Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 153
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(10): e2115973119, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35235463

RESUMO

White matter disorders of the central nervous system (CNS), such as multiple sclerosis (MS), lead to failure of nerve conduction and long-lasting neurological disabilities affecting a variety of sensory and motor systems, including vision. While most disease-modifying therapies target the immune and inflammatory response, the promotion of remyelination has become a new therapeutic avenue to prevent neuronal degeneration and promote recovery. Most of these strategies have been developed in short-lived rodent models of demyelination, which spontaneously repair and do not reflect the size, organization, and biology of the human CNS. Thus, well-defined nonhuman primate models are required to efficiently advance therapeutic approaches for patients. Here, we followed the consequence of long-term toxin-induced demyelination of the macaque optic nerve on remyelination and axon preservation, as well as its impact on visual functions. Findings from oculomotor behavior, ophthalmic examination, electrophysiology, and retinal imaging indicate visual impairment involving the optic nerve and retina. These visual dysfunctions fully correlated at the anatomical level, with sustained optic nerve demyelination, axonal degeneration, and alterations of the inner retinal layers. This nonhuman primate model of chronic optic nerve demyelination associated with axonal degeneration and visual dysfunction, recapitulates several key features of MS lesions and should be instrumental in providing the missing link to translate emerging repair promyelinating/neuroprotective therapies to the clinic for myelin disorders, such as MS.


Assuntos
Axônios , Nervo Óptico/patologia , Remielinização , Retina/patologia , Transtornos da Visão/patologia , Animais , Modelos Animais de Doenças , Potenciais Evocados Visuais , Macaca fascicularis , Masculino , Esclerose Múltipla/patologia , Reflexo Pupilar , Retina/diagnóstico por imagem , Retina/fisiopatologia , Tomografia de Coerência Óptica
2.
Int J Mol Sci ; 25(5)2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38474284

RESUMO

N-retinylidene-N-retinylethanolamine (A2E) has been associated with age-related macular degeneration (AMD) physiopathology by inducing cell death, angiogenesis and inflammation in retinal pigmented epithelial (RPE) cells. It was previously thought that the A2E effects were solely mediated via the retinoic acid receptor (RAR)-α activation. However, this conclusion was based on experiments using the RAR "specific" antagonist RO-41-5253, which was found to also be a ligand and partial agonist of the peroxisome proliferator-activated receptor (PPAR)-γ. Moreover, we previously reported that inhibiting PPAR and retinoid X receptor (RXR) transactivation with norbixin also modulated inflammation and angiogenesis in RPE cells challenged in the presence of A2E. Here, using several RAR inhibitors, we deciphered the respective roles of RAR, PPAR and RXR transactivations in an in vitro model of AMD. We showed that BMS 195614 (a selective RAR-α antagonist) displayed photoprotective properties against toxic blue light exposure in the presence of A2E. BMS 195614 also significantly reduced the AP-1 transactivation and mRNA expression of the inflammatory interleukin (IL)-6 and vascular endothelial growth factor (VEGF) induced by A2E in RPE cells in vitro, suggesting a major role of RAR in these processes. Surprisingly, however, we showed that (1) Norbixin increased the RAR transactivation and (2) AGN 193109 (a high affinity pan-RAR antagonist) and BMS 493 (a pan-RAR inverse agonist), which are photoprotective against toxic blue light exposure in the presence of A2E, also inhibited PPARs transactivation and RXR transactivation, respectively. Therefore, in our in vitro model of AMD, several commercialized RAR inhibitors appear to be non-specific, and we propose that the phototoxicity and expression of IL-6 and VEGF induced by A2E in RPE cells operates through the activation of PPAR or RXR rather than by RAR transactivation.


Assuntos
Carotenoides , Degeneração Macular , Receptores Ativados por Proliferador de Peroxissomo , Quinolinas , para-Aminobenzoatos , Anti-Inflamatórios , Agonismo Inverso de Drogas , Inflamação , Degeneração Macular/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptor alfa de Ácido Retinoico/metabolismo , Receptores X de Retinoides/metabolismo , Retinoides/metabolismo , Ativação Transcricional , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
J Neuroinflammation ; 20(1): 28, 2023 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-36755326

RESUMO

Retinal melanosome/melanolipofuscin-containing cells (MCCs), clinically visible as hyperreflective foci (HRF) and a highly predictive imaging biomarker for the progression of age-related macular degeneration (AMD), are widely believed to be migrating retinal pigment epithelial (RPE) cells. Using human donor tissue, we identify the vast majority of MCCs as melanophages, melanosome/melanolipofuscin-laden mononuclear phagocytes (MPs). Using serial block-face scanning electron microscopy, RPE flatmounts, bone marrow transplantation and in vitro experiments, we show how retinal melanophages form by the transfer of melanosomes from the RPE to subretinal MPs when the "don't eat me" signal CD47 is blocked. These melanophages give rise to hyperreflective foci in Cd47-/--mice in vivo, and are associated with RPE dysmorphia similar to intermediate AMD. Finally, we show that Cd47 expression in human RPE declines with age and in AMD, which likely participates in melanophage formation and RPE decline. Boosting CD47 expression in AMD might protect RPE cells and delay AMD progression.


Assuntos
Antígeno CD47 , Degeneração Macular , Humanos , Animais , Camundongos , Antígeno CD47/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Degeneração Macular/metabolismo , Retina/metabolismo , Tomografia de Coerência Óptica/métodos
4.
Proc Natl Acad Sci U S A ; 117(25): 14453-14463, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32513717

RESUMO

Deep regions of the brain are not easily accessible to investigation at the mesoscale level in awake animals or humans. We have recently developed a functional ultrasound (fUS) technique that enables imaging hemodynamic responses to visual tasks. Using fUS imaging on two awake nonhuman primates performing a passive fixation task, we constructed retinotopic maps at depth in the visual cortex (V1, V2, and V3) in the calcarine and lunate sulci. The maps could be acquired in a single-hour session with relatively few presentations of the stimuli. The spatial resolution of the technology is illustrated by mapping patterns similar to ocular dominance (OD) columns within superficial and deep layers of the primary visual cortex. These acquisitions using fUS suggested that OD selectivity is mostly present in layer IV but with extensions into layers II/III and V. This imaging technology provides a new mesoscale approach to the mapping of brain activity at high spatiotemporal resolution in awake subjects within the whole depth of the cortex.


Assuntos
Mapeamento Encefálico/métodos , Córtex Visual/fisiologia , Vigília/fisiologia , Animais , Dominância Ocular/fisiologia , Feminino , Macaca mulatta , Masculino , Estimulação Luminosa , Reprodutibilidade dos Testes , Análise Espaço-Temporal , Ultrassonografia/métodos , Córtex Visual/diagnóstico por imagem
5.
Proc Natl Acad Sci U S A ; 116(52): 26280-26287, 2019 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-31871177

RESUMO

Retinal degenerative diseases caused by photoreceptor cell death are major causes of irreversible vision loss. As only primates have a macula, the nonhuman primate (NHP) models have a crucial role not only in revealing biological mechanisms underlying high-acuity vision but also in the development of therapies. Successful translation of basic research findings into clinical trials and, moreover, approval of the first therapies for blinding inherited and age-related retinal dystrophies has been reported in recent years. This article explores the value of the NHP models in understanding human vision and reviews their contribution to the development of innovative therapeutic strategies to save and restore vision.

6.
Int J Mol Sci ; 24(1)2022 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-36613663

RESUMO

Mutations in GPR179 are one of the most common causes of autosomal recessive complete congenital stationary night blindness (cCSNB). This retinal disease is characterized in patients by impaired dim and night vision, associated with other ocular symptoms, including high myopia. cCSNB is caused by a complete loss of signal transmission from photoreceptors to ON-bipolar cells. In this study, we hypothesized that the lack of Gpr179 and the subsequent impaired ON-pathway could lead to myopic features in a mouse model of cCSNB. Using ultra performance liquid chromatography, we show that adult Gpr179-/- mice have a significant decrease in both retinal dopamine and 3,4-dihydroxyphenylacetic acid, compared to Gpr179+/+ mice. This alteration of the dopaminergic system is thought to be correlated with an increased susceptibility to lens-induced myopia but does not affect the natural refractive development. Altogether, our data added a novel myopia model, which could be used to identify therapeutic interventions.


Assuntos
Doenças Genéticas Ligadas ao Cromossomo X , Miopia , Cegueira Noturna , Camundongos , Animais , Eletrorretinografia/métodos , Cegueira Noturna/genética , Retina , Miopia/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Receptores Acoplados a Proteínas G/genética
7.
PLoS Comput Biol ; 16(7): e1007857, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32667921

RESUMO

In many cases of inherited retinal degenerations, ganglion cells are spared despite photoreceptor cell death, making it possible to stimulate them to restore visual function. Several studies have shown that it is possible to express an optogenetic protein in ganglion cells and make them light sensitive, a promising strategy to restore vision. However the spatial resolution of optogenetically-reactivated retinas has rarely been measured, especially in the primate. Since the optogenetic protein is also expressed in axons, it is unclear if these neurons will only be sensitive to the stimulation of a small region covering their somas and dendrites, or if they will also respond to any stimulation overlapping with their axon, dramatically impairing spatial resolution. Here we recorded responses of mouse and macaque retinas to random checkerboard patterns following an in vivo optogenetic therapy. We show that optogenetically activated ganglion cells are each sensitive to a small region of visual space. A simple model based on this small receptive field predicted accurately their responses to complex stimuli. From this model, we simulated how the entire population of light sensitive ganglion cells would respond to letters of different sizes. We then estimated the maximal acuity expected by a patient, assuming it could make an optimal use of the information delivered by this reactivated retina. The obtained acuity is above the limit of legal blindness. Our model also makes interesting predictions on how acuity might vary upon changing the therapeutic strategy, assuming an optimal use of the information present in the retinal activity. Optogenetic therapy could thus potentially lead to high resolution vision, under conditions that our model helps to determinine.


Assuntos
Cegueira , Optogenética/métodos , Células Ganglionares da Retina/fisiologia , Animais , Cegueira/fisiopatologia , Cegueira/terapia , Terapia Genética , Macaca , Camundongos , Modelos Biológicos , Retina/fisiologia , Acuidade Visual/fisiologia
8.
Brain ; 143(10): 2911-2928, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33103737

RESUMO

Human post-natal neurodevelopmental delay is often associated with cerebral alterations that can lead, by themselves or associated with peripheral deficits, to premature death. Here, we report the clinical features of 10 patients from six independent families with mutations in the autosomal YIF1B gene encoding a ubiquitous protein involved in anterograde traffic from the endoplasmic reticulum to the cell membrane, and in Golgi apparatus morphology. The patients displayed global developmental delay, motor delay, visual deficits with brain MRI evidence of ventricle enlargement, myelination alterations and cerebellar atrophy. A similar profile was observed in the Yif1b knockout (KO) mouse model developed to identify the cellular alterations involved in the clinical defects. In the CNS, mice lacking Yif1b displayed neuronal reduction, altered myelination of the motor cortex, cerebellar atrophy, enlargement of the ventricles, and subcellular alterations of endoplasmic reticulum and Golgi apparatus compartments. Remarkably, although YIF1B was not detected in primary cilia, biallelic YIF1B mutations caused primary cilia abnormalities in skin fibroblasts from both patients and Yif1b-KO mice, and in ciliary architectural components in the Yif1b-KO brain. Consequently, our findings identify YIF1B as an essential gene in early post-natal development in human, and provide a new genetic target that should be tested in patients developing a neurodevelopmental delay during the first year of life. Thus, our work is the first description of a functional deficit linking Golgipathies and ciliopathies, diseases so far associated exclusively to mutations in genes coding for proteins expressed within the primary cilium or related ultrastructures. We therefore propose that these pathologies should be considered as belonging to a larger class of neurodevelopmental diseases depending on proteins involved in the trafficking of proteins towards specific cell membrane compartments.


Assuntos
Cílios/genética , Complexo de Golgi/genética , Mutação/genética , Transtornos do Neurodesenvolvimento/genética , Proteínas de Transporte Vesicular/genética , Animais , Células Cultivadas , Cílios/patologia , Feminino , Complexo de Golgi/patologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Transtornos do Neurodesenvolvimento/diagnóstico por imagem
9.
Int J Mol Sci ; 23(1)2021 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-35008772

RESUMO

We investigate glial cell activation and oxidative stress induced by taurine deficiency secondary to ß-alanine administration and light exposure. Two months old Sprague-Dawley rats were divided into a control group and three experimental groups that were treated with 3% ß-alanine in drinking water (taurine depleted) for two months, light exposed or both. Retinal and external thickness were measured in vivo at baseline and pre-processing with Spectral-Domain Optical Coherence Tomography (SD-OCT). Retinal cryostat cross sections were immunodetected with antibodies against various antigens to investigate microglial and macroglial cell reaction, photoreceptor outer segments, synaptic connections and oxidative stress. Taurine depletion caused a decrease in retinal thickness, shortening of photoreceptor outer segments, microglial cell activation, oxidative stress in the outer and inner nuclear layers and the ganglion cell layer and synaptic loss. These events were also observed in light exposed animals, which in addition showed photoreceptor death and macroglial cell reactivity. Light exposure under taurine depletion further increased glial cell reaction and oxidative stress. Finally, the retinal pigment epithelial cells were Fluorogold labeled and whole mounted, and we document that taurine depletion impairs their phagocytic capacity. We conclude that taurine depletion causes cell damage to various retinal layers including retinal pigment epithelial cells, photoreceptors and retinal ganglion cells, and increases the susceptibility of the photoreceptor outer segments to light damage. Thus, beta-alanine supplements should be used with caution.


Assuntos
Luz , Neuroglia/patologia , Neuroglia/efeitos da radiação , Estresse Oxidativo/efeitos da radiação , Degeneração Retiniana/patologia , Taurina/metabolismo , Animais , Contagem de Células , Sobrevivência Celular , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Microglia/patologia , Neuroglia/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Ratos Sprague-Dawley , Degeneração Retiniana/sangue , Degeneração Retiniana/diagnóstico por imagem , Epitélio Pigmentado da Retina/diagnóstico por imagem , Epitélio Pigmentado da Retina/patologia , Sinapses/metabolismo , Taurina/sangue , Tomografia de Coerência Óptica , beta-Alanina
10.
J Neuroinflammation ; 17(1): 1, 2020 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-31900165

RESUMO

BACKGROUND: Age-related macular degeneration is characterized by the accumulation of subretinal macrophages and the degeneration of cones, but mainly of rods. We have previously shown that Mononuclear Phagocytes-derived IL-1ß induces rod photoreceptor cell death during experimental subretinal inflammation and in retinal explants exposed to IL-1ß but the mechanism is unknown. METHODS: Retinal explants were culture in the presence of human monocytes or IL-1ß and photoreceptor cell survival was analyzed by TUNEL labeling. Glutamate concentration and transcription levels of gene involved in the homeostasis of glutamate were analyzed in cell fractions of explant cultured or not in the presence of IL-1ß. Glutamate receptor antagonists were evaluated for their ability to reduce photoreceptor cell death in the presence of IL1-ß or monocytes. RESULTS: We here show that IL-1ß does not induce death in isolated photoreceptors, suggesting an indirect effect. We demonstrate that IL-1ß leads to glutamate-induced rod photoreceptor cell death as it increases the extracellular glutamate concentrations in the retina through the inhibition of its conversion to glutamine in Müller cells, increased release from Müller cells, and diminished reuptake. The inhibition of non-NMDA receptors completely and efficiently prevented rod apoptosis in retinal explants cultured in the presence of IL-1ß or, more importantly, in vivo, in a model of subretinal inflammation. CONCLUSIONS: Our study emphasizes the importance of inflammation in the deregulation of glutamate homeostasis and provides a comprehensive mechanism of action for IL-1ß-induced rod degeneration.


Assuntos
Ácido Glutâmico/metabolismo , Homeostase/fisiologia , Interleucina-1beta/toxicidade , Degeneração Retiniana/induzido quimicamente , Degeneração Retiniana/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Animais , Técnicas de Cocultura , Homeostase/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/efeitos dos fármacos
11.
Exp Eye Res ; 188: 107781, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31473259

RESUMO

To study the effect of taurine depletion induced by ß-alanine supplementation in the retinal nerve fiber layer (RNFL), and retinal ganglion cell (RGC) survival and axonal transport. Albino Sprague-Dawley rats were divided into two groups: one group received ß-alanine supplementation (3%) in the drinking water during 2 months to induce taurine depletion, and the other group received regular water. After one month, half of the rats from each group were exposed to light. Retinas were analyzed in-vivo using Spectral-Domain Optical Coherence Tomography (SD-OCT). Prior to processing, RGCs were retrogradely traced with fluorogold (FG) applied to both superior colliculi, to assess the state of their retrograde axonal transport. Retinas were dissected as wholemounts, surviving RGCs were immunoidentified with Brn3a, and the RNFL with phosphorylated high-molecular-weight subunit of the neurofilament triplet (pNFH) antibodies. ß-alanine supplementation decreases significantly taurine plasma levels and causes a significant reduction of the RNFL thickness that is increased after light exposure. An abnormal pNFH immunoreactivity in some RGC bodies, their proximal dendrites and axons, and a further diminution of the mean number of FG-traced RGCs compared with Brn3a+RGCs, indicate that their retrograde axonal transport is affected. In conclusion, taurine depletion causes RGC loss and axonal transport impairment. Finally, our results suggest that care should be taken when ingesting ß-alanine supplements due to the limited understanding of their potential adverse effects.


Assuntos
Transporte Axonal/efeitos dos fármacos , Luz/efeitos adversos , Fibras Nervosas/efeitos dos fármacos , Degeneração Retiniana/etiologia , Células Ganglionares da Retina/efeitos dos fármacos , Taurina/deficiência , beta-Alanina/toxicidade , Animais , Fibras Nervosas/metabolismo , Fibras Nervosas/patologia , Proteínas de Neurofilamentos/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Taurina/sangue , Tomografia de Coerência Óptica , Fator de Transcrição Brn-3A/metabolismo
12.
Vis Neurosci ; 36: E005, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-31199212

RESUMO

At the first retinal synapse, horizontal cells (HCs) contact both photoreceptor terminals and bipolar cell dendrites, modulating information transfer between these two cell types to enhance spatial contrast and mediate color opponency. The synaptic mechanisms through which these modulations occur are still debated. The initial hypothesis of a GABAergic feedback from HCs to cones has been challenged by pharmacological inconsistencies. Surround antagonism has been demonstrated to occur via a modulation of cone calcium channels through ephaptic signaling and pH changes in the synaptic cleft. GABAergic transmission between HCs and cones has been reported in some lower vertebrates, like the turtle and tiger salamander. In these reports, it was revealed that GABA is released from HCs through reverse transport and target GABA receptors are located at the cone terminals. In mammalian retinas, there is growing evidence that HCs can release GABA through conventional vesicular transmission, acting both on autaptic GABA receptors and on receptors expressed at the dendritic tips of the bipolar cells. The presence of GABA receptors on mammalian cone terminals remains equivocal. Here, we looked specifically for functional GABA receptors in mouse photoreceptors by recording in the whole-cell or amphotericin/gramicidin-perforated patch clamp configurations. Cones could be differentiated from rods through morphological criteria. Local GABA applications evoked a Cl- current in cones but not in rods. It was blocked by the GABAA receptor antagonist bicuculline methiodide and unaffected by the GABAC receptor antagonist TPMPA [(1,2,5,6-tetrahydropyridin-4-yl)methylphosphinic acid]. The voltage dependency of the current amplitude was as expected from a direct action of GABA on cone pedicles but not from an indirect modulation of cone currents following the activation of the GABA receptors of HCs. This supports a direct role of GABA released from HCs in the control of cone activity in the mouse retina.


Assuntos
Receptores de GABA-A/metabolismo , Receptores de GABA/metabolismo , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Horizontais da Retina/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Antagonistas GABAérgicos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Receptores de GABA/efeitos dos fármacos , Receptores de GABA-A/efeitos dos fármacos , Células Fotorreceptoras Retinianas Cones/metabolismo
13.
Mol Ther ; 25(11): 2546-2560, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28807567

RESUMO

The majority of inherited retinal degenerations converge on the phenotype of photoreceptor cell death. Second- and third-order neurons are spared in these diseases, making it possible to restore retinal light responses using optogenetics. Viral expression of channelrhodopsin in the third-order neurons under ubiquitous promoters was previously shown to restore visual function, albeit at light intensities above illumination safety thresholds. Here, we report (to our knowledge, for the first time) activation of macaque retinas, up to 6 months post-injection, using channelrhodopsin-Ca2+-permeable channelrhodopsin (CatCh) at safe light intensities. High-level CatCh expression was achieved due to a new promoter based on the regulatory region of the gamma-synuclein gene (SNCG) allowing strong expression in ganglion cells across species. Our promoter, in combination with clinically proven adeno-associated virus 2 (AAV2), provides CatCh expression in peri-foveolar ganglion cells responding robustly to light under the illumination safety thresholds for the human eye. On the contrary, the threshold of activation and the proportion of unresponsive cells were much higher when a ubiquitous promoter (cytomegalovirus [CMV]) was used to express CatCh. The results of our study suggest that the inclusion of optimized promoters is key in the path to clinical translation of optogenetics.


Assuntos
Channelrhodopsins/genética , Vetores Genéticos/administração & dosagem , Regiões Promotoras Genéticas , Recuperação de Função Fisiológica , Degeneração Retiniana/terapia , Animais , Channelrhodopsins/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Injeções Intravítreas , Luz , Macaca fascicularis , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Optogenética , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Transdução Genética , Transgenes , Visão Ocular/fisiologia
14.
Neuroimage ; 149: 267-274, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28167348

RESUMO

3D functional imaging of the whole brain activity during visual task is a challenging task in rodents due to the complex tri-dimensional shape of involved brain regions and the fine spatial and temporal resolutions required to reveal the visual tract. By coupling functional ultrasound (fUS) imaging with a translational motorized stage and an episodic visual stimulation device, we managed to accurately map and to recover the activity of the visual cortices, the Superior Colliculus (SC) and the Lateral Geniculate Nuclei (LGN) in 3D. Cerebral Blood Volume (CBV) responses during visual stimuli were found to be highly correlated with the visual stimulus time profile in visual cortices (r=0.6), SC (r=0.7) and LGN (r=0.7). These responses were found dependent on flickering frequency and contrast, and optimal stimulus parameters for largest CBV increases were obtained. In particular, increasing the flickering frequency higher than 7Hz revealed a decrease of visual cortices response while the SC response was preserved. Finally, cross-correlation between CBV signals exhibited significant delays (d=0.35s +/-0.1s) between blood volume response in SC and visual cortices in response to our visual stimulus. These results emphasize the interest of fUS imaging as a whole brain neuroimaging modality for brain vision studies in rodent models.


Assuntos
Mapeamento Encefálico/métodos , Imageamento Tridimensional/métodos , Ultrassonografia/métodos , Córtex Visual/diagnóstico por imagem , Vias Visuais/diagnóstico por imagem , Animais , Masculino , Ratos , Ratos Long-Evans
15.
Neurobiol Dis ; 100: 52-61, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28057519

RESUMO

The HANAC syndrome is caused by mutations in the gene coding for collagen4a1, a major component of blood vessel basement membranes. Ocular symptoms include an increase in blood vessel tortuosity and occasional hemorrhages. To examine how vascular defects can affect neuronal function, we analyzed the retinal phenotype of a HANAC mouse model. Heterozygous mutant mice displayed both a thinning of the basement membrane in retinal blood vessels and in Bruch's membrane resulting in vascular leakage. Homozygous mice had additional vascular changes, including greater vessel coverage and tortuosity. This greater tortuosity was associated to higher expression levels of vascular endothelial growth factor (VEGF). These major changes to the blood vessels were correlated with photoreceptor dysfunction and degeneration. The neuronal damage was associated with reactive gliosis in astrocytes and Müller glial cells, and by the migration of microglial cells into the outer retina. This study illustrates how vascular changes can trigger neuronal degeneration in a new model of HANAC syndrome that can be used to further study dysfunctions of neurovascular coupling. SUMMARY STATEMENT: This study provides a phenotypic analysis of a novel mouse model of HANAC syndrome focusing on the retinal aspect. It recapitulates most of the aspects of the human disease and is therefore a great tool to study and to address this condition.


Assuntos
Colágeno Tipo IV/genética , Cãibra Muscular/genética , Mutação/genética , Neurônios/patologia , Doença de Raynaud/genética , Vasos Retinianos/anormalidades , Animais , Modelos Animais de Doenças , Camundongos Transgênicos , Neuroglia/metabolismo , Neurônios/metabolismo , Retina/metabolismo , Vasos Retinianos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Adv Exp Med Biol ; 975 Pt 2: 687-701, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28849492

RESUMO

Retinal ganglion cell (RGC) degeneration occurs in numerous retinal diseases, either as a primary process like in glaucoma, or secondary to photoreceptor loss and no efficient compound targeting directly RGC neuroprotection is yet available. We previously described that taurine exerts a direct protective effect on RGCs cultured under serum-deprived conditions. Because taurine was known to have an agonist-like activity for GABA/glycine receptors, we investigated here if the taurine-elicited neuroprotective effect may be mediated through the activation of these receptors using selective antagonist ligands. RGCs were purified, seeded in 96-well plate and maintained in culture during 6 days in vitro. Viable cells were labelled with calcein and densities in full-well area were then automatically counted. Here we show that the protective effect of taurine against RGC loss observed under serum deprivation can be mediated through the GABAB receptor stimulation. Hence, two selective agonists, including baclofen, at this metabotropic GABAB receptor were found to reproduce taurine action by enhancing RGC survival in culture. This study suggests that GABAB receptor stimulation provides direct neuroprotection for RGCs. Accordingly, drugs targeting GABAB receptor may represent a new way for the prevention of RGC degeneration.


Assuntos
Agonistas dos Receptores de GABA-B/farmacologia , Fármacos Neuroprotetores/farmacologia , Receptores de GABA-B/efeitos dos fármacos , Células Ganglionares da Retina/efeitos dos fármacos , Taurina/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Masculino , Ratos , Ratos Long-Evans , Receptores de GABA-B/metabolismo , Células Ganglionares da Retina/metabolismo
17.
Hum Mol Genet ; 23(7): 1742-53, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24234651

RESUMO

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.


Assuntos
Anormalidades do Olho/genética , Fatores de Transcrição Otx/genética , Células Bipolares da Retina/citologia , Células Fotorreceptoras Retinianas Cones/citologia , Células Horizontais da Retina/citologia , Animais , Diferenciação Celular/genética , Linhagem Celular , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Penetrância , Acuidade Visual/genética
18.
Mol Ther ; 23(1): 7-16, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25095892

RESUMO

Most inherited retinal dystrophies display progressive photoreceptor cell degeneration leading to severe visual impairment. Optogenetic reactivation of retinal neurons mediated by adeno-associated virus (AAV) gene therapy has the potential to restore vision regardless of patient-specific mutations. The challenge for clinical translatability is to restore a vision as close to natural vision as possible, while using a surgically safe delivery route for the fragile degenerated retina. To preserve the visual processing of the inner retina, we targeted ON bipolar cells, which are still present at late stages of disease. For safe gene delivery, we used a recently engineered AAV variant that can transduce the bipolar cells after injection into the eye's easily accessible vitreous humor. We show that AAV encoding channelrhodopsin under the ON bipolar cell-specific promoter mediates long-term gene delivery restricted to ON-bipolar cells after intravitreal administration. Channelrhodopsin expression in ON bipolar cells leads to restoration of ON and OFF responses at the retinal and cortical levels. Moreover, light-induced locomotory behavior is restored in treated blind mice. Our results support the clinical relevance of a minimally invasive AAV-mediated optogenetic therapy for visual restoration.


Assuntos
Cegueira/terapia , Dependovirus/genética , Terapia Genética/métodos , Células Bipolares da Retina/metabolismo , Degeneração Retiniana/terapia , Animais , Comportamento Animal , Cegueira/genética , Cegueira/patologia , Channelrhodopsins , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Engenharia Genética , Vetores Genéticos , Injeções Intravítreas , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas , Células Bipolares da Retina/patologia , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Percepção Visual/genética , Corpo Vítreo
19.
Analyst ; 139(13): 3281-9, 2014 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-24699623

RESUMO

Direct interfacing of neurons with electronic devices has been investigated for both prosthetic and neuro-computing applications. In vitro neuronal networks provide great tools not only for improving neuroprostheses but also to take advantage of their computing abilities. However, it is often difficult to organize neuronal networks according to specific cell distributions. Our aim was to develop a cell-type specific immobilization of neurons on individual electrodes to produce organized in vitro neuronal networks on multi-electrode arrays (MEAs). We demonstrate the selective capture of retinal neurons on antibody functionalized surfaces following the formation of self-assembled monolayers from protein-thiol conjugates by simple contact and protein-polypyrrole deposits by electrochemical functionalization. This neuronal selection was achieved on gold for either cone photoreceptors or retinal ganglion neurons using a PNA lectin or a Thy1 antibody, respectively. Anti-fouling of un-functionalized gold surfaces was optimized to increase the capture efficiencies. The technique was extended to electrode arrays by addressing electropolymerization of pyrrole monomers and pyrrole-protein conjugates to active electrodes. Retinal ganglion cell recording on the array further demonstrated the integrity of these neurons following their selection on polypyrrole-coated electrodes. Therefore, this protein-polypyrrole electrodeposition could provide a new approach to generate organized in vitro neuronal networks.


Assuntos
Rede Nervosa , Células Ganglionares da Retina/citologia , Análise Serial de Tecidos/instrumentação , Animais , Anticorpos Imobilizados/química , Técnicas de Cultura de Células/instrumentação , Células Cultivadas , Células Imobilizadas/citologia , Desenho de Equipamento , Microeletrodos , Polimerização , Polímeros/química , Pirróis/química , Ratos Long-Evans , Compostos de Sulfidrila/química
20.
Nanoscale Horiz ; 9(4): 544-554, 2024 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-38323517

RESUMO

Current methodology used to investigate how shifts in brain states associated with regional cerebral blood volume (CBV) change in deep brain areas, are limited by either the spatiotemporal resolution of the CBV techniques, and/or compatibility with electrophysiological recordings; particularly in relation to spontaneous brain activity and the study of individual events. Additionally, infraslow brain signals (<0.1 Hz), including spreading depolarisations, DC-shifts and infraslow oscillations (ISO), are poorly captured by traditional AC-coupled electrographic recordings; yet these very slow brain signals can profoundly change CBV. To gain an improved understanding of how infraslow brain signals couple to CBV we present a new method for concurrent CBV with wide bandwidth electrophysiological mapping using simultaneous functional ultrasound imaging (fUS) and graphene-based field effect transistor (gFET) DC-coupled electrophysiological acquisitions. To validate the feasibility of this methodology visually-evoked neurovascular coupling (NVC) responses were examined. gFET recordings are not affected by concurrent fUS imaging, and epidural placement of gFET arrays within the imaging window did not deteriorate fUS signal quality. To examine directly the impact of infra-slow potential shifts on CBV, cortical spreading depolarisations (CSDs) were induced. A biphasic pattern of decreased, followed by increased CBV, propagating throughout the ipsilateral cortex, and a delayed decrease in deeper subcortical brain regions was observed. In a model of acute seizures, CBV oscillations were observed prior to seizure initiation. Individual seizures occurred on the rising phase of both infraslow brain signal and CBV oscillations. When seizures co-occurred with CSDs, CBV responses were larger in amplitude, with delayed CBV decreases in subcortical structures. Overall, our data demonstrate that gFETs are highly compatible with fUS and allow concurrent examination of wide bandwidth electrophysiology and CBV. This graphene-enabled technological advance has the potential to improve our understanding of how infraslow brain signals relate to CBV changes in control and pathological brain states.


Assuntos
Grafite , Humanos , Encéfalo/diagnóstico por imagem , Convulsões , Eletrofisiologia , Circulação Cerebrovascular/fisiologia , Ultrassonografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA