Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 199
Filtrar
1.
Exp Eye Res ; 231: 109485, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37080381

RESUMO

The vesicant sulfur mustard (SM) is a chemical warfare agent that causes acute and chronic injury to the cornea and proximal anterior segment structures. Despite clinical evidence of SM-exposure causing unexplained retinal deficits, there have been no animal studies conducted to examine the retinal toxicity of this vesciant. The cardinal hallmark of retinal response to stressors or injury is the activation of reactive gliosis, a cellular process largely governed by Müller glia. Previously we showed that corneal exposure to sodium hydroxide elicits rapid induction of reactive gliosis and results in retinal degeneration in a dose-related manner. Based on this evidence, we hypothesized that the vesicant nitrogen mustard (NM), an analog of SM, may also elicit reactive gliosis. To test this idea, we developed a mouse model of NM ocular injury and investigated corneal and retinal effects focusing on citrullination, a posttranslational modification (PTM) of proteins. This PTM was recently linked to alkali injury and has also been shown to occur in retinal degenerative conditions. Here, we demonstrate that corneal exposure to 1% NM causes a synchronous activation of citrullination in both the cornea and retina with hypercitrullination becoming apparent temporally and manifesting with altered cellular expression characteristics. A key finding is that ocular citrullination occurs acutely as early as 1-h post-injury in both the cornea and retina, which underscores a need for expeditious interception of this acute corneal and retinal response. Moreover, exploiting dose response and temporal studies, we uncoupled NM-induced retinal citrullination from its induction of retinal gliosis. Our findings demonstrate that hypercitrullination is a common corneo-retinal mechanism that sensitizes the eye to NM injury and suggests that counteracting hypercitrullination may provide a suitable countermeasure to vesicant injury.


Assuntos
Traumatismos Oculares , Gás de Mostarda , Doenças Retinianas , Animais , Camundongos , Mecloretamina/toxicidade , Irritantes/efeitos adversos , Irritantes/metabolismo , Gliose/induzido quimicamente , Gliose/metabolismo , Córnea/metabolismo , Traumatismos Oculares/induzido quimicamente , Traumatismos Oculares/metabolismo , Retina , Gás de Mostarda/toxicidade , Doenças Retinianas/induzido quimicamente , Doenças Retinianas/metabolismo
2.
Int J Mol Sci ; 23(22)2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-36430547

RESUMO

Neurotrophins are a family of closely related secreted proteins that promote differentiation, development, and survival of neurons, which include nerve growth factor (NGF), brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4. All neurotrophins signal through tropomyosin receptor kinases (TrkA, TrkB, and TrkC) which are more selective to NGF, brain-derived neurotrophic factor, and neurotrophin-3, respectively. NGF is the most studied neurotrophin in the ocular surface and a human recombinant NGF has reached clinics, having been approved to treat neurotrophic keratitis. Brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4 are less studied neurotrophins in the ocular surface, even though brain-derived neurotrophic factor is well characterized in glaucoma, retina, and neuroscience. Recently, neurotrophin analogs with panTrk activity and TrkC selectivity have shown promise as novel drugs for treating dry eye disease. In this review, we discuss the biology of the neurotrophin family, its role in corneal homeostasis, and its use in treating ocular surface diseases. There is an unmet need to investigate parenteral neurotrophins and its analogs that activate TrkB and TrkC selectively.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Traumatismos Oculares , Fator de Crescimento Neural , Receptores Proteína Tirosina Quinases , Humanos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Olho/metabolismo , Olho/patologia , Ligantes , Fator de Crescimento Neural/metabolismo , Fator de Crescimento Neural/farmacologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Receptor trkC/metabolismo , Traumatismos Oculares/tratamento farmacológico , Traumatismos Oculares/genética , Traumatismos Oculares/metabolismo
3.
Cells ; 11(21)2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36359852

RESUMO

Cataracts are treated by lens fiber cell removal followed by intraocular lens (IOL) implantation into the lens capsule. While effective, this procedure leaves behind numerous lens epithelial cells (LECs) which undergo a wound healing response that frequently leads to posterior capsular opacification (PCO). In order to elucidate the acute response of LECs to lens fiber cell removal which models cataract surgery (post cataract surgery, PCS), RNA-seq was conducted on LECs derived from wild type mice at 0 and 6 h PCS. This analysis found that LECs upregulate the expression of numerous proinflammatory cytokines and profibrotic regulators by 6 h PCS suggesting rapid priming of pathways leading to inflammation and fibrosis PCS. LECs also highly upregulate the expression of numerous immediate early transcription factors (IETFs) by 6 h PCS and immunolocalization found elevated levels of these proteins by 3 h PCS, and this was preceded by the phosphorylation of ERK1/2 in injured LECs. Egr1 and FosB were among the highest expressed of these factors and qRT-PCR revealed that they also upregulate in explanted mouse lens epithelia suggesting potential roles in the LEC injury response. Analysis of lenses lacking either Egr1 or FosB revealed that both genes may regulate a portion of the acute LEC injury response, although neither gene was essential for expression of either proinflammatory or fibrotic markers at later times PCS suggesting that IETFs may work in concert to mediate the LEC injury response following cataract surgery.


Assuntos
Opacificação da Cápsula , Extração de Catarata , Traumatismos Oculares , Cápsula do Cristalino , Cristalino , Camundongos , Animais , Cápsula do Cristalino/metabolismo , Cápsula do Cristalino/patologia , Cristalino/metabolismo , Células Epiteliais/metabolismo , Opacificação da Cápsula/metabolismo , Traumatismos Oculares/metabolismo , Fatores de Transcrição/metabolismo , Fibrose
4.
Exp Eye Res ; 223: 109209, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35961426

RESUMO

Sulfur mustard (SM) is a notorious, bifunctional alkylating vesicant that was first used in warfare during World War I in 1917 and since then has been deployed in numerous skirmishes with its most recent documented use being during the Middle Eastern conflicts. Apart from its use in combat and terrorist activities, continual threat of accidental exposure from old stockpiles and improperly discarded munitions is ever present, especially to the innocent and unassuming civilian populations. SM can cause devastating injuries, depending on the dosage of SM exposure, route of exposure, as well as the physiological conditions of the individuals exposed. The most common routes of exposure are ocular, dermal, and exposure to the lungs and respiratory tissues through inhalation. Eyes are the most susceptible organ to SM-induced toxicities owing to their high moisture content and rapidly dividing cells. Additionally, ocular injury causes the most expeditious disablement of individuals even upon whole-body exposures. Therefore, it is imperative to understand the mechanisms underlying SM-induced ocular toxicity and design therapeutic interventions to prevent/mitigate ocular injuries. Ocular SM exposure may cause a wide range of symptoms such as inflammation, lacrimation, itching, dryness, photophobia, edema of the cornea/sclera/retina/iris, conjunctivitis, degradation of the corneal layer, fusion of two or more ocular layers, neovascularization, fibrosis, and temporary or permanent structural damage to one or more ocular layers. These symptoms may lead to vision impairments, resulting in partial or complete blindness that may be permanent. The highly toxic and exceedingly notorious nature of SM makes it a highly regulated chemical, requiring very expensive licensing, security, and safety requirements; thus, the more easily accessible analogue, nitrogen mustard (NM) that mimics SM-induced toxicity and injuries is employed in plethora of studies conducted in different animal models and culture systems. This review provides a comprehensive account of the injuries and symptoms that occur upon ocular SM exposures in human patients as well as studies in animal (in vivo, ex vivo) and cell (in vitro) models of SM and NM ocular exposures. Special emphasis has been laid on highlighting the strengths and lacunae in the research as well as the possible unexplored avenues of mechanisms underlying mustard-induced ocular injury that can be explored in future research endeavors. Furthermore, development of therapeutic interventions and targets of interest in the ocular system exposed to SM and NM, based on studies in human patients as well as in vivo, ex vivo, and in vitro models has been discussed in great depth, providing a valuable knowledge database to delineate pathways associated with vesicant-induced toxicity, and strategies/diagnostic tools against SM-induced toxicity.


Assuntos
Substâncias para a Guerra Química , Traumatismos Oculares , Gás de Mostarda , Animais , Substâncias para a Guerra Química/toxicidade , Córnea/metabolismo , Traumatismos Oculares/induzido quimicamente , Traumatismos Oculares/metabolismo , Humanos , Irritantes/efeitos adversos , Irritantes/metabolismo , Mecloretamina/toxicidade , Gás de Mostarda/toxicidade
5.
Exp Eye Res ; 220: 109104, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35577069

RESUMO

Glutamate-induced excitotoxic injury is widely described as a prominent pathophysiological mechanism in several neurodegenerative diseases including glaucoma. Glaucoma, the leading cause of irreversible blindness, is characterized by loss of retinal ganglion cells (RGC). Currently, the treatment focuses on lowering intraocular pressure (IOP) and no neuroprotective therapies are available. Since excessive glutamate-mediated neurotransmission underlies glaucomatous RGC apoptosis, enhancing synaptic glutamate clearance by glutamate transporters in glial cells is expected to protect against excitotoxic injury. Trans-resveratrol is known for its neuroprotective effects; however, its effects on the expression of glutamate transporters and glutamate clearance in retina remain unclear. Hence, in the current study, we investigated the protective effects of trans-resveratrol against glutamate-induced retinal injury in rats. Rats were intravitreally injected with glutamate alone or glutamate with trans-resveratrol as pre- and post-treatment. Animals were subjected to Open Field Test (OFT) on day six and a two-chamber mirror test on day seven post-injection. Subsequently, rats were sacrificed and retinal expression of excitatory amino acid transporter (EAAT)1 and EAAT2 gene and protein was determined using PCR and ELISA, respectively. Retinal glutamate concentration was measured using ELISA and retinal morphology was studied on H&E-stained retinal sections. It was observed that pre-treatment with trans-resveratrol causes gene expression for EAAT1 and EAAT2 to increase by 2.51 and 1.93 folds compared to glutamate-treated group (p < 0.001 and p < 0.01, respectively); while the same in trans-resveratrol post-treatment group showed a 1.58- and 1.44 folds upregulation (p < 0.05).The retinal EAAT1 and EAAT2 protein expression was significantly greater in trans-resveratrol pre-treatment group compared to glutamate-treated group (p < 0.05) but not in post-treatment group. Retinal glutamate concentration was1.64 folds greater in glutamate-treated group than the vehicle-treated group (p < 0.01) but the same was 1.27-fold lower in trans-resveratrol pre-treatment group compared to glutamate-treated group (p < 0.01). Corresponding to these findings, we observed preservation of retinal morphology and visual behaviour in trans-resveratrol pre-treatment group compared to glutamate-treated group. We did not observe similar effects of trans-resveratrol when it was given as post-treatment after glutamate administration. In conclusion, current study showed that pre-treatment with trans-resveratrol protects against glutamate induced changes in retinal morphology and visual behaviour by increasing the expression of EAAT1 and EAAT2 and increasing glutamate clearance in rat retinas. The results of this study may be relevant to disease conditions involving excitotoxic neuronal injury.


Assuntos
Traumatismos Oculares , Glaucoma , Doenças Retinianas , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Traumatismos Oculares/metabolismo , Glaucoma/tratamento farmacológico , Glaucoma/metabolismo , Ácido Glutâmico/metabolismo , Ratos , Resveratrol/farmacologia , Doenças Retinianas/metabolismo , Células Ganglionares da Retina/metabolismo
6.
Invest Ophthalmol Vis Sci ; 63(2): 30, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35201262

RESUMO

Purpose: The purpose of this study was to explore the therapeutic role of heat shock protein 90 (Hsp90) in wound healing of injury cornea epithelium. Methods: The right eye of C57BL/6N male mice were performed the debridement wounds in the center of the cornea using an algerbrush II blade. The injured area was determined by staining the cornea with fluorescein sodium and measured with image-J. Immunoblotting, ELISA and immunochemistry were used for determining protein expression. The quantitation PCR was performed to measure mRNA expression. Results: Hsp90α is upregulated at both the mRNA and protein levels, and is secreted extracellularly into the corneal stroma and tear film during the healing process after corneal injury in mice. This upregulation is associated with activation of HSF1. Administration of recombinant exogenous Hsp90α (eHsp90α) speeds up wound healing of injured corneal epithelium. The eHsp90α binds to low-density lipoprotein (LDL)-related protein-1 (LRP-1) on the corneal epithelial cells and increases phosphorylation of AKT at S473, which is associated with proliferation and migration corneal epithelial cells in vitro or vivo. Inhibition of AKT by its inhibitor LY294002 abolishes eHsp90α-induced migration and proliferation of corneal epithelial cells. Conclusion: Hsp90α is upregulated and secreted after corneal injury and acts to promote the healing process. Recombinant Hsp90α may be a promising therapeutic drug candidate for corneal injury.


Assuntos
Epitélio Corneano/lesões , Traumatismos Oculares/tratamento farmacológico , Proteínas de Choque Térmico HSP90/uso terapêutico , Cicatrização/efeitos dos fármacos , Animais , Western Blotting , Linhagem Celular , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Desbridamento , Ensaio de Imunoadsorção Enzimática , Epitélio Corneano/efeitos dos fármacos , Epitélio Corneano/metabolismo , Traumatismos Oculares/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas de Choque Térmico HSP90/genética , Fatores de Transcrição de Choque Térmico/metabolismo , Humanos , Imuno-Histoquímica , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapêutico
7.
Exp Eye Res ; 218: 108966, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35143834

RESUMO

Visual deficits after ocular blast injury (OBI) are common, but pharmacological approaches to improve long-term outcomes have not been identified. Blast forces frequently damage the retina and optic nerves, and work on experimental animals has shown the pro-inflammatory actions of microglia can further exacerbate such injuries. Cannabinoid type-2 receptor (CB2) inverse agonists specifically target activated microglia, biasing them away from the harmful pro-inflammatory M1 state toward the helpful reparative M2 state. We previously found that treating mice with CB2 inverse agonists after traumatic brain injury, produced by either focal cranial air blast or dorsal cranial impact, greatly attenuated the visual deficits and pathology that otherwise resulted. Here we examined the consequences of single and repeat OBI and the benefit provided by raloxifene, an FDA-approved estrogen receptor drug that possesses noteworthy CB2 inverse agonism. After single OBI, although the amplitudes of the A- and B-waves of the electroretinogram and pupil light response appeared to be normal, the mice showed hints of deficits in contrast sensitivity and visual acuity, a trend toward optic nerve axon loss, and significantly increased light aversion, which were reversed by 2 weeks of daily treatment with raloxifene. Mice subjected to repeat OBI (5 blasts spaced 1 min apart), exhibited more severe visual deficits, including decreases in contrast sensitivity, visual acuity, the amplitudes of the A- and B-waves of the electroretinogram, light aversion, and resting pupil diameter (i.e. hyperconstriction), accompanied by the loss of photoreceptor cells and optic nerve axons, nearly all of which were mitigated by raloxifene. Interestingly, optic nerve axon abundance was strongly correlated with contrast sensitivity and visual acuity across all groups of experimental mice in the repeat OBI study, suggesting optic nerve axon loss with repeat OBI and its attenuation with raloxifene are associated with the extent of these two deficits while photoreceptor abundance was highly correlated with A-wave amplitude and resting pupil size, suggesting a prominent role for photoreceptors in these two deficits. Quantitative PCR (qPCR) showed levels of M1-type microglial markers (e.g. iNOS, IL1ß, TNFα, and CD32) in retina, optic nerve, and thalamus were increased 3 days after repeat OBI. With raloxifene treatment, the overall expression of M1 markers was more similar to that in sham mice. Raloxifene treatment was also associated with the elevation of IL10 transcripts in all three tissues compared to repeat OBI alone, but the results for the three other M2 microglial markers we examined were more varied. Taken together, the qPCR results suggest that raloxifene benefit for visual function and pathology was associated with a lessening of the pro-inflammatory actions of microglia. The benefit we find for raloxifene following OBI provides a strong basis for phase-2 efficacy testing in human clinical trials for treating ocular injury.


Assuntos
Traumatismos por Explosões , Canabinoides , Traumatismos Oculares , Animais , Traumatismos por Explosões/metabolismo , Agonistas de Receptores de Canabinoides , Traumatismos Oculares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Cloridrato de Raloxifeno/metabolismo , Cloridrato de Raloxifeno/farmacologia , Cloridrato de Raloxifeno/uso terapêutico
8.
Exp Eye Res ; 217: 108979, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35143835

RESUMO

Excitotoxicity-induced retinal neuronal death is characterized by the progressive retinal ganglion cell (RGC) apoptosis. Strategies are needed to reduce neurodegeneration. Recent investigations have indicated the potential effects of metformin on multiple systems, especially in the networks. However, it also remains unclear whether mitophagy contributes to the neuroprotective effect of metformin on the retina. In this study, excitotoxicity-induced retinal injury models were constructed. In vitro, R28 cells were treated with calcium ionophore and metformin/phosphate-buffer saline (PBS). Cell viability, lactate dehydrogenase release, and the cellular apoptosis rate were assessed. In vivo, rats received intravitreal injection of N-methyl-D-aspartate and metformin/PBS. Comprehensive examinations including retrograde fluorescent gold labelling, Nissl's staining, full-field electroretinography, photopic negative response, optic coherence tomography and retinal imaging, transmission electron microscopy, western blotting, and quantitative polymerase chain reaction were conducted during the observation period. The viability of R28 cells was significantly increased in the metformin-treated group compared with the negative control group, while, the release of lactate dehydrogenase and R28 cell apoptosis showed a significant decrease. In vivo, metformin treatment significantly increased the number of surviving RGCs, the b/NR wave amplitude and the thickness of the inner retina but had no obvious adverse effects on the fundus. In the metformin-treated group, the morphology and number of mitochondria were better preserved, as observed for RGCs; mitochondrial autophagosomes were located in RGCs, as indicated by transmission electron microscopy; and the expression of mitophagy-related genes and proteins presented was significant regulated. These data indicated that the regulation of mitophagy by metformin improved the structure and function of RGCs.


Assuntos
Traumatismos Oculares , Metformina , Doenças Retinianas , Animais , Apoptose , Traumatismos Oculares/metabolismo , Lactato Desidrogenases/metabolismo , Metformina/metabolismo , Metformina/farmacologia , Mitofagia , N-Metilaspartato/farmacologia , Ratos , Doenças Retinianas/metabolismo , Células Ganglionares da Retina/metabolismo
9.
Exp Eye Res ; 215: 108930, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35016886

RESUMO

Visual deficits are a common concern among subjects with head trauma. Stem cell therapies have gained recent attention in treating visual deficits following head trauma. Previously, we have shown that adipose-derived stem cell (ASC) concentrated conditioned medium (ASC-CCM), when delivered via an intravitreal route, yielded a significant improvement in vision accompanied by a decrease in retinal neuroinflammation in a focal cranial blast model that indirectly injures the retina. The purpose of the current study is to extend our previous studies to a direct ocular blast injury model to further establish the preclinical efficacy of ASC-CCM. Adult C57BL/6J mice were subjected to repetitive ocular blast injury (rOBI) of 25 psi to the left eye, followed by intravitreal delivery of ASC-CCM (∼200 ng protein/2 µl) or saline within 2-3 h. Visual function and histological changes were measured 4 weeks after injury and treatment. In vitro, Müller cells were used to evaluate the antioxidant effect of ASC-CCM. Visual acuity, contrast sensitivity, and b-wave amplitudes in rOBI mice receiving saline were significantly decreased compared with age-matched sham blast mice. Immunohistological analyses demonstrated a significant increase in glial fibrillary acidic protein (a retinal injury marker) in Müller cell processes, DNA/RNA damage, and nitrotyrosine (indicative of oxidative stress) in the retina, while qPCR analysis revealed a >2-fold increase in pro-inflammatory cytokines (TNF-α, ICAM1, and Ccl2) in the retina, as well as markers for microglia/macrophage activation (IL-1ß and CD86). Remarkably, rOBI mice that received ASC-CCM demonstrated a significant improvement in visual function compared to saline-treated rOBI mice, with visual acuity, contrast sensitivity, and b-wave amplitudes that were not different from those in sham mice. This improvement in visual function also was associated with a significant reduction in retinal GFAP, neuroinflammation markers, and oxidative stress compared to saline-treated rOBI mice. In vitro, Müller cells exposed to oxidative stress via increasing doses of hydrogen peroxide demonstrated decreased viability, increased GFAP mRNA expression, and reduced activity for the antioxidant catalase. On the other hand, oxidatively stressed Müller cells pre-incubated with ASC-CCM showed normalized GFAP, viability, and catalase activity. In conclusion, our study demonstrates that a single intravitreal injection of ASC-CCM in the rOBI can significantly rescue retinal injury and provide significant restoration of visual function. Our in vitro studies suggest that the antioxidant catalase may play a major role in the protective effects of ASC-CCM, uncovering yet another aspect of the multifaceted benefits of ASC secretome therapies in neurotrauma.


Assuntos
Traumatismos por Explosões , Traumatismos Oculares , Células-Tronco Mesenquimais , Animais , Antioxidantes/farmacologia , Traumatismos por Explosões/metabolismo , Catalase/metabolismo , Traumatismos Oculares/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Retina/metabolismo , Secretoma
10.
Invest Ophthalmol Vis Sci ; 62(3): 32, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33755044

RESUMO

Purpose: New lasers with a continuous wave power exceeding 15 W are currently investigated for retinal therapies, promising highly localized effects at and close to the Retinal Pigment Epithelium (RPE). The goal of this work is to evaluate mechanisms and thresholds for RPE cell damage by means of pulse durations up to 50 µs. Methods: A diode laser with a wavelength of 514 nm, a power of 15 W, and adjustable pulse durations between 2 µs and 50 µs was used. Porcine RPE-choroidal explants (ex vivo) and chinchilla bastard rabbits (in vivo) were irradiated to determine threshold radiant exposures for RPE damage \({\bar H_{Cell}}\) by calcein vitality staining and fluorescence angiography, respectively. Thresholds for microbubble formation (MBF) \({\bar H_{MBF}}\) were evaluated by time-resolved optoacoustics. Exemplary histologies support the findings. Results: \({\bar H_{{{MBF}}}}\) is significantly higher than \({\bar H_{Cell}}\) at pulse durations ≥ 5 µs (P < 0.05) ex vivo, while at 2 µs, no statistically significant difference was found. The ratios between \({\bar H_{{{MBF}}}}\) and \({\bar H_{Cell}}\) increase with pulse duration from 1.07 to 1.48 ex vivo and 1.1 to 1.6 in vivo, for 5.2 and 50 µs. Conclusions: Cellular damage with and without MBF related disintegration are both present and very likely to play a role for pulse durations ≥ 5 µs. With the lower µs pulses, selective RPE disruption might be possible, while higher values allow achieving spatially limited thermal effects without MBF. However, both modi require a very accurate real-time dosing control in order to avoid extended retinal disintegration in this power range.


Assuntos
Traumatismos Oculares/etiologia , Fotocoagulação a Laser/efeitos adversos , Lasers Semicondutores/efeitos adversos , Epitélio Pigmentado da Retina/lesões , Animais , Sobrevivência Celular , Traumatismos Oculares/metabolismo , Traumatismos Oculares/patologia , Angiofluoresceinografia , Fluoresceínas/metabolismo , Microbolhas , Microscopia de Fluorescência , Coelhos , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Suínos
11.
Exp Eye Res ; 197: 108102, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32522477

RESUMO

Primary blast injury (caused by the initial rapid increase in pressure following an explosive blast) to the retina and optic nerve (ON) causes progressive visual loss and neurodegeneration. Military personnel are exposed to multiple low-overpressure blast waves, which may be in quick succession, such as during breacher training or in combat. We investigated the necroptotic cell death pathway in the retina in a mouse repeated primary ocular blast injury (rPBI) model using immunohistochemistry. We further evaluated whether intravitreal injections of a potent necroptosis inhibitor, Necrostatin-1s (Nec-1s), protects the retina and ON axons by retinal ganglion cells (RGC) counts, ON axonal counting and optical coherence tomography (OCT) analysis of vitreous haze. Receptor interacting protein kinase (RIPK) 3, increased in the inner plexiform layer 2 days post injury (dpi) and persisted until 14 dpi, whilst RIPK1 protein expression did not change after injury. The number of degenerating ON axons was increased at 28 dpi but there was no evidence of a reduction in the number of intact ON axons or RNA-binding protein with multiple splicing (RBPMS)+ RGC in the retina by 28 dpi in animals not receiving any intravitreal injections. But, when intravitreal injections (vehicle or Nec-1s) were given there was a significant reduction in RBPMS+ RGC numbers, suggesting that rPBI with intraocular injections is damaging to RGC. There were fewer RGC lost after Nec-1s than vehicle injection, but there was no effect of Nec-1s or vehicle treatment on the number of degenerating axons. OCT analysis demonstrated no effect of rPBI on vitreous haze, but intravitreal injection combined with rPBI increased vitreous haze (P = 0.004). Whilst necroptosis may be an active cell death signalling pathway after rPBI, its inhibition did not prevent cell death, and intravitreal injections in combination with rPBI increased vitreous inflammation and reduced RBPMS+ RGC numbers, implying intravitreal injection is not an ideal method for drug delivery after rPBI.


Assuntos
Traumatismos por Explosões/patologia , Traumatismos Oculares/patologia , Necroptose , Retina/patologia , Animais , Traumatismos por Explosões/metabolismo , Morte Celular , Modelos Animais de Doenças , Eletrorretinografia , Traumatismos Oculares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Retina/metabolismo , Tomografia de Coerência Óptica
12.
Ann N Y Acad Sci ; 1479(1): 65-74, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-31967686

RESUMO

The possibility of chemical terrorism within the United States is a rising concern, with the eye being one of the most sensitive tissues to toxicant exposure. We sought to develop mouse models of toxicant-induced ocular injury for the purpose of evaluating potential therapeutics. Chloropicrin (CP) and hydrogen fluoride (HF) were selected for the study owing to their reportedly high potential to induce ocular injury. Eyes of female BALB/c mice were exposed to CP or HF vapor in order to produce a moderate injury, as defined by acute corneal epithelial loss followed by progressive corneal pathology with the absence of injury to deeper eye structures. Clinical injury progression was evaluated up to 12 weeks postexposure, where a significant dose-dependent induction of corneal neovascularization was measured. Histopathology noted epithelial necrosis and stromal edema as early as 24 h after exposure but was resolved by 12 weeks. A significant increase in inflammatory cytokine concentrations was measured in the cornea 24 h after exposure and returned to baseline by day 14. The ocular injury models we developed here for CP and HF exposure should serve as a valuable tool for the future evaluation of novel therapeutics and the molecular mechanisms of injury.


Assuntos
Neovascularização da Córnea , Traumatismos Oculares , Hidrocarbonetos Clorados/toxicidade , Ácido Fluorídrico/toxicidade , Animais , Neovascularização da Córnea/induzido quimicamente , Neovascularização da Córnea/metabolismo , Neovascularização da Córnea/patologia , Modelos Animais de Doenças , Traumatismos Oculares/induzido quimicamente , Traumatismos Oculares/metabolismo , Traumatismos Oculares/patologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C
13.
Int Immunopharmacol ; 77: 105844, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31669888

RESUMO

BACKGROUND: The serum and tear levels of four inflammatory chemokines were evaluated in sulfur mustard (SM)-exposed with serious ocular problems. MATERIALS AND METHODS: In this study, 128 SM-exposed patients and 31 healthy control participants participated. Tear and serum levels of chemokines were assessed by ELISA method. RESULTS: There was no significant difference in the serum level of IL-8/CXCL8, CX3CL1/fractalkine, CCL2/MCP-1, and CCL5/RANTES between all SM-exposed subjects and control groups. The tear level of IL-8 in the SM-exposed group was lower than the control group, but the difference was not significant. In the SM-exposed group with the abnormalities in tear breakup time (TBUT) test, fundus and pannus formation were significantly higher than SM-exposed patients without these problems. CX3CL1 levels have significantly increased in SM-exposed group with blepharitis, pterygium, and conjunctival pigmentation as compared with the control group. Besides, significantly higher levels of CX3CL1 were observed in SM-exposed group with or without bulbar conjunctival hyperemia and abnormal vessels a well as with fundus abnormality compared to the control group. Only, SM-exposed group with subconjunctival fibrosis had significantly lower levels of CCL5 than SM-exposed group without this problem. CONCLUSION: The higher level of CX3CL1 and consistent levels of IL-8/CXCL8, MCP-1/CCL2, and RANTES/CCL5 in SM-exposed individuals may indicate an anti-inflammatory response against the destructive effects of SM gas. High tear level of IL-8/CXCL8 reflects the severity of ocular surface abnormalities, yet significantly low tear level found in mild SM-exposed subgroup compared with the control group. The lower levels of CX3CL1 and RANTES/CCL5 may represent the different pathophysiology which requires further studies.


Assuntos
Substâncias para a Guerra Química/toxicidade , Citocinas/metabolismo , Traumatismos Oculares/metabolismo , Gás de Mostarda/toxicidade , Lágrimas/metabolismo , Adulto , Citocinas/sangue , Traumatismos Oculares/sangue , Traumatismos Oculares/induzido quimicamente , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença
14.
Cells ; 8(12)2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31779177

RESUMO

Closed-globe injury can cause visual loss in military and civilian populations, with retinal cell death, including retinal ganglion cell (RGC) degeneration, leading to irreversible blindness. RGC and optic nerve (ON) degeneration after eye or head injury is termed traumatic optic neuropathy (TON). There are currently no treatments for RGC loss, therefore novel therapeutics to prevent RGC death or promote axonal regeneration are a priority. We investigated necroptotic signaling mechanisms in a rat blunt ocular injury model. After bilateral blunt trauma, protein expression and retinal localization of necroptosis pathway members (receptor interacting protein kinase 1, RIPK1; receptor interacting protein kinase 3, RIPK3; and mixed lineage kinase domain like pseudokinase, MLKL) were assessed by Western blot and immunohistochemistry (IHC), and potent necroptosis inhibitor Necrostatin-1s (Nec-1s) was delivered by intravitreal injection to one eye and vehicle to the contralateral eye. RGC and photoreceptor survival were assessed by cell counting and outer nuclear layer (ONL) thickness measurements on histology. The neuroprotective effects of Nec-1s were assessed in primary retinal culture by ßIII-tubulin+ RGC cell counts. MLKL protein expression were upregulated at 48 h after injury and MLKL immunolocalised to retinal binding protein with multiple splice (RBPMS)+ RGC, inner nuclear cells and ONL cells, specifically at the retinal injury site. RIPK3 expression did not increase but RIPK3 co-immunolocalised with RBPMS+ RGC in intact and injured retinae. In vitro, a Nec-1s concentration of 0.01 pg/µL was RGC neuroprotective. In the blunt ocular injury rat model, Nec-1s prevented RGC death at the center of the impact site but did not protect against ONL thinning or provide functional restitution. RGC degeneration in our blunt ocular injury model is site-specific, with necroptosis driving death at the center of the focal impact site.


Assuntos
Traumatismos Oculares/etiologia , Traumatismos Oculares/metabolismo , Necroptose , Células Ganglionares da Retina/metabolismo , Animais , Biomarcadores , Sobrevivência Celular , Modelos Animais de Doenças , Traumatismos Oculares/patologia , Expressão Gênica , Imuno-Histoquímica , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Ratos , Proteína Serina-Treonina Quinases de Interação com Receptores , Células Ganglionares da Retina/patologia , Ferimentos não Penetrantes
15.
Int Immunopharmacol ; 77: 105812, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31677500

RESUMO

INTRODUCTION: Sulfur mustard (SM) intoxication produces local and systemic changes in the human body. In this study, the relationship between tear and serum matrix metalloproteinase (MMP)-9 and serum tissue inhibitors of metalloproteinases (TIMPs) are assessed in serious eye-injured SM-exposed casualties. METHODS: A group of 128 SM-exposed patients with serious ocular injuries in three subgroups (19 mild, 31 moderate, and 78 severe cases) is compared with 31 healthy controls. Tear and ocular status and serum MMPs and MMP-9/TIMPs complex levels were evaluated using enzyme-linked immunosorbent assay (ELISA). RESULTS: Serum level of MMP-9 was significantly higher in the SM-exposed group compared to the control group (P = 0.009). Mean serum MMP-9 level in the SM-exposed group with ocular abnormalities was significantly higher than that in the SM-exposed group without ocular abnormalities. SM-exposed people with corneal calcification had significantly higher serum MMP-9/TIMP-1 level compared to the SM-exposed ones without this problem (P = 0.045). The SM-exposed group with severe ocular injuries had significantly higher MMP-9/TIMP-1 than the controls (P = 0.046). The SM-exposed group had significantly lower levels of MMP-9/TIMP-4 complex than the controls (P < 0.001). The SM-exposed group with tear meniscus and fundus abnormality had significantly higher MMP-9/TIMP-4 levels than the SM-exposed group without these problems (P = 0.009 and P = 0.020). CONCLUSION: Serum MMP-9 level had increased in SM-exposed groups with ocular problems, while TIMP-1 and TIMP-2 levels had remained unchanged. Serum TIMP-4 drastically decreased in SM-exposed group, which clearly explains the severity of the systemic and ocular damages.


Assuntos
Substâncias para a Guerra Química/toxicidade , Traumatismos Oculares/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Gás de Mostarda/toxicidade , Lágrimas/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo , Traumatismos Oculares/sangue , Traumatismos Oculares/induzido quimicamente , Humanos , Metaloproteinase 9 da Matriz/sangue , Índice de Gravidade de Doença , Inibidores Teciduais de Metaloproteinases/sangue
16.
Invest Ophthalmol Vis Sci ; 60(13): 4436-4450, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31652328

RESUMO

Purpose: The purpose of this study was to systematically characterize and correlate the transcriptome and DNA methylome signatures of mouse Müller cells that may underlie the development, physiological functions, and regeneration capacity of these cells. Methods: Mouse Müller cells under normal, injury, and aging conditions were sorted from Müller cell-specific green fluorescent protein (GFP)-expressing mice. RNA sequencing was used to sequence transcriptomes, and reduced representation bisulfite sequencing was used to sequence DNA methylomes. Various bioinformatics tools were used to compare and correlate the transcriptomes and DNA methylomes. Results: Müller cells express a distinct transcriptome that is in line with their retinal supporting roles and dormant retinogenic status. Injury changes the Müller cell transcriptome dramatically but fails to stimulate the cell cycle machinery and retinogenic factors to the states observed in early retinal progenitor cells (RPCs). Müller cells exhibit a less methylated genome than that of early RPCs, but most regulatory elements for Müller cell- and RPC-specific genes are similarly hypomethylated in both Müller cells and RPCs, except for a subset of Müller cell-specific functional genes. Aging only subtly affects the transcriptome and DNA methylome of Müller cells. Conclusions: Failure to reactivate the cell cycle machinery and retinogenic factors to necessary levels might be key barriers blocking Müller cells from entering an RPC-like regeneration state. DNA methylation might regulate the expression of a subset of Müller cell-specific functional genes during development but is likely not involved in restricting the regeneration activity of Müller cells.


Assuntos
Senescência Celular/fisiologia , Células Ependimogliais/metabolismo , Epigenoma/genética , Traumatismos Oculares/metabolismo , Neuroglia/metabolismo , Retina/lesões , Transcriptoma/genética , Animais , Ilhas de CpG/genética , Metilação de DNA , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Regeneração/fisiologia , Células-Tronco/metabolismo
17.
Cell Transplant ; 28(9-10): 1257-1270, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31208228

RESUMO

While limbal epithelial cells are used for treating ocular surface wounds, the therapeutic potential of mesenchymal cells cultivated from the limbal stroma (LMSC) is less clear. We have therefore examined the effects of LMSC when applied to acute ocular surface wounds. LMSC derived from male rabbits (RLMSC) were applied to the ocular surface of female rabbits immediately following removal of the corneal and limbal epithelium. Human amniotic membrane (HAM) was used as the vehicle for implanting the RLMSC. The effects of RLMSC were examined when applied alone (n = 3) and in conjunction with a stratified culture of human limbal epithelial cells (HLE) grown on the opposing surface of the HAM (n = 3). Outcomes were monitored over 3 months in comparison with animals receiving no treatment (n = 3) or treatment with HLE alone on HAM (n = 3). Animals treated with RLMSC (n = 6) displayed faster re-epithelialization (∼90% versus 70% healing after 12 weeks), with best results being observed when RLMSC were pre-cultivated and implanted in the presence of HLE (p < 0.01; 90% healing by 7 weeks). While all animals displayed conjunctival cells on the corneal surface (by presence of goblet cells and/or keratin 13 expression) and corneal neovascularization, evidence of corneal epithelial regeneration was observed in animals that received RLMSC in the presence of HLE (by staining for keratin 3 and the absence of goblet cells). Conversely, corneal neovascularization was significantly greater when RLMSC were applied in the absence of HLE (<0.05; 90% of cornea compared with 20-30% in other cohorts). Nevertheless, neither human nuclear antigen nor rabbit Y chromosome were detected within the regenerated epithelium. Our results demonstrate that while cultured LMSC encourage corneal re-epithelialization, healing is improved by the pre-cultivation and implantation of these mesenchymal cells in the presence of limbal epithelial cells.


Assuntos
Células Epiteliais , Epitélio Corneano , Traumatismos Oculares , Limbo da Córnea , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Cicatrização , Doença Aguda , Animais , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitélio Corneano/lesões , Epitélio Corneano/metabolismo , Epitélio Corneano/patologia , Traumatismos Oculares/metabolismo , Traumatismos Oculares/patologia , Traumatismos Oculares/terapia , Feminino , Humanos , Limbo da Córnea/lesões , Limbo da Córnea/metabolismo , Limbo da Córnea/patologia , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Coelhos
18.
Exp Eye Res ; 181: 232-239, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30738069

RESUMO

Proliferative vitreoretinopathy (PVR) is the leading cause of retinal detachment failure. The mechanism of PVR development is complex and still not completely elucidated. There are no proven methods for early prevention or clinical treatment. Retinal proteins are abnormally expressed during the entire PVR disease process. Due to the limitations of research methods and techniques, we do not fully understand the retinal protein changes in PVR. This proteomics study systemically analyzed and identified differential protein expression between retinas of PVR and non-PVR (normal) eyes. Retinal samples were subjected to sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) coupled with mass spectrometry. Raw data were processed and analyzed by Maxquant software and then searched against the human UniProKB (201510) protein database. Differentially expressed proteins were selected and further validated in a human retinal pigment epithelial (RPE) cell line. The effects of dysregulated proteins on cell proliferation, apoptosis, and migration were studied. Systemic proteomics analysis identified several PVR-enriched proteins. The differentially expressed proteins were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) annotation to find abnormal pathways involved in PVR. Retinal-specific ATP-binding cassette transporter (ABCA4) expression was one of the most increased proteins in PVR tissue. ABCA4 knockdown significantly reduced proliferation and affected the cell cycle in the human RPE cell line. ABCA4 knockdown also induced apoptosis and inhibited retinal cell migration. In conclusion, systemic proteomics analysis identified differentially expressed proteins in traumatic PVR, with ABCA4 being highly expressed. Disruption of ABCA4 expression induced apoptosis and inhibited cell proliferation and migration in a human RPE cell line.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Traumatismos Oculares/complicações , Regulação da Expressão Gênica , Proteômica , Epitélio Pigmentado da Retina/metabolismo , Vitreorretinopatia Proliferativa/genética , Transportadores de Cassetes de Ligação de ATP/biossíntese , Western Blotting , Ciclo Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Traumatismos Oculares/metabolismo , Traumatismos Oculares/patologia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Oftalmoscopia , RNA/genética , Epitélio Pigmentado da Retina/patologia , Estudos Retrospectivos , Segmento Externo da Célula Bastonete , Vitreorretinopatia Proliferativa/etiologia , Vitreorretinopatia Proliferativa/metabolismo
19.
Exp Eye Res ; 176: 59-68, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30008389

RESUMO

Alkali burn (AB) is one of the most serious ocular traumas in the world, characterized by extreme ocular surface disorders, critical secondary dry eye and irreversible vision loss. The exact mechanisms involved are unknown. Innate immunity, including the involvement of Toll-like receptors (TLRs) and NOD-like receptors (NLRs), is believed to participate in the pathogenesis of the epithelia, but the exact mechanisms by which TLRs transduce signals to NLRs and downstream molecules to initiate innate immunity remain poorly defined. In this present study, we used murine models of AB and AB concomitant desiccating stress (DS) to investigate the potential functions and mechanisms of TLR4 in regulating NLRP3 and NLRP6 during AB injury and secondary dry eye. We demonstrated that AB injury induced activation of the TLR4-MyD88 pathway, leading to imbalanced NLRP3 and NLRP6 via the activation of caspase-8 signaling. DS worsened ocular surface disorders post-AB injury by magnifying this phenomenon. Caspase-8 signaling promoted NLRP3 upregulation via the nuclear factor (NF)-κB pathway, while NLRP6 suppressed NF-κB activation. Our findings also revealed that TLR4-MyD88 knockout can alleviate AB-induced or DS-worsened ocular surface disorders, shedding light on the potential therapeutic strategies in the future for AB injury. Taken together, our findings demonstrate that AB promotes the TLR4-MyD88-caspase-8 axis to cause imbalanced NLRP3/NLRP6, and DS exacerbates ocular surface damage via magnifying this imbalance.


Assuntos
Queimaduras Químicas/metabolismo , Caspase 8/metabolismo , Traumatismos Oculares/induzido quimicamente , Fator 88 de Diferenciação Mieloide/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Receptores de Superfície Celular/metabolismo , Receptor 4 Toll-Like/fisiologia , Animais , Western Blotting , Modelos Animais de Doenças , Traumatismos Oculares/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Glibureto/farmacologia , Humanos , Hipoglicemiantes/farmacologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Superfície Celular/genética , Transdução de Sinais/fisiologia , Hidróxido de Sódio
20.
Pharmacol Res ; 123: 62-72, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28687341

RESUMO

Glaucoma is one of the leading causes of preventable blindness diseases, affecting more than 2 million people in the United States. Recently, 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) inhibitors were found to exert preventive effects against glaucoma. Therefore, we investigated whether carbenoxolone (CBX), an 11ß-HSD1 inhibitor, prevents chemical ischemia-reperfusion-induced cell death in human trabecular meshwork (HTM) cells. The present study demonstrated that CBX inhibited cell death caused by iodoacetic acid (IAA)-induced ischemia-reperfusion, and its effect was associated with the inhibition of 11ß-HSD1 expression and activity. Furthermore, CBX reversed the IAA-induced structural damage on filamentous actin in HTM cells. In IAA-treated cells, the levels of 11ß-HSD1 and the apoptosis-related factors Bax and FASL were increased throughout the reperfusion period, and CBX was able to attenuate the expression of 11ß-HSD1 and the apoptosis-related factors. CBX also effectively suppressed IAA-induced intracellular ROS formation and cytochrome c release, which are involved in the mitochondrial apoptosis pathway. In addition, IAA-induced chemical ischemia-reperfusion stimulated TNF-α expression and NF-κB p65 phosphorylation, and these effects were attenuated by CBX. 11ß-HSD1 RNAi also suppressed IAA-induced cell apoptosis via reduction of oxidative stress and inhibition of the pro-inflammatory pathway. Taken together, the present study demonstrated that the inhibition of 11ß-HSD1 protected the TM against chemical ischemia-reperfusion injury, suggesting that the use of 11ß-HSD1 inhibitors could be a useful strategy for glaucoma therapy.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/antagonistas & inibidores , Carbenoxolona/farmacologia , Traumatismos Oculares/prevenção & controle , Substâncias Protetoras/farmacologia , Traumatismo por Reperfusão/prevenção & controle , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocromos c/metabolismo , Traumatismos Oculares/induzido quimicamente , Traumatismos Oculares/metabolismo , Humanos , Ácido Iodoacético , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/induzido quimicamente , Traumatismo por Reperfusão/metabolismo , Malha Trabecular/citologia , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA