Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.166
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732096

ABSTRACT

Alterations in intraocular and external pressure critically involve the pathogenesis of glaucoma, traumatic retinal injury (TRI), and other retinal disorders, and retinal neurons have been reported to express multiple mechanical-sensitive channels (MSCs) in recent decades. However, the role of MSCs in visual functions and pressure-related retinal conditions has been unclear. This review will focus on the variety and functional significance of the MSCs permeable to K+, Na+, and Ca2+, primarily including the big potassium channel (BK); the two-pore domain potassium channels TRAAK and TREK; Piezo; the epithelial sodium channel (ENaC); and the transient receptor potential channels vanilloid TRPV1, TRPV2, and TRPV4 in retinal photoreceptors, bipolar cells, horizontal cells, amacrine cells, and ganglion cells. Most MSCs do not directly mediate visual signals in vertebrate retinas. On the other hand, some studies have shown that MSCs can open in physiological conditions and regulate the activities of retinal neurons. While these data reasonably predict the crossing of visual and mechanical signals, how retinal light pathways deal with endogenous and exogenous mechanical stimulation is uncertain.


Subject(s)
Ion Channels , Retinal Neurons , Humans , Animals , Ion Channels/metabolism , Retinal Neurons/metabolism , Mechanotransduction, Cellular , Retina/metabolism , Retina/cytology
2.
Sci Rep ; 14(1): 3380, 2024 02 09.
Article in English | MEDLINE | ID: mdl-38336828

ABSTRACT

Optical coherence tomography (OCT) has become a key method for diagnosing and staging radiation retinopathy, based mainly on the presence of fluid in the central macula. A robust retinal layer segmentation method is required for identification of the specific layers involved in radiation-induced pathology in individual eyes over time, in order to determine damage driven by radiation injury to the microvessels and to the inner retinal neurons. Here, we utilized OCT, OCT-angiography, visual field testing, and patient-specific dosimetry models to analyze abnormal retinal layer thickening and thinning relative to microvessel density, visual function, radiation dose, and time from radiotherapy in a cross-sectional cohort of uveal melanoma patients treated with 125I-plaque brachytherapy. Within the first 24 months of radiotherapy, we show differential thickening and thinning of the two inner retinal layers, suggestive of microvessel leakage and neurodegeneration, mostly favoring thickening. Four out of 13 eyes showed decreased inner retinal capillary density associated with a corresponding normal inner retinal thickness, indicating early microvascular pathology. Two eyes showed the opposite: significant inner retinal layer thinning and normal capillary density, indicating early neuronal damage preceding a decrease in capillary density. At later time points, inner retinal thinning becomes the dominant pathology and correlates significantly with decreased vascularity, vision loss, and dose to the optic nerve. Stable multiple retinal layer segmentation provided by 3D graph-based methods aids in assessing the microvascular and neuronal response to radiation, information needed to target therapeutics for radiation retinopathy and vision loss.


Subject(s)
Radiation Injuries , Retinal Degeneration , Retinal Neurons , Humans , Visual Field Tests , Tomography, Optical Coherence/methods , Cross-Sectional Studies , Retina/diagnostic imaging , Retina/pathology , Retinal Neurons/pathology , Retinal Degeneration/pathology , Radiation Injuries/etiology , Radiation Injuries/pathology
3.
Int J Mol Sci ; 25(3)2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38339191

ABSTRACT

We generated a novel Cre mouse strain for cell-specific deletion of floxed genes in ribbon synapse-forming retinal neurons. Previous studies have shown that the RIBEYE promotor targets the expression of recombinant proteins such as fluorescently tagged RIBEYE to photoreceptors and retinal bipolar cells and generates fluorescent synaptic ribbons in situ in these neurons. Here, we used the same promotor to generate a novel transgenic mouse strain in which the RIBEYE promotor controls the expression of a Cre-ER(T2) recombinase (RIBEYE-Cre). To visualize Cre expression, the RIBEYE-Cre animals were crossed with ROSA26 tau-GFP (R26-τGFP) reporter mice. In the resulting RIBEYE-Cre/R26 τGFP animals, Cre-mediated removal of a transcriptional STOP cassette results in the expression of green fluorescent tau protein (tau-GFP) that binds to cellular microtubules. We detected robust tau-GFP expression in retinal bipolar cells. Surprisingly, we did not find fluorescent tau-GFP expression in mouse photoreceptors. The lack of tau-GFP reporter protein in these cells could be based on the previously reported absence of tau protein in mouse photoreceptors which could lead to the degradation of the recombinant tau protein. Consistent with this, we detected Cre and tau-GFP mRNA in mouse photoreceptor slices by RT-PCR. The transgenic RIBEYE-Cre mouse strain provides a new tool to study the deletion of floxed genes in ribbon synapse-forming neurons of the retina and will also allow for analyzing gene deletions that are lethal if globally deleted in neurons.


Subject(s)
Retinal Neurons , tau Proteins , Mice , Animals , tau Proteins/metabolism , Mice, Transgenic , Retinal Neurons/metabolism , Synapses/metabolism , Integrases/genetics , Integrases/metabolism , Green Fluorescent Proteins/metabolism
4.
Neurol Sci ; 45(6): 2615-2623, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38216851

ABSTRACT

PURPOSE: To compare the peripapillary retinal nerve fiber layer (pRNFL), retinal nerve fiber layer (RNFL), and ganglion cell complex (GCC) thickness measurement in early-onset Alzheimer's disease (EOAD) and controls using spectral domain optical coherence tomography (SD-OCT). We also assessed the relationship between SD-OCT measurements and cognitive measures, serum biomarkers for Alzheimer's disease (AD), and cerebral microstructural volume. METHODS: pRNFL, RNFL, and GCC thicknesses were measured in 43 EOAD and 42 controls using SD-OCT. Montreal Cognitive Assessment (MoCA) and Mini-Mental State Examination (MMSE) were used to assess cognitive status, magnetic resonance imaging (MRI) tool was used to quantify cerebral microstructural volume, and serum biomarkers were quantified from peripheral blood. RESULTS: EOAD patients had thinner pRNFL (P < 0.001), RNFL (P = 0.008), and GCC (P = 0.018) thicknesses compared to controls after adjusting for multiple factors. pRNFL thickness correlated (P = 0.016) with serum t-tau level. Serum Aß42 (P < 0.05) concentration correlated with RNFL thickness. Importantly, occipital lobe volume (P = 0.010) correlated with GCC thicknesses in EOAD patients. CONCLUSION: Our findings suggest that retinal thickness may be useful markers for assessing neurodegenerative process in EOAD.


Subject(s)
Alzheimer Disease , Biomarkers , Brain , Tomography, Optical Coherence , Humans , Alzheimer Disease/blood , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Male , Female , Biomarkers/blood , Middle Aged , Brain/diagnostic imaging , Brain/pathology , Magnetic Resonance Imaging , Amyloid beta-Peptides/blood , tau Proteins/blood , Retina/pathology , Retina/diagnostic imaging , Aged , Retinal Neurons/pathology , Nerve Fibers/pathology , Peptide Fragments/blood
5.
Ann Clin Lab Sci ; 53(6): 905-913, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38182143

ABSTRACT

OBJECTIVE: This study aimed to investigate the role and mechanism of microRNA (miR)-193a in promoting apoptosis of retinal neuronal cells in early diabetic (DM) rats. METHODS: Seventy-two male SD-grade rats were selected to establish a DM model by intraperitoneal injection of streptozotocin (STZ), and randomly divided into a control group (blank control group), a DM group (diabetic model group), a DM+miR-NC inhibitor group (miR-193a inhibition negative control group), a DM+miR-193a inhibitor group (miR-193a inhibitor group), DM+miR-NC mimic group (miR-193a overexpression negative control group), DM+miR-193a mimic group (miR-193a overexpression group), with12 rats in each group. RESULTS: The miR-193a expression, apoptosis rate, and Bax, Caspase3, and Caspase9 protein expression levels were elevated, and Bcl-2 protein expression was decreased in the retinal tissues of DM rats and high glucose-induced rat retinal neuronal cells, while miR-193a inhibitors reversed these processes. These dual luciferase reporter assay showed that WT1CDS, and WT1Mut were lower in the miR-193a group than in the miR-NC group (P<0.05); WT1 protein expression was reduced in the retinal tissues of DM rat and high glucose-induced rat retinal neuronal cells, and miR-193a inhibitors increased WT1 protein expression. Compared with cells co-transfected with miR-193a and WT1vector, miR-193a and WT1 cotransfection inhibited high glucose-induced apoptosis in retinal neuronal cells and regulated apoptotic protein expression. miR-193a was highly expressed and WT1 was lowly expressed in retinal tissues of DM rats and high glucose-induced rat retinal neuronal cells. CONCLUSION: miR-193a could inhibit early retinal neuronal cell apoptosis in DM rats by targeting and negatively regulating WT1 expression.


Subject(s)
Apoptosis , Diabetes Mellitus , MicroRNAs , Retinal Neurons , Animals , Male , Rats , Apoptosis/genetics , Genes, Wilms Tumor , Glucose , MicroRNAs/genetics , WT1 Proteins , Retinal Neurons/metabolism
6.
Proc Natl Acad Sci U S A ; 120(52): e2315282120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38109525

ABSTRACT

Intrinsically photosensitive retinal ganglion cells (ipRGCs) serve as primary photoceptors by expressing the photopigment, melanopsin, and also as retinal relay neurons for rod and cone signals en route to the brain, in both cases for the purpose of non-image vision as well as aspects of image vision. So far, six subtypes of ipRGCs (M1 through M6) have been characterized. Regarding their phototransduction mechanisms, we have previously found that, unconventionally, rhabdomeric (microvillous) and ciliary signaling motifs co-exist within a given M1-, M2-, and M4-ipRGC, with the first mechanism involving PLCß4 and TRPC6,7 channels and the second involving cAMP and HCN channels. We have now examined M3-, M5-, and M6-cells and found that each cell likewise uses both signaling pathways for phototransduction, despite differences in the percentage representation by each pathway in a given ipRGC subtype for bright-flash responses (and saturated except for M6-cells). Generally, M3- and M5-cells show responses quite similar in kinetics to M2-responses, and M6-cell responses resemble broadly those of M1-cells although much lower in absolute sensitivity and amplitude. Therefore, similar to rod and cone subtypes in image vision, ipRGC subtypes possess the same phototransduction mechanism(s) even though they do not show microvilli or cilia morphologically.


Subject(s)
Retinal Neurons , Vision, Ocular , Light Signal Transduction/physiology , Retinal Ganglion Cells/physiology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Neurons/metabolism , Rod Opsins/metabolism
7.
Proc Natl Acad Sci U S A ; 120(42): e2307380120, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37831740

ABSTRACT

In patients blinded by geographic atrophy, a subretinal photovoltaic implant with 100 µm pixels provided visual acuity closely matching the pixel pitch. However, such flat bipolar pixels cannot be scaled below 75 µm, limiting the attainable visual acuity. This limitation can be overcome by shaping the electric field with 3-dimensional (3-D) electrodes. In particular, elevating the return electrode on top of the honeycomb-shaped vertical walls surrounding each pixel extends the electric field vertically and decouples its penetration into tissue from the pixel width. This approach relies on migration of the retinal cells into the honeycomb wells. Here, we demonstrate that majority of the inner retinal neurons migrate into the 25 µm deep wells, leaving the third-order neurons, such as amacrine and ganglion cells, outside. This enables selective stimulation of the second-order neurons inside the wells, thus preserving the intraretinal signal processing in prosthetic vision. Comparable glial response to that with flat implants suggests that migration and separation of the retinal cells by the walls does not cause additional stress. Furthermore, retinal migration into the honeycombs does not negatively affect its electrical excitability, while grating acuity matches the pixel pitch down to 40 µm and reaches the 27 µm limit of natural resolution in rats with 20 µm pixels. These findings pave the way for 3-D subretinal prostheses with pixel sizes of cellular dimensions.


Subject(s)
Porifera , Retinal Neurons , Visual Prosthesis , Humans , Rats , Animals , Prosthesis Implantation , Retina/physiology , Vision, Ocular , Electric Stimulation
8.
Stem Cell Reports ; 18(11): 2203-2221, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37802075

ABSTRACT

Intercellular cytoplasmic material transfer (MT) occurs between transplanted and developing photoreceptors and ambiguates cell origin identification in developmental, transdifferentiation, and transplantation experiments. Whether MT is a photoreceptor-specific phenomenon is unclear. Retinal ganglion cell (RGC) replacement, through transdifferentiation or transplantation, holds potential for restoring vision in optic neuropathies. During careful assessment for MT following human stem cell-derived RGC transplantation into mice, we identified RGC xenografts occasionally giving rise to labeling of donor-derived cytoplasmic, nuclear, and mitochondrial proteins within recipient Müller glia. Critically, nuclear organization is distinct between human and murine retinal neurons, which enables unequivocal discrimination of donor from host cells. MT was greatly facilitated by internal limiting membrane disruption, which also augments retinal engraftment following transplantation. Our findings demonstrate that retinal MT is not unique to photoreceptors and challenge the isolated use of species-specific immunofluorescent markers for xenotransplant identification. Assessment for MT is critical when analyzing neuronal replacement interventions.


Subject(s)
Retina , Retinal Neurons , Animals , Humans , Mice , Retina/metabolism , Retinal Ganglion Cells , Neuroglia/metabolism , Photoreceptor Cells
9.
Int J Mol Sci ; 24(17)2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37686457

ABSTRACT

The human eye plays a critical role in vision perception, but various retinal degenerative diseases such as retinitis pigmentosa (RP), glaucoma, and age-related macular degeneration (AMD) can lead to vision loss or blindness. Although progress has been made in understanding retinal development and in clinical research, current treatments remain inadequate for curing or reversing these degenerative conditions. Animal models have limited relevance to humans, and obtaining human eye tissue samples is challenging due to ethical and legal considerations. Consequently, researchers have turned to stem cell-based approaches, specifically induced pluripotent stem cells (iPSCs), to generate distinct retinal cell populations and develop cell replacement therapies. iPSCs offer a novel platform for studying the key stages of human retinogenesis and disease-specific mechanisms. Stem cell technology has facilitated the production of diverse retinal cell types, including retinal ganglion cells (RGCs) and photoreceptors, and the development of retinal organoids has emerged as a valuable in vitro tool for investigating retinal neuron differentiation and modeling retinal diseases. This review focuses on the protocols, culture conditions, and techniques employed in differentiating retinal neurons from iPSCs. Furthermore, it emphasizes the significance of molecular and functional validation of the differentiated cells.


Subject(s)
Induced Pluripotent Stem Cells , Retinal Degeneration , Retinal Neurons , Animals , Humans , Retina , Cell Differentiation , Retinal Degeneration/therapy , Blindness
10.
Invest Ophthalmol Vis Sci ; 64(12): 9, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37669061

ABSTRACT

Purpose: This study aimed to explore the neuroprotective effects of sigma-1 receptor (S1R) on optic nerve crush (ONC) mice by upregulating its expression through intravitreal injection of adeno-associated virus (AAV). Methods: The animals were divided into four groups. Mice that underwent ONC were administered an intravitreal injection with blank vector (ONC group), with AAV targeting downregulation of S1R (S1R-sh group), or with AAV targeting overexpression of S1R (S1R-AAV group). Mice in the control group underwent intravitreal injection with blank vector. The thickness of each layer of the retina was measured through optical coherence tomography, and the apoptotic rate of retinal neurons was determined using the TUNEL assay. The expression levels of brain-derived neurotrophic factor (BDNF) and S1R were quantified through western blot. Electroretinogram (ERG) was performed to evaluate the visual function. Results: The thickness of the total retina (P = 0.001), ganglion cell layer (P = 0.017), and inner nuclear layer (P = 0.002) in S1R-AAV group was significantly thicker than that of the ONC group. The number of retinal apoptotic cells in the S1R-AAV group was 23% lower than that in the ONC group (P = 0.002). ERG results showed that, compared to the ONC group, the amplitudes of the a- and b-waves were higher in the S1R-AAV group (a-wave, P < 0.001; b-wave, P = 0.007). Western blot showed that the expression of BDNF in the S1R-AAV group was higher than that in the ONC group (P < 0.001). Conclusions: Activation of S1R in the retina through intravitreal injection of AAV can effectively maintain the retina structure, promote neuronal cell survival, and protect visual function.


Subject(s)
Neuroprotective Agents , Retinal Neurons , Animals , Mice , Brain-Derived Neurotrophic Factor , Optic Nerve , Retina , Dependovirus , Sigma-1 Receptor
11.
Annu Rev Vis Sci ; 9: 131-153, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37713276

ABSTRACT

Rod and cone photoreceptors degenerate in retinitis pigmentosa and age-related macular degeneration, robbing the visual system of light-triggered signals necessary for sight. However, changes in the retina do not stop with the photoreceptors. A stereotypical set of morphological and physiological changes, known as remodeling, occur in downstream retinal neurons. Some aspects of remodeling are homeostatic, with structural or functional changes compensating for partial loss of visual inputs. However, other aspects are nonhomeostatic, corrupting retinal information processing to obscure vision mediated naturally by surviving photoreceptors or artificially by vision-restoration technologies. In this review, I consider the mechanism of remodeling and its consequences for residual and restored visual function; discuss the role of retinoic acid, a critical molecular trigger of detrimental remodeling; and discuss strategies for suppressing retinoic acid biosynthesis or signaling as therapeutic possibilities for mitigating vision loss.


Subject(s)
Retinal Neurons , Vision Disorders , Humans , Retina , Retinal Cone Photoreceptor Cells , Tretinoin
12.
Med ; 4(9): 583-590, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37689055

ABSTRACT

The translation of regenerative therapies to neuronal eye diseases requires a roadmap specific to the nature of the target diseases, patient population, methodologies for assessing outcome, and other factors. This commentary focuses on critical issues for translating regenerative eye therapies relevant to retinal neurons to human clinical trials.


Subject(s)
Eye Diseases , Retinal Neurons , Humans , Eye Diseases/therapy , Translations
13.
Invest Ophthalmol Vis Sci ; 64(11): 29, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37610761

ABSTRACT

Purpose: The isolated ex vivo retina is the standard model in retinal physiology and neuroscience. During isolation, the retina is peeled from the retinal pigment epithelium (RPE), which plays a key role in the visual cycle. Here we introduce the choroid-attached bovine retina as an in vivo-like model for retinal physiology. We find that-in the bovine eye-the choroid and retina can be peeled from the sclera as a single thin sheet. Importantly, the retina remains tightly associated with the RPE, which is sandwiched between the retina and the choroid. Furthermore, bovine tissue is readily available and cheap, and there are no ethical concerns related to the use of animals solely for research purposes. Methods: We combine multi-electrode array and single-cell patch-clamp recordings to characterize light responses in the choroid-attached bovine ex vivo retina. Results: We demonstrate robust and consistent light responses in choroid-attached preparations. Importantly, light responses adapt to different levels of background illumination and rapidly recover from photobleaching. The choroid-attached retina is also thin enough to permit targeted electrophysiological recording from individual retinal neurons using standard differential interference contrast microscopy. We also characterize light responses and membrane properties of bovine retinal ganglion cells and compare data obtained from bovine and murine retinas. Conclusions: The choroid-attached retinal model retains the advantages of the isolated retina but with an intact visual cycle and represents a useful tool to elucidate retinal physiology.


Subject(s)
Retina , Retinal Neurons , Cattle , Animals , Mice , Retinal Pigment Epithelium , Retinal Ganglion Cells , Choroid
14.
Life Sci Alliance ; 6(9)2023 09.
Article in English | MEDLINE | ID: mdl-37308277

ABSTRACT

The present study focused on the effect of miR-93-5p on apoptosis of retinal neurons in acute ocular hypertension (AOH) model by regulating PDCD4 and explored its related mechanism. We detected that miR-93-5p expression was decreased and PDCD4 expression was increased in the AOH retina by qRT-PCR. Therefore, we explored the role of miR-93-5p and PDCD4. MiR-93-5p overexpression inhibited the apoptosis of retinal neurons and the expression of PDCD4 in vivo and in vitro. Inhibiting the expression of PDCD4 via transfected interfering RNA decreased the apoptosis of retinal cells and increased the expression of PI3K/Akt pathway-related proteins in vitro. However, the addition of PI3K protein inhibitor LY294002 reversed this effect, leading to a decrease of PI3K/Akt pathway protein expression and an increase of apoptosis-related protein Bax/Bcl-2 expression ratio. Finally, up-regulating miR-93-5p or down-regulating PDCD4 increased the expression of PI3K/Akt pathway protein in vivo. In conclusion, under the condition of AOH injury, miR-93-5p-inhibiting PDCD4 expression reduced the apoptosis of retinal neurons by activating PI3K/Akt pathway.


Subject(s)
MicroRNAs , Ocular Hypertension , Retinal Neurons , Humans , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Apoptosis , RNA-Binding Proteins , Apoptosis Regulatory Proteins
15.
Annu Rev Vis Sci ; 9: 91-109, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37164027

ABSTRACT

Although diabetic retinopathy (DR) is clinically diagnosed as a vascular disease, many studies find retinal neuronal and visual dysfunction before the onset of vascular DR. This suggests that DR should be viewed as a neurovascular disease. Prior to the onset of DR, human patients have compromised electroretinograms that indicate a disruption of normal function, particularly in the inner retina. They also exhibit reduced contrast sensitivity. These early changes, especially those due to dysfunction in the inner retina, are also seen in rodent models of diabetes in the early stages of the disease. Rodent models of diabetes exhibit several neuronal mechanisms, such as reduced evoked GABA release, increased excitatory glutamate signaling, and reduced dopamine signaling, that suggest specific neuronal deficits. This suggests that understanding neuronal deficits may lead to early diabetes treatments to ameliorate neuronal dysfunction.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Retinal Neurons , Humans , Retina , Vision Disorders , Dopamine
16.
Cells ; 12(9)2023 05 08.
Article in English | MEDLINE | ID: mdl-37174742

ABSTRACT

Diabetic retinopathy (DR), the most common microvascular complication that occurs in diabetes mellitus (DM), is the leading cause of vision loss in working-age adults. The prevalence of diabetic retinopathy is approximately 30% of the diabetic population and untreated DR can eventually cause blindness. For decades, diabetic retinopathy was considered a microvascular complication and clinically staged by its vascular manifestations. In recent years, emerging evidence has shown that diabetic retinopathy causes early neuronal dysfunction and neurodegeneration that may precede vascular pathology and affect retinal neurons as well as glial cells. This knowledge leads to new therapeutic strategies aiming to prevent dysfunction of retinal neurons at the early stage of DR. Early detection and timely treatment to protect retinal neurons are critical to preventing visual loss in DR. This review provides an overview of DR and the structural and functional changes associated with DR, and discusses neuronal degeneration during diabetic retinopathy, the mechanisms underlying retinal neurodegeneration and microvascular complications, and perspectives on current and future clinic therapies.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Retinal Neurons , Humans , Diabetic Retinopathy/pathology , Retina/pathology , Retinal Neurons/pathology , Neuroglia/pathology , Vision Disorders
17.
Cell Rep ; 42(5): 112476, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37141093

ABSTRACT

The wiring of visual circuits requires that retinal neurons functionally connect to specific brain targets, a process that involves activity-dependent signaling between retinal axons and their postsynaptic targets. Vision loss in various ophthalmological and neurological diseases is caused by damage to the connections from the eye to the brain. How postsynaptic brain targets influence retinal ganglion cell (RGC) axon regeneration and functional reconnection with the brain targets remains poorly understood. Here, we established a paradigm in which the enhancement of neural activity in the distal optic pathway, where the postsynaptic visual target neurons reside, promotes RGC axon regeneration and target reinnervation and leads to the rescue of optomotor function. Furthermore, selective activation of retinorecipient neuron subsets is sufficient to promote RGC axon regeneration. Our findings reveal a key role for postsynaptic neuronal activity in the repair of neural circuits and highlight the potential to restore damaged sensory inputs via proper brain stimulation.


Subject(s)
Axons , Retinal Neurons , Axons/metabolism , Nerve Regeneration/physiology , Retina/metabolism , Retinal Ganglion Cells/metabolism
18.
Cogn Sci ; 47(4): e13265, 2023 04.
Article in English | MEDLINE | ID: mdl-37029513

ABSTRACT

How does neuronal activity give rise to cognitive capacities? To address this question, neuroscientists hypothesize about what neurons "represent," "encode," or "compute," and test these hypotheses empirically. This process is similar to the assessment of hypotheses in other fields of science and as such is subject to the same limitations and difficulties that have been discussed at length by philosophers of science. In this paper, we highlight an additional difficulty in the process of empirical assessment of hypotheses that is unique to the cognitive sciences. We argue that, unlike in other scientific fields, comparing hypotheses according to the extent to which they explain or predict empirical data can lead to absurd results. Other considerations, which are perhaps more subjective, must be taken into account. We focus on one such consideration, which is the purposeful function of the neurons as part of a biological system. We believe that progress in neuroscience critically depends on properly addressing this difficulty.


Subject(s)
Cognition , Retinal Neurons , Humans , Retinal Neurons/physiology
19.
Biochem Biophys Res Commun ; 663: 87-95, 2023 06 30.
Article in English | MEDLINE | ID: mdl-37119770

ABSTRACT

miR-183/96/182 cluster plays a critical role in the developing retina by regulating many target genes involved in signaling pathways. This study aimed to survey the miR-183/96/182 cluster-target interactions that, potentially contribute to human retinal pigmented epithelial (hRPE) cell differentiation into photoreceptors. Target genes of the miR-183/96/182 cluster were obtained from miRNA-target databases and applied to construct miRNA-target networks. Gene ontology and KEGG pathway analysis was performed. miR-183/96/182 cluster sequence was cloned into an eGFP-intron splicing cassette in an AAV2 vector and overexpressed in hRPE cells. The expression level of target genes including HES1, PAX6, SOX2, CCNJ, and RORΒ was evaluated using qPCR. Our results showed that miR-183, miR-96, and miR-182 share 136 target genes that are involved in cell proliferation pathways such as PI3K/AKT and MAPK pathway. qPCR data indicated a 22-, 7-, and 4-fold overexpression of miR-183, miR-96, and miR-182, respectively, in infected hRPE cells. Consequently, the downregulation of several key targets such as PAX6, CCND2, CDK5R1, and CCNJ and upregulation of a few retina-specific neural markers such as Rhodopsin, red opsin, and CRX was detected. Our findings suggest that the miR-183/96/182 cluster may induce hRPE transdifferentiation by targeting key genes that involve in the cell cycle and proliferation pathways.


Subject(s)
MicroRNAs , Retinal Neurons , Humans , Cell Transdifferentiation/genetics , Phosphatidylinositol 3-Kinases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Retinal Neurons/metabolism , Epithelial Cells/metabolism , Retinal Pigments/metabolism
20.
Redox Biol ; 63: 102713, 2023 07.
Article in English | MEDLINE | ID: mdl-37120931

ABSTRACT

Glaucoma is the leading cause of blindness worldwide. However, our insufficient understanding of the pathogenesis of glaucoma has limited the development of effective treatments. Because recent research has highlighted the importance of non-coding RNAs (ncRNAs) in various diseases, we investigated their roles in glaucoma. Specifically, we detected expression changes of ncRNAs in cell and animal models of acute glaucoma. Further analysis revealed that the Ier2/miR-1839/TSPO axis was critical to cell loss and retinal damage. The knockdown of Ier2, the overexpression of miR-1839, and the silencing of TSPO effectively prevented retinal damage and cell loss. Furthermore, we found that the Ier2/miR-1839/TSPO axis regulated the pyroptosis and apoptosis of retinal neurons through the NLRP3/caspase1/GSDMD, cleaved-caspase3 pathways. In addition to high expression in the retina, TSPO expression was found to be significantly higher in the dorsal lateral geniculate nucleus (DLG) of the brain in the pathologically high intraocular pressure (ph-IOP) rat model, as well as in the peripheral blood mononuclear cells (PBMCs) of glaucoma patients with high IOP. These results indicate that TSPO, which is regulated by Ier2/miR-1839, plays an important role in the pathogenesis of glaucoma, and this study provides a theoretical basis and a new target for the diagnosis and treatment of glaucoma.


Subject(s)
Glaucoma , MicroRNAs , Retinal Neurons , Rats , Animals , Retinal Ganglion Cells/metabolism , Pyroptosis/genetics , Leukocytes, Mononuclear/metabolism , Glaucoma/genetics , Retina/metabolism , Apoptosis/genetics , Carrier Proteins/metabolism , Retinal Neurons/metabolism , Retinal Neurons/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL
...