Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Cell Dev Biol ; 11: 1045759, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37351276

RESUMEN

Sigma 1 Receptor (S1R) is a therapeutic target for a wide spectrum of pathological conditions ranging from neurodegenerative diseases to cancer and COVID-19. S1R is ubiquitously expressed throughout the visceral organs, nervous, immune and cardiovascular systems. It is proposed to function as a ligand-dependent molecular chaperone that modulates multiple intracellular signaling pathways. The purpose of this study was to define the S1R proximatome under native conditions and upon binding to well-characterized ligands. This was accomplished by fusing the biotin ligase, Apex2, to the C terminus of S1R. Cells stably expressing S1R-Apex or a GFP-Apex control were used to map proximal proteins. Biotinylated proteins were labeled under native conditions and in a ligand dependent manner, then purified and identified using quantitative mass spectrometry. Under native conditions, S1R biotinylates over 200 novel proteins, many of which localize within the endomembrane system (endoplasmic reticulum, Golgi, secretory vesicles) and function within the secretory pathway. Under conditions of cellular exposure to either S1R agonist or antagonist, results show enrichment of proteins integral to secretion, extracellular matrix formation, and cholesterol biosynthesis. Notably, Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) displays increased binding to S1R under conditions of treatment with Haloperidol, a well-known S1R antagonist; whereas Low density lipoprotein receptor (LDLR) binds more efficiently to S1R upon treatment with (+)-Pentazocine ((+)-PTZ), a classical S1R agonist. Furthermore, we demonstrate that the ligand bound state of S1R correlates with specific changes to the cellular secretome. Our results are consistent with the postulated role of S1R as an intracellular chaperone and further suggest important and novel functionalities related to secretion and cholesterol metabolism.

2.
Exp Eye Res ; 226: 109308, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36400283

RESUMEN

PURPOSE: Glaucoma is a worldwide leading cause of irreversible blindness. Standard treatments lower intraocular pressure (IOP). Novel treatments to prevent optic nerve (ON) degeneration are needed. Here, we investigate the hypothesis that sigma-1 receptor (S1R) agonist (+)-pentazocine (PTZ) is neuroprotective in a Brown Norway (BN) rat, microbead model of glaucoma. METHODS: BN rats (9-11 weeks, male and female) were treated by intraperitoneal injection, 3 times per week with (+)-PTZ (2 mg/kg) or vehicle (VEH) alone. Treatment started 1 week prior to intraocular injection of polystyrene microbeads to elevate IOP. IOP was measured 2-3 times per week. Five weeks post microbead injection, rats were euthanized. ONs were removed, then fixed and processed for 63x oil, light microscope imaging of toluidine blue stained ON cross sections. To facilitate comparison of ON morphology from VEH and (+)-PTZ treated rats with similar ocular hypertensive insults, rats were assigned to low (IOP ≤15.8 mmHg), moderate (15.8 < IOP <28.0 mmHg), and high (IOP ≥28.0 mmHg) groups based on average IOP in the microbead injected eye. Axon numbers, axon density, axonal and glial areas, axon loss, and axon size distributions of naïve, bead, and contralateral ONs were assessed using QuPath program for automated image analysis. RESULTS: (+)-PTZ treatment of BN rats protected ONs from damage caused by moderate IOP elevation. Treatment with (+)-PTZ significantly reduced axon loss and glial areas, and increased axon density and axonal areas compared to ONs from VEH treated rats with moderate IOP. (+)-PTZ-mediated neuroprotection was independent of IOP lowering effects. At average IOP ≥28.0 mmHg, (+)-PTZ treatment did not provide measurable neuroprotection. ONs from contralateral eyes exhibited subtle, complex changes in response to conditions in the bead eyes. CONCLUSIONS: S1R agonist (+)-PTZ shows promise as a neuroprotective treatment for glaucoma. Future studies to understand the complex molecular mechanisms by which (+)-PTZ provides this neuroprotection are needed.


Asunto(s)
Glaucoma , Pentazocina , Ratas , Masculino , Femenino , Animales , Ratas Endogámicas BN , Microesferas , Pentazocina/farmacología , Pentazocina/uso terapéutico , Neuroprotección , Células Ganglionares de la Retina , Presión Intraocular , Inyecciones Intraoculares/efectos adversos , Modelos Animales de Enfermedad , Receptor Sigma-1
3.
Invest Ophthalmol Vis Sci ; 63(2): 1, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35103752

RESUMEN

Purpose: Sigma 1 receptor (S1R) is expressed in retinal ganglion cells (RGCs) and astrocytes, and its activation is neuroprotective. We evaluated the contribution of S1R within optic nerve head astrocytes (ONHAs) to growth and survival of RGCs in vitro. Methods: Wild-type (WT) RGCs and WT or S1R knockout (S1R KO) ONHAs were cocultured for 2, 4, or 7 days. Total and maximal neurite length, neurite root, and extremity counts were measured. Cell death was measured using a TUNEL assay. Signal transducer and activator of transcription 3 phosphorylation levels were evaluated in ONHA-derived lysates by immunoblotting. Results: The coculture of WT RGCs with WT or S1R KO ONHAs increased the total and maximal neurite length. Neurite root and extremity counts increased at 4 and 7 days when WT RGCs were cocultured with WT or S1R KO ONHAs. At all timepoints, the total and maximal neurite length decreased for WT RGCs in coculture with S1R KO ONHAs compared with WT ONHAs. Root and extremity counts decreased for WT RGCs in coculture with S1R KO ONHAs compared with WT ONHAs at 2 and 7, but not 4 days. RGC apoptosis increased in S1R KO ONHA coculture and S1R KO-conditioned medium, compared with WT ONHA coculture or WT-conditioned medium. S1R KO ONHA-derived lysates showed decreased phosphorylated signal transducer and activator of transcription 3 levels compared with WT ONHA-derived lysates. Conclusions: The absence of S1R within ONHAs has a deleterious effect on RGC neurite growth and RGC survival, reflected in analysis of WT RGC + S1R KO ONHA indirect cocultures. The data suggest that S1R may enhance ganglion cell survival via glia-mediated mechanisms.


Asunto(s)
Apoptosis , Astrocitos/metabolismo , Neuroprotección/fisiología , Estrés Oxidativo , Receptores sigma/metabolismo , Enfermedades de la Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Astrocitos/patología , Western Blotting , Muerte Celular , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Disco Óptico/metabolismo , Disco Óptico/patología , Enfermedades de la Retina/patología , Células Ganglionares de la Retina/patología , Receptor Sigma-1
4.
Invest Ophthalmol Vis Sci ; 62(7): 5, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34086045

RESUMEN

Purpose: Stimulation of Sigma 1 Receptor (S1R) is neuroprotective in retina and optic nerve. S1R is expressed in both neurons and glia. The purpose of this work is to evaluate the ability of S1R to modulate reactivity responses of optic nerve head astrocytes (ONHAs) by investigating the extent to which S1R activation alters ONHA reactivity under conditions of ischemic cellular stress. Methods: Wild type (WT) and S1R knockout (KO) ONHAs were derived and treated with vehicle or S1R agonist, (+)-pentazocine ((+)-PTZ). Cells were subjected to six hours of oxygen glucose deprivation (OGD) followed by 18 hours of re-oxygenation (OGD/R). Astrocyte reactivity responses were measured. Molecules that regulate ONHA reactivity, signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappa B (NF-kB), were evaluated. Results: Baseline glial fibrillary acidic protein (GFAP) levels were increased in nonstressed KO ONHAs compared with WT cultures. Baseline cellular migration was also increased in nonstressed KO ONHAs compared with WT. Treatment with (+)-PTZ increased cellular migration in nonstressed WT ONHAs but not in KO ONHAs. Exposure of both WT and KO ONHAs to ischemia (OGD/R), increased GFAP levels and cellular proliferation. However, (+)-PTZ treatment of OGD/R-exposed ONHAs enhanced GFAP levels, cellular proliferation, and cellular migration in WT but not KO cultures. The (+)-PTZ treatment of WT ONHAs also enhanced the OGD/R-induced increase in cellular pSTAT3 levels. However, treatment of WT ONHAs with (+)-PTZ abrogated the OGD/R-induced rise in NF-kB(p65) activation. Conclusions: Under ischemic stress conditions, S1R activation enhanced ONHA reactivity characteristics. Future studies should address effects of these responses on RGC survival.


Asunto(s)
Astrocitos/metabolismo , Disco Óptico , Receptores sigma , Células Ganglionares de la Retina/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Noqueados , Neuroprotección/fisiología , Fármacos Neuroprotectores/farmacología , Disco Óptico/metabolismo , Disco Óptico/patología , Neuropatía Óptica Isquémica/metabolismo , Pentazocina/farmacología , Receptores sigma/agonistas , Receptores sigma/metabolismo , Resultado del Tratamiento , Receptor Sigma-1
5.
Transl Vis Sci Technol ; 9(3): 22, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32714648

RESUMEN

Purpose: A novel application of QuPath open-source digital analysis software is used to provide in-depth morphological analysis of progressive optic nerve (ON) degeneration in rats. Methods: QuPath software was adapted to assess axon and gliotic morphology in toluidine blue-stained, Brown Norway rat ON light micrographs. QuPath axon numbers, density, size distributions, and gliotic areas were obtained from test images and ON cross-sections separated by damage grade. QuPath results were compared with manual counting, AxonJ, and electron microscopy axon estimates. Results: QuPath-derived axon number, density, and diameter decreased with increasing ON damage. Axon density negatively correlated with gliotic areas in test images (R2 = 0.759; P < 0.0001; N = 40) and in ON cross-sections (R2 = 0.803; P < 0.0004; N = 10). Although axon losses occurred across most axon diameters, large axons were more susceptible to degeneration. The exception was swollen axons > 2 µm, which increased in moderately but not severely damaged images. QuPath axon counts correlated strongly with manual counts of test images (R2 = 0.956; P < 0.0001). QuPath outperformed AxonJ on test images and total ON axon counts. Compared to electron microscopy analysis, QuPath undercounted ON axons; however, correlation between the methods was robust (R2 = 0.797; P < 0.001; N = 10). Conclusions: QuPath analysis reliably identified axon loss, axon morphology changes, and gliotic expansion that occurred in degenerating ONs. Translational Relevance: QuPath is a valuable tool for rapid, automated, analysis of healthy and degenerating ONs. Reproducible preclinical studies for new glaucoma treatments depend on unbiased in-depth analysis of ON pathology. This was provided by the QuPath approach.


Asunto(s)
Traumatismos del Nervio Óptico , Nervio Óptico , Animales , Axones/patología , Degeneración Nerviosa/patología , Nervio Óptico/diagnóstico por imagen , Traumatismos del Nervio Óptico/patología , Ratas , Ratas Endogámicas BN
6.
Sci Rep ; 9(1): 19406, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31852976

RESUMEN

Keratoconus (KC) is the most common corneal ectatic disorder affecting >300,000 people in the US. KC normally has its onset in adolescence, progressively worsening through the third to fourth decades of life. KC patients report significant impaired vision-related quality of life. Genetic factors play an important role in KC pathogenesis. To identify novel genes in familial KC patients, we performed whole exome and genome sequencing in a four-generation family. We identified potential variants in the PPIP5K2 and PCSK1 genes. Using in vitro cellular model and in vivo gene-trap mouse model, we found critical evidence to support the role of PPIP5K2 in normal corneal function and KC pathogenesis. The gene-trap mouse showed irregular corneal surfaces and pathological corneal thinning resembling KC. For the first time, we have integrated corneal tomography and pachymetry mapping into characterization of mouse corneal phenotypes which could be widely implemented in basic and translational research for KC diagnosis and therapy in the future.


Asunto(s)
Predisposición Genética a la Enfermedad , Queratocono/genética , Fosfotransferasas (Aceptor del Grupo Fosfato)/genética , Proproteína Convertasa 1/genética , Adulto , Animales , Mapeo Cromosómico , Córnea/diagnóstico por imagen , Córnea/patología , Topografía de la Córnea/métodos , Modelos Animales de Enfermedad , Femenino , Ligamiento Genético , Genoma Humano/genética , Genotipo , Humanos , Queratocono/patología , Masculino , Ratones , Mutación/genética , Linaje , Calidad de Vida , Secuenciación del Exoma
7.
Prog Retin Eye Res ; 67: 130-149, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30075336

RESUMEN

Retinal degenerative diseases are major causes of untreatable blindness worldwide and efficacious treatments for these diseases are sorely needed. A novel target for treatment of retinal disease is the transmembrane protein Sigma 1 Receptor (Sig1R). This enigmatic protein is an evolutionary isolate with no known homology to any other protein. Sig1R was originally thought to be an opioid receptor. That notion has been dispelled and more recent pharmacological and molecular studies suggest that it is a pluripotent modulator with a number of biological functions, many of which are relevant to retinal disease. This review provides an overview of the discovery of Sig1R and early pharmacologic studies that led to the cloning of the Sig1R gene and eventual elucidation of its crystal structure. Studies of Sig1R in the eye were not reported until the late 1990s, but since that time there has been increasing interest in the potential role of Sig1R as a target for retinal disease. Studies have focused on elucidating the mechanism(s) of Sig1R function in retina including calcium regulation, modulation of oxidative stress, ion channel regulation and molecular chaperone activity. Mechanistic studies have been performed in isolated retinal cells, such as Müller glial cells, microglial cells, optic nerve head astrocytes and retinal ganglion cells as well as in the intact retina. Several compelling studies have provided evidence of powerful in vivo neuroprotective effects against ganglion cell loss as well as photoreceptor cell loss. Also described are studies that have examined retinal structure/function in various models of retinal disease in which Sig1R is absent and reveal that these phenotypes are accelerated compared to retinas of animals that express Sig1R. The collective evidence from analysis of studies over the past 20 years is that Sig1R plays a key role in modulating retinal cellular stress and that it holds great promise as a target in retinal neurodegenerative disease.


Asunto(s)
Terapia Molecular Dirigida/métodos , Fármacos Neuroprotectores/uso terapéutico , Receptores sigma/fisiología , Degeneración Retiniana , Animales , Humanos , Canales Iónicos/fisiología , Chaperonas Moleculares/fisiología , Neuroprotección/fisiología , Estrés Oxidativo/fisiología , Degeneración Retiniana/tratamiento farmacológico , Degeneración Retiniana/metabolismo , Receptor Sigma-1
8.
Exp Eye Res ; 167: 25-30, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29031856

RESUMEN

Glaucoma is an incurable optic neuropathy characterized by dysfunction and death of retinal ganglion cells (RGCs). Brain derived neurotrophic factor (BDNF) is an essential neurotrophin that supports RGC function and survival. Despite BDNF's importance, our knowledge of molecular mechanisms that modulate BDNF processing and secretion is incomplete. Sigma-1 receptor (S1R) is associated with increased BDNF in hippocampus and with BDNF secretion by brain-derived astrocytes and neuronal cell lines. Much less is known about the relationship between S1R and BDNF in the visual system. Here, we examine how S1R activation and deletion alter expression of mature BDNF (mBDNF) and proBDNF in retina and cultured optic nerve head (ONH) astrocytes. For S1R activation, the S1R agonist (+)-pentazocine (PTZ, 0.5 mg/kg) was administered by intraperitoneal injection to C57BL/6J mice, 3 times per week, for 5 weeks. Expression of proBDNF and mBDNF was also examined in S1R knockout and age-matched C57BL/6J mice. In vitro, cultured ONH astrocytes were treated with 3 µM PTZ for 24 h followed by collection of media and ONH astrocyte lysates. Results showed that treatment with (+)-PTZ increased mBDNF protein in both retina and hippocampus. In contrast, S1R deletion was associated with retinal mBDNF deficits. In ONH astrocytes S1R agonist (+)-PTZ significantly increased levels of secreted BDNF and proBDNF in cell lysates. These findings support a role for S1R in the modulation of BDNF levels within the retina and optic nerve head. Treatment with S1R agonists might provide benefit in diseases such as glaucoma by increasing BDNF levels from endogenous sources.


Asunto(s)
Astrocitos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Receptores sigma/fisiología , Retina/metabolismo , Analgésicos Opioides/farmacología , Animales , Western Blotting , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Glaucoma/metabolismo , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Disco Óptico/citología , Pentazocina/farmacología , Ratas , Ratas Sprague-Dawley , Receptores sigma/agonistas , Receptor Sigma-1
9.
PLoS One ; 12(9): e0184421, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28898265

RESUMEN

The sigma 1 receptor (S1R) is a unique transmembrane protein that has been shown to regulate neuronal differentiation and cellular survival. It is expressed within several cell types throughout the nervous system and visceral organs, including neurons and glia within the eye. S1R ligands are therapeutic targets for diseases ranging from neurodegenerative conditions to neoplastic disorders. However, effects of S1R activation and inhibition within glia cells are not well characterized. Within the eye, the astrocytes at the optic nerve head are crucial to the health and survival of the neurons that send visual information to the brain. In this study, we used the S1R-specific agonist, (+)-pentazocine, to evaluate S1R activation within optic nerve head-derived astrocytes (ONHAs). Treatment of ONHAs with (+)-pentazocine attenuated the level and duration of stress-induced ERK phosphorylation following oxidative stress exposure and promoted survival of ONHAs. These effects were specific to S1R activation because they were not observed in ONHAs that were depleted of S1R using siRNA-mediated knockdown. Collectively, our results suggest that S1R activation suppresses ERK1/2 phosphorylation and protects ONHAs from oxidative stress-induced death.


Asunto(s)
Astrocitos/metabolismo , Nervio Óptico/metabolismo , Receptores sigma/metabolismo , Analgésicos Opioides/farmacología , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Células HeLa , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Nervio Óptico/citología , Estrés Oxidativo , Pentazocina/farmacología , Ratas , Ratas Sprague-Dawley , Receptores sigma/agonistas , Receptor Sigma-1
10.
Invest Ophthalmol Vis Sci ; 57(2): 453-61, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26868747

RESUMEN

PURPOSE: To evaluate, in vivo, the effects of the sigma-1 receptor (σR1) agonist, (+)-pentazocine, on N-methyl-D-aspartate (NMDA)-mediated retinal excitotoxicity. METHODS: Intravitreal NMDA injections were performed in C57BL/6J mice (wild type [WT]) and σR1-/- (σR1 knockout [KO]) mice. Fellow eyes were injected with phosphate-buffered saline (PBS). An experimental cohort of WT and σR1 KO mice was administered (+)-pentazocine by intraperitoneal injection, and untreated animals served as controls. Retinas derived from mice were flat-mounted and labeled for retinal ganglion cells (RGCs). The number of RGCs was compared between NMDA and PBS-injected eyes for all groups. Apoptosis was assessed using TUNEL assay. Levels of extracellular-signal-regulated kinases (ERK1/2) were analyzed by Western blot. RESULTS: N-methyl-D-aspartate induced a significant increase in TUNEL-positive nuclei and a dose-dependent loss of RGCs. Mice deficient in σR1 showed greater RGC loss (≈80%) than WT animals (≈50%). (+)-Pentazocine treatment promoted neuronal survival, and this effect was prevented by deletion of σR1. (+)-Pentazocine treatment resulted in enhanced activation of ERK at the 6-hour time point following NMDA injection. The (+)-pentazocine-induced ERK activation was diminished in σR1 KO mice. CONCLUSIONS: Targeting σR1 activation prevented RGC death while enhancing activation of the mitogen-activated protein kinase (MAPK), ERK1/2. Sigma-1 receptor is a promising therapeutic target for retinal neurodegenerative diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/toxicidad , N-Metilaspartato/toxicidad , Antagonistas de Narcóticos/farmacología , Pentazocina/farmacología , Receptores sigma/metabolismo , Degeneración Retiniana/prevención & control , Células Ganglionares de la Retina/patología , Animales , Western Blotting , Recuento de Células , Relación Dosis-Respuesta a Droga , Etiquetado Corte-Fin in Situ , Inyecciones Intraperitoneales , Inyecciones Intravítreas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo , Receptor Sigma-1
11.
Curr Eye Res ; 41(8): 1105-1112, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26642738

RESUMEN

PURPOSE: Sigma receptors 1 (σR1) and 2 (σR2) are thought to be two distinct proteins which share the ability to bind multiple ligands, several of which are common to both receptors. Whether σR1 and σR2 share overlapping biological functions is unknown. Recently, progesterone receptor membrane component 1 (PGRMC1) was shown to contain the putative σR2 binding site. PGRMC1 has not been studied in retina. We hypothesize that biological interactions between σR1 and PGRMC1 will be evidenced by compensatory upregulation of PGRMC1 in σR1-/- mice. METHODS: Immunofluorescence, RT-PCR, and immunoblotting methods were used to analyze expression of PGRMC1 in wild-type mouse retina. Tissues from σR1-/- mice were used to investigate whether a biological interaction exists between σR1 and PGRMC1. RESULTS: In the eye, PGRMC1 is expressed in corneal epithelium, lens, ciliary body epithelium, and retina. In retina, PGRMC1 is present in Müller cells and retinal pigment epithelium. This expression pattern is similar, but not identical to σR1. PGRMC1 protein levels in neural retina and eye cup from σR1-/- mice did not differ from wild-type mice. Nonocular tissues, lung, heart, and kidney showed similar Pgrmc1 gene expression in wild-type and σR1-/- mice. In contrast, liver, brain, and intestine showed increased Pgrmc1 gene expression in σR1-/- mice. CONCLUSION: Despite potential biological overlap, deletion of σR1 did not result in a compensatory change in PGRMC1 protein levels in σR1-/- mouse retina. Increased Pgrmc1 gene expression in organs with high lipid content such as liver, brain, and intestine indicates a possible tissue-specific interaction between σR1 and PGRMC1. The current studies establish the presence of PGRMC1 in retina and lay the foundation for analysis of its biological function.


