Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 27(7): 2002-2013.e5, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31091440

RESUMEN

Microglia have important remodeling functions in neurodevelopment, aging, and disease, with evidence for molecular diversity. However, the signaling pathways and environmental cues that drive diverse states of microglia are incompletely understood. We profiled microglia of a discrete developing CNS region, the murine retina. We found distinct transcriptional signatures for retinal microglia across development and peak postnatal density of a population that resembles aging and disease-associated microglia (DAM) and CD11c+ microglia of developing white matter. While TREM2 signaling modulates the expression of select genes, the DAM-related signature is significantly reduced in retinas lacking Bax, a proapoptotic factor required for neuronal death. Furthermore, we found postnatal retinal microglia highly expressing CD11c are resistant to loss or inhibition of colony stimulating factor 1 receptor (CSF1R), while most microglia can be eliminated in Bax knockout retina. Thus, developmental apoptosis promotes a microglia gene signature linked to CSF1R independence that shares features with microglia in developing white matter and in disease.


Asunto(s)
Apoptosis , Diferenciación Celular , Microglía/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Retina/metabolismo , Enfermedades de la Retina/metabolismo , Transducción de Señal , Transcriptoma , Animales , Ratones , Ratones Noqueados , Microglía/patología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Retina/patología , Enfermedades de la Retina/genética , Enfermedades de la Retina/patología
2.
J Neurosci ; 39(11): 2025-2040, 2019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30647151

RESUMEN

Microglia play important roles in shaping the developing CNS, and at early stages they have been proposed to regulate progenitor proliferation, differentiation, and neuronal survival. However, these studies reveal contradictory outcomes, highlighting the complexity of these cell-cell interactions. Here, we investigate microglia function during embryonic mouse retina development, where only microglia, progenitors, and neurons are present. In both sexes, we determine that microglia primarily interact with retinal neurons and find that depletion of microglia via conditional KO of the Csf1 receptor results in increased density of retinal ganglion cells (RGCs). Pharmacological inhibition of microglia also results in an increase in RGCs, with no effect on retinal progenitor proliferation, RGC genesis, or apoptosis. We show that microglia in the embryonic retina are enriched for phagocytic markers and observe engulfment of nonapoptotic Brn3-labeled RGCs. We investigate the molecular pathways that can mediate cell engulfment by microglia and find selective downregulation of complement pathway components with microglia inhibition, and further show that C1q protein marks a subset of RGCs in the embryonic retina. KO of complement receptor 3 (CR3; Itgam), which is only expressed by microglia, results in increased RGC density, similar to what we observed after depletion or inhibition of microglia. Thus, our data suggest that microglia regulate neuron elimination in the embryonic mouse retina by complement-mediated phagocytosis of non-apoptotic newborn RGCs.SIGNIFICANCE STATEMENT Microglia are emerging as active and important participants in regulating neuron number in development, during adult neurogenesis, and following stem cell therapies. However, their role in these contexts and the mechanisms involved are not fully defined. Using a well-characterized in vivo system, we provide evidence that microglia regulate neuronal elimination by complement-mediated engulfment of nonapoptotic neurons. This work provides a significant advancement of the field by defining in vivo molecular mechanisms for microglia-mediated cell elimination. Our data add to a growing body of evidence that microglia are essential for proper nervous system development. In addition, we elucidate microglia function in the developing retina, which may shed light on microglia involvement in the context of retinal injury and disease.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Microglía/fisiología , Fagocitosis/fisiología , Retina/crecimiento & desarrollo , Células Ganglionares de la Retina/fisiología , Animales , Recuento de Células , Femenino , Factor Estimulante de Colonias de Macrófagos/genética , Masculino , Ratones Noqueados
3.
Mol Ther ; 26(10): 2379-2396, 2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30217731

RESUMEN

Dysregulation of the complement system is implicated in neurodegeneration, including human and animal glaucoma. Optic nerve and retinal damage in glaucoma is preceded by local complement upregulation and activation, but whether targeting this early innate immune response could have therapeutic benefit remains undefined. Because complement signals through three pathways that intersect at complement C3 activation, here we targeted this step to restore complement balance in the glaucomatous retina and to determine its contribution to degeneration onset and/or progression. To achieve this, we combined adeno-associated virus retinal gene therapy with the targeted C3 inhibitor CR2-Crry. We show that intravitreal injection of AAV2.CR2-Crry produced sustained Crry overexpression in the retina and reduced deposition of the activation product complement C3d on retinal ganglion cells and the inner retina of DBA/2J mice. This resulted in neuroprotection of retinal ganglion cell axons and somata despite continued intraocular pressure elevation, suggesting a direct restriction of neurodegeneration onset and progression and significant delay to terminal disease stages. Our study uncovers a damaging effect of complement C3 or downstream complement activation in glaucoma, and it establishes AAV2.CR2-Crry as a viable therapeutic strategy to target pathogenic C3-mediated complement activation in the glaucomatous retina.


Asunto(s)
Complemento C3/genética , Glaucoma/terapia , Degeneración Nerviosa/terapia , Proteínas Recombinantes de Fusión/genética , Animales , Complemento C3/antagonistas & inhibidores , Dependovirus/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Terapia Genética , Glaucoma/genética , Glaucoma/patología , Humanos , Presión Intraocular/efectos de los fármacos , Inyecciones Intravítreas , Ratones , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Proteínas Recombinantes de Fusión/administración & dosificación , Retina/efectos de los fármacos , Retina/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología
4.
Front Neurosci ; 10: 526, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27932942

RESUMEN

Neurodegeneration in glaucoma results in decline and loss of retinal ganglion cells (RGCs), and is associated with activation of myeloid cells such as microglia and macrophages. The chemokine fractalkine (FKN or Cx3cl1) mediates communication from neurons to myeloid cells. Signaling through its receptor Cx3cr1 has been implicated in multiple neurodegenerative diseases, but the effects on neuronal pathology are variable. Since it is unknown how FKN-mediated crosstalk influences RGC degeneration in glaucoma, we assessed this in a chronic mouse model, DBA/2J. We analyzed a DBA/2J substrain deficient in Cx3cr1, and compared compartmentalized RGC degeneration and myeloid cell responses to those in standard DBA/2J mice. We found that loss of FKN signaling exacerbates axon transport dysfunction, an early event in neurodegeneration, with a significant increase in RGCs with somal accumulation of the axonal protein phosphorylated neurofilament, and reduced retinal expression of genes involved in axon transport, Kif1b, and Atp8a2. There was no change in the loss of Brn3-positive RGCs, and no difference in the extent of damage to the proximal optic nerve, suggesting that the loss of fractalkine signaling primarily affects axon transport. Since Cx3cr1 is specifically expressed in myeloid cells, we assessed changes in retinal microglial number and activation, changes in gene expression, and the extent of macrophage infiltration. We found that loss of fractalkine signaling led to innate immune changes within the retina, including increased infiltration of peripheral macrophages and upregulated nitric oxide synthase-2 (Nos-2) expression in myeloid cells, which contributes to the production of NO and can promote axon transport deficits. In contrast, resident retinal microglia appeared unchanged either in number, morphology, or expression of the myeloid activation marker ionized calcium binding adaptor molecule 1 (Iba1). There was also no significant increase in the proinflammatory gene interleukin 1 beta (Il1ß). We conclude that loss of fractalkine signaling causes a selective worsening of axon transport dysfunction in RGCs, which is linked to enhanced Nos-2 expression in myeloid cells. Our findings suggest that distinct mechanisms may contribute to different aspects of RGC decline in glaucoma, with axonal transport selectively altered after loss of Cx3cr1 in microglia and/or macrophages.

5.
Exp Eye Res ; 150: 34-43, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26851485

RESUMEN

Within the white matter, axonal loss by neurodegeneration is coupled to glial cell changes in gene expression, structure and function commonly termed gliosis. Recently, we described the highly variable expansion of gliosis alebosco@neuro.utah.edu in degenerative optic nerves from the DBA/2J mouse model of chronic, age-related glaucoma. Here, to estimate and compare the levels of axonal loss with the expansion of glial coverage and axonal degeneration in DBA/2J nerves, we combined semiautomatic axon counts with threshold-based segmentation of total glial/scar areas and degenerative axonal profiles in plastic cross-sections. In nerves ranging from mild to severe degeneration, we found that the progression of axonal dropout is coupled to an increase of gliotic area. We detected a strong correlation between axon loss and the aggregate coverage by glial cells and scar, whereas axon loss did not correlate with the small fraction of degenerating profiles. Nerves with low to medium levels of axon loss displayed moderate glial reactivity, consisting of hypertrophic astrocytes, activated microglia and normal distribution of oligodendrocytes, with minimal reorganization of the tissue architecture. In contrast, nerves with extensive axonal loss showed prevalent rearrangement of the nerve, with loss of axon fascicle territories and enlarged or almost continuous gliotic and scar domains, containing reactive astrocytes, oligodendrocytes and activated microglia. These findings support the value of optic nerve gliotic expansion as a quantitative estimate of optic neuropathy that correlates with axon loss, applicable to grade the severity of optic nerve damage in mouse chronic glaucoma.


Asunto(s)
Glaucoma/patología , Gliosis/complicaciones , Neuroglía/patología , Enfermedades del Nervio Óptico/patología , Nervio Óptico/patología , Células Ganglionares de la Retina/patología , Animales , Astrocitos/patología , Axones/patología , Enfermedad Crónica , Modelos Animales de Enfermedad , Femenino , Glaucoma/complicaciones , Gliosis/patología , Masculino , Ratones , Ratones Endogámicos DBA , Microscopía Confocal , Enfermedades del Nervio Óptico/etiología , Fotomicrografía
6.
Dis Model Mech ; 8(5): 443-55, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25755083

RESUMEN

Microglia serve key homeostatic roles, and respond to neuronal perturbation and decline with a high spatiotemporal resolution. The course of all chronic CNS pathologies is thus paralleled by local microgliosis and microglia activation, which begin at early stages of the disease. However, the possibility of using live monitoring of microglia during early disease progression to predict the severity of neurodegeneration has not been explored. Because the retina allows live tracking of fluorescent microglia in their intact niche, here we investigated their early changes in relation to later optic nerve neurodegeneration. To achieve this, we used the DBA/2J mouse model of inherited glaucoma, which develops progressive retinal ganglion cell degeneration of variable severity during aging, and represents a useful model to study pathogenic mechanisms of retinal ganglion cell decline that are similar to those in human glaucoma. We imaged CX3CR1(+/GFP) microglial cells in vivo at ages ranging from 1 to 5 months by confocal scanning laser ophthalmoscopy (cSLO) and quantified cell density and morphological activation. We detected early microgliosis at the optic nerve head (ONH), where axonopathy first manifests, and could track attenuation of this microgliosis induced by minocycline. We also observed heterogeneous and dynamic patterns of early microglia activation in the retina. When the same animals were aged and analyzed for the severity of optic nerve pathology at 10 months of age, we found a strong correlation with the levels of ONH microgliosis at 3 to 4 months. Our findings indicate that live imaging and monitoring the time course and levels of early retinal microgliosis and microglia activation in glaucoma could serve as indicators of future neurodegeneration severity.


Asunto(s)
Glaucoma/patología , Microglía/patología , Degeneración Nerviosa/patología , Animales , Receptor 1 de Quimiocinas CX3C , Modelos Animales de Enfermedad , Femenino , Gliosis/patología , Rayos Láser , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Oftalmoscopía , Disco Óptico/patología , Receptores de Quimiocina/metabolismo , Retina/patología
7.
PLoS One ; 7(8): e43602, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22952717

RESUMEN

Glaucoma is a neurodegenerative disease that results in the progressive decline and ultimate death of retinal ganglion cells (RGCs). While multiple risk factors are associated with glaucoma, the mechanisms leading to onset and progression of the disease remain unknown. Molecular analysis in various glaucoma models has revealed involvement of non-neuronal cell populations, including astrocytes, Mueller glia and microglia, at early stages of glaucoma. High-dose irradiation was reported to have a significant long-term protective effect in the DBA/2J (D2) mouse model of glaucoma, although the cellular and molecular basis for this effect remains unclear. In particular, the acute effects of irradiation on specific cell populations, including non-neuronal cells, in the D2 retina and nerve have not been assessed. Here we report that irradiation induces transient reduction in proliferating microglia within the optic nerve head and glial lamina within the first week post-irradiation. This was accompanied by reduced microglial activation, with no effect on astrocyte gliosis in those regions. At later stages we confirm that early high-dose irradiation of the mouse head results in improvement of axonal structural integrity and anterograde transport function, without reduction of intraocular pressure. Thus reduced microglial activation induced by irradiation at early stages is associated with reduced optic nerve and retinal neurodegeneration in the D2 mouse model of glaucoma.


Asunto(s)
Glaucoma/patología , Microglía/patología , Microglía/efectos de la radiación , Animales , Axones/patología , Axones/efectos de la radiación , Proliferación Celular/efectos de la radiación , Enfermedad Crónica , Modelos Animales de Enfermedad , Relación Dosis-Respuesta en la Radiación , Femenino , Gliosis/patología , Masculino , Ratones , Ratones Endogámicos DBA , Nervio Óptico/patología , Nervio Óptico/efectos de la radiación , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/efectos de la radiación , Factores de Tiempo
8.
J Comp Neurol ; 519(4): 599-620, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21246546

RESUMEN

Changes in microglial cell activation and distribution are associated with neuronal decline in the central nervous system (CNS), particularly under pathological conditions. Activated microglia converge on the initial site of axonal degeneration in human glaucoma, yet their part in its pathophysiology remains unresolved. To begin with, it is unknown whether microglia activation precedes or is a late consequence of retinal ganglion cell (RGC) neurodegeneration. Here we address this critical element in DBA/2J (D2) mice, an established model of chronic inherited glaucoma, using as a control the congenic substrain DBA/2J Gpnmb(+/SjJ) (D2G), which is not affected by glaucoma. We analyzed the spatial distribution and timecourse of microglial changes in the retina, as well as within the proximal optic nerve prior to and throughout ages when neurodegeneration has been reported. Exclusively in D2 mice, we detected early microglia clustering in the inner central retina and unmyelinated optic nerve regions, with microglia activation peaking by 3 months of age. Between 5 and 8 months of age, activated microglia persisted and concentrated in the optic disc, but also localized to the retinal periphery. Collectively, our findings suggest microglia activation is an early alteration in the retina and optic nerve in D2 glaucoma, potentially contributing to disease onset or progression. Ultimately, detection of microglial activation may have value in early disease diagnosis, while modulation of microglial responses may alter disease progression.


Asunto(s)
Glaucoma/fisiopatología , Microglía/fisiología , Animales , Receptor 1 de Quimiocinas CX3C , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Presión Intraocular , Ratones , Ratones Endogámicos DBA , Ratones Transgénicos , Microglía/citología , Nervio Óptico/citología , Nervio Óptico/crecimiento & desarrollo , Nervio Óptico/fisiopatología , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Retina/citología , Retina/crecimiento & desarrollo , Retina/fisiopatología
9.
Invest Ophthalmol Vis Sci ; 49(4): 1437-46, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18385061

RESUMEN

PURPOSE: In the context of the retinal ganglion cell (RGC) axon degeneration in the optic nerve that occurs in glaucoma, microglia become activated, then phagocytic, and redistribute in the optic nerve head. The authors investigated the potential contribution of retinal microglia activation to glaucoma progression in the DBA/2J chronic mouse glaucoma model. METHODS: The authors treated 6-week-old DBA/2J mice for 25 weeks with minocycline, a tetracycline derivative known to reduce microglia activation and to improve neuronal survival in other models of neurodegenerative disease. They quantified RGC numbers and characterized microglia activation, gliosis, and both axonal integrity and retrograde tracer transport by RGCs in mice systemically treated with minocycline or vehicle only. RESULTS: Minocycline reduced microglial activation and improved RGC axonal transport and integrity, yet it had no effect on the characteristic age-related ocular changes that lead to chronically elevated pressure and did not alter Müller or astrocyte gliosis. Specifically, minocycline increased the fraction of microglia with resting ramified morphology and reduced levels of Iba1 mRNA and protein, a microglia-specific calcium ligand linked to activation. The reduction in microglial activation was coupled to significant improvement in RGC axonal transport, as measured by neuronal retrograde tracing from the superior colliculus. Finally, minocycline treatment significantly decoupled RGC axon loss from increased intraocular pressure. CONCLUSIONS: These observations suggest that in glaucoma, retina and optic nerve head microglia activation may be a factor in the early decline in function of the optic nerve and its subsequent degeneration.


Asunto(s)
Modelos Animales de Enfermedad , Glaucoma/prevención & control , Microglía/efectos de los fármacos , Minociclina/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Enfermedades del Nervio Óptico/metabolismo , Retina/citología , Animales , Transporte Axonal/efectos de los fármacos , Proteínas de Unión al Calcio/genética , Supervivencia Celular , Glaucoma/metabolismo , Glaucoma/patología , Gliosis/metabolismo , Inyecciones Intraperitoneales , Presión Intraocular , Espectrometría de Masas , Ratones , Ratones Endogámicos DBA , Proteínas de Microfilamentos , Microglía/metabolismo , Enfermedades del Nervio Óptico/patología , ARN Mensajero/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
J Neurosci ; 28(11): 2735-44, 2008 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-18337403

RESUMEN

Glaucoma is characterized by retinal ganglion cell (RGC) pathology and a progressive loss of vision. Previous studies suggest RGC death is responsible for vision loss in glaucoma, yet evidence from other neurodegenerative diseases suggests axonal degeneration, in the absence of neuronal loss, can significantly affect neuronal function. To characterize RGC degeneration in the DBA/2 mouse model of glaucoma, we quantified RGCs in mice of various ages using neuronal-specific nuclear protein (NeuN) immunolabeling, retrograde labeling, and optic nerve axon counts. Surprisingly, the number of NeuN-labeled RGCs did not decline significantly until 18 months of age, at which time a significant decrease in RGC somal size was also observed. Axon dysfunction and degeneration occurred before loss of NeuN-positive RGCs, because significant declines in RGC number assayed by retrograde tracers and axon counts were observed at 13 months. To examine whether axonal dysfunction/degeneration affected gene expression in RGC axons or somas, NeuN and neurofilament-heavy (NF-H) immunolabeling was performed along with quantitative reverse transcription-PCR for RGC-specific genes in retinas of aged DBA/2 mice. Although these mice had similar numbers of NeuN-positive RGCs, the expression of neurofilament light, Brn-3b, and Sncg mRNA varied; this variation in RGC-specific gene expression was correlated with the appearance of NF-H immunoreactive RGC axons. Together, these data support a progression of RGC degeneration in this model of glaucoma, beginning with loss of retrograde label, where axon dysfunction and degeneration precede neuronal loss. This progression of degeneration suggests a need to examine the RGC axon as a locus of pathology in glaucoma.


Asunto(s)
Modelos Animales de Enfermedad , Glaucoma/patología , Degeneración Nerviosa/patología , Neuronas/patología , Células Ganglionares de la Retina/patología , Animales , Recuento de Células/métodos , Muerte Celular/fisiología , Progresión de la Enfermedad , Glaucoma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Degeneración Nerviosa/genética , Neuronas/fisiología , Células Ganglionares de la Retina/fisiología
11.
J Neurosci ; 28(2): 548-61, 2008 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-18184797

RESUMEN

Little is known about molecular changes occurring within retinal ganglion cells (RGCs) before their death in glaucoma. Taking advantage of the fact that gamma-synuclein (Sncg) mRNA is expressed specifically and highly in adult mouse RGCs, we show in the DBA/2J mouse model of glaucoma that there is not only a loss of cells expressing this gene, but also a downregulation of gene expression of Sncg and many other genes within large numbers of RGCs. This downregulation of gene expression within RGCs occurs together with reductions in FluoroGold (FG) retrograde transport. Surprisingly, there are also large numbers of Sncg-expressing cells without any FG labeling, and among these many that have a marker previously associated with disconnected RGCs, accumulation of phosphorylated neurofilaments in their somas. These same diseased retinas also have large numbers of RGCs that maintain the intraocular portion while losing the optic nerve portion of their axons, and these disconnected axons terminate within the optic nerve head. Our data support the view that RGC degeneration in glaucoma has two separable stages: the first involves atrophy of RGCs, whereas the second involves an insult to axons, which causes the degeneration of axon portions distal to the optic nerve head but does not cause the immediate degeneration of intraretinal portions of axons or the immediate death of RGCs.


Asunto(s)
Axones/patología , Regulación hacia Abajo/fisiología , Glaucoma/patología , Disco Óptico/patología , Disco Óptico/fisiopatología , Células Ganglionares de la Retina/fisiología , Factores de Edad , Animales , Axotomía/métodos , Modelos Animales de Enfermedad , Glaucoma/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Degeneración Nerviosa/etiología , Degeneración Nerviosa/fisiopatología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero/metabolismo , Especificidad de la Especie , Estilbamidinas/metabolismo , gamma-Sinucleína/genética , gamma-Sinucleína/metabolismo
12.
Invest Ophthalmol Vis Sci ; 47(3): 977-85, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16505032

RESUMEN

PURPOSE: The DBA/2J mouse is a model for secondary angle-closure glaucoma, due to iris atrophy and pigment dispersion, which ultimately lead to increased intraocular pressure (IOP). The study was undertaken to correlate changes in retinal gene expression with IOP elevation by performing microarray analysis of retinal RNA from DBA/2J mice at 3 months before disease onset and at 8 months after IOP elevation. METHODS: IOP was monitored monthly in DBA/2J animals, and animals with normal (3 months) or elevated IOP (8 months) were identified. RNA was prepared from three individual retinas at each age, and the RNA was amplified and used to generate biotin-labeled probe for high-density mouse gene microarrays (U430.2; Affymetrix, Santa Clara, CA). A subset of genes was selected for confirmation by quantitative RT-PCR, by using independent retina samples from DBA/2J animals at 3, 5, and 8 months of age and compared to retinas from C57BL/6J control animals at 3 and 8 months. RESULTS: There were changes in expression of 68 genes, with 32 genes increasing and 36 genes decreasing at 8 months versus 3 months. Upregulated genes were associated with immune response, glial activation, signaling, and gene expression, whereas downregulated genes included multiple crystallin genes. Significant changes in nine upregulated genes and two downregulated genes were confirmed by quantitative RT-PCR, with some showing changes in expression by 5 months. CONCLUSIONS: DBA/2J retina shows evidence of glial activation and an immune-related response after IOP elevation, similar to what has been reported after acute elevation of IOP in other models.


Asunto(s)
Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Expresión Génica , Glaucoma de Ángulo Cerrado/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Retina/metabolismo , Animales , Femenino , Presión Intraocular , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tonometría Ocular , Regulación hacia Arriba
13.
Dev Dyn ; 234(3): 802-7, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16217738

RESUMEN

Synaptic vesicle-associated proteins are important regulators of neurotransmitter release at synaptic terminals in mature animals. Some synaptic vesicle-associated proteins are also expressed during development, although their contribution to development is not as clear. Here, we describe the cloning and developmental expression pattern of the Xenopus laevis synaptic vesicle-associated protein SVOP, a gene first identified as an immediate target for proneural basic helix-loop-helix factors. Alignment analysis revealed a high level of identity between the SVOP protein sequences from Xenopus and other vertebrates. In developing Xenopus embryos, SVOP expression is restricted to the nervous system and is first detectable at the mid-neurula stage. As development progresses SVOP becomes broadly expressed throughout the central nervous system. Our observation that SVOP is expressed in the developing Xenopus nervous system suggests that it may be involved in neuron formation, maturation, or neuronal function.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Proteínas del Tejido Nervioso/metabolismo , Sistema Nervioso/crecimiento & desarrollo , Sistema Nervioso/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/crecimiento & desarrollo , Xenopus laevis/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Filogenia , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Proteínas de Xenopus/química , Proteínas de Xenopus/genética
14.
Dev Biol ; 285(2): 570-83, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16112102

RESUMEN

Proneural basic helix-loop-helix (bHLH) transcription factors are critical positive regulators of neuronal differentiation in a variety of species and are required for proper differentiation of various subtypes of neurons. Although bHLH factors demonstrate some unique functions during neural development, they share the ability to regulate neuronal differentiation, potentially by targeting overlapping sets of genes. To assess this, we performed a screen in ectoderm animal cap tissue to identify direct transcriptional targets shared by two Xenopus ato-related bHLH factors, Xath5 and XNeuroD. Candidate target genes identified in this screen include several transcriptional regulators (Xebf2, Xebf3, XETOR and NKL), an RNA binding protein (elrC), a cell cycle component (Xgadd45gamma) and several novel genes. Overexpression of either Xath5 or XNeuroD induced ectopic in vivo expression of these candidate target genes. Conversely, blocking ato-related bHLH activity prevented endogenous nervous system expression of these genes. Therefore, we have identified a set of genes that can be regulated by multiple ato-related bHLH factors and may function as critical effectors of proneural bHLH-mediated differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ectodermo/metabolismo , Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes/genética , Proteínas del Tejido Nervioso/metabolismo , Sistema Nervioso/embriología , Proteínas de Xenopus/metabolismo , Xenopus/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cartilla de ADN , Proteínas del Ojo/genética , Perfilación de la Expresión Génica , Hibridación in Situ , Proteínas del Tejido Nervioso/genética , Sistema Nervioso/metabolismo , Reacción en Cadena de la Polimerasa , Xenopus/genética , Proteínas de Xenopus/genética
15.
J Biol Chem ; 278(35): 33351-63, 2003 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-12810716

RESUMEN

The cyclic AMP-specific phosphodiesterase (PDE4) isoform PDE4A5 interacted with the immunophilin XAP2 in a yeast two-hybrid assay. The interaction was confirmed in biochemical pull-down analyses. The interaction was specific, in that PDE4A5 did not interact with the closely related immunophilins AIPL1, FKBP51, or FKBP52. XAP2 also did not interact with other PDE4A isoforms or typical isoforms from the three other PDE4 subfamilies. Functionally, XAP2 reversibly inhibited the enzymatic activity of PDE4A5, increased the sensitivity of PDE4A5 to inhibition by the prototypical PDE4 inhibitor 4-[3-(cyclopentyloxy)-4-methoxyphenyl]-2-pyrrolidinone (rolipram) and attenuated the ability of cAMP-dependent protein kinase to phosphorylate PDE4A5 in intact cells. XAP2 maximally inhibited PDE4A5 by approximately 60%, with an IC50 of 120 nm, and reduced the IC50 for rolipram from 390 nm to 70-90 nm. Co-expression of XAP2 and PDE4A5 in COS7 cells showed that they could be co-immunoprecipitated and also reduced both the enzymatic activity of PDE4A5 and its IC50 for rolipram. Native XAP2 and PDE4A5 could be co-immunoprecipitated from the brain. The isolated COOH-terminal half of XAP2 (amino acids 170-330), containing its tetratricopeptide repeat domain, but not the isolated NH2-terminal half (amino acids 1-169), containing the immunophilin homology region, similarly reduced PDE4A5 activity and its IC50 for rolipram. Mutation of Arg271 to alanine, in the XAP2 tetratricopeptide repeat region, attenuated its ability to both interact with PDE4A5 in two-hybrid assays and to inhibit PDE4A5 activity. Either the deletion of a specific portion of the unique amino-terminal region or specific mutations in the regulatory UCR2 domain of PDE4A5 attenuated its ability be inhibited by XAP2. We suggest that XAP2 functionally interacts with PDE4A5 in cells.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/química , AMP Cíclico/metabolismo , Proteínas/química , 1-Metil-3-Isobutilxantina/farmacología , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Alanina/química , Secuencia de Aminoácidos , Animales , Células COS , Clonación Molecular , Colforsina/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Eliminación de Gen , Glutatión Transferasa/metabolismo , Humanos , Immunoblotting , Concentración 50 Inhibidora , Péptidos y Proteínas de Señalización Intracelular , Datos de Secuencia Molecular , Mutación , Sistemas de Lectura Abierta , Fosforilación , Pruebas de Precipitina , Unión Proteica , Isoformas de Proteínas , Estructura Terciaria de Proteína , Proteínas/metabolismo , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Rolipram/farmacología , Saccharomyces cerevisiae/metabolismo , Homología de Secuencia de Aminoácido , Factores de Tiempo , Técnicas del Sistema de Dos Híbridos
16.
BMC Biochem ; 3: 24, 2002 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-12193273

RESUMEN

BACKGROUND: The cyclic AMP specific phosphodiesterase, PDE4D5 interacts with the beta-propeller protein RACK1 to form a signaling scaffold complex in cells. Two-hybrid analysis of truncation and mutant constructs of the unique N-terminal region of the cAMP-specific phosphodiesterase, PDE4D5 were used to define a domain conferring interaction with the signaling scaffold protein, RACK1. RESULTS: Truncation and mutagenesis approaches showed that the RACK1-interacting domain on PDE4D5 comprised a cluster of residues provided by Asn-22/Pro-23/Trp-24/Asn-26 together with a series of hydrophobic amino acids, namely Leu-29, Val-30, Leu-33, Leu-37 and Leu-38 in a 'Leu-Xaa-Xaa-Xaa-Leu' repeat. This was done by 2-hybrid analyses and then confirmed in biochemical pull down analyses using GST-RACK1 and mutant PDE4D5 forms expressed in COS cells. Mutation of Arg-34, to alanine, in PDE4D5 attenuated its interaction with RACK1 both in 2-hybrid screens and in pull down analyses. A 38-mer peptide, whose sequence reflected residues 12 through 49 of PDE4D5, bound to RACK1 with similar affinity to native PDE4D5 itself (Ka circa 6 nM). CONCLUSIONS: The RACK1 Interaction Domain on PDE4D5, that we here call RAID1, is proposed to form an amphipathic helical structure that we suggest may interact with the C-terminal beta-propeller blades of RACK1 in a manner akin to the interaction of the helical G-gamma signal transducing protein with the beta-propeller protein, G-beta.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , AMP Cíclico/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión/fisiología , Células COS , Proteína Adaptadora de Señalización CRADD , Chlorocebus aethiops , AMP Cíclico/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Proteínas de Unión al GTP/genética , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Hidrolasas Diéster Fosfóricas/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Cinasa C Activada , Receptores de Superficie Celular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...