Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Exp Eye Res ; 224: 109222, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36041511

RESUMO

Retinal pigment epithelium (RPE) cells, essential for preserving retina homeostasis, also contribute to the development of retina proliferative diseases, through their exacerbated migration, epithelial to mesenchymal transition (EMT) and inflammatory response. Uncovering the mechanisms inducing these changes is crucial for designing effective treatments for these pathologies. Sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P) are bioactive sphingolipids that promote migration and inflammation in several cell types; we recently established that they stimulate the migration of retina Müller glial cells (Simón et al., 2015; Vera et al., 2021). We here analyzed whether S1P and C1P regulate migration, inflammation and EMT in RPE cells. We cultured two human RPE cell lines, ARPE-19 and D407 cells, and supplemented them with either 5 µM S1P or 10 µM C1P, or their vehicles, for 24 h. Analysis of cell migration by the scratch wound assay showed that S1P addition significantly enhanced migration in both cell lines. Pre-treatment with W146 and BML-241, antagonists for S1P receptor 1 (S1P1) and 3 (S1P3), respectively, blocked exogenous S1P-induced migration. Inhibiting sphingosine kinase 1 (SphK1), the enzyme involved in S1P synthesis, significantly reduced cell migration and exogenous S1P only partially restored it. Addition of C1P markedly stimulated cell migration. Whereas inhibiting C1P synthesis did not affect C1P-induced migration, inhibiting S1P synthesis strikingly decreased it; noteworthy, addition of C1P promoted the transcription of SphK1. These results suggest that S1P and C1P stimulate RPE cell migration and their effect requires S1P endogenous synthesis. Both S1P and C1P increase the transcription of pro-inflammatory cytokines IL-6 and IL-8, and of EMT marker α-smooth muscle actin (α-SMA) in ARPE-19 cells. Collectively, our results suggest new roles for S1P and C1P in the regulation of RPE cell migration and inflammation; since the deregulation of sphingolipid metabolism is involved in several proliferative retinopathies, targeting their metabolism might provide new tools for treating these pathologies.


Assuntos
Actinas , Epitélio Pigmentado da Retina , Humanos , Receptores de Esfingosina-1-Fosfato , Epitélio Pigmentado da Retina/metabolismo , Transição Epitelial-Mesenquimal , Interleucina-6 , Interleucina-8 , Lisofosfolipídeos/farmacologia , Lisofosfolipídeos/metabolismo , Esfingosina/farmacologia , Esfingosina/metabolismo , Ceramidas/farmacologia , Ceramidas/metabolismo , Inflamação/metabolismo , Fosfatos
2.
J Lipid Res ; 62: 100037, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32948663

RESUMO

Sphingolipids have emerged as bioactive lipids involved in the regulation of many physiological and pathological processes. In the retina, they have been established to participate in numerous processes, such as neuronal survival and death, proliferation and migration of neuronal and vascular cells, inflammation, and neovascularization. Dysregulation of sphingolipids is therefore crucial in the onset and progression of retinal diseases. This review examines the involvement of sphingolipids in retinal physiology and diseases. Ceramide (Cer) has emerged as a common mediator of inflammation and death of neuronal and retinal pigment epithelium cells in animal models of retinopathies such as glaucoma, age-related macular degeneration (AMD), and retinitis pigmentosa. Sphingosine-1-phosphate (S1P) has opposite roles, preventing photoreceptor and ganglion cell degeneration but also promoting inflammation, fibrosis, and neovascularization in AMD, glaucoma, and pro-fibrotic disorders. Alterations in Cer, S1P, and ceramide 1-phosphate may also contribute to uveitis. Notably, use of inhibitors that either prevent Cer increase or modulate S1P signaling, such as Myriocin, desipramine, and Fingolimod (FTY720), preserves neuronal viability and retinal function. These findings underscore the relevance of alterations in the sphingolipid metabolic network in the etiology of multiple retinopathies and highlight the potential of modulating their metabolism for the design of novel therapeutic approaches.