Asunto(s)
Regulación de la Expresión Génica , Proteínas de la Membrana/genética , ARN Mensajero/genética , Receptores de Progesterona/genética , Células Ganglionares de la Retina/metabolismo , Animales , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Immunoblotting , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Receptores de Progesterona/biosíntesis , Células Ganglionares de la Retina/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Mol Ther Methods Clin Dev ; 2: 15013, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26029724

RESUMEN

Accumulation of the nerve growth factor precursor (proNGF) and its receptor p75(NTR) have been associated with several neurodegenerative diseases in both brain and retina. However, whether proNGF contributes to microvascular degeneration remain unexplored. This study seeks to investigate the mechanism by which proNGF/p75(NTR) induce endothelial cell (EC) death and development of acellular capillaries, a surrogate marker of retinal ischemia. Stable overexpression of the cleavage-resistant proNGF and molecular silencing of p75(NTR) were utilized in human retinal EC and rat retinas in vivo. Stable overexpression of proNGF decreased NGF levels and induced retinal vascular cell death evident by 1.9-fold increase in acellular capillaries and activation of JNK and cleaved-PARP that were mitigated by p75(NTR)shRNA. In vitro, overexpression of proNGF did not alter TNF-α level, reduced NGF, however induced EC apoptosis evident by activation of JNK and p38 MAPK, cleaved-PARP. Silencing p75(NTR) using siRNA restored expression of NGF and TrkA activation and prevented EC apoptosis. Treatment of EC with human-mutant proNGF induced apoptosis that coincided with marked protein interaction and nuclear translocation of p75(NTR) and the neurotrophin receptor interacting factor. These effects were abolished by a selective p75(NTR) antagonist. Therefore, targeting p75(NTR) represents a potential therapeutic strategy for diseases associated with aberrant expression of proNGF.

13.
Middle East Afr J Ophthalmol ; 22(2): 135-44, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25949069

RESUMEN

Diabetic retinopathy (DR) is the leading cause of blindness in working-age adults in United States. Research indicates an association between oxidative stress and the development of diabetes complications. However, clinical trials with general antioxidants have failed to prove effective in diabetic patients. Mounting evidence from experimental studies that continue to elucidate the damaging effects of oxidative stress and inflammation in both vascular and neural retina suggest its critical role in the pathogenesis of DR. This review will outline the current management of DR as well as present potential experimental therapeutic interventions, focusing on molecules that link oxidative stress to inflammation to provide potential therapeutic targets for treatment or prevention of DR. Understanding the biochemical changes and the molecular events under diabetic conditions could provide new effective therapeutic tools to combat the disease.


Asunto(s)
Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/etiología , Inhibidores de la Angiogénesis/uso terapéutico , Complicaciones de la Diabetes , Retinopatía Diabética/metabolismo , Humanos , NADPH Oxidasas/efectos adversos , NADPH Oxidasas/metabolismo , Estrés Oxidativo , Ácido Peroxinitroso/efectos adversos , Ácido Peroxinitroso/metabolismo , Estados Unidos
15.
Expert Rev Ophthalmol ; 9(2): 99-107, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25031607

RESUMEN

Diabetic retinopathy (DR), a major ocular complication of diabetes, is a leading cause of blindness in US working age adults with limited treatments. Neurotrophins (NTs), a family of proteins essential for growth, differentiation and survival of retinal neurons, have emerged as potential players in the pathogenesis of DR. NTs can signal through their corresponding tropomyosin kinase related receptor to mediate cell survival or through the p75 neurotrophin receptor with the co-receptor, sortilin, to mediate cell death. This review focuses on the role of NGF, the first discovered NT, in the development of DR. Impaired processing of proNGF has been found in ocular fluids from diabetic patients as well as experimental models. Evidence from literature and our studies support the notion that NTs appear to play multiple potential roles in DR, hence, understanding their contribution to DR may lead to promising therapeutic approaches for this devastating disease.

16.
Diabetologia ; 56(10): 2329-39, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23918145

RESUMEN

AIMS/HYPOTHESIS: Diabetic retinopathy is characterised by early blood-retina barrier (BRB) breakdown and neurodegeneration. Diabetes causes imbalance of nerve growth factor (NGF), leading to accumulation of the NGF precursor (proNGF), as well as the NGF receptor, p75 neurotrophin receptor (p75(NTR)), suggesting a possible pathological role of the proNGF-p75(NTR) axis in the diabetic retina. To date, the role of this axis in diabetes-induced retinal inflammation and BRB breakdown has not been explored. We hypothesised that modulating p75(NTR) would prevent diabetes- and proNGF-induced retinal inflammation and BRB breakdown. METHODS: Diabetes was induced by streptozotocin in wild-type and p75(NTR) knockout (p75KO) mice. After 5 weeks, the expression of inflammatory mediators, ganglion cell loss and BRB breakdown were determined. Cleavage-resistant proNGF was overexpressed in rodent retinas with and without p75(NTR) short hairpin RNA or with pharmacological inhibitors. In vitro, the effects of proNGF were investigated in retinal Müller glial cell line (rMC-1) and primary Müller cells. RESULTS: Deletion of p75(NTR) blunted the diabetes-induced decrease in retinal NGF expression and increases in proNGF, nuclear factor κB (NFκB), p-NFκB and TNF-α. Deletion of p75(NTR) also abrogated diabetes-induced glial fibrillary acidic protein expression, ganglion cell loss and vascular permeability. Inhibited expression or cleavage of p75(NTR) blunted proNGF-induced retinal inflammation and vascular permeability. In vitro, proNGF induced p75(NTR)-dependent production of inflammatory mediators in primary wild-type Müller and rMC-1 cultures, but not in p75KO Müller cells. CONCLUSIONS/INTERPRETATION: The proNGF-p75(NTR) axis contributes to retinal inflammation and vascular dysfunction in the rodent diabetic retina. These findings underscore the importance of p75(NTR) as a novel regulator of inflammation and potential therapeutic target in diabetic retinopathy.


Asunto(s)
Barrera Hematorretinal/metabolismo , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/metabolismo , Receptor de Factor de Crecimiento Nervioso/metabolismo , Animales , Western Blotting , Células Cultivadas , Diabetes Mellitus Experimental/inmunología , Ensayo de Inmunoadsorción Enzimática , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Noqueados , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Factor de Crecimiento Nervioso/genética
17.
PLoS One ; 8(1): e54692, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23365678

RESUMEN

Our previous studies showed positive correlation between accumulation of proNGF, activation of RhoA and neuronal death in diabetic models. Here, we examined the neuroprotective effects of selective inhibition of RhoA kinase in the diabetic rat retina and in a model that stably overexpressed the cleavage-resistance proNGF plasmid in the retina. Male Sprague-Dawley rats were rendered diabetic using streptozotocin or stably express cleavage-resistant proNGF plasmid. The neuroprotective effects of the intravitreal injection of RhoA kinase inhibitor Y27632 were examined in vivo. Effects of proNGF were examined in freshly isolated primary retinal ganglion cell (RGC) cultures and RGC-5 cell line. Retinal neurodegeneration was assessed by counting TUNEL-positive and Brn-3a positive retinal ganglion cells. Expression of proNGF, p75(NTR), cleaved-PARP, caspase-3 and p38MAPK/JNK were examined by Western-blot. Activation of RhoA was assessed by pull-down assay and G-LISA. Diabetes and overexpression of proNGF resulted in retinal neurodegeneration as indicated by 9- and 6-fold increase in TUNEL-positive cells, respectively. In vitro, proNGF induced 5-fold cell death in RGC-5 cell line, and it induced >10-fold cell death in primary RGC cultures. These effects were associated with significant upregulation of p75(NTR) and activation of RhoA. While proNGF induced TNF-α expression in vivo, it selectively activated RhoA in primary RGC cultures and RGC-5 cell line. Inhibiting RhoA kinase with Y27632 significantly reduced diabetes- and proNGF-induced activation of proapoptotic p38MAPK/JNK, expression of cleaved-PARP and caspase-3 and prevented retinal neurodegeneration in vivo and in vitro. Taken together, these results provide compelling evidence for a causal role of proNGF in diabetes-induced retinal neurodegeneration through enhancing p75(NTR) expression and direct activation of RhoA and p38MAPK/JNK apoptotic pathways.


Asunto(s)
Diabetes Mellitus Experimental/genética , Factor de Crecimiento Nervioso/genética , Precursores de Proteínas/genética , Degeneración Retiniana/genética , Células Ganglionares de la Retina/metabolismo , Proteína de Unión al GTP rhoA/genética , Amidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/genética , Caspasa 3/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Regulación de la Expresión Génica/efectos de los fármacos , Inyecciones Intravítreas , Masculino , Factor de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso , Poli(ADP-Ribosa) Polimerasas , Cultivo Primario de Células , Inhibidores de Proteínas Quinasas/farmacología , Precursores de Proteínas/metabolismo , Proteolisis/efectos de los fármacos , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Transducción de Señal/efectos de los fármacos , Estreptozocina , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
18.
Mol Vis ; 18: 2993-3003, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23288991

RESUMEN

PURPOSE: Neurotrophins, including nerve growth factor (NGF), are secreted by glia as a pro-form (proNGF) that is normally cleaved into the mature ligand. Increases of proNGF has been well documented in retinal neurodegenerative diseases. Since systemic overexpression of proNGF exhibits embryonic lethality, we aimed to establish a model that specifically and stably overexpresses a cleavage-resistant mutant of proNGF (proNGF123) plasmid in the retina using electroporation. METHODS: Male Sprague-Dawley rats were injected intravitreally with pGFP or pGFP-proNGF123 plasmids, then electroporated with various settings for optimization. Retinal cell death and ganglion cell count were assessed by TUNEL and immunostaining with anti-Brn3. Expression of proNGF, NGF, and their receptors was examined by western blot. Retinal vascular permeability was assessed by extravasation of bovine serum albumin-fluorescein. Development of acellular capillaries was assessed by periodic acid-Schiff and hematoxylin staining. RESULTS: Successful pGFP-proNGF123 gene delivery and expression of proNGF was demonstrated by western blot and extensive proNGF immunostaining in retina sections. Overexpression of proNGF reduced NGF expression while inducing the expression of neurotrophin receptors, including p75(NTR) and tyrosine receptor kinase A, but not sortilin. Overexpression of proNGF resulted in ~50% reduction in ganglion cell count and fivefold increase in TUNEL-positive cells in rat retina. In addition, overexpression of proNGF induced breakdown of the blood-retina barrier evident by twofold increase in extravasation of bovine serum albumin-fluorescein after 1 week and induced the development of acellular capillaries after 4 weeks. CONCLUSIONS: Electroporation can successfully incorporate and express biologically active cleavage-resistant proNGF locally in rat retinas. Overexpression of cleavage-resistant proNGF can be a useful tool to investigate specific molecular mechanisms by which proNGF causes neurodegeneration and vascular injury in the retina.


Asunto(s)
Barrera Hematorretinal/patología , Factores de Crecimiento Nervioso/metabolismo , Precursores de Proteínas/metabolismo , Neuronas Retinianas/patología , Vasos Retinianos/patología , Animales , Barrera Hematorretinal/metabolismo , Permeabilidad Capilar , Supervivencia Celular , Electroporación , Expresión Génica , Técnicas de Transferencia de Gen , Inyecciones Intravítreas , Masculino , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso , Precursores de Proteínas/genética , Proteolisis , Ratas , Ratas Sprague-Dawley , Receptor trkA/genética , Receptor trkA/metabolismo , Receptores de Factores de Crecimiento , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Neuronas Retinianas/metabolismo , Vasos Retinianos/metabolismo , Factor de Transcripción Brn-3/genética , Factor de Transcripción Brn-3/metabolismo , Transgenes
20.
Invest Ophthalmol Vis Sci ; 50(9): 4460-70, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19357353

RESUMEN

PURPOSE: To determine the effects of endogenous elevation of homocysteine on the retina using the cystathionine beta-synthase (cbs) mutant mouse. METHODS: Retinal homocysteine in cbs mutant mice was measured by high-performance liquid chromatography (HPLC). Retinal cryosections from cbs(-/-) mice and cbs(+/-) mice were examined for histologic changes by light and electron microscopy. Morphometric analysis was performed on retinas of cbs(+/-) mice maintained on a high-methionine diet (cbs(+/-) HM). Changes in retinal gene expression were screened by microarray. RESULTS: HPLC analysis revealed an approximate twofold elevation in retinal homocysteine in cbs(+/-) mice and an approximate sevenfold elevation in cbs(-/-) mice. Distinct alterations in the ganglion, inner plexiform, inner nuclear, and epithelial layers were observed in retinas of cbs(-/-) and 1-year-old cbs(+/-) mice. Retinas of cbs(+/-) HM mice demonstrated an approximate 20% decrease in cells of the ganglion cell layer (GCL), which occurred as early as 5-weeks after onset of the HM diet. Microarray analysis revealed alterations in expression of several genes, including increased expression of Aven, Egr1, and Bat3 in retinas of cbs(+/-) HM mice. CONCLUSIONS: This study provides the first analysis of morphologic and molecular effects of endogenous elevations of retinal homocysteine in an in vivo model. Increased retinal homocysteine alters inner and outer retinal layers in cbs homozygous mice and older cbs heterozygous mice, and it primarily affects the cells of the GCL in younger heterozygous mice. Elevated retinal homocysteine alters expression of genes involved in endoplasmic reticular stress, N-methyl-d-aspartate (NMDA) receptor activation, cell cycle, and apoptosis.


Asunto(s)
Apoptosis , Cistationina betasintasa/genética , Homocisteína/fisiología , Hiperhomocisteinemia/patología , Mutación , Enfermedades de la Retina/patología , Células Ganglionares de la Retina/patología , Animales , Cromatografía Líquida de Alta Presión , Cistationina betasintasa/deficiencia , Cistationina betasintasa/metabolismo , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Expresión Génica/fisiología , Glutatión/metabolismo , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/metabolismo , Masculino , Ratones , Ratones Noqueados , Análisis por Micromatrices , Enfermedades de la Retina/genética , Enfermedades de la Retina/metabolismo , Células Ganglionares de la Retina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...