Tag Archives: ultrastructure

Impact of Retinal Degeneration on Response of ON and OFF Cone Bipolar Cells to Electrical Stimulation

We have a new manuscript from the lab in IEEE, Impact of Retinal Degeneration on Response of ON and OFF Cone Bipolar Cells to Electrical Stimulation. This manuscript is in collaboration with the Lazzi lab out of USC.  The first author, Shayan Farzad, Pragya Kosta, Ege Iseri, Steven T Walston, Jean-Marie C. Bouteiller,  Rebecca L. Pfeiffer @BeccaPfeiffer19, Crystal L. Sigulinsky @CSigulinsky, Jia-Hui Yang, Jessica C. Garcia, James R. Anderson, Bryan W. Jones @BWJones, and Gianluca Lazzi. The PDF is here.

Abstract: In retinal degenerative diseases, such as retinitis pigmentosa (RP) and age-related macular degeneration (AMD), the photoreceptors become stressed and start to degenerate in the early stages of the disease. Retinal prosthetic devices have been developed to restore vision in patients by applying electrical stimulation to the surviving retinal cells. However, these devices provide limited visual perception as the therapeutic interventions are generally considered in the later stages of the disease when only inner retinal layer cells are left. A potential treatment option for retinal degenerative diseases in the early stages can be stimulating bipolar cells, which receive presynaptic signals from photoreceptors. In this work, we constructed computational models of healthy and degenerated (both ON and OFF-type) cone bipolar cells (CBCs) with realistic morphologies extracted from connectomes of the healthy and early-stage degenerated rabbit retina. We examined these cells’ membrane potential and axon terminal calcium current differences when subjected to electrical stimulation. In addition, we investigated how differently healthy and degenerated cells behave with respect to various stimulation parameters, including pulse duration and cells’ distance from the stimulating electrode. The results suggested that regardless of the position of the OFF CBCs in the retina model, there is not a significant difference between the membrane potential of healthy and degenerate cells when electrically stimulated. However, the healthy ON CBC axon terminal membrane potential rising time-constant is shorter (0.29 ± 0.03 ms) than the degenerated cells (0.8 ± 0.07 ms). Moreover, the ionic calcium channels at the axon terminals of the cells have a higher concentration and higher current in degenerated cells (32.24 ± 6.12 pA) than the healthy cells (13.64 ± 2.88 pA) independently of the cell’s position.

Mitochondrial Transfer Between Inner Retinal Neurons

This abstract was presented today, April 26th at the 2023 Association for Research in Vision and Opthalmology (ARVO) meetings in New Orleans, Louisiana by Selena Wirthlin, Crystal Sigulinsky, James Anderson, and Bryan William Jones.

Full resolution version here.

Purpose
Intercellular mitochondrial transfer has been reported across a variety of cells and tissues under both physiological and pathological conditions. Such transfer has shown broad therapeutic potential. The effectiveness of this therapy, however, is limited by a lack of understanding of the cellular and molecular mechanisms. Here, the ultrastructural features of mitochondrial transfer between inner retinal neurons discovered through retinal connectomics analysis is shown.

Methods
Retinal Connectome 2 (RC2) was built by automated transmission electron microscopy at ultrastructural (2nm/pixel) resolution. RC2 is a 0.25mm diameter volume of retina obtained from a 5-month-old female C57BL/6J mouse. The Viking application was used to visualize and annotate inter- and intracellular features of interest in the connectome.

Results
Exploration of RC2 revealed material transfer between apposing neural processes within the OFF subliminal of the inner plexiform layer. The transferred material can be defined as a mitochondria, confirmed by the presence of crustae. At the transfer site, a short, electron-dense 140-nm diameter tube with a curved cap tightly associated with the inner mitochondrial membrane of one neuritis extends into a vacuole within the apposing neuritis formed by the plasma membranes of the two cells. Thin cytoskeletal components consistent with actin microfilaments extend into the mitochondrion. Morphology and synaptology of the acceptor cell confirm it is an Aii amacrine cell, while preliminary findings suggest the donor cell is a type of ON/OFF ganglion cell.

