Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Glia ; 72(8): 1469-1483, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38771121

ABSTRACT

Myelination is the terminal step in a complex and precisely timed program that orchestrates the proliferation, migration and differentiation of oligodendroglial cells. It is thought that Sonic Hedgehog (Shh) acting on Smoothened (Smo) participates in regulating this process, but that these effects are highly context dependent. Here, we investigate oligodendroglial development and remyelination from three specific transgenic lines: NG2-CreERT2 (control), Smofl/fl/NG2-CreERT2 (loss of function), and SmoM2/NG2-CreERT2 (gain of function), as well as pharmacological manipulation that enhance or inhibit the Smo pathway (Smoothened Agonist (SAG) or cyclopamine treatment, respectively). To explore the effects of Shh/Smo on differentiation and myelination in vivo, we developed a highly quantifiable model by transplanting oligodendrocyte precursor cells (OPCs) in the retina. We find that myelination is greatly enhanced upon cyclopamine treatment and hypothesize that Shh/Smo could promote OPC proliferation to subsequently inhibit differentiation. Consistent with this hypothesis, we find that the genetic activation of Smo significantly increased numbers of OPCs and decreased oligodendrocyte differentiation when we examined the corpus callosum during development and after cuprizone demyelination and remyelination. However, upon loss of function with the conditional ablation of Smo, myelination in the same scenarios are unchanged. Taken together, our present findings suggest that the Shh pathway is sufficient to maintain OPCs in an undifferentiated state, but is not necessary for myelination and remyelination.


Subject(s)
Cell Differentiation , Hedgehog Proteins , Mice, Transgenic , Myelin Sheath , Oligodendrocyte Precursor Cells , Smoothened Receptor , Animals , Hedgehog Proteins/metabolism , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/drug effects , Smoothened Receptor/metabolism , Smoothened Receptor/genetics , Myelin Sheath/metabolism , Cell Differentiation/physiology , Cell Differentiation/drug effects , Veratrum Alkaloids/pharmacology , Mice , Remyelination/physiology , Remyelination/drug effects , Oligodendroglia/metabolism , Oligodendroglia/drug effects , Oligodendroglia/physiology , Mice, Inbred C57BL , Signal Transduction/physiology , Signal Transduction/drug effects
2.
Br J Pharmacol ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38742374

ABSTRACT

BACKGROUND AND PURPOSE: ApTOLL is an aptamer selected to antagonize toll-like receptor 4 (TLR4), a relevant actor for innate immunity involved in inflammatory responses in multiple sclerosis (MS) and other diseases. The currently available therapeutic arsenal to treat MS is composed of immunomodulators but, to date, there are no (re)myelinating drugs available in clinics. In our present study, we studied the effect of ApTOLL on different animal models of MS. EXPERIMENTAL APPROACH: The experimental autoimmune encephalomyelitis (EAE) model was used to evaluate the effect of ApTOLL on reducing the inflammatory component. A more direct effect on oligodendroglia was studied with the cuprizone model and purified primary cultures of murine and human oligodendrocyte precursor cells (OPCs) isolated through magnetic-activated cell sorting (MACS) from samples of brain cortex. Also, we tested these effects in an ex vivo model of organotypic cultures demyelinated with lysolecithin (LPC). KEY RESULTS: ApTOLL treatment positively impacted the clinical symptomatology of mice in the EAE and cuprizone models, which was associated with better preservation plus restoration of myelin and oligodendrocytes in the demyelinated lesions of animals. Restoration was corroborated on purified cultures of rodent and human OPCs. CONCLUSION AND IMPLICATIONS: Our findings reveal a new therapeutic approach for the treatment of inflammatory and demyelinating diseases such as MS. The molecular nature of the aptamer exerts not only an anti-inflammatory effect but also neuroprotective and remyelinating effects. The excellent safety profile demonstrated by ApTOLL in animals and humans opens the door to future clinical trials in MS patients.

3.
Brain Struct Funct ; 228(3-4): 907-920, 2023 May.
Article in English | MEDLINE | ID: mdl-36995433

ABSTRACT

The development and survival of dopaminergic neurons are influenced by the fibroblast growth factor (FGF) pathway. Anosmin-1 (A1) is an extracellular matrix protein that acts as a major regulator of this signaling pathway, controlling FGF diffusion, and receptor interaction and shuttling. In particular, previous work showed that A1 overexpression results in more dopaminergic neurons in the olfactory bulb. Prompted by those intriguing results, in this study, we investigated the effects of A1 overexpression on different populations of catecholaminergic neurons in the central (CNS) and the peripheral nervous systems (PNS). We found that A1 overexpression increases the number of dopaminergic substantia nigra pars compacta (SNpc) neurons and alters the striosome/matrix organization of the striatum. Interestingly, these numerical and morphological changes in the nigrostriatal pathway of A1-mice did not confer an altered susceptibility to experimental MPTP-parkinsonism with respect to wild-type controls. Moreover, the study of the effects of A1 overexpression was extended to different dopaminergic tissues associated with the PNS, detecting a significant reduction in the number of dopaminergic chemosensitive carotid body glomus cells in A1-mice. Overall, our work shows that A1 regulates the development and survival of dopaminergic neurons in different nuclei of the mammalian nervous system.


Subject(s)
Parkinson Disease , Mice , Animals , Parkinson Disease/pathology , Substantia Nigra/metabolism , Extracellular Matrix Proteins/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Peripheral Nervous System/metabolism , Peripheral Nervous System/pathology , Mice, Inbred C57BL , Mammals
4.
Cell Death Dis ; 13(4): 383, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35444190

ABSTRACT

Synaptic loss, neuronal death, and circuit remodeling are common features of central nervous system neurodegenerative disorders. Retinitis pigmentosa (RP), the leading cause of inherited blindness, is a group of retinal dystrophies characterized by photoreceptor dysfunction and death. The insulin receptor, a key controller of metabolism, also regulates neuronal survival and synaptic formation, maintenance, and activity. Indeed, deficient insulin receptor signaling has been implicated in several brain neurodegenerative pathologies. We present evidence linking impaired insulin receptor signaling with RP. We describe a selective decrease in the levels of the insulin receptor and its downstream effector phospho-S6 in retinal horizontal cell terminals in the rd10 mouse model of RP, as well as aberrant synapses between rod photoreceptors and the postsynaptic terminals of horizontal and bipolar cells. A gene therapy strategy to induce sustained proinsulin, the insulin precursor, production restored retinal insulin receptor signaling, by increasing S6 phosphorylation, without peripheral metabolic consequences. Moreover, proinsulin preserved photoreceptor synaptic connectivity and prolonged visual function in electroretinogram and optomotor tests. These findings point to a disease-modifying role of insulin receptor and support the therapeutic potential of proinsulin in retinitis pigmentosa.


Subject(s)
Proinsulin , Retinitis Pigmentosa , Animals , Disease Models, Animal , Insulin , Mice , Mice, Inbred C57BL , Proinsulin/pharmacology , Receptor, Insulin , Retinitis Pigmentosa/pathology , Synapses/metabolism
5.
Dev Dyn ; 251(2): 240-275, 2022 02.
Article in English | MEDLINE | ID: mdl-34241926

ABSTRACT

Membrane-type matrix metalloproteinases (MT-MMPs) are cell membrane-tethered proteinases that belong to the family of the MMPs. Apart from their roles in degradation of the extracellular milieu, MT-MMPs are able to activate through proteolytic processing at the cell surface distinct molecules such as receptors, growth factors, cytokines, adhesion molecules, and other pericellular proteins. Although most of the information regarding these enzymes comes from cancer studies, our current knowledge about their contribution in distinct developmental processes occurring in the embryo is limited. In this review, we want to summarize the involvement of MT-MMPs in distinct processes during embryonic morphogenesis, including cell migration and proliferation, epithelial-mesenchymal transition, cell polarity and branching, axon growth and navigation, synapse formation, and angiogenesis. We also considered information about MT-MMP functions from studies assessed in pathological conditions and compared these data with those relevant for embryonic development.


Subject(s)
Matrix Metalloproteinases , Neoplasms , Cell Membrane , Embryonic Development , Extracellular Matrix/metabolism , Humans , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases, Membrane-Associated/metabolism , Neoplasms/pathology
6.
Nat Commun ; 12(1): 3098, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035282

ABSTRACT

The human Alzheimer's disease (AD) brain accumulates angiogenic markers but paradoxically, the cerebral microvasculature is reduced around Aß plaques. Here we demonstrate that angiogenesis is started near Aß plaques in both AD mouse models and human AD samples. However, endothelial cells express the molecular signature of non-productive angiogenesis (NPA) and accumulate, around Aß plaques, a tip cell marker and IB4 reactive vascular anomalies with reduced NOTCH activity. Notably, NPA induction by endothelial loss of presenilin, whose mutations cause familial AD and which activity has been shown to decrease with age, produced a similar vascular phenotype in the absence of Aß pathology. We also show that Aß plaque-associated NPA locally disassembles blood vessels, leaving behind vascular scars, and that microglial phagocytosis contributes to the local loss of endothelial cells. These results define the role of NPA and microglia in local blood vessel disassembly and highlight the vascular component of presenilin loss of function in AD.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Blood Vessels/metabolism , Brain/metabolism , Neovascularization, Pathologic/genetics , Plaque, Amyloid/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Blood Vessels/pathology , Brain/blood supply , Brain/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Female , Gene Expression Profiling/methods , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neovascularization, Pathologic/metabolism , Plaque, Amyloid/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
7.
J Vis Exp ; (136)2018 06 26.
Article in English | MEDLINE | ID: mdl-30010638

ABSTRACT

The Escherichia coli LacZ gene, encoding ß-galactosidase, is largely used as a reporter for gene expression and as a tracer in cell lineage studies. The classical histochemical reaction is based on the hydrolysis of the substrate X-gal in combination with ferric and ferrous ions, which produces an insoluble blue precipitate that is easy to visualize. Therefore, ß-galactosidase activity serves as a marker for the expression pattern of the gene of interest as the development proceeds. Here we describe the standard protocol for the detection of ß-galactosidase activity in early whole mouse embryos and the subsequent method for paraffin sectioning and counterstaining. Additionally, a procedure for clarifying whole embryos is provided to better visualize X-gal staining in deeper regions of the embryo. Consistent results are obtained by performing this procedure, although optimization of reaction conditions is needed to minimize background activity. Limitations in the assay should be also considered, particularly regarding the size of the embryo in whole mount staining. Our protocol provides a sensitive and a reliable method for ß-galactosidase detection during the mouse development that can be further applied to the cryostat sections as well as whole organs. Thus, the dynamic gene expression patterns throughout development can be easily analyzed by using this protocol in whole embryos, but also detailed expression at the cellular level can be assessed after paraffin sectioning.


Subject(s)
Gene Expression/genetics , Mice/embryology , beta-Galactosidase/genetics , Animals , beta-Galactosidase/metabolism
8.
Mol Neurodegener ; 13(1): 19, 2018 04 16.
Article in English | MEDLINE | ID: mdl-29661219

ABSTRACT

BACKGROUND: Retinitis pigmentosa (RP) is a group of hereditary retinal neurodegenerative conditions characterized by primary dysfunction and death of photoreceptor cells, resulting in visual loss and, eventually, blindness. To date, no effective therapies have been transferred to clinic. Given the diverse genetic etiology of RP, targeting common cellular and molecular retinal alterations has emerged as a potential therapeutic strategy. METHODS: Using the Pde6b rd10/rd10 mouse model of RP, we investigated the effects of daily intraperitoneal administration of VP3.15, a small-molecule heterocyclic GSK-3 inhibitor. Gene expression was analyzed by quantitative PCR and protein expression and phosphorylation by Western blot. Photoreceptor preservation was evaluated by histological analysis and visual function was assessed by electroretinography. RESULTS: In rd10 retinas, increased expression of pro-inflammatory markers and reactive gliosis coincided with the early stages of retinal degeneration. Compared with wild-type controls, GSK-3ß expression (mRNA and protein) remained unchanged during the retinal degeneration period. However, levels of GSK-3ßSer9 and its regulator AktSer473 were increased in rd10 versus wild-type retinas. In vivo administration of VP3.15 reduced photoreceptor cell loss and preserved visual function. This neuroprotective effect was accompanied by a decrease in the expression of neuroinflammatory markers. CONCLUSIONS: These results provide proof of concept of the therapeutic potential of VP3.15 for the treatment of retinal neurodegenerative conditions in general, and RP in particular.


Subject(s)
Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Retinitis Pigmentosa/pathology , Thiadiazoles/pharmacology , Animals , Apoptosis/drug effects , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Retina/drug effects
9.
PLoS One ; 13(3): e0194218, 2018.
Article in English | MEDLINE | ID: mdl-29543853

ABSTRACT

Light causes damage to the retina (phototoxicity) and decreases photoreceptor responses to light. The most harmful component of visible light is the blue wavelength (400-500 nm). Different filters have been tested, but so far all of them allow passing a lot of this wavelength (70%). The aim of this work has been to prove that a filter that removes 94% of the blue component may protect the function and morphology of the retina significantly. Three experimental groups were designed. The first group was unexposed to light, the second one was exposed and the third one was exposed and protected by a blue-blocking filter. Light damage was induced in young albino mice (p30) by exposing them to white light of high intensity (5,000 lux) continuously for 7 days. Short wavelength light filters were used for light protection. The blue component was removed (94%) from the light source by our filter. Electroretinographical recordings were performed before and after light damage. Changes in retinal structure were studied using immunohistochemistry, and TUNEL labeling. Also, cells in the outer nuclear layer were counted and compared among the three different groups. Functional visual responses were significantly more conserved in protected animals (with the blue-blocking filter) than in unprotected animals. Also, retinal structure was better kept and photoreceptor survival was greater in protected animals, these differences were significant in central areas of the retina. Still, functional and morphological responses were significantly lower in protected than in unexposed groups. In conclusion, this blue-blocking filter decreases significantly photoreceptor damage after exposure to high intensity light. Actually, our eyes are exposed for a very long time to high levels of blue light (screens, artificial light LED, neons…). The potential damage caused by blue light can be palliated.


Subject(s)
Eye Injuries/prevention & control , Light/adverse effects , Radiation Injuries, Experimental/prevention & control , Retina/radiation effects , Retinal Degeneration/prevention & control , Animals , Color , Electroretinography , Eye Injuries/diagnosis , Eye Injuries/etiology , In Situ Nick-End Labeling , Mice , Photoreceptor Cells, Vertebrate/radiation effects , Radiation Injuries, Experimental/diagnosis , Radiation Injuries, Experimental/etiology , Retina/cytology , Retina/injuries , Retinal Degeneration/etiology
10.
Cell Death Dis ; 8(7): e2922, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28703796

ABSTRACT

ProNGF signaling through p75NTR has been associated with neurodegenerative disorders. Retinitis pigmentosa (RP) comprises a group of inherited retinal dystrophies that causes progressive photoreceptor cell degeneration and death, at a rate dependent on the genetic mutation. There are more than 300 mutations causing RP, and this is a challenge to therapy. Our study was designed to explore a common mechanism for p75NTR in the progression of RP, and assess its potential value as a therapeutic target. The proNGF/p75NTR system is present in the dystrophic retina of the rd10 RP mouse model. Compared with wild-type (WT) retina, the levels of unprocessed proNGF were increased in the rd10 retina at early degenerative stages, before the peak of photoreceptor cell death. Conversely, processed NGF levels were similar in rd10 and WT retinas. ProNGF remained elevated throughout the period of photoreceptor cell loss, correlating with increased expression of α2-macroglobulin, an inhibitor of proNGF processing. The neuroprotective effect of blocking p75NTR was assessed in organotypic retinal cultures from rd10 and RhoP mouse models. Retinal explants treated with p75NTR antagonists showed significantly reduced photoreceptor cell death, as determined by the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay and by preservation of the thickness of the outer nuclear layer (ONL), where photoreceptor nuclei are located. This effect was accompanied by decreased retinal-reactive gliosis and reduced TNFα secretion. Use of p75NTR antagonist THX-B (1,3-diisopropyl-1-[2-(1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-purin-7-yl)-acetyl]-urea) in vivo in the rd10 and RhoP mouse models, by a single intravitreal or subconjunctival injection, afforded neuroprotection to photoreceptor cells, with preservation of the ONL. This study demonstrates a role of the p75NTR/proNGF axis in the progression of RP, and validates these proteins as therapeutic targets in two different RP models, suggesting utility irrespective of etiology.


Subject(s)
Apoptosis/drug effects , Neuroprotective Agents/pharmacology , Receptors, Nerve Growth Factor/antagonists & inhibitors , Retinitis Pigmentosa/pathology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Inbred C57BL , Nerve Growth Factor/analysis , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Neuroprotective Agents/chemistry , Photoreceptor Cells/cytology , Photoreceptor Cells/drug effects , Photoreceptor Cells/metabolism , Protein Precursors/analysis , Protein Precursors/metabolism , Purines/chemistry , Purines/pharmacology , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinitis Pigmentosa/metabolism , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Urea/analogs & derivatives , Urea/chemistry , Urea/pharmacology
11.
J Enzyme Inhib Med Chem ; 32(1): 522-526, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28114834

ABSTRACT

Retinitis pigmentosa (RP) is an inherited retinal dystrophy that courses with progressive degeneration of retinal tissue and loss of vision. Currently, RP is an unpreventable, incurable condition. We propose glycogen synthase kinase 3 (GSK-3) inhibitors as potential leads for retinal cell neuroprotection, since the retina is also a part of the central nervous system and GSK-3 inhibitors are potent neuroprotectant agents. Using a chemical genetic approach, diverse small molecules with different potency and binding mode to GSK-3 have been used to validate and confirm GSK-3 as a pharmacological target for RP. Moreover, this medicinal chemistry approach has provided new leads for the future disease-modifying treatment of RP.


Subject(s)
Glycogen Synthase Kinase 3/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Retinitis Pigmentosa/drug therapy , Small Molecule Libraries/pharmacology , Animals , Cell Death/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3/metabolism , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Retinitis Pigmentosa/enzymology , Retinitis Pigmentosa/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
12.
Invest Ophthalmol Vis Sci ; 57(8): 3610-8, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27391551

ABSTRACT

PURPOSE: The induction of proinsulin expression by transgenesis or intramuscular gene therapy has been shown previously to retard retinal degeneration in mouse and rat models of retinitis pigmentosa (RP), a group of inherited conditions that result in visual impairment. We investigated whether intraocular treatment with biodegradable poly (lactic-co-glycolic) acid microspheres (PLGA-MS) loaded with proinsulin has cellular and functional neuroprotective effects in the retina. METHODS: Experiments were performed using the Pde6brd10 mouse model of RP. Methionylated human recombinant proinsulin (hPI) was formulated in PLGA-MS, which were administered by intravitreal injection on postnatal days (P) 14 to 15. Retinal neuroprotection was assessed at P25 by electroretinography, and by evaluating outer nuclear layer (ONL) cellular preservation. The attenuation of photoreceptor cell death by hPI was determined by TUNEL assay in cultured P22 retinas, as well as Akt phosphorylation by immunoblotting. RESULTS: We successfully formulated hPI PLGA-MS to deliver the active molecule for several weeks in vitro. The amplitude of b-cone and mixed b-waves in electroretinographic recording was significantly higher in eyes injected with hPI-PLGA-MS compared to control eyes. Treatment with hPI-PLGA-MS attenuated photoreceptor cell loss, as revealed by comparing ONL thickness and the number of cell rows in this layer in treated versus untreated retinas. Finally, hPI prevented photoreceptor cell death and increased AktThr308 phosphorylation in organotypic cultured retinas. CONCLUSIONS: Retinal degeneration in the rd10 mouse was slowed by a single intravitreal injection of hPI-PLGA-MS. Human recombinant proinsulin elicited a rapid and effective neuroprotective effect when administered in biodegradable microspheres, which may constitute a future potentially feasible delivery method for proinsulin-based treatment of RP.


Subject(s)
Blindness/physiopathology , Neuroprotective Agents/pharmacology , Proinsulin/pharmacology , Retinal Cone Photoreceptor Cells/pathology , Retinitis Pigmentosa/pathology , Animals , Biodegradable Plastics , Blindness/pathology , Cell Count , Cell Death/physiology , Cells, Cultured , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Intravitreal Injections , MAP Kinase Signaling System/physiology , Male , Mice, Transgenic , Microspheres , Neuroprotective Agents/administration & dosage , Phosphorylation , Proinsulin/administration & dosage , Retinal Cone Photoreceptor Cells/drug effects , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology
13.
Hum Mol Genet ; 22(14): 2775-84, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23512985

ABSTRACT

Down syndrome (DS) results from the triplication of approximately 300 human chromosome 21 (Hsa21) genes and affects almost all body organs. Children with DS have defects in visual processing that may have a negative impact on their daily life and cognitive development. However, there is little known about the genes and pathogenesis underlying these defects. Here, we show morphometric in vivo data indicating that the neural retina is thicker in DS individuals than in the normal population. A similar thickening specifically affecting the inner part of the retina was also observed in a trisomic model of DS, the Ts65Dn mouse. Increased retinal size and cellularity in this model correlated with abnormal retinal function and resulted from an impaired caspase-9-mediated apoptosis during development. Moreover, we show that mice bearing only one additional copy of Dyrk1a have the same retinal phenotype as Ts65Dn mice and normalization of Dyrk1a gene copy number in Ts65Dn mice completely rescues both, morphological and functional phenotypes. Thus, triplication of Dyrk1a is necessary and sufficient to cause the retinal phenotype described in the trisomic model. Our data demonstrate for the first time the implication of DYRK1A overexpression in a developmental alteration of the central nervous system associated with DS, thereby providing insights into the aetiology of neurosensorial dysfunction in a complex disease.


Subject(s)
Down Syndrome/enzymology , Down Syndrome/genetics , Gene Dosage , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Retina/anatomy & histology , Adult , Animals , Apoptosis , Caspase 9/genetics , Caspase 9/metabolism , Disease Models, Animal , Down Syndrome/physiopathology , Female , Gene Amplification , Humans , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Retina/cytology , Retina/enzymology , Young Adult , Dyrk Kinases
14.
Exp Eye Res ; 93(5): 607-17, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21824473

ABSTRACT

In this work, we used the pcd (Purkinje cell degeneration) mutant mouse with a slow temporal progression of photoreceptor degeneration in order to analyze the structural and functional modifications in the neuronal populations of the outer and inner retina. Retinal immunocytochemistry and functional electroretinography were performed on the pcd/pcd mutant mice and control wild type animals of the C57/DBA strain at 45, 90, 180 and 270 post-natal days. Immunohistochemical studies were performed for a series of protein markers: calbindin, calretinin, PKCα, bassoon, synapsin, syntaxin and islet1. Full field electroretinography recordings were performed on control and dystrophic mice. Rod and mixed responses, and oscillatory potentials, were recorded in dark adapted conditions; cone and flicker responses were recorded under light adaptation. Our results show significant structural modifications in the photoreceptor populations and neurons of the inner retina. Changes in cell morphology affect mainly to the bipolar cells, which gradually lose their dendritic tufts. The electroretinography records reveal that in the pcd retinas the rod and cone systems show a reduction in the amplitude of the electrical signals. This decrease progresses slowly with the passage of time, although for the most advanced stage of photoreceptor degeneration considered, 270 post-natal days, it is still possible to record light induced responses. We conclude that pcd mice experience a loss of retinal function in correlation with the loss of photoreceptors with age, and significant changes in retinal synaptic processes.


Subject(s)
Disease Models, Animal , Photoreceptor Cells, Vertebrate/pathology , Purkinje Cells/pathology , Retina/physiopathology , Retinal Dystrophies/pathology , Retinal Ganglion Cells/pathology , Animals , Biomarkers/metabolism , Dark Adaptation , Electroretinography , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Mutant Strains , Photic Stimulation , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...