Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Int J Mol Sci ; 24(15)2023 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-37569482

RESUMEN

To explore the temporal profile of retinal proteomes specific to primary and secondary retinal ganglion cell (RGC) loss. Unilateral partial optic nerve transection (pONT) was performed on the temporal side of the rat optic nerve. Temporal and nasal retinal samples were collected at 1, 4 and 8 weeks after pONT (n = 4 each) for non-biased profiling with a high-resolution hybrid quadrupole time-of-flight mass spectrometry running on label-free SWATHTM acquisition (SCIEX). An information-dependent acquisition ion library was generated using ProteinPilot 5.0 and OneOmics cloud bioinformatics. Combined proteome analysis detected 2531 proteins with a false discovery rate of <1%. Compared to the nasal retina, 10, 25 and 61 significantly regulated proteins were found in the temporal retina at 1, 4, and 8 weeks, respectively (p < 0.05, FC ≥ 1.4 or ≤0.7). Eight proteins (ALDH1A1, TRY10, GFAP, HBB-B1, ALB, CDC42, SNCG, NEFL) were differentially expressed for at least two time points. The expressions of ALDH1A1 and SNCG at nerve fibers were decreased along with axonal loss. Increased ALDH1A1 localization in the inner nuclear layer suggested stress response. Increased GFAP expression demonstrated regional reactivity of astrocytes and Muller cells. Meta-analysis of gene ontology showed a pronounced difference in endopeptidase and peptidase inhibitor activity. Temporal proteomic profiling demonstrates established and novel protein targets associated with RGC damage.

2.
Int J Mol Sci ; 22(16)2021 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-34445296

RESUMEN

To investigate the retinal proteins associated with primary and secondary retinal ganglion cell (RGC) degeneration and explore their molecular pathways, SWATH label-free and target-based mass spectrometry was employed to identify the proteomes in various retinal locations in response to localized optic nerve injury. Unilateral partial optic nerve transection (pONT) was performed on adult Wistar rats and their retinas were harvested 2 weeks later. To confirm the separation of primary and secondary RGC degeneration, immunohistochemistry of RNA binding protein with multiple splicing (RBPMS) and glial fibrillary acidic protein (GFAP) was performed on retinal whole-mounts. Retinal proteomes in the temporal and nasal quadrants were evaluated with high resolution hybrid quadrupole time-of-flight mass spectrometry (QTOF-MS), and SWATH-based acquisition, and their expression was compared to the corresponding retinal quadrant in contralateral control eyes and further validated by multiple reaction monitoring mass spectrometry (MRM-MS). A total of 3641 proteins (FDR < 1%) were identified using QTOF-MS. The raw data are available via ProteomeXchange with the identifier PXD026783. Bioinformatics data analysis showed that there were 37 upregulated and 25 downregulated proteins in the temporal quadrant, whereas 20 and five proteins were upregulated and downregulated, respectively, in the nasal quadrant, respectively (n = 4, p < 0.05; fold change ≥ 1.4-fold or ≤0.7). Six proteins were regulated in both the temporal and the nasal quadrants, including CLU, GFAP, GNG5, IRF2BPL, L1CAM, and CPLX1. Linear regression analysis indicated a strong association between the data obtained by means of SWATH-MS and MRM-MS (temporal, R2 = 0.97; nasal, R2 = 0.96). Gene ontology analysis revealed statistically significant changes in the biological processes and cellular components of primary RGC degeneration. The majority of the significant changes in structural, signaling, and cell death proteins were associated with the loss of RGCs in the area of primary RGC degeneration. The combined use of SWATH-MS and MRM-MS methods detects and quantifies regional changes of retinal protein expressions after localized injury. Future investigation with this integrated approach will significantly increase the understanding of diverse processes of progressive RGC degeneration from a proteomic prospective.


Asunto(s)
Proteínas del Ojo/metabolismo , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Proteínas del Ojo/análisis , Espectrometría de Masas/métodos , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Traumatismos del Nervio Óptico/complicaciones , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/patología , Proteoma/análisis , Proteoma/metabolismo , Proteómica/métodos , Ratas , Ratas Wistar , Retina/química , Retina/metabolismo , Degeneración Retiniana/etiología , Degeneración Retiniana/patología , Células Ganglionares de la Retina/patología
3.
Exp Eye Res ; 140: 1-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26277582

RESUMEN

Programmed cell death-1 (PD-1) is a key negative receptor inducibly expressed on T cells, B cells and dendritic cells. It was discovered on T cells undergoing classical programmed cell death. Studies showed that PD-1 ligation promotes retinal ganglion cell (RGC) death during retinal development. The purpose of this present study is to characterize PD-1 regulation in the retina after optic nerve crush (ONC). C57BL/6 mice were subjected to ONC and RGC loss was monitored by immunolabelling with RNA-binding protein with multiple splicing (Rbpms). Time course of PD-1 mRNA expression was determined by real-time PCR. PD-1 expression was detected with anti-PD-1 antibody on whole mount retinas. PD-1 staining intensity was quantitated. Colocalization of PD-1 and cleaved-caspase-3 after ONC was analyzed. Real-time PCR results demonstrated that PD-1 gene expression was significantly upregulated at day 1, 3, 7, 10 and 14 after ONC. Immunofluorescent staining revealed a dramatic increase of PD-1 expression following ONC. In both control and injured retinas, PD-1 tended to be up-expressed in a subtype of RGCs, whose somata size were significantly larger than others. Compared to control, PD-1 intensity in large RGCs was increased by 82% in the injured retina. None of the large RGCs expressed cleaved-caspase-3 at day 5 after ONC. Our work presents the first evidence of PD-1 induction in RGCs after ONC. This observation supports further investigation into the role of PD-1 expression during RGC death or survival following injury.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Traumatismos del Nervio Óptico/genética , Receptor de Muerte Celular Programada 1/genética , Células Ganglionares de la Retina/metabolismo , Animales , Apoptosis , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente Indirecta , Ratones , Ratones Endogámicos C57BL , Compresión Nerviosa , Traumatismos del Nervio Óptico/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
4.
Exp Eye Res ; 112: 21-8, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23603611

RESUMEN

Intraocular pressure (IOP) elevation is considered as a major risk factor causing the progression of vision deterioration in glaucoma. Although it is known that the IOP level changes widely throughout the day and night, how the dark or light phase IOP elevation contributes to retinal ganglion cell (RGC) degeneration is still largely unclear. To examine the profile of IOP, modified laser photocoagulation was applied to the trabecular meshwork of Brown Norway rats and both light and dark phase IOPs were monitored approximately 1-2 times a week. The relationship between IOP elevation and RGC degeneration was investigated while RGC body loss was analyzed with Rbpms immunolabeling on retinal wholemount and axonal injury in the optic nerve was semi-quantified. The baseline awake dark and light IOPs were 30.4 ± 2.7 and 20.2 ± 2.1 mmHg respectively. The average dark IOP was increased to 38.2 ± 3.2 mmHg for five weeks after the laser treatment on 270° trabecular meshwork. However, there was no significant loss of RGC body and axonal injury. After laser treatment on 330° trabecular meshwork, the dark and light IOPs were significantly increased to 43.8 ± 4.6 and 23 ± 3.7 mmHg respectively for 5 weeks. The cumulative dark and light IOP elevations were 277 ± 86 and 113 ± 50 mmHg days respectively while the cumulative total (light and dark) IOP elevation was 213 ± 114 mmHg days. After 5 weeks, regional RGC body loss of 29.5 ± 15.5% and moderate axonal injury were observed. Axonal injury and loss of RGC body had a high correlation with the cumulative total IOP elevation (R(2) = 0.60 and 0.65 respectively). There was an association between the cumulative dark IOP elevation and RGC body loss (R(2) = 0.37) and axonal injury (R(2) = 0.51) whereas the associations between neuronal damages and the cumulative light IOP elevation were weak (for RGC body loss, R(2) = 0.01; for axonal injury, R(2) = 0.26). Simple linear regression model analysis showed statistical significance for the relationships between the total cumulative IOP elevation and RGC body loss (P = 0.009), and axonal injury (P = 0.016). To examine the role of light and dark IOP elevation in RGC body loss and axonal injury, analyses for the association between different light/dark IOP factors and percentage of RGC body loss/axonal injury grading were performed and only the association between the cumulative dark IOP elevation and axonal injury showed statistical significance (P = 0.033). The findings demonstrated that the cumulative total (light and dark) IOP elevation is a risk factor to RGC degeneration in a rat model of experimental glaucoma using modified partial laser photocoagulation at 330° trabecular meshwork. Further investigations are required to understand the role of longer term light and dark phase IOP elevation contributing to the progression of degeneration in different compartments of RGCs.


Asunto(s)
Adaptación a la Oscuridad , Modelos Animales de Enfermedad , Glaucoma/fisiopatología , Presión Intraocular/fisiología , Enfermedades del Nervio Óptico/fisiopatología , Degeneración Retiniana/fisiopatología , Células Ganglionares de la Retina/patología , Animales , Axones/patología , Biomarcadores/metabolismo , Glaucoma/metabolismo , Coagulación con Láser , Masculino , Enfermedades del Nervio Óptico/metabolismo , Proteínas de Unión al ARN/metabolismo , Ratas , Ratas Endogámicas BN , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo , Tonometría Ocular , Malla Trabecular/cirugía
5.
J Biol Chem ; 286(37): 32563-74, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21705333

RESUMEN

The choroid plexus lining the four ventricles in the brain is where the majority of cerebrospinal fluid (CSF) is produced. The secretory function of the choroid plexus is mediated by specific transport systems that allow the directional flux of nutrients and ions into the CSF and the removal of toxins. Normal CSF dynamics and chemistry ensure that the environment for neural function is optimal. Here, we report that targeted disruption of the Slc4a5 gene encoding the electrogenic sodium bicarbonate cotransporter NBCe2 results in significant remodeling of choroid plexus epithelial cells, including abnormal mitochondrial distribution, cytoskeletal protein expression, and ion transporter polarity. These changes are accompanied by very significant abnormalities in intracerebral ventricle volume, intracranial pressure, and CSF electrolyte levels. The Slc4a5(-/-) mice are significantly more resistant to induction of seizure behavior than wild-type controls. In the retina of Slc4a5(-/-) mice, loss of photoreceptors, ganglion cells, and retinal detachment results in visual impairment assessed by abnormal electroretinogram waveforms. Our findings are the first demonstration of the fundamental importance of NBCe2 in the biology of the nervous system.


