Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.161
Filter
1.
Invest Ophthalmol Vis Sci ; 65(11): 8, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39230992

ABSTRACT

Purpose: This study investigates alterations in intrinsically photosensitive retinal ganglion cells (ipRGCs) and dopaminergic amacrine cells (DACs) in lid suture myopia (LSM) rats. Methods: LSM was induced in rats by suturing the right eyes for 4 weeks. Double immunofluorescence staining of ipRGCs and DACs in whole-mount retinas was performed to analyze changes in the density and morphology of control, LSM, and fellow eyes. Real-time quantitative PCR and Western blotting were used to detect related genes and protein expression levels. Results: Significant myopia was induced in the lid-sutured eye, but the fellow eye was not different to control. Decreased ipRGC density with paradoxically increased overall melanopsin expression and enlarged dendritic beads was observed in both the LSM and fellow eyes of the LSM rat retinas. In contrast, DAC changes occurred only in the LSM eyes, with reduced DAC density and tyrosine hydroxylase (TH) expression, sparser dendritic processes, and fewer varicosities. Interestingly, contacts between ipRGCs and DACs in the inner plexiform layer (IPL) and the expression of pituitary adenylate cyclase-activating polypeptide (PACAP) and vesicular monoamine transporter protein 2 (VMAT2) mRNA were decreased in the LSM eyes. Conclusions: The ipRGCs and DACs in LSM rat retinas undergo multiple alterations in density, morphology, and related molecule expressions. However, the ipRGC changes alone appear not to be required for the development of myopia, given that myopia is only induced in the lid-sutured eye, and they are unlikely alone to drive the DAC changes. Reduced contacts between ipRGCs and DACs in the LSM eyes may be the structural foundation for the impaired signaling between them. PACAP and VMAT2, strongly associated with ipRGCs and DACs, may play important roles in LSM through complex mechanisms.


Subject(s)
Amacrine Cells , Blotting, Western , Disease Models, Animal , Myopia , Retinal Ganglion Cells , Rod Opsins , Animals , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Rats , Myopia/metabolism , Amacrine Cells/metabolism , Amacrine Cells/pathology , Rod Opsins/metabolism , Tyrosine 3-Monooxygenase/metabolism , Real-Time Polymerase Chain Reaction , Vesicular Monoamine Transport Proteins/metabolism , Vesicular Monoamine Transport Proteins/genetics , Male , Rats, Sprague-Dawley , Eyelids/pathology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Cell Count , Vesicular Glutamate Transport Protein 2
2.
Cell Rep ; 43(8): 114615, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39133615

ABSTRACT

In vertebrate retina, individual neurons of the same type are distributed regularly across the tissue in a pattern known as a mosaic. Establishment of mosaics during development requires cell-cell repulsion among homotypic neurons, but the mechanisms underlying this repulsion remain unknown. Here, we show that two mouse retinal cell types, OFF and ON starburst amacrine cells, establish mosaic spacing by using their dendritic arbors to repel neighboring homotypic somata. Using transgenic tools and single-cell labeling, we identify a developmental period when starburst somata are contacted by neighboring starburst dendrites; these serve to exclude somata from settling within the neighbor's dendritic territory. Dendrite-soma exclusion is mediated by MEGF10, a cell-surface molecule required for starburst mosaic patterning. Our results implicate dendrite-soma exclusion as a key mechanism underlying starburst mosaic spacing and raise the possibility that this could be a general mechanism for mosaic patterning across many cell types and species.


Subject(s)
Dendrites , Animals , Dendrites/metabolism , Mice , Amacrine Cells/metabolism , Amacrine Cells/cytology , Retina/cytology , Retina/metabolism , Mosaicism , Retinal Neurons/cytology , Retinal Neurons/metabolism , Mice, Transgenic , Mice, Inbred C57BL
3.
ACS Appl Mater Interfaces ; 16(35): 46454-46460, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39169757

ABSTRACT

Event-based imaging represents a new paradigm in visual information processing that addresses the speed and energy efficiency shortcomings inherently present in the current complementary metal oxide semiconductor-based machine vision. Realizing such imaging systems has previously been sought using very large-scale integration technologies that have complex circuitries consisting of many photodiodes, differential amplifiers, capacitors, and resistors. Here, we demonstrate that event-driven sensing can be achieved using a simple one-resistor, one-capacitor (1R1C) circuit, where the capacitor is modified with colloidal quantum dots (CQDs) to have a photoresponse. This sensory circuit emulates the motion-tracking function of the biological retina, in which the amacrine cells in the bipolar-to-ganglion synaptic pathway produce a transient spiking signal only in response to changes in light intensity but remain inactive under constant illumination. When extended to a 2D imaging array, the individual sensors work independently and output signals only when a change in the light intensity is detected; hence, the concept of the frame in image processing is thereby removed. In this work, we present the fabrication and characterization of a CQD photocapacitor-based 1R1C circuit that has a spectral response at 1550 nm in the short-wave infrared (SWIR). We report on the key performance parameters including peak responsivity, noise, and optical noise equivalent power and discuss the operating mechanism that is responsible for spiking responses in these artificial retinal circuits. The present work sets the foundation for expanding the bioinspired vision sensor capability toward midwave infrared (MWIR) and long-wave infrared (LWIR) spectral regions that are invisible to human eyes and mainstream semiconductor technologies.


Subject(s)
Amacrine Cells , Quantum Dots , Amacrine Cells/physiology , Quantum Dots/chemistry , Retina/physiology , Semiconductors , Humans
4.
Curr Biol ; 34(18): 4224-4239.e4, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39214087

ABSTRACT

Neurons form cell-type-specific morphologies that are shaped by cell-surface molecules and their cellular events governing dendrite growth. One growth rule is dendrite self-avoidance, whereby dendrites distribute uniformly within a neuron's territory by avoiding sibling branches. In mammalian neurons, dendrite self-avoidance is regulated by a large family of cell-recognition molecules called the clustered protocadherins (cPcdhs). Genetic and molecular studies suggest that the cPcdhs mediate homophilic recognition and repulsion between self-dendrites. However, this model has not been tested through direct investigation of self-avoidance during development. Here, we performed live imaging and four-dimensional (4D) quantifications of dendrite morphogenesis to define the dynamics and cPcdh-dependent mechanisms of self-avoidance. We focused on the mouse retinal starburst amacrine cell (SAC), which requires the gamma-Pcdhs (Pcdhgs) and self/non-self-recognition to establish a stereotypic radial morphology while permitting dendritic interactions with neighboring SACs. Through morphogenesis, SACs extend dendritic protrusions that iteratively fill the growing arbor and contact and retract from nearby self-dendrites. Compared to non-self-contacting protrusions, self-contacting events have longer lifetimes, and a subset persists as loops. In the absence of the Pcdhgs, non-self-contacting dynamics are unaffected but self-contacting retractions are significantly diminished. Self-contacting bridges accumulate, leading to the bundling of dendritic processes and disruption to the arbor shape. By tracking dendrite self-avoidance in real time, our findings establish that the γ-Pcdhs mediate self-recognition and retraction between contacting sibling dendrites. Our results also illustrate how self-avoidance shapes stochastic and space-filling dendritic outgrowth for robust pattern formation in mammalian neurons.


Subject(s)
Amacrine Cells , Cadherin Related Proteins , Cadherins , Dendrites , Animals , Dendrites/physiology , Dendrites/metabolism , Mice , Cadherins/metabolism , Cadherins/genetics , Amacrine Cells/metabolism , Amacrine Cells/physiology , Retina/metabolism , Morphogenesis
5.
Sci Adv ; 10(31): eado0866, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093964

ABSTRACT

As part of the central nervous system, the optic nerve, composed of axons from retinal ganglion cells (RGCs), generally fails to regenerate on its own when injured in adult mammals. An innovative approach to promoting optic nerve regeneration involves manipulating the interactions between amacrine cells (ACs) and RGCs. Here, we identified a unique AC subtype, dopaminergic ACs (DACs), that responded early after optic nerve crush by down-regulating neuronal activity and reducing retinal dopamine (DA) release. Activating DACs or augmenting DA release with levodopa demonstrated neuroprotective effects and modestly enhanced axon regeneration. Within this context, we pinpointed the DA receptor D1 (DRD1) as a critical mediator of DAC-derived DA and showed that RGC-specific Drd1 overexpression effectively overcame subtype-specific barriers to regeneration. This strategy markedly boosted RGC survival and axon regeneration after crush and preserved vision in a glaucoma model. This study unveils the crucial role of DAC-derived DA signaling in optic nerve regeneration, holding promise for therapeutic insights into neural repair.


Subject(s)
Amacrine Cells , Dopamine , Nerve Regeneration , Optic Nerve , Retinal Ganglion Cells , Signal Transduction , Animals , Amacrine Cells/metabolism , Dopamine/metabolism , Nerve Regeneration/drug effects , Optic Nerve/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/drug effects , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/pathology , Mice , Axons/metabolism , Axons/physiology , Receptors, Dopamine D1/metabolism , Vision, Ocular/physiology , Disease Models, Animal
6.
Am J Physiol Cell Physiol ; 327(3): C716-C727, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39010839

ABSTRACT

Gap junctions are channels that allow for direct transmission of electrical signals between cells. However, the ability of one cell to be impacted or controlled by other cells through gap junctions remains unclear. In this study, heterocellular coupling between ON α retinal ganglion cells (α-RGCs) and displaced amacrine cells (ACs) in the mouse retina was used as a model. The impact of the extent of coupling of interconnected ACs on the synchronized firing between coupled ON α-RGC-AC pair was investigated using the dopamine 1 receptor (D1R) antagonist-SCH23390 and agonist-SKF38393. It was observed that the synchronized firing between the ON α-RGC-ACs pairs was increased by the D1R antagonist SCH23390, whereas it was eradicated by the agonist SKF38393. Subsequently, the signaling drive was investigated by infecting coupled ON α-RGC-AC pairs with the channelrhodopsin-2(ChR2) mutation L132C engineered to enhance light sensitivities. The results demonstrated that the spikes of ON α-RGCs (without ChR2) could be triggered by ACs (with ChR2) through the gap junction, and vice versa. Furthermore, it was observed that ON α-RGCs stimulated with 3-10 Hz currents by whole cell patch could elicit synchronous spikes in the coupled ACs, and vice versa. This provided direct evidence that the firing of one cell could be influenced by another cell through gap junctions. However, this phenomenon was not observed between OFF α-RGC pairs. The study implied that the synchronized firing between ON α-RGC-AC pairs could potentially be affected by the coupling of interconnected ACs. Additionally, one cell type could selectively control the firing of another cell type, thereby forcefully transmitting information. The key role of gap junctions in synchronizing firing and driving cells between α-RGCs and coupled ACs in the mouse retina was highlighted.NEW & NOTEWORTHY This study investigates the role of gap junctions in transmitting electrical signals between cells and their potential for cell control. Using ON α retinal ganglion cells (α-RGCs) and amacrine cells (ACs) in the mouse retina, the researchers find that the extent of coupling between ACs affects synchronized firing. Bidirectional signaling occurs between ACs and ON α-RGCs through gap junctions.


Subject(s)
Action Potentials , Amacrine Cells , Gap Junctions , Retinal Ganglion Cells , Animals , Amacrine Cells/metabolism , Amacrine Cells/physiology , Amacrine Cells/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/physiology , Mice , Gap Junctions/metabolism , Gap Junctions/drug effects , Action Potentials/drug effects , Mice, Inbred C57BL , Retina/metabolism , Retina/physiology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/agonists , Cell Communication/physiology , Male , Channelrhodopsins/metabolism , Channelrhodopsins/genetics
7.
J Gen Physiol ; 156(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38836782

ABSTRACT

Cholinergic signaling in the retina is mediated by acetylcholine (ACh) released from starburst amacrine cells (SACs), which are key neurons for motion detection. SACs comprise ON and OFF subtypes, which morphologically show mirror symmetry to each other. Although many physiological studies on SACs have targeted ON cells only, the synaptic computation of ON and OFF SACs is assumed to be similar. Recent studies demonstrated that gene expression patterns and receptor types differed between ON and OFF SACs, suggesting differences in their functions. Here, we compared cholinergic signaling pathways between ON and OFF SACs in the mouse retina using the patch clamp technique. The application of ACh increased GABAergic feedback, observed as postsynaptic currents to SACs, in both ON and OFF SACs; however, the mode of GABAergic feedback differed. Nicotinic receptors mediated GABAergic feedback in both ON and OFF SACs, while muscarinic receptors mediated GABAergic feedback in ON SACs only in adults. Neither tetrodotoxin, which blocked action potentials, nor LY354740, which blocked neurotransmitter release from SACs, eliminated ACh-induced GABAergic feedback in SACs. These results suggest that ACh-induced GABAergic feedback in ON and OFF SACs is regulated by different feedback mechanisms in adults and mediated by non-spiking amacrine cells other than SACs.


