Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Luis Pérez de Sevilla Müller is active.

Publication


Featured researches published by Luis Pérez de Sevilla Müller.


The Journal of Comparative Neurology | 2014

The RNA binding protein RBPMS is a selective marker of ganglion cells in the mammalian retina.

Allen Rodriguez; Luis Pérez de Sevilla Müller; Nicholas C. Brecha

There are few neurochemical markers that reliably identify retinal ganglion cells (RGCs), which are a heterogeneous population of cells that integrate and transmit the visual signal from the retina to the central visual nuclei. We have developed and characterized a new set of affinity‐purified guinea pig and rabbit antibodies against RNA‐binding protein with multiple splicing (RBPMS). On western blots these antibodies recognize a single band at 〜24 kDa, corresponding to RBPMS, and they strongly label RGC and displaced RGC (dRGC) somata in mouse, rat, guinea pig, rabbit, and monkey retina. RBPMS‐immunoreactive cells and RGCs identified by other techniques have a similar range of somal diameters and areas. The density of RBPMS cells in mouse and rat retina is comparable to earlier semiquantitative estimates of RGCs. RBPMS is mainly expressed in medium and large DAPI‐, DRAQ5‐, NeuroTrace‐ and NeuN‐stained cells in the ganglion cell layer (GCL), and RBPMS is not expressed in syntaxin (HPC‐1)‐immunoreactive cells in the inner nuclear layer (INL) and GCL, consistent with their identity as RGCs, and not displaced amacrine cells. In mouse and rat retina, most RBPMS cells are lost following optic nerve crush or transection at 3 weeks, and all Brn3a‐, SMI‐32‐, and melanopsin‐immunoreactive RGCs also express RBPMS immunoreactivity. RBPMS immunoreactivity is localized to cyan fluorescent protein (CFP)‐fluorescent RGCs in the B6.Cg‐Tg(Thy1‐CFP)23Jrs/J mouse line. These findings show that antibodies against RBPMS are robust reagents that exclusively identify RGCs and dRGCs in multiple mammalian species, and they will be especially useful for quantification of RGCs. J. Comp. Neurol. 522:1411–1443, 2014.


PLOS ONE | 2014

Melanopsin Ganglion Cells Are the Most Resistant Retinal Ganglion Cell Type to Axonal Injury in the Rat Retina

Luis Pérez de Sevilla Müller; Allison Sargoy; Allen Rodriguez; Nicholas C. Brecha

We report that the most common retinal ganglion cell type that remains after optic nerve transection is the M1 melanopsin ganglion cell. M1 ganglion cells are members of the intrinsically photosensitive retinal ganglion cell population that mediates non-image-forming vision, comprising ∼2.5% of all ganglion cells in the rat retina. In the present study, M1 ganglion cells comprised 1.7±1%, 28±14%, 55±13% and 82±8% of the surviving ganglion cells 7, 14, 21 and 60 days after optic nerve transection, respectively. Average M1 ganglion cell somal diameter and overall morphological appearance remained unchanged in non-injured and injured retinas, suggesting a lack of injury-induced degeneration. Average M1 dendritic field size increased at 7 and 60 days following optic nerve transection, while average dendritic field size remained similar in non-injured retinas and in retinas at 14 and 21 days after optic nerve transection. These findings demonstrate that M1 ganglion cells are more resistant to injury than other ganglion cell types following optic nerve injury, and provide an opportunity to develop pharmacological or genetic therapeutic approaches to mitigate ganglion cell death and save vision following optic nerve injury.


Investigative Ophthalmology & Visual Science | 2014

Loss of Outer Retinal Neurons and Circuitry Alterations in the DBA/2J Mouse

Laura Fernández-Sánchez; Luis Pérez de Sevilla Müller; Nicholas C. Brecha; Nicolás Cuenca

PURPOSE The DBA/2J mouse line develops essential iris atrophy, pigment dispersion, and glaucomatous age-related changes, including an increase of IOP, optic nerve atrophy, and retinal ganglion cell (RGC) death. The aim of this study was to evaluate possible morphological changes in the outer retina of the DBA/2J mouse concomitant with disease progression and aging, based on the reduction of both the a- and b-waves and photopic flicker ERGs in this mouse line. METHODS Vertically sectioned DBA/2J mice retinas were evaluated at 3, 8, and 16 months of age using photoreceptor, horizontal, and bipolar cell markers. Sixteen-month-old C57BL/6 mice retinas were used as controls. RESULTS The DBA/2J mice had outer retinal degeneration at all ages, with the most severe degeneration in the oldest retinas. At 3 months of age, the number of photoreceptor cells and the thickness of the OPL were reduced. In addition, there was a loss of horizontal and ON-bipolar cell processes. At 8 months of age, RGC degeneration occurred in patches, and in the outer retina overlying these patches, cone morphology was impaired with a reduction in size as well as loss of outer segments and growth of horizontal and bipolar cell processes into the outer nuclear layer. At 16 months of age, connectivity between photoreceptors and horizontal and bipolar cell processes overlying these patches was lost. CONCLUSIONS Retinal degeneration in DBA/2J mice includes photoreceptor death, loss of bipolar and horizontal cell processes, and loss of synaptic contacts in an aging-dependent manner.


eNeuro | 2016

Targeted Deletion of Vesicular GABA Transporter from Retinal Horizontal Cells Eliminates Feedback Modulation of Photoreceptor Calcium Channels.

Arlene A. Hirano; Xue Liu; Jim Boulter; James Grove; Luis Pérez de Sevilla Müller; Steven Barnes; Nicholas C. Brecha

Abstract The cellular mechanisms underlying feedback signaling from horizontal cells to photoreceptors, which are important for the formation of receptive field surrounds of early visual neurons, remain unsettled. Mammalian horizontal cells express a complement of synaptic proteins that are necessary and sufficient for calcium-dependent exocytosis of inhibitory neurotransmitters at their contacts with photoreceptor terminals, suggesting that they are capable of releasing GABA via vesicular release. To test whether horizontal cell vesicular release is involved in feedback signaling, we perturbed inhibitory neurotransmission in these cells by targeted deletion of the vesicular GABA transporter (VGAT), the protein responsible for the uptake of inhibitory transmitter by synaptic vesicles. To manipulate horizontal cells selectively, an iCre mouse line with Cre recombinase expression controlled by connexin57 (Cx57) regulatory elements was generated. In Cx57-iCre mouse retina, only horizontal cells expressed Cre protein, and its expression occurred in all retinal regions. After crossing with a VGATflox/flox mouse line, VGAT was selectively eliminated from horizontal cells, which was confirmed immunohistochemically. Voltage-gated ion channel currents in horizontal cells of Cx57-VGAT−/− mice were the same as Cx57-VGAT+/+ controls, as were the cell responses to the ionotropic glutamate receptor agonist kainate, but the response to the GABAA receptor agonist muscimol in Cx57-VGAT−/− mice was larger. In contrast, the feedback inhibition of photoreceptor calcium channels, which in control animals is induced by horizontal cell depolarization, was completely absent in Cx57-VGAT−/− mice. The results suggest that vesicular release of GABA from horizontal cells is required for feedback inhibition of photoreceptors.


The Journal of Comparative Neurology | 2013

Expression of voltage-gated calcium channel α2δ4 subunits in the mouse and rat retina†

Luis Pérez de Sevilla Müller; Janelle Liu; Alexander Solomon; Allen Rodriguez; Nicholas C. Brecha

High‐voltage activated Ca channels participate in multiple cellular functions, including transmitter release, excitation, and gene transcription. Ca channels are heteromeric proteins consisting of a pore‐forming α1 subunit and auxiliary α2δ and β subunits. Although there are reports of α2δ4 subunit mRNA in the mouse retina and localization of the α2δ4 subunit immunoreactivity to salamander photoreceptor terminals, there is a limited overall understanding of its expression and localization in the retina. α2δ4 subunit expression and distribution in the mouse and rat retina were evaluated by using reverse transcriptase polymerase chain reaction, western blot, and immunohistochemistry with specific primers and a well‐characterized antibody to the α2δ4 subunit. α2δ4 subunit mRNA and protein are present in mouse and rat retina, brain, and liver homogenates. Immunostaining for the α2δ4 subunit is mainly localized to Müller cell processes and endfeet, photoreceptor terminals, and photoreceptor outer segments. This subunit is also expressed in a few displaced ganglion cells and bipolar cell dendrites. These findings suggest that the α2δ4 subunit participates in the modulation of L‐type Ca2+ current regulating neurotransmitter release from photoreceptor terminals and Ca2+‐dependent signaling pathways in bipolar and Müller cells. J. Comp. Neurol. 521:2486–2501, 2013.


The Journal of Comparative Neurology | 2015

Expression and cellular localization of the voltage-gated calcium channel α2δ3 in the rodent retina.

Luis Pérez de Sevilla Müller; Allison Sargoy; Laura Fernández-Sánchez; Allen Rodriguez; Janelle Liu; Nicolás Cuenca; Nicholas C. Brecha

High‐voltage‐activated calcium channels are hetero‐oligomeric protein complexes that mediate multiple cellular processes, including the influx of extracellular Ca2+, neurotransmitter release, gene transcription, and synaptic plasticity. These channels consist of a primary α1 pore‐forming subunit, which is associated with an extracellular α2δ subunit and an intracellular β auxiliary subunit, which alter the gating properties and trafficking of the calcium channel. The cellular localization of the α2δ3 subunit in the mouse and rat retina is unknown. In this study using RT‐PCR, a single band at ∼305 bp corresponding to the predicted size of the α2δ3 subunit fragment was found in mouse and rat retina and brain homogenates. Western blotting of rodent retina and brain homogenates showed a single 123‐kDa band. Immunohistochemistry with an affinity‐purified antibody to the α2δ3 subunit revealed immunoreactive cell bodies in the ganglion cell layer and inner nuclear layer and immunoreactive processes in the inner plexiform layer and the outer plexiform layer. α2δ3 immunoreactivity was localized to multiple cell types, including ganglion, amacrine, and bipolar cells and photoreceptors, but not horizontal cells. The expression of the α2δ3 calcium channel subunit to multiple cell types suggests that this subunit participates widely in Ca‐channel‐mediated signaling in the retina. J. Comp. Neurol. 523:1443–1460, 2015.


Frontiers in Neural Circuits | 2016

Peripheral Sensory Neurons Expressing Melanopsin Respond to Light.

Anna Matynia; Eileen Nguyen; Xiaoping Sun; Frank W. Blixt; Sachin Parikh; Jason Kessler; Luis Pérez de Sevilla Müller; Samer Habib; Paul Kim; Zhe Z. Wang; Allen Rodriguez; Andrew Charles; Steven Nusinowitz; Lars Edvinsson; Steven Barnes; Nicholas C. Brecha; Michael B. Gorin

The ability of light to cause pain is paradoxical. The retina detects light but is devoid of nociceptors while the trigeminal sensory ganglia (TG) contain nociceptors but not photoreceptors. Melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) are thought to mediate light-induced pain but recent evidence raises the possibility of an alternative light responsive pathway independent of the retina and optic nerve. Here, we show that melanopsin is expressed in both human and mouse TG neurons. In mice, they represent 3% of small TG neurons that are preferentially localized in the ophthalmic branch of the trigeminal nerve and are likely nociceptive C fibers and high-threshold mechanoreceptor Aδ fibers based on a strong size-function association. These isolated neurons respond to blue light stimuli with a delayed onset and sustained firing, similar to the melanopsin-dependent intrinsic photosensitivity observed in ipRGCs. Mice with severe bilateral optic nerve crush exhibit no light-induced responses including behavioral light aversion until treated with nitroglycerin, an inducer of migraine in people and migraine-like symptoms in mice. With nitroglycerin, these same mice with optic nerve crush exhibit significant light aversion. Furthermore, this retained light aversion remains dependent on melanopsin-expressing neurons. Our results demonstrate a novel light-responsive neural function independent of the optic nerve that may originate in the peripheral nervous system to provide the first direct mechanism for an alternative light detection pathway that influences motivated behavior.


Frontiers in Neuroanatomy | 2017

Prox1 Is a Marker for AII Amacrine Cells in the Mouse Retina

Luis Pérez de Sevilla Müller; Shaghauyegh S. Azar; Janira de los Santos; Nicholas C. Brecha

The transcription factor Prox1 is expressed in multiple cells in the retina during eye development. This study has focused on neuronal Prox1 expression in the inner nuclear layer (INL) of the adult mouse retina. Prox1 immunostaining was evaluated in vertical retinal sections and whole mount preparations using a specific antibody directed to the C-terminus of Prox1. Strong immunostaining was observed in numerous amacrine cell bodies and in all horizontal cell bodies in the proximal and distal INL, respectively. Some bipolar cells were also weakly immunostained. Prox1-immunoreactive amacrine cells expressed glycine, and they formed 35 ± 3% of all glycinergic amacrine cells. Intracellular Neurobiotin injections into AII amacrine cells showed that all gap junction-coupled AII amacrine cells express Prox1, and no other Prox1-immunostained amacrine cells were in the immediate area surrounding the injected AII amacrine cell. Prox1-immunoreactive amacrine cell bodies were distributed across the retina, with their highest density (3887 ± 160 cells/mm2) in the central retina, 0.5 mm from the optic nerve head, and their lowest density (3133 ± 350 cells/mm2) in the mid-peripheral retina, 2 mm from the optic nerve head. Prox1-immunoreactive amacrine cell bodies comprised ~9.8% of the total amacrine cell population, and they formed a non-random mosaic with a regularity index (RI) of 3.4, similar to AII amacrine cells in the retinas of other mammals. Together, these findings indicate that AII amacrine cells are the predominant and likely only amacrine cell type strongly expressing Prox1 in the adult mouse retina, and establish Prox1 as a marker of AII amacrine cells.


The Journal of Comparative Neurology | 2017

Multiple cell types form the VIP amacrine cell population

Luis Pérez de Sevilla Müller; Alexander Solomon; Kristopher Sheets; Hinekura Hapukino; Allen Rodriguez; Nicholas C. Brecha

Amacrine cells are a heterogeneous group of interneurons that form microcircuits with bipolar, amacrine and ganglion cells to process visual information in the inner retina. This study has characterized the morphology, neurochemistry and major cell types of a VIP‐ires‐Cre amacrine cell population. VIP‐tdTomato and ‐Confetti (Brainbow2.1) mouse lines were generated by crossing a VIP‐ires‐Cre line with either a Cre‐dependent tdTomato or Brainbow2.1 reporter line. Retinal sections and whole‐mounts were evaluated by quantitative, immunohistochemical, and intracellular labeling approaches. The majority of tdTomato and Confetti fluorescent cell bodies were in the inner nuclear layer (INL) and a few cell bodies were in the ganglion cell layer (GCL). Fluorescent processes ramified in strata 1, 3, 4, and 5 of the inner plexiform layer (IPL). All tdTomato fluorescent cells expressed syntaxin 1A and GABA‐immunoreactivity indicating they were amacrine cells. The average VIP‐tdTomato fluorescent cell density in the INL and GCL was 535 and 24 cells/mm2, respectively. TdTomato fluorescent cells in the INL and GCL contained VIP‐immunoreactivity. The VIP‐ires‐Cre amacrine cell types were identified in VIP‐Brainbow2.1 retinas or by intracellular labeling in VIP‐tdTomato retinas. VIP‐1 amacrine cells are bistratified, wide‐field cells that ramify in strata 1, 4, and 5, VIP‐2A and 2B amacrine cells are medium‐field cells that mainly ramify in strata 3 and 4, and VIP‐3 displaced amacrine cells are medium‐field cells that ramify in strata 4 and 5 of the IPL. VIP‐ires‐Cre amacrine cells form a neuropeptide‐expressing cell population with multiple cell types, which are likely to have distinct roles in visual processing.


Journal of Visualized Experiments | 2014

Immunohistochemical and Calcium Imaging Methods in Wholemount Rat Retina

Allison Sargoy; Steven Barnes; Nicholas C. Brecha; Luis Pérez de Sevilla Müller

In this paper we describe the tools, reagents, and the practical steps that are needed for: 1) successful preparation of wholemount retinas for immunohistochemistry and, 2) calcium imaging for the study of voltage gated calcium channel (VGCC) mediated calcium signaling in retinal ganglion cells. The calcium imaging method we describe circumvents issues concerning non-specific loading of displaced amacrine cells in the ganglion cell layer.

Collaboration


Dive into the Luis Pérez de Sevilla Müller's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Allison Sargoy

University of California

View shared research outputs
Top Co-Authors

Avatar

Steven Barnes

University of California

View shared research outputs
Top Co-Authors

Avatar

Janelle Liu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steven Barnes

University of California

View shared research outputs
Top Co-Authors

Avatar

Alex Solomon

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge