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.
Exp Eye Res ; 202: 108342, 2021 01.
Article in English | MEDLINE | ID: mdl-33144094

ABSTRACT

B-N-methylamino-L-alanine (BMAA), a cyanotoxin produced by most cyanobacteria, has been proposed to cause long term damages leading to neurodegenerative diseases, including Amyotrophic Lateral Sclerosis/Parkinsonism Dementia complex (ALS/PDC) and retinal pathologies. Previous work has shown diverse mechanisms leading to BMAA-induced degeneration; however, the underlying mechanisms of toxicity affecting retina cells are not fully elucidated. We here show that BMAA treatment of rat retina neurons in vitro induced nuclear fragmentation and cell death in both photoreceptors (PHRs) and amacrine neurons, provoking mitochondrial membrane depolarization. Pretreatment with the N-Methyl-D-aspartate (NMDA) receptor antagonist MK-801 prevented BMAA-induced death of amacrine neurons, but not that of PHRs, implying activation of NMDA receptors participated only in amacrine cell death. Noteworthy, BMAA stimulated a selective axonal outgrowth in amacrine neurons, simultaneously promoting growth cone destabilization. BMAA partially decreased the viability of Müller glial cells (MGC), the main glial cell type in the retina, induced marked alterations in their actin cytoskeleton and impaired their capacity to protect retinal neurons. BMAA also induced cell death and promoted axonal outgrowth in differentiated rat pheochromocytoma (PC12) cells, implying these effects were not limited to amacrine neurons. These results suggest that BMAA is toxic for retina neurons and MGC and point to the involvement of NMDA receptors in amacrine cell death, providing new insight into the mechanisms involved in BMAA neurotoxic effects in the retina.


Subject(s)
Amino Acids, Diamino/toxicity , Ependymoglial Cells/drug effects , Excitatory Amino Acid Agonists/toxicity , Retinal Diseases/chemically induced , Retinal Neurons/drug effects , Animals , Animals, Newborn , Cell Survival/drug effects , Cyanobacteria Toxins , DNA Fragmentation/drug effects , Dizocilpine Maleate/pharmacology , Ependymoglial Cells/pathology , Excitatory Amino Acid Antagonists/pharmacology , Membrane Potential, Mitochondrial/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Retinal Diseases/metabolism , Retinal Diseases/prevention & control , Retinal Neurons/pathology
2.
Exp Eye Res ; 202: 108359, 2021 01.
Article in English | MEDLINE | ID: mdl-33197453

ABSTRACT

Müller glial cells, the major glial cell type in the retina, are activated by most retina injuries, leading to an increased proliferation and migration that contributes to visual dysfunction. The molecular cues involved in these processes are still ill defined. We demonstrated that sphingosine-1-phosphate (S1P), a bioactive sphingolipid, promotes glial migration. We now investigated whether ceramide-1-phosphate (C1P), also a bioactive sphingolipid, was involved in Müller glial cell migration. We evaluated cell migration in primary Müller glial cultures, prepared from newborn rat retinas, by the scratch wound assay. Addition of either 10 µM C8-ceramide-1-phosphate (C8-C1P) or 5 µM C16-C1P (a long chain, natural C1P) stimulated glial migration. Inhibiting PI3K almost completely blocked C8-C1P-elicited migration whereas inhibition of ERK1-2/MAPK pathway diminished it and p38MAPK inhibition did not affect it. Pre-treatment with a cytoplasmic phospholipase A2 (cPLA2) inhibitor markedly reduced C8-C1P-induced migration. Inhibiting ceramide kinase (CerK), the enzyme catalyzing C1P synthesis, partially decreased glial migration. Combined addition of S1P and C8-C1P promoted glial migration to the same extent as when they were added separately, suggesting they converge on their downstream signaling to stimulate Müller glia migration. These results suggest that C1P addition stimulated migration of glial Müller cells, promoting the activation of cPLA2, and the PI3K and ERK/MAPK pathways. They also suggest that CerK-dependent C1P synthesis was one of the factors contributing to glial migration, thus uncovering a novel role for C1P in controlling glial motility.


Subject(s)
Ceramides/pharmacology , Ependymoglial Cells/cytology , Retinal Ganglion Cells/cytology , Animals , Animals, Newborn , Cell Movement/drug effects , Ependymoglial Cells/drug effects , Models, Animal , Rats , Rats, Wistar , Retinal Ganglion Cells/drug effects , Signal Transduction
3.
J Lipid Res ; 51(6): 1247-62, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20100817

ABSTRACT

Many sphingolipids have key functions in the regulation of crucial cellular processes. Ceramide (Cer) and sphingosine (Sph) induce growth arrest and cell death in multiple situations of cellular stress. On the contrary, sphingosine-1-phosphate (S1P), the product of Sph phosphorylation, promotes proliferation, differentiation, and survival in different cell systems. This review summarizes the roles of these simple sphingolipids in different tissues and then analyzes their possible functions in the retina. Alterations in proliferation, neovascularization, differentiation, and cell death are critical in major retina diseases and collective evidence points to a role for sphingolipids in these processes. Cer induces inflammation and apoptosis in endothelial and retinal pigmented epithelium cells, leading to several retinopathies. S1P can prevent this death but also promotes cell proliferation that might lead to neovascularization and fibrosis. Recent data support Cer and Sph as crucial mediators in the induction of photoreceptor apoptosis in diverse models of oxidative damage and neurodegeneration, and suggest that regulating their metabolism can prevent this death. New evidence proposes a central role for S1P controlling photoreceptor survival and differentiation. Finally, this review discusses the ability of trophic factors to regulate sphingolipid metabolism and transactivate S1P signaling pathways to control survival and development in retina photoreceptors.


Subject(s)
Retina/cytology , Retina/growth & development , Sphingolipids/metabolism , Animals , Apoptosis , Cell Survival , Humans , Retina/metabolism , Signal Transduction , Sphingolipids/chemistry
4.
J Neurosci Res ; 87(4): 964-77, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18855938

ABSTRACT

Oxidative damage is involved in triggering neuronal death in several retinal neurodegenerative diseases. The recent finding of stem cells in the retina suggests that both preventing neuronal death and replacing lost neurons might be useful strategies for treating these diseases. We have previously shown that oxidative stress induces apoptosis in cultured retinal neurons. We now investigated the response of Müller cells, proposed as retina stem cells, to this damage. Treatment of glial cell cultures prepared from rat retinas with the oxidant paraquat (PQ) did not induce glial cell apoptosis. Instead, PQ promoted their rapid dedifferentiation and proliferation. PQ decreased expression of a marker of differentiated glial cells, simultaneously increasing the expression of smooth muscle actin, shown to increase with glial dedifferentiation, the levels of cell-cycle markers, and the number of glial cells in the cultures. In addition, glial cells protected neurons in coculture from apoptosis induced by PQ and H(2)O(2). In pure neuronal cultures, PQ induced apoptosis of photoreceptors and amacrine neurons, simultaneously decreasing the percentage of neurons preserving mitochondrial membrane potential; coculturing neurons with glial cells completely prevented PQ-induced apoptosis and preserved mitochondrial potential in both neuronal types. These results demonstrate that oxidative damage activated different responses in Müller glial cells; they rapidly dedifferentiated and enhanced their proliferation, concurrently preventing neuronal apoptosis. Glial cells might not only preserve neuronal survival but also activate their cell cycle in order to provide a pool of new progenitor cells that might eventually be manipulated to preserve retinal functionality.


Subject(s)
Cell Dedifferentiation , Cell Proliferation , Neuroglia/physiology , Oxidative Stress/physiology , Retina/cytology , Retinal Neurons/cytology , Actins/metabolism , Animals , Apoptosis/drug effects , Cell Dedifferentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Hydrogen Peroxide/pharmacology , Membrane Potential, Mitochondrial , Neuroglia/cytology , Neuroglia/drug effects , Oxidants/pharmacology , Paraquat/pharmacology , Rats , Rats, Wistar , Retina/drug effects , Retinal Neurons/physiology
5.
Invest Ophthalmol Vis Sci ; 44(5): 2235-44, 2003 May.
Article in English | MEDLINE | ID: mdl-12714666

ABSTRACT

PURPOSE: A recent study has shown that glia-derived neurotrophic factor (GDNF) and docosahexaenoic acid (DHA) promote the survival and differentiation of retina photoreceptors. The current study was undertaken to investigate whether these molecules participate in cell cycle regulation in retinal progenitors in vitro. METHODS: Developmental changes in the expression of the stem cell marker nestin and of cell cycle and differentiated neuron markers were analyzed in neuroblasts obtained from 1-day-old rat retinas. The effects of GDNF and DHA on those changes were then determined. RESULTS: Expression of nestin, found in more than one third of neuroblasts at day 1, rapidly decreased during development, with most neuroblasts acquiring the photoreceptor phenotype. GDNF increased the percentage of photoreceptor progenitors expressing nestin, whereas DHA reduced it, simultaneously enhancing photoreceptor differentiation. Several markers of cell cycle progression indicated that photoreceptor progenitors maintained an active cell cycle during the first 2 days in vitro. GDNF stimulated the cell cycle, increasing the number of dividing cells and generating more photoreceptor progenitors, whereas DHA induced cell cycle exit and photoreceptor differentiation. Analysis of the expression of the cyclin-Cdk inhibitor p27(Kip1) confirmed these results. CONCLUSIONS: GDNF and DHA acted as molecular cues, counterbalancing the decision of photoreceptors to remain in or exit the cell cycle. The results strongly suggest that both factors participate in determining the number of photoreceptors in vitro, regulating the cell cycle and survival at early and late stages of development, respectively. Hence, GDNF and DHA may coordinately control the histogenesis of photoreceptors in the retina by modulating both neurogenesis and apoptosis.


Subject(s)
Cell Cycle/drug effects , Docosahexaenoic Acids/pharmacology , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins , Retina/cytology , Stem Cells/cytology , Animals , Animals, Newborn , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p27 , Down-Regulation , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes/metabolism , Glial Cell Line-Derived Neurotrophic Factor , Indoles/metabolism , Intermediate Filament Proteins/metabolism , Nestin , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar , Retina/metabolism , Rod Opsins/metabolism , Stem Cells/metabolism , Tumor Suppressor Proteins/metabolism
6.
Invest Ophthalmol Vis Sci ; 44(5): 2252-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12714668

ABSTRACT

PURPOSE: In a recent study, it was demonstrated that docosahexaenoic acid (DHA) promotes the survival of retinal photoreceptors in vitro, delaying apoptosis. However, lipid enrichment in DHA is known to contribute to retina vulnerability to oxidative stress. In this study, the effect of oxidative damage on rat retina neurons in vitro and whether DHA enhances or diminishes this damage were investigated. METHODS: Rat retina neurons in 3-day cultures, with or without DHA, were treated with the oxidant paraquat. After 24 hours, apoptosis, mitochondrial membrane integrity, and Bcl-2 and Bax expression were immunocytochemically determined. RESULTS: Paraquat induced apoptosis in amacrine and photoreceptor neurons, major neuronal types in the culture. Neuronal apoptosis was accompanied by mitochondrial membrane depolarization, an increase in the amount of photoreceptors expressing Bax, and a decrease in those expressing Bcl-2. Addition of DHA reduced photoreceptor apoptosis by almost half, simultaneously preserving their mitochondrial membrane integrity. DHA blocked the paraquat-induced increase in Bax expression and remarkably upregulated Bcl-2 expression. Glia-derived neurotrophic factor, a photoreceptor trophic factor, only slightly increased Bcl-2 expression and did not protect photoreceptors from oxidative damage. Similarly, other fatty acids tested did not prevent photoreceptor apoptosis. CONCLUSIONS: These results show that oxidative damage induces apoptosis in retinal neurons during their early development in culture and suggest that the loss of mitochondrial membrane integrity is crucial in the apoptotic death of these cells. DHA activates intracellular mechanisms that prevent this loss and by modulating the levels of pro- and antiapoptotic proteins of the Bcl-2 family selectively protect photoreceptors from oxidative stress.


Subject(s)
Apoptosis/drug effects , Cytoprotection/drug effects , Docosahexaenoic Acids/pharmacology , Herbicides/toxicity , Oxidative Stress/drug effects , Paraquat/toxicity , Photoreceptor Cells, Vertebrate/cytology , Animals , Cell Culture Techniques , Cell Survival , Membrane Potentials/physiology , Mitochondria/physiology , Nerve Growth Factors/metabolism , Neurons/cytology , Neurons/drug effects , Photoreceptor Cells, Vertebrate/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Up-Regulation , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...