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












Intervalo de año de publicación
1.
Sci Rep ; 13(1): 5665, 2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-37024714

RESUMEN

Purkinje cell (PC) loss occurs at an early age in patients and animal models of Niemann-Pick Type C (NPC), a lysosomal storage disease caused by mutations in the Npc1 or Npc2 genes. Although degeneration of PCs occurs early in NPC, little is known about how NPC1 deficiency affects the postnatal development of PCs. Using the Npc1nmf164 mouse model, we found that NPC1 deficiency significantly affected the postnatal development of PC dendrites and synapses. The developing dendrites of Npc1nmf164 PCs were significantly deficient in mitochondria and lysosomes. Furthermore, anabolic (mTORC1) and catabolic (TFEB) signaling pathways were not only perturbed but simultaneously activated in NPC1-deficient PCs, suggesting a loss of metabolic balance. We also found that mice with conditional heterozygous deletion of the Phosphatase and Tensin Homolog Deleted on Chromosome 10 gene (Pten-cHet), an inhibitor of mTORC1, showed similar early dendritic alterations in PCs to those found in Npc1-deficient mice. However, in contrast to Npc1nmf164 mice, Pten-cHet mice exhibited the overactivation of the mTORC1 pathway but with a strong inhibition of TFEB signaling, along with no dendritic mitochondrial reductions by the end of their postnatal development. Our data suggest that disruption of the lysosomal-metabolic signaling in PCs causes dendritic and synaptic developmental deficits that precede and promote their early degeneration in NPC.


Asunto(s)
Enfermedad de Niemann-Pick Tipo C , Células de Purkinje , Ratones , Animales , Células de Purkinje/metabolismo , Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Animales de Enfermedad , Lisosomas/metabolismo
2.
PLoS One ; 16(11): e0255196, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34739478

RESUMEN

Retinoic acid (RA) plays major roles during nervous system development, and during regeneration of the adult nervous system. We have previously shown that components of the RA signaling pathway are upregulated after optic nerve injury, and that exogenous application of all-trans retinoic acid (ATRA) greatly increases the survival of axotomized retinal ganglion cells (RGCs). The objective of the present study is to investigate the effects of ATRA application on the macrophages in the optic nerve after injury, and to determine whether this affects axonal regeneration. The optic nerve was crushed and treated with PBS, ATRA and/or clodronate-loaded liposomes. Nerves were examined at one and two weeks after axotomy with light microscopy, immunocytochemistry and electron microscopy. ATRA application to the optic nerve caused transient increases in the number of macrophages and microglia one week after injury. The macrophages are consistently labeled with M2-type markers, and have considerable phagocytic activity. ATRA increased ultrastructural features of ongoing phagocytic activity in macrophages at one and two weeks. ATRA treatment also significantly increased the numbers of regenerating GAP-43-labeled axons. Clodronate liposome treatment depleted macrophage numbers by 80%, completely eliminated the ATRA-mediated increase in axonal regeneration, and clodronate treatment alone decreased axonal numbers by 30%. These results suggest that the success of axon regeneration is partially dependent on the presence of debris-phagocytosing macrophages, and that the increases in regeneration caused by ATRA are in part due to their increased numbers. Further studies will examine whether macrophage depletion affects RGC survival.


Asunto(s)
Macrófagos/efectos de los fármacos , Regeneración Nerviosa/efectos de los fármacos , Traumatismos del Nervio Óptico/tratamiento farmacológico , Células Ganglionares de la Retina/efectos de los fármacos , Tretinoina/farmacología , Animales , Liposomas , Traumatismos del Nervio Óptico/fisiopatología , Rana pipiens , Células Ganglionares de la Retina/fisiología , Tretinoina/uso terapéutico
3.
Sci Rep ; 9(1): 14722, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31605022

RESUMEN

Niemann Pick Type-C disease (NPC) is an inherited lysosomal storage disease (LSD) caused by pathogenic variants in the Npc1 or Npc2 genes that lead to the accumulation of cholesterol and lipids in lysosomes. NPC1 deficiency causes neurodegeneration, dementia and early death. Cerebellar Purkinje cells (PCs) are particularly hypersensitive to NPC1 deficiency and degenerate earlier than other neurons in the brain. Activation of microglia is an important contributor to PCs degeneration in NPC. However, the mechanisms by which activated microglia promote PCs degeneration in NPC are not completely understood. Here, we are demonstrating that in the Npc1nmf164 mouse cerebellum, microglia in the molecular layer (ML) are activated and contacting dendrites at early stages of NPC, when no loss of PCs is detected. During the progression of PCs degeneration in Npc1nmf164 mice, accumulation of phagosomes and autofluorescent material in microglia at the ML coincided with the degeneration of dendrites and PCs. Feeding Npc1nmf164 mice a western diet (WD) increased microglia activation and corresponded with a more extensive degeneration of dendrites but not PC somata. Together our data suggest that microglia contribute to the degeneration of PCs by interacting, engulfing and phagocytosing their dendrites while the cell somata are still present.


Asunto(s)
Dendritas/metabolismo , Microglía/metabolismo , Degeneración Nerviosa/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Células de Purkinje/metabolismo , Animales , Cerebelo/metabolismo , Cerebelo/patología , Dieta Occidental , Modelos Animales de Enfermedad , Femenino , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/genética , Proteína Niemann-Pick C1 , Fagocitosis/genética , Fagosomas/metabolismo
4.
PLoS One ; 14(5): e0209733, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31048836

RESUMEN

We have previously shown that a single application of the growth factors ciliary neurotrophic factor (CNTF) or fibroblast growth factor 2 (FGF-2) to the crushed optic nerve of the frog, Rana pipiens, increases the numbers and elongation rate of regenerating retinal ganglion cell axons. Here we investigate the effects of these factors on the numbers and types of macrophages that invade the regeneration zone. In control PBS-treated nerves, many macrophages are present 100 µm distal to the crush site at 1 week after injury; their numbers halve by 2 weeks. A single application of CNTF at the time of injury triples the numbers of macrophages at 1 week, with this increase compared to control being maintained at 2 weeks. Application of FGF-2 is equally effective at 1 week, but the macrophage numbers have fallen to control levels at 2 weeks. Immunostaining with a pan-macrophage marker, ED1, and a marker for M2-like macrophages, Arg-1, showed that the proportion of the putative M2 phenotype remained at approximately 80% with all treatments. Electron microscopy of the macrophages at 1 week shows strong phagocytic activity with all treatments, with many vacuoles containing axon fragments and membrane debris. At 2 weeks with PBS or FGF-2 treatment the remaining macrophages are less phagocytically active, containing mainly lipid inclusions. With CNTF treatment, at 2 weeks many of the more numerous macrophages are still phagocytosing axonal debris, although they also contain lipid inclusions. We conclude that the increase in macrophage influx seen after growth factor application is beneficial for the regenerating axons, probably due to more extensive removal of degenerating distal axons, but also perhaps to secretion of growth-promoting substances.


Asunto(s)
Factor Neurotrófico Ciliar/farmacología , Factor Neurotrófico Ciliar/uso terapéutico , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/uso terapéutico , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Traumatismos del Nervio Óptico/tratamiento farmacológico , Traumatismos del Nervio Óptico/metabolismo , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Axones/ultraestructura , Inmunohistoquímica , Microscopía Electrónica , Rana pipiens , Vacuolas/efectos de los fármacos , Vacuolas/metabolismo , Vacuolas/ultraestructura
5.
PLoS One ; 11(9): e0162626, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27611191

RESUMEN

After lesions to the mammalian optic nerve, the great majority of retinal ganglion cells (RGCs) die before their axons have even had a chance to regenerate. Frog RGCs, on the other hand, suffer only an approximately 50% cell loss, and we have previously investigated the mechanisms by which the application of growth factors can increase their survival rate. Retinoic acid (RA) is a vitamin A-derived lipophilic molecule that plays major roles during development of the nervous system. The RA signaling pathway is also present in parts of the adult nervous system, and components of it are upregulated after injury in peripheral nerves but not in the CNS. Here we investigate whether RA signaling affects long-term RGC survival at 6 weeks after axotomy. Intraocular injection of all-trans retinoic acid (ATRA), the retinoic acid receptor (RAR) type-α agonist AM80, the RARß agonist CD2314, or the RARγ agonist CD1530, returned axotomized RGC numbers to almost normal levels. On the other hand, inhibition of RA synthesis with disulfiram, or of RAR receptors with the pan-RAR antagonist Ro-41-5253, or the RARß antagonist LE135E, greatly reduced the survival of the axotomized neurons. Axotomy elicited a strong activation of the MAPK, STAT3 and AKT pathways; this activation was prevented by disulfiram or by RAR antagonists. Finally, addition of exogenous ATRA stimulated the activation of the first two of these pathways. Future experiments will investigate whether these strong survival-promoting effects of RA are mediated via the upregulation of neurotrophins.


Asunto(s)
Regeneración Nerviosa/efectos de los fármacos , Traumatismos del Nervio Óptico/metabolismo , Nervio Óptico/efectos de los fármacos , Tretinoina/metabolismo , Animales , Anuros , Benzoatos/farmacología , Cromanos/farmacología , Naftoles/farmacología , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/antagonistas & inhibidores , Receptores de Ácido Retinoico/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Tetrahidronaftalenos/farmacología
6.
J Chem Neuroanat ; 77: 80-92, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27242163

RESUMEN

Retinoic acid (RA) is important during development, in neuronal plasticity, and also in peripheral nervous system regeneration. Here we use the frog visual system as a model to investigate the changes in RA signaling that take place after axonal injury to the central nervous system. Immunocytochemistry was used to localize different components of RA signaling within sections of the retina and optic tectum, namely, the synthetic enzyme retinaldehyde dehydrogenase (RALDH), the RA binding proteins CRABPI and II, the retinoic acid receptors RARα, ß and γ, and finally the catabolic enzyme CYP26A1. The levels of these proteins were quantified in extracts of retina and tectum using Western blotting. Animals were studied at 1 week, 3 weeks and 6 weeks after optic nerve transection. At the latter time point the RGC axons were re-entering the optic tectum. All the components of RA signaling were present at low to moderate levels in retinas and tecta of control, unoperated animals. In retina, soon after optic nerve injury there was a large increase in RALDH, some increase in the CRABPs, and a large increase in RGC RARß and (expression. These increases continued as the RGC axons were regenerating, with the addition of later RARα expression at 6 weeks. At no stage did CYP26A1 expression significantly change. In the tectum the levels of RALDH increased after axotomy and during regrowth of axons (3 weeks), then decreased at 6 weeks, at which time the levels of CYP26A1 increased. Axotomy did not cause an immediate increase in tectal RAR levels but RARα and RARß increased after 3 weeks and RARγ only after 6 weeks. These results are consistent with RA signaling playing an important role in the survival and regeneration of frog RGCs.


Asunto(s)
Traumatismos del Nervio Óptico/fisiopatología , Transducción de Señal , Tretinoina/metabolismo , Vías Visuales/fisiopatología , Animales , Femenino , Regulación de la Expresión Génica , Inmunohistoquímica , Masculino , Rana pipiens , Receptores de Ácido Retinoico/biosíntesis , Retina/fisiopatología , Retinal-Deshidrogenasa/biosíntesis , Células Ganglionares de la Retina/metabolismo , Ácido Retinoico 4-Hidroxilasa/biosíntesis , Ácido Retinoico 4-Hidroxilasa/genética , Receptores X Retinoide/biosíntesis , Colículos Superiores/fisiopatología
7.
J Neurosci Res ; 92(1): 13-23, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24166589

RESUMEN

Neurotrophins such as ciliary neurotrophic factor (CNTF) and brain-derived neurotrophic factor (BDNF) and growth factors such as fibroblast growth factor (FGF-2) play important roles in neuronal survival and in axonal outgrowth during development. However, whether they can modulate regeneration after optic nerve injury in the adult animal is less clear. The present study investigates the effects of application of these neurotrophic factors on the speed, number, and distribution of regenerating axons in the frog Rana pipiens after optic nerve crush. Optic nerves were crushed and the factors, or phosphate-buffered saline, were applied to the stump or intraocularly. The nerves were examined at different times after axotomy, using anterograde labeling with biotin dextran amine and antibody against growth-associated protein 43. We measured the length, number, and distribution of axons projecting beyond the lesion site. Untreated regenerating axons show an increase in elongation rate over 3 weeks. CNTF more than doubles this rate, FGF-2 increases it, and BDNF has little effect. In contrast, the numbers of regenerating axons that have reached 200 µm at 2 weeks were more than doubled by FGF-2, increased by CNTF, and barely affected by BDNF. The regenerating axons were preferentially distributed in the periphery of the nerve; although the numbers of axons were increased by neurotrophic factor application, this overall distribution was substantially unaffected.


Asunto(s)
Axones/efectos de los fármacos , Factor Neurotrófico Ciliar/uso terapéutico , Factores de Crecimiento de Fibroblastos/uso terapéutico , Regeneración Nerviosa/efectos de los fármacos , Traumatismos del Nervio Óptico/tratamiento farmacológico , Animales , Axones/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Factor Neurotrófico Ciliar/farmacología , Factores de Crecimiento de Fibroblastos/farmacología , Compresión Nerviosa , Regeneración Nerviosa/fisiología , Traumatismos del Nervio Óptico/metabolismo , Rana pipiens
8.
J Chem Neuroanat ; 46(1-2): 35-44, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22940608

RESUMEN

We have previously shown that application of fibroblast growth factor-2 (FGF-2) to cut optic nerve axons enhances retinal ganglion cell (RGC) survival in the adult frog visual system. These actions are mediated via activation of its high affinity receptor FGFR1, enhanced BDNF and TrkB expression, increased CREB phosphorylation, and by promoting MAPK and PKA signaling pathways. The role of endogenous FGF-2 in this system is less well understood. In this study, we determine the distribution of FGF-2 and its receptors in normal animals and in animals at different times after optic nerve cut. Immunohistochemistry and Western blot analysis were conducted using specific antibodies against FGF-2 and its receptors in control retinas and optic tecta, and after one, three, and six weeks post nerve injury. FGF-2 was transiently increased in the retina while it was reduced in the optic tectum just one week after optic nerve transection. Axotomy induced a prolonged upregulation of FGFR1 and FGFR3 in both retina and tectum. FGFR4 levels decreased in the retina shortly after axotomy, whereas a significant increase was detected in the optic tectum. FGFR2 distribution was not affected by the optic nerve lesion. Changes in the presence of these proteins after axotomy suggest a potential role during regeneration.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regeneración Nerviosa/fisiología , Nervio Óptico/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Retina/metabolismo , Colículos Superiores/metabolismo , Animales , Femenino , Masculino , Rana pipiens
9.
J Neurosci Res ; 86(15): 3382-92, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18655198

RESUMEN

Application of basic fibroblast growth factor (FGF-2) to the optic nerve after axotomy promotes the survival of retinal ganglion cells (RGCs) in the frog Rana pipiens and results in a rapid up-regulation of brain-derived neurotrophic factor (BDNF) and TrkB synthesis by the RGCs. Here we investigate whether this up-regulation is maintained over the long term and whether it is required for FGF-2's survival effect. At 6 weeks after axotomy and FGF-2 treatment, we found more RGCs immunopositive for BDNF protein and higher intensity of BDNF and TrkB immunostaining, accompanied by increases in BDNF and TrkB mRNA in RGCs. Application of fluorescently labeled siRNA targeted against BDNF to the cut RGC axons showed that it was transported to the cell bodies. Axonal siRNA treatment eliminated the increases in BDNF immunostaining and mRNA that were induced by FGF-2 and had no effect on TrkB mRNA. This reduction in BDNF synthesis by siRNA greatly reduced the long-term survival effect of FGF-2 on RGCs. This, taken together with previous results, suggests that, although FGF-2 may initially activate survival pathways via ERK signaling, its main long-term survival effects are mediated via its up-regulation of BDNF synthesis by the RGCs.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Nervio Óptico/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Axotomía , Western Blotting , Supervivencia Celular/fisiología , Inmunohistoquímica , Hibridación in Situ , Nervio Óptico/patología , ARN Mensajero/análisis , ARN Interferente Pequeño , Rana pipiens , Receptor trkB/metabolismo , Células Ganglionares de la Retina/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
10.
Brain Res ; 1103(1): 65-75, 2006 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-16808907

RESUMEN

We have shown previously that application of fibroblast growth factor-2 (FGF-2) to the cut optic nerve of the frog, Rana pipiens, augments the survival of retinal ganglion cells (RGCs). In this study, we examine the effects of axotomy and FGF-2 treatment upon the distribution of nitric oxide synthase (NOS) and NADPH diaphorase (NADPH-d) activity in the frog retina and tectum. We find that NOS and NADPH-d are largely absent from RGCs but present in amacrine neurons and in retinorecipient tectal layers. Axotomy alone has little effect on NOS expression or diaphorase activity, apart from slightly increasing the levels of expression in a subpopulation of amacrine cells that arborize in the On sublamina of the inner plexiform layer. FGF-2 application to the optic nerve down-regulates NOS expression and activity in the retina and up-regulates it in the tectum, particularly in retinorecipient layers. Electron microscopy of the optic nerve and neurofilament immunostaining of the tectum suggests that FGF-2 treatment increases the number of regenerating retinal axons arriving at the tectum. The effects in the retina and tectum are probably indirect, that in the retina being due to retrograde signaling from RGCs to amacrine neurons, and that in the tectum being due to re-induction of NOS expression in tectal neurons by the arrival of regenerating axons. At this stage, it appears unlikely that these changes in NOS play a role in the FGF-2's survival effect on RGCs.


Asunto(s)
Axotomía , Factor 2 de Crecimiento de Fibroblastos/farmacología , NADPH Deshidrogenasa/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Retina/efectos de los fármacos , Retina/enzimología , Colículos Superiores/efectos de los fármacos , Colículos Superiores/enzimología , Animales , Axones/fisiología , Western Blotting , Recuento de Células , Inmunohistoquímica , Regeneración Nerviosa/efectos de los fármacos , Proteínas de Neurofilamentos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/enzimología , Rana pipiens , Retina/citología , Células Ganglionares de la Retina/efectos de los fármacos , Colículos Superiores/citología
11.
J Neurochem ; 96(1): 82-96, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16269011

RESUMEN

Application of basic fibroblast growth factor (FGF-2) to the optic nerve after axotomy promotes the survival of retinal ganglion cells (RGCs) in the frog, Rana pipiens. Here we investigate the effects of FGF-2 treatment upon the synthesis of brain-derived neurotrophic factor (BDNF) and its receptor, tyrosine receptor kinase B (TrkB). Axotomy alone increased the amounts of BDNF and TrkB mRNA in RGCs after 1 week and 48 h, respectively; FGF-2 treatment to the nerve accelerated and increased this up-regulation of both. FGF-2 also increased the amounts of phosphorylated cAMP response element binding protein (pCREB) in the retina. Blocking extracellular-regulated kinase (ERK) activation with PD98059 or U0126 prevented the FGF-2-induced up-regulation of BDNF transcription but had no effect on TrkB. However, blocking protein kinase A (PKA) with H89 or Rp-8-Cl-cAMPS reduced the up-regulation of both BDNF and TrkB, and reduced pCREB. In addition, H89 inhibited ERK activation, indicating cross-talk between the pathways. Finally, axonal application of blocking antibody against the FGF receptor 1 (FGFR1) prevented the FGF-2-induced up-regulation of BDNF and TrkB. Our results suggest that FGF-2 acts on RGCs via FGFR1, activating the ERK pathway and CREB to increase BDNF synthesis, and PKA and CREB to increase TrkB synthesis.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Nervio Óptico/efectos de los fármacos , Receptor trkB/biosíntesis , Células Ganglionares de la Retina/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Axotomía , Western Blotting , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , ADN Complementario/biosíntesis , ADN Complementario/genética , Activación Enzimática/efectos de los fármacos , Exones/genética , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Inmunohistoquímica , Hibridación in Situ , Proteínas Quinasas Activadas por Mitógenos/fisiología , Rana pipiens , Células Ganglionares de la Retina/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos
12.
J Neurochem ; 93(6): 1422-33, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15935058

RESUMEN

We have shown that application of basic fibroblast growth factor (FGF-2) to axotomized optic nerve promotes the survival of frog retinal ganglion cells (RGCs). In the present study we used western blotting and immunocytochemistry to investigate the effects of this FGF-2 treatment upon the activation of the extracellular signal-regulated kinase (ERK) pathway, the amounts and distribution of Bcl-2 family proteins, and the activation of caspase-3. Axotomy alone temporarily increased ERK activation; FGF-2 treatment to the nerve prolonged this activation. This effect was blocked by U0126, a selective ERK kinase (MEK) inhibitor. Axotomy caused a decrease in Bcl-2 and a small increase in Bcl-x(L). FGF-2 treatment caused an ERK-dependent increase in Bcl-2 and an ERK-independent increase in Bcl-x(L). The pro-apoptotic Bax was increased by axotomy; FGF-2 treatment greatly decreased Bax levels, an effect that was inhibited by U0126. Axotomy induced the cleavage of caspase-3; FGF-2 treatment blocked this effect in an ERK-dependent manner. Finally, intraocular application of the MEK inhibitor caused a large reduction in the survival-promoting effect that FGF-2 application to the nerve stump had on RGCs. Our results suggest that FGF-2 acts, at least in part, via the ERK pathway to prevent apoptosis of axotomized RGCs not only by increasing amounts of anti-apoptotic proteins, but also by a striking reduction in the levels of apoptotic effectors themselves.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Degeneración Nerviosa/metabolismo , Regeneración Nerviosa/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Axotomía , Caspasa 3 , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Regulación hacia Abajo/fisiología , Inhibidores Enzimáticos/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/prevención & control , Regeneración Nerviosa/efectos de los fármacos , Traumatismos del Nervio Óptico/tratamiento farmacológico , Traumatismos del Nervio Óptico/enzimología , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Rana pipiens , Células Ganglionares de la Retina/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Proteína X Asociada a bcl-2 , Proteína bcl-X
13.
J Neurosci Res ; 73(4): 507-17, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12898535

RESUMEN

Basic fibroblast growth factor (bFGF or FGF-2) has been implicated as a trophic factor that promotes survival and neurite outgrowth of neurons. We found previously that application of FGF-2 to the proximal stump of the injured axon increases retinal ganglion cell (RGC) survival. We determine here the effect of FGF-2 on expression of the axonal growth-associated phosphoprotein (GAP)-43 in retinal ganglion cells and tectum of Rana pipiens during regeneration of the optic nerve. In control retinas, GAP-43 protein was found in the optic fiber layer and in optic nerve; mRNA levels were low. After axotomy, mRNA levels increased sevenfold and GAP-43 protein was significantly increased. GAP-43 was localized in retinal axons and in a subset of RGC cell bodies and dendrites. This upregulation of GAP-43 was sustained through the period in which retinal axons reconnect with their target in the tectum. FGF-2 application to the injured nerve, but not to the eyeball, increased GAP-43 mRNA in the retina but decreased GAP-43 protein levels and decreased the number of immunopositive cell bodies. In the tectum, no treatment affected GAP-43 mRNA but FGF-2 application to the axotomized optic nerve increased GAP-43 protein in regenerating retinal projections. We conclude that FGF-2 upregulates the synthesis and alters the distribution of the axonal growth-promoting protein GAP-43, suggesting that it may enhance axonal regrowth.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/fisiología , Proteína GAP-43/metabolismo , Traumatismos del Nervio Óptico/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Axotomía/métodos , Western Blotting/métodos , Proteína GAP-43/genética , Inmunohistoquímica , Hibridación in Situ/métodos , Traumatismos del Nervio Óptico/patología , ARN Mensajero/biosíntesis , Rana pipiens , Retina/citología , Retina/metabolismo , Células Ganglionares de la Retina/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Colículos Superiores/metabolismo , Colículos Superiores/patología , Factores de Tiempo
14.
Brain Res ; 982(1): 54-63, 2003 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-12915240

RESUMEN

Neurotrophins are potent regulators of the survival of different neuronal populations in the CNS. Little is known of the immunodistribution of neurotrophin-3 (NT-3) and tyrosine kinase C (TrkC) receptor in the frog visual system, which can successfully regenerate and recover vision after injury. In this study we show that both NT-3 and TrkC are present in the frog retina and tectum, and that their distribution changes after optic nerve transection. Both NT-3 and TrkC are present in the ganglion cell layer, inner nuclear layer, nerve fiber layer and outer plexiform layer, and in Müller cells of control retinas. Quantification of identified RGCs shows that there are only small changes in the proportion, or intensity, of NT-3 immunostained cells surviving after axotomy and regeneration. Müller cell staining, however, is increased. TrkC staining in the retina does not change after axotomy. In the tectum, NT-3 immunoreactivity is present in the retinorecipient layer 9, and in radial processes of neurons and ependymoglia. TrkC is present in ependymoglia and in tectal neurons. After axotomy or colchicine treatment fewer NT-3-immunoreactive processes are present in layer 9 and there is decreased staining of tectal neurons. These data are consistent with the hypothesis that NT-3 is synthesized in the retina and anterogradely transported to the tectum. TrkC immunostaining, on the other hand, increases in tectal cells after optic nerve transection, suggesting that it may be regulated by the supply of NT-3 from the retina.


Asunto(s)
Neurotrofina 3/metabolismo , Receptor trkC/metabolismo , Vías Visuales/metabolismo , Animales , Axotomía , Inmunohistoquímica , Traumatismos del Nervio Óptico , Rana pipiens , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Colículos Superiores/metabolismo , Factores de Tiempo
15.
J Comp Neurol ; 454(4): 456-69, 2002 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-12455009

RESUMEN

In this study we used immunocytochemistry to investigate the distribution of brain-derived neurotrophic factor (BDNF) and its receptor tyrosine kinase (trkB) in retina and optic tectum of the frog Rana pipiens during regeneration after axotomy. We also measured changes in BDNF mRNA in retina and tectum. Retrograde labeling was used to identify retinal ganglion cells (RGCs) prior to quantification of the BDNF immunoreactivity. In control animals, BDNF was found in the majority of RGCs and displaced amacrine cells and in some cells in the inner nuclear layer (INL). After axotomy, BDNF immunoreactivity was reduced in RGCs but increased in the INL. BDNF mRNA levels in the retina remained high before and after axotomy. Three months after axotomy, after reconnection to the target, the staining intensity of many of the surviving RGCs had partially recovered. In the control tectum, BDNF staining was present in ependymoglial cells and in neurons throughout layers 4, 6, 8, and 9. After axotomy, BDNF staining in tectal neurons became more intense, even though mRNA synthesis was transiently down-regulated. In control retinas, trkB receptor immunostaining was present in most RGCs; no significant changes were observed after axotomy. In control tectum, trkB was detected only in ependymoglial cells. After axotomy, many neuronal cell bodies were transiently labeled. Our data are consistent with the hypothesis that a considerable fraction of the BDNF normally present in RGCs is acquired from their targets in the tectum. However, there are also intraretinal sources of BDNF that could contribute to the survival of RGCs.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Regeneración Nerviosa , Rana pipiens , Receptor trkB/metabolismo , Retina/metabolismo , Colículos Superiores/metabolismo , Animales , Axotomía , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/inmunología , Inmunohistoquímica , Nervio Óptico/cirugía , ARN Mensajero/análisis , Receptor trkB/genética , Receptor trkB/inmunología , Retina/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Colículos Superiores/patología
16.
P. R. health sci. j ; 12(2): 143-6, jun. 1993.
Artículo en Inglés | LILACS | ID: lil-176753

RESUMEN

Tyrosine hydroxylase (TH), the rate limiting enzyme in the conversion of tyrosine to DOPA, is a reliable marker for catecholaminergic (dopaminergic) neurons. To investigate the distribution of dopamine in the retina of the thirteen-lined ground squirrel (Spermophilus tridecemlineatus), retinal sections and wholemounts were incubated with an antiserum directed against TH and then processed using the avidin-biotin immunohistochemical method. TH-like immunoreactivity was exhibited by amacrine and interplexiform-like cells in the innermost portion of the inner nuclear layer (INL) and by cells we presume to be displaced amacrines in the ganglion cell layer (GCL). Their somata were 12 to 20 microns in diameter, with the majority measuring approximately 18 microns. In transverse sections the processes of the three types of neurons were seen to extend into lamina 1 of the inner plexiform layer (IPL). In horizontal sections 2-3 primary dendrites were seen to ramify and the branches extended for considerable distances, with overlap between the dendritic fields of neighboring TH cells. A distance to the nearest neighbor analysis suggests the TH-neurons in the INL are distributed in a non-random fashion


Asunto(s)
Animales , Células Ganglionares de la Retina/citología , Retina/citología , Sciuridae/anatomía & histología , Tirosina 3-Monooxigenasa/análisis , Células Ganglionares de la Retina/química , Células Ganglionares de la Retina/metabolismo , Retina/química , Retina/metabolismo , Sciuridae/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...