Assuntos
Esfingolipídeos
3.
J Neurochem ; 159(5): 840-856, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34133756

RESUMO

Pigment epithelium-derived factor (PEDF) is a cytoprotective protein for the retina. We hypothesize that this protein acts on neuronal survival and differentiation of photoreceptor cells in culture. The purpose of the present study was to evaluate the neurotrophic effects of PEDF and its fragments in an in vitro model of cultured primary retinal neurons that die spontaneously in the absence of trophic factors. We used Wistar albino rats. Cell death was assayed by immunofluorescence and flow cytometry through TUNEL assay, propidium iodide, mitotracker, and annexin V. Immunofluorescence of cells for visualizing rhodopsin, CRX, and antisyntaxin under confocal microscopy was performed. Neurite outgrowth was also quantified. Results show that PEDF protected photoreceptor precursors from apoptosis, preserved mitochondrial function and promoted polarization of opsin enhancing their developmental process, as well as induced neurite outgrowth in amacrine neurons. These effects were abolished by an inhibitor of the PEDF receptor or receptor-derived peptides that block ligand/receptor interactions. While all the activities were specifically conferred by short peptide fragments (17 amino acid residues) derived from the PEDF neurotrophic domain, no effects were triggered by peptides from the PEDF antiangiogenic region. The observed effects on retinal neurons imply a specific activation of the PEDF receptor by a small neurotrophic region of PEDF. Our findings support the neurotrophic PEDF peptides as neuronal guardians for the retina, highlighting their potential as promoters of retinal differentiation, and inhibitors of retinal cell death and its blinding consequences. Cover Image for this issue: https://doi.org/10.1111/jnc.15089.


Assuntos
Células Amácrinas/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proteínas do Olho/farmacologia , Fatores de Crescimento Neural/farmacologia , Crescimento Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Serpinas/farmacologia , Células Amácrinas/fisiologia , Sequência de Aminoácidos , Animais , Diferenciação Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Proteínas do Olho/genética , Feminino , Masculino , Fatores de Crescimento Neural/genética , Crescimento Neuronal/fisiologia , Neurônios/fisiologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Células Fotorreceptoras de Vertebrados/fisiologia , Ratos , Ratos Wistar , Serpinas/genética
4.
Exp Eye Res ; 202: 108342, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33144094

RESUMO

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.


Assuntos
Diamino Aminoácidos/toxicidade , Células Ependimogliais/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/toxicidade , Doenças Retinianas/induzido quimicamente , Neurônios Retinianos/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Sobrevivência Celular/efeitos dos fármacos , Toxinas de Cianobactérias , Fragmentação do DNA/efeitos dos fármacos , Maleato de Dizocilpina/farmacologia , Células Ependimogliais/patologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Doenças Retinianas/metabolismo , Doenças Retinianas/prevenção & controle , Neurônios Retinianos/patologia
5.
Exp Eye Res ; 202: 108359, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33197453

RESUMO

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.


Assuntos
Ceramidas/farmacologia , Células Ependimogliais/citologia , Células Ganglionares da Retina/citologia , Animais , Animais Recém-Nascidos , Movimento Celular/efeitos dos fármacos , Células Ependimogliais/efeitos dos fármacos , Modelos Animais , Ratos , Ratos Wistar , Células Ganglionares da Retina/efeitos dos fármacos , Transdução de Sinais
6.
J Integr Neurosci ; 17(3-4): 347-353, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29081421

RESUMO

Microsaccade are sensitive to changes of perceptual inputs as well as modulations of cognitive states. There are just a few works analyzing microsaccade while subjects are processing complex information and fewer when doing predictions about upcoming events. To evaluate whether contextual predictability would change microsaccadic behavior, we evaluated microsaccade of twenty one persons when reading 40 regular sentences and 40 proverbs. Analysis of microsaccade during reading proverbs and regular sentences revealed that microsaccade rate on words before maxjump, during maxjump and words after maxjump varied depending on the kind of sentence and on the word predictability. Maxjump was defined as the word with the largest difference between the cloze predictability of two consecutive words. Low and high predictable words demanded less or more microsaccade on words previous, during and on maxjump depending of the semantic context and of the readers' predictions of upcoming words.In summary, the present study shows that microsaccade' rate evidenced significant differences when reading proverbs and regular sentences. Hence, evaluation of microsaccade during reading sentences with different contextual predictability might provide information about specific effect of cue attention on complex task.


Assuntos
Antecipação Psicológica , Leitura , Movimentos Sacádicos , Adulto , Aforismos e Provérbios como Assunto , Atenção , Humanos
7.
Biochim Biophys Acta ; 1863(6 Pt A): 1134-45, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26883505

RESUMO

Age-related macular degeneration (AMD) is among the main pathologies leading to blindness in adults and has currently no cure or effective treatment. Selective apoptosis of retina pigment epithelial (RPE) cells results in the progressive loss of photoreceptor neurons, with the consequent gradual vision loss. Oxidative stress plays an important role in this process. We have previously determined that activation of RXRs protects rat photoreceptor neurons from oxidative stress-induced apoptosis. In this study we investigated whether RXR ligands prevented apoptosis in an RPE cell line, D407 cells, exposed to hydrogen peroxide (H2O2). H2O2 induced apoptosis of D407 cells, promoting p65NFκB nuclear translocation, increasing Bax mRNA expression, activating caspase-3 and altering cell morphology. We show, for the first time, that HX630, a RXR pan-agonist, protected D407 cells from H2O2-induced apoptosis, preventing p65NFκB nuclear translocation, increasing Bclxl and PPARγ mRNA levels and simultaneously decreasing Bax mRNA levels and caspase-3 activation. Pretreatment with a RXR antagonist blocked HX630 protection. LG100754, which binds RXRs but only activates heterodimers and is an antagonist of RXR homodimers, also had a protective effect. In addition, only agonists known to bind to RXR/PPARγ were protective. As a whole, our results suggest that RXR activation protects RPE cells from oxidative stress-induced apoptosis and this protection might involve signaling through a heterodimeric receptor, such as RXR/PPARγ. These data also imply that RXR agonists might provide potential pharmacological tools for treating retina degenerative diseases.


Assuntos
Apoptose/fisiologia , Epitélio Pigmentado da Retina/metabolismo , Receptores X de Retinoides/metabolismo , Transdução de Sinais/fisiologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Benzazepinas/farmacologia , Benzoatos/farmacologia , Western Blotting , Caspase 3/metabolismo , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Microscopia Confocal , Oxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , PPAR gama/genética , PPAR gama/metabolismo , Substâncias Protetoras/farmacologia , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Receptores X de Retinoides/agonistas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
8.
J Neurochem ; 136(5): 931-46, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26662863

RESUMO

Oxidative stress is involved in activating photoreceptor death in several retinal degenerations. Docosahexaenoic acid (DHA), the major polyunsaturated fatty acid in the retina, protects cultured retina photoreceptors from apoptosis induced by oxidative stress and promotes photoreceptor differentiation. Here, we investigated whether eicosapentaenoic acid (EPA), a metabolic precursor to DHA, had similar effects and whether retinal neurons could metabolize EPA to DHA. Adding EPA to rat retina neuronal cultures increased opsin expression and protected photoreceptors from apoptosis induced by the oxidants paraquat and hydrogen peroxide (H2 O2 ). Palmitic, oleic, and arachidonic acids had no protective effect, showing the specificity for DHA. We found that EPA supplementation significantly increased DHA percentage in retinal neurons, but not EPA percentage. Photoreceptors and glial cells expressed Δ6 desaturase (FADS2), which introduces the last double bond in DHA biosynthetic pathway. Pre-treatment of neuronal cultures with CP-24879 hydrochloride, a Δ5/Δ6 desaturase inhibitor, prevented EPA-induced increase in DHA percentage and completely blocked EPA protection and its effect on photoreceptor differentiation. These results suggest that EPA promoted photoreceptor differentiation and rescued photoreceptors from oxidative stress-induced apoptosis through its elongation and desaturation to DHA. Our data show, for the first time, that isolated retinal neurons can synthesize DHA in culture. Docosahexaenoic acid (DHA), the major polyunsaturated fatty acid in retina photoreceptors, and its precursor, eicosapentaenoic acid (EPA) have multiple beneficial effects. Here, we show that retina neurons in vitro express the desaturase FADS2 and can synthesize DHA from EPA. Moreover, addition of EPA to these cultures protects photoreceptors from oxidative stress and promotes their differentiation through its metabolization to DHA.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Ácido Eicosapentaenoico/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Mitocôndrias/metabolismo , Paraquat/farmacologia , Substâncias Protetoras/farmacologia , Ratos Wistar , Retina/metabolismo
9.
Photochem Photobiol Sci ; 14(9): 1737-53, 2015 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-26204250

RESUMO

Due to its constant exposure to light and its high oxygen consumption the retina is highly sensitive to oxidative damage, which is a common factor in inducing the death of photoreceptors after light damage or in inherited retinal degenerations. The high content of docosahexaenoic acid (DHA), the major polyunsaturated fatty acid in the retina, has been suggested to contribute to this sensitivity. DHA is crucial for developing and preserving normal visual function. However, further roles of DHA in the retina are still controversial. Current data support that it can tilt the scale either towards degeneration or survival of retinal cells. DHA peroxidation products can be deleterious to the retina and might lead to retinal degeneration. However, DHA has also been shown to act as, or to be the source of, a survival molecule that protects photoreceptors and retinal pigment epithelium cells from oxidative damage. We have established that DHA protects photoreceptors from oxidative stress-induced apoptosis and promotes their differentiation in vitro. DHA activates the retinoid X receptor (RXR) and the ERK/MAPK pathway, thus regulating the expression of anti and pro-apoptotic proteins. It also orchestrates a diversity of signaling pathways, modulating enzymatic pathways that control the sphingolipid metabolism and activate antioxidant defense mechanisms to promote photoreceptor survival and development. A deeper comprehension of DHA signaling pathways and context-dependent behavior is required to understand its dual functions in retinal physiology.


Assuntos
Ácidos Docosa-Hexaenoicos/metabolismo , Luz , Peróxidos Lipídicos/metabolismo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/efeitos da radiação , Animais , Morte Celular/fisiologia , Morte Celular/efeitos da radiação , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Humanos , Luz/efeitos adversos , Estresse Oxidativo/fisiologia , Estresse Oxidativo/efeitos da radiação , Degeneração Retiniana/etiologia , Degeneração Retiniana/metabolismo , Receptores X de Retinoides/metabolismo
10.
Biochim Biophys Acta Mol Cell Res ; 1871(8): 119816, 2024 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-39159686

RESUMO

Exposure to the non-protein amino acid cyanotoxin ß-N-methylamino-L-alanine (BMAA), released by cyanobacteria found in many water reservoirs has been associated with neurodegenerative diseases. We previously demonstrated that BMAA induced cell death in both retina photoreceptors (PHRs) and amacrine neurons by triggering different molecular pathways, as activation of NMDA receptors and formation of carbamate-adducts was only observed in amacrine cell death. We established that activation of Retinoid X Receptors (RXR) protects retinal cells, including retina pigment epithelial (RPE) cells from oxidative stress-induced apoptosis. We now investigated the mechanisms underlying BMAA toxicity in these cells and those involved in RXR protection. BMAA addition to rat retinal neurons during early development in vitro increased reactive oxygen species (ROS) generation and polyADP ribose polymers (PAR) formation, while pre-treatment with serine (Ser) before BMAA addition decreased PHR death. Notably, RXR activation with the HX630 agonist prevented BMAA-induced death in both neuronal types, reducing ROS generation, preserving mitochondrial potential, and decreasing TUNEL-positive cells and PAR formation. This suggests that BMAA promoted PHR death by substituting Ser in polypeptide chains and by inducing polyADP ribose polymerase activation. BMAA induced cell death in ARPE-19 cells, a human epithelial cell line; RXR activation prevented this death, decreasing ROS generation and caspase 3/7 activity. These findings suggest that RXR activation prevents BMAA harmful effects on retinal neurons and RPE cells, supporting this activation as a broad-spectrum strategy for treating retina degenerations.

11.
J Lipid Res ; 54(8): 2236-2246, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23723389

RESUMO

We have established that docosahexaenoic acid (DHA), the major polyunsaturated fatty acid in the retina, promotes survival of rat retina photoreceptors during early development in vitro and upon oxidative stress by activating the ERK/MAPK signaling pathway. Here we have investigated whether DHA turns on this pathway through activation of retinoid X receptors (RXRs) or by inducing tyrosine kinase (Trk) receptor activation. We also evaluated whether DHA release from phospholipids was required for its protective effect. Addition of RXR antagonists (HX531, PA452) to rat retinal neuronal cultures inhibited DHA protection during early development in vitro and upon oxidative stress induced with Paraquat or H2O2. In contrast, the Trk inhibitor K252a did not affect DHA prevention of photoreceptor apoptosis. These results imply that activation of RXRs was required for DHA protection whereas Trk receptors were not involved in this protection. Pretreatment with 4-bromoenol lactone, a phospholipase A2 inhibitor, blocked DHA prevention of oxidative stress-induced apoptosis of photoreceptors. It is noteworthy that RXR agonists (HX630, PA024) also rescued photoreceptors from H2O2-induced apoptosis. These results provide the first evidence that activation of RXRs prevents photoreceptor apoptosis and suggest that DHA is first released from phospholipids and then activates RXRs to promote the survival of photoreceptors.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Células Fotorreceptoras Retinianas Bastonetes/efeitos dos fármacos , Receptores X de Retinoides/metabolismo , Animais , Apoptose/efeitos dos fármacos , Benzoatos/farmacologia , Compostos de Bifenilo/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/química , Relação Dose-Resposta a Droga , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Células Fotorreceptoras Retinianas Bastonetes/citologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Receptores X de Retinoides/antagonistas & inibidores , Relação Estrutura-Atividade
12.
J Biol Chem ; 287(29): 24092-102, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22645143

RESUMO

Leukemia inhibitory factor (LIF), an interleukin-6 family neurocytokine, is up-regulated in response to different types of retinal stress and has neuroprotective activity through activation of the gp130 receptor/STAT3 pathway. We observed that LIF induces rapid, robust, and sustained activation of STAT3 in both the retina and retinal pigmented epithelium (RPE). Here, we tested whether LIF-induced STAT3 activation within the RPE can down-regulate RPE65, the central enzyme in the visual cycle that provides the 11-cis-retinal chromophore to photoreceptors in vivo. We generated conditional knock-out mice to specifically delete STAT3 or gp130 in RPE, retina, or both RPE and retina. After intravitreal injection of LIF, we analyzed the expression levels of visual cycle genes and proteins, isomerase activity of RPE65, levels of rhodopsin protein, and the rates of dark adaptation and rhodopsin regeneration. We found that RPE65 protein levels and isomerase activity were reduced and recovery of bleachable rhodopsin was delayed in LIF-injected eyes. In mice with functional gp130/STAT3 signaling in the retina, rhodopsin protein was also reduced by LIF. However, the LIF-induced down-regulation of RPE65 required a functional gp130/STAT3 cascade intrinsic to RPE. Our data demonstrate that a single cytokine, LIF, can simultaneously and independently affect both RPE and photoreceptors through the same signaling cascade to reduce the generation and utilization of 11-cis-retinal.


Assuntos
Fator Inibidor de Leucemia/farmacologia , Epitélio Pigmentado da Retina/metabolismo , Animais , Western Blotting , Receptor gp130 de Citocina/genética , Receptor gp130 de Citocina/metabolismo , Regulação para Baixo/efeitos dos fármacos , Eletrorretinografia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Retina/efeitos dos fármacos , Retina/metabolismo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , cis-trans-Isomerases/genética , cis-trans-Isomerases/metabolismo
13.
J Neurosci Res ; 90(2): 407-21, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21972118

RESUMO

Using stem cells to replace lost neurons is a promising strategy for treating retinal neurodegenerative diseases. Among their multiple functions, Müller glial cells are retina stem cells, with a robust regenerative potential in lower vertebrates, which is much more restricted in mammals. In rodents, most retina progenitors exit the cell cycle immediately after birth, differentiate as neurons, and then cannot reenter the cell cycle. Here we demonstrate that, in mixed cultures with Müller glial cells, rat retina progenitor cells expressed stem cell properties, maintained their proliferative potential, and were able to preserve these properties and remain mitotically active after several consecutive passages. Notably, these progenitors retained the capacity to differentiate as photoreceptors, even after successive reseedings. Müller glial cells markedly stimulated differentiation of retina progenitors; these cells initially expressed Crx and then developed as mature photoreceptors that expressed characteristic markers, such as opsin and peripherin. Moreover, they were light responsive, insofar as they decreased their cGMP levels when exposed to light, and they also showed high-affinity glutamate uptake, a characteristic of mature photoreceptors. Our present findings indicate that, in addition to giving rise to new photoreceptors, Müller glial cells might instruct a pool of undifferentiated cells to develop and preserve stem cell characteristics, even after successive reseedings, and then stimulate their differentiation as functional photoreceptors. This complementary mechanism might contribute to enlarge the limited regenerative capacity of mammalian Müller cells.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Neuroglia/fisiologia , Células Fotorreceptoras/fisiologia , Retina/citologia , Retina/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Células Cultivadas , Técnicas de Cocultura , Células-Tronco Neurais/citologia , Neuroglia/classificação , Células Fotorreceptoras/citologia , Ratos , Ratos Wistar , Células-Tronco/fisiologia
14.
World J Stem Cells ; 13(10): 1446-1479, 2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34786153

RESUMO

Retinal degeneration is a major contributor to visual dysfunction worldwide. Although it comprises several eye diseases, loss of retinal pigment epithelial (RPE) and photoreceptor cells are the major contributors to their pathogenesis. Early therapies included diverse treatments, such as provision of anti-vascular endothelial growth factor and many survival and trophic factors that, in some cases, slow down the progression of the degeneration, but do not effectively prevent it. The finding of stem cells (SC) in the eye has led to the proposal of cell replacement strategies for retina degeneration. Therapies using different types of SC, such as retinal progenitor cells (RPCs), embryonic SC, pluripotent SCs (PSCs), induced PSCs (iPSCs), and mesenchymal stromal cells, capable of self-renewal and of differentiating into multiple cell types, have gained ample support. Numerous preclinical studies have assessed transplantation of SC in animal models, with encouraging results. The aim of this work is to revise the different preclinical and clinical approaches, analyzing the SC type used, their efficacy, safety, cell attachment and integration, absence of tumor formation and immunorejection, in order to establish which were the most relevant and successful. In addition, we examine the questions and concerns still open in the field. The data demonstrate the existence of two main approaches, aimed at replacing either RPE cells or photoreceptors. Emerging evidence suggests that RPCs and iPSC are the best candidates, presenting no ethical concerns and a low risk of immunorejection. Clinical trials have already supported the safety and efficacy of SC treatments. Serious concerns are pending, such as the risk of tumor formation, lack of attachment or integration of transplanted cells into host retinas, immunorejection, cell death, and also ethical. However, the amazing progress in the field in the last few years makes it possible to envisage safe and effective treatments to restore vision loss in a near future.

15.
Biochim Biophys Acta Mol Cell Res ; 1868(11): 119098, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34271041

RESUMO

Photoreceptor cell (PHR) death is a hallmark of most retinal neurodegenerative diseases, in which inflammation plays a critical role. Activation of retinoid X receptors (RXR) modulates and integrates multiple cell functions, and has beneficial effects in animal models of chronic inflammatory diseases. Nonetheless, the mechanisms involved and their role in retina neuroprotection are poorly understood. In this work we assessed whether RXR activation prevents inflammation and/or PHR death in retinitis pigmentosa, an inherited retina neurodegeneration, using as an ex vivo model, retinas from the rd1 mice, a murine model of this disease. We demonstrated that rd1 retinas had lower levels of RXR alpha isoform than their wt counterparts at early developmental times, whereas its distribution pattern remained similar. In mixed neuro-glial cultures obtained from either rd1 or wt retinas, both PHR and Müller glial cells (MGC) expressed RXRalpha, and RXR activation by its synthetic pan-agonist PA024 selectively increased mRNA levels of RXRgamma isoform. PA024 decreased PHR death in rd1 mixed cultures; it reduced the amount of non-viable neurons, delayed the onset of PHR apoptosis, and decreased Bax mRNA levels. PA024 also reduced MGC reactivity in vitro before and at the onset of degeneration, decreasing GFAP expression, increasing glutamine synthetase mRNA levels, and promoting the transcription of the anti-inflammatory cytokine, Il-10. These results suggest that RXR activation rescues rd1 PHR and decreases MGC reactivity, promoting an anti-inflammatory environment in the rd1 retina, thus supporting the potential of RXR agonists as pharmacological tools for treating retina degenerative diseases.


Assuntos
Modelos Animais de Doenças , Inflamação/metabolismo , Células Fotorreceptoras/metabolismo , Retinose Pigmentar/metabolismo , Receptores X de Retinoides/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
16.
J Lipid Res ; 51(6): 1247-62, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20100817

RESUMO

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.


Assuntos
Retina/citologia , Retina/crescimento & desenvolvimento , Esfingolipídeos/metabolismo , Animais , Apoptose , Sobrevivência Celular , Humanos , Retina/metabolismo , Transdução de Sinais , Esfingolipídeos/química
17.
J Neurochem ; 110(5): 1648-60, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19575708

RESUMO

The insulin receptor (IR) and IR signaling proteins are widely distributed throughout the CNS. IR signaling provides a trophic signal for transformed retinal neurons in culture and we recently reported that deletion of IR in rod photoreceptors by Cre/lox system resulted in stress-induced photoreceptor degeneration. These studies suggest a neuroprotective role of IR in rod photoreceptor cell function. However, there are no studies available on the role of insulin-induced IR signaling in the development of normal photoreceptors. To examine the role of insulin-induced IR signaling, we analyzed cultured neuronal cells isolated from newborn rodent retinas. In insulin-lacking cultures, photoreceptors from wild-type rat retinas exhibited an abnormal morphology with a wide axon cone and disorganization of the actin and tubulin cytoskeleton. Photoreceptors from IR knockout mouse retinas also exhibited a similar abnormal morphology. A novel finding in this study was that addition of docosahexaenoic acid, a photoreceptor trophic factor, restored normal axonal outgrowth in insulin-lacking cultures. These data suggest that IR signaling pathways regulate actin and tubulin cytoskeletal organization in photoreceptors; they also imply that insulin and docosahexaenoic acid activate at least partially overlapping signaling pathways that are essential for the development of normal photoreceptors.


Assuntos
Actinas , Citoesqueleto/metabolismo , Células Fotorreceptoras/fisiologia , Receptor de Insulina/fisiologia , Transdução de Sinais , Actinas/antagonistas & inibidores , Actinas/metabolismo , Actinas/fisiologia , Animais , Células Cultivadas , Citoesqueleto/patologia , Citoesqueleto/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Células Fotorreceptoras/citologia , Células Fotorreceptoras/patologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptor de Insulina/deficiência , Receptor de Insulina/genética , Transdução de Sinais/genética
18.
J Neurosci Res ; 87(4): 964-77, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18855938

RESUMO

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.


Assuntos
Desdiferenciação Celular , Proliferação de Células , Neuroglia/fisiologia , Estresse Oxidativo/fisiologia , Retina/citologia , Neurônios Retinianos/citologia , Actinas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Desdiferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Peróxido de Hidrogênio/farmacologia , Potencial da Membrana Mitocondrial , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Oxidantes/farmacologia , Paraquat/farmacologia , Ratos , Ratos Wistar , Retina/efeitos dos fármacos , Neurônios Retinianos/fisiologia
19.
Front Cell Neurosci ; 13: 246, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31244608

RESUMO

The sphingolipids ceramide (Cer), sphingosine-1-phosphate (S1P), sphingosine (Sph), and ceramide-1-phosphate (C1P) are key signaling molecules that regulate major cellular functions. Their roles in the retina have gained increasing attention during the last decade since they emerge as mediators of proliferation, survival, migration, neovascularization, inflammation and death in retina cells. As exacerbation of these processes is central to retina degenerative diseases, they appear as crucial players in their progression. This review analyzes the functions of these sphingolipids in retina cell types and their possible pathological roles. Cer appears as a key arbitrator in diverse retinal pathologies; it promotes inflammation in endothelial and retina pigment epithelium (RPE) cells and its increase is a common feature in photoreceptor death in vitro and in animal models of retina degeneration; noteworthy, inhibiting Cer synthesis preserves photoreceptor viability and functionality. In turn, S1P acts as a double edge sword in the retina. It is essential for retina development, promoting the survival of photoreceptors and ganglion cells and regulating proliferation and differentiation of photoreceptor progenitors. However, S1P has also deleterious effects, stimulating migration of Müller glial cells, angiogenesis and fibrosis, contributing to the inflammatory scenario of proliferative retinopathies and age related macular degeneration (AMD). C1P, as S1P, promotes photoreceptor survival and differentiation. Collectively, the expanding role for these sphingolipids in the regulation of critical processes in retina cell types and in their dysregulation in retina degenerations makes them attractive targets for treating these diseases.

20.
Mol Neurobiol ; 56(7): 4760-4777, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30387075

RESUMO

Ceramide (Cer) has a key role inducing cell death and has been proposed as a messenger in photoreceptor cell death in the retina. Here, we explored the pathways induced by C2-acetylsphingosine (C2-Cer), a cell-permeable Cer, to elicit photoreceptor death. Treating pure retina neuronal cultures with 10 µM C2-Cer for 6 h selectively induced photoreceptor death, decreasing mitochondrial membrane potential and increasing the formation of reactive oxygen species (ROS). In contrast, amacrine neurons preserved their viability. Noteworthy, the amount of TUNEL-labeled cells and photoreceptors expressing cleaved caspase-3 remained constant and pretreatment with a pan-caspase inhibitor did not prevent C2-Cer-induced death. C2-Cer provoked polyADP ribosyl polymerase-1 (PARP-1) overactivation. Inhibiting PARP-1 decreased C2-Cer-induced photoreceptor death; C2-Cer increased polyADP ribose polymer (PAR) levels and induced the translocation of apoptosis inducing factor (AIF) from mitochondria to photoreceptor nuclei, which was prevented by PARP-1 inhibition. Pretreatment with a calpain and cathepsin inhibitor and with a calpain inhibitor reduced photoreceptor death, whereas selective cathepsin inhibitors granted no protection. Combined pretreatment with a PARP-1 and a calpain inhibitor evidenced the same protection as each inhibitor by itself. Neither autophagy nor necroptosis was involved in C2-Cer-elicited death; no increase in LDH release was observed upon C2-Cer treatment and pretreatment with inhibitors of necroptosis and autophagy did not rescue photoreceptors. These results suggest that C2-Cer induced photoreceptor death by a novel, caspase-independent mechanism, involving activation of PARP-1, decline of mitochondrial membrane potential, calpain activation, and AIF translocation, all of which are biochemical features of parthanatos.


Assuntos
Ceramidas/farmacologia , Parthanatos/efeitos dos fármacos , Células Fotorreceptoras de Vertebrados/patologia , Animais , Fator de Indução de Apoptose/metabolismo , Calpaína/metabolismo , Caspases/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Modelos Biológicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA