Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
iScience ; 26(8): 107262, 2023 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-37609630

RESUMO

In the nervous system, parallel circuits are organized in part by the lamina-specific compartmentalization of synaptic connections. In sensory systems such as mammalian retina, degenerating third-order neurons remodel their local presynaptic connectivity with second-order neurons. To determine whether there are sublamina-specific perturbations after injury of adult retinal ganglion cells, we comprehensively analyzed excitatory synapses across the inner plexiform layer (IPL) where bipolar cells connect to ganglion cells. Here, we show that pre- and postsynaptic component loss occurs throughout the IPL in a sublamina-dependent fashion after transient intraocular pressure elevation. Partnered synaptic components are lost as neurodegeneration progresses, while unpartnered synaptic components remain stable. Furthermore, presynaptic components are either lost first or simultaneously with the postsynaptic component. Our results demonstrate that this degenerating neural circuit exhibits differential vulnerability of excitatory synapses depending on IPL depth, highlighting the ordered disassembly of synapses that is specific to laminar compartments of the retina.

2.
Front Pharmacol ; 14: 1112318, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36755945

RESUMO

Introduction: Ocular and periocular traumatisms may result in loss of vision. Our previous work showed that therapeutic hypothermia prevents retinal damage caused by traumatic neuropathy. We also generated and characterized small molecules that elicit the beneficial effects of hypothermia at normal body temperature. Here we investigate whether one of these mimetic molecules, zr17-2, is able to preserve the function of eyes exposed to trauma. Methods: Intraorbital optic nerve crush (IONC) or sham manipulation was applied to Sprague-Dawley rats. One hour after surgery, 5.0 µl of 330 nmol/L zr17-2 or PBS, as vehicle, were injected in the vitreum of treated animals. Electroretinograms were performed 21 days after surgery and a- and b-wave amplitude, as well as oscillatory potentials (OP), were calculated. Some animals were sacrificed 6 days after surgery for TUNEL analysis. All animal experiments were approved by the local ethics board. Results: Our previous studies showed that zr17-2 does not cross the blood-ocular barrier, thus preventing systemic treatment. Here we show that intravitreal injection of zr17-2 results in a very significant prevention of retinal damage, providing preclinical support for its pharmacological use in ocular conditions. As previously reported, IONC resulted in a drastic reduction in the amplitude of the b-wave (p < 0.0001) and OPs (p < 0.05), a large decrease in the number of RGCs (p < 0.0001), and a large increase in the number of apoptotic cells in the GCL and the INL (p < 0.0001). Interestingly, injection of zr17-2 largely prevented all these parameters, in a very similar pattern to that elicited by therapeutic hypothermia. The small molecule was also able to reduce oxidative stress-induced retinal cell death in vitro. Discussion: In summary, we have shown that intravitreal injection of the hypothermia mimetic, zr17-2, significantly reduces the morphological and electrophysiological consequences of ocular traumatism and may represent a new treatment option for this cause of visual loss.

3.
Front Pharmacol ; 14: 1252184, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37767403

RESUMO

Introduction: Perinatal asphyxia (PA) represents a major problem in perinatology and may cause visual losses, including blindness. We, and others, have shown that hypothermia prevents retinal symptoms associated to PA. In the present work, we evaluate whether a hypothermia mimetic small molecule, zr17-2, has similar effects in the context of PA. Methods: Four experimental groups were studied in male rats: Naturally born rats as controls (CTL), naturally born rats injected s.c. with 50 µL of 330 nmols/L zr17-2 (ZR), animals that were exposed to PA for 20 min at 37°C (PA), and rats that were exposed to PA and injected with zr17-2 (PA-ZR). Forty-five days after treatment, animals were subjected to electroretinography. In addition, morphological techniques (TUNEL, H&E, multiple immunofluorescence) were applied to the retinas. Results: A reduction in the amplitude of the a- and b-wave and oscillatory potentials (OP) of the electroretinogram (ERG) was detected in PA animals. Treatment with zr17-2 resulted in a significant amelioration of these parameters (p < 0.01). In PA animals, a large number of apoptotic cells was found in the GCL. This number was significantly reduced by treatment with the small molecule (p < 0.0001). In a similar way, the thickness of the inner retina and the intensity of GFAP immunoreactivity (gliosis) increased in PA retinas (p < 0.0001). These parameters were corrected by the administration of zr17-2 (p < 0.0001). Furthermore, injection of the small molecule in the absence of PA did not modify the ERG nor the morphological parameters studied, suggesting a lack of toxicity. Discussion: In conclusion, our results indicate that a single s.c. injection of zr17-2 in asphyctic neonates may provide a novel and efficacious method to prevent the visual sequelae of PA.

4.
Front Pharmacol ; 13: 840134, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35387355

RESUMO

Continuous illumination induces the degeneration of photoreceptors. This animal model of light-induced retinal degeneration resembles many characteristics of human degenerative diseases of the outer retina, such as age-related macular degeneration. This work aimed to evaluate the potential neuroprotective effect of the modulation of adenosine A2A receptor in the model of light-induced retinal degeneration. Sprague-Dawley rats were intravitreally injected in the right eye with either CGS 21680, an adenosine A2A receptor agonist, or SCH 58261, an adenosine A2A receptor antagonist. Contralateral eyes were injected with respective vehicles as control. Then, rats were subjected to continuous illumination (12,000 lux) for 24 h. Retinas were processed by glial fibrillary acidic protein (GFAP) immunohistochemistry, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) technique, Western blotting (WB), and quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Another group of rats was subjected to functional studies by electroretinography. Animals treated with CGS21680 showed a significant increase of apoptotic nuclei in the outer nuclear layer and a significant increase of GFAP immunoreactive area of the retinas but did not alter WB nor electroretinography results. qRT-PCR showed that CGS 21680 significantly increased the expression of interleukin-1ß. On the opposite, SCH 58261 significantly decreased apoptotic nuclei in the outer nuclear layer and GFAP immunoreactive area of the retinas. It also significantly decreased GFAP and activated caspase-3 levels as measured by WB and preserved retinal function, as treated eyes showed significantly greater amplitudes of a- and b-waves and oscillatory potentials. qRT-PCR revealed that SCH 58261 significantly decreased the expression of tumor necrosis factor-α. These results show that the blockade of the A2A receptor before the start of the pathogenic process is neuroprotective, as it prevents light-induced retinal damage. The use of A2A receptor antagonists deserves to be evaluated in retinal degenerative diseases.

5.
Adv Drug Alcohol Res ; 2: 10734, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-38390616

RESUMO

In the last few years, an increasing interest in the neuroprotective effect of cannabinoids has taken place. The aim of the present work was to study the effects of modulating cannabinoid receptor 1 (CB1) in the context of light induced retinal degeneration (LIRD), using an animal model that resembles many characteristics of human age-related macular degeneration (AMD) and other degenerative diseases of the outer retina. Sprague Dawley rats (n = 28) were intravitreally injected in the right eye with either a CB1 agonist (ACEA), or an antagonist (AM251). Contralateral eyes were injected with respective vehicles as controls. Then, rats were subjected to continuous illumination (12,000 lux) for 24 h. Retinas from 28 animals were processed by GFAP-immunohistochemistry (IHC), TUNEL technique, Western blotting (WB), or qRT-PCR. ACEA-treated retinas showed a significantly lower number of apoptotic nuclei in the outer nuclear layer (ONL), lower levels of activated Caspase-3 by WB, and lower levels of glial reactivity by both GFAP-IHC and WB. qRT-PCR revealed that ACEA significantly decreased the expression of Bcl-2 and CYP1A1. Conversely, AM251-treated retinas showed a higher number of apoptotic nuclei in the ONL, higher levels of activated Caspase-3 by WB, and higher levels of glial reactivity as determined by GFAP-IHC and WB. AM251 increased the expression of Bcl-2, Bad, Bax, Aryl hydrocarbon Receptor (AhR), GFAP, and TNFα. In summary, the stimulation of the CB1 receptor, previous to the start of the pathogenic process, improved the survival of photoreceptors exposed to LIRD. The modulation of CB1 activity may be used as a neuroprotective strategy in retinal degeneration and deserves further studies.

6.
Cell Rep ; 36(5): 109463, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34348156

RESUMO

Specificity and timing of synapse disassembly in the CNS are essential to learning how individual circuits react to neurodegeneration of the postsynaptic neuron. In sensory systems such as the mammalian retina, synaptic connections of second-order neurons are known to remodel and reconnect in the face of sensory cell loss. Here we analyzed whether degenerating third-order neurons can remodel their local presynaptic connectivity. We injured adult retinal ganglion cells by transiently elevating intraocular pressure. We show that loss of presynaptic structures occurs before postsynaptic density proteins and accounts for impaired transmission from presynaptic neurons, despite no evidence of presynaptic cell loss, axon terminal shrinkage, or reduced functional input. Loss of synapses is biased among converging presynaptic neuron types, with preferential loss of the major excitatory cone-driven partner and increased connectivity with rod-driven presynaptic partners, demonstrating that this adult neural circuit is capable of structural plasticity while undergoing neurodegeneration.


Assuntos
Rede Nervosa/patologia , Ferimentos e Lesões/patologia , Animais , Feminino , Pressão Intraocular , Luz , Masculino , Camundongos , Terminações Pré-Sinápticas/patologia , Células Bipolares da Retina/patologia , Células Ganglionares da Retina/patologia , Sinapses/patologia
7.
Front Pharmacol ; 12: 651599, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33897437

RESUMO

Perinatal asphyxia (PA) can cause retinopathy and different degrees of visual loss, including total blindness. In a rat model of PA, we have previously shown a protective effect of hypothermia on the retina when applied simultaneously with the hypoxic insult. In the present work, we evaluated the possible protective effect of hypothermia on the retina of PA rats when applied immediately after delivery. Four experimental groups were studied: Rats born naturally as controls (CTL), animals that were exposed to PA for 20 min at 37°C (PA), animals exposed to PA for 20 min at 15°C (HYP), and animals that were exposed to PA for 20 min at 37°C and, immediately after birth, kept for 15 min at 8°C (HYP-PA). To evaluate the integrity of the visual pathway, animals were subjected to electroretinography at 45 days of age. Molecular (real time PCR) and histological (immunohistochemistry, immunofluorescence, TUNEL assay) techniques were applied to the eyes of all experimental groups collected at 6, 12, 24, and 48 h, and 6 days after birth. PA resulted in a significant reduction in the amplitude of the a- and b-wave and oscillatory potentials (OP) of the electroretinogram. All animals treated with hypothermia had a significant correction of the a-wave and OP, but the b-wave was fully corrected in the HYP group but only partially in the HYP-PA group. The number of TUNEL-positive cells increased sharply in the ganglion cell layer of the PA animals and this increase was significantly prevented by both hypothermia treatments. Expression of the cold-shock proteins, cold-inducible RNA binding protein (CIRP) and RNA binding motif protein 3 (RBM3), was undetectable in retinas of the CTL and PA groups, but they were highly expressed in ganglion neurons and cells of the inner nuclear layer of the HYP and HYP-PA groups. In conclusion, our results suggest that a post-partum hypothermic shock could represent a useful and affordable method to prevent asphyxia-related vision disabling sequelae.

8.
J Comp Neurol ; 528(16): 2679-2694, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32301107

RESUMO

GABAergic medium spiny neurons are the main neuronal population in the striatum. Calbindin is preferentially expressed in medium spiny neurons involved in the indirect pathway. The aim of the present work is to analyze the effect of perinatal asphyxia on different subpopulations of GABAergic neurons in the striatum and to assess the outcome of deep therapeutic hypothermia. The uterus of pregnant rats was removed by cesarean section and the fetuses were exposed to hypoxia by immersion in water (19 min) at 37°C (perinatal asphyxia). The hypothermic group was exposed to 10°C during 30 min after perinatal asphyxia. The rats were euthanized at the age of one month (adolescent/adult rats), their brains were dissected out and coronal sections were immunolabeled for calbindin, calretinin, NeuN, and reelin. Reelin+ cells showed no staining in the striatum besides subventricular zone. The perinatal asphyxia (PA) group showed a significant decrease in calbindin neurons and a paradoxical increase in neurons estimated by NeuN staining. Moreover, calretinin+ cells, a specific subpopulation of GABAergic neurons, showed an increase caused by PA. Deep hypothermia reversed most of these alterations probably by protecting calbindin neurons. Similarly, there was a reduction of the diameter of the anterior commissure produced by the asphyxia that was prevented by hypothermic treatment.


Assuntos
Asfixia Neonatal/terapia , Corpo Estriado/patologia , Discinesias/prevenção & controle , Hipotermia Induzida/métodos , Transtornos Psicóticos/prevenção & controle , Animais , Animais Recém-Nascidos , Comissura Anterior/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Calbindina 2/metabolismo , Calbindinas/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Corpo Estriado/metabolismo , Discinesias/etiologia , Proteínas da Matriz Extracelular/metabolismo , Feminino , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/patologia , Masculino , Proteínas do Tecido Nervoso/metabolismo , Gravidez , Transtornos Psicóticos/etiologia , Ratos , Ratos Sprague-Dawley , Proteína Reelina , Serina Endopeptidases/metabolismo
9.
Front Cell Neurosci ; 14: 157, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32581722

RESUMO

Perinatal asphyxia (PA) is responsible for a large proportion of neonatal deaths and numerous neurological sequelae, including visual dysfunction and blindness. In PA, the retina is exposed to ischemia/reoxygenation, which results in nitric oxide (NO) overproduction and neurotoxicity. We hypothesized that methylene blue (MB), a guanylyl cyclase inhibitor, and free-radical scavenger currently used in the clinic, may block this pathway and prevent PA-induced retinal degeneration. Male rat pups were subjected to an experimental model of PA. Four groups were studied: normally delivered (CTL), normally delivered treated with 2 mg Kg-1 MB (MB), exposed to PA for 20 min at 37°C (PA), and exposed to PA and, then, treated with MB (PA-MB). Scotopic electroretinography performed 45 days after birth showed that PA animals had significant defects in the a- and b-waves and oscillatory potentials (OP). The same animals presented a significant increase in the thickness of the inner retina and a large number of TUNEL-positive cells. All these physiological and morphological parameters were significantly prevented by the treatment with MB. Gene expression analysis demonstrated significant increases in iNOS, MMP9, and VEGF in the eyes of PA animals, which were prevented by MB treatment. In conclusion, MB regulates key players of inflammation, matrix remodeling, gliosis, and angiogenesis in the eye and could be used as a treatment to prevent the deleterious visual consequences of PA. Given its safety profile and low cost, MB may be used clinically in places where alternative treatments may be unavailable.

10.
Neurosci Lett ; 687: 259-267, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30291879

RESUMO

The modulation of adenosine receptors, A1 (A1R) and A2A (A2AR), is neuroprotective in different models of retinal injury. In order to understand the processes underlying retinal degeneration, we studied the expression of adenosine receptors in the retinas of control and continuously illuminated (CI) rats by qRT-PCR, Western blot (WB) and immunohistochemistry (IHC). Significant increases of A1R, A2AR, and A2BR mRNAs at 1, 5, and 7 days of CI (P < 0.0001) were observed by qRT-PCR. Also, a significant increase of A3R mRNA was detected after 5 and 7 days of CI. WB studies showed a significant rise of A1R on day 1 of CI and on days 5 and 7 (P < 0.0001), while A2AR increase was seen from 2 days of CI on (P < 0.001). After 1 day of CI, A1R immunoreactivity (A1R-IR) increased in ganglion cell layer, inner nuclear layer, and in both the outer and inner plexiform layers. After 2 days of CI, the A1R-IR went back to control levels. After 5 days of CI, a second rise in A1R, which persisted until 7 days of CI, was measured (P < 0.0001). A significant rise of A2aR immunoreactivity was also observed at day 2 of CI at GCL and INL and subsided at days 5 and 7 (P < 0.0001). The observed up-regulation of A1R after 1 day of CI, corresponds with the peak of oxidative stress; while the rise of A2aR at day 2 of CI, coincides with the massive apoptosis of photoreceptors. We postulate that an early modulation of adenosine receptors could delay or prevent the degeneration of photoreceptors.


Assuntos
Adenosina/metabolismo , Luz/efeitos adversos , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Degeneração Retiniana/metabolismo , Animais , Iluminação/métodos , Masculino , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Degeneração Retiniana/patologia
11.
PLoS One ; 13(6): e0198838, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29912966

RESUMO

Light induced retinal degeneration (LIRD) is a useful model that resembles human retinal degenerative diseases. The modulation of adenosine A1 receptor is neuroprotective in different models of retinal injury. The aim of this work was to evaluate the potential neuroprotective effect of the modulation of A1 receptor in LIRD. The eyes of rats intravitreally injected with N6-cyclopentyladenosine (CPA), an A1 agonist, which were later subjected to continuous illumination (CI) for 24 h, showed retinas with a lower number of apoptotic nuclei and a decrease of Glial Fibrillary Acidic Protein (GFAP) immunoreactive area than controls. Lower levels of activated Caspase 3 and GFAP were demonstrated by Western Blot (WB) in treated animals. Also a decrease of iNOS, TNFα and GFAP mRNA was demonstrated by RT-PCR. A decrease of Iba 1+/MHC-II+ reactive microglial cells was shown by immunohistochemistry. Electroretinograms (ERG) showed higher amplitudes of a-wave, b-wave and oscillatory potentials after CI compared to controls. Conversely, the eyes of rats intravitreally injected with dipropylcyclopentylxanthine (DPCPX), an A1 antagonist, and subjected to CI for 24 h, showed retinas with a higher number of apoptotic nuclei and an increase of GFAP immunoreactive area compared to controls. Also, higher levels of activated Caspase 3 and GFAP were demonstrated by Western Blot. The mRNA levels of iNOS, nNOS and inflammatory cytokines (IL-1ß and TNFα) were not modified by DPCPX treatment. An increase of Iba 1+/MHC-II+ reactive microglial cells was shown by immunohistochemistry. ERG showed that the amplitudes of a-wave, b-wave, and oscillatory potentials after CI were similar to control values. A single pharmacological intervention prior illumination stress was able to swing retinal fate in opposite directions: CPA was neuroprotective, while DPCPX worsened retinal damage. In summary, A1 receptor agonism is a plausible neuroprotective strategy in LIRD.


Assuntos
Agonistas do Receptor A1 de Adenosina/uso terapêutico , Adenosina/análogos & derivados , Receptor A1 de Adenosina/efeitos dos fármacos , Degeneração Retiniana/tratamento farmacológico , Adenosina/administração & dosagem , Adenosina/uso terapêutico , Agonistas do Receptor A1 de Adenosina/administração & dosagem , Animais , Western Blotting , Caspase 3/metabolismo , Modelos Animais de Doenças , Eletrorretinografia , Proteína Glial Fibrilar Ácida/metabolismo , Injeções Intravítreas , Masculino , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Receptor A1 de Adenosina/fisiologia , Retina/efeitos dos fármacos , Retina/efeitos da radiação , Fator de Necrose Tumoral alfa/metabolismo
12.
Sci Rep ; 7(1): 6966, 2017 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-28761115

RESUMO

Ocular and periocular traumatisms may result in loss of vision. Hypothermia provides a beneficial intervention for brain and heart conditions and, here, we study whether hypothermia can prevent retinal damage caused by traumatic neuropathy. Intraorbital optic nerve crush (IONC) or sham manipulation was applied to male rats. Some animals were subjected to hypothermia (8 °C) for 3 h following surgery. Thirty days later, animals were subjected to electroretinography and behavioral tests. IONC treatment resulted in amplitude reduction of the b-wave and oscillatory potentials of the electroretinogram, whereas the hypothermic treatment significantly (p < 0.05) reversed this process. Using a descending method of limits in a two-choice visual task apparatus, we demonstrated that hypothermia significantly (p < 0.001) preserved visual acuity. Furthermore, IONC-treated rats had a lower (p < 0.0001) number of retinal ganglion cells and a higher (p < 0.0001) number of TUNEL-positive cells than sham-operated controls. These numbers were significantly (p < 0.0001) corrected by hypothermic treatment. There was a significant (p < 0.001) increase of RNA-binding motif protein 3 (RBM3) and of BCL2 (p < 0.01) mRNA expression in the eyes exposed to hypothermia. In conclusion, hypothermia constitutes an efficacious treatment for traumatic vision-impairing conditions, and the cold-shock protein pathway may be involved in mediating the beneficial effects shown in the retina.


Assuntos
Hipotermia Induzida/métodos , Traumatismos do Nervo Óptico/terapia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas de Ligação a RNA/genética , Doenças Retinianas/prevenção & controle , Animais , Modelos Animais de Doenças , Eletrorretinografia , Perfilação da Expressão Gênica , Masculino , Traumatismos do Nervo Óptico/complicações , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/patologia , Ratos , Doenças Retinianas/etiologia , Doenças Retinianas/genética , Doenças Retinianas/patologia , Regulação para Cima , Acuidade Visual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA