Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO Rep ; 24(8): e56525, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37534581

RESUMEN

Several homeoprotein transcription factors transfer between cells and regulate gene expression, protein translation, and chromatin organization in recipient cells. ENGRAILED-1 is one such homeoprotein expressed in spinal V1 interneurons that synapse on α-motoneurons. Neutralizing extracellular ENGRAILED-1 by expressing a secreted single-chain antibody blocks its capture by spinal motoneurons resulting in α-motoneuron loss and limb weakness. A similar but stronger phenotype is observed in the Engrailed-1 heterozygote mouse, confirming that ENGRAILED-1 exerts a paracrine neurotrophic activity on spinal cord α-motoneurons. Intrathecal injection of ENGRAILED-1 leads to its specific internalization by spinal motoneurons and has long-lasting protective effects against neurodegeneration and weakness. Midbrain dopaminergic neurons express Engrailed-1 and, similarly to spinal cord α-motoneurons, degenerate in the heterozygote. We identify genes expressed in spinal cord motoneurons whose expression changes in mouse Engrailed-1 heterozygote midbrain neurons. Among these, p62/SQSTM1 shows increased expression during aging in spinal cord motoneurons in the Engrailed-1 heterozygote and upon extracellular ENGRAILED-1 neutralization. We conclude that ENGRAILED-1 might regulate motoneuron aging and has non-cell-autonomous neurotrophic activity.


Asunto(s)
Neuronas Motoras , Factores de Transcripción , Ratones , Animales , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Neuronas Motoras/metabolismo , Médula Espinal/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Interneuronas/metabolismo
2.
Neural Regen Res ; 17(3): 690-696, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34380911

RESUMEN

Retinal ganglion cell (RGC) axons provide the only link between the light sensitive and photon transducing neural retina and visual centers of the brain. RGC axon degeneration occurs in a number of blinding diseases and the ability to stimulate axon regeneration from surviving ganglion cells could provide the anatomic substrate for restoration of vision. OTX2 is a homeoprotein transcription factor expressed in the retina and previous studies showed that, in response to stress, exogenous OTX2 increases the in vitro and in vivo survival of RGCs. Here we examined and quantified the effects of OTX2 on adult RGC axon regeneration in vitro and in vivo. The results show that exogenous OTX2 stimulates the regrowth of axons from RGCs in cultures of dissociated adult retinal cells and from explants of adult retinal tissue and that RGCs respond directly to OTX2 as regrowth is observed in cultures of purified adult rat RGCs. Importantly, after nerve crush in vivo, we observed a positive effect of OTX2 on the number of regenerating axons up to the optic chiasm within 14 days post crush and a very modest level of acuity absent in control mice. The effect of OTX2 on RGC survival and regeneration is of potential interest for degenerative diseases affecting this cell type. All animal procedures were approved by the local "Comié d'éιthique en expérimentation animale n°59" and authorization n° 00702.01 delivered March 28, 2014 by the French "Ministére de l'enseignement supérieur et de la recherche".

3.
eNeuro ; 8(5)2021.
Artículo en Inglés | MEDLINE | ID: mdl-34475267

RESUMEN

In the mature mouse retina, Otx2 is expressed in both retinal pigmented epithelium (RPE) and photoreceptor (PR) cells, and Otx2 knock-out (KO) in the RPE alone results in PR degeneration. To study the cell-autonomous function of OTX2 in PRs, we performed PR-specific Otx2 KO (cKO) in adults. As expected, the protein disappears completely from PR nuclei but is still observed in PR inner and outer segments while its level concomitantly decreases in the RPE, suggesting a transfer of OTX2 from RPE to PRs in response to Otx2 ablation in PRs. The ability of OTX2 to transfer from RPE to PRs was verified by viral expression of tagged-OTX2 in the RPE. Transferred OTX2 distributed across the PR cytoplasm, suggesting functions distinct from nuclear transcription regulation. PR-specific Otx2 cKO did not alter the structure of the retina but impaired the translocation of PR arrestin-1 on illumination changes, making mice photophobic. RNA-seq analyses following Otx2 KO revealed downregulation of genes involved in the cytoskeleton that might account for the arrestin-1 translocation defect, and of genes involved in extracellular matrix (ECM) and signaling factors that may participate in the enhanced transfer of OTX2. Interestingly, several RPE-specific OTX2 target genes involved in melanogenesis were downregulated, lending weight to a decrease of OTX2 levels in the RPE following PR-specific Otx2 cKO. Our study reveals a new role of endogenous OTX2 in PR light adaptation and demonstrates the existence of OTX2 transfer from RPE to PR cells, which is increased on PR-specific Otx2 ablation and might participate in PR neuroprotection.


Asunto(s)
Fotofobia , Degeneración Retiniana , Animales , Ratones , Factores de Transcripción Otx/genética , Células Fotorreceptoras , Retina
4.
Neural Regen Res ; 16(10): 2002-2003, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33642378
5.
eNeuro ; 7(5)2020.
Artículo en Inglés | MEDLINE | ID: mdl-32737182

RESUMEN

OTX2 is a homeoprotein transcription factor expressed in photoreceptors and bipolar cells in the retina. OTX2, like many other homeoproteins, transfers between cells and exerts non-cell autonomous effects such as promoting the survival of retinal ganglion cells that do not express the protein. Here we used a genetic approach to target extracellular OTX2 in the retina by conditional expression of a secreted single-chain anti-OTX2 antibody. Compared with control mice, the expression of this antibody by parvalbumin-expressing neurons in the retina is followed by a reduction in visual acuity in 1-month-old mice with no alteration of the retinal structure or cell type number or aspect. The a-waves and b-waves measured by electroretinogram were also indistinguishable from those of control mice, suggesting no functional deficit of photoreceptors and bipolar cells. Mice expressing the OTX2-neutralizing antibody did show a significant doubling in the flicker amplitude and a reduction in oscillatory potential, consistent with a change in inner retinal function. Our results show that interfering in vivo with OTX2 non-cell autonomous activity in the postnatal retina leads to an alteration in inner retinal cell functions and causes a deficit in visual acuity.


Asunto(s)
Factores de Transcripción Otx , Retina , Animales , Electrorretinografía , Ratones , Factores de Transcripción Otx/genética , Células Fotorreceptoras , Factores de Transcripción
6.
eNeuro ; 6(5)2019.
Artículo en Inglés | MEDLINE | ID: mdl-31451602

RESUMEN

Most homeoprotein transcription factors have a highly conserved internalization domain used in intercellular transfer. Internalization of homeoproteins ENGRAILED1 or ENGRAILED2 promotes the survival of adult dopaminergic cells, whereas that of OTX2 protects adult retinal ganglion cells. Here we characterize the in vitro neuroprotective activity of several homeoproteins in response to H2O2 Protection is observed with ENGRAILED1, ENGRAILED2, OTX2, GBX2, and LHX9 on midbrain and striatal embryonic neurons, whereas cell-permeable c-MYC shows no protective effects. Therefore, five homeoproteins belonging to three different classes (ANTENNAPEDIA, PAIRED, and LIM) share the ability to protect embryonic neurons from midbrain and striatum. Because midbrain and striatal neurons do not express the same repertoire of the four proteins, a lack of neuronal specificity together with a general protective activity can be proposed. Interestingly, hEN1 and GBX2 provided protection to primary midbrain astrocytes but not to non-neural cells, including mouse embryo fibroblasts, macrophages or HeLa cells. For the four proteins, protection against cell death correlated with a reduction in the number of H2O2-induced DNA break foci in midbrain and striatal neurons. In conclusion, within the limit of the number of cell types and homeoproteins tested, homeoprotein protection against oxidative stress-induced DNA breaks and death is specific to neurons and astrocytes but shows no homeoprotein or neuronal type specificity.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/biosíntesis , Células-Madre Neurales/metabolismo , Neuroprotección/fisiología , Estrés Oxidativo/fisiología , Animales , Línea Celular Transformada , Células Cultivadas , Pollos , Células Madre Embrionarias/efectos de los fármacos , Femenino , Células HeLa , Proteínas de Homeodominio/antagonistas & inhibidores , Humanos , Peróxido de Hidrógeno/toxicidad , Ratones , Células-Madre Neurales/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Embarazo
8.
Physiol Rev ; 98(4): 1943-1982, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30067157

RESUMEN

The homeoprotein family comprises ~300 transcription factors and was long seen as primarily involved in developmental programs through cell autonomous regulation. However, recent evidence reveals that many of these factors are also expressed in the adult where they exert physiological functions not yet fully deciphered. Furthermore, the DNA-binding domain of most homeoproteins contains two signal sequences allowing their secretion and internalization, thus intercellular transfer. This review focuses on this new-found signaling in cell migration, axon guidance, and cerebral cortex physiological homeostasis and speculates on how it may play important roles in early arealization of the neuroepithelium. It also describes the use of homeoproteins as therapeutic proteins in mouse models of diseases affecting the central nervous system, in particular Parkinson disease and glaucoma.


Asunto(s)
Proteínas de Homeodominio/fisiología , Transducción de Señal/fisiología , Animales , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/fisiología , Epitelio/metabolismo , Epitelio/fisiología , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción/metabolismo
9.
Cell Mol Life Sci ; 72(8): 1433-45, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25432704

RESUMEN

Engrailed is a homeoprotein transcription factor. This family of transcription factors is characterized by their DNA-binding homeodomain and some members, including Engrailed, can transfer between cells and regulate protein translation in addition to gene transcription. Engrailed is intimately involved in the development of the vertebrate visual system. Early expression of Engrailed in dorsal mesencephalon contributes to the development and organization of a visual structure, the optic tectum/superior colliculus. This structure is an important target for retinal ganglion cell axons that carry visual information from the retina. Engrailed regulates the expression of Ephrin axon guidance cues in the tectum/superior colliculus. More recently it has been reported that Engrailed itself acts as an axon guidance cue in synergy with the Ephrin system and is proposed to enhance retinal topographic precision.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Retina/crecimiento & desarrollo , Adenosina Trifosfato/metabolismo , Animales , Proteínas de Homeodominio/química , Mitocondrias/metabolismo , Retina/metabolismo , Transducción de Señal , Colículos Superiores/crecimiento & desarrollo , Colículos Superiores/metabolismo , Factores de Transcripción/metabolismo
10.
Semin Cell Dev Biol ; 35: 165-72, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25042849

RESUMEN

The construction of the brain is a highly regulated process, requiring coordination of various cellular and molecular mechanisms that together ensure the stability of the cerebrum architecture and functions. The mature brain is an organ that performs complex computational operations using specific sensory information from the outside world and this requires precise organization within sensory networks and a separation of sensory modalities during development. We review here the role of homeoproteins in the arealization of the brain according to sensorimotor functions, the micropartition of its cytoarchitecture, and the maturation of its sensory circuitry. One of the most interesting observation about homeoproteins in recent years concerns their ability to act both in a cell-autonomous and non-cell-autonomous manner. The highlights in the present review collectively show how these two modes of action of homeoproteins confer various functions in shaping cortical maps.


Asunto(s)
Encéfalo/fisiología , Proteínas de Homeodominio/fisiología , Modelos Neurológicos , Vías Nerviosas/fisiología , Corteza Visual/fisiología , Animales , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Mapeo Encefálico , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Vías Nerviosas/embriología , Vías Nerviosas/crecimiento & desarrollo , Corteza Somatosensorial/embriología , Corteza Somatosensorial/crecimiento & desarrollo , Corteza Somatosensorial/fisiología , Corteza Visual/embriología , Corteza Visual/crecimiento & desarrollo , Vías Visuales/embriología , Vías Visuales/crecimiento & desarrollo , Vías Visuales/fisiología
11.
Hum Mol Genet ; 23(7): 1742-53, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24234651

RESUMEN

In the human, mutations of OTX2 (Orthodenticle homeobox 2 transcription factor) translate into eye malformations of variable expressivity (even between the two eyes of the same individual) and incomplete penetrance, suggesting the existence of subtle thresholds in OTX2 activity. We have addressed this issue by analyzing retinal structure and function in six mutant mice with graded Otx2 activity: Otx2(+/+), Otx2(+/AA), Otx2(+/GFP), Otx2(AA/AA), Otx2(AA/GFP) and Otx2(GFP/GFP). Null mice (Otx2(GFP/GFP)) fail to develop the head and are embryonic lethal, and compound heterozygous Otx2(AA/GFP) mice show a truncated head and die at birth. All other genotypes develop until adulthood. We analyzed eye structure and visual physiology in the genotypes that develop until adulthood and report that phenotype severity parallels Otx2 activity. Otx2(+/AA) are only mildly affected whereas Otx2(+/GFP) are more affected than Otx2(+/AA) but less than Otx2(AA/AA) mice. Otx2(AA/AA) mice later manifest the most severe defects, with variable expressivity. Electrophysiological and histological analyses of the mouse retina revealed progressive death of bipolar cells and cone photoreceptors that is both Otx2 activity- and age-dependent with the same ranking of phenotypic severity. This study demonstrates the importance of gene dosage in the development of age-dependent pathologies and underscores the fact that small gene dosage differences can cause significant pathological states.


Asunto(s)
Anomalías del Ojo/genética , Factores de Transcripción Otx/genética , Células Bipolares de la Retina/citología , Células Fotorreceptoras Retinianas Conos/citología , Células Horizontales de la Retina/citología , Animales , Diferenciación Celular/genética , Línea Celular , Dosificación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Penetrancia , Agudeza Visual/genética
12.
Pharmacol Rev ; 65(1): 90-104, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23300132

RESUMEN

Homeoproteins constitute a major class of transcription factors active throughout development and in adulthood. Their membrane transduction properties were discovered over 20 years ago, opening an original field of research in the domain of vector peptides and signal transduction. In early development, homeoprotein transfer participates in tissue patterning, cell/axon guidance, and migration. In the axon guidance model, homeoproteins exert their non-cell autonomous activity through the regulation of translation, in particular, that of nuclear-transcribed mitochondrial mRNAs. An important aspect of these studies on patterning and migration is that homeoproteins sensitize the cells to the action of other growth factors, thus cooperating with established signaling pathways. The role of homeoprotein signaling at later developmental stages is also of interest. In particular, the transfer of homeoprotein Otx2 into parvalbumin-expressing inhibitory neurons (PV-cells) in the visual cortex regulates cortical plasticity. The molecular deciphering of the interaction of Otx2 with binding sites at the surface of PV-cells has allowed the development of a specific Otx2 antagonist that reopens plasticity in the adult cortex and cures mice from experimental amblyopia, a neurodevelopmental disease. Finally, the use of homeoproteins as therapeutic proteins in mouse models of glaucoma and Parkinson disease is reviewed. In the latter case, engrailed homeoproteins protect mesencephalic dopaminergic neurons by increasing the local translation of complex I mitochondrial mRNAs. In conclusion, this review synthesizes 20 years of work on the fundamental and potentially translational aspects of homeoprotein signaling.


Asunto(s)
Proteínas de Homeodominio/fisiología , Animales , Axones/fisiología , Proteínas Portadoras/metabolismo , Movimiento Celular , Péptidos de Penetración Celular , Plasticidad Neuronal , Transducción de Señal , Corteza Visual/fisiología
13.
Eur J Neurosci ; 35(12): 1837-45, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22708594

RESUMEN

Several homeoproteins can function in a direct cell non-autonomous fashion to control various biological processes. In the developing nervous system, this mode of signaling has been well documented for Engrailed in the guidance of retinal ganglion cell axons and retino-tectal patterning. Engrailed is also a key factor for mesencephalic dopaminergic (mDA) neurons, not only during development but also in the adult. Haplodeficiency for Engrailed1 leads to progressive adult-onset loss of mDA neurons and several phenotypic alterations reminiscent of Parkinson's disease (PD). Thanks to its transduction properties, Engrailed has been shown to confer neuroprotection in several experimental models of PD. Study of the mechanisms underlying these two Engrailed-mediated effects has revealed a key role of the translation regulation by Engrailed and uncovered an unsuspected link between a homeoprotein and mitochondrial activity. These studies highlight the crucial role of cellular energetic metabolism in neuron development, survival and neurodegeneration, and may help to identify novel therapeutic targets.


Asunto(s)
Axones/fisiología , Neuronas Dopaminérgicas/metabolismo , Proteínas de Homeodominio/metabolismo , Neuronas Retinianas/metabolismo , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiopatología , Supervivencia Celular , Proteínas de Homeodominio/genética , Humanos , Ratones , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Transducción de Señal
14.
Development ; 139(1): 215-24, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22147955

RESUMEN

Engrailed 1 and engrailed 2 homeoprotein transcription factors (collectively Engrailed) display graded expression in the chick optic tectum where they participate in retino-tectal patterning. In vitro, extracellular Engrailed guides retinal ganglion cell (RGC) axons and synergises with ephrin A5 to provoke the collapse of temporal growth cones. In vivo disruption of endogenous extracellular Engrailed leads to misrouting of RGC axons. Here we characterise the signalling pathway of extracellular Engrailed. Our results show that Engrailed/ephrin A5 synergy in growth cone collapse involves adenosine A1 receptor activation after Engrailed-dependent ATP synthesis, followed by ATP secretion and hydrolysis to adenosine. This is, to our knowledge, the first evidence for a role of the adenosine A1 receptor in axon guidance. Based on these results, together with higher expression of the adenosine A1 receptor in temporal than nasal growth cones, we propose a computational model that illustrates how the interaction between Engrailed, ephrin A5 and adenosine could increase the precision of the retinal projection map.


Asunto(s)
Efrina-A5/metabolismo , Conos de Crecimiento/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptor de Adenosina A1/metabolismo , Retina/embriología , Transducción de Señal/fisiología , Adenosina Trifosfato/metabolismo , Animales , Western Blotting , Embrión de Pollo , Técnica del Anticuerpo Fluorescente , Microscopía Fluorescente , Modelos Biológicos , Proteómica , Retina/metabolismo
15.
Nat Neurosci ; 14(10): 1260-6, 2011 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-21892157

RESUMEN

Mice heterozygous for the homeobox gene Engrailed-1 (En1) display progressive loss of mesencephalic dopaminergic (mDA) neurons. We report that exogenous Engrailed-1 and Engrailed-2 (collectively Engrailed) protect mDA neurons from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a mitochondrial complex I toxin used to model Parkinson's disease in animals. Engrailed enhances the translation of nuclearly encoded mRNAs for two key complex I subunits, Ndufs1 and Ndufs3, and increases complex I activity. Accordingly, in vivo protection against MPTP by Engrailed is antagonized by Ndufs1 small interfering RNA. An association between Engrailed and complex I is further confirmed by the reduced expression of Ndufs1 and Ndufs3 in the substantia nigra pars compacta of En1 heterozygous mice. Engrailed also confers in vivo protection against 6-hydroxydopamine and α-synuclein-A30P. Finally, the unilateral infusion of Engrailed into the midbrain increases striatal dopamine content, resulting in contralateral amphetamine-induced turning. Therefore, Engrailed is both a survival factor for adult mDA neurons and a regulator of their physiological activity.


Asunto(s)
Dopamina/metabolismo , Proteínas de Homeodominio/metabolismo , Mesencéfalo/citología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Neurotoxinas/toxicidad , Animales , Recuento de Células/métodos , Células Cultivadas , Cromatografía Líquida de Alta Presión/métodos , Maleato de Dizocilpina/farmacología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Embrión de Mamíferos , Proteínas de Homeodominio/farmacología , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NADH Deshidrogenasa/farmacología , Proteínas del Tejido Nervioso/farmacología , Neuronas/efectos de los fármacos , Nitrocompuestos/toxicidad , Oxidopamina/toxicidad , Propionatos/toxicidad , ARN Interferente Pequeño/farmacología , Rotenona/toxicidad , Conducta Estereotipada/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo , alfa-Sinucleína/metabolismo
16.
FEBS Lett ; 585(11): 1573-8, 2011 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-21565195

RESUMEN

Homeoproteins transcription factors can transfer between cells and play important roles in development. However, some of these homeoproteins are expressed in the adult, but their function is unknown. The loss of mesencephalic dopaminergic (mDA) neurons is the cause of Parkinson's disease. In mice lacking a functional allele for the Engrailed 1 homeoprotein, mDA neurons progressively die starting about 6 weeks after birth. Infusion of recombinant Engrailed stops the death of these neurons demonstrating that homeoproteins can be neuroprotective. This has been extended to retinal ganglion cell neurons (RGCs), which die in glaucoma and optic neuropathies. The homeoprotein Otx2 promotes the survival of injured adult RGCs both in vitro and in vivo. These examples raise the possibility that homeoproteins may provide neuroprotection to neurons vulnerable in other neurodegenerative diseases.


Asunto(s)
Encéfalo/citología , Encéfalo/metabolismo , Proteínas de Homeodominio/metabolismo , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Glaucoma/metabolismo , Glaucoma/patología , Glaucoma/fisiopatología , Proteínas de Homeodominio/genética , Humanos , Factores de Transcripción Otx/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Enfermedad de Parkinson/terapia , Transducción Genética
17.
J Neurosci ; 31(14): 5495-503, 2011 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-21471386

RESUMEN

Retinal ganglion cells (RGCs) are the projection neurons from the eye to the brain and their loss results in visual impairment in a number of diseases. Transcription factors with a homeodomain can translocate between cells and, in at least one reported case, can stimulate neuronal survival. Otx2 is a homeoprotein transcription factor expressed in the retina that is taken up by RGCs. We thus hypothesized that Otx2 capture could regulate the survival of adult RGCs. We report that Otx2 stimulates the survival of adult mouse and rat RGCs in vitro and protects RGCs against NMDA-induced toxicity in vivo in mice. In the latter model, Otx2 also preserves visual acuity.


Asunto(s)
Fármacos Neuroprotectores/farmacología , Factores de Transcripción Otx/farmacología , Células Ganglionares de la Retina/efectos de los fármacos , Agudeza Visual/efectos de los fármacos , Análisis de Varianza , Animales , Recuento de Células/métodos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , N-Metilaspartato/toxicidad , Proteínas de Neurofilamentos/metabolismo , Neurotoxinas/toxicidad , Oligopéptidos/farmacología , Optometría/métodos , Factores de Transcripción Otx/genética , ARN Mensajero/metabolismo , Ratas , Ratas Long-Evans , Retina/citología , Células Ganglionares de la Retina/metabolismo , Factores de Tiempo , Factor de Transcripción Brn-3A/genética , Factor de Transcripción Brn-3A/metabolismo , Agudeza Visual/fisiología
18.
J Mol Neurosci ; 34(1): 9-15, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18157653

RESUMEN

The cellular prion protein (PrP(C)) is found prominently at the synapse. However, its role at the nerve termini and elsewhere is unknown. Here we discuss research presented at the 2005 International Institute for Complex Adaptive Matter (I2CAM) first Annual Amyloid Conference that provides insight into the role of synaptic PrP(C). The prion protein can interact and facilitate copper uptake at the synapse, is presumed to oligodimerize to facilitate putative cell-cell adhesion, and it transports toward the synapse by fast microtubule-based anterograde transport. While PrP(C) appears to be involved in all these processes, the mechanisms of PrP(C) function in each of them remain unclear. A role for PrP(C) in these distinct processes suggests a complex role for this protein at the synapse. Unraveling PrP(C) function will likely entail employing combined approaches that take into account its possible multifaceted functions.


Asunto(s)
Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , Plasticidad Neuronal/fisiología , Proteínas PrPC/metabolismo , Sinapsis/metabolismo , Animales , Transporte Axonal/fisiología , Adhesión Celular/fisiología , Cobre/metabolismo , Humanos , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Sistema Nervioso/citología , Enfermedades por Prión/metabolismo , Enfermedades por Prión/fisiopatología , Sinapsis/ultraestructura
19.
J Comp Neurol ; 504(6): 646-58, 2007 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-17722030

RESUMEN

Cellular prion protein (PrP(c)) is a cell surface glycoprotein highly expressed in neurons, and a protease-resistant conformer of the protein accumulates in the brain parenchyma in prion diseases. In human prion diseases, visual cortex and visual function can be affected. We examined both the levels and the localization of PrP(c) in developing visual cortex of the common marmoset. Western blot analysis showed that PrP(c) increased from the day of birth through adulthood, and this increase correlated with the progression of synapse formation. Immunohistochemistry showed that PrP(c) was present in fiber tracts of the neonate, and this immunoreactivity was lost with maturation. Within the neuropil, the laminar distribution of PrP(c) changed with age. In the neonate, PrP(c) immunoreactivity was strongest in layer 1, where the earliest synapses form. At the end of the first postnatal week, layer 4C, as identified by its strong cytochrome oxidase activity, was noticeably lighter in terms of PrP(c) immunoreactivity than the adjacent layers. The contrast between the strong immunoreactivity in both supragranular and infragranular layers and weak immunoreactivity in layer 4C increased with age. Layers 2/3 and 5 contained more intense PrP(c) immunoreactivity; these layers receive thalamic input from the koniocellular division of the LGN, and these layers of the LGN also had strong PrP(c) immunoreactivity. Together, these results provide evidence for PrP(c) localization in an identified functional pathway and may shed some light on prion disease pathogenesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas PrPC/metabolismo , Primates/metabolismo , Corteza Visual/crecimiento & desarrollo , Corteza Visual/metabolismo , Animales , Animales Recién Nacidos , Callithrix , Macaca fascicularis , Primates/anatomía & histología
20.
Biol Chem ; 387(3): 297-300, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16542151

RESUMEN

A key molecular event in prion diseases is the conversion of cellular prion protein (PrP(c)) into an abnormal misfolded conformer (PrP(sc)). The PrP(c) N-terminal domain plays a central role in PrP(c) functions and in prion propagation. Because mammalian PrP(c) is found as a full-length and N-terminally truncated form, we examined the presence and amount of PrP(c) C-terminal fragment in the brain of different species. We found important variations between primates and rodents. In addition, our data show that the PrP(c) fragment is present in detergent-resistant raft domains, a membrane domain of critical importance for PrP(c) functions and its conversion into PrP(sc).


Asunto(s)
Encéfalo/metabolismo , Membrana Celular/metabolismo , Microdominios de Membrana/metabolismo , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Enfermedades por Prión/metabolismo , Animales , Cricetinae , Detergentes/farmacología , Electroforesis en Gel de Poliacrilamida , Papio , Proteínas PrPC/química , Proteínas PrPC/genética , Proteínas PrPSc/química , Proteínas PrPSc/genética , Primates , Enfermedades por Prión/etiología , Roedores , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...