Conclusions
These findings demonstrate active mitochondrial transfer between different classes of endogenous inner retinal neurons and suggests it may represent an important component of tissue homeostasis in the retina. Features of this transfer differ from previously reported mitochondrial transfer between photoreceptors upon transplantation, which may indicate cell type- or context-dependent differences in the cellular or molecular mechanisms. Our findings demonstrate active mitochondrial transfer between different classes of endogenous inner retinal neurons and suggest it may represent an important component of tissue homeostasis in the retina. Features of this transfer differ from previous reports by the Wallace and Pearson groups of material transfer between photoreceptors upon transplantation through tunneling nanotubes (Ortin- Martinez et al., 2021; Kalargyrou et al., 2021), which may indicate cell type- or context-dependent differences in the cellular or molecular mechanisms. Understanding these mechanisms could serve as a catalyst for development of novel therapeutics for disease in the retina and beyond.

Species-Specific Connectivity In The Aii Connectome

This talk was presented today, April 25th at the 2023 Association for Research in Vision and Opthalmology (ARVO) meetings in New Orleans, Louisiana by Crystal Sigulinsky as part of an ARVO Minisymposium organized by Bryan William Jones.

Abstract: Biomedical research relies heavily on animal models to study human disease and develop therapeutics. Understanding the architectural diversity in neural networks between humans and these model species is essential for choosing a relevant study model and interpreting conflicting results. Using comparative connectomics, we sought to map and compare the local neural network architecture of rabbit and mouse retinal Aii amacrine cells. This specialized narrow-field, multistratified, glycinergic interneuron has critical feedforward and feedback roles in both the photopic and scotopic retinal networks spanning the ON and OFF pathways, making it an ideal candidate for investigating species-specific differences in retinal networks. High-resolution, serial-section transmission electron microscopy (TEM) volumes of rabbit (RC1: female, 13- month, Dutch Belted) and mouse (RC2: female, 5-month, C57BL/6J) retinal tissue provided spatially-registered synaptic maps of Aii connectivity at directly comparable resolution and completeness. These reveal that despite species-specific morphologies, gross synaptology and compartmentalization appear conserved. Yet, rabbit and mouse Aii cells diverge in the weighting of their partnerships, most notably in their coupling profiles. Opposing biases in gap junction partnerships and their respective sizing rules indicate a greater relative output by mouse Aii cells to ON pathways than in rabbit. However, a unique topological conformation for a subset of conventional presynapses formed by Aii cell lobular dendrites with species-specific features and prevalence may influence signal output to specific partner classes within the OFF pathway and either nullify or exacerbate this difference in ON/OFF output. Additionally, rabbit Aii cells in RC1 showed greater Aii-Aii coupling than in mouse, which may suggest greater signal-to-noise compensation. Lastly, preliminary data suggest mouse Aii cells receive greater excitatory, but not inhibitory input/feedback from the OFF pathway than in rabbit. Together these data indicate that precise neural circuit architectures diverge between species and require detailed, comprehensive mapping to begin to dissect potential influence on signal flow.

Structural Motifs Of Excitatory Synapses In The Mammalian Retina

This abstract was presented today, April 24th at the 2023 Association for Research in Vision and Opthalmology (ARVO) meetings in New Orleans, Louisiana by Taylor Otterness, Crystal Sigulinsky, James Anderson, and Bryan William Jones.

Full resolution version here.

Purpose
Connectivity within the nervous system is precise and disruptions lead to degraded performance and disease, yet the rules that govern connectivity remain unknown. Recent efforts reveal that different types of cone bipolar cells in the neural retina show preferences in the selection and frequency of presynaptic structure types used for signal transmission. However, it is not yet known how these differences are related to the quantity or type of postsynaptic partner. We used Retinal Connectome 1 (RC1) to analyze the synaptic output of rabbit CBb6 cells, a type of ON cone bipolar cell that forms excitatory synapses via diverse presynaptic structure types, to identify patterns in how these cells interact with their postsynaptic partners.

Methods
RC1 is a 0.25 mm diameter volume sampled from mid-peripheral retina of a 13 month old female Dutch-Belted rabbit, serially sectioned at 70 nm, and imaged at ultrastructural resolution (2nm/px) using transmission electron microscopy. Postsynaptic partners of CBb6 cell 6156’s presynaptic structures were annotated using the Viking Viewer for Connectomics. Statistical analyses were conducted in Microsoft Excel and investigated further with 3D rendering and graph visualization of connectivity.

Results
The factors tracked for comparison included presynaptic structure type, target number, and postsynaptic partner type. Multiribbon synapses of CBb6 cell 6156 trended towards having a greater number of output partners, with a greater proportion of dyads than monads. Despite this, triads and quadrads were only found opposing single ribbon synapses. As the different presynaptic structure types may differ in the strength of neurotransmitter release (ribbonless < single ribbon < multiribbon), these findings are inconsistent with scaling of output to the number of postsynaptic targets. Both amacrine cells (AC) and ganglion cells (GC) are postsynaptic partners of 6156. However, single ribbon and ribbonless structures appear biased towards AC only targets, while multiribbon synapses appear biased toward mixed AC and GC targets.

Conclusions
Target type relationships appear more important than the number of targets in determining presynaptic structure type in CBb6. Future efforts will examine size differences of postsynaptic structures and presynaptic ribbon size, and even compare across bipolar cell classes, in order to provide further insight on the connectivity rules underlying excitatory synapses.

Optic Cup Morphogenesis Requires Neural Crest-Mediated Basement Membrane Assembly

We have a new manuscript out in Development, Optic cup morphogenesis requires neural crest-mediated basement membrane assembly.

Authors: Chase D. Bryan @CDBE30, Macaulie A. Casey, Rebecca L. Pfeiffer @BeccaPfeiffer19, Bryan W. Jones @BWJones, and Kristen M. Kwan @BlockInTheBack

This is a collaborative project out of the Kwan lab that we helped out with some of the ultrastructural work and analysis.

Abstract: Organogenesis requires precise interactions between a developing tissue and its environment. In vertebrates, the developing eye is surrounded by a complex extracellular matrix as well as multiple mesenchymal cell populations. Disruptions to either the matrix or periocular mesenchyme can cause defects in early eye development, yet in many cases, the underlying mechanism is unknown. Here, using multidimensional imaging and computational analyses in zebrafish, we establish that cell movements in the developing optic cup require neural crest. Ultrastructural analysis reveals that basement membrane formation around the developing eye is also dependent on neural crest, but only specifically around the retinal pigment epithelium. Neural crest cells produce the extracellular matrix protein nidogen: impairing nidogen function disrupts eye development, and strikingly, expression of nidogen in the absence of neural crest partially restores optic cup morphogenesis. These results demonstrate that eye formation is regulated in part by extrinsic control of extracellular matrix assembly.

SEM vs. TEM

SEM vs. TEM is a tradeoff of convenience, resolution, cost and speed. The very physics of SEM signal integration means that the fundamental acquisition time for large canonical volume datasets are incompatible with 5 year grant cycles. SEM based approaches can potentially rival TEM, but dwell time/pixel increases logarithmically with resolution.

To give you some idea for the resolution differences at routine capture speeds, both of these above images capture a region within the inner plexiform layer of retina, looking at bipolar cell terminals. The TEM image was captured at a standard operating resolution of 2nm/pixel. The SEM image was captured at 16nm/pixel. You cannot see any gap junctions that might be present in the SEM image and you can only infer or guess at synaptic ribbons. And look at the texture!

You *can* get better resolution with SEM, but as I said before, the capture time increases logarithmically. To accomplish what we perform in 8-10 hours with a TEM, would take 108-115 hours on a current, cutting edge multi beam SEM. There are many other advantages of TEM including the ability to capture higher resolution images faster, be able to re-image in goniometric tilt series, be able to integrate molecular markers inside connectome volumes, and a TEM is about 1/3rd the cost of an SEM. Also, SEM images tend to be texturally poor as they are made from capturing electron backscatter of surfaces rather than made by projection of electrons through a small volume, and there is tremendous value in the texture of ultrastructural images. Ergo, this is why we use TEM.

This is not to say that SEM is not a great tool. It is just not the best tool for large scale connectomics where you have to have the resolution to capture all synapses and gap junctions, over large areas. For smaller volumes that do not require a canonical sampling of cell classes, SEM is absolutely an appropriate tool.

This content was originally published on Jonesblog.

The Rod-Cone Crossover Connectome of Mammalian Bipolar Cells

We have a new publication out (direct link), The rod-cone crossover connectome of mammalian bipolar cells authored by Scott Lauritzen, Crystal Sigulinsky, James Anderson, Michael Kalloniatis, Noah Nelson, Danny Emrich, Chris Rapp, Nicolas McCarthy, Ethan Kerzner, Mariah Meyer, Bryan W. Jones, and Robert Marc.

Abstract: The basis of cross-suppression between rod and cone channels has long been an enigma. Using rabbit retinal connectome RC1, we show that all cone bipolar cell (BC) classes inhibit rod BCs via amacrine cell (AC) motifs (C1-6); that all cone BC classes are themselves inhibited by AC motifs (R1-5, R25) driven by rod BCs. A sparse symmetric AC motif (CR) is presynaptic and postsynaptic to both rod and cone BCs. ON cone BCs of all classes drive inhibition of rod BCs via motif C1 wide-field GABAergic ACs (γACs) and motif C2 narrow field glycinergic ON ACs (GACs). Each rod BC receives ≈ 10 crossover AC synapses and each ON cone BC can target ≈ 10 or more rod BCs via separate AC processes. OFF cone BCs mediate monosynaptic inhibition of rod BCs via motif C3 driven by OFF γACs and GACs and disynaptic inhibition via motifs C4 and C5 driven by OFF wide-field γACs and narrow-field GACs, respectively. Motifs C4 and C5 form halos of 60-100 inhibitory synapses on proximal dendrites of AI γACs. Rod BCs inhibit surrounding arrays of cone BCs through AII GAC networks that access ON and OFF cone BC patches via motifs R1, R2, R4 R5 and a unique ON AC motif R3 that collects rod BC inputs and targets ON cone BCs. Crossover synapses for motifs C1, C4, C5 and R3 are 3-4x larger than typical feedback synapses, which may be a signature for synaptic winner-take-all switches.

2-nm Resolution Anatomy of Retinal Neuro-Glial-Vascular Architecture

This abstract was presented today, May 2th at the 2016 Association for Research in Vision and Opthalmology (ARVO) meetings in Seattle, Washington by Jefferson R. Brown, Rebecca L. Pfeiffer, Crystal Sigulinsky, Felix Vazquez-Chona, Daniel Emrich, Bryan W. Jones, Robert E. Marc.

Abstract Number: 995

Author Block: Jefferson R. Brown, Rebecca L. Pfeiffer, Crystal Sigulinsky, Felix Vazquez-Chona, Daniel Emrich, Bryan W. Jones, Robert E. Marc
1 Dept of Ophthalmology, University of Utah, Salt Lake City, Utah, United States

Disclosure Block:Jefferson R. Brown, None; Rebecca L. Pfeiffer, None; Crystal Sigulinsky, None; Felix Vazquez-Chona, None; Daniel Emrich, None; Bryan W. Jones, None; Robert E. Marc, Signature Immunologics (Code I (Personal Financial Interest) )

Purpose:Retinal vasculature is strongly affected by degenerative pathologies and in turn, may also contribute to their progression. However, much of what we understand about the normal, healthy interaction between neurons, glia, and blood vessels at the ultrastructural level is limited to single section electron microscopy. The technology of serial section transmission electron microscopy (ssTEM) extends the high definition of TEM imaging into three dimensions to create volumes, allowing for more thorough visualization and analysis of the vascular-glial-neuronal complex.

Methods:RC2 is a 40TB ssTEM volume of over 1,400 horizontal sections of retinal tissue derived from an adult female C57BL/6J mouse. The tissue sample is 250 um in diameter and spans the outer nuclear layer to the vitreal surface. Baseline resolution is 2.18nm per pixel. Visualization, navigation and metadata annotations of the database are made via the Viking software suite.

Results:Much of the retinal vascular basement membrane directly contacts Muller cells. In the ganglion cell layer, direct basement membrane contact with astrocytes is frequent. Microglia commonly contact the basement membrane, and occasionally direct contact of neurons onto basement membrane was observed. Full 3D reconstruction of all vascular pathways with associated endothelia and pericytes within the volume was completed, demonstrating that all the retinal capillary layers are continuous with one another [Figure].

Conclusions:The presence of occasional direct neuronal contact onto vascular basement membrane supports earlier work by Ochs and colleagues (2000) and suggests the blood-retina barrier does not universally involve retinal glia. However, since such contacts are extremely sparse, it remains to be seen whether this finding has biologic significance, though their existence suggests significance. The RC2 volume is a valuable resource to aid in discovery of defining characteristics of wild type neurovascular architecture.


The intro figure is a side view of reconstruction of all vasculature within the RC2 volume. Vessels at the top of the figure correspond to the outer plexiform layer, while those at the bottom correspond to the ganglion cell layer. This capillary plexus is one continuous structure. Visualization by VikingView software.

A Targeted Inhibitor Of The Alternative Complement Pathway Accelerates Recovery From Smoke-Induced Ocular Injury

We have a new publication out, A Targeted Inhibitor Of The Alternative Complement Pathway Accelerates Recovery From Smoke-Induced Ocular Injury authored by Alex Woodell, Bryan W. Jones, Tucker Williamson, Gloriane Schnabolk, Stephen Tomlinson, Carl Atkinson and Bärbel Rohrer.

Abstract:
PURPOSE. Morphological and genetic evidence exists that an overactive complement system driven by the complement alternative pathway (CAP) is involved in pathogenesis of age- related macular degeneration (AMD). Smoking is the only modifiable risk factor for AMD. As we have shown that smoke-related ocular pathology can be prevented in mice that lack an essential activator of CAP, we ask here whether this pathology can be reversed by increasing inhibition in CAP.

METHODS. Mice were exposed to either cigarette smoke (CS) or filtered air (6 hours/day, 5 days/week, 6 months). Smoke-exposed animals were then treated with the CAP inhibitor (CR2-fH) or vehicle control (PBS) for 3 months. Spatial frequency and contrast sensitivity were assessed by optokinetic response paradigms at 6 and 9 months; additional readouts included assessment of retinal morphology by electron microscopy (EM) and gene expression analysis by quantitative PCR.

RESULTS. The CS mice treated with CR2-fH showed significant improvement in contrast threshold compared to PBS-treated mice, whereas spatial frequency was unaffected by CS or pharmacological intervention. Treatment with CR2-fH in CS animals reversed thinning of the retina observed in PBS-treated mice as analyzed by spectral-domain optical coherence tomography, and reversed most morphological changes in RPE and Bruch’s membrane seen in CS animals by EM.

CONCLUSIONS. Taken together, these findings suggest that CAP inhibitors not only prevent, but have the potential to accelerate, the clearance of complement-mediated ocular injury. Improving our understanding of the regulation of the CAP pathway is paramount to developing novel treatment approaches for AMD.

Retinal Remodeling in Human Retinitis Pigmentosa

We have a new publication out (Direct Link, Free Open Access), Retinal Remodeling in Human Retinitis Pigmentosa authored by Bryan W. Jones, Rebecca Pfeiffer, Drew Ferrell, Carl Watt, Michael Marmor and Robert Marc.

Abstract: Retinitis Pigmentosa (RP) in the human is a progressive, currently irreversible neural degenerative disease usually caused by gene defects that disrupt the function or architecture of the photoreceptors. While RP can initially be a disease of photoreceptors, there is increasing evidence that the inner retina becomes progressively disorganized as the outer retina degenerates. These alterations have been extensively described in animal models, but remodeling in humans has not been as well characterized. This study, using computational molecular phenotyping (CMP) seeks to advance our understanding of the retinal remodeling process in humans. We describe cone mediated preservation of overall topology, retinal reprogramming in the earliest stages of the disease in retinal bipolar cells, and alterations in both small molecule and protein signatures of neurons and glia. Furthermore, while Müller glia appear to be some of the last cells left in the degenerate retina, they are also one of the first cell classes in the neural retina to respond to stress which may reveal mechanisms related to remodeling and cell death in other retinal cell classes. Also fundamentally important is the finding that retinal network topologies are altered. Our results suggest interventions that presume substantial preservation of the neural retina will likely fail in late stages of the disease. Even early intervention offers no guarantee that the interventions will be immune to progressive remodeling. Fundamental work in the biology and mechanisms of disease progression are needed to support vision rescue strategies.