cGMP analogue 8-br-cGMP (1 mM) did not change the baseline LP over 4 hours. Also, the PKG inhibitor KT5823 (1 microM) did not inhibit the response of BREC LP to VEGF. CONCLUSIONS: These experiments indicate that VEGF elevates hydraulic conductivity in BRECs through a signaling mechanism that involves NO but not the GC/cGMP/PKG pathway.


Subject(s)
Body Water/metabolism , Carbazoles , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/metabolism , Indoles , Lymphokines/pharmacology , Nitric Oxide/physiology , Retinal Vessels/metabolism , Alkaloids/pharmacology , Aminoquinolines/pharmacology , Animals , Biological Transport/drug effects , Cattle , Cells, Cultured , Cyclic GMP/antagonists & inhibitors , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Nitrates/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type III , Nitrites/metabolism , Permeability/drug effects , Retinal Vessels/cytology , Retinal Vessels/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , omega-N-Methylarginine/pharmacology
7.
Invest Ophthalmol Vis Sci ; 41(11): 3561-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11006253

ABSTRACT

PURPOSE: To investigate how diabetes alters vascular endothelial cell tight junction protein and glial cell morphology at the blood-retinal barrier (BRB). METHODS: The distribution of the glial marker, glial fibrillary acidic protein (GFAP), and the endothelial cell tight junction protein occludin were explored by immunofluorescence histochemistry in flatmounted retinas of streptozotocin (STZ)-diabetic and age-matched control rats, and in BB/Wor diabetes-prone and age-matched diabetes-resistant rats. RESULTS: GFAP immunoreactivity was limited to astrocytes in control retinas. Two months of STZ-diabetes reduced GFAP immunoreactivity in astrocytes and increased GFAP immunoreactivity in small groups of Müller cells. After 4 months of STZ-induced diabetes, all Müller cells had intense GFAP immunoreactivity, whereas there was virtually none in the astrocytes. BB/Wor diabetic rats had similar changes in GFAP immunoreactivity. Occludin immunoreactivity in normal rats was greatest in the capillary bed of the outer plexiform layer and arterioles of the inner retina but much less intense in the postcapillary venules. Diabetes reduced occludin immunoreactivity in the capillaries and induced redistribution from continuous cell border to interrupted, punctate immunoreactivity in the arterioles. Forty-eight hours of insulin treatment reversed the pattern of GFAP and occludin immunoreactivity in the STZ-diabetic rats. CONCLUSIONS: Diabetes alters GFAP expression in retinal glial cells, accompanied by reduction and redistribution of occludin in endothelial cells. These changes are consistent with the concept that altered glial-endothelial cell interactions at the BRB contribute to diabetic retinopathy.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Eye Proteins/metabolism , Glial Fibrillary Acidic Protein/metabolism , Membrane Proteins/metabolism , Retina/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Blood-Retinal Barrier , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Male , Microscopy, Confocal , Neuroglia/metabolism , Neuroglia/pathology , Occludin , Rats , Rats, Inbred BB , Rats, Sprague-Dawley , Retina/pathology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Tight Junctions/metabolism , Tight Junctions/pathology
8.
Exp Eye Res ; 70(6): 723-30, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10843776

ABSTRACT

Retinas of diabetic individuals develop early functional changes measurable by electrophysiological and psychometric testing. Using a rat model of diabetes, we previously identified diabetes-induced alterations in metabolism of the neurotransmitter glutamate which may ultimately lead to accumulation of glutamate in the retina (Diabetes, 47: 815, 1998). We therefore investigated the function of enzymes that mediate the synthesis and breakdown of glutamate in retinas from rats made diabetic by injection of streptozotocin. De novo synthesis of nitrogen-containing amino acids including glutamate, glutamine and aspartate was assessed by measuring the rate of carbon fixation in freshly dissected retinas, and was unchanged by diabetes. In contrast, the oxidation of glutamate was significantly reduced in retinas from diabetic rats (62%, P < 0.05). Furthermore, diabetic retinas were less susceptible to inhibition of glutamate oxidation by the transaminase inhibitor aminoxyacetate (80%, N.S.), compared to the significant decrease seen in control rats (61%, P < 0.001). The activity and content of glutamine synthetase were also significantly reduced in retinas from rats diabetic for 2-6 months [range of 48% (P < 0.005) to 83% (P < 0.05) compared to control]. The activity of glutamine synthetase was normalized by acute injections of insulin, but not by reducing blood sugar levels with injections of phlorizin. These results indicate two enzymatic abnormalities in the glutamate metabolism pathway in the retina during diabetes: transamination to alpha-ketoglutarate and amination to glutamine. The reduced flux through these pathways may be associated with the accumulation of glutamate. These results are also consistent with the possibility that some of the glial changes in the retina during diabetes may be caused by hypoinsulinemia rather than hyperglycemia.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Glutamic Acid/metabolism , Glutamine/biosynthesis , Retina/metabolism , Animals , Blotting, Western/methods , Culture Techniques , Glutamic Acid/biosynthesis , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Time Factors
9.
Clin Exp Ophthalmol ; 28(1): 3-8, 2000 Feb.
Article in English | MEDLINE | ID: mdl-11345341

ABSTRACT

Normal vision depends on the normal function of retinal neurons, so vision loss in diabetes must ultimately be explained in terms of altered neuronal function. However to date relatively little attention has been paid to the impact of diabetes on the neural retina. Instead, the focus of most research has been primarily on retinal vascular changes, with the assumption that they cause altered neuronal function and consequently vision loss. An increasing body of evidence suggests that alterations in neuronal function and viability may contribute to the pathogenic mechanisms of diabetic retinopathy beginning shortly after the onset of diabetes. This view arises from neurophysiological, psychometric, histopathological and biochemical observations in humans and experimental animals. The collective evidence from past and recent studies supports the hypothesis that neurodegeneration, together with functional changes in the vasculature, is an important component of diabetic retinopathy. The authors invite other investigators to include the neural retina as a component of their studies so that the pathogenesis of diabetic retinopathy can be understood more clearly.


Subject(s)
Diabetic Retinopathy/diagnosis , Neuroglia/pathology , Retinal Degeneration/diagnosis , Apoptosis , Electroretinography , Humans
10.
Diabetes Technol Ther ; 2(4): 601-8, 2000.
Article in English | MEDLINE | ID: mdl-11469624

ABSTRACT

Diabetic retinopathy, a leading cause of vision impairment, is classically defined by its vascular lesions. This review examines how diabetes affects vascular cells, as well as neurons, macroglia, and microglia. The cellular and clinical elements of diabetic retinopathy have many features of chronic inflammation. Understanding the individual cell-specific and global inflammatory changes in the retina may lead to novel therapeutic approaches to prevent vision loss.


Subject(s)
Diabetes Mellitus/physiopathology , Diabetic Retinopathy/physiopathology , Diabetic Retinopathy/therapy , Diabetes Mellitus/pathology , Diabetic Retinopathy/prevention & control , Humans , Inflammation , Microglia/pathology , Neurons/pathology , Retina/cytology , Retina/pathology , Retina/physiopathology , Vision Disorders/prevention & control
11.
J Biol Chem ; 274(51): 36168-75, 1999 Dec 17.
Article in English | MEDLINE | ID: mdl-10593901

ABSTRACT

Leucine, glutamine, and tyrosine, three amino acids playing key modulatory roles in hepatic proteolysis, were evaluated for activation of signaling pathways involved in regulation of liver protein synthesis. Furthermore, because leucine signals to effectors that lie distal to the mammalian target of rapamycin, these downstream factors were selected for study as candidate mediators of amino acid signaling. Using the perfused rat liver as a model system, we observed a 25% stimulation of protein synthesis in response to balanced hyperaminoacidemia, whereas amino acid imbalance due to elevated concentrations of leucine, glutamine, and tyrosine resulted in a protein synthetic depression of roughly 50% compared with normoaminoacidemic controls. The reduction in protein synthesis accompanying amino acid imbalance became manifest at high physiologic concentrations and was dictated by the guanine nucleotide exchange activity of translation initiation factor eIF2B. Paradoxically, this phenomenon occurred concomitantly with assembly of the mRNA cap recognition complex, eIF4F as well as activation of the 70-kDa ribosomal S6 kinase, p70(S6k). Dual and reciprocal modulation of eIF4F and eIF2B was leucine-specific because isoleucine, a structural analog, was ineffective in these regards. Thus, we conclude that amino acid imbalance, heralded by leucine, initiates a liver-specific translational fail-safe mechanism that deters protein synthesis under unfavorable circumstances despite promotion of the eIF4F complex.


Subject(s)
Eukaryotic Initiation Factor-2B/metabolism , Glutamine , Leucine , Liver/metabolism , Peptide Initiation Factors/metabolism , Tyrosine , Animals , Eukaryotic Initiation Factor-4F , Male , Perfusion , Rats , Rats, Sprague-Dawley
12.
J Biol Chem ; 274(33): 23463-7, 1999 Aug 13.
Article in English | MEDLINE | ID: mdl-10438525

ABSTRACT

Vascular endothelial growth factor (VEGF) may have a physiologic role in regulating vessel permeability and contributes to the pathophysiology of diabetic retinopathy as well as tumor development. We set out to ascertain the mechanism by which VEGF regulates paracellular permeability in rats. Intra-ocular injection of VEGF caused a post-translational modification of occludin as determined by a gel shift from 60 to 62 kDa. This event began by 15 min post-injection and was maximal by 45 min. Alkaline phosphatase treatment revealed this modification was caused by a change in occludin phosphorylation. In addition, the quantity of extracted occludin increased 2-fold in the same time frame. The phosphorylation and increased extraction of occludin was recapitulated in retinal endothelial cells in culture after VEGF stimulation. The data presented herein are the first demonstration of a change in the phosphorylation of this transmembrane protein under conditions of increased endothelial permeability. In addition, intra-ocular injection of VEGF also caused tyrosine phosphorylation of ZO-1 as early as 15 min and increased phosphorylation 4-fold after 90 min. In conclusion, VEGF rapidly increases occludin phosphorylation as well as the tyrosine phosphorylation of ZO-1. Phosphorylation of occludin and ZO-1 likely contribute to regulated endothelial paracellular permeability.


Subject(s)
Capillary Permeability/physiology , Diabetic Retinopathy/physiopathology , Endothelial Growth Factors/physiology , Lymphokines/physiology , Membrane Proteins/metabolism , Neoplasms, Experimental/physiopathology , Phosphoproteins/metabolism , Animals , Cattle , Cells, Cultured , Diabetic Retinopathy/metabolism , Male , Neoplasms, Experimental/metabolism , Occludin , Phosphorylation , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Zonula Occludens-1 Protein
14.
Semin Ophthalmol ; 14(4): 240-8, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10758225

ABSTRACT

Diabetes leads to a wide array of complications in humans, including kidney failure, vascular disease, peripheral nerve degeneration, and vision loss. Diabetic retinopathy causes blindness in more working-age people in the United States than any other disease and contributes greatly to blindness in the young and old as well. The increasing rate of diabetes occurring in our society can only bring about a further decrease in the visual health of this country unless new modalities are discovered to prevent and cure diabetic retinopathy. Breakdown of the blood-retinal barrier and the resultant vascular permeability remains one of the first observable alterations in diabetic retinopathy and strongly correlates with vision loss. In this article, we examine the molecular components that form this blood-retinal barrier and explore how changes in the production of growth factors in the neural parenchyma cause an increase in vascular permeability and contribute to retinal degeneration.


Subject(s)
Blood-Retinal Barrier/physiology , Capillary Permeability/physiology , Diabetic Retinopathy/physiopathology , Retinal Vessels/physiopathology , Biological Transport , Diabetic Retinopathy/prevention & control , Growth Substances/metabolism , Humans
15.
Diabetes ; 47(12): 1953-9, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9836530

ABSTRACT

Blood-retinal barrier (BRB) breakdown is a hallmark of diabetic retinopathy, but the molecular changes that cause this pathology are unclear. Occludin is a transmembrane component of interendothelial tight junctions that may regulate permeability at the BRB. In this study, we examined the effects of vascular endothelial growth factor (VEGF) and diabetes on vascular occludin content and barrier function. Sprague-Dawley rats were made diabetic by intravenous streptozotocin injection, and age-matched animals served as controls. After 3 months, BRB permeability was quantified by intravenous injection of fluorescein isothiocyanate-bovine serum albumin (FITC-BSA), Mr 66 kDa, and 10-kDa rhodamine-dextran (R-D), followed by digital image analysis of retinal sections. Retinal fluorescence intensity for FITC-BSA increased 62% (P < or = 0.05), but R-D fluorescence did not change significantly. Occludin localization at interendothelial junctions was confirmed by immunofluorescence, and relative protein content was determined by immunoblotting of retinal homogenates. Retinal occludin content decreased approximately 35% (P < or = 0.03) in the diabetic versus the control animals, whereas the glucose transporter GLUT1 content was unchanged in rat retinas. Additionally, treatment of bovine retinal endothelial cells in culture with 0.12 nmol/l or 12 nmol/l VEGF for 6 h reduced occludin content 46 and 54%, respectively. These data show that diabetes selectively reduces retinal occludin protein expression and increases BRB permeability. Our findings suggest that the elevated VEGF in the vitreous of patients with diabetic retinopathy increases vascular permeability by downregulating occludin content. Decreased tight junction protein expression may be an important means by which diabetes causes increased vascular permeability and contributes to macular edema.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endothelium, Vascular/metabolism , Membrane Proteins/metabolism , Animals , Blood-Retinal Barrier/drug effects , Capillary Permeability , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Immunohistochemistry , Lymphokines/pharmacology , Male , Membrane Proteins/drug effects , Occludin , Rats , Rats, Sprague-Dawley , Retina/chemistry , Retina/drug effects , Retinal Vessels/cytology , Retinal Vessels/drug effects , Retinal Vessels/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
J Clin Invest ; 102(4): 783-91, 1998 Aug 15.
Article in English | MEDLINE | ID: mdl-9710447

ABSTRACT

This study determined whether retinal degeneration during diabetes includes retinal neural cell apoptosis. Image analysis of retinal sections from streptozotocin (STZ) diabetic rats after 7.5 months of STZ diabetes identified 22% and 14% reductions in the thickness of the inner plexiform and inner nuclear layers, respectively (P < 0. 001). The number of surviving ganglion cells was also reduced by 10% compared to controls (P < 0.001). In situ end labeling of DNA terminal dUTP nick end labeling (TUNEL) identified a 10-fold increase in the frequency of retinal apoptosis in whole-mounted rat retinas after 1, 3, 6, and 12 months of diabetes (P < 0.001, P < 0. 001, P < 0.01, and P < 0.01, respectively). Most TUNEL-positive cells were not associated with blood vessels and did not colocalize with the endothelial cell-specific antigen, von Willebrand factor. Insulin implants significantly reduced the number of TUNEL-positive cells (P < 0.05). The number of TUNEL-positive cells was also increased in retinas from humans with diabetes. These data indicate that retinal neural cell death occurs early in diabetes. This is the first quantitative report of an increase in neural cell apoptosis in the retina during diabetes, and indicates that neurodegeneration is an important component of diabetic retinopathy.


Subject(s)
Apoptosis , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Diabetic Retinopathy/pathology , Retinal Degeneration/pathology , Adult , Animals , Blood Glucose/analysis , Body Weight , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/complications , Diabetic Retinopathy/complications , Female , Glycated Hemoglobin/analysis , Humans , Insulin/therapeutic use , Male , Middle Aged , Neuroglia/pathology , Neurons/pathology , Rats , Rats, Sprague-Dawley , Retinal Degeneration/complications , Retinal Ganglion Cells/pathology , Streptozocin
17.
J Biol Chem ; 272(38): 23696-702, 1997 Sep 19.
Article in English | MEDLINE | ID: mdl-9295312

ABSTRACT

Following phosphorylation by the insulin receptor kinase, the insulin receptor substrates (IRS)-1 and IRS-2 bind to and activate several Src homology 2 (SH2) domain proteins. To identify novel proteins that interact with IRS proteins in muscle, a human skeletal muscle cDNA expression library was created in the lambdaEXlox system and probed with baculovirus-produced and tyrosine-phosphorylated human IRS-1. One clone of the 10 clones which was positive through three rounds of screening represented the C terminus of the human homologue of the adult fast twitch skeletal muscle Ca2+-ATPase (SERCA1) including the cytoplasmic tail and part of transmembrane region 10. Western blot analysis of extracts of rat muscle demonstrated co-immunoprecipitation of both IRS-1 and IRS-2 with the skeletal muscle Ca2+-ATPase (SERCA1) and the cardiac muscle isoform (SERCA2). In both cases, injection of insulin stimulated a 2- to 6-fold increase in association of which was maximal within 5 min. In primary cultures of aortic smooth muscle cells and C2C12 cells, the insulin-stimulated interaction between IRS proteins and SERCA1 and -2 was dose-dependent with a maximum induction at 100 nM insulin. This interaction was confirmed in a "pull down" experiment using a glutathione S-transferase fusion protein containing the C terminus of the human SERCA isoform and phosphorylated IRS-1 in vitro and could be blocked by a FLVRES-like domain peptide present in the human SERCA sequence. Affinity chromatography of phosphopeptide libraries using the glutathione S-transferase fusion protein of the C terminus of SERCA1 indicated a consensus sequence for binding of XpYGSS; this is identical to potential tyrosine phosphorylation sites at position 431 of human IRS-1 and at position 500 of human IRS-2. In streptozotocin diabetic rats the interaction between IRS proteins and SERCA1 in skeletal muscle and SERCA2 in cardiac muscle was significantly reduced. Taken together, these results indicate that the IRS proteins bind to the Ca2+-ATPase of the sarcoplasmic reticulum in an insulin-regulated fashion, thus creating a potential link between the tyrosine phosphorylation cascade and effects of insulin on calcium.


Subject(s)
Calcium-Transporting ATPases/metabolism , Isoenzymes/metabolism , Muscle, Skeletal/metabolism , Myocardium/metabolism , Phosphoproteins/metabolism , Tyrosine/metabolism , Animals , Binding, Competitive , Calcium-Transporting ATPases/chemistry , Cell Line , Diabetes Mellitus, Experimental/metabolism , Humans , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Ion Transport , Isoenzymes/chemistry , Male , Muscle, Skeletal/enzymology , Myocardium/enzymology , Peptides/metabolism , Phosphorylation , Precipitin Tests , Rats , Rats, Sprague-Dawley
18.
Mol Cell Biol ; 16(5): 2195-203, 1996 May.
Article in English | MEDLINE | ID: mdl-8628286

ABSTRACT

We have identified two novel alternatively spliced forms of the p85alpha regulatory subunit of phosphatidylinositol (PI) 3-kinase by expression screening of a human skeletal muscle library with phosphorylated baculovirus- produced human insulin receptor substrate 1. One form is identical to p85alpha throughout the region which encodes both Src homology 2 (SH2) domains and the inter-SH2 domain/p110 binding region but diverges in sequence from p85alpha on the 5' side of nucleotide 953, where the entire break point cluster gene and SH3 regions are replaced by a unique 34-amino-acid N terminus. This form has an estimated molecular mass of approximately 53 kDa and has been termed p85/AS53. The second form is identical to p85 and p85/AS53 except for a 24-nucleotide insert between the SH2 domains that results in a replacement of aspartic acid 605 with nine amino acids, adding two potential serine phosphorylation sites in the vicinity of the known serine autophosphorylation site (Ser-608). Northern (RNA) analyses reveal a wide tissue distribution of p85alpha, whereas p85/AS53 is dominant in skeletal muscle and brain, and the insert isoforms are restricted to cardiac muscle and skeletal muscle. Western blot (immunoblot) analyses using an anti-p85 polyclonal antibody and a specific anti-p85/AS53 antibody confirmed the tissue distribution of p85/AS53 protein and indicate a approximately 7-fold higher expression of p85/AS53 protein than of p85 in skeletal muscle. Both p85 and p85/AS53 bind to p110 in coprecipitation experiments, but p85alpha itself appears to have preferential binding to insulin receptor substrate 1 following insulin stimulation. These data indicate that the gene for the p85alpha regulatory subunit of PI 3-kinase can undergo tissue-specific alternative splicing. Two novel splice variants of the regulatory subunit of PI 3-kinase are present in skeletal muscle, cardiac muscle, and brain; these variants may have important functional differences in activity and may play a role in tissue-specific signals such as insulin-stimulated glucose transport or control of neurotransmitter secretion or action.


Subject(s)
Alternative Splicing , Brain/metabolism , Muscle, Skeletal/enzymology , Phosphoproteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Cloning, Molecular , Gene Expression , Genetic Variation , Humans , Insulin Receptor Substrate Proteins , Isoenzymes/biosynthesis , Isoenzymes/chemistry , Isoenzymes/metabolism , Macromolecular Substances , Molecular Sequence Data , Multigene Family , Phosphatidylinositol 3-Kinases , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Spodoptera , Transfection , src Homology Domains
19.
J Biol Chem ; 271(2): 768-73, 1996 Jan 12.
Article in English | MEDLINE | ID: mdl-8557685

ABSTRACT

Rad, a prototypic member of a subfamily of Ras-related GTPases, is overexpressed in skeletal muscle of type II diabetic humans. By expression screening of mouse embryo and human skeletal muscle cDNA libraries, we found that Rad interacted with skeletal muscle beta-tropomyosin. In the mouse skeletal muscle cell line C2C12, this interaction was significantly increased by the calcium ionophore A23187. A23187 also caused a time- and concentration-dependent decrease in total cellular Rad with increased interaction between tropomyosin and Rad in the detergent-soluble fraction and the appearance of Rad in the cytoskeleton. In C2C12 cells stably overexpressing a putative dominant negative mutant of Rad (S105N), there was an increase in the amount of tropomyosin in Rad immunoprecipitates. In cells overexpressing wild type Rad, much of Rad was associated with the cytoskeleton and was no longer responsive to A23187. In far-Western blotting and guanine nucleotide saturation studies, GDP-Rad bound to tropomyosin far better than GTP-Rad. We conclude that Rad interacts with skeletal muscle beta-tropomyosin and the cytoskeleton in a guanine nucleotide-dependent manner. These data suggest that Rad may be involved in skeletal muscle motor function and cytoskeletal organization.


Subject(s)
GTP-Binding Proteins/metabolism , Muscle, Skeletal/metabolism , Tropomyosin/metabolism , ras Proteins , Animals , Calcimycin/pharmacology , Cell Line , Cytoskeleton/metabolism , Humans , Ionophores/pharmacology , Mice
20.
J Clin Invest ; 95(3): 1383-8, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7533791

ABSTRACT

Screening subtraction libraries from normal and type II diabetic human skeletal muscle, we identified four different mitochondrially encoded genes which were increased in expression in diabetes. The genes were cytochrome oxidase I, cytochrome oxidase III, NADH dehydrogenase IV, and 12s rRNA, all of which are located on the heavy strand of the mitochondrial genome. There was a 1.5- to 2.2-fold increase in the expression of these mRNA molecules relative to total RNA in both type I and type II diabetes as assessed by Northern blot analyses. Since there was approximately 50% decrease in mitochondrial DNA copy number as estimated by Southern blot analyses, mitochondrial gene expression increased approximately 2.5-fold when expressed relative to mitochondrial DNA copy number. For cytochrome oxidase I similar changes in mitochondrial gene expression were observed in muscle of nonobese diabetic and ob/ob mice, models of type I and type II diabetes, respectively. By contrast there was no change or a slight decrease in expression of cytochrome oxidase 7a, a nuclear-encoded subunit of cytochrome oxidase, and the expression of mitochondrial transcription factor 1 in human skeletal muscle did not change with type I or type II diabetes. The increased mitochondrial gene expression may contribute to the increase in mitochondrial respiration observed in uncontrolled diabetes.


Subject(s)
DNA, Mitochondrial/genetics , Diabetes Mellitus, Type 2/genetics , Gene Expression Regulation , Mitochondria/genetics , Mitochondrial Proteins , Muscle, Skeletal/physiology , Nuclear Proteins , Animals , Cell Nucleus/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 1/genetics , Electron Transport Complex IV/biosynthesis , Electron Transport Complex IV/genetics , Gene Dosage , Gene Library , High Mobility Group Proteins , Humans , Isoenzymes/genetics , Mice , Mice, Inbred NOD , Mice, Obese , NADH Dehydrogenase/biosynthesis , NADH Dehydrogenase/genetics , RNA/biosynthesis , RNA/genetics , RNA, Mitochondrial , RNA, Ribosomal/biosynthesis , RNA, Ribosomal/genetics , Rats , Streptozocin/pharmacology , Tissue Distribution , Transcription Factors/biosynthesis , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...