Subject(s)
Acetylcholine , Amacrine Cells , Animals , Amacrine Cells/metabolism , Mice , Acetylcholine/pharmacology , Acetylcholine/metabolism , Mice, Inbred C57BL , gamma-Aminobutyric Acid/metabolism , Receptors, Muscarinic/metabolism , Receptors, Nicotinic/metabolism
8.
Exp Eye Res ; 245: 109985, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38945518

ABSTRACT

Aging is a major risk factor for the development or the worsening of retinal degenerative conditions. The intricate network of the neural retina determined that the retinal aging is a complicated process. The aim of this study is to delineate the transcriptomic changes of major retinal neurons during aging in C57BL/6 mice at single-cell level. We analyzed the transcriptional profiles of the photoreceptor, bipolar, amacrine, and Müller glial cells of 1.5-2 and 24-30 months old mice using single-cell RNA sequencing technique. We selectively confirmed the differences in gene expression using immunofluorescence staining and RNA in situ hybridization analysis. We found that each retinal cell type had unique changes upon aging. However, they all showed signs of dysregulated glucose and energy metabolism, and perturbed proteostasis. In particular, old Müller glia exhibited the most profound changes, including the upregulation of cell metabolism, stress-responses, antigen-presentation and immune responses and metal ion homeostasis. The dysregulated gliogenesis and differentiation was confirmed by the presence of Müller glia expressing rod-specific genes in the inner nuclear layer and the outer plexiform layer of the old retina. We further pinpointed the specific loss of GABAergic amacrine cells in old retina. Our study emphasized changes of amacrine and Müller glia during retinal aging, provided resources for further research on the molecular and cellular regulatory mechanisms underlying aging-associated retinal deterioration.


Subject(s)
Aging , Amacrine Cells , Energy Metabolism , Mice, Inbred C57BL , Proteostasis , Animals , Amacrine Cells/metabolism , Mice , Aging/physiology , Energy Metabolism/physiology , Ependymoglial Cells/metabolism , Retina/metabolism , GABAergic Neurons/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/genetics , In Situ Hybridization , Homeostasis/physiology
9.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38719453

ABSTRACT

Retinal prosthetics are one of the leading therapeutic strategies to restore lost vision in patients with retinitis pigmentosa and age-related macular degeneration. Much work has described patterns of spiking in retinal ganglion cells (RGCs) in response to electrical stimulation, but less work has examined the underlying retinal circuitry that is activated by electrical stimulation to drive these responses. Surprisingly, little is known about the role of inhibition in generating electrical responses or how inhibition might be altered during degeneration. Using whole-cell voltage-clamp recordings during subretinal electrical stimulation in the rd10 and wild-type (wt) retina, we found electrically evoked synaptic inputs differed between ON and OFF RGC populations, with ON cells receiving mostly excitation and OFF cells receiving mostly inhibition and very little excitation. We found that the inhibition of OFF bipolar cells limits excitation in OFF RGCs, and a majority of both pre- and postsynaptic inhibition in the OFF pathway arises from glycinergic amacrine cells, and the stimulation of the ON pathway contributes to inhibitory inputs to the RGC. We also show that this presynaptic inhibition in the OFF pathway is greater in the rd10 retina, compared with that in the wt retina.


Subject(s)
Electric Stimulation , Retinal Ganglion Cells , Animals , Retinal Ganglion Cells/physiology , Retinal Degeneration/physiopathology , Mice, Inbred C57BL , Retinal Bipolar Cells/physiology , Patch-Clamp Techniques , Visual Pathways/physiology , Visual Pathways/physiopathology , Neural Inhibition/physiology , Female , Male , Retina/physiology , Amacrine Cells/physiology
10.
Brain Struct Funct ; 229(5): 1279-1298, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703218

ABSTRACT

ß-synuclein, a member of the synuclein family, is frequently co-expressed with α-synuclein in the neural system, where it serves to inhibit abnormal aggregation of α-synuclein in neurodegenerative diseases. Beyond its role in pathological conditions, ß-synuclein plays various functions independently of α-synuclein. In our investigation, we discovered a broader expression of ß-synuclein in the mouse retina compared to α-synuclein. This widespread pattern implies its potential significance in the retina. Through detailed examination via light- and electron-microscopic immunocytochemistry, we identified ß-synuclein expression from the inner segment (IS) and outer segment (OS) of photoreceptor cells to the ganglion cell layer (GCL). Our findings unveiled unique features, including ß-synuclein immunoreactive IS and OS of cones, higher expression in cone pedicles than in rod spherules, absence in horizontal cells, limited expression in cone bipolar dendrites and somas, higher expression in cone bipolar terminals, presence in most amacrine cells, and expression in almost majority of somas in GCL with an absence in intrinsically photosensitive retinal ganglion cell (ipRGCs) processes. Notably, all cholinergic amacrine cells express high ß- but not α-synuclein, while dopaminergic amacrine cells express α-synuclein exclusively. These distinctive expression patterns offer valuable insights for further exploration into the functions of ß-synuclein and its potential role in synuclein pathology within the retina.


Subject(s)
Mice, Inbred C57BL , Retina , Retinal Ganglion Cells , alpha-Synuclein , beta-Synuclein , Animals , Male , Mice , alpha-Synuclein/metabolism , Amacrine Cells/metabolism , beta-Synuclein/metabolism , Retina/metabolism , Retinal Bipolar Cells/metabolism , Retinal Ganglion Cells/metabolism
11.
Neural Comput ; 36(6): 1041-1083, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38669693

ABSTRACT

We consider a model of basic inner retinal connectivity where bipolar and amacrine cells interconnect and both cell types project onto ganglion cells, modulating their response output to the brain visual areas. We derive an analytical formula for the spatiotemporal response of retinal ganglion cells to stimuli, taking into account the effects of amacrine cells inhibition. This analysis reveals two important functional parameters of the network: (1) the intensity of the interactions between bipolar and amacrine cells and (2) the characteristic timescale of these responses. Both parameters have a profound combined impact on the spatiotemporal features of retinal ganglion cells' responses to light. The validity of the model is confirmed by faithfully reproducing pharmacogenetic experimental results obtained by stimulating excitatory DREADDs (Designer Receptors Exclusively Activated by Designer Drugs) expressed on ganglion cells and amacrine cells' subclasses, thereby modifying the inner retinal network activity to visual stimuli in a complex, entangled manner. Our mathematical model allows us to explore and decipher these complex effects in a manner that would not be feasible experimentally and provides novel insights in retinal dynamics.


Subject(s)
Retina , Retinal Ganglion Cells , Retinal Ganglion Cells/physiology , Retina/physiology , Animals , Models, Neurological , Amacrine Cells/physiology , Computer Simulation , Humans , Visual Pathways/physiology , Photic Stimulation/methods , Nerve Net/physiology , Visual Fields/physiology , Retinal Bipolar Cells/physiology
12.
Nat Commun ; 15(1): 2965, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38580652

ABSTRACT

VGluT3-expressing mouse retinal amacrine cells (VG3s) respond to small-object motion and connect to multiple types of bipolar cells (inputs) and retinal ganglion cells (RGCs, outputs). Because these input and output connections are intermixed on the same dendrites, making sense of VG3 circuitry requires comparing the distribution of synapses across their arbors to the subcellular flow of signals. Here, we combine subcellular calcium imaging and electron microscopic connectomic reconstruction to analyze how VG3s integrate and transmit visual information. VG3s receive inputs from all nearby bipolar cell types but exhibit a strong preference for the fast type 3a bipolar cells. By comparing input distributions to VG3 dendrite responses, we show that VG3 dendrites have a short functional length constant that likely depends on inhibitory shunting. This model predicts that RGCs that extend dendrites into the middle layers of the inner plexiform encounter VG3 dendrites whose responses vary according to the local bipolar cell response type.


Subject(s)
Amacrine Cells , Retina , Mice , Animals , Amacrine Cells/physiology , Retina/physiology , Retinal Ganglion Cells/physiology , Synapses/metabolism , Microscopy, Electron , Dendrites/physiology
13.
J Neurosci ; 44(18)2024 May 01.
Article in English | MEDLINE | ID: mdl-38514178

ABSTRACT

An organizational feature of neural circuits is the specificity of synaptic connections. A striking example is the direction-selective (DS) circuit of the retina. There are multiple subtypes of DS retinal ganglion cells (DSGCs) that prefer motion along one of four preferred directions. This computation is mediated by selective wiring of a single inhibitory interneuron, the starburst amacrine cell (SAC), with each DSGC subtype preferentially receiving input from a subset of SAC processes. We hypothesize that the molecular basis of this wiring is mediated in part by unique expression profiles of DSGC subtypes. To test this, we first performed paired recordings from isolated mouse retinas of both sexes to determine that postnatal day 10 (P10) represents the age at which asymmetric synapses form. Second, we performed RNA sequencing and differential expression analysis on isolated P10 ON-OFF DSGCs tuned for either nasal or ventral motion and identified candidates which may promote direction-specific wiring. We then used a conditional knock-out strategy to test the role of one candidate, the secreted synaptic organizer cerebellin-4 (Cbln4), in the development of DS tuning. Using two-photon calcium imaging, we observed a small deficit in directional tuning among ventral-preferring DSGCs lacking Cbln4, though whole-cell voltage-clamp recordings did not identify a significant change in inhibitory inputs. This suggests that Cbln4 does not function primarily via a cell-autonomous mechanism to instruct wiring of DS circuits. Nevertheless, our transcriptomic analysis identified unique candidate factors for gaining insights into the molecular mechanisms that instruct wiring specificity in the DS circuit.


Subject(s)
Mice, Inbred C57BL , Retina , Retinal Ganglion Cells , Synapses , Animals , Mice , Retina/metabolism , Retina/physiology , Male , Synapses/physiology , Synapses/metabolism , Female , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/physiology , Amacrine Cells/physiology , Amacrine Cells/metabolism , Motion Perception/physiology , Nerve Net/physiology , Nerve Net/metabolism , Visual Pathways/physiology , Visual Pathways/metabolism
14.
Cell Rep ; 43(4): 114005, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38551961

ABSTRACT

The retina is exquisitely patterned, with neuronal somata positioned at regular intervals to completely sample the visual field. Here, we show that phosphatase and tensin homolog (Pten) controls starburst amacrine cell spacing by modulating vesicular trafficking of cell adhesion molecules and Wnt proteins. Single-cell transcriptomics and double-mutant analyses revealed that Pten and Down syndrome cell adhesion molecule Dscam) are co-expressed and function additively to pattern starburst amacrine cell mosaics. Mechanistically, Pten loss accelerates the endocytic trafficking of DSCAM, FAT3, and MEGF10 off the cell membrane and into endocytic vesicles in amacrine cells. Accordingly, the vesicular proteome, a molecular signature of the cell of origin, is enriched in exocytosis, vesicle-mediated transport, and receptor internalization proteins in Pten conditional knockout (PtencKO) retinas. Wnt signaling molecules are also enriched in PtencKO retinal vesicles, and the genetic or pharmacological disruption of Wnt signaling phenocopies amacrine cell patterning defects. Pten thus controls vesicular trafficking of cell adhesion and signaling molecules to establish retinal amacrine cell mosaics.


Subject(s)
Amacrine Cells , Cell Adhesion , Endocytosis , PTEN Phosphohydrolase , Retina , Wnt Signaling Pathway , Animals , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Retina/metabolism , Mice , Amacrine Cells/metabolism , Mice, Knockout , Protein Transport , Wnt Proteins/metabolism , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics
15.
Int J Mol Sci ; 25(5)2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38474095

ABSTRACT

We recently identified PKN1 as a developmentally active gatekeeper of the transcription factor neuronal differentiation-2 (NeuroD2) in several brain areas. Since NeuroD2 plays an important role in amacrine cell (AC) and retinal ganglion cell (RGC) type formation, we aimed to study the expression of NeuroD2 in the postnatal retina of WT and Pkn1-/- animals, with a particular focus on these two cell types. We show that PKN1 is broadly expressed in the retina and that the gross retinal structure is not different between both genotypes. Postnatal retinal NeuroD2 levels were elevated upon Pkn1 knockout, with Pkn1-/- retinae showing more NeuroD2+ cells in the lower portion of the inner nuclear layer. Accordingly, immunohistochemical analysis revealed an increased amount of AC in postnatal and adult Pkn1-/- retinae. There were no differences in horizontal cell, bipolar cell, glial cell and RGC numbers, nor defective axon guidance to the optic chiasm or tract upon Pkn1 knockout. Interestingly, we did, however, see a specific reduction in SMI-32+ α-RGC in Pkn1-/- retinae. These results suggest that PKN1 is important for retinal cell type formation and validate PKN1 for future studies focusing on AC and α-RGC specification and development.


Subject(s)
Retina , Retinal Ganglion Cells , Animals , Retina/metabolism , Retinal Ganglion Cells/metabolism , Amacrine Cells/metabolism , Optic Chiasm/metabolism , Transcription Factors/metabolism
16.
J Fish Biol ; 104(5): 1299-1307, 2024 May.
Article in English | MEDLINE | ID: mdl-38308449

ABSTRACT

We studied the topography of retinal ganglion cells (GCs) and estimated spatial resolving power (SRP) in the pajama cardinalfish Sphaeramia nematoptera (Bleeker, 1856), a relatively small brightly colored fish inhabiting coral reefs and lagoons in the Western Pacific. S. nematoptera is an active night predator feeding on near-bottom animal plankton and benthos. DAPI staining was used to label nuclei of GCs and non-GCs in the inner plexiform and ganglion cell layers. Non-GCs were distinguished from GCs in Nissl-stained retinal wholemounts based on cell size, shape, and staining intensity. The proportion of displaced amacrine cells (DACs) varied from 15.46 ± 1.12 (visual streak [VS]) to 17.99 ± 1.06% (dorsal periphery) (mean ± S.E.M., N = 5); the respective proportions of glial cells were 6.61 ± 0.84 and 5.89 ± 0.76%. Thus, 76%-78% of cells in the ganglion cell layer and inner plexiform layer were GCs. The minimum spatial coverage of GCs (3600-4600 cells/mm2) was detected in the dorsal and ventral periphery. It gradually increased toward the central retina to form a moderate VS. The maximum GC density (11,400-12,400 cells/mm2) was registered in the central portion of the VS. No pronounced concentric retinal specializations were found. The total number of GCs ranged within 595.2-635.9 × 103. The anatomical spatial resolving power was minimum in the ventral periphery (4.91-5.53 cpd) and maximum in the central portion of the VS (8.47-9.07 cpd). The respective minimum separable angles were 0.18-0.20° and 0.11-0.12°. The relatively high spatial resolving power and presence of the VS in the pajama cardinalfish are in line with its highly visual behavior.


Subject(s)
Retinal Ganglion Cells , Animals , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology , Amacrine Cells/physiology , Amacrine Cells/cytology
17.
PLoS Biol ; 22(2): e3002506, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38363811

ABSTRACT

In mammals, retinal direction selectivity originates from GABAergic/cholinergic amacrine cells (ACs) specifically expressing the sox2 gene. However, the cellular diversity of GABAergic/cholinergic ACs of other vertebrate species remains largely unexplored. Here, we identified 2 morphologically and genetically distinct GABAergic/cholinergic AC types in zebrafish, a previously undescribed bhlhe22+ type and a mammalian counterpart sox2+ type. Notably, while sole sox2 disruption removed sox2+ type, the codisruption of bhlhe22 and bhlhe23 was required to remove bhlhe22+ type. Also, both types significantly differed in dendritic arbors, lamination, and soma position. Furthermore, in vivo two-photon calcium imaging and the behavior assay suggested the direction selectivity of both AC types. Nevertheless, the 2 types showed preferential responses to moving bars of different sizes. Thus, our findings provide new cellular diversity and functional characteristics of GABAergic/cholinergic ACs in the vertebrate retina.


Subject(s)
Amacrine Cells , Zebrafish , Animals , Amacrine Cells/metabolism , Retina/metabolism , Cholinergic Agents/metabolism , Transcription Factors/metabolism , Mammals
18.
Cell Mol Neurobiol ; 44(1): 19, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38315298

ABSTRACT

Retinal vasoactive intestinal peptide amacrine cells (VIP-ACs) play an important role in various retinal light-mediated pathological processes related to different developmental ocular diseases and even mental disorders. It is important to characterize the developmental changes in VIP-ACs to further elucidate their mechanisms of circuit function. We bred VIP-Cre mice with Ai14 and Ai32 to specifically label retinal VIP-ACs. The VIP-AC soma and spine density generally increased, from postnatal day (P)0 to P35, reaching adult levels at P14 and P28, respectively. The VIP-AC soma density curve was different with the VIP-AC spine density curve. The total retinal VIP content reached a high level plateau at P14 but was decreased in adults. From P14 to P16, the resting membrane potential (RMP) became more negative, and the input resistance decreased. Cell membrane capacitance (MC) showed three peaks at P7, P12 and P16. The RMP and MC reached a stable level similar to the adult level at P18, whereas input resistance reached a stable level at P21. The percentage of sustained voltage-dependent potassium currents peaked at P16 and remained stable thereafter. The spontaneous excitatory postsynaptic current and spontaneous inhibitory postsynaptic current frequencies and amplitudes, as well as charge transfer, peaked at P12 to P16; however, there were also secondary peaks at different time points. In conclusion, we found that the second, third and fourth weeks after birth were important periods of VIP-AC development. Many developmental changes occurred around eye opening. The development of soma, dendrite and electrophysiological properties showed uneven dynamics of progression. Cell differentiation may contribute to soma development whereas the changes of different ion channels may play important role for spine development.


Subject(s)
Amacrine Cells , Vasoactive Intestinal Peptide , Animals , Mice , Cell Differentiation , Membrane Potentials/physiology , Retina/metabolism , Vasoactive Intestinal Peptide/metabolism
19.
Am J Pathol ; 194(5): 796-809, 2024 05.
Article in English | MEDLINE | ID: mdl-38395146

ABSTRACT

α-Synuclein (α-Syn) is a key determinator of Parkinson disease (PD) pathology, but synapse and microcircuit pathologies in the retina underlying visual dysfunction are poorly understood. Herein, histochemical and ultrastructural analyses and ophthalmologic measurements in old transgenic M83 PD model (mice aged 16 to 18 months) indicated that abnormal α-Syn aggregation in the outer plexiform layer (OPL) was associated with degeneration in the C-terminal binding protein 2 (CtBP2)+ ribbon synapses of photoreceptor terminals and protein kinase C alpha (PKCα)+ rod bipolar cell terminals, whereas α-Syn aggregates in the inner retina correlated with the reduction and degeneration of tyrosine hydroxylase- and parvalbumin-positive amacrine cells. Phosphorylated Ser129 α-synuclein expression was strikingly restricted in the OPL, with the most severe degenerations in the entire retina, including mitochondrial degeneration and loss of ribbon synapses in 16- to 18-month-old mice. These synapse- and microcircuit-specific deficits of the rod pathway at the CtBP2+ rod terminals and PKCα+ rod bipolar and amacrine cells were associated with attenuated a- and b-wave amplitudes and oscillatory potentials on the electroretinogram. They were also associated with the impairment of visual functions, including reduced contrast sensitivity and impairment of the middle range of spatial frequencies. Collectively, these findings demonstrate that α-Syn aggregates cause the synapse- and microcircuit-specific deficits of the rod pathway and the most severe damage to the OPL, providing the retinal synaptic and microcircuit basis for visual dysfunctions in PD.


Subject(s)
Protein Kinase C-alpha , alpha-Synuclein , Animals , Mice , alpha-Synuclein/metabolism , Amacrine Cells/metabolism , Protein Kinase C-alpha/metabolism , Retina/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/ultrastructure , Synapses/metabolism , Transcription Factors/metabolism
20.
PLoS Biol ; 22(2): e3002538, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38422167

ABSTRACT

In mammals, starburst amacrine cells are centrally involved in motion vision and a new study in PLOS Biology, by Yan and colleagues finds that zebrafish have them, too. They coexist with a second pair of starburst-like neurons, but neither appears to be strongly motion selective.


Subject(s)
Amacrine Cells , Zebrafish , Animals , Amacrine Cells/physiology , Retina/physiology , Mammals , Cholinergic Agents
SELECTION OF CITATIONS
SEARCH DETAIL