Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2708: 175-194, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37558971

RESUMO

The identification of distinct retinal ganglion cell (RGC) populations in flat-mounted retinas is key to investigating pathological or pharmacological effects in these cells. In this chapter, we review the main techniques for detecting the total population of RGCs and various of their subtypes in whole-mounted retinas of pigmented and albino rats and mice, four of the animal strains most studied by the scientific community in the retina field. These methods are based on the studies published by the Vidal-Sanz's laboratory.


Assuntos
Retina , Células Ganglionares da Retina , Ratos , Camundongos , Animais , Células Ganglionares da Retina/patologia , Retina/patologia
2.
bioRxiv ; 2023 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-36909547

RESUMO

Specific wiring is essential for sensory systems to precisely relay information to higher brain regions. The retina, an approachable part of the brain, is an ideal model for studying neural circuits due to its well-organized structure. In the retina, S-cone photoreceptors sense and relay short-wavelength (e.g., blue) light signals for encoding color information and other environmental cues. S-cones usually account for less than 10% of cones and are precisely connected to S-cone bipolar cells (SCBCs). This connection is ancient and highly conserved across species, indicating essential functions. How this wiring specificity is formed and maintained, however, is not understood. To unveil the molecular mechanisms underlying this highly specific connection, we sequenced the transcriptomes of thirteen-lined ground squirrel (TLGS) photoreceptors. We chose TLGS for their cone-rich retina and the absence of cones that co-express multiple opsin proteins, as compared to mice. We used a targeted SMART-seq approach to obtain high-resolution transcriptomes from S- and M-cone photoreceptors and identified a cell-adhesion molecule, Nrxn3, as a potential candidate mediating the S-cone to SCBC connection. Given the limitations of genetic manipulation in TLGS, we utilized mouse models to study the function of Nrxn3 in S-cones. In 'true' S-cones (S-opsin+/M-opsin-) that lack Nrxn3 expression, the number of connections with SCBCs was drastically reduced, indicating a critical role of Nrxn3 for this synapse. While neurexins are well known for their diverse roles in regulating various synapses, this study is the first to document its crucial role in mediating or maintaining a specific synapse in the central nervous system. In addition, the differentially expressed genes identified here provide a valuable resource for further investigating cone subtype-specific functions.

3.
Zool Res ; 44(1): 226-248, 2023 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-36594396

RESUMO

Univocal identification of retinal ganglion cells (RGCs) is an essential prerequisite for studying their degeneration and neuroprotection. Before the advent of phenotypic markers, RGCs were normally identified using retrograde tracing of retinorecipient areas. This is an invasive technique, and its use is precluded in higher mammals such as monkeys. In the past decade, several RGC markers have been described. Here, we reviewed and analyzed the specificity of nine markers used to identify all or most RGCs, i.e., pan-RGC markers, in rats, mice, and macaques. The best markers in the three species in terms of specificity, proportion of RGCs labeled, and indicators of viability were BRN3A, expressed by vision-forming RGCs, and RBPMS, expressed by vision- and non-vision-forming RGCs. NEUN, often used to identify RGCs, was expressed by non-RGCs in the ganglion cell layer, and therefore was not RGC-specific. γ-SYN, TUJ1, and NF-L labeled the RGC axons, which impaired the detection of their somas in the central retina but would be good for studying RGC morphology. In rats, TUJ1 and NF-L were also expressed by non-RGCs. BM88, ERRß, and PGP9.5 are rarely used as markers, but they identified most RGCs in the rats and macaques and ERRß in mice. However, PGP9.5 was also expressed by non-RGCs in rats and macaques and BM88 and ERRß were not suitable markers of viability.


Assuntos
Traumatismos do Nervo Óptico , Ratos , Camundongos , Animais , Células Ganglionares da Retina , Macaca mulatta , Traumatismos do Nervo Óptico/veterinária , Retina , Mamíferos , Biomarcadores
5.
Dev Cell ; 56(20): 2871-2885.e6, 2021 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-34428430

RESUMO

The mechanism of vulnerability to pediatric low-grade gliomas (pLGGs)-the most common brain tumor in children-during development remains largely unknown. Using mouse models of neurofibromatosis type 1 (NF1)-associated pLGGs in the optic pathway (NF1-OPG), we demonstrate that NF1-OPG arose from the vulnerability to the dependency of Mek-Erk/MAPK signaling during gliogenesis of one of the two developmentally transient precursor populations in the optic nerve, brain-derived migrating glial progenitors (GPs), but not local progenitors. Hyperactive Erk/MAPK signaling by Nf1 loss overproduced GPs by disrupting the balance between stem-cell maintenance and gliogenesis of hypothalamic ventricular zone radial glia (RG). Persistence of RG-like GPs initiated NF1-OPG, causing Bax-dependent apoptosis in retinal ganglion cells. Removal of three Mek1/Mek2 alleles or transient post-natal treatment with a low-dose MEK inhibitor normalized differentiation of Nf1-/- RG-like GPs, preventing NF1-OPG formation and neuronal degeneration. We provide the proof-of-concept evidence for preventing pLGGs before tumor-associated neurological damage enters an irreversible phase.


Assuntos
Sistema de Sinalização das MAP Quinases/fisiologia , Neurofibromatose 1/terapia , Glioma do Nervo Óptico/terapia , Células-Tronco/citologia , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Modelos Animais de Doenças , Olho/metabolismo , Camundongos , Neurofibromatose 1/metabolismo , Neurofibromatose 1/patologia , Neuroglia/metabolismo , Nervo Óptico/patologia , Glioma do Nervo Óptico/metabolismo , Glioma do Nervo Óptico/patologia
6.
Lab Invest ; 101(9): 1289-1303, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34253851

RESUMO

Retinal ganglion cell (RGC) death occurs after optic nerve injury due to acute trauma or chronic degenerative conditions such as optic neuropathies (e.g., glaucoma). Currently, there are no effective therapies to prevent permanent vision loss resulting from RGC death, underlining the need for research on the pathogenesis of RGC disorders. Modeling human RGC/optic nerve diseases in non-human primates is ideal because of their similarity to humans, but has practical limitations including high cost and ethical considerations. In addition, many retinal degenerative disorders are age-related making the study in primate models prohibitively slow. For these reasons, mice and rats are commonly used to model RGC injuries. However, as nocturnal mammals, these rodents have retinal structures that differ from primates - possessing less than one-tenth of the RGCs found in the primate retina. Here we report the diurnal thirteen-lined ground squirrel (TLGS) as an alternative model. Compared to other rodent models, the number and distribution of RGCs in the TLGS retina are closer to primates. The TLGS retina possesses ~600,000 RGCs with the highest density along the equatorial retina matching the location of the highest cone density (visual streak). TLGS and primate retinas also share a similar interlocking pattern between RGC axons and astrocyte processes in the retina nerve fiber layer (RNFL). In addition, using TLGS we establish a new partial optic nerve injury model that precisely controls the extent of injury while sparing a portion of the retina as an ideal internal control for investigating the pathophysiology of axon degeneration and RGC death. Moreover, in vivo optical coherence tomography (OCT) imaging and ex vivo microscopic examinations of the retina in optic nerve injured TLGS confirm RGC loss precedes proximal axon degeneration, recapitulating human pathology. Thus, the TLGS retina is an excellent model, for translational research in neurodegeneration and therapeutic neuroprotection.


Assuntos
Modelos Animais de Doenças , Doenças do Nervo Óptico , Doenças Retinianas , Células Ganglionares da Retina , Sciuridae/fisiologia , Animais , Feminino , Macaca mulatta , Camundongos , Ratos , Retina/citologia , Retina/patologia , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/patologia , Células Ganglionares da Retina/fisiologia
7.
J Neurosci ; 2021 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-34083252

RESUMO

Amacrine cells are interneurons composing the most diverse cell class in the mammalian retina. They help encode visual features such as edges or directed motion by mediating excitatory and inhibitory interactions between input (i.e. bipolar) and output (i.e. ganglion) neurons in the inner plexiform layer (IPL). Like other brain regions, the retina also contains glial cells that contribute to neurotransmitter uptake, metabolic regulation and neurovascular control. Here, we report that in mouse retina (of either sex), an abundant, though previously unstudied inhibitory amacrine cell is coupled directly to Müller glia. Electron microscopic reconstructions of this amacrine type revealed chemical synapses with known retinal cell types and extensive associations with Müller glia, the processes of which often completely ensheathe the neurites of this amacrine cell. Microinjecting small tracer molecules into the somas of these amacrine cells led to selective labelling of nearby Müller glia, leading us to suggest the name "Müller glia-coupled amacrine cell," or MAC. Our data also indicate that MACs release glycine at conventional chemical synapses, and viral retrograde transsynaptic tracing from the dorsal lateral geniculate nucleus (dLGN) showed selective connections between MACs and a subpopulation of RGC types. Visually-evoked responses revealed a strong preference for light increments; these "ON" responses were primarily mediated by excitatory chemical synaptic input and direct electrical coupling with other cells. This initial characterization of the MAC provides the first evidence for neuron-glia coupling in the mammalian retina and identifies the MAC as a potential link between inhibitory processing and glial function.Significance Statement:Gap junctions between pairs of neurons or glial cells are commonly found throughout the nervous system and play multiple roles, including electrical coupling and metabolic exchange. In contrast, gap junctions between neurons and glia cells have rarely been reported and are poorly understood. Here we report the first evidence for neuron-glia coupling in the mammalian retina, specifically between an abundant (but previously unstudied) inhibitory interneuron and Müller glia. Moreover, viral tracing, optogenetics and serial electron microscopy provide new information about the neuron's synaptic partners and physiological responses.

8.
Elife ; 92020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32463363

RESUMO

Color, an important visual cue for survival, is encoded by comparing signals from photoreceptors with different spectral sensitivities. The mouse retina expresses a short wavelength-sensitive and a middle/long wavelength-sensitive opsin (S- and M-opsin), forming opposing, overlapping gradients along the dorsal-ventral axis. Here, we analyzed the distribution of all cone types across the entire retina for two commonly used mouse strains. We found, unexpectedly, that 'true S-cones' (S-opsin only) are highly concentrated (up to 30% of cones) in ventral retina. Moreover, S-cone bipolar cells (SCBCs) are also skewed towards ventral retina, with wiring patterns matching the distribution of true S-cones. In addition, true S-cones in the ventral retina form clusters, which may augment synaptic input to SCBCs. Such a unique true S-cone and SCBC connecting pattern forms a basis for mouse color vision, likely reflecting evolutionary adaptation to enhance color coding for the upper visual field suitable for mice's habitat and behavior.


Many primates, including humans, can see color better than most other mammals. This difference is due to the variety of light-detecting proteins ­ called opsins ­ that are produced in the eye by cells known as cones. While humans have three, mice only have two different opsins, known as S and M, which detect blue/UV and green light, respectively. Mouse cones produce either S-opsins, M-opsins or both. Fewer than 10 percent of cone cells in mice produce just the S-opsin, and these cells are essential for color vision. Mice are commonly used in scientific research, and so their vision has been well studied. However, previous research has produced conflicting results. Some studies report that cone cells that contain only S-opsin are evenly spread out across the retina. Other evidence suggests that color vision in mice exists only for the upper field of their vision, in other words, that mice can only distinguish colors that appeared above them. Nadal-Nicolás et al. set out to understand how to reconcile these contrasting findings. Molecular tools were used to detect S- and M-opsin in the retina of mice and revealed large differences between the lower part, known as the ventral retina, and the upper part, known as the dorsal retina. The ventral retina detects light coming from above the animal, and about a third of cone cells in this region produced exclusively S-opsin, compared to only 1 percent of cones in the dorsal retina. These S-opsin cone cells in the ventral retina group into clusters, where they connect with a special type of nerve cells that transmit this signal. To better understand these findings, Nadal-Nicolás et al. also studied albino mice. Although albino mice have a different distribution of S-opsin protein in the retina, the cone cells producing only S-opsin are similarly clustered in the ventral retina. This suggests that the concentration of S-opsin cone cells in the ventral retina is an important feature in mouse sight. This new finding corrects the misconception that S-opsin-only cone cells are evenly spread throughout the retina and supports the previous evidence that mouse color vision is greatest in the upper part of their field of vision. Nadal-Nicolás et al. suggest this arrangement could help the mice to detect predators that may attack them from above during the daytime. Together, these new findings could help to improve the design of future studies involving vision in mice and potentially other similar species.


Assuntos
Visão de Cores , Retina/metabolismo , Opsinas de Bastonetes/metabolismo , Animais , Opsinas dos Cones/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Campos Visuais
9.
Int J Mol Sci ; 20(12)2019 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-31226772

RESUMO

We studied short- and long-term effects of intravitreal injection of N-methyl-d-aspartate (NMDA) on melanopsin-containing (m+) and non-melanopsin-containing (Brn3a+) retinal ganglion cells (RGCs). In adult SD-rats, the left eye received a single intravitreal injection of 5µL of 100nM NMDA. At 3 and 15 months, retinal thickness was measured in vivo using Spectral Domain-Optical Coherence Tomography (SD-OCT). Ex vivo analyses were done at 3, 7, or 14 days or 15 months after damage. Whole-mounted retinas were immunolabelled for brain-specific homeobox/POU domain protein 3A (Brn3a) and melanopsin (m), the total number of Brn3a+RGCs and m+RGCs were quantified, and their topography represented. In control retinas, the mean total numbers of Brn3a+RGCs and m+RGCs were 78,903 ± 3572 and 2358 ± 144 (mean ± SD; n = 10), respectively. In the NMDA injected retinas, Brn3a+RGCs numbers diminished to 49%, 28%, 24%, and 19%, at 3, 7, 14 days, and 15 months, respectively. There was no further loss between 7 days and 15 months. The number of immunoidentified m+RGCs decreased significantly at 3 days, recovered between 3 and 7 days, and were back to normal thereafter. OCT measurements revealed a significant thinning of the left retinas at 3 and 15 months. Intravitreal injections of NMDA induced within a week a rapid loss of 72% of Brn3a+RGCs, a transient downregulation of melanopsin expression (but not m+RGC death), and a thinning of the inner retinal layers.


Assuntos
N-Metilaspartato/toxicidade , Células Ganglionares da Retina/efeitos dos fármacos , Opsinas de Bastonetes/metabolismo , Animais , Contagem de Células , Feminino , Injeções Intravítreas , N-Metilaspartato/administração & dosagem , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Opsinas de Bastonetes/análise , Fator de Transcrição Brn-3A/análise , Fator de Transcrição Brn-3A/metabolismo
10.
Exp Eye Res ; 182: 156-159, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30940447

RESUMO

Optic nerve axotomy in rodents allows detailed studies of the effect of different treatments on the survival of central nervous system neurons, the retinal ganglion cells (RGCs). Here we have analyzed the neuroprotective effect of topical bromfenac treatment, a nonsteroidal anti-inflammatory drug (NSAID) used in clinic to ameliorate post-operative inflammation, on axotomized rat RGCs. The left optic nerve of adult rats was subjected to optic nerve crush (ONC). Half of the rats were treated with a topical instillation of saline. On the other half, immediately after the surgery, 2 drops of bromfenac (0.09% Yellox; Bausch & Lomb) were instilled, and then every 12 h until analysis. Retinas in both groups were dissected 3, 5, 7, 9 and 14 days after ONC (n = 4-8/time point/group). Toxicity of bromfenac was assessed in intact retinas treated during 14 days (n = 6). Intact untreated retinas were used as control of the RGC population. RGCs were identified by Brn3a immunodetection and automatically quantified. Our results show that bromfenac does not cause RGC loss in intact retinas. In the injured groups, the number of RGCs at 7, 9 and 14 days after the lesion was significantly higher in treated vs. untreated retinas. To our knowledge this is the first report showing that a topical treatment with a NSAIDs delays axotomy-induced RGC loss and indicates that treatment with NSAIDs could be used as conjunctive therapy in diseases that proceed with optic nerve damage.


Assuntos
Benzofenonas/administração & dosagem , Bromobenzenos/administração & dosagem , Traumatismos do Nervo Óptico/tratamento farmacológico , Nervo Óptico/patologia , Células Ganglionares da Retina/efeitos dos fármacos , Administração Tópica , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Axotomia , Contagem de Células , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Traumatismos do Nervo Óptico/patologia , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/patologia
12.
Sci Rep ; 8(1): 16299, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30389962

RESUMO

Mesenchymal stem cell (MSC) transplantation is emerging as an ideal tool to restore the wounded central nervous system (CNS). MSCs isolated from extra-embryonic tissues have some advantages compared to MSCs derived from adult ones, such as an improved proliferative capacity, life span, differentiation potential and immunomodulatory properties. In addition, they are more immunoprivileged, reducing the probability of being rejected by the recipient. Umbilical cords (UCs) are a good source of MSCs because they are abundant, safe, non-invasively harvested after birth and, importantly, they are not encumbered with ethical problems. Here we show that the intravitreal transplant of Wharton´s jelly mesenchymal stem cells isolated from three different human UCs (hWJMSCs) delays axotomy-induced retinal ganglion cell (RGC) loss. In vivo, hWJMSCs secrete anti-inflammatory molecules and trophic factors, the latter alone may account for the elicited neuroprotection. Interestingly, this expression profile differs between naive and injured retinas, suggesting that the environment in which the hWJMSCs are modulates their secretome. Finally, even though the transplant itself is not toxic for RGCs, it is not innocuous as it triggers a transient but massive infiltration of Iba1+cells from the choroid to the retina that alters the retinal structure.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Degeneração Retiniana/terapia , Células Ganglionares da Retina/patologia , Geleia de Wharton/citologia , Animais , Anti-Inflamatórios/metabolismo , Axotomia/efeitos adversos , Modelos Animais de Doenças , Feminino , Humanos , Injeções Intravítreas , Células-Tronco Mesenquimais/metabolismo , Fatores de Crescimento Neural/metabolismo , Ratos , Ratos Sprague-Dawley , Degeneração Retiniana/etiologia , Degeneração Retiniana/patologia , Resultado do Tratamento , Cordão Umbilical/citologia
13.
Adv Exp Med Biol ; 1074: 457-464, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29721976

RESUMO

Oxidative stress has been implicated in neurodegenerative diseases, such as age-related macular degeneration. Hydrogen peroxide and sodium iodate can mediate oxidative injury. Sodium iodate induces a selective retinal degeneration targeting the RPE. We describe a method of chronic sodium iodate-mediated injury on RPE cells that may serve to evaluate protective factors against oxidative stress. Cytotoxicity and cell viability curves of ARPE-19 cells with sodium iodate were generated. The antioxidant pigment epithelium-derived factor decreased sodium iodate-mediated cytotoxicity without affecting ARPE-19 cell viability. A cell culture system to evaluate protection against oxidative stress injury with PEDF is discussed.


Assuntos
Antioxidantes/farmacologia , Proteínas do Olho/farmacologia , Fatores de Crescimento Neural/farmacologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Serpinas/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Células Epiteliais/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/toxicidade , Iodatos/toxicidade , Degeneração Macular/patologia , Estresse Oxidativo , Proteínas Recombinantes/farmacologia , Epitélio Pigmentado da Retina/citologia
14.
Neurobiol Aging ; 61: 146-168, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29080498

RESUMO

In healthy beings, age is the ultimate reason of cellular malfunction and death. In the rat retina, age causes a functional decline and loss of specific neuronal populations. In this regard, controversial conclusions have been reported for the innermost retina. Here, we have studied the albino and pigmented retina for the duration of the rat life-span. Independent of age (21 days-22 months), the electroretinographic recordings and the volume of the retina and its layers are smaller in albinos. Functionally, aging causes in both strains a loss of cone- and rod-mediated responses. Anatomically, cell density decreases with age because the retina grows linearly with time; no cell loss is observed in the ganglion cell layer; and only in the pigmented rat, there is a decrease in cone photoreceptors. In old animals of both strains, there is gliosis in the superior colliculi and a diminution of the area innervated by retinal ganglion cells. In conclusion, this work provides the basis for further studies linking senescence to neurodegenerative retinal diseases.


Assuntos
Envelhecimento/patologia , Envelhecimento/fisiologia , Retina/patologia , Retina/fisiopatologia , Animais , Transporte Axonal , Eletrorretinografia , Feminino , Microglia , Ratos Sprague-Dawley , Retina/citologia , Retina/diagnóstico por imagem , Células Fotorreceptoras Retinianas Cones/patologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Segmento Interno das Células Fotorreceptoras da Retina/patologia , Segmento Interno das Células Fotorreceptoras da Retina/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/patologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Tomografia de Coerência Óptica
15.
J Neuroinflammation ; 14(1): 218, 2017 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-29121969

RESUMO

BACKGROUND: Microglial cells (MCs) are the sentries of the central nervous system. In health, they are known as surveying MCs because they examine the tissue to maintain the homeostasis. In disease, they activate and, among other functions, become phagocytic to clean the cellular debris. In this work, we have studied the behavior of rat retinal MCs in two models of unilateral complete intraorbital optic nerve axotomy which elicit a different time course of retinal ganglion cell (RGC) loss. METHODS: Albino Sprague-Dawley rats were divided into these groups: (a) intact (no surgery), (b) fluorogold (FG) tracing from the superior colliculi, and (c) FG tracing + crush or transection of the left optic nerve. The retinas were dissected from 2 days to 2 months after the lesions (n = 4-12 group/lesion and time point) and then were subjected to Brn3a and Iba1 double immunodetection. In each intact retina, the total number of Brn3a+RGCs and Iba+MCs was quantified. In each traced retina (b and c groups), FG-traced RGCs and phagocytic microglial cells (PMCs, FG+Iba+) were also quantified. Topographical distribution was assessed by neighbor maps. RESULTS: In intact retinas, surveying MCs are homogenously distributed in the ganglion cell layer and the inner plexiform layer. Independently of the axotomy model, RGC death occurs in two phases, one quick and one protracted, and there is a lineal and topographical correlation between the appearance of PMCs and the loss of traced RGCs. Furthermore, the clearance of FG+RGCs by PMCs occurs 3 days after the actual loss of Brn3a expression that marks RGC death. In addition, almost 50% of MCs from the inner plexiform layer migrate to the ganglion cell layer during the quick phase of RGC loss, returning to the inner plexiform layer during the slow degeneration phase. Finally, in contrast to what happens in mice, in rats, there is no microglial phagocytosis in the contralateral uninjured retina. CONCLUSIONS: Axotomy-induced RGC death occurs earlier than RGC clearance and there is an inverse correlation between RGC loss and PMC appearance, both numerically and topographically, suggesting that phagocytosis occurs as a direct response to RGC death rather than to axonal damage.


Assuntos
Microglia/metabolismo , Traumatismos do Nervo Óptico/patologia , Fagocitose/fisiologia , Células Ganglionares da Retina/patologia , Animais , Axotomia , Morte Celular , Feminino , Nervo Óptico/patologia , Nervo Óptico/cirurgia , Ratos , Ratos Sprague-Dawley
16.
Front Neurosci ; 11: 235, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28491019

RESUMO

Glaucoma, one of the leading causes of blindness worldwide, affects primarily retinal ganglion cells (RGCs) and their axons. The pathophysiology of glaucoma is not fully understood, but it is currently believed that damage to RGC axons at the optic nerve head plays a major role. Rodent models to study glaucoma include those that mimic either ocular hypertension or optic nerve injury. Here we review the anatomical loss of the general population of RGCs (that express Brn3a; Brn3a+RGCs) and of the intrinsically photosensitive RGCs (that express melanopsin; m+RGCs) after chronic (LP-OHT) or acute (A-OHT) ocular hypertension and after complete intraorbital optic nerve transection (ONT) or crush (ONC). Our studies show that all of these insults trigger RGC death. Compared to Brn3a+RGCs, m+RGCs are more resilient to ONT, ONC, and A-OHT but not to LP-OHT. There are differences in the course of RGC loss both between these RGC types and among injuries. An important difference between the damage caused by ocular hypertension or optic nerve injury appears in the outer retina. Both axotomy and LP-OHT induce selective loss of RGCs but LP-OHT also induces a protracted loss of cone photoreceptors. This review outlines our current understanding of the anatomical changes occurring in rodent models of glaucoma and discusses the advantages of each one and their translational value.

17.
Acta Ophthalmol ; 95(1): e10-e21, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27535721

RESUMO

PURPOSE: To analyse miRNA regulation in a rat model of acute ocular hypertension (AOH). METHODS: Acute ocular hypertension (AOH) was induced in the left eye of adult albino rats by inserting a cannula connected with a saline container into the anterior chamber. The contralateral eye served as a control. Seven days later, animals were killed. Retinas were used either for quantitative analysis of retinal ganglion cells (RGCs) and microglial cells or for miRNA array hybridization, qRT-PCR and Western blotting. RESULTS: Anatomically, AOH caused axonal degeneration, a significant loss of RGCs and a significant increase in microglial cells in the ganglion cell layer. The miRNAs microarray analysis revealed 31 differentially expressed miRNAs in the AOH versus control group, and the regulation of 12 selected microRNAs was further confirmed by qRT-PCR. Bioinformatic analysis indicates that several signalling pathways are putatively regulated by the validated miRNAs. Of particular interest was the inflammatory pathway signalled by mitogen-activated protein kinases (MAPKs). In agreement with the in silico analysis, p38 MAP kinase, tumour necrosis factor-alpha (TNF-α) and iNOS proteins were significantly upregulated in the AOH retinas. CONCLUSIONS: Acute IOP elevation led to changes in the expression of miRNAs, whose target genes were associated with the regulation of microglia-mediated neuroinflammation or neural apoptosis. Addressing miRNAs in the process of retinal ischaemia and optic nerve damage in association with high IOP elevation may open new avenues in preventing retinal ganglion cell apoptosis and may serve as target for future therapeutic regimen in acute ocular hypertension and retinal ischaemic conditions.


Assuntos
Modelos Animais de Doenças , MicroRNAs/metabolismo , Microglia/metabolismo , Hipertensão Ocular/metabolismo , Células Ganglionares da Retina/metabolismo , Doença Aguda , Animais , Western Blotting , Sobrevivência Celular , Feminino , Pressão Intraocular , Microglia/patologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Hibridização de Ácido Nucleico , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Células Ganglionares da Retina/patologia , Transdução de Sinais , Regulação para Cima
18.
Invest Ophthalmol Vis Sci ; 57(15): 6652-6661, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27930778

RESUMO

Purpose: To analyze the responses of different retinal ganglion cell (RGC) types to acute ocular hypertension (AOH) and intravitreal administration of brain-derived neurotrophic factor (BDNF). Methods: In adult albino rats, the anterior chamber of the left eye was cannulated with a needle connected to a saline container elevated 1½ meters above the eye for 75 minutes. Rats received 12 hours before a 5 µl intravitreal injection containing 5 µg BDNF in 1% albumin PBS or vehicle and were analyzed 3, 7, 14, or 45 days later. Both retinas were dissected as wholemounts and immunolabeled for melanopsin (to identify intrinsically photosensitive RGCs) or Brn3a (to identify all RGCs except melanopsin +RGCs). Results: During AOH there is ischemic damage and mechanical eye-globe deformation. Acute ocular hypertension results in a progressive loss of Brn3a+RGCs in the vehicle-treated retinas (39%, 35%, 25%, and 13% of the original value, at 3, 7, 14, or 45 days, respectively), whereas BDNF increases their survival to 81%, 73%, 59%, or 57% at the same time periods. In vehicle-treated retinas, 37% or 39% of m+RGCs survive at 14 or 45 days, respectively, whereas BDNF treatment increases their survival to 40% or 78% at the same time points. Conclusions: Different types of RGCs respond differently to AOH because Brn3a+RGCs die progressively, but m+RGCs do not. After a transient downregulation of melanopsin expression, their number remains constant and their survival is proportionally higher than that of Brn3a+RGCs. BDNF affords a permanent protection up to 45 days after AOH injury in both types of RGCs.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/administração & dosagem , Pressão Intraocular/fisiologia , Neuroproteção , Hipertensão Ocular/diagnóstico , Doenças Retinianas/prevenção & controle , Células Ganglionares da Retina/metabolismo , Opsinas de Bastonetes/metabolismo , Doença Aguda , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Injeções Intravítreas , Hipertensão Ocular/tratamento farmacológico , Hipertensão Ocular/metabolismo , Ratos , Ratos Sprague-Dawley , Doenças Retinianas/diagnóstico , Doenças Retinianas/metabolismo , Células Ganglionares da Retina/patologia , Ultrassonografia Doppler
19.
Sci Rep ; 6: 38499, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27929040

RESUMO

Axonal injury is a common feature of central nervous system insults that culminates with the death of the affected neurons, and an irreversible loss of function. Inflammation is an important component of the neurodegenerative process, where the microglia plays an important role by releasing proinflammatory factors as well as clearing the death neurons by phagocytosis. Here we have identified the purinergic signaling through the P2X7 receptor as an important component for the neuronal death in a model of optic nerve axotomy. We have found that in P2X7 receptor deficient mice there is a delayed loss of retinal ganglion cells and a decrease of phagocytic microglia at early times points after axotomy. In contralateral to the axotomy retinas, P2X7 receptor controlled the numbers of phagocytic microglia, suggesting that extracellular ATP could act as a danger signal activating the P2X7 receptor in mediating the loss of neurons in contralateral retinas. Finally, we show that intravitreal administration of the selective P2X7 receptor antagonist A438079 also delays axotomy-induced retinal ganglion cell death in retinas from wild type mice. Thus, our work demonstrates that P2X7 receptor signaling is involved in neuronal cell death after axonal injury, being P2X7 receptor antagonism a potential therapeutic strategy.


Assuntos
Degeneração Neural/metabolismo , Traumatismos do Nervo Óptico/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Animais , Axotomia , Contagem de Células , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Degeneração Neural/patologia , Nervo Óptico/efeitos dos fármacos , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Traumatismos do Nervo Óptico/patologia , Fagocitose/efeitos dos fármacos , Antagonistas do Receptor Purinérgico P2X/farmacologia , Piridinas/farmacologia , Receptores Purinérgicos P2X7/deficiência , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Tetrazóis/farmacologia , Fatores de Tempo
20.
Invest Ophthalmol Vis Sci ; 57(14): 6098-6106, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27832276

RESUMO

PURPOSE: To study the effect of topical administration of bromfenac, a nonsteroidal anti-inflammatory drug (NSAID), on retinal gliosis and levels of prostaglandin E2 (PGE2) after complete optic nerve crush (ONC). METHODS: Adult albino rats were divided into the following groups (n = 8 retinas/group): (1) intact, (2) intact and bromfenac treatment (twice a day during 7 days), (3) ONC (7 days), and (4) ONC (7 days) + bromfenac treatment (twice a day during 7 days). Animals from groups 3 and 4 were imaged in vivo with spectral-domain optical coherence tomography (SD-OCT) before the procedure and 15 minutes, 3, 5, or 7 days later. Retinas from all groups were analyzed by immunodetection, Western blotting, or enzyme-linked immunoabsorbent assay (ELISA). RESULTS: Quantification of Brn3a (brain-specific homeobox/POU domain protein 3A) +RGCs (retinal ganglion cells) in cross sections showed that bromfenac treatment does not accelerate ONC-induced degeneration. Cellular retinaldehyde binding protein 1 regulation indicated that bromfenac improves retinal homeostasis in injured retinas. Spectral-domain OCT showed that the thickness of the retina and the retinal nerve fiber layer at 7 days post ONC was significantly reduced in bromfenac-treated animals when compared to untreated animals. In agreement with these data, hypertrophy of astrocytes and Müller cells and expression of glial fibrillary acidic protein and vimentin were greatly diminished by bromfenac treatment. While no changes in cyclooxygenase (COX) enzyme COX1 and COX2 expression were observed, there was a significant increase of PGE2 after ONC that was controlled by bromfenac treatment. CONCLUSIONS: Topical administration of bromfenac is an efficient and noninvasive treatment to control the retinal gliosis and release of proinflammatory mediators that follow a massive insult to the RGC population.


Assuntos
Benzofenonas/administração & dosagem , Bromobenzenos/administração & dosagem , Traumatismos do Nervo Óptico/tratamento farmacológico , Neurite Óptica/tratamento farmacológico , Células Ganglionares da Retina/patologia , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Western Blotting , Contagem de Células , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Gliose/tratamento farmacológico , Gliose/etiologia , Gliose/patologia , Soluções Oftálmicas , Disco Óptico/efeitos dos fármacos , Disco Óptico/metabolismo , Disco Óptico/patologia , Traumatismos do Nervo Óptico/complicações , Traumatismos do Nervo Óptico/patologia , Neurite Óptica/etiologia , Neurite Óptica/patologia , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Tomografia de Coerência Óptica/métodos , Fator de Transcrição Brn-3A/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...