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










Database
Language
Publication year range
1.
Front Aging Neurosci ; 9: 43, 2017.
Article in English | MEDLINE | ID: mdl-28298893

ABSTRACT

Age-related macular degeneration (ARMD) is the leading cause of vision loss in developed countries. Hallmarks of the disease are well known; indeed, this pathology is characterized by lipofuscin accumulation, is principally composed of lipid-containing residues of lysosomal digestion. The N-retinyl-N-retinylidene ethanolamine (A2E) retinoid which is thought to be a cytotoxic component for RPE is the best-characterized component of lipofuscin so far. Even if no direct correlation between A2E spatial distribution and lipofuscin fluorescence has been established in aged human RPE, modified forms or metabolites of A2E could be involved in ARMD pathology. Mitogen-activated protein kinase (MAPK) pathways have been involved in many pathologies, but not in ARMD. Therefore, we wanted to analyze the effects of A2E on MAPKs in polarized ARPE19 and isolated mouse RPE cells. We showed that long-term exposure of polarized ARPE19 cells to low A2E dose induces a strong decrease of the extracellular signal-regulated kinases' (ERK1/2) activity. In addition, we showed that A2E, via ERK1/2 decrease, induces a significant decrease of the retinal pigment epithelium-specific protein 65 kDa (RPE65) expression in ARPE19 cells and isolated mouse RPE. In the meantime, we showed that the decrease of ERK1/2 activity mediates an increase of basic fibroblast growth factor (bFGF) mRNA expression and secretion that induces an increase in phagocytosis via a paracrine effect. We suggest that the accumulation of deposits coming from outer segments (OS) could be explained by both an increase of bFGF-induced phagocytosis and by the decrease of clearance by A2E. The bFGF angiogenic protein may therefore be an attractive target to treat ARMD.

2.
PLoS One ; 11(2): e0150266, 2016.
Article in English | MEDLINE | ID: mdl-26918849

ABSTRACT

Glucose is the most important metabolic substrate of the retina and maintenance of normoglycemia is an essential challenge for diabetic patients. Chronic, exaggerated, glycemic excursions could lead to cardiovascular diseases, nephropathy, neuropathy and retinopathy. We recently showed that hypoglycemia induced retinal cell death in mouse via caspase 3 activation and glutathione (GSH) decrease. Ex vivo experiments in 661W photoreceptor cells confirmed the low-glucose induction of death via superoxide production and activation of caspase 3, which was concomitant with a decrease of GSH content. We evaluate herein retinal gene expression 4 h and 48 h after insulin-induced hypoglycemia. Microarray analysis demonstrated clusters of genes whose expression was modified by hypoglycemia and we discuss the potential implication of those genes in retinal cell death. In addition, we identify by gene set enrichment analysis, three important pathways, including lysosomal function, GSH metabolism and apoptotic pathways. Then we tested the effect of recurrent hypoglycemia (three successive 4h periods of hypoglycemia spaced by 48 h recovery) on retinal cell death. Interestingly, exposure to multiple hypoglycemic events prevented GSH decrease and retinal cell death, or adapted the retina to external stress by restoring GSH level comparable to control situation. We hypothesize that scavenger GSH is a key compound in this apoptotic process, and maintaining "normal" GSH level, as well as a strict glycemic control, represents a therapeutic challenge in order to avoid side effects of diabetes, especially diabetic retinopathy.


Subject(s)
Gene Expression Regulation/drug effects , Hypoglycemia/chemically induced , Insulin/toxicity , Multigene Family/genetics , Retina/metabolism , Acute Disease , Adaptation, Physiological , Animals , Apoptosis/genetics , DNA Fragmentation , Diabetic Retinopathy/prevention & control , Eye Proteins/biosynthesis , Eye Proteins/genetics , Female , Free Radical Scavengers , Gene Expression Regulation/physiology , Glucose Clamp Technique , Glutathione/metabolism , Hypoglycemia/genetics , Hypoglycemia/metabolism , Insulin/pharmacology , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Microarray Analysis , Oxidation-Reduction , Random Allocation , Retina/pathology , Time Factors
3.
PLoS One ; 8(9): e74162, 2013.
Article in English | MEDLINE | ID: mdl-24066113

ABSTRACT

Glucose is an important metabolic substrate of the retina and diabetic patients have to maintain a strict normoglycemia to avoid diabetes secondary effects, including cardiovascular disease, nephropathy, neuropathy and retinopathy. Others and we recently demonstrated the potential role of hypoglycemia in diabetic retinopathy. We showed acute hypoglycemia to induce retinal cell death both in vivo during an hyperinsulinemic/hypoglycemic clamp and in vitro in 661W photoreceptor cells cultured at low glucose concentration. In the present study, we showed low glucose to induce a decrease of BCL2 and BCL-XL anti-apoptotic proteins expression, leading to an increase of free pro-apoptotic BAX. In parallel, we showed that, in retinal cells, low glucose-induced apoptosis is involved in the process of autophagosomes formation through the AMPK/RAPTOR/mTOR pathway. Moreover, the decrease of LAMP2a expression led to a defect in the autophagosome/lysosome fusion process. Specific inhibition of autophagy, either by 3-methyladenine or by down-regulation of ATG5 or ATG7 proteins expression, increased caspase 3 activation and 661W cell death. We show that low glucose modifies the delicate equilibrium between apoptosis and autophagy. Cells struggled against low nutrient condition-induced apoptosis by starting an autophagic process, which led to cell death when inhibited. We conclude that autophagy defect is associated with low glucose-induced 661W cells death that could play a role in diabetic retinopathy. These results could modify the way of addressing negative effects of hypoglycemia. Short-term modulation of autophagy could be envisioned to treat diabetic patients in order to avoid secondary complications of the disease.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Glucose/pharmacology , Photoreceptor Cells/cytology , Photoreceptor Cells/drug effects , Animals , Blotting, Western , Cells, Cultured , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , Mice , Photoreceptor Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction
4.
PLoS One ; 6(6): e21586, 2011.
Article in English | MEDLINE | ID: mdl-21738719

ABSTRACT

BACKGROUND: Glucose is the most important metabolic substrate of the retina and maintenance of normoglycemia is an essential challenge for diabetic patients. Glycemic excursions could lead to cardiovascular disease, nephropathy, neuropathy and retinopathy. A vast body of literature exists on hyperglycemia namely in the field of diabetic retinopathy, but very little is known about the deleterious effect of hypoglycemia. Therefore, we decided to study the role of acute hypoglycemia in mouse retina. METHODOLOGY/PRINCIPAL FINDINGS: To test effects of hypoglycemia, we performed a 5-hour hyperinsulinemic/hypoglycemic clamp; to exclude an effect of insulin, we made a hyperinsulinemic/euglycemic clamp as control. We then isolated retinas from each group at different time-points after the clamp to analyze cells apoptosis and genes regulation. In parallel, we used 661W photoreceptor cells to confirm in vivo results. We showed herein that hypoglycemia induced retinal cell death in mouse via caspase 3 activation. We then tested the mRNA expression of glutathione transferase omega 1 (Gsto1) and glutathione peroxidase 3 (Gpx3), two genes involved in glutathione (GSH) homeostasis. The expression of both genes was up-regulated by low glucose, leading to a decrease of reduced glutathione (GSH). In vitro experiments confirmed the low-glucose induction of 661W cell death via superoxide production and activation of caspase 3, which was concomitant with a decrease of GSH content. Moreover, decrease of GSH content by inhibition with buthionine sulphoximine (BSO) at high glucose induced apoptosis, while complementation with extracellular glutathione ethyl ester (GSHee) at low glucose restored GSH level and reduced apoptosis. CONCLUSIONS/SIGNIFICANCE: We showed, for the first time, that acute insulin-induced hypoglycemia leads to caspase 3-dependant retinal cell death with a predominant role of GSH content.


Subject(s)
Cell Death/physiology , Hyperglycemia/physiopathology , Retina/cytology , Retina/pathology , Adenosine Triphosphate/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Carrier Proteins/genetics , Caspase 3/genetics , Caspase 3/metabolism , Cells, Cultured , Female , Flow Cytometry , Glutathione/metabolism , Glutathione Peroxidase/genetics , Glutathione Transferase/genetics , In Vitro Techniques , Mice , Mice, Inbred C57BL , Retina/metabolism , Reverse Transcriptase Polymerase Chain Reaction
5.
Diabetes ; 55(4): 988-95, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16567520

ABSTRACT

A role for glucose in the control of feeding has been proposed, but its precise physiological importance is unknown. Here, we evaluated feeding behavior in glut2-null mice, which express a transgenic glucose transporter in their beta-cells to rescue insulin secretion (ripglut1;glut2-/- mice). We showed that in the absence of GLUT2, daily food intake was increased and feeding initiation and termination following a fasting period were abnormal. This was accompanied by suppressed regulation of hypothalamic orexigenic and anorexigenic neuropeptides expression during the fast-to-refed transition. In these conditions, however, there was normal regulation of the circulating levels of insulin, leptin, or glucose but a loss of regulation of plasma ghrelin concentrations. To evaluate whether the abnormal feeding behavior was due to suppressed glucose sensing, we evaluated feeding in response to intraperitoneal or intracerebroventricular glucose or 2-deoxy-D-glucose injections. We showed that in GLUT2-null mice, feeding was no longer inhibited by glucose or activated by 2-deoxy-D-glucose injections and the regulation of hypothalamic neuropeptide expression by intracerebroventricular glucose administration was lost. Together, these data demonstrate that absence of GLUT2 suppressed the function of central glucose sensors, which control feeding probably by regulating the hypothalamic melanocortin pathway. Furthermore, inactivation of these glucose sensors causes overeating.


Subject(s)
Feeding Behavior/physiology , Glucose Transporter Type 2/physiology , Animals , Base Sequence , DNA Primers , Ghrelin , Glucose Transporter Type 2/deficiency , Glucose Transporter Type 2/genetics , Hypothalamus/physiology , Insulin/blood , Leptin/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/genetics , Peptide Hormones/blood , Pro-Opiomelanocortin/genetics , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , alpha-MSH/physiology
6.
J Clin Invest ; 115(12): 3545-53, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16322792

ABSTRACT

Ripglut1;glut2-/- mice have no endogenous glucose transporter type 2 (glut2) gene expression but rescue glucose-regulated insulin secretion. Control of glucagon plasma levels is, however, abnormal, with fed hyperglucagonemia and insensitivity to physiological hypo- or hyperglycemia, indicating that GLUT2-dependent sensors control glucagon secretion. Here, we evaluated whether these sensors were located centrally and whether GLUT2 was expressed in glial cells or in neurons. We showed that ripglut1;glut2-/- mice failed to increase plasma glucagon levels following glucoprivation induced either by i.p. or intracerebroventricular 2-deoxy-D-glucose injections. This was accompanied by failure of 2-deoxy-D-glucose injections to activate c-Fos-like immunoreactivity in the nucleus of the tractus solitarius and the dorsal motor nucleus of the vagus. When glut2 was expressed by transgenesis in glial cells but not in neurons of ripglut1;glut2-/- mice, stimulated glucagon secretion was restored as was c-Fos-like immunoreactive labeling in the brainstem. When ripglut1;glut2-/- mice were backcrossed into the C57BL/6 genetic background, fed plasma glucagon levels were also elevated due to abnormal autonomic input to the alpha cells; glucagon secretion was, however, stimulated by hypoglycemic stimuli to levels similar to those in control mice. These studies identify the existence of central glucose sensors requiring glut2 expression in glial cells and therefore functional coupling between glial cells and neurons. These sensors may be activated at different glycemic levels depending on the genetic background.


Subject(s)
Astrocytes/metabolism , Gene Expression Regulation , Glucagon/metabolism , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/physiology , Glucose/metabolism , Animals , Blood Glucose/metabolism , Blotting, Northern , Blotting, Southern , Blotting, Western , DNA, Complementary/metabolism , Deoxyglucose/chemistry , Glucagon/blood , Glucagon/chemistry , Glucagon-Secreting Cells/metabolism , Glucose Transporter Type 2/metabolism , Hypoglycemia/pathology , Immunohistochemistry , Insulin/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Models, Genetic , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Time Factors , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...