Asunto(s)
Plexo Coroideo/metabolismo , Mitocondrias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Simportadores de Sodio-Bicarbonato/metabolismo , Equilibrio Hidroelectrolítico , Animales , Plexo Coroideo/patología , Presión Intracraneal/genética , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/patología , Proteínas del Tejido Nervioso/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Desprendimiento de Retina/líquido cefalorraquídeo , Desprendimiento de Retina/genética , Simportadores de Sodio-Bicarbonato/genética
6.
J Neuroophthalmol ; 32(3): 227-37, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22635166

RESUMEN

OBJECTIVES: During mouse retina maturation, the final number of retinal ganglion cells (RGCs) is determined by highly regulated programmed cell death. Previous studies demonstrated that the immunoregulatory receptor programmed cell death-1 (PD-1) promotes developmental RGC death. To identify the functional signaling partner(s) for PD-1, we identified retinal expression of PD-1 ligands and examined the effect of PD-1 ligand expression on RGC number. We also explored the hypothesis that PD-1 signaling promotes the development of functional visual circuitry. METHODS: Characterization of retinal and brain programmed cell death-1 ligand 1 (PD-L1) expression were examined by immunofluorescence on tissue sections. The contribution of PD-ligands, PD-L1, and programmed cell death-1 ligand 2 (PD-L2) to RGC number was examined in PD-ligand knockout mice lacking 1 or both ligands. Retinal architecture was assessed by spectral-domain optical coherence tomography, and retinal function was analyzed by electroretinography in wild-type and PD-L1/L2 double-deficient mice. RESULTS: PD-L1 expression is found throughout the neonatal retina and persists in adult RGCs, bipolar interneurons, and Müller glia. In the absence of both PD-ligands, there is a significant numerical increase in RGCs (34% at postnatal day 2 [P2] and 18% in adult), as compared to wild type, and PD-ligands have redundant function in this process. Despite the increased RGC number, adult PD-L1/L2 double-knockout mice have normal retinal architecture and outer retina function. CONCLUSION: This study demonstrates that PD-L1 and PD-L2 together impact the final number of RGCs in adult mice and supports a novel role for active promotion of neuronal cell death through PD-1 receptor-ligand engagement.


Asunto(s)
Envejecimiento , Antígeno B7-H1/metabolismo , Retina/citología , Células Ganglionares de la Retina/metabolismo , Animales , Axones/metabolismo , Antígeno B7-H1/deficiencia , Electrorretinografía , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nervio Óptico/metabolismo , Proteína 2 Ligando de Muerte Celular Programada 1/deficiencia , Receptor de Muerte Celular Programada 1/deficiencia , Análisis Espectral
7.
Cells ; 11(21)2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36359797

RESUMEN

Rbfox1 is a multifunctional RNA binding protein that regulates various aspects of RNA metabolism important for neuronal differentiation and normal physiology. Rbfox1 has been associated with neurodevelopmental and neurological conditions as well as age-related neurodegenerative diseases such as Alzheimer's and Parkinson's. We have shown that in mammalian retinas Rbfox1 is expressed in retinal ganglion cells (RGCs) and in amacrine cells (ACs). This study investigates the effect of advanced age (22-month-old mice) on visual function, retinal morphology and survival of injured retinal ganglion cells (RGC) in Rbfox1 knockout (KO) animals. A visual cliff test, which was used to evaluate visual function, showed that 22-month old Rbfox1 KO mice have profound depth perception deficiency. Retinal gross morphology in these animals appeared to be normal. Optic nerve crush (ONC) induced axonal injury resulted in approximately 50% of RGC loss in both Rbfox1 KO and age-matched control animals: the average RGC densities in uninjured control and Rbfox1 KO animals were 6274 ± 1673 cells/mm2 and 6004 ± 1531 cells/mm2, respectively, whereas 1 week after ONC, RGC numbers in the retinas of control and Rbfox1 KO mice were reduced to 2998 ± 858 cells/mm2 and 3036 ± 857 cells/mm2, respectively (Rbfox1 KO vs. Rbfox1 KO + ONC, p < 0.0001 and control vs. control + ONC, p < 0.0001). No significant difference between RGC numbers in Rbfox1 KO + ONC and age-matched control + ONC animals was observed, suggesting that Rbfox1 has no effect on the survival of injured RGCs. Interestingly, however, contrary to a commonly accepted view that the number of RGCs in old (18 month of age) compared to young animals is reduced by approximately 40%, the RGC densities in 22-month-old mice in this study were similar to those of 4-month-old counterparts.


Asunto(s)
Traumatismos del Nervio Óptico , Células Ganglionares de la Retina , Animales , Ratones , Modelos Animales de Enfermedad , Mamíferos , Ratones Noqueados , Compresión Nerviosa , Traumatismos del Nervio Óptico/genética , Células Ganglionares de la Retina/metabolismo , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo
8.
Adv Exp Med Biol ; 664: 355-63, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20238036

RESUMEN

Oxidative damage has been implicated in retinal ganglion cell (RGC) death after optic nerve transection (ONT) and during glaucomatous neuropathy. Here, we analyzed the expression and cell protective role of thioredoxins (TRX), key regulators of the cellular redox state, in RGCs damaged by pharmacologically induced oxidative stress, ONT and elevated intraocular pressure (IOP). The endogenous level of thioredoxin-1 (TRX1) and thioredoxin-2 (TRX2) in RGCs after axotomy and in RGC-5 cells after glutamate/buthionine sulfoximine (BSO) treatment showed upregulation of TRX2, whereas no significant change was observed in TRX1 expression. The increased level TRX-interacting protein (TXNIP) in the retinas was observed 2 and 5 weeks after IOP elevation. TRX1 level was decreased at 2 weeks and more prominently at 5 weeks after IOP increase. No change in TRX2 levels in response to IOP change was observed. Overexpression of TRX1 and TRX2 in RGC-5 treated with glutamate/BSO increased the cell survival by 2- and 3-fold 24 and 48 h after treatment, respectively. Overexpression of these proteins in the retina increased the survival of RGCs by 35 and 135% 7 and 14 days after ONT, respectively. In hypertensive eyes, RGC loss was approximately 27% 5 weeks after IOP elevation compared to control. TRX1 and TRX2 overexpression preserved approximately 45 and 37% of RGCs, respectively, that were destined to die due to IOP increase.


Asunto(s)
Citoprotección , Hipertensión Ocular/complicaciones , Traumatismos del Nervio Óptico/complicaciones , Estrés Oxidativo , Células Ganglionares de la Retina/patología , Tiorredoxinas/metabolismo , Animales , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Supervivencia Celular , Presión Intraocular/fisiología , Masculino , Hipertensión Ocular/fisiopatología , Hipertensión Ocular/prevención & control , Traumatismos del Nervio Óptico/fisiopatología , Traumatismos del Nervio Óptico/prevención & control , Ratas , Ratas Wistar
9.
Data Brief ; 24: 103950, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31193141

RESUMEN

In adult mammals, retinal ganglion cells (RGCs) fail to regenerate following damage. As a result, RGCs die after acute injury and in progressive degenerative diseases such as glaucoma; this can lead to permanent vision loss and, eventually, blindness. Lipids are crucial for the development and maintenance of cell membranes, myelin sheaths, and cellular signaling pathways, however, little is known about their role in axon injury and repair. Studies examining changes to the lipidome during optic nerve (ON) regeneration could greatly inform treatment strategies, yet these are largely lacking. Experimental animal models of ON regeneration have facilitated the exploration of the molecular determinants that affect RGC axon regeneration. Here, we analyzed lipid profiles of the ON and retina in an ON crush rat model using liquid chromatography-mass spectrometry. Furthermore, we investigated lipidome changes after ON crush followed by intravitreal treatment with Zymosan, a yeast cell wall derivative known to enhance RGC regeneration. This data is available at the NIH Common Fund's Metabolomics Data Repository and Coordinating Center (supported by NIH grant, U01-DK097430) website, the Metabolomics Workbench, http://www.metabolomicsworkbench.org, where it has been assigned Project ID: PR000661. The data can be accessed directly via it's Project DOI: doi: 10.21,228/M87D53.

10.
J Neurosci Res ; 86(13): 2943-51, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18521932

RESUMEN

Autophagy has been shown to be activated in neuronal cells in response to injury and suggested to have a cell-protective role in neurodegenerative diseases. In this study, we investigated the activation of autophagy in retinal ganglion cells (RGCs) following optic nerve transection (ONT) and evaluated its effect on RGC survival. Expression of several autophagy-related genes, including Atg5, Atg7, and Atg12, and autophagy markers microtubule-associated protein 1 light chain 3-II (LC3-II) and beclin-1 were analyzed at the transcriptional or protein level 1, 3, and 7 days after ONT. Transcription of the Atg5, Atg7, and Atg12 genes was up-regulated 1.5- to 1.8-fold in the retina 3 days after ONT compared with that in the controls. Expression of Atg12 mRNA was increased 1.6-fold 1 day after ONT. Seven days after ONT, expression of Atg5, Atg7, and Atg12 mRNA was comparable to that in the untreated retinas. Western blot analysis of proteins isolated from RGCs showed 1.6-, 2.7-, and 1.7-fold increases in LC3-II level 1, 3, and 7 days after ONT, respectively, compared with those in the controls. Expression of beclin-1 was 1.7-fold higher 1 day after RGCs were axotomized, but 3 and 7 days after ONT it was comparable to that of the control. Inhibition of autophagy with bafilomycin A1, 3-methyladenine, and Wortmannin in RGC-5 cells under serum-deprived conditions decreased cell viability by approximately 40%. These results suggest possible activation of autophagy in RGCs after optic nerve transection and demonstrate its protective role in RGC-5 cells maintained under conditions of serum deprivation.


Asunto(s)
Autofagia/fisiología , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/fisiología , Animales , Axotomía , Western Blotting , Expresión Génica , Inmunohistoquímica , Masculino , Nervio Óptico/fisiología , ARN Mensajero/análisis , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Neurosci Lett ; 433(1): 11-6, 2008 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-18242848

RESUMEN

The heat shock response is a genetically well-ordered process for cell to generate heat shock protein (HSP). Various stressors can trigger the response through heat shock transcriptional factor (HSF) regulation. Recent studies demonstrated that preconditioning of N-methyl-d-aspartate (NMDA) at non-lethal levels has neuroprotective effects, but the exact mechanisms are unclear. We hypothesize that the protective mechanisms of NMDA preconditioning could involve HSP expression. To understand the regulatory mechanisms of HSP under stress, we examined the expression of Hsp72, HSF1 and HSF2 in the adult rat retina after intravitreal injection of NMDA. Retinal ganglion cell (RGC) counting with retrograde labeling showed that 8 nmol, but not 0.8 nmol, of intravitreal NMDA reduced RGC survival. Western blotting and immunohistochemistry showed that non-lethal (0.8 nmol) doses of NMDA induced a time-dependent expression of HSF1 and HSF2, and that the expression of HSF1 and HSF2 in the RGC layer peaked between 9 and 18 h after injection. Parallel to the increased HSF expression, immunohistochemistry and in situ hybridization demonstrated that Hsp72 mRNA and protein expression increased 9 and 12 h after non-lethal NMDA injection, respectively. Our findings suggest that the expression of HSF1 and HSF2 is associated with the Hsp72-related stress response.


Asunto(s)
Citoprotección/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas del Choque Térmico HSP72/metabolismo , Respuesta al Choque Térmico/fisiología , Retina/metabolismo , Estrés Fisiológico/metabolismo , Factores de Transcripción/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Citoprotección/efectos de los fármacos , Proteínas de Unión al ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Agonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Proteínas del Choque Térmico HSP72/efectos de los fármacos , Factores de Transcripción del Choque Térmico , Proteínas de Choque Térmico/efectos de los fármacos , Proteínas de Choque Térmico/metabolismo , Respuesta al Choque Térmico/efectos de los fármacos , Inmunohistoquímica , Masculino , N-Metilaspartato/farmacología , Fármacos Neuroprotectores/farmacología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Retina/efectos de los fármacos , Retina/fisiopatología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Estrés Fisiológico/inducido químicamente , Estrés Fisiológico/fisiopatología , Factores de Tiempo , Factores de Transcripción/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Cuerpo Vítreo/efectos de los fármacos
12.
Neurosci Lett ; 670: 89-93, 2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29398521

RESUMEN

Celastrol, a quinine methide triterpene extracted from the perennial vine Tripterygium wilfordii, has been identified as a neuroprotective agent in various models of neurodegenerative disorders. We have reported earlier that systemic and intravitreal administration of celastrol stimulate the survival of retinal ganglion cells (RGCs) injured by optic nerve crush (ONC) and that mechanisms underlying celastrol׳s RGC protection may be associated with inhibition of TNF-alpha-mediated cell death. The present study evaluates the effect of celastrol on the survival of RGCs injured by ocular hypertension. Intraocular pressure (IOP) elevation resulted in approximately 23% of RGCs loss. Reduction in RGC numbers was observed in all four retinal quadrants: 30% in superior, 17% in inferior, 11% in nasal and 35% in temporal regions. Celastrol (1 mg/kg) or vehicle (DMSO) was administered three times per week by intraperitoneal injection, starting on the day of laser photocoagulation of the TM and continued for the entire duration of the experiment (5 weeks). Celastrol treatment stimulated RGC survival by an average of 24% in the entire retina compared to the vehicle-treated group. RGC numbers were increased in all four quadrants: approximately 40%, 17%, 15% and 30% more RGCs were counted in the superior, inferior, nasal and temporal regions, respectively. The average RGC numbers for the entire retinas of the celastrol/IOP group were only ∼5% and 10% lower than that in vehicle- or celastrol-injected animals with normal IOP, respectively. Our data indicate a significant celastrol-mediated neuroprotection against elevated IOP-induced injury.


Asunto(s)
Degeneración Nerviosa/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Hipertensión Ocular/complicaciones , Células Ganglionares de la Retina/efectos de los fármacos , Triterpenos/uso terapéutico , Animales , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Degeneración Nerviosa/etiología , Degeneración Nerviosa/patología , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Hipertensión Ocular/patología , Triterpenos Pentacíclicos , Ratas , Células Ganglionares de la Retina/patología , Resultado del Tratamiento , Triterpenos/farmacología
13.
Invest Ophthalmol Vis Sci ; 59(1): 212-222, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29340649

RESUMEN

Purpose: Mammalian central nervous system axons fail to regenerate after injury. Contributing factors include limited intrinsic growth capacity and an inhibitory glial environment. Inflammation-induced optic nerve regeneration (IIR) is thought to boost retinal ganglion cell (RGC) intrinsic growth capacity through progrowth gene expression, but effects on the inhibitory glial environment of the optic nerve are unexplored. To investigate progrowth molecular changes associated with reactive gliosis during IIR, we developed an imaging mass spectrometry (IMS)-based approach that identifies discriminant molecular signals in and around optic nerve crush (ONC) sites. Methods: ONC was performed in rats, and IIR was established by intravitreal injection of a yeast cell wall preparation. Optic nerves were collected at various postcrush intervals, and longitudinal sections were analyzed with matrix-assisted laser desorption/ionization (MALDI) IMS and data mining. Immunohistochemistry and confocal microscopy were used to compare discriminant molecular features with cellular features of reactive gliosis. Results: IIR increased the area of the crush site that was occupied by a dense cellular infiltrate and mass spectral features consistent with lysosome-specific lipids. IIR also increased immunohistochemical labeling for microglia and macrophages. IIR enhanced clearance of lipid sulfatide myelin-associated inhibitors of axon growth and accumulation of simple GM3 gangliosides in a spatial distribution consistent with degradation of plasma membrane from degenerated axons. Conclusions: IIR promotes a robust phagocytic response that improves clearance of myelin and axon debris. This growth-permissive molecular remodeling of the crush injury site extends our current understanding of IIR to include mechanisms extrinsic to the RGC.


Asunto(s)
Compresión Nerviosa , Regeneración Nerviosa/fisiología , Plasticidad Neuronal/fisiología , Traumatismos del Nervio Óptico/fisiopatología , Nervio Óptico/fisiología , Animales , Axones , Recuento de Células , Supervivencia Celular , Modelos Animales de Enfermedad , Gliosis , Metabolismo de los Lípidos/fisiología , Masculino , Microscopía Confocal , Traumatismos del Nervio Óptico/metabolismo , Ratas , Ratas Endogámicas F344 , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
14.
Brain Res ; 1141: 1-9, 2007 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-17316577

RESUMEN

The expression of alpha (alphaA and alphaB) and beta (betaA1/A3, betaA2, betaA4, and betaB2) crystallin genes were analyzed at the mRNA and protein levels in rat retinas with ocular hypertension-induced ganglion cell death. An animal model with progressive loss of retinal ganglion cells (RGC) was generated by elevation of intraocular pressure (IOP). The estimated RGC loss was approximately 8% and 20% at 2 and 5 weeks post IOP elevation, respectively. mRNA and protein quantification showed that alpha and beta crystallin genes were downregulated at both transcriptional (alphaA, alphaB, betaA1/A3, betaA4, and betaB2 approximately 50% and betaA2~40%) and protein (alphaA~50%, alphaB~63%, betaA1/A3~70%, and betaB2~38%) levels 2 weeks after IOP elevation. In experimental retinas 5 weeks after IOP elevation, the levels of crystallin mRNAs were higher than at 2 weeks and were comparable to that of control retinas. However, the levels of the corresponding proteins were still lower (alphaA, alphaB, and betaB2 approximately 37% and betaA1/A3~70%) than in control retinas. Furthermore, we found that the expression of these genes in the retina is predominantly localized to the cells in the GCL and to a lesser degree in the INL and ONL. Colocalization of the crystallin-positive and Fluorogold retrogradely labeled cells indicated that the cells expressing alpha and beta crystallins in the GCL are RGCs. In summary, we showed that alpha and beta crystallins are expressed in the retina predominantly by RGCs and that their expression is affected by ocular hypertension.


Asunto(s)
Cristalinas/metabolismo , Regulación de la Expresión Génica/fisiología , Degeneración Nerviosa/patología , Retina/patología , Células Ganglionares de la Retina/metabolismo , Análisis de Varianza , Animales , Recuento de Células/métodos , Cristalinas/clasificación , Cristalinas/genética , Hibridación in Situ/métodos , Masculino , Degeneración Nerviosa/etiología , Degeneración Nerviosa/metabolismo , Hipertensión Ocular/complicaciones , Hipertensión Ocular/patología , Ratas , Ratas Wistar , Retina/metabolismo , Estilbamidinas/metabolismo , Factores de Tiempo
15.
Invest Ophthalmol Vis Sci ; 47(4): 1448-57, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16565378

RESUMEN

PURPOSE: To characterize a model of optic nerve axonal degeneration induced by tumor necrosis factor (TNF)-alpha and to determine the role of nuclear factor (NF)-kappaB p65 in axonal degeneration. METHODS: Groups of rats were euthanatized at 1 day, 1 or 2 weeks, or 1 or 2 months after intravitreal injection of TNF-alpha. Morphometric analyses of neurofilament- or Thy-1-positive cells, retinal ganglion cells (flat preparations stained with cresyl violet or retrograde labeling with a neurotracer), the number of axons, immunostaining for myelin basic protein, and TUNEL assays were performed. Levels of NF-kappaB p65 protein in retina and optic nerve were determined by Western blot analysis and immunohistochemistry. The effects of antisense oligodeoxynucleotide (AS ODN) against NF-kappaB p65 and helenalin, an inhibitor of NF-kappaB p65 activation, on TNF-alpha-induced optic nerve degeneration were determined by counting the number of axons. RESULTS: Intravitreal injections of TNF-alpha induced obvious axonal loss and extensive degeneration of the axons from 2 weeks to 2 months after injection, whereas significant retinal ganglion cell loss was noted only at 2 months after injection. NF-kappaB p65 was increased in the optic nerve but not in the retina and was found to colocalize with ED-1 and Iba1, markers of microglia. Inhibition of NF-kappaB p65 with AS ODN or helenalin significantly ameliorated the effects of TNF-alpha-mediated axonal loss. CONCLUSIONS: TNF-alpha causes axonal degeneration with probable delayed loss of retinal ganglion cell bodies. NF-kappaB p65 may play a pivotal role in axonal degeneration, with the possible involvement of microglial cells.


Asunto(s)
Degeneración Nerviosa/inducido químicamente , Nervio Óptico/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/toxicidad , Animales , Axones/metabolismo , Western Blotting , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Inyecciones , Masculino , Proteína Básica de Mielina/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Oligodesoxirribonucleótidos Antisentido/farmacología , Nervio Óptico/metabolismo , Nervio Óptico/patología , Ratas , Ratas Endogámicas Lew , Retina/metabolismo , Sesquiterpenos/farmacología , Sesquiterpenos de Guayano , Cuerpo Vítreo
16.
Mol Vis ; 12: 1660-73, 2006 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-17200666

RESUMEN

PURPOSE: To obtain and analyze the gene expression profiles of the retina following optic nerve transection (ONT). METHODS: An axotomy animal model was generated by taking a cross section of the optic nerve with care not to damage the adjacent blood supply. The extent of cell loss was evaluated by counting cells in the ganglion cell layer (GCL) of flat-mounted retinas stained with cresyl violet. Gene expression profiles of control and ONT retinas were analyzed with Agilent's rat oligo microarray slides. Differentially expressed genes were identified from three independent experiments and clustered based on their functions with expression analysis systematic explorer software. Real-time quantitative and semiquantitative RT-PCR were used to validate changes in gene expression levels. RESULTS: Gene expression changes in axotomized retinas were analyzed with rat oligo microarray slides containing 22,575 oligonucleotide probes that represent over 20,000 genes and expressed sequence tags (ESTs). The expression of 493 genes was increased more than 1.5 fold, including 85 that were upregulated more than 2 fold, and the expression of 113 genes was decreased 1.5 fold or more, including 21 that were downregulated more than 2 fold. Differentially expressed genes were clustered based on their functions. Several novel genes expressed in the GCL have been identified and their expression patterns in different tissues were analyzed. Among the genes differentially regulated in retinas with induced retinal ganglion cell (RGC) death, we have identified 13 genes that are mapped to known glaucoma loci and can be considered for mutation screening in patients with inherited forms of glaucoma. CONCLUSIONS: The gene expression profiles of the ONT retinas can be used to identify RGC-expressed genes, which may be essential for the morphological and physiological characteristics of RGCs. The results of this study can also be used to evaluate the molecular changes taking place in the retina in response to RGC loss.


Asunto(s)
Expresión Génica , Traumatismos del Nervio Óptico/genética , Retina/metabolismo , Animales , Axotomía , Mapeo Cromosómico , Análisis Mutacional de ADN/métodos , Perfilación de la Expresión Génica , Glaucoma/genética , Hibridación in Situ , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Wistar , Células Ganglionares de la Retina/metabolismo
17.
Brain Res ; 1081(1): 211-8, 2006 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-16530172

RESUMEN

We previously demonstrated that endogenous interleukin-6 (IL-6) is upregulated and may be neuroprotective after retinal ischemia. The purpose of this study is to investigate the role of nuclear factor kappa-B (NF-kappaB) in regulating IL-6 expression after ischemia. NF-kappaB p65 mRNA levels were significantly elevated between 2 and 12 h after the insult. A high number of NF-kappaB p65 positive cells were detected in the inner retina at 12 h after ischemia. Activated nuclear NF-kappaB p65 and IL-6 were colocalized in cells, which were also marked by a microglial/phagocytic cell marker (ED1) in the inner retina. Carbobenzoxy-L-leucyl-L-leucyl-L-leucinal (MG-132, a proteasome inhibitor, which inhibits IkappaB degradation and hence prevents the activation and translocation of NF-kappaB into the nucleus) abolished the increase in NF-kappaB p65 mRNA levels after the insult, while there was no effect by helenalin (an inhibitor which inhibits NF-kappaB activity by alkylation of the p65 subunit, thereby blocking its binding to the target DNA). However, MG-132 and/or helenalin significantly diminished the increase in IL-6 mRNA levels after the insult. Small interfering RNAs (siRNAs, inhibit target gene expression through the sequence-specific destruction of the target messenger RNA) against NF-kappaB p65 significantly reduced the increase in NF-kappaB p65 mRNA levels as well as IL-6 mRNA levels after ischemia. The number of retinal ganglion cells (RGCs) was also significantly decreased using the inhibitors of NF-kappaB compared with those of the controls after ischemia. These findings support the hypothesis that upregulation of endogenous retinal IL-6 in retinal I/R injury in microglial/phagocytic cells is controlled predominantly by NF-kappaB p65.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Interleucina-6/metabolismo , FN-kappa B/metabolismo , Daño por Reperfusión/metabolismo , Retina/metabolismo , Retina/fisiopatología , Análisis de Varianza , Animales , Antiinflamatorios no Esteroideos/farmacología , Recuento de Células/métodos , Inhibidores de Cisteína Proteinasa/farmacología , Interacciones Farmacológicas , Regulación de la Expresión Génica/efectos de los fármacos , Inmunohistoquímica/métodos , Interleucina-6/genética , Leupeptinas/farmacología , Masculino , FN-kappa B/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Ratas Endogámicas Lew , Daño por Reperfusión/fisiopatología , Retina/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Sesquiterpenos/farmacología , Sesquiterpenos de Guayano , Factores de Tiempo
18.
Neurosci Lett ; 405(1-2): 40-5, 2006 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-16870336

RESUMEN

The RNA binding protein with multiple splicing 2, or hermes, is a member of the RRM (RNA recognition motif) family of RNA-binding proteins. In this study, we show that the hermes gene is expressed in the rat retina, and its expression is restricted to the ganglion cell layer. Double in situ hybridization with riboprobes corresponding to the hermes gene and Thy-1, the RGC marker in the retina, showed that the majority of the Thy-1 positive cells in the ganglion cell layer were also hermes positive. This was also shown by co-localization of the hermes in situ hybridization signals with the retrogradely labeled RGCs. Our observations suggest that hermes is expressed in the majority, if not all, of RGCs and is not restricted to only certain RGC types. Hermes in situ hybridization signals were not detected in the retinal sections of optic nerve transected animals, which are characterized by rapid and specific RGC degeneration. The dramatic reduction of the hermes mRNA level in axotomized retinas was also observed by semi-quantitative RT-PCR. The specific expression of hermes in retinal ganglion cells qualifies this gene as a potential RGC marker in the retina. Outside the retina, hermes is expressed in the heart, liver, and kidney, and to a lesser degree in the cerebellum, cortex, lung, and small intestine.


Asunto(s)
Proteínas de Unión al ARN/biosíntesis , Células Ganglionares de la Retina/metabolismo , Animales , Axotomía , Inmunohistoquímica , Hibridación in Situ , Masculino , Traumatismos del Nervio Óptico/patología , Especificidad de Órganos , Ratas , Ratas Wistar , Células Ganglionares de la Retina/patología
19.
Neurosci Lett ; 405(3): 191-5, 2006 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-16889897

RESUMEN

Heat shock protein (HSP) plays an important role in the maintenance of neuronal survival during harmful conditions. Previously, we reported that metabolic stress induces HSP72 in retinal ganglion cells (RGCs) and protects against excitotoxicity, hypoxia and experimental glaucoma. To understand heat shock protein transcriptional mechanisms, we examined the cellular expression of heat shock factors 1 (HSF1) and 2 (HSF2) in the unstressed adult rat retina. Western blotting, immunohistochemistry and RT-PCR showed that mRNA and protein of HSF1 and HSF2 were present in the rat retina and predominantly expressed in RGC layer cells. Western blotting of dissociated RGC suspensions harvested with Thy-1 immuno-labeled magnetic beads confirmed that RGCs expressed HSF1, HSF2 and HSP72. Our findings suggest that both heat shock transcription factors 1 and 2 are linked to the heat shock response in retinal ganglion cells.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Expresión Génica/fisiología , Células Ganglionares de la Retina/metabolismo , Factores de Transcripción/metabolismo , Animales , Northern Blotting/métodos , Western Blotting/métodos , Proteínas de Unión al ADN/genética , Técnica del Anticuerpo Fluorescente/métodos , Factores de Transcripción del Choque Térmico , Proteínas de Choque Térmico/metabolismo , Masculino , Transporte de Proteínas , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores de Transcripción/genética
20.
Prog Retin Eye Res ; 52: 22-46, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27017896

RESUMEN

Heat shock proteins (HSPs) belong to a superfamily of stress proteins that are critical constituents of a complex defense mechanism that enhances cell survival under adverse environmental conditions. Cell protective roles of HSPs are related to their chaperone functions, antiapoptotic and antinecrotic effects. HSPs' anti-apoptotic and cytoprotective characteristics, their ability to protect cells from a variety of stressful stimuli, and the possibility of their pharmacological induction in cells under pathological stress make these proteins an attractive therapeutic target for various neurodegenerative diseases; these include Alzheimer's, Parkinson's, Huntington's, prion disease, and others. This review discusses the possible roles of HSPs, particularly HSP70 and small HSPs (alpha A and alpha B crystallins) in enhancing the survival of retinal ganglion cells (RGCs) in optic neuropathies such as glaucoma, which is characterized by progressive loss of vision caused by degeneration of RGCs and their axons in the optic nerve. Studies in animal models of RGC degeneration induced by ocular hypertension, optic nerve crush and axotomy show that upregulation of HSP70 expression by hyperthermia, zinc, geranyl-geranyl acetone, 17-AAG (a HSP90 inhibitor), or through transfection of retinal cells with AAV2-HSP70 effectively supports the survival of injured RGCs. RGCs survival was also stimulated by overexpression of alpha A and alpha B crystallins. These findings provide support for translating the HSP70- and alpha crystallin-based cell survival strategy into therapy to protect and rescue injured RGCs from degeneration associated with glaucomatous and other optic neuropathies.


Asunto(s)
Proteínas del Choque Térmico HSP72/metabolismo , Células Ganglionares de la Retina/fisiología , alfa-Cristalinas/metabolismo , Animales , Supervivencia Celular/fisiología , Humanos , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA