Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38293142

RESUMEN

Macropinocytosis has emerged as a nutrient-scavenging pathway that cancer cells exploit to survive the nutrient-deprived conditions of the tumor microenvironment. Cancer cells are especially reliant on glutamine for their survival, and in pancreatic ductal adenocarcinoma (PDAC) cells, glutamine deficiency can enhance the stimulation of macropinocytosis, allowing the cells to escape metabolic stress through the production of extracellular-protein-derived amino acids. Here, we identify the atypical protein kinase C (aPKC) enzymes, PKCζ and PKCι as novel regulators of macropinocytosis. In normal epithelial cells, aPKCs are known to regulate cell polarity in association with the scaffold proteins Par3 and Par6, controlling the function of several targets, including the Par1 kinases. In PDAC cells, we identify that each of these cell polarity proteins are required for glutamine stress-induced macropinocytosis. Mechanistically, we find that the aPKCs are regulated by EGFR signaling or by the transcription factor CREM to promote the relocation of Par3 to microtubules, facilitating macropinocytosis in a dynein-dependent manner. Importantly, we determine that cell fitness impairment caused by aPKC depletion is rescued by the restoration of macropinocytosis and that aPKCs support PDAC growth in vivo. These results identify a previously unappreciated role for cell polarity proteins in the regulation of macropinocytosis and provide a better understanding of the mechanistic underpinnings that control macropinocytic uptake in the context of metabolic stress.

2.
Nat Cancer ; 5(1): 100-113, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37814011

RESUMEN

In pancreatic ductal adenocarcinoma (PDAC), glutamine is a critical nutrient that drives a wide array of metabolic and biosynthetic processes that support tumor growth. Here, we elucidate how 6-diazo-5-oxo-L-norleucine (DON), a glutamine antagonist that broadly inhibits glutamine metabolism, blocks PDAC tumor growth and metastasis. We find that DON significantly reduces asparagine production by inhibiting asparagine synthetase (ASNS), and that the effects of DON are rescued by asparagine. As a metabolic adaptation, PDAC cells upregulate ASNS expression in response to DON, and we show that ASNS levels are inversely correlated with DON efficacy. We also show that L-asparaginase (ASNase) synergizes with DON to affect the viability of PDAC cells, and that DON and ASNase combination therapy has a significant impact on metastasis. These results shed light on the mechanisms that drive the effects of glutamine mimicry and point to the utility of cotargeting adaptive responses to control PDAC progression.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Glutamina/metabolismo , Asparagina/metabolismo , Línea Celular Tumoral , Asparaginasa/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Procesos Neoplásicos
3.
Aging Cell ; 18(5): e13000, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31267651

RESUMEN

Dual-specificity tyrosine phosphorylation-regulated kinase-1A (DYRK1A) is known to phosphorylate the microtubule-associated tau protein. Overexpression is correlated with tau hyperphosphorylation and neurofibrillary tangle (NFT) formation in Alzheimer's disease (AD). This study assessed the potential of SM07883, an oral DYRK1A inhibitor, to inhibit tau hyperphosphorylation, aggregation, NFT formation, and associated phenotypes in mouse models. Exploratory neuroinflammatory effects were also studied. SM07883 specificity was tested in a kinase panel screen and showed potent inhibition of DYRK1A (IC50  = 1.6 nM) and GSK-3ß (IC50  = 10.8 nM) kinase activity. Tau phosphorylation measured in cell-based assays showed a reduction in phosphorylation of multiple tau epitopes, especially the threonine 212 site (EC50  = 16 nM). SM07883 showed good oral bioavailability in multiple species and demonstrated a dose-dependent reduction of transient hypothermia-induced phosphorylated tau in the brains of wild-type mice compared to vehicle (47%, p < 0.001). Long-term efficacy assessed in aged JNPL3 mice overexpressing the P301L human tau mutation (3 mg/kg, QD, for 3 months) exhibited significant reductions in tau hyperphosphorylation, oligomeric and aggregated tau, and tau-positive inclusions compared to vehicle in brainstem and spinal cord samples. Reduced gliosis compared to vehicle was further confirmed by ELISA. SM07883 was well tolerated with improved general health, weight gain, and functional improvement in a wire-hang test compared to vehicle-treated mice (p = 0.048). SM07883, a potent, orally bioavailable, brain-penetrant DYRK1A inhibitor, significantly reduced effects of pathological tau overexpression and neuroinflammation, while functional endpoints were improved compared to vehicle in animal models. This small molecule has potential as a treatment for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Isoquinolinas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas tau/metabolismo , Administración Oral , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Isoquinolinas/administración & dosificación , Isoquinolinas/química , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Estructura Molecular , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/química , Proteínas tau/genética , Proteínas tau/toxicidad , Quinasas DyrK
4.
Sci Rep ; 6: 29523, 2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27389831

RESUMEN

Abnormal α-synuclein (α-syn) accumulation in the CNS may underlie neuronal cell and synaptic dysfunction leading to motor and cognitive deficits in synucleinopathies including Parkinson's disease (PD) and Dementia with Lewy Bodies (DLB). Multiple groups demonstrated α-syn accumulation in CNS accessory structures, including the eyes and olfactory terminals, as well as in peripheral organs of Parkinsonian patients. Retinal imaging studies of mice overexpressing fused α-syn::GFP were conducted to evaluate the presence and progression of retinal pathology in a PD/DLB transgenic mouse model. Bright-field image retinal maps and fluorescent images were acquired at 1-month intervals for 3 months. Retinal imaging revealed the accumulation of GFP-tagged α-syn in retinal ganglion cell layer and in the edges of arterial blood vessels in the transgenic mice. Double labeling studies confirmed that the α-syn::GFP-positive cells were retinal ganglion cells containing α-syn. Accumulation of α-syn persisted in the same cells and increased with age. Accumulation of α-syn::GFP was reduced by immunization with single chain antibodies against α-syn. In conclusion, longitudinal live imaging of the retina in the PDGF-α-syn::GFP mice might represent a useful, non-invasive tool to monitor the fate of α-syn accumulation in the CNS and to evaluate the therapeutic effects of compounds targeting α-syn.


Asunto(s)
Proteínas Fluorescentes Verdes/metabolismo , Enfermedad por Cuerpos de Lewy/diagnóstico por imagen , Enfermedad de Parkinson/diagnóstico por imagen , Retina/diagnóstico por imagen , Anticuerpos de Cadena Única/administración & dosificación , alfa-Sinucleína/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunoterapia , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/terapia , Masculino , Ratones , Ratones Transgénicos , Imagen Óptica/instrumentación , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , Proteínas Recombinantes de Fusión/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Anticuerpos de Cadena Única/farmacología , alfa-Sinucleína/efectos de los fármacos , alfa-Sinucleína/genética
5.
Neurobiol Aging ; 36(5): 1969-81, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25772060

RESUMEN

Long-term dietary supplementation with resveratrol protects against cardiovascular disease, osteoporesis, and metabolic decline. This study determined how long-term dietary resveratrol treatment protects against retinal ganglion cell (RGC) dendrite loss after optic nerve injury and alters the resolution of the unfolded protein response. Associated changes in markers of endoplasmic reticulum stress in RGCs also were investigated. Young-adult Thy1-yellow fluorescent protein (YFP) and C57BL/6 mice received either control diet or diet containing resveratrol for approximately 1 year. Both groups then received optic nerve crush (ONC). Fluorescent RGC dendrites in the Thy1-YFP mice were imaged weekly for 4 weeks after ONC. There was progressive loss of dendrite length in all RGC types within the mice that received control diet. Resveratrol delayed loss of dendrite complexity and complete dendrite loss for most RGC types. However, there were variations in the rate of retraction among different RGC types. Three weeks after ONC, cytoplasmic binding immunoglobulin protein (BiP) suppression observed in control diet ganglion cell layer neurons was reversed in mice that received resveratrol, nuclear C/EBP homologous protein (CHOP) was near baseline in control diet eyes but was moderately increased by resveratrol; and increased nuclear X-box-binding protein-1 (XBP-1) observed in control diet eyes was reduced in eyes that received resveratrol to the same level as in control diet uncrushed eyes. These results indicate that protection of dendrites by resveratrol after ONC differs among RGC types and suggest that alterations in long-term expression of binding immunoglobulin protein, CHOP, and XBP-1 may contribute to the resveratrol-mediated protection of RGC dendrites after ONC.


Asunto(s)
Dendritas/metabolismo , Dendritas/patología , Suplementos Dietéticos , Traumatismos del Nervio Óptico/patología , Desplegamiento Proteico/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Estilbenos/administración & dosificación , Estilbenos/farmacología , Animales , Proteínas de Unión al ADN/metabolismo , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteínas de Choque Térmico/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores de Transcripción del Factor Regulador X , Resveratrol , Células Ganglionares de la Retina/citología , Factores de Tiempo , Factor de Transcripción CHOP/metabolismo , Factores de Transcripción/metabolismo , Proteína 1 de Unión a la X-Box
6.
Invest Ophthalmol Vis Sci ; 56(3): 1789-804, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25634981

RESUMEN

PURPOSE: To determine whether brimonidine protects against the retraction and loss of retinal ganglion cell (RGC) dendrites after optic nerve crush (ONC). METHODS: Fluorescent RGCs of mice expressing yellow fluorescent protein (YFP) under the control of the Thy-1 promoter (Thy1-YFP mice) were imaged in vivo and assigned to one of six groups according to dendrite structure. The mice then received brimonidine every other day starting 2 days before, or 2 or 6 days after, unilateral ONC. Control animals received vehicle every other day starting 2 days before ONC. Control animals received vehicle every other day starting 2 days before ONC. Total dendrite length, dendrite branching complexity, and the time until complete loss of dendrites were assessed weekly for 4 weeks. RESULTS: Overall, brimonidine treatment significantly slowed the complete loss of RGC dendrites and significantly slowed the reduction of total dendrite length and branching complexity. Separate analysis of each RGC group showed brimonidine significantly delayed the time until complete loss of dendrites in four of the RGC groups. These delays generally were similar when treatment started either 2 days before or 2 days after ONC, but were smaller or absent when treatment started 6 days after ONC Protection against loss of total dendrite length and loss of branching complexity was observed in three of the RGC groups. In two of these RGC groups, protective effects persisted until the end of the study. CONCLUSIONS: Brimonidine protects many RGC types against dendrite retraction, loss of branching complexity, and complete loss of dendrites following ONC. However, the pattern and magnitude of this protection differs substantially among different RGC types. These results indicate that requirements for RGC-protective therapies following optic nerve injury may differ among RGC types.


Asunto(s)
Citoprotección/efectos de los fármacos , Dendritas/efectos de los fármacos , Compresión Nerviosa , Traumatismos del Nervio Óptico/patología , Quinoxalinas/farmacología , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Tartrato de Brimonidina , Dendritas/patología , Femenino , Masculino , Ratones , Ratones Endogámicos , Microscopía Fluorescente , Células Ganglionares de la Retina/patología
7.
PLoS One ; 8(8): e65966, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23940507

RESUMEN

Thy-1 is a cell surface protein that is expressed during the differentiation of retinal ganglion cells (RGCs). Optic nerve injury induces progressive loss in the number of RGCs expressing Thy-1. The rate of this loss is fastest during the first week after optic nerve injury and slower in subsequent weeks. This study was undertaken to determine whether oral treatment with a water-soluble N-hydroxy-2,2,6,6-tetramethylpiperidine derivative (OT-440) protects against loss of Thy-1 promoter activation following optic nerve crush and whether this effect targets the earlier quick phase or the later slow phase. The retina of mice expressing cyan fluorescent protein under control of the Thy-1 promoter (Thy1-CFP mice) was imaged using a blue-light confocal scanning laser ophthalmoscope (bCSLO). These mice then received oral OT-440 prepared in cream cheese or dissolved in water, or plain vehicle, for two weeks and were imaged again prior to unilateral optic nerve crush. Treatments and weekly imaging continued for four more weeks. Fluorescent neurons were counted in the same defined retinal areas imaged at each time point in a masked fashion. When the counts at each time point were directly compared, the numbers of fluorescent cells at each time point were greater in the animals that received OT-440 in cream cheese by 8%, 27%, 52% and 60% than in corresponding control animals at 1, 2, 3 and 4 weeks after optic nerve crush. Similar results were obtained when the vehicle was water. Rate analysis indicated the protective effect of OT-440 was greatest during the first two weeks and was maintained in the second two weeks after crush for both the cream cheese vehicle study and water vehicle study. Because most of the fluorescent cells detected by bCSLO are RGCs, these findings suggest that oral OT-440 can either protect against or delay early degenerative responses occurring in RGCs following optic nerve injury.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Hidroxilamina/química , Hidroxilamina/farmacología , Traumatismos del Nervio Óptico/tratamiento farmacológico , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/efectos de los fármacos , Administración Oral , Animales , Peso Corporal/efectos de los fármacos , Femenino , Hidroxilamina/administración & dosificación , Hidroxilamina/uso terapéutico , Masculino , Ratones , Microscopía Confocal , Compresión Nerviosa , Células Ganglionares de la Retina/metabolismo , Antígenos Thy-1/metabolismo
8.
BMC Ophthalmol ; 13(1): 26, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23805828

RESUMEN

BACKGROUND: The loss of RGCs expressing Thy-1 after optic nerve injury has an initial phase of rapid decline followed by a longer phase with slower reduction rate. This study used longitudinal retinal imaging of mice expressing cyan fluorescent protein under control of the Thy-1 promoter (Thy1-CFP mice) to determine how the α2-adrenergic agonist brimonidine influences loss of Thy1 promoter activation. METHODS: Baseline images of the fluorescent retinal neurons in 30 Thy1-CFP mice were obtained using a modified confocal scanning laser ophthalmoscope. Next, brimonidine (100 ug/kg, IP) was administered either one time immediately after optic nerve crush, or immediately after optic nerve crush and then every 2 days for four weeks. A control group received a single saline injection immediately after optic nerve crush. All animals were imaged weekly for four weeks after optic nerve crush. Loss of fluorescent retinal neurons within specific retinal areas was determined by counting. RESULTS: At one week after optic nerve crush, the proportion of fluorescent retinal neurons retaining fluorescence was 44±7% of baseline in control mice, 51±6% after one brimonidine treatment, and 55±6% after brimonidine treatment every other day (P<0.05 for both brimonidine treatment groups compared to the control group). Subsequently, the number of fluorescent retinal neurons in the group that received one treatment differed insignificantly from the control group. In contrast, the number of fluorescent retinal neurons in the group that received repeated brimonidine treatments was greater than the control group by 28% at two weeks after crush and by 32% at three weeks after crush (P<0.05 at both time points). Rate analysis showed that brimonidine slowed the initial rate of fluorescent cell decline in the animals that received multiple treatments (P<0.05). Differences in the rate of loss among the treatment groups were insignificant after the second week. CONCLUSION: Repeated brimonidine treatments protect against loss of fluorescence within fluorescent retinal neurons of Thy1-CFP mice after optic nerve crush. As most of fluorescent retinal neurons in this system are RGCs, these findings indicate that repeated brimonidine treatments may protect RGC health following optic nerve crush.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/uso terapéutico , Compresión Nerviosa , Traumatismos del Nervio Óptico/tratamiento farmacológico , Regiones Promotoras Genéticas , Sustancias Protectoras/uso terapéutico , Quinoxalinas/uso terapéutico , Antígenos Thy-1/fisiología , Análisis de Varianza , Animales , Tartrato de Brimonidina , Recuento de Células , Modelos Animales de Enfermedad , Femenino , Estudios Longitudinales , Masculino , Ratones , Microscopía Fluorescente , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/patología , Células Ganglionares de la Retina/efectos de los fármacos
9.
Magn Reson Imaging ; 31(6): 865-74, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23541073

RESUMEN

Injection of manganese into the eye will enhance the contrast of visual system neuronal pathways imaged by MRI (MEMRI). The present study was undertaken to determine the effect of a range of MnCl2 doses upon the integrity of various ocular structures. Anesthetized mice received ocular anterior chamber injections of 50-500 nmol of MnCl2. One week later, the eyes were fixed, embedded in paraffin, sectioned, and stained with hematoxylin and eosin. Additional animals received 50 nmol of MnCl2 injected into the anterior chamber and were later imaged using T1-weighted 7T MRI. Following 500 and 300nmol MnCl2, the corneal stroma and endothelium were degenerated, the anterior chamber contained a dense fibrin matrix with extensive inflammatory cell infiltration, a plaque often formed on the anterior lens, and significant retinal degeneration was observed. Following 100nmol MnCl2, retinal preservation of ocular structures was significantly better than at higher doses. In addition, there was no difference from vehicle control retina in cell counts within the ganglion cell layer, or in the width of the inner nuclear layer or outer nuclear layer. Also, there was no difference in the thickness of the inner plexiform layer. However, there was thinning of the peripheral outer plexiform layer, as well as in the outer segment layer. Visual system elements labeled in MRI of mice that received 100nmol MnCl2 included the retina, optic nerve, lateral geniculate nucleus, and superior colliculus. The preservation of ganglion cell layer cell counts and inner plexiform layer thickness following 100nmol MnCl2 suggests there was negligible injury to RGCs following this dose. These results support using 100nmol MnCl2 in mouse eyes for in vivo assessment of the integrity of RGC projections to target neurons in the brain.


Asunto(s)
Cloruros/efectos adversos , Imagen por Resonancia Magnética/efectos adversos , Compuestos de Manganeso/efectos adversos , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/efectos de los fármacos , Vías Visuales/anatomía & histología , Vías Visuales/efectos de los fármacos , Animales , Cloruros/administración & dosificación , Medios de Contraste/administración & dosificación , Medios de Contraste/efectos adversos , Imagen por Resonancia Magnética/métodos , Compuestos de Manganeso/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Enfermedades de la Retina/inducido químicamente , Enfermedades de la Retina/prevención & control , Coloración y Etiquetado/métodos
10.
Invest Ophthalmol Vis Sci ; 54(1): 96-102, 2013 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-23197683

RESUMEN

PURPOSE: Thy-1 is a marker of retinal ganglion cell (RGC) differentiation. Optic nerve injury triggers reduction of Thy-1 promoter activation followed by retinal ganglion cell (RGC) death. This study determined whether MS-275, an inhibitor of the histone deacetylases 1 and 3, can inhibit these changes. METHODS: Mice expressing cyan fluorescent protein (CFP) under control of the Thy-1 promoter received MS-275 (subcutaneous) or vehicle three times per week starting 1 week before optic nerve crush and continuing for 6 weeks. The same retinal area was imaged using the blue-light confocal scanning laser ophthalmoscope before and after optic nerve crush every week, and fluorescent spots were counted manually. The eyes were then processed for histopathologic analysis. RESULTS: The mean proportions of fluorescent retinal neurons remaining in the vehicle group following optic nerve crush were 36 ± 8, 18 ± 6, 13 ± 10, 12 ± 4, 13 ± 5, and 13 ± 5% at weeks 1 through 6, respectively (n = 6). In contrast, the mean proportions of fluorescent retinal neurons remaining in the group treated with MS-275 were 59 ± 19, 39 ± 11, 34 ± 12, 33 ± 15, 32 ± 13, and 27 ± 15% at weeks 1 through 6, respectively (n = 7, P < 0.05 at weeks 1 through 5). Rate analysis showed that MS-275 slowed the rate of loss during the first 2 weeks by 23% (P < 0.05) and subsequently was similar. Histopathologic analysis revealed 27 ± 13% greater ganglion cell layer (GCL) neurons in the eyes from mice that received MS-275 treatment (P < 0.02). CONCLUSIONS: These results indicate that treatment with MS-275 protects against the loss of RGC differentiation and promotes RGC survival following optic nerve injury.


Asunto(s)
Benzamidas/farmacología , Histona Desacetilasa 1/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Traumatismos del Nervio Óptico/tratamiento farmacológico , Piridinas/farmacología , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Proteínas Fluorescentes Verdes/genética , Histona Desacetilasa 1/metabolismo , Masculino , Ratones , Ratones Mutantes , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/patología , Células Ganglionares de la Retina/enzimología , Células Ganglionares de la Retina/patología , Antígenos Thy-1/genética
11.
Mol Vis ; 16: 1331-42, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20664796

RESUMEN

PURPOSE: The goal of this study is to determine whether increased optic atrophy type 1 (OPA1) expression protects against retinal ganglion cell (RGC) death in glaucomatous DBA/2J mice. METHODS: Intraocular pressure in DBA/2J mice was measured, and pre-glaucomatous DBA/2J mice eyes were transfected with recombinant adeno-associated virus serotype 2 (AAV2) constructs including AAV2-wild type (WT) mOPA1 for two months. Increased OPA1 expression was confirmed by western blotting and RGC survival was assessed by retrograde labeling with FluoroGold. In addition, apoptotic cell death and mitochondrial structure were determined in AAV2-WT mOPA1-transfected differentiated RGC-5 cells exposed to elevated hydrostatic pressure (30 mmHg) for three days. RESULTS: WT AAV2-mOPA1 transfection significantly increased 90 kDa and 80 kDa OPA1 isoforms in the retina of glaucomatous DBA/2J mice. OPA1 immunoreactivity was increased in the inner nuclear layer, inner plexiform layer, and ganglion cell layer in nine month-old glaucomatous DBA/2J mice transfected with AAV2-WT mOPA1. Overexpression of OPA1 significantly increased RGC survival at two months after AAV2-WT mOPA1 transfection, and decreased activation of both astroglia and microglia in the retina of glaucomatous DBA/2J mice. Also, overexpression of OPA1 in differentiated RGC-5 cells resulted in less apoptotic cell death and blocked mitochondrial fission following elevated hydrostatic pressure. CONCLUSIONS: OPA1 can directly modulate RGC survival, and increasing OPA1 expression may protect against RGC death in glaucomatous optic neuropathy.


Asunto(s)
Citoprotección , GTP Fosfohidrolasas/metabolismo , Glaucoma/enzimología , Glaucoma/patología , Células Ganglionares de la Retina/enzimología , Animales , Apoptosis , Astrocitos/enzimología , Astrocitos/patología , Supervivencia Celular , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Glaucoma/fisiopatología , Proteínas Fluorescentes Verdes/metabolismo , Presión Intraocular/fisiología , Ratones , Ratones Endogámicos C57BL , Microglía/enzimología , Microglía/patología , Mitocondrias/metabolismo , Células Ganglionares de la Retina/patología , Transfección
12.
Proc Natl Acad Sci U S A ; 106(21): 8760-5, 2009 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-19433784

RESUMEN

Many ocular pathologies, including retinopathy of prematurity (ROP), diabetic retinopathy, and age-related macular degeneration, result in vision loss because of aberrant neoangiogenesis. A common feature of these conditions is the presence of hypoxic areas and overexpression of the proangiogenic vascular endothelial growth factor (VEGF). The prevailing current treatment, laser ablation of the retina, is destructive and only partially effective. Preventive and less destructive therapies are much more desirable. Here, we show that mice lacking c-Jun N-terminal kinase 1 (JNK1) exhibit reduced pathological angiogenesis and lower levels of retinal VEGF production in a murine model of ROP. We found that hypoxia induces JNK activation and regulates VEGF expression by enhancing the binding of phospho-c-Jun to the VEGF promoter. Intravitreal injection of a specific JNK inhibitor decreases retinal VEGF expression and reduces pathological retinal neovascularization without obvious side effects. These results strongly suggest that JNK1 plays a key role in retinal neoangiogenesis and that it represents a new pharmacological target for treatment of diseases where excessive neoangiogenesis is the underlying pathology.


Asunto(s)
Hipoxia/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Enfermedades de la Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Activación Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Hipoxia/genética , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Proteína Quinasa 8 Activada por Mitógenos/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Inhibidores de Proteínas Quinasas/farmacología , Enfermedades de la Retina/genética , Enfermedades de la Retina/patología , Transcripción Genética/genética , Factor A de Crecimiento Endotelial Vascular/genética
13.
Invest Ophthalmol Vis Sci ; 50(2): 707-16, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18936150

RESUMEN

PURPOSE: To determine whether intraocular pressure (IOP) elevation alters OPA1 expression and triggers OPA1 release, as well as whether the uncompetitive N-methyl-d-aspartate (NMDA) glutamate receptor antagonist memantine blocks OPA1 release and subsequent apoptotic cell death in glaucomatous DBA/2J mouse retina. METHODS: Preglaucomatous DBA/2J mice received memantine (5 mg/kg, intraperitoneal injection, twice daily for 3 months) and IOP in the eyes was measured monthly. RGC loss was counted after FluoroGold labeling. OPA1, Dnm1, Bcl-2, and Bax mRNA were measured by qPCR. OPA1 protein was assessed by immunohistochemistry and Western blot. Apoptotic cell death was assessed by TUNEL staining. RESULTS: Memantine treatment significantly increased RGC survival in glaucomatous DBA/2J mice and increased the 75-kDa OPA1 isoform, but did not alter the 80- and 90-kDa isoforms. The isoforms of OPA1 were significantly increased in the cytosol of the vehicle-treated glaucomatous retinas but were significantly decreased in memantine-treated glaucomatous retinas. OPA1 immunoreactivity was decreased in the photoreceptors of both vehicle- and memantine-treated glaucomatous retinas, but was increased in the outer plexiform layer of only the memantine-treated glaucomatous retinas. Memantine blocked apoptotic cell death in the GCL, increased Bcl-2 gene expression, and decreased Bax gene expression. CONCLUSIONS: OPA1 release from mitochondria in glaucomatous mouse retina is inhibited by blockade of glutamate receptor activation. Because this OPA1 effect was accompanied by increased Bcl-2 expression, decreased Bax expression, and apoptosis blockade, glutamate receptor activation in the glaucomatous retina may involve a distinct mitochondria-mediated cell death pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Citocromos c/metabolismo , GTP Fosfohidrolasas/metabolismo , Glaucoma/prevención & control , Memantina/farmacología , Mitocondrias/efectos de los fármacos , Enfermedades de la Retina/prevención & control , Animales , Western Blotting , Supervivencia Celular , Citocromos c/genética , Dinamina I/genética , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , GTP Fosfohidrolasas/genética , Expresión Génica , Glaucoma/genética , Glaucoma/metabolismo , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Inyecciones Intraperitoneales , Presión Intraocular , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Mitocondrias/metabolismo , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Enfermedades de la Retina/genética , Enfermedades de la Retina/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Proteína X Asociada a bcl-2/genética
14.
Invest Ophthalmol Vis Sci ; 49(11): 4903-11, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18469184

RESUMEN

PURPOSE: To determine whether elevation of intraocular pressure (IOP) triggers mitochondrial fission and ultrastructural changes and alters optic atrophy type 1 (OPA1) expression and distribution in the optic nerve (ON) of glaucomatous DBA/2J mice. METHODS: IOP in the eyes of DBA/2J mice was measured, and mitochondrial structural changes were assessed by conventional electron microscopy (EM) and EM tomography. Cytochrome c oxidase IV subunit 1 (COX), OPA1, and Dnm1, a rat homologue of dynamin-related protein-1, mRNA were measured by quantitative (q)PCR. COX and OPA1 protein distribution was assessed by immunocytochemistry and Western blot. RESULTS: Excavation of the optic nerve head (ONH), axon loss, and COX reduction were evident in 10-month-old glaucomatous ONHs of eyes with >20 mm Hg IOP elevation. EM analysis showed mitochondrial fission, matrix swelling, substantially reduced cristae volume, and abnormal cristae depletion in 10-month-old glaucomatous ONH axons. The mean length of mitochondrial cross section in these axons decreased from 858.2 +/- 515.3 nm in 3-month-old mice to 583.3 +/- 298.6 nm in 10-month-old glaucomatous mice (P < 0.001). Moderate reductions of COX mRNA were observed in the 10-month-old DBA/2J mice's ONHs. Larger reductions of OPA1 immunoreactivity and gene expression were coupled with larger increases of Dnm1 gene expression in 10-month-old glaucomatous ONH. Subcellular fractionation analysis indicates increased release of both OPA1 and cytochrome c from mitochondria in 10-month-old glaucomatous ONs. CONCLUSIONS: IOP elevation may directly damage mitochondria in the ONH axons by promoting reduction of COX, mitochondrial fission and cristae depletion, alterations of OPA1 and Dnm1 expression, and induction of OPA1 release. Thus, interventions to preserve mitochondria may be useful for protecting against ON degeneration in glaucoma.


Asunto(s)
GTP Fosfohidrolasas/biosíntesis , Expresión Génica , Glaucoma/patología , Presión Intraocular/fisiología , Mitocondrias/ultraestructura , Nervio Óptico/patología , ARN Mensajero/genética , Animales , Western Blotting , Modelos Animales de Enfermedad , Dinamina I/biosíntesis , Dinamina I/genética , Complejo IV de Transporte de Electrones/biosíntesis , Complejo IV de Transporte de Electrones/genética , Femenino , GTP Fosfohidrolasas/genética , Glaucoma/metabolismo , Glaucoma/fisiopatología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Nervio Óptico/metabolismo , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA