Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Biomed Pharmacother ; 178: 117270, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39126773

RESUMO

The blood supply in the retina ensures photoreceptor function and maintains regular vision. Leber's hereditary optic neuropathy (LHON), caused by the mitochondrial DNA mutations that deteriorate complex I activity, is characterized by progressive vision loss. Although some reports indicated retinal vasculature abnormalities as one of the comorbidities in LHON, the paracrine influence of LHON-affected retinal ganglion cells (RGCs) on vascular endothelial cell physiology remains unclear. To address this, we established an in vitro model of mitochondrial complex I deficiency using induced pluripotent stem cell-derived RGCs (iPSC-RGCs) treated with a mitochondrial complex I inhibitor rotenone (Rot) to recapitulate LHON pathologies. The secretomes from Rot-treated iPSC-RGCs (Rot-iPSC-RGCs) were collected, and their treatment effect on human umbilical vein endothelial cells (HUVECs) was studied. Rot induced LHON-like characteristics in iPSC-RGCs, including decreased mitochondrial complex I activity and membrane potential, and increased mitochondrial reactive oxygen species (ROS) and apoptosis, leading to mitochondrial dysfunction. When HUVECs were exposed to conditioned media (CM) from Rot-iPSC-RGCs, the angiogenesis of HUVECs was suppressed compared to those treated with CM from control iPSC-RGCs (Ctrl-iPSC-RGCs). Angiogenesis-related proteins were altered in the secretomes from Rot-iPSC-RGC-derived CM, particularly angiopoietin, MMP-9, uPA, collagen XVIII, and VEGF were reduced. Notably, GeneMANIA analysis indicated that VEGFA emerged as the pivotal angiogenesis-related protein among the identified proteins secreted by health iPSC-RGCs but reduced in the secretomes from Rot-iPSC-RGCs. Quantitative real-time PCR and western blots confirmed the reduction of VEGFA at both transcription and translation levels, respectively. Our study reveals that Rot-iPSC-RGCs establish a microenvironment to diminish the angiogenic potential of vascular cells nearby, shedding light on the paracrine regulation of LHON-affected RGCs on retinal vasculature.


Assuntos
Células Endoteliais da Veia Umbilical Humana , Células-Tronco Pluripotentes Induzidas , Atrofia Óptica Hereditária de Leber , Células Ganglionares da Retina , Humanos , Atrofia Óptica Hereditária de Leber/metabolismo , Atrofia Óptica Hereditária de Leber/patologia , Atrofia Óptica Hereditária de Leber/genética , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Rotenona/farmacologia , Meios de Cultivo Condicionados/farmacologia , Apoptose/efeitos dos fármacos , Complexo I de Transporte de Elétrons/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Neovascularização Patológica/metabolismo , Angiogênese
2.
Antioxidants (Basel) ; 13(6)2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38929182

RESUMO

Oxidative stress is a key factor causing mitochondrial dysfunction and retinal ganglion cell (RGC) death in glaucomatous neurodegeneration. The cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) signaling pathway is involved in mitochondrial protection, promoting RGC survival. Soluble adenylyl cyclase (sAC) is a key regulator of the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) signaling pathway, which is known to protect mitochondria and promote RGC survival. However, the precise molecular mechanisms connecting the sAC-mediated signaling pathway with mitochondrial protection in RGCs against oxidative stress are not well characterized. Here, we demonstrate that sAC plays a critical role in protecting RGC mitochondria from oxidative stress. Using mouse models of oxidative stress induced by ischemic injury and paraquat administration, we found that administration of bicarbonate, as an activator of sAC, protected RGCs, blocked AMP-activated protein kinase activation, inhibited glial activation, and improved visual function. Moreover, we found that this is the result of preserving mitochondrial dynamics (fusion and fission), promoting mitochondrial bioenergetics and biogenesis, and preventing metabolic stress and apoptotic cell death. Notably, the administration of bicarbonate ameliorated mitochondrial dysfunction in RGCs by enhancing mitochondrial biogenesis, preserving mitochondrial structure, and increasing ATP production in oxidatively stressed RGCs. These findings suggest that activating sAC enhances the mitochondrial structure and function in RGCs to counter oxidative stress, consequently promoting RGC protection. We propose that modulation of the sAC-mediated signaling pathway has therapeutic potential acting on RGC mitochondria for treating glaucoma and other retinal diseases.

3.
Exp Eye Res ; 244: 109929, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38750783

RESUMO

Optic nerve injuries are severely disrupt the structural and functional integrity of the retina, often leading to visual impairment or blindness. Despite the profound impact of these injuries, the molecular mechanisms involved remain poorly understood. In this study, we performed a comprehensive whole-transcriptome analysis of mouse retina samples after optic nerve crush (ONC) to elucidate changes in gene expression and regulatory networks. Transcriptome analysis revealed a variety of molecular alterations, including 256 mRNAs, 530 lncRNAs, and 37 miRNAs, associated with metabolic, inflammatory, signaling, and biosynthetic pathways in the injured retina. The integrated analysis of co-expression and protein-protein interactions identified an active interconnected module comprising 5 co-expressed proteins (Fga, Serpina1a, Hpd, Slc38a4, and Ahsg) associated with the complement and coagulation cascades. Finally, 5 mRNAs (Fga, Serpinala, Hpd, Slc38a4, and Ahsg), 2 miRNAs (miR-671-5p and miR-3057-5p), and 6 lncRNAs (MSTRG. 1830.1, Gm10814, A530013C23Rik, Gm40634, MSTRG.9514.1, A330023F24Rik) were identified by qPCR in the injured retina, and some of them were validated as critical components of a ceRNA network active in 661W and HEK293T cells through dual-luciferase reporter assays. In conclusion, our study provides comprehensive insight into the complex and dynamic biological mechanisms involved in retinal injury responses and highlights promising potential targets to enhance neuroprotection and restore vision.


Assuntos
Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Camundongos Endogâmicos C57BL , Traumatismos do Nervo Óptico , RNA Mensageiro , Retina , Animais , Camundongos , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/genética , Retina/metabolismo , RNA Mensageiro/genética , Modelos Animais de Doenças , Transcriptoma , MicroRNAs/genética , Regulação da Expressão Gênica/fisiologia , Masculino , Humanos , RNA Longo não Codificante/genética
4.
Int J Mol Sci ; 25(6)2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-38542089

RESUMO

Glaucoma is a neurodegenerative disease that causes blindness. In this study, we aimed to evaluate the protective role of cilastatin (CIL), generally used in the treatment of nephropathologies associated with inflammation, in an experimental mouse model based on unilateral (left) laser-induced ocular hypertension (OHT). Male Swiss mice were administered CIL daily (300 mg/kg, i.p.) two days before OHT surgery until sacrifice 3 or 7 days later. Intraocular Pressure (IOP), as well as retinal ganglion cell (RGC) survival, was registered, and the inflammatory responses of macroglial and microglial cells were studied via immunohistochemical techniques. Results from OHT eyes were compared to normotensive contralateral (CONTRA) and naïve control eyes considering nine retinal areas and all retinal layers. OHT successfully increased IOP values in OHT eyes but not in CONTRA eyes; CIL did not affect IOP values. Surgery induced a higher loss of RGCs in OHT eyes than in CONTRA eyes, while CIL attenuated this loss. Similarly, surgery increased macroglial and microglial activation in OHT eyes and to a lesser extent in CONTRA eyes; CIL prevented both macroglial and microglial activation in OHT and CONTRA eyes. Therefore, CIL arises as a potential effective strategy to reduce OHT-associated damage in the retina of experimental mice.


Assuntos
Glaucoma , Doenças Neurodegenerativas , Hipertensão Ocular , Masculino , Camundongos , Animais , Doenças Neurodegenerativas/complicações , Glaucoma/etiologia , Hipertensão Ocular/tratamento farmacológico , Hipertensão Ocular/patologia , Pressão Intraocular , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Cilastatina/uso terapêutico , Modelos Animais de Doenças
5.
Exp Eye Res ; 241: 109834, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38382575

RESUMO

Myopia and astigmatism are two primary types of refractive errors characterized by inaccurate focusing images on the retina. This study aimed to investigate the response characteristics of Retinal Ganglion Cells (RGCs), represented by alpha (α) RGCs, when exposed to focused, simulated spherically defocused images and astigmatically defocused images projected onto mouse retinas. Negative pressure was applied to stretch the soma of RGC in vitro to simulate myopia using a 7-8 µm diameter glass microelectrode, resulting in a 5% increase in the cell's diameter. A custom-made device was utilized to project spherically (equal to ±10 and ± 20 D) and astigmatically (+6.00 D) defocused images onto the retinas. As a control for a deficient intact retinal circuit, αRGCs of connexin 36 knockout (Cx36 KO) mice were used. The response of αRGCs varied significantly in terms of spikes, excitatory postsynaptic currents (EPSCs) and capacitances under stretching conditions to mimic myopia. Significant differences in the amplitudes of EPSCs were observed in the majority of αRGCs when exposed to focused and spherically defocused images in normal and mechanically simulated myopic retinas. However, this difference was not observed in αRGCs of Cx36 KO mice. αRGCs demonstrated significant differences in response between focused and astigmatically defocused images. Once again, αRGCs of Cx36 KO mice did not display differences. αRGCs have the ability to detect focused, spherically, and astigmatically defocused images and exhibit differential responses ex vivo. Gap junction subunit Cx36 may play a crucial role in transmitting visual signals associated with developing and perceiving refractive errors.


Assuntos
Miopia , Células Ganglionares da Retina , Animais , Camundongos , Retina , Junções Comunicantes
6.
Exp Eye Res ; 239: 109781, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38184223

RESUMO

In addition to regulating cholesterol synthesis, statins have neuroprotective effects. Apoptosis of retinal ganglion cells (RGCs) causes a gradual loss of visual function in glaucoma. This study aimed to investigate the neuroprotective effect of statins on the RGC apoptosis induced by activated Müller glia. Primary Müller cells and RGCs were cultured from the retina of C57BL6 mice. Müller cells were activated with GSK101, a transient receptor potential vanilloid 4 (TRPV4) agonist, and tumor necrosis factor-alpha (TNF-α) released to the medium was measured using an enzyme-linked immunosorbent assay. Cells were pretreated with simvastatin or lovastatin before GSK101. RGCs were treated with conditioned media from Müller glia cultures, and apoptosis was determined using flow cytometry. TRPV4 activation through GSK101 treatment induced gliosis of Müller cells, and the conditioned media from activated Müller cells was potent to induce RGC apoptosis. Statins suppress both gliosis in Müller cells and subsequent RGC apoptosis. TNF-α release to the media was increased in GSK101-treated Müller cells, and TNF-α in the conditioned media was the critical factor causing RGC apoptosis. The increase in TRPV4-mediated TNF-α expression occurred through the nuclear factor kappa-light chain enhancer of activated B cell pathway activation, which was inhibited by statins. Herein, we showed that statins can modulate gliosis and TNF-α expression in Müller cells, protecting RGCs. These data further support the neuroprotective effect of statins, promoting them as a potential treatment for glaucoma.


Assuntos
Antineoplásicos , Glaucoma , Inibidores de Hidroximetilglutaril-CoA Redutases , Fármacos Neuroprotetores , Animais , Camundongos , Antineoplásicos/farmacologia , Apoptose , Meios de Cultivo Condicionados/farmacologia , Células Ependimogliais/metabolismo , Glaucoma/tratamento farmacológico , Glaucoma/patologia , Gliose/patologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Canais de Cátion TRPV/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
Biomed Mater ; 19(2)2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38181445

RESUMO

Ciliary neurotrophic factor (CNTF) promotes survival and/or differentiation of a variety of neuronal cells including retinal ganglion cells (RGCs). Delivery of CNTF requires a suitable medium capable of mediating diffusion and premature release of CNTF within the target tissue. Polymeric tissue-engineered scaffolds have been readily used as substrates for cell transplantation, expansion, and differentiation and, as carriers of cell growth factors. Their functions to CNTF release for RGC proliferation have remained so far unexplored, especially to CNTF affinity to the scaffold and subsequent RGC fate. Electrospunpoly(glycerol sebacate)/poly(ϵ-caprolactone) (PGS/PCL) biopolymer scaffolds have recently shown promising results in terms of supporting regeneration of RGC neurites. This work explores covalent immobilization of CNTF on PGS/PCL scaffold and the way immobilised CNTF mediates growth of RGC axons on the scaffold. Anex-vivothree-dimensional model of rodent optic nerve on PGS/PCL revealed that RGC explants cultured in CNTF mediated environment increased their neurite extensions after 20 d of cell culture employing neurite outgrowth measurements. The CNTF secretion on PGS/PCL scaffold was found bio-mimicking natural extracellular matrix of the cell target tissue and, consequently, has shown a potential to improve the overall efficacy of the RGC regeneration process.


Assuntos
Fator Neurotrófico Ciliar , Células Ganglionares da Retina , Células Ganglionares da Retina/metabolismo , Fator Neurotrófico Ciliar/metabolismo , Axônios/fisiologia , Neuritos/metabolismo , Proliferação de Células , Regeneração Nervosa/fisiologia , Sobrevivência Celular/fisiologia
8.
Exp Eye Res ; 234: 109616, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37580002

RESUMO

The etiology of myopia remains unclear. This study investigated whether retinal ganglion cells (RGCs) in the myopic retina encode visual information differently from the normal retina and to determine the role of Connexin (Cx) 36 in this process. Generalized linear models (GLMs), which can capture stimulus-dependent changes in real neurons with spike timing precision and reliability, were used to predict RGCs responses to focused and defocused images in the retinas of wild-type (normal) and Lens-Induced Myopia (LIM) mice. As the predominant subunit of gap junctions in the mouse retina and a plausible modulator in myopia development, Cx36 knockout (KO) mice were used as a control for an intact retinal circuit. The kinetics of excitatory postsynaptic currents (EPSCs) of a single αRGC could reflect projection of both focused and defocused images in the retinas of normal and LIM, but not in the Cx36 knockout mice. Poisson GLMs revealed that RGC encoding of visual stimuli in the LIM retina was similar to that of the normal retina. In the LIM retinas, the linear-Gaussian GLM model with offset was a better fit for predicting the spike count under a focused image than the defocused image. Akaike information criterion (AIC) indicated that nonparametric GLM (np-GLM) model predicted focused/defocused images better in both LIM and normal retinas. However, the spike counts in 33% of αRGCs in LIM retinas were better fitted by exponential GLM (exp-GLM) under defocus, compared to only 13% αRGCs in normal retinas. The differences in encoding performance between LIM and normal retinas indicated the possible amendment and plasticity of the retinal circuit in myopic retinas. The absence of a similar response between Cx36 KO mice and normal/LIM mice might suggest that Cx36, which is associated with myopia development, plays a role in encoding focused and defocused images.


Assuntos
Miopia , Células Ganglionares da Retina , Animais , Camundongos , Células Ganglionares da Retina/fisiologia , Reprodutibilidade dos Testes , Retina , Miopia/etiologia , Camundongos Knockout
9.
Methods Mol Biol ; 2708: 155-174, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37558970

RESUMO

The injection of therapies into the eye is common practice, both clinically and pre-clinically. The most straightforward delivery route is via an intravitreal injection, which introduces the treatment into the largest cavity at the posterior of the eye. This technique is frequently used to deliver gene therapies, including those containing recombinant adeno-associated viral vectors (AAVs), to the back of the eye to enable inner retinal targeting. This chapter provides detailed methodology on how to successfully perform an intravitreal injection in mice. The chapter covers vector preparation considerations, advice on how to minimize vector loss in the injection device, and ways to reduce vector reflux from the eye when administering a therapy. Finally, a protocol is provided on common retinal histology processing techniques to assess vector-mediated expression in retinal ganglion cells. It is hoped that this chapter will enable researchers to carry out effective and consistent intravitreal injections that transduce the inner retinal surface while avoiding common pitfalls.


Assuntos
Retina , Células Ganglionares da Retina , Camundongos , Animais , Injeções Intravítreas , Transdução Genética , Retina/metabolismo , Terapia Genética/métodos , Dependovirus/genética , Vetores Genéticos/genética
11.
J Ocul Pharmacol Ther ; 39(8): 509-518, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37172141

RESUMO

Purpose: There is increasing interest in nonpharmacologic approaches to protect retinal ganglion cells (RGCs) after injury and enhance the efficacy of therapeutic molecules. Accumulating evidence demonstrates neuroprotection by the high-fat low-carbohydrate ketogenic diet (KD) in humans and animal models of neurologic diseases. However, no studies to date have examined whether the KD protects RGCs and promotes axonal regrowth after traumatic injury to the optic nerve (ON) or whether it increases efficacy of experimental proregenerative molecules. In this study, we investigated whether the KD promoted RGC survival and axonal regeneration after ON injury in the presence and absence of neuroprotective Wnt3a ligand. Methods: Adult mice were placed on a KD or control diet before ON crush injury and remained on the diet until the end of the experiment. Nutritional ketosis was confirmed by measuring serum beta-hydroxybutyrate levels. Mice were intravitreally injected with Wnt3a ligand or phosphate-buffered saline (PBS), and RGC survival, function, axonal regeneration, and inflammatory responses were measured. Results: Mice fed the KD showed increased RGC survival and reduced inflammatory cells in PBS-injected mice. Also, mice fed the KD had increased RGC functional responses but not increased RGC numbers in the presence of Wnt3a, indicating that the KD did not enhance the prosurvival effect of Wnt3a. The KD did not promote axonal regeneration in the presence or absence of Wnt3a. Conclusions: The KD has a complex protective effect after ON injury and cotreatment with Wnt3a. This work sets the foundation for studies identifying underlying molecular mechanisms.


Assuntos
Dieta Cetogênica , Traumatismos do Nervo Óptico , Humanos , Camundongos , Animais , Traumatismos do Nervo Óptico/tratamento farmacológico , Células Ganglionares da Retina , Ligantes , Regeneração Nervosa/fisiologia , Sobrevivência Celular , Modelos Animais de Doenças
12.
CNS Neurosci Ther ; 29 Suppl 1: 146-160, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36924268

RESUMO

INTRODUCTION: Optic nerve injury is a leading cause of irreversible blindness worldwide. The retinal ganglion cells (RGCs) and their axons cannot be regenerated once damaged. Therefore, reducing RGC damage is crucial to prevent blindness. Accordingly, we aimed to investigate the potential influence of the gut microbiota on RGC survival, as well as the associated action mechanisms. METHODS: We evaluated the effects of microbiota, specifically Bifidobacterium, on RGC. Optic nerve crush (ONC) was used as a model of optic nerve injury. Vancomycin and Bifidobacterium were orally administered to specific pathogen-free (SPF) mice. RESULTS: Bifidobacterium promoted RGC survival and optic nerve regeneration. The administration of Bifidobacterium inhibited microglia activation but promoted Müller cell activation, which was accompanied by the downregulation of inflammatory cytokines and upregulation of neurotrophic factors and retinal ERK/Fos signaling pathway activation. CONCLUSIONS: Our study demonstrates that Bifidobacterium-induced changes in intestinal flora promote RGC survival. The protective effect of Bifidobacterium on RGC can be attributed to the inhibition of microglia activation and promotion of Müller cell activation and the secondary regulation of inflammatory and neurotrophic factors.


Assuntos
Traumatismos do Nervo Óptico , Células Ganglionares da Retina , Camundongos , Animais , Células Ganglionares da Retina/metabolismo , Traumatismos do Nervo Óptico/terapia , Traumatismos do Nervo Óptico/metabolismo , Neuroglia/metabolismo , Axônios/metabolismo , Fatores de Crescimento Neural/metabolismo , Cegueira/metabolismo , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças
13.
Appl Psychophysiol Biofeedback ; 48(3): 311-321, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-36971985

RESUMO

Light-induced effects on the autonomic nervous system (ANS) are assumed to be mediated by retinal projections to the hypothalamic suprachiasmatic nucleus (SCN) via different routes. Light information for the circadian system is detected by a subset of intrinsically photosensitive retinal ganglion cells (ipRGCs), however, inconsistency exists in research concerning the effects of light exposure on heart rate variability (HRV). Two within-subject experiments were conducted in a standardized sleep laboratory to investigate effects of light intensity (study I, n = 29: 2 days dim vs. bright light) and spectral composition (study II, n = 24: 3 days using red vs. blue vs. green light) on HRV parameters (RMSSD, LF, HF-HRV, LF/HF ratio). Light exposure was conducted for one-hour in the post-awakening phase at 5:00 AM. Results revealed no significant light intensity effect comparing dim light versus bright white light on HRV parameters. Light color of different wavelengths significantly influenced all HRV parameters except the low frequency, with moderate to large effect sizes. RMSSD values were elevated for all three colors compared to norm values, indicating stronger parasympathetic activation. LED light of different spectral compositions demonstrated bidirectional effects on spectral components of the HRV. Red light decreased the LF/HF ratio within 30 min, whereas with blue light, LF/HF ratio consistently increased across 40 min of light exposure.


Assuntos
Sistema Nervoso Autônomo , Sono , Humanos , Masculino , Frequência Cardíaca/fisiologia
14.
BMC Ophthalmol ; 22(1): 502, 2022 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-36539722

RESUMO

BACKGROUND: Optic nerve trauma caused by crush injury is frequently used for investigating experimental treatments that protect retinal ganglion cells (RGCs) and induce axonal regrowth. Retaining outer retinal light responses is essential for therapeutic rescue of RGCs after injury. However, whether optic nerve crush also damages the structure or function of photoreceptors has not been systematically investigated. In this study, we investigated whether outer retinal thickness and visual function are altered by optic nerve crush in the mouse. METHODS: Wildtype mice underwent optic nerve crush and intravitreal injection of a control solution in one eye with the fellow eye remaining uninjured. Two weeks after injury, the thickness of the ganglion cell region (GCL to IPL) and photoreceptor layer (bottom of the OPL to top of the RPE) were measured using OCT. Retinal function was assessed using flash ERGs. Immunodetection of RGCs was performed on retinal cryosections and RGCs and ONL nuclei rows were counted. Multiple comparison analyses were conducted using Analysis of Variance (ANOVA) with Tukey's post hoc test and P values less than 0.05 were considered statistically significant. RESULTS: Optic nerve crush injury induced RGC death as expected, demonstrated by thinning of the ganglion cell region and RGC loss. In contrast, outer retinal thickness, photopic and scotopic a-wave and b-wave amplitudes and photoreceptor nuclei counts, were equivalent between injured and uninjured eyes. CONCLUSIONS: Secondary degeneration of the outer retina was not detected after optic nerve injury in the presence of significant RGC death, suggesting that the retina has the capacity to compartmentalize damage. These findings also indicate that experimental treatments to preserve the GCL and rescue vision using this optic nerve injury model would not require additional strategies to preserve the ONL.


Assuntos
Lesões por Esmagamento , Traumatismos do Nervo Óptico , Camundongos , Animais , Retina , Células Ganglionares da Retina , Nervo Óptico , Lesões por Esmagamento/complicações , Lesões por Esmagamento/metabolismo , Compressão Nervosa , Modelos Animais de Doenças
15.
Ann Transl Med ; 10(6): 312, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35433984

RESUMO

Background: Glaucoma is the second leading cause of blindness in the world and is characterized by optic neuropathy and degeneration of retinal ganglion cells (RGCs). Our preliminary research found that acteoside can inhibit autophagy-induced apoptosis of RGCs via the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway. However, it is unclear how acteoside activates the PI3K/AKT signaling pathway to prevents RGCs autophagic apoptosis. Methods: Animal and cell models were used in this study. Hematoxylin-eosin staining revealed pathological histology of retinas. The number of RGCs in retinas was counted using immunofluorescence. Malondialdehyde and superoxide dismutase were determined using enzyme-linked immunosorbent assay kits. Flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining were used to detect cell apoptosis. The reactive oxygen species was determined by the Flow cytometry. The proteins were determined by Western blot. Results: The results showed that acteoside treatment significantly reduced RGC loss, oxidative stress, and autophagy, thereby preventing glaucoma exacerbation. Acteoside reversed caveolin 1 (Cav1) expression and PI3K/AKT signaling activation, according to Western blot results. Cav1 knockdown also reversed acteoside's effects on RGC loss, PI3K/AKT signaling pathway activation, autophagy and oxidative stress. Notably, 3-methyladenine, a PI3K inhibitor, reversed the effects of acteoside and Cav1 overexpression on RGC loss, oxidative stress, and autophagy. Conclusions: These finding imply that acteoside alleviates RGC loss and oxidative stress by activating of the PI3K/AKT signaling pathway by upregulating Cav1.

16.
Int J Mol Sci ; 23(6)2022 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-35328623

RESUMO

Patients with congenital nystagmus, involuntary eye movements, often have a reduced visual acuity. Some of these patients have a retinal-specific mutation in the protein nyctalopin, which is also present in the Nyxnob mouse. In these mice, retinal ganglion cells (RGCs) have oscillatory activity, which leads to expanded axonal projections towards the dLGN and consequently to a desegregation of retinal projections to the brain. In this study, we investigate whether the receptive fields of Nyxnob RGCs have also expanded by measuring the size of their receptive fields using MEA recordings. Contrary to our expectation, relative to wild-type (WT) mice we found receptive field sizes in the Nyxnob retina had not increased but instead had decreased for green-light preferring RGCs. Additionally, we also found the receptive fields of UV-light preferring RGCs are larger than green-light preferring RGCs in both WT and Nyxnob mice.


Assuntos
Retina , Células Ganglionares da Retina , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteoglicanas/genética
17.
Eur J Ophthalmol ; 32(5): 2589-2597, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34967226

RESUMO

PURPOSE: To explore the optimized concentration of AAV2-GFP for sparse transfection of retinal ganglion cells (RGCs) and optic nerve (ON), and to examine the changes of microglial morphology and distribution in the retina, optic nerve and chiasm after injection. METHODS: We defined the optimal concentration of AAV2-GFP for sparse labeling of RGCs and axons in WT mice. We further explored the changes of microglial morphology and distribution in the retina, optic nerve and chiasm after intravitreal injection in CX3CR1+/GFP mice. RESULTS: 14 days after intravitreal injection of AAV2-GFP, live imaging of the retina showed that fundus fluorescence was very strong and dense at 2.16 × 1011 VG/retina, 2.16 × 1010 VG/retina, 2.16 × 109 VG/retina. RGCs were sparsely marked at a concentration 1:1000 (2.16 × 108 VG/retina) and fundus fluorescence was weak. The transfected RGCs and axons were unevenly distributed in the retina and significantly more RGCs were transfected near the injection site of AAV2-GFP compared to the other sites of the flat-mounted retina. Microglia density increased significantly in the retina and part of optic nerve, but not in the optic chiasm. The morphology of microglia was largely unchanged. CONCLUSIONS: AAV2-GFP was highly efficient and the optimal concentration of sparsely labeled RGCs was 1:1000 (2.16 × 108 VG/retina). After intravitreal injection of AAV2-GFP, the number of microglia increased partly. The morphology of microglia was comparable.


Assuntos
Microglia , Nervo Óptico , Animais , Injeções Intravítreas , Camundongos , Retina , Células Ganglionares da Retina
18.
Cell Biosci ; 11(1): 97, 2021 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-34039431

RESUMO

BACKGROUND: The mechanisms leading to retinal ganglion cell (RGC) death after optic nerve injury have not been fully elucidated. Current evidence indicates that microglial activation and M1- and M2-like dynamics may be an important factor in RGC apoptosis after optic nerve crush (ONC). Semaphorin3A (Sema3A) is a classic axonal guidance protein,which has been found to have a role in neuroinflammation processes. In this study, we investigated the contribution of microglial-derived Sema3A to progressive RGC apoptosis through regulating paradigm of M1- and M2-like microglia after ONC. METHOD: A mouse ONC model and a primary microglial-RGC co-culture system were used in the present study. The expression of M1- and M2-like microglial activation markers were assessed by real-time polymerase chain reaction (RT-qPCR). Histological and Western blot (WB) analyses were used to investigate the polarization patterns of microglia transitions and the levels of Sema3A. RGC apoptosis was investigated by TUNEL staining and caspase-3 detection. RESULTS: Levels of Sema3A in the mouse retina increased after ONC. Treatment of mice with the stimulating factor 1 receptor antagonist PLX3397 resulted in a decrease of retinal microglia. The levels of CD16/32 (M1) were up-regulated at days 3 and 7 post-ONC. However, CD206 (M2) declined on day 7 after ONC. Exposure to anti-Sema3A antibodies (anti-Sema3A) resulted in a decrease in the number of M1-like microglia, an increase in the number of M2-like microglia, and the amelioration of RGC apoptosis. CONCLUSIONS: An increase in microglia-derived Sema3A in the retina after ONC partially leads to a continuous increase of M1-like microglia and plays an important role in RGC apoptosis. Inhibition of Sema3A activity may be a novel approach to the prevention of RGC apoptosis after optic nerve injury.

19.
Front Cell Neurosci ; 15: 664491, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34025362

RESUMO

Low-dose atropine helps to control myopia progression with few side effects. However, the impact of atropine, a non-selective muscarinic Acetylcholine (ACh) receptor antagonist, on retinal ganglion cells (RGCs) remains unclear. After immersing the cornea and adjacent conjunctiva of enucleated eyes in 0.05% (approximately 800 µM) atropine solution for 30 min, the atropine concentration reached in the retina was below 2 µM. After direct superfusion of the retina with 1 µM atropine (considering that the clinical application of 0.05% atropine eye drops will be diluted over time due to tear flow for 30 min), no noticeable changes in the morphology of ON and OFF alpha RGCs (αRGCs) were observed. Atropine affected the light-evoked responses of ON and OFF αRGCs in a dose- and time-dependent fashion. Direct application of less than 100 µM atropine on the retina did not affect light-evoked responses. The time latency of light-induced responses of ON or OFF αRGCs did not change after the application of 0.05-100 µM atropine for 5 min. However, 50 µM atropine extended the threshold of joint inter-spike interval (ISI) distribution of the RGCs. These results indicated that low-dose atropine (<0.5 µM; equal to 1% atropine topical application) did not interfere with spike frequency, the pattern of synchronized firing between OFF αRGCs, or the threshold of joint ISI distribution of αRGCs. The application of atropine unmasked inhibition to induce ON responses from certain OFF RGCs, possibly via the GABAergic pathway, potentially affecting visual information processing.

20.
Cent Eur J Immunol ; 46(1): 105-110, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33897291

RESUMO

Glaucoma is the neurodegenerative disease of retinal ganglion cells. The main risk factor for glaucoma is increased intraocular pressure. The processes leading to cell death due to presence of the injury factor comprise multiple molecular mechanisms, as well as the immunological response. The knowledge of immunological mechanisms occurring in glaucomatous degeneration makes it possible to introduce glaucoma treatment modulating the cellular degradation. The glaucoma treatment of the future will make it possible not only to lower the intraocular pressure, but also to moderate the intracellular mechanisms in order to prevent retinal cell degeneration. Citicoline is a drug modulating glutamate excitotoxicity that is already in use. Rho kinase inhibitors were found to stimulate neurite growth and axon regeneration apart from lowering intraocular pressure. The complementary action of brimonidine is to increase neurotrophic factor (NTF) concentrations and inhibit glutamate toxicity. Immunomodulatory therapies with antibodies and gene therapies show promising effects in the current studies. The supplementation of NTFs prevents glaucomatous damage. Resveratrol and other antioxidants inhibit reactive oxygen species formation. Cell transplantation of stem cells, Schwann cells and nerve extracts was reported to be successful so far. Our review presents the most promising new strategies of neuroprotection and immunomodulation in glaucoma.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...