1
|
Dey H, Perez-Hurtado M, Heidelberger R. Syntaxin 3B: A SNARE Protein Required for Vision. Int J Mol Sci 2024; 25:10665. [PMID: 39408994 PMCID: PMC11476516 DOI: 10.3390/ijms251910665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Syntaxin 3 is a member of a large protein family of syntaxin proteins that mediate fusion between vesicles and their target membranes. Mutations in the ubiquitously expressed syntaxin 3A splice form give rise to a serious gastrointestinal disorder in humans called microvillus inclusion disorder, while mutations that additionally involve syntaxin 3B, a splice form that is expressed primarily in retinal photoreceptors and bipolar cells, additionally give rise to an early onset severe retinal dystrophy. In this review, we discuss recent studies elucidating the roles of syntaxin 3B and the regulation of syntaxin 3B functionality in membrane fusion and neurotransmitter release in the vertebrate retina.
Collapse
Affiliation(s)
| | | | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (H.D.)
| |
Collapse
|
2
|
Thoreson WB, Zenisek D. Presynaptic Proteins and Their Roles in Visual Processing by the Retina. Annu Rev Vis Sci 2024; 10:347-375. [PMID: 38621251 PMCID: PMC11536687 DOI: 10.1146/annurev-vision-101322-111204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
The sense of vision begins in the retina, where light is detected and processed through a complex series of synaptic connections into meaningful information relayed to the brain via retinal ganglion cells. Light responses begin as tonic and graded signals in photoreceptors, later emerging from the retina as a series of spikes from ganglion cells. Processing by the retina extracts critical features of the visual world, including spatial frequency, temporal frequency, motion direction, color, contrast, and luminance. To achieve this, the retina has evolved specialized and unique synapse types. These include the ribbon synapses of photoreceptors and bipolar cells, the dendritic synapses of amacrine and horizontal cells, and unconventional synaptic feedback from horizontal cells to photoreceptors. We review these unique synapses in the retina with a focus on the presynaptic molecules and physiological properties that shape their capabilities.
Collapse
Affiliation(s)
- Wallace B Thoreson
- Departments of Ophthalmology & Visual Sciences and Pharmacology & Experimental Neuroscience, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA;
| | - David Zenisek
- Departments of Cellular and Molecular Physiology, Ophthalmology and Visual Sciences, and Neuroscience, Yale University, New Haven, Connecticut, USA;
| |
Collapse
|
3
|
Tebbe L, Kakakhel M, Al-Ubaidi MR, Naash MI. The role of syntaxins in retinal function and health. Front Cell Neurosci 2024; 18:1380064. [PMID: 38799985 PMCID: PMC11119284 DOI: 10.3389/fncel.2024.1380064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024] Open
Abstract
The soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein (SNAP) receptor (SNARE) superfamily plays a pivotal role in cellular trafficking by facilitating membrane fusion events. These SNARE proteins, including syntaxins, assemble into complexes that actively facilitate specific membrane fusion events. Syntaxins, as integral components of the SNARE complex, play a crucial role in initiating and regulating these fusion activities. While specific syntaxins have been extensively studied in various cellular processes, including neurotransmitter release, autophagy and endoplasmic reticulum (ER)-to-Golgi protein transport, their roles in the retina remain less explored. This review aims to enhance our understanding of syntaxins' functions in the retina by shedding light on how syntaxins mediate membrane fusion events unique to the retina. Additionally, we seek to establish a connection between syntaxin mutations and retinal diseases. By exploring the intricate interplay of syntaxins in retinal function and health, we aim to contribute to the broader comprehension of cellular trafficking in the context of retinal physiology and pathology.
Collapse
Affiliation(s)
| | | | | | - Muna I. Naash
- *Correspondence: Muna I. Naash, ; Muayyad R. Al-Ubaidi,
| |
Collapse
|
4
|
Huang M, Chow CH, Gurdita A, Harada H, Pham Truong VQB, Eide S, Sun HS, Feng ZP, Monnier PP, Wallace VA, Sugita S. SNAP-25, but not SNAP-23, is essential for photoreceptor development, survival, and function in mice. Commun Biol 2024; 7:34. [PMID: 38182732 PMCID: PMC10770054 DOI: 10.1038/s42003-023-05760-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
SNARE-mediated vesicular transport is thought to play roles in photoreceptor glutamate exocytosis and photopigment delivery. However, the functions of Synaptosomal-associated protein (SNAP) isoforms in photoreceptors are unknown. Here, we revisit the expression of SNAP-23 and SNAP-25 and generate photoreceptor-specific knockout mice to investigate their roles. Although we find that SNAP-23 shows weak mRNA expression in photoreceptors, SNAP-23 removal does not affect retinal morphology or vision. SNAP-25 mRNA is developmentally regulated and undergoes mRNA trafficking to photoreceptor inner segments at postnatal day 9 (P9). SNAP-25 knockout photoreceptors develop normally until P9 but degenerate by P14 resulting in severe retinal thinning. Photoreceptor loss in SNAP-25 knockout mice is associated with abolished electroretinograms and vision loss. We find mistrafficked photopigments, enlarged synaptic vesicles, and abnormal synaptic ribbons which potentially underlie photoreceptor degeneration. Our results conclude that SNAP-25, but not SNAP-23, mediates photopigment delivery and synaptic functioning required for photoreceptor development, survival, and function.
Collapse
Affiliation(s)
- Mengjia Huang
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Chun Hin Chow
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Akshay Gurdita
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Donald K. Johnson Eye Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Hidekiyo Harada
- Donald K. Johnson Eye Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Victor Q B Pham Truong
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Donald K. Johnson Eye Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Sarah Eide
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Anatomy, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Philippe P Monnier
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Donald K. Johnson Eye Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Department of Ophthalmology & Vision Sciences, University of Toronto, Toronto, ON, M5T 3A9, Canada
| | - Valerie A Wallace
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Donald K. Johnson Eye Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Department of Ophthalmology & Vision Sciences, University of Toronto, Toronto, ON, M5T 3A9, Canada
| | - Shuzo Sugita
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON, M5T 0S8, Canada.
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
5
|
Sanchez-Bretano A, Keeling E, Scott JA, Lynn SA, Soundara-Pandi SP, Macdonald SL, Newall T, Griffiths H, Lotery AJ, Ratnayaka JA, Self JE, Lee H. Human equivalent doses of L-DOPA rescues retinal morphology and visual function in a murine model of albinism. Sci Rep 2023; 13:17173. [PMID: 37821525 PMCID: PMC10567794 DOI: 10.1038/s41598-023-44373-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/07/2023] [Indexed: 10/13/2023] Open
Abstract
L-DOPA is deficient in the developing albino eye, resulting in abnormalities of retinal development and visual impairment. Ongoing retinal development after birth has also been demonstrated in the developing albino eye offering a potential therapeutic window in humans. To study whether human equivalent doses of L-DOPA/Carbidopa administered during the crucial postnatal period of neuroplasticity can rescue visual function, OCA C57BL/6 J-c2J OCA1 mice were treated with a 28-day course of oral L-DOPA/Carbidopa at 3 different doses from 15 to 43 days postnatal age (PNA) and for 3 different lengths of treatment, to identify optimum dosage and treatment length. Visual electrophysiology, acuity, and retinal morphology were measured at 4, 5, 6, 12 and 16 weeks PNA and compared to untreated C57BL/6 J (WT) and OCA1 mice. Quantification of PEDF, βIII-tubulin and syntaxin-3 expression was also performed. Our data showed impaired retinal morphology, decreased retinal function and lower visual acuity in untreated OCA1 mice compared to WT mice. These changes were diminished or eliminated when treated with higher doses of L-DOPA/Carbidopa. Our results demonstrate that oral L-DOPA/Carbidopa supplementation at human equivalent doses during the postnatal critical period of retinal neuroplasticity can rescue visual retinal morphology and retinal function, via PEDF upregulation and modulation of retinal synaptogenesis, providing a further step towards developing an effective treatment for albinism patients.
Collapse
Affiliation(s)
- Aida Sanchez-Bretano
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton University Hospital, South Block Mail Point 806, Level D, Southampton, SO16 6YD, UK
| | - Eloise Keeling
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton University Hospital, South Block Mail Point 806, Level D, Southampton, SO16 6YD, UK
| | - Jennifer A Scott
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton University Hospital, South Block Mail Point 806, Level D, Southampton, SO16 6YD, UK
| | - Savannah A Lynn
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton University Hospital, South Block Mail Point 806, Level D, Southampton, SO16 6YD, UK
| | - Sudha Priya Soundara-Pandi
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton University Hospital, South Block Mail Point 806, Level D, Southampton, SO16 6YD, UK
| | - Sarah L Macdonald
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton University Hospital, South Block Mail Point 806, Level D, Southampton, SO16 6YD, UK
| | - Tutte Newall
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton University Hospital, South Block Mail Point 806, Level D, Southampton, SO16 6YD, UK
| | - Helen Griffiths
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton University Hospital, South Block Mail Point 806, Level D, Southampton, SO16 6YD, UK
| | - Andrew J Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton University Hospital, South Block Mail Point 806, Level D, Southampton, SO16 6YD, UK
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton, SO16 6YD, UK
| | - J Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton University Hospital, South Block Mail Point 806, Level D, Southampton, SO16 6YD, UK
| | - Jay E Self
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton University Hospital, South Block Mail Point 806, Level D, Southampton, SO16 6YD, UK
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton, SO16 6YD, UK
| | - Helena Lee
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Sir Henry Wellcome Laboratories, Southampton University Hospital, South Block Mail Point 806, Level D, Southampton, SO16 6YD, UK.
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton, SO16 6YD, UK.
| |
Collapse
|
6
|
Nishad R, Betancourt-Solis M, Dey H, Heidelberger R, McNew JA. Regulation of Syntaxin3B-Mediated Membrane Fusion by T14, Munc18, and Complexin. Biomolecules 2023; 13:1463. [PMID: 37892145 PMCID: PMC10604575 DOI: 10.3390/biom13101463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Retinal neurons that form ribbon-style synapses operate over a wide dynamic range, continuously relaying visual information to their downstream targets. The remarkable signaling abilities of these neurons are supported by specialized presynaptic machinery, one component of which is syntaxin3B. Syntaxin3B is an essential t-SNARE protein of photoreceptors and bipolar cells that is required for neurotransmitter release. It has a light-regulated phosphorylation site in its N-terminal domain at T14 that has been proposed to modulate membrane fusion. However, a direct test of the latter has been lacking. Using a well-controlled in vitro fusion assay, we found that a phosphomimetic T14 syntaxin3B mutation leads to a small but significant enhancement of SNARE-mediated membrane fusion following the formation of the t-SNARE complex. While the addition of Munc18a had only a minimal effect on membrane fusion mediated by SNARE complexes containing wild-type syntaxin3B, a more significant enhancement was observed in the presence of Munc18a when the SNARE complexes contained a syntaxin3B T14 phosphomimetic mutant. Finally, we showed that the retinal-specific complexins (Cpx III and Cpx IV) inhibited membrane fusion mediated by syntaxin3B-containing SNARE complexes in a dose-dependent manner. Collectively, our results establish that membrane fusion mediated by syntaxin3B-containing SNARE complexes is regulated by the T14 residue of syntaxin3B, Munc18a, and Cpxs III and IV.
Collapse
Affiliation(s)
- Rajkishor Nishad
- Department of BioSciences, Rice University, 6500 Main Street, MS 601, Houston, TX 77005, USA;
| | - Miguel Betancourt-Solis
- Department of BioSciences, Rice University, 6500 Main Street, MS 601, Houston, TX 77005, USA;
- Lonza Biologics, 14905 Kirby Dr, Houston, TX 77047, USA
| | - Himani Dey
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center, Houston (UTHealth Houston), 6431 Fannin Street, Houston, TX 77030, USA;
| | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center, Houston (UTHealth Houston), 6431 Fannin Street, Houston, TX 77030, USA;
| | - James A. McNew
- Department of BioSciences, Rice University, 6500 Main Street, MS 601, Houston, TX 77005, USA;
| |
Collapse
|
7
|
Perez-Hurtado M, Dao C, Saenz AE, Heidelberger R. Syntaxin 3 is haplosufficient for long-term photoreceptor survival in the mouse retina. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1208805. [PMID: 37609371 PMCID: PMC10443939 DOI: 10.3389/fopht.2023.1208805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Biallelic loss-of-function mutations in the syntaxin 3 gene have been linked to a severe retinal dystrophy in humans that presents in early childhood. In mouse models, biallelic inactivation of the syntaxin 3 gene in photoreceptors rapidly leads to their death. What is not known is whether a monoallelic syntaxin 3 loss-of-function mutation might cause photoreceptor loss with advancing age. To address this question, we compared the outer nuclear layer of older adult mice (≈ 20 months of age) that were heterozygous for syntaxin 3 with those of similarly-aged control mice. We found that the photoreceptor layer maintains its thickness in mice that are heterozygous for syntaxin 3 relative to controls and that photoreceptor somatic counts are comparable. In addition, dendritic sprouting of the rod bipolar cell dendrites into the outer nuclear layer, which occurs following the loss of functional rod targets, was similar between genotypes. Thus, syntaxin 3 appears to be haplosufficient for photoreceptor survival, even with advancing age.
Collapse
Affiliation(s)
- Mariajose Perez-Hurtado
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX
| | - Calvin Dao
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX
| | - Amanda E. Saenz
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX
| | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX
- The University of Texas MDAnderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston TX
| |
Collapse
|
8
|
Davison A, Lux UT, Brandstätter JH, Babai N. T-Type Ca 2+ Channels Boost Neurotransmission in Mammalian Cone Photoreceptors. J Neurosci 2022; 42:6325-6343. [PMID: 35803735 PMCID: PMC9398539 DOI: 10.1523/jneurosci.1878-21.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 06/13/2022] [Accepted: 06/22/2022] [Indexed: 11/21/2022] Open
Abstract
It is a commonly accepted view that light stimulation of mammalian photoreceptors causes a graded change in membrane potential instead of developing a spike. The presynaptic Ca2+ channels serve as a crucial link for the coding of membrane potential variations into neurotransmitter release. Cav1.4 L-type Ca2+ channels are expressed in photoreceptor terminals, but the complete pool of Ca2+ channels in cone photoreceptors appears to be more diverse. Here, we discovered, employing whole-cell patch-clamp recording from cone photoreceptor terminals in both sexes of mice, that their Ca2+ currents are composed of low- (T-type Ca2+ channels) and high- (L-type Ca2+ channels) voltage-activated components. Furthermore, Ca2+ channels exerted self-generated spike behavior in dark membrane potentials, and spikes were generated in response to light/dark transition. The application of fast and slow Ca2+ chelators revealed that T-type Ca2+ channels are located close to the release machinery. Furthermore, capacitance measurements indicated that they are involved in evoked vesicle release. Additionally, RT-PCR experiments showed the presence of Cav3.2 T-type Ca2+ channels in cone photoreceptors but not in rod photoreceptors. Altogether, we found several crucial functions of T-type Ca2+ channels, which increase the functional repertoire of cone photoreceptors. Namely, they extend cone photoreceptor light-responsive membrane potential range, amplify dark responses, generate spikes, increase intracellular Ca2+ levels, and boost synaptic transmission.SIGNIFICANCE STATEMENT Photoreceptors provide the first synapse for coding light information. The key elements in synaptic transmission are the voltage-sensitive Ca2+ channels. Here, we provide evidence that mouse cone photoreceptors express low-voltage-activated Cav3.2 T-type Ca2+ channels in addition to high-voltage-activated L-type Ca2+ channels. The presence of T-type Ca2+ channels in cone photoreceptors appears to extend their light-responsive membrane potential range, amplify dark response, generate spikes, increase intracellular Ca2+ levels, and boost synaptic transmission. By these functions, Cav3.2 T-type Ca2+ channels increase the functional repertoire of cone photoreceptors.
Collapse
Affiliation(s)
- Adam Davison
- Department of Biology, Animal Physiology/Neurobiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Uwe Thorsten Lux
- Department of Biology, Animal Physiology/Neurobiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Johann Helmut Brandstätter
- Department of Biology, Animal Physiology/Neurobiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Norbert Babai
- Department of Biology, Animal Physiology/Neurobiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| |
Collapse
|
9
|
Ciliary Proteins Repurposed by the Synaptic Ribbon: Trafficking Myristoylated Proteins at Rod Photoreceptor Synapses. Int J Mol Sci 2022; 23:ijms23137135. [PMID: 35806143 PMCID: PMC9266639 DOI: 10.3390/ijms23137135] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/25/2022] Open
Abstract
The Unc119 protein mediates transport of myristoylated proteins to the photoreceptor outer segment, a specialized primary cilium. This transport activity is regulated by the GTPase Arl3 as well as by Arl13b and Rp2 that control Arl3 activation/inactivation. Interestingly, Unc119 is also enriched in photoreceptor synapses and can bind to RIBEYE, the main component of synaptic ribbons. In the present study, we analyzed whether the known regulatory proteins, that control the Unc119-dependent myristoylated protein transport at the primary cilium, are also present at the photoreceptor synaptic ribbon complex by using high-resolution immunofluorescence and immunogold electron microscopy. We found Arl3 and Arl13b to be enriched at the synaptic ribbon whereas Rp2 was predominantly found on vesicles distributed within the entire terminal. These findings indicate that the synaptic ribbon could be involved in the discharge of Unc119-bound lipid-modified proteins. In agreement with this hypothesis, we found Nphp3 (Nephrocystin-3), a myristoylated, Unc119-dependent cargo protein enriched at the basal portion of the ribbon in close vicinity to the active zone. Mutations in Nphp3 are known to be associated with Senior–Løken Syndrome 3 (SLS3). Visual impairment and blindness in SLS3 might thus not only result from ciliary dysfunctions but also from malfunctions of the photoreceptor synapse.
Collapse
|
10
|
Gething C, Ferrar J, Misra B, Howells G, Andrzejewski AL, Bowen ME, Choi UB. Conformational change of Syntaxin-3b in regulating SNARE complex assembly in the ribbon synapses. Sci Rep 2022; 12:9261. [PMID: 35661757 PMCID: PMC9166750 DOI: 10.1038/s41598-022-09654-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 03/10/2022] [Indexed: 11/09/2022] Open
Abstract
Neurotransmitter release of synaptic vesicles relies on the assembly of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex, consisting of syntaxin and SNAP-25 on the plasma membrane and synaptobrevin on the synaptic vesicle. The formation of the SNARE complex progressively zippers towards the membranes, which drives membrane fusion between the plasma membrane and the synaptic vesicle. However, the underlying molecular mechanism of SNARE complex regulation is unclear. In this study, we investigated the syntaxin-3b isoform found in the retinal ribbon synapses using single-molecule fluorescence resonance energy transfer (smFRET) to monitor the conformational changes of syntaxin-3b that modulate the SNARE complex formation. We found that syntaxin-3b is predominantly in a self-inhibiting closed conformation, inefficiently forming the ternary SNARE complex. Conversely, a phosphomimetic mutation (T14E) at the N-terminal region of syntaxin-3b promoted the open conformation, similar to the constitutively open form of syntaxin LE mutant. When syntaxin-3b is bound to Munc18-1, SNARE complex formation is almost completely blocked. Surprisingly, the T14E mutation of syntaxin-3b partially abolishes Munc18-1 regulation, acting as a conformational switch to trigger SNARE complex assembly. Thus, we suggest a model where the conformational change of syntaxin-3b induced by phosphorylation initiates the release of neurotransmitters in the ribbon synapses.
Collapse
Affiliation(s)
- Claire Gething
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Joshua Ferrar
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Bishal Misra
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Giovanni Howells
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA
| | | | - Mark E Bowen
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, 11794, USA.,Quantum-Si, Inc, Guilford, CT, 06437, USA
| | - Ucheor B Choi
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA. .,Quantum-Si, Inc, Guilford, CT, 06437, USA.
| |
Collapse
|
11
|
Complexin Suppresses Spontaneous Exocytosis by Capturing the Membrane-Proximal Regions of VAMP2 and SNAP25. Cell Rep 2021; 32:107926. [PMID: 32698012 PMCID: PMC7116205 DOI: 10.1016/j.celrep.2020.107926] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 02/28/2020] [Accepted: 06/26/2020] [Indexed: 01/29/2023] Open
Abstract
The neuronal protein complexin contains multiple domains that exert clamping and facilitatory functions to tune spontaneous and action potential-triggered synaptic release. We address the clamping mechanism and show that the accessory helix of complexin arrests assembly of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex that forms the core machinery of intracellular membrane fusion. In a reconstituted fusion assay, site-and stage-specific photo-cross-linking reveals that, prior to fusion, the complexin accessory helix laterally binds the membrane-proximal C-terminal ends of SNAP25 and VAMP2. Corresponding complexin interface mutants selectively increase spontaneous release of neuro-transmitters in living neurons, implying that the accessory helix suppresses final zippering/assembly of the SNARE four-helix bundle by restraining VAMP2 and SNAP25.
Collapse
|
12
|
Burger CA, Jiang D, Mackin RD, Samuel MA. Development and maintenance of vision's first synapse. Dev Biol 2021; 476:218-239. [PMID: 33848537 DOI: 10.1016/j.ydbio.2021.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 12/21/2022]
Abstract
Synapses in the outer retina are the first information relay points in vision. Here, photoreceptors form synapses onto two types of interneurons, bipolar cells and horizontal cells. Because outer retina synapses are particularly large and highly ordered, they have been a useful system for the discovery of mechanisms underlying synapse specificity and maintenance. Understanding these processes is critical to efforts aimed at restoring visual function through repairing or replacing neurons and promoting their connectivity. We review outer retina neuron synapse architecture, neural migration modes, and the cellular and molecular pathways that play key roles in the development and maintenance of these connections. We further discuss how these mechanisms may impact connectivity in the retina.
Collapse
Affiliation(s)
- Courtney A Burger
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Danye Jiang
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Robert D Mackin
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Melanie A Samuel
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
13
|
Thoreson WB. Transmission at rod and cone ribbon synapses in the retina. Pflugers Arch 2021; 473:1469-1491. [PMID: 33779813 DOI: 10.1007/s00424-021-02548-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/29/2022]
Abstract
Light-evoked voltage responses of rod and cone photoreceptor cells in the vertebrate retina must be converted to a train of synaptic vesicle release events for transmission to downstream neurons. This review discusses the processes, proteins, and structures that shape this critical early step in vision, focusing on studies from salamander retina with comparisons to other experimental animals. Many mechanisms are conserved across species. In cones, glutamate release is confined to ribbon release sites although rods are also capable of release at non-ribbon sites. The role of non-ribbon release in rods remains unclear. Release from synaptic ribbons in rods and cones involves at least three vesicle pools: a readily releasable pool (RRP) matching the number of membrane-associated vesicles along the ribbon base, a ribbon reserve pool matching the number of additional vesicles on the ribbon, and an enormous cytoplasmic reserve. Vesicle release increases in parallel with Ca2+ channel activity. While the opening of only a few Ca2+ channels beneath each ribbon can trigger fusion of a single vesicle, sustained release rates in darkness are governed by the rate at which the RRP can be replenished. The number of vacant release sites, their functional status, and the rate of vesicle delivery in turn govern replenishment. Along with an overview of the mechanisms of exocytosis and endocytosis, we consider specific properties of ribbon-associated proteins and pose a number of remaining questions about this first synapse in the visual system.
Collapse
Affiliation(s)
- Wallace B Thoreson
- Truhlsen Eye Institute, Departments of Ophthalmology & Visual Sciences and Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
14
|
Hirano AA, Vuong HE, Kornmann HL, Schietroma C, Stella SL, Barnes S, Brecha NC. Vesicular Release of GABA by Mammalian Horizontal Cells Mediates Inhibitory Output to Photoreceptors. Front Cell Neurosci 2020; 14:600777. [PMID: 33335476 PMCID: PMC7735995 DOI: 10.3389/fncel.2020.600777] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Feedback inhibition by horizontal cells regulates rod and cone photoreceptor calcium channels that control their release of the neurotransmitter glutamate. This inhibition contributes to synaptic gain control and the formation of the center-surround antagonistic receptive fields passed on to all downstream neurons, which is important for contrast sensitivity and color opponency in vision. In contrast to the plasmalemmal GABA transporter found in non-mammalian horizontal cells, there is evidence that the mechanism by which mammalian horizontal cells inhibit photoreceptors involves the vesicular release of the inhibitory neurotransmitter GABA. Historically, inconsistent findings of GABA and its biosynthetic enzyme, L-glutamate decarboxylase (GAD) in horizontal cells, and the apparent lack of surround response block by GABAergic agents diminished support for GABA's role in feedback inhibition. However, the immunolocalization of the vesicular GABA transporter (VGAT) in the dendritic and axonal endings of horizontal cells that innervate photoreceptor terminals suggested GABA was released via vesicular exocytosis. To test the idea that GABA is released from vesicles, we localized GABA and GAD, multiple SNARE complex proteins, synaptic vesicle proteins, and Cav channels that mediate exocytosis to horizontal cell dendritic tips and axonal terminals. To address the perceived relative paucity of synaptic vesicles in horizontal cell endings, we used conical electron tomography on mouse and guinea pig retinas that revealed small, clear-core vesicles, along with a few clathrin-coated vesicles and endosomes in horizontal cell processes within photoreceptor terminals. Some small-diameter vesicles were adjacent to the plasma membrane and plasma membrane specializations. To assess vesicular release, a functional assay involving incubation of retinal slices in luminal VGAT-C antibodies demonstrated vesicles fused with the membrane in a depolarization- and calcium-dependent manner, and these labeled vesicles can fuse multiple times. Finally, targeted elimination of VGAT in horizontal cells resulted in a loss of tonic, autaptic GABA currents, and of inhibitory feedback modulation of the cone photoreceptor Cai, consistent with the elimination of GABA release from horizontal cell endings. These results in mammalian retina identify the central role of vesicular release of GABA from horizontal cells in the feedback inhibition of photoreceptors.
Collapse
Affiliation(s)
- Arlene A. Hirano
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Helen E. Vuong
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Helen L. Kornmann
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cataldo Schietroma
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Salvatore L. Stella
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Steven Barnes
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Doheny Eye Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nicholas C. Brecha
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
15
|
Campbell JR, Li H, Wang Y, Kozhemyakin M, Hunt AJ, Liu X, Janz R, Heidelberger R. Phosphorylation of the Retinal Ribbon Synapse Specific t-SNARE Protein Syntaxin3B Is Regulated by Light via a Ca 2 +-Dependent Pathway. Front Cell Neurosci 2020; 14:587072. [PMID: 33192329 PMCID: PMC7606922 DOI: 10.3389/fncel.2020.587072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/11/2020] [Indexed: 12/27/2022] Open
Abstract
Neurotransmitter release at retinal ribbon-style synapses utilizes a specialized t-SNARE protein called syntaxin3B (STX3B). In contrast to other syntaxins, STX3 proteins can be phosphorylated in vitro at T14 by Ca2+/calmodulin-dependent protein kinase II (CaMKII). This modification has the potential to modulate SNARE complex formation required for neurotransmitter release in an activity-dependent manner. To determine the extent to which T14 phosphorylation occurs in vivo in the mammalian retina and characterize the pathway responsible for the in vivo phosphorylation of T14, we utilized quantitative immunofluorescence to measure the levels of STX3 and STX3 phosphorylated at T14 (pSTX3) in the synaptic terminals of mouse retinal photoreceptors and rod bipolar cells (RBCs). Results demonstrate that STX3B phosphorylation at T14 is light-regulated and dependent upon the elevation of intraterminal Ca2+. In rod photoreceptor terminals, the ratio of pSTX3 to STX3 was significantly higher in dark-adapted mice, when rods are active, than in light-exposed mice. By contrast, in RBC terminals, the ratio of pSTX3 to STX3 was higher in light-exposed mice, when these terminals are active, than in dark-adapted mice. These results were recapitulated in the isolated eyecup preparation, but only when Ca2+ was included in the external medium. In the absence of external Ca2+, pSTX3 levels remained low regardless of light/dark exposure. Using the isolated RBC preparation, we next showed that elevation of intraterminal Ca2+ alone was sufficient to increase STX3 phosphorylation at T14. Furthermore, both the non-specific kinase inhibitor staurosporine and the selective CaMKII inhibitor AIP inhibited the Ca2+-dependent increase in the pSTX3/STX3 ratio in isolated RBC terminals, while in parallel experiments, AIP suppressed RBC depolarization-evoked exocytosis, measured using membrane capacitance measurements. Our data support a novel, illumination-regulated modulation of retinal ribbon-style synapse function in which activity-dependent Ca2+ entry drives the phosphorylation of STX3B at T14 by CaMKII, which in turn, modulates the ability to form SNARE complexes required for exocytosis.
Collapse
Affiliation(s)
- Joseph R Campbell
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Hongyan Li
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yanzhao Wang
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Maxim Kozhemyakin
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Albert J Hunt
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xiaoqin Liu
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Roger Janz
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
16
|
Hunter DD, Manglapus MK, Bachay G, Claudepierre T, Dolan MW, Gesuelli KA, Brunken WJ. CNS synapses are stabilized trans-synaptically by laminins and laminin-interacting proteins. J Comp Neurol 2017; 527:67-86. [PMID: 29023785 DOI: 10.1002/cne.24338] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/21/2017] [Accepted: 09/29/2017] [Indexed: 01/05/2023]
Abstract
The retina expresses several laminins in the outer plexiform layer (OPL), where they may provide an extracellular scaffold for synapse stabilization. Mice with a targeted deletion of the laminin β2 gene (Lamb2) exhibit retinal disruptions: photoreceptor synapses in the OPL are disorganized and the retinal physiological response is attenuated. We hypothesize that laminins are required for proper trans-synaptic alignment. To test this, we compared the distribution, expression, association and modification of several pre- and post-synaptic elements in wild-type and Lamb2-null retinae. A potential laminin receptor, integrin α3, is at the presynaptic side of the wild-type OPL. Another potential laminin receptor, dystroglycan, is at the post-synaptic side of the wild-type OPL. Integrin α3 and dystroglycan can be co-immunoprecipitated with the laminin β2 chain, demonstrating that they may bind laminins. In the absence of the laminin β2 chain, the expression of many pre-synaptic components (bassoon, kinesin, among others) is relatively undisturbed although their spatial organization and anchoring to the membrane is disrupted. In contrast, in the Lamb2-null, β-dystroglycan (β-DG) expression is altered, co-localization of β-DG with dystrophin and the glutamate receptor mGluR6 is disrupted, and the post-synaptic bipolar cell components mGluR6 and GPR179 become dissociated, suggesting that laminins mediate scaffolding of post-synaptic components. In addition, although pikachurin remains associated with β-DG, pikachurin is no longer closely associated with mGluR6 or α-DG in the Lamb2-null. These data suggest that laminins act as links among pre- and post-synaptic laminin receptors and α-DG and pikachurin in the synaptic space to maintain proper trans-synaptic alignment.
Collapse
Affiliation(s)
- Dale D Hunter
- Department of Anatomy and Cellular Biology, Tufts University and Tufts Center for Vision Research, Boston, Massachusetts.,Department of Ophthalmology and the SUNY Eye Institute, Upstate Medical University, Syracuse, New York
| | - Mary K Manglapus
- Department of Anatomy and Cellular Biology, Tufts University and Tufts Center for Vision Research, Boston, Massachusetts
| | - Galina Bachay
- Department of Ophthalmology and the SUNY Eye Institute, Upstate Medical University, Syracuse, New York
| | - Thomas Claudepierre
- Department of Anatomy and Cellular Biology, Tufts University and Tufts Center for Vision Research, Boston, Massachusetts
| | - Michael W Dolan
- Department of Ophthalmology and the SUNY Eye Institute, Upstate Medical University, Syracuse, New York
| | - Kelly-Ann Gesuelli
- Department of Ophthalmology and the SUNY Eye Institute, Upstate Medical University, Syracuse, New York
| | - William J Brunken
- Department of Anatomy and Cellular Biology, Tufts University and Tufts Center for Vision Research, Boston, Massachusetts.,Department of Ophthalmology and the SUNY Eye Institute, Upstate Medical University, Syracuse, New York
| |
Collapse
|
17
|
Wen X, Saltzgaber GW, Thoreson WB. Kiss-and-Run Is a Significant Contributor to Synaptic Exocytosis and Endocytosis in Photoreceptors. Front Cell Neurosci 2017; 11:286. [PMID: 28979188 PMCID: PMC5611439 DOI: 10.3389/fncel.2017.00286] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 09/01/2017] [Indexed: 11/13/2022] Open
Abstract
Accompanying sustained release in darkness, rod and cone photoreceptors exhibit rapid endocytosis of synaptic vesicles. Membrane capacitance measurements indicated that rapid endocytosis retrieves at least 70% of the exocytotic membrane increase. One mechanism for rapid endocytosis is kiss-and-run fusion where vesicles briefly contact the plasma membrane through a small fusion pore. Release can also occur by full-collapse in which vesicles merge completely with the plasma membrane. We assessed relative contributions of full-collapse and kiss-and-run in salamander photoreceptors using optical techniques to measure endocytosis and exocytosis of large vs. small dye molecules. Incubation with small dyes (SR101, 1 nm; 3-kDa dextran-conjugated Texas Red, 2.3 nm) loaded rod and cone synaptic terminals much more readily than larger dyes (10-kDa Texas Red, 4.6 nm; 10-kDa pHrodo, 4.6 nm; 70-kDa Texas Red, 12 nm) consistent with significant uptake through 2.3–4.6 nm fusion pores. By using total internal reflection fluorescence microscopy (TIRFM) to image individual vesicles, when rods were incubated simultaneously with Texas Red and AlexaFluor-488 dyes conjugated to either 3-kDa or 10-kDa dextran, more vesicles loaded small molecules than large molecules. Using TIRFM to detect release by the disappearance of dye-loaded vesicles, we found that SR101 and 3-kDa Texas Red were released from individual vesicles more readily than 10-kDa and 70-kDa Texas Red. Although 10-kDa pHrodo was endocytosed poorly like other large dyes, the fraction of release events was similar to SR101 and 3-kDa Texas Red. We hypothesize that while 10-kDa pHrodo may not exit through a fusion pore, release of intravesicular protons can promote detection of fusion events by rapidly quenching fluorescence of this pH-sensitive dye. Assuming that large molecules can only be released by full-collapse whereas small molecules can be released by both modes, our results indicate that 50%–70% of release from rods involves kiss-and-run with 2.3–4.6 nm fusion pores. Rapid retrieval of vesicles by kiss-and-run may limit membrane disruption of release site function during ongoing release at photoreceptor ribbon synapses.
Collapse
Affiliation(s)
- Xiangyi Wen
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical CenterOmaha, NE, United States.,Ophthalmology & Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical CenterOmaha, NE, United States
| | - Grant W Saltzgaber
- Ophthalmology & Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical CenterOmaha, NE, United States
| | - Wallace B Thoreson
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical CenterOmaha, NE, United States.,Ophthalmology & Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical CenterOmaha, NE, United States
| |
Collapse
|
18
|
Datta P, Gilliam J, Thoreson WB, Janz R, Heidelberger R. Two Pools of Vesicles Associated with Synaptic Ribbons Are Molecularly Prepared for Release. Biophys J 2017; 113:2281-2298. [PMID: 28863864 DOI: 10.1016/j.bpj.2017.08.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/28/2017] [Accepted: 08/07/2017] [Indexed: 11/17/2022] Open
Abstract
Neurons that form ribbon-style synapses are specialized for continuous exocytosis. To this end, their synaptic terminals contain numerous synaptic vesicles, some of which are ribbon associated, that have difference susceptibilities for undergoing Ca2+-dependent exocytosis. In this study, we probed the relationship between previously defined vesicle populations and determined their fusion competency with respect to SNARE complex formation. We found that both the rapidly releasing vesicle pool and the releasable vesicle pool of the retinal bipolar cell are situated at the ribbon-style active zones, where they functionally interact. A peptide inhibitor of SNARE complex formation failed to block exocytosis from either pool, suggesting that these two vesicle pools have formed the SNARE complexes necessary for fusion. By contrast, a third, slower component of exocytosis was blocked by the peptide, as was the functional replenishment of vesicle pools, indicating that few vesicles outside of the ribbon-style active zones were initially fusion competent. In cone photoreceptors, similar to bipolar cells, fusion of the initial ribbon-associated synaptic vesicle cohort was not blocked by the SNARE complex-inhibiting peptide, whereas a later phase of exocytosis, attributable to the recruitment and subsequent fusion of vesicles newly arrived at the synaptic ribbons, was blocked. Together, our results support a model in which stimulus-evoked exocytosis in retinal ribbon synapses is SNARE-dependent; where vesicles higher up on the synaptic ribbon replenish the rapidly releasing vesicle pool; and at any given time, there are sufficient SNARE complexes to support the fusion of the entire ribbon-associated cohort of vesicles. An important implication of these results is that ribbon-associated vesicles can form intervesicular SNARE complexes, providing mechanistic insight into compound fusion at ribbon-style synapses.
Collapse
Affiliation(s)
- Proleta Datta
- Department of Neurobiology and Anatomy, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas; The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Jared Gilliam
- Department of Neurobiology and Anatomy, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas
| | - Wallace B Thoreson
- Truhlsen Eye Institute, Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska; Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Roger Janz
- Department of Neurobiology and Anatomy, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas; The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas; The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, Texas.
| |
Collapse
|
19
|
Abstract
Abstract
Vision begins in highly specialized light-sensing neurons, the rod and cone photoreceptors. Their task is to absorb photons, transduce the physical stimulus into neuronal signals, transmit the signals to the parallel signal processing pathways of the subsequent retinal network with the highest possible fidelity and continuously adapt to changes in stimulus intensities. If you imagine a pitch-black night with only a few photons hitting the retina and being absorbed by the photoreceptors and a bright sunny day with the photoreceptors being bombarded by billions of photons, you realize that a photoreceptor faces two fundamental challenges: it has to detect the light signal with the greatest sensitivity, e.g. a single photon leads to a change in the membrane potential of a rod photoreceptor and, at the same time, encode light intensities covering a broad dynamic range of several orders of magnitude. To fulfill these demands, photoreceptors have developed separate, structurally and functionally specialized compartments, which are the topic of this article: the outer segment for signal transduction and the terminal with its highly complex ribbon synapse for signal transmission.
Collapse
|
20
|
Honma S, Kadono K, Kawano A, Wakisaka S. Immunohistochemical localization of SNARE core proteins in intrapulpal and intradentinal nerve fibers of rat molar teeth. Arch Oral Biol 2017; 73:248-252. [DOI: 10.1016/j.archoralbio.2016.10.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 10/15/2016] [Accepted: 10/24/2016] [Indexed: 11/29/2022]
|
21
|
Vila A, Whitaker CM, O'Brien J. Membrane-associated guanylate kinase scaffolds organize a horizontal cell synaptic complex restricted to invaginating contacts with photoreceptors. J Comp Neurol 2016; 525:850-867. [PMID: 27558197 DOI: 10.1002/cne.24101] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/02/2016] [Accepted: 08/15/2016] [Indexed: 12/21/2022]
Abstract
Synaptic processes and plasticity of synapses are mediated by large suites of proteins. In most cases, many of these proteins are tethered together by synaptic scaffold proteins. Scaffold proteins have a large number and typically a variety of protein interaction domains that allow many different proteins to be assembled into functional complexes. Because each scaffold protein has a different set of protein interaction domains and a unique set of interacting partners, the presence of synaptic scaffolds can provide insight into the molecular mechanisms that regulate synaptic processes. In studies of rabbit retina, we found SAP102 and Chapsyn110 selectively localized in the tips of B-type horizontal cell processes, where they contact cone and rod photoreceptors. We further identified some known SAP102 binding partners, kainate receptor GluR6/7 and inward rectifier potassium channel Kir2.1, closely associated with SAP102 in photoreceptor invaginations. The kainate receptor occupies a position distinct from that of the majority of AMPA receptors that dominate the horizontal cell postsynaptic response. GluR6/7 and Kir2.1 presumably are involved in synaptic processes that govern cell-to-cell communication and could both contribute in different ways to synaptic currents that mediate feedback signaling. Notably, we failed to find evidence for the presence of Cx57 or Cx59 that might be involved in ephaptic feedback signaling in this complex. The presence of SAP102 and its binding partners in both cone and rod invaginating synapses suggests that whatever mechanism is supported by this protein complex is present in both types of photoreceptors. J. Comp. Neurol. 525:850-867, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alejandro Vila
- Richard S. Ruiz M.D. Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas, 77030.,University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, 77030
| | - Christopher M Whitaker
- Richard S. Ruiz M.D. Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas, 77030
| | - John O'Brien
- Richard S. Ruiz M.D. Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas, 77030.,University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, 77030
| |
Collapse
|
22
|
Lin SY, Vollrath MA, Mangosing S, Shen J, Cardenas E, Corey DP. The zebrafish pinball wizard gene encodes WRB, a tail-anchored-protein receptor essential for inner-ear hair cells and retinal photoreceptors. J Physiol 2015; 594:895-914. [PMID: 26593130 DOI: 10.1113/jp271437] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/17/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The zebrafish pinball wizard (pwi) mutant is deaf and blind. The pwi phenotype includes a reduced auditory startle response and reduced visual evoked potentials, suggesting fatigue of synaptic release at ribbon synapses in hair cells and photoreceptors. The gene defective in the pwi mutant is WRB, a protein homologous to the yeast protein Get1, which is involved in the insertion of 'tail-anchored' membrane proteins. Many tail-anchored proteins are associated with synaptic vesicles, and both vesicles and synaptic ribbons are reduced in synaptic regions of hair cells in pwi. Abnormal processing of synaptic vesicle proteins important for ribbon synapses can explain the pwi phenotype. ABSTRACT In a large-scale zebrafish insertional mutagenesis screen, we identified the pinball wizard (pwi) line, which displays a deafness and blindness phenotype. Although the gross morphology and structure of the pwi larval inner ear was near normal, acoustic startle stimuli evoked smaller postsynaptic responses in afferent neurons, which rapidly fatigued. In the retina, similarly, an abnormal electroretinogram suggested reduced transmission at the photoreceptor ribbon synapse. A functional deficit in these specialized synapses was further supported by a reduction of synaptic marker proteins Rab3 and cysteine-string protein (CSP/Dnajc5) in hair cells and photoreceptors, as well as by a reduction of the number of both ribbons and vesicles surrounding the ribbons in hair cells. The pwi gene encodes a homologue of the yeast Get1 and human tryptophan-rich basic (WRB) proteins, which are receptors for membrane insertion of tail-anchored (TA) proteins. We identified more than 100 TA proteins expressed in hair cells, including many synaptic proteins. The expression of synaptobrevin and syntaxin 3, TA proteins essential for vesicle fusion, was reduced in the synaptic layers of mutant retina, consistent with a role for the pwi/WRB protein in TA-protein processing. The WRB protein was located near the apical domain and the ribbons in hair cells, and in the inner segment and the axon of the photoreceptor, consistent with a role in vesicle biogenesis or trafficking. Taken together, our results suggest that WRB plays a critical role in synaptic functions in these two sensory cells, and that disrupted processing of synaptic vesicle TA proteins explains much of the mutant phenotype.
Collapse
Affiliation(s)
- Shuh-Yow Lin
- Department of Surgery, UC San Diego School of Medicine, La Jolla, CA, USA
| | - Melissa A Vollrath
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Sara Mangosing
- Department of Surgery, UC San Diego School of Medicine, La Jolla, CA, USA
| | - Jun Shen
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Elena Cardenas
- Department of Surgery, UC San Diego School of Medicine, La Jolla, CA, USA
| | - David P Corey
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
23
|
Wichmann C, Moser T. Relating structure and function of inner hair cell ribbon synapses. Cell Tissue Res 2015; 361:95-114. [PMID: 25874597 PMCID: PMC4487357 DOI: 10.1007/s00441-014-2102-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/18/2014] [Indexed: 01/28/2023]
Abstract
In the mammalian cochlea, sound is encoded at synapses between inner hair cells (IHCs) and type I spiral ganglion neurons (SGNs). Each SGN receives input from a single IHC ribbon-type active zone (AZ) and yet SGNs indefatigably spike up to hundreds of Hz to encode acoustic stimuli with submillisecond precision. Accumulating evidence indicates a highly specialized molecular composition and structure of the presynapse, adapted to suit these high functional demands. However, we are only beginning to understand key features such as stimulus-secretion coupling, exocytosis mechanisms, exo-endocytosis coupling, modes of endocytosis and vesicle reformation, as well as replenishment of the readily releasable pool. Relating structure and function has become an important avenue in addressing these points and has been applied to normal and genetically manipulated hair cell synapses. Here, we review some of the exciting new insights gained from recent studies of the molecular anatomy and physiology of IHC ribbon synapses.
Collapse
Affiliation(s)
- C. Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University Medical Center Göttingen, Göttingen, Germany
| | - T. Moser
- Collaborative Research Center 889, University Medical Center Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University of Göttingen, Göttingen, Germany
- Bernstein Center for Computational Neuroscience, University of Göttingen, Göttingen, Germany
| |
Collapse
|
24
|
Chograni M, Alkuraya FS, Ourteni I, Maazoul F, Lariani I, Chaabouni HB. Autosomal recessive congenital cataract, intellectual disability phenotype linked to STX3 in a consanguineous Tunisian family. Clin Genet 2014; 88:283-7. [PMID: 25358429 DOI: 10.1111/cge.12489] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/08/2014] [Accepted: 08/18/2014] [Indexed: 11/27/2022]
Abstract
The aim of this study is to investigate the genetic basis of autosomal recessive congenital cataract and intellectual disability phenotype in a consanguineous Tunisian family. The whole genome scan of the studied family was performed with single nucleotide polymorphisms (SNPs). The resulted runs of homozygosity (ROH) were analyzed through the integrated Systems Tool for Eye gene discovery (iSyTE) in order to prioritize candidate genes associated with congenital cataract. Selected genes were amplified and sequenced. Bioinformatic analysis was conducted to predict the function of the mutant gene. We identified a new specific lens gene named syntaxin 3 linked to the studied phenotype. The direct sequencing of this gene revealed a novel missense mutation c.122A>G which results in p.E41G. Bioinformatic analysis suggested a deleterious effect of this mutation on protein structure and function. Here, we report for the first time a missense mutation of a novel lens specific gene STX3 in a phenotype associating autosomal recessive congenital cataract and intellectual disability.
Collapse
Affiliation(s)
- M Chograni
- Laboratory of Human Genetics, Faculty of Medicine of Tunis, University Tunis ElManar, Tunis, Tunisia
| | - F S Alkuraya
- Developmental Genetics Unit, King Faisal Specialist Hospital and Research Center, Riyad, Saudi Arabia
| | - I Ourteni
- Congenital and Hereditary Disorders Department, Charles Nicolle hospital, Tunis, Tunisia
| | - F Maazoul
- Congenital and Hereditary Disorders Department, Charles Nicolle hospital, Tunis, Tunisia
| | - I Lariani
- Laboratory of Human Genetics, Faculty of Medicine of Tunis, University Tunis ElManar, Tunis, Tunisia
| | - H B Chaabouni
- Laboratory of Human Genetics, Faculty of Medicine of Tunis, University Tunis ElManar, Tunis, Tunisia.,Congenital and Hereditary Disorders Department, Charles Nicolle hospital, Tunis, Tunisia
| |
Collapse
|
25
|
Chen X, Thomson H, Cooke J, Scott J, Hossain P, Lotery A. Adult limbal neurosphere cells: a potential autologous cell resource for retinal cell generation. PLoS One 2014; 9:e108418. [PMID: 25271851 PMCID: PMC4182722 DOI: 10.1371/journal.pone.0108418] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 08/27/2014] [Indexed: 01/15/2023] Open
Abstract
The Corneal limbus is a readily accessible region at the front of the eye, separating the cornea and sclera. Neural colonies (neurospheres) can be generated from adult corneal limbus invitro. We have previously shown that these neurospheres originate from neural crest stem/progenitor cells and that they can differentiate into functional neurons invitro. The aim of this study was to investigate whether mouse and human limbal neurosphere cells (LNS) could differentiate towards a retinal lineage both invivo and invitro following exposure to a developing retinal microenvironment. In this article we show that LNS can be generated from adult mice and aged humans (up to 97 years) using a serum free culture assay. Following culture with developing mouse retinal cells, we detected retinal progenitor cell markers, mature retinal/neuronal markers and sensory cilia in the majority of mouse LNS experiments. After transplantation into the sub-retinal space of neonatal mice, mouse LNS cells expressed photoreceptor specific markers, but no incorporation into host retinal tissue was seen. Human LNS cells also expressed retinal progenitor markers at the transcription level but mature retinal markers were not observed invitro or invivo. This data highlights that mouse corneal limbal stromal progenitor cells can transdifferentiate towards a retinal lineage. Complete differentiation is likely to require more comprehensive regulation; however, the accessibility and plasticity of LNS makes them an attractive cell resource for future study and ultimately therapeutic application.
Collapse
Affiliation(s)
- Xiaoli Chen
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Heather Thomson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Jessica Cooke
- School of Clinical Sciences, University of Bristol, Bristol Eye Hospital, Bristol, United Kingdom
| | - Jennifer Scott
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Parwez Hossain
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Andrew Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- * E-mail:
| |
Collapse
|
26
|
Regus-Leidig H, Fuchs M, Löhner M, Leist SR, Leal-Ortiz S, Chiodo VA, Hauswirth WW, Garner CC, Brandstätter JH. In vivo knockdown of Piccolino disrupts presynaptic ribbon morphology in mouse photoreceptor synapses. Front Cell Neurosci 2014; 8:259. [PMID: 25232303 PMCID: PMC4153300 DOI: 10.3389/fncel.2014.00259] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 08/13/2014] [Indexed: 01/08/2023] Open
Abstract
Piccolo is the largest known cytomatrix protein at active zones of chemical synapses. A growing number of studies on conventional chemical synapses assign Piccolo a role in the recruitment and integration of molecules relevant for both endo- and exocytosis of synaptic vesicles, the dynamic assembly of presynaptic F-actin, as well as the proteostasis of presynaptic proteins, yet a direct function in the structural organization of the active zone has not been uncovered in part due to the expression of multiple alternatively spliced isoforms. We recently identified Piccolino, a Piccolo splice variant specifically expressed in sensory ribbon synapses of the eye and ear. Here we down regulated Piccolino in vivo via an adeno-associated virus-based RNA interference approach and explored the impact on the presynaptic structure of mouse photoreceptor ribbon synapses. Detailed immunocytochemical light and electron microscopical analysis of Piccolino knockdown in photoreceptors revealed a hitherto undescribed photoreceptor ribbon synaptic phenotype with striking morphological changes of synaptic ribbon ultrastructure.
Collapse
Affiliation(s)
- Hanna Regus-Leidig
- Department of Biology, Animal Physiology, Friedrich-Alexander-University of Erlangen-Nuremberg Erlangen, Germany
| | - Michaela Fuchs
- Department of Biology, Animal Physiology, Friedrich-Alexander-University of Erlangen-Nuremberg Erlangen, Germany
| | - Martina Löhner
- Department of Biology, Animal Physiology, Friedrich-Alexander-University of Erlangen-Nuremberg Erlangen, Germany
| | - Sarah R Leist
- Department of Biology, Animal Physiology, Friedrich-Alexander-University of Erlangen-Nuremberg Erlangen, Germany ; Department of Infection Genetics, Helmholtz Centre for Infection Research Braunschweig, Germany
| | - Sergio Leal-Ortiz
- Department of Psychiatry and Behavioral Sciences, Stanford University Palo Alto, CA, USA
| | - Vince A Chiodo
- Department of Ophthalmology, College of Medicine, University of Florida Gainesville, FL, USA
| | - William W Hauswirth
- Department of Ophthalmology, College of Medicine, University of Florida Gainesville, FL, USA
| | - Craig C Garner
- Department of Psychiatry and Behavioral Sciences, Stanford University Palo Alto, CA, USA ; Deutsches Zentrum für Neurodegenerative Erkrankungen Charité, Berlin, Germany
| | - Johann H Brandstätter
- Department of Biology, Animal Physiology, Friedrich-Alexander-University of Erlangen-Nuremberg Erlangen, Germany
| |
Collapse
|
27
|
Liu X, Heidelberger R, Janz R. Phosphorylation of syntaxin 3B by CaMKII regulates the formation of t-SNARE complexes. Mol Cell Neurosci 2014; 60:53-62. [PMID: 24680688 DOI: 10.1016/j.mcn.2014.03.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 02/04/2014] [Accepted: 03/09/2014] [Indexed: 10/25/2022] Open
Abstract
Ribbon synapses in the retina lack the t-SNARE (target-soluble N-ethylmaleimide-sensitive factor attachment protein receptor) syntaxin 1A that is found in conventional synapses of the nervous system, but instead contain the related isoform syntaxin 3B. Previous studies have demonstrated that syntaxin 3B is essential for synaptic vesicle exocytosis in ribbon synapses, but syntaxin 3B is less efficient than syntaxin 1A in binding the t-SNARE protein SNAP-25 and catalyzing vesicle fusion. We demonstrate here that syntaxin 3B is localized mainly on the presynaptic membrane of retinal ribbon synapses and that a subset of syntaxin 3B is localized in close proximity to the synaptic ribbon. We show further, that syntaxin 3B can be phosphorylated by the Ca(2+)/calmodulin-dependent protein kinase II (CaMKII). We determine that the phosphorylation site is located close to the N-terminus at T14. Syntaxin 3B with a phosphomimetic mutation (T14E) had a stronger binding affinity for SNAP-25 compared with wild type syntaxin 3B. We propose that phosphorylation of syntaxin 3B by CaMKII can modulate the assembly of the SNARE complex in ribbon synapses of the retina, and might regulate the exocytosis of synaptic vesicles in ribbon synapses.
Collapse
Affiliation(s)
- Xiaoqin Liu
- Department of Neurobiology and Anatomy, The University of Texas Medical School, Houston, United States
| | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, The University of Texas Medical School, Houston, United States
| | - Roger Janz
- Department of Neurobiology and Anatomy, The University of Texas Medical School, Houston, United States.
| |
Collapse
|
28
|
Sherry DM, Blackburn BA. P-Rex2, a Rac-guanine nucleotide exchange factor, is expressed selectively in ribbon synaptic terminals of the mouse retina. BMC Neurosci 2013; 14:70. [PMID: 23844743 PMCID: PMC3716592 DOI: 10.1186/1471-2202-14-70] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 07/10/2013] [Indexed: 12/16/2022] Open
Abstract
Background Phosphatidylinositol (3,4,5)-trisphosphate-dependent Rac Exchanger 2 (P-Rex2) is a guanine nucleotide exchange factor (GEF) that specifically activates Rac GTPases, important regulators of actin cytoskeleton remodeling. P-Rex2 is known to modulate cerebellar Purkinje cell architecture and function, but P-Rex2 expression and function elsewhere in the central nervous system is unclear. To better understand potential roles for P-Rex2 in neuronal cytoskeletal remodeling and function, we performed widefield and confocal microscopy of specimens double immunolabeled for P-Rex2 and cell- and synapse-specific markers in the mouse retina. Results P-Rex2 was restricted to the plexiform layers of the retina and colocalized extensively with Vesicular Glutamate Transporter 1 (VGluT1), a specific marker for photoreceptor and bipolar cell terminals. Double labeling for P-Rex2 and peanut agglutinin, a cone terminal marker, confirmed that P-Rex2 was present in both rod and cone terminals. Double labeling with markers for specific bipolar cell types showed that P-Rex2 was present in the terminals of rod bipolar cells and multiple ON- and OFF-cone bipolar cell types. In contrast, P-Rex2 was not expressed in the processes or conventional synapses of amacrine or horizontal cells. Conclusions P-Rex2 is associated specifically with the glutamatergic ribbon synaptic terminals of photoreceptors and bipolar cells that transmit visual signals vertically through the retina. The Rac-GEF function of P-Rex2 implies a specific role for P-Rex2 and Rac-GTPases in regulating the actin cytoskeleton in glutamatergic ribbon synaptic terminals of retinal photoreceptors and bipolar cells and appears to be ideally positioned to modulate the adaptive plasticity of these terminals.
Collapse
Affiliation(s)
- David M Sherry
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, BMSB-553, Oklahoma City, OK 73104, USA.
| | | |
Collapse
|
29
|
Molecular anatomy and physiology of exocytosis in sensory hair cells. Cell Calcium 2012; 52:327-37. [DOI: 10.1016/j.ceca.2012.05.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 05/08/2012] [Accepted: 05/14/2012] [Indexed: 11/23/2022]
|
30
|
Vila A, Satoh H, Rangel C, Mills SL, Hoshi H, O'Brien J, Marshak DR, Macleish PR, Marshak DW. Histamine receptors of cones and horizontal cells in Old World monkey retinas. J Comp Neurol 2012; 520:528-43. [PMID: 21800315 DOI: 10.1002/cne.22731] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In primates the retina receives input from histaminergic neurons in the posterior hypothalamus that are active during the day. In order to understand how this input contributes to information processing in Old World monkey retinas, we have been localizing histamine receptors (HR) and studying the effects of histamine on the neurons that express them. Previously, we localized HR3 to the tips of ON bipolar cell dendrites and showed that histamine hyperpolarizes the cells via this receptor. We raised antisera against synthetic peptides corresponding to an extracellular domain of HR1 between the 4th and 5th transmembrane domains and to an intracellular domain near the carboxyl terminus of HR2. Using these, we localized HR1 to horizontal cells and a small number of amacrine cells and localized HR2 to puncta closely associated with synaptic ribbons inside cone pedicles. Consistent with this, HR1 mRNA was detected in horizontal cell perikarya and primary dendrites and HR2 mRNA was found in cone inner segments. We studied the effect of 5 μM exogenous histamine on primate cones in macaque retinal slices. Histamine reduced I(h) at moderately hyperpolarized potentials, but not the maximal current. This would be expected to increase the operating range of cones and conserve ATP in bright, ambient light. Thus, all three major targets of histamine are in the outer plexiform layer, but the retinopetal axons containing histamine terminate in the inner plexiform layer. Taken together, the findings in these three studies suggest that histamine acts primarily via volume transmission in primate retina.
Collapse
Affiliation(s)
- Alejandro Vila
- Department of Neurobiology and Anatomy, Medical School, University of Texas at Health Science Center at Houston, Houston, Texas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ramakrishnan NA, Drescher MJ, Drescher DG. The SNARE complex in neuronal and sensory cells. Mol Cell Neurosci 2012; 50:58-69. [PMID: 22498053 PMCID: PMC3570063 DOI: 10.1016/j.mcn.2012.03.009] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Revised: 03/23/2012] [Accepted: 03/26/2012] [Indexed: 11/28/2022] Open
Abstract
Transmitter release at synapses ensures faithful chemical coding of information that is transmitted in the sub-second time frame. The brain, the central unit of information processing, depends upon fast communication for decision making. Neuronal and neurosensory cells are equipped with the molecular machinery that responds reliably, and with high fidelity, to external stimuli. However, neuronal cells differ markedly from neurosensory cells in their signal transmission at synapses. The main difference rests in how the synaptic complex is organized, with active zones in neuronal cells and ribbon synapses in sensory cells (such as photoreceptors and hair cells). In exocytosis/neurosecretion, SNAREs (soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptors) and associated proteins play a critical role in vesicle docking, priming, fusion and synchronization of neurotransmitter release. Recent studies suggest differences between neuronal and sensory cells with respect to the molecular components of their synaptic complexes. In this review, we will cover current findings on neuronal and sensory-cell SNARE proteins and their modulators. We will also briefly discuss recent investigations on how deficits in the expression of SNARE proteins in humans impair function in brain and sense organs.
Collapse
Affiliation(s)
| | - Marian J. Drescher
- Department of Otolaryngology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Dennis G. Drescher
- Department of Otolaryngology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
32
|
The dynamic architecture of photoreceptor ribbon synapses: cytoskeletal, extracellular matrix, and intramembrane proteins. Vis Neurosci 2012; 28:453-71. [PMID: 22192503 DOI: 10.1017/s0952523811000356] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Rod and cone photoreceptors possess ribbon synapses that assist in the transmission of graded light responses to second-order bipolar and horizontal cells of the vertebrate retina. Proper functioning of the synapse requires the juxtaposition of presynaptic release sites immediately adjacent to postsynaptic receptors. In this review, we focus on the synaptic, cytoskeletal, and extracellular matrix proteins that help to organize photoreceptor ribbon synapses in the outer plexiform layer. We examine the proteins that foster the clustering of release proteins, calcium channels, and synaptic vesicles in the presynaptic terminals of photoreceptors adjacent to their postsynaptic contacts. Although many proteins interact with one another in the presynaptic terminal and synaptic cleft, these protein-protein interactions do not create a static and immutable structure. Instead, photoreceptor ribbon synapses are remarkably dynamic, exhibiting structural changes on both rapid and slow time scales.
Collapse
|
33
|
Abstract
Vision is the most important of the senses for humans, and the retina is the first stage in the processing of light signals in the visual system. In the retina, highly specialized light-sensing neurons, the rod and cone photoreceptors, convert light into neural signals. These signals are extensively processed and filtered in the subsequent retinal network before transmitted to the higher visual centres in the brain, where the perception of viewed objects and scenes is finally constructed. A key feature of signal processing in the mammalian retina is parallel processing. Visual information is segregated in parallel pathways already at the rod and cone photoreceptor terminals, which provide multiple output synapses for the faithful encoding and transfer of the visual signals to the post-receptoral retinal network. This review aims at highlighting the current knowledge about the structural and functional pre- and post-synaptic specializations of rod and cone photoreceptor ribbon synapses, which belong to the most complex chemical synapses in the central nervous system.
Collapse
Affiliation(s)
- H Regus-Leidig
- Animal Physiology, Department of Biology, University of Erlangen-Nuremberg, Germany
| | | |
Collapse
|
34
|
Karan S, Tam BM, Moritz OL, Baehr W. Targeting of mouse guanylate cyclase 1 (Gucy2e) to Xenopus laevis rod outer segments. Vision Res 2011; 51:2304-11. [PMID: 21945483 DOI: 10.1016/j.visres.2011.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 09/02/2011] [Accepted: 09/03/2011] [Indexed: 11/18/2022]
Abstract
Photoreceptor guanylate cyclase (GC1) is a transmembrane protein and responsible for synthesis of cGMP, the secondary messenger of phototransduction. It consists of an extracellular domain, a single transmembrane domain, and an intracellular domain. It is unknown how GC1 targets to the outer segments where it resides. To identify a putative GC1 targeting signal, we generated a series of peripheral membrane and transmembrane constructs encoding extracellular and intracellular mouse GC1 fragments fused to EGFP. The constructs were expressed in Xenopus laevis rod photoreceptors under the control of the rhodopsin promoter. We examined the localization of GFP-GC1 fusion proteins containing the complete GC1 sequence, or partial GC1 sequences, which were membrane-associated via either the GC1 transmembrane domain or the rhodopsin C-terminal palmitoyl chains. Full-length GFP-GC1 targeted to the rod outer segment disk rims. As a group, fusion proteins containing the entire cytoplasmic domain of GC1 targeted to the OS, whereas other fusion proteins containing portions of the cytoplasmic or the extracellular domains did not. We conclude that GC1 likely has no single linear peptide-based OS targeting signal. Our results suggest targeting is due to either multiple weak signals in the cytoplasmic domain of GC1, or co-transport to the OS with an accessory protein.
Collapse
Affiliation(s)
- Sukanya Karan
- Department of Ophthalmology, John A. Moran Eye Center, University of Utah Health Science Center, Salt Lake City, UT 84132, USA
| | | | | | | |
Collapse
|
35
|
Magharious M, D'Onofrio PM, Hollander A, Zhu P, Chen J, Koeberle PD. Quantitative iTRAQ analysis of retinal ganglion cell degeneration after optic nerve crush. J Proteome Res 2011; 10:3344-62. [PMID: 21627321 DOI: 10.1021/pr2004055] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Retinal ganglion cells (RGCs) are central nervous system (CNS) neurons that transmit visual information from the retina to the brain. Apoptotic RGC degeneration causes visual impairment that can be modeled by optic nerve crush. Neuronal apoptosis is also a salient feature of CNS trauma, ischemia (stroke), and diseases of the CNS such as Alzheimer's, Parkinson's, multiple sclerosis, and amyotrophic lateral sclerosis. Optic nerve crush induces the apoptotic cell death of ∼ 70% of RGCs within the first 14 days after injury. This model is particularly attractive for studying adult neuron apoptosis because the time-course of RGC death is well established and axon regeneration within the myelinated optic nerve can be concurrently evaluated. Here, we performed a large scale iTRAQ proteomic study to identify and quantify proteins of the rat retina at 1, 3, 4, 7, 14, and 21 days after optic nerve crush. In total, 337 proteins were identified, and 110 were differentially regulated after injury. Of these, 58 proteins were upregulated (>1.3 ×), 46 were downregulated (<0.7 ×), and 6 showed both positive and negative regulation over 21 days, relative to normal retinas. Among the differentially expressed proteins, Thymosin-β4 showed an early upregulation at 3 days, the time-point that immediately precedes the induction of RGC apoptosis after injury. We examined the effect of exogenous Thymosin-β4 administration on RGC death after optic nerve injury. Intraocular injections of Thymosin-β4 significantly increased RGC survival by ∼ 3-fold compared to controls and enhanced axon regeneration after crush, demonstrating therapeutic potential for CNS insults. Overall, our study identified numerous proteins that are differentially regulated at key time-points after optic nerve crush, and how the temporal profiles of their expression parallel RGC death. This data will aid in the future development of novel therapeutics to promote neuronal survival and regeneration in the adult CNS.
Collapse
Affiliation(s)
- Mark Magharious
- Graduate Department of Rehabilitation Science, University of Toronto, Canada
| | | | | | | | | | | |
Collapse
|
36
|
Schubert V, Bouvier D, Volterra A. SNARE protein expression in synaptic terminals and astrocytes in the adult hippocampus: a comparative analysis. Glia 2011; 59:1472-88. [PMID: 21656854 DOI: 10.1002/glia.21190] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 04/27/2011] [Indexed: 01/02/2023]
Abstract
Several evidences suggest that astrocytes release small transmitter molecules, peptides, and protein factors via regulated exocytosis, implying that they function as specialized neurosecretory cells. However, very little is known about the molecular and functional properties of regulated secretion in astrocytes in the adult brain. Establishing these properties is central to the understanding of the communication mode(s) of these cells and their role(s) in the control of synaptic functions and of cerebral blood flow. In this study, we have set-up a high-resolution confocal microscopy approach to distinguish protein expression in astrocytic structures and neighboring synaptic terminals in adult brain tissue. This approach was applied to investigate the expression pattern of core SNARE proteins for vesicle fusion in the dentate gyrus and CA1 regions of the mouse hippocampus. Our comparative analysis shows that astrocytes abundantly express, in their cell body and main processes, all three protein partners necessary to form an operational SNARE complex but not in the same isoforms expressed in neighbouring synaptic terminals. Thus, SNAP25 and VAMP2 are absent from astrocytic processes and typically concentrated in terminals, while SNAP23 and VAMP3 have the opposite expression pattern. Syntaxin 1 is present in both synaptic terminals and astrocytes. These data support the view that astrocytes in the adult hippocampus can communicate via regulated exocytosis and also indicates that astrocytic exocytosis may differ in its properties from action potential-dependent exocytosis at neuronal synapses, as it relies on a distinctive set of SNARE proteins.
Collapse
Affiliation(s)
- Vanessa Schubert
- Department of Cell Biology and Morphology, University of Lausanne, Lausanne, Switzerland
| | | | | |
Collapse
|
37
|
Hirano AA, Brandstätter JH, Morgans CW, Brecha NC. SNAP25 expression in mammalian retinal horizontal cells. J Comp Neurol 2011; 519:972-88. [PMID: 21280047 DOI: 10.1002/cne.22562] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Horizontal cells mediate inhibitory feedforward and feedback lateral interactions in the outer retina at photoreceptor terminals and bipolar cell dendrites; however, the mechanisms that underlie synaptic transmission from mammalian horizontal cells are poorly understood. The localization of a vesicular γ-aminobutyric acid (GABA) transporter (VGAT) to horizontal cell processes in primate and rodent retinae suggested that mammalian horizontal cells release transmitter in a vesicular manner. Toward determining whether the molecular machinery for vesicular transmitter release is present in horizontal cells, we investigated the expression of SNAP25 (synaptosomal-associated protein of 25 kDa), a key SNARE protein, by immunocytochemistry with cell type-specific markers in the retinae of mouse, rat, rabbit, and monkey. Different commercial antibodies to SNAP25 were tested on vertical sections of retina. We report the robust expression of SNAP25 in both plexiform layers. Double labeling with SNAP25 and calbindin antibodies demonstrated that horizontal cell processes and their endings in photoreceptor triad synapses were strongly labeled for both proteins in mouse, rat, rabbit, and monkey retinae. Double labeling with parvalbumin antibodies in monkey retina verified SNAP25 immunoreactivity in all horizontal cells. Pre-embedding immunoelectron microscopy in rabbit retina confirmed expression of SNAP25 in lateral elements within photoreceptor triad synapses. The SNAP25 immunoreactivity in the plexiform layers and outer nuclear layer fell into at least three patterns depending on the antibody, suggesting a differential distribution of SNAP25 isoforms. The presence of SNAP25a and SNAP25b isoforms in mouse retina was established by reverse transcriptase-polymerase chain reaction. SNAP25 expression in mammalian horizontal cells along with other SNARE proteins is consistent with vesicular exocytosis.
Collapse
Affiliation(s)
- Arlene A Hirano
- Department of Neurobiology, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California 90095, USA.
| | | | | | | |
Collapse
|
38
|
Lee H, Brecha NC. Immunocytochemical evidence for SNARE protein-dependent transmitter release from guinea pig horizontal cells. Eur J Neurosci 2010; 31:1388-401. [PMID: 20384779 DOI: 10.1111/j.1460-9568.2010.07181.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Horizontal cells are lateral interneurons that participate in visual processing in the outer retina but the cellular mechanisms underlying transmitter release from these cells are not fully understood. In non-mammalian horizontal cells, GABA release has been shown to occur by a non-vesicular mechanism. However, recent evidence in mammalian horizontal cells favors a vesicular mechanism as they lack plasmalemmal GABA transporters and some soluble NSF attachment protein receptor (SNARE) core proteins have been identified in rodent horizontal cells. Moreover, immunoreactivity for GABA and the molecular machinery to synthesize GABA have been found in guinea pig horizontal cells, suggesting that if components of the SNARE complex are expressed they could contribute to the vesicular release of GABA. In this study we investigated whether these vesicular and synaptic proteins are expressed by guinea pig horizontal cells using immunohistochemistry with well-characterized antibodies to evaluate their cellular distribution. Components of synaptic vesicles including vesicular GABA transporter, synapsin I and synaptic vesicle protein 2A were localized to horizontal cell processes and endings, along with the SNARE core complex proteins, syntaxin-1a, syntaxin-4 and synaptosomal-associated protein 25 (SNAP-25). Complexin I/II, a cytosolic protein that stabilizes the activated SNARE fusion core, strongly immunostained horizontal cell soma and processes. In addition, the vesicular Ca(2+)-sensor, synaptotagmin-2, which is essential for Ca(2+)-mediated vesicular release, was also localized to horizontal cell processes and somata. These morphological findings from guinea pig horizontal cells suggest that mammalian horizontal cells have the capacity to utilize a regulated Ca(2+)-dependent vesicular pathway to release neurotransmitter, and that this mechanism may be shared among many mammalian species.
Collapse
Affiliation(s)
- Helen Lee
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095-1763, USA.
| | | |
Collapse
|
39
|
Curtis L, Datta P, Liu X, Bogdanova N, Heidelberger R, Janz R. Syntaxin 3B is essential for the exocytosis of synaptic vesicles in ribbon synapses of the retina. Neuroscience 2010; 166:832-41. [PMID: 20060037 PMCID: PMC2837114 DOI: 10.1016/j.neuroscience.2009.12.075] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Revised: 12/30/2009] [Accepted: 12/30/2009] [Indexed: 10/20/2022]
Abstract
Ribbon synapses of the vertebrate retina are specialized synapses that release neurotransmitter by synaptic vesicle exocytosis in a manner that is proportional to the level of depolarization of the cell. This release property is different from conventional neurons, in which the release of neurotransmitter occurs as a short-lived burst triggered by an action potential. Synaptic vesicle exocytosis is a calcium regulated process that is dependent on a set of interacting synaptic proteins that form the so-called SNARE (soluble N-ethylmaleimide sensitive factor attachment protein receptor) complex. Syntaxin 3B has been identified as a specialized SNARE molecule in ribbon synapses of the rodent retina. However, the best physiologically-characterized neuron that forms ribbon-style synapses is the rod-dominant or Mb1 bipolar cell of the goldfish retina. We report here the molecular characterization of syntaxin 3B from the goldfish retina. Using a combination of reverse transcription (RT) polymerase chain reaction (PCR) and immunostaining with a specific antibody, we show that syntaxin 3B is highly enriched in the plasma membrane of bipolar cell synaptic terminals of the goldfish retina. Using membrane capacitance measurements we demonstrate that a peptide derived from goldfish syntaxin 3B inhibits synaptic vesicle exocytosis. These experiments demonstrate that syntaxin 3B is an important factor for synaptic vesicle exocytosis in ribbon synapses of the vertebrate retina.
Collapse
Affiliation(s)
- L Curtis
- Department of Neurobiology and Anatomy, The University of Texas Medical School, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
40
|
Honma S, Taki K, Lei S, Niwa H, Wakisaka S. Immunohistochemical Localization of SNARE Proteins in Dental Pulp and Periodontal Ligament of the Rat Incisor. Anat Rec (Hoboken) 2010; 293:1070-80. [DOI: 10.1002/ar.21106] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
41
|
Reim K, Regus-Leidig H, Ammermüller J, El-Kordi A, Radyushkin K, Ehrenreich H, Brandstätter JH, Brose N. Aberrant function and structure of retinal ribbon synapses in the absence of complexin 3 and complexin 4. J Cell Sci 2009; 122:1352-61. [PMID: 19386896 DOI: 10.1242/jcs.045401] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Complexins regulate the speed and Ca(2+) sensitivity of SNARE-mediated synaptic vesicle fusion at conventional synapses. Two of the vertebrate complexins, Cplx3 and Cplx4, are specifically localized to retinal ribbon synapses. To test whether Cplx3 and Cplx4 contribute to the highly efficient transmitter release at ribbon synapses, we studied retina function and structure in Cplx3 and Cplx4 single- and double-knockout mice. Electroretinographic recordings from single and double mutants revealed a cooperative perturbing effect of Cplx3 and Cplx4 deletion on the b-wave amplitude, whereas most other detected effects in both plexiform synaptic layers were additive. Light and electron microscopic analyses uncovered a disorganized outer plexiform layer in the retinae of mice lacking Cplx3 and Cplx4, with a significant proportion of photoreceptor terminals containing spherical free-floating ribbons. These structural and functional aberrations were accompanied by behavioural deficits indicative of a vision deficit. Our results show that Cplx3 and Cplx4 are essential regulators of transmitter release at retinal ribbon synapses. Their loss leads to aberrant adjustment and fine-tuning of transmitter release at the photoreceptor ribbon synapse, alterations in transmission at bipolar cell terminals, changes in the temporal structure of synaptic processing in the inner plexiform layer of the retina and perturbed vision.
Collapse
Affiliation(s)
- Kerstin Reim
- Department of Molecular Neurobiology and Center for the Molecular Physiology of the Brain, Max Planck Institute of Experimental Medicine, D-37075 Göttingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Aartsen WM, Arsanto JP, Chauvin JP, Vos RM, Versteeg I, Cardozo BN, Bivic AL, Wijnholds J. PSD95β regulates plasma membrane Ca(2+) pump localization at the photoreceptor synapse. Mol Cell Neurosci 2009; 41:156-65. [DOI: 10.1016/j.mcn.2009.02.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Revised: 01/27/2009] [Accepted: 02/10/2009] [Indexed: 01/05/2023] Open
|
43
|
Mazelova J, Ransom N, Astuto-Gribble L, Wilson MC, Deretic D. Syntaxin 3 and SNAP-25 pairing, regulated by omega-3 docosahexaenoic acid, controls the delivery of rhodopsin for the biogenesis of cilia-derived sensory organelles, the rod outer segments. J Cell Sci 2009; 122:2003-13. [PMID: 19454479 DOI: 10.1242/jcs.039982] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The biogenesis of cilia-derived sensory organelles, the photoreceptor rod outer segments (ROS), is mediated by rhodopsin transport carriers (RTCs). The small GTPase Rab8 regulates ciliary targeting of RTCs, but their specific fusion sites have not been characterized. Here, we report that the Sec6/8 complex, or exocyst, is a candidate effector for Rab8. We also show that the Qa-SNARE syntaxin 3 is present in the rod inner segment (RIS) plasma membrane at the base of the cilium and displays a microtubule-dependent concentration gradient, whereas the Qbc-SNARE SNAP-25 is uniformly distributed in the RIS plasma membrane and the synapse. Treatment with omega-3 docosahexaenoic acid [DHA, 22:6(n-3)] causes increased co-immunoprecipitation and colocalization of SNAP-25 and syntaxin 3 at the base of the cilium, which results in the increased delivery of membrane to the ROS. This is particularly evident in propranolol-treated retinas, in which the DHA-mediated increase in SNARE pairing overcomes the tethering block, including dissociation of Sec8 into the cytosol. Together, our data indicate that the Sec6/8 complex, syntaxin 3 and SNAP-25 regulate rhodopsin delivery, probably by mediating docking and fusion of RTCs. We show further that DHA, an essential polyunsaturated fatty acid of the ROS, increases pairing of syntaxin 3 and SNAP-25 to regulate expansion of the ciliary membrane and ROS biogenesis.
Collapse
Affiliation(s)
- Jana Mazelova
- Department of Surgery, Division of Ophthalmology, University of New Mexico, Albuquerque, New Mexico 87131, USA
| | | | | | | | | |
Collapse
|
44
|
Zanazzi G, Matthews G. The molecular architecture of ribbon presynaptic terminals. Mol Neurobiol 2009; 39:130-48. [PMID: 19253034 PMCID: PMC2701268 DOI: 10.1007/s12035-009-8058-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Accepted: 02/04/2009] [Indexed: 12/24/2022]
Abstract
The primary receptor neurons of the auditory, vestibular, and visual systems encode a broad range of sensory information by modulating the tonic release of the neurotransmitter glutamate in response to graded changes in membrane potential. The output synapses of these neurons are marked by structures called synaptic ribbons, which tether a pool of releasable synaptic vesicles at the active zone where glutamate release occurs in response to calcium influx through L-type channels. Ribbons are composed primarily of the protein, RIBEYE, which is unique to ribbon synapses, but cytomatrix proteins that regulate the vesicle cycle in conventional terminals, such as Piccolo and Bassoon, also are found at ribbons. Conventional and ribbon terminals differ, however, in the size, molecular composition, and mobilization of their synaptic vesicle pools. Calcium-binding proteins and plasma membrane calcium pumps, together with endomembrane pumps and channels, play important roles in calcium handling at ribbon synapses. Taken together, emerging evidence suggests that several molecular and cellular specializations work in concert to support the sustained exocytosis of glutamate that is a hallmark of ribbon synapses. Consistent with its functional importance, abnormalities in a variety of functional aspects of the ribbon presynaptic terminal underlie several forms of auditory neuropathy and retinopathy.
Collapse
Affiliation(s)
- George Zanazzi
- Department of Neurobiology & Behavior, State Universtiy of New York, Stony Brook, NY 11794-5230, USA
| | | |
Collapse
|
45
|
Baker SA, Haeri M, Yoo P, Gospe SM, Skiba NP, Knox BE, Arshavsky VY. The outer segment serves as a default destination for the trafficking of membrane proteins in photoreceptors. ACTA ACUST UNITED AC 2008; 183:485-98. [PMID: 18981232 PMCID: PMC2575789 DOI: 10.1083/jcb.200806009] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Photoreceptors are compartmentalized neurons in which all proteins responsible for evoking visual signals are confined to the outer segment. Yet, the mechanisms responsible for establishing and maintaining photoreceptor compartmentalization are poorly understood. Here we investigated the targeting of two related membrane proteins, R9AP and syntaxin 3, one residing within and the other excluded from the outer segment. Surprisingly, we have found that only syntaxin 3 has targeting information encoded in its sequence and its removal redirects this protein to the outer segment. Furthermore, proteins residing in the endoplasmic reticulum and mitochondria were similarly redirected to the outer segment after removing their targeting signals. This reveals a pattern where membrane proteins lacking specific targeting information are delivered to the outer segment, which is likely to reflect the enormous appetite of this organelle for new material necessitated by its constant renewal. This also implies that every protein residing outside the outer segment must have a means to avoid this "default" trafficking flow.
Collapse
Affiliation(s)
- Sheila A Baker
- Albert Eye Research Institute, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Curtis LB, Doneske B, Liu X, Thaller C, McNew JA, Janz R. Syntaxin 3b is a t-SNARE specific for ribbon synapses of the retina. J Comp Neurol 2008; 510:550-9. [PMID: 18683220 PMCID: PMC2893228 DOI: 10.1002/cne.21806] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Previous studies have demonstrated that ribbon synapses in the retina do not contain the t-SNARE (target-soluble N-ethylmaleimide-sensitive factor attachment protein receptor) syntaxin 1A that is found in conventional synapses of the nervous system. In contrast, ribbon synapses of the retina contain the related isoform syntaxin 3. In addition to its localization in ribbon synapses, syntaxin 3 is also found in nonneuronal cells, where it has been implicated in the trafficking of transport vesicles to the apical plasma membrane of polarized cells. The syntaxin 3 gene codes for four different splice forms, syntaxins 3A, 3B, 3C, and 3D. We demonstrate here by using analysis of EST databases, RT-PCR, in situ hybridization, and Northern blot analysis that cells in the mouse retina express only syntaxin 3B. In contrast, nonneuronal tissues, such as kidney, express only syntaxin 3A. The two major syntaxin isoforms (3A and 3B) have an identical N-terminal domain but differ in the C-terminal half of the SNARE domain and the C-terminal transmembrane domain. These two domains are thought to be directly involved in synaptic vesicle fusion. The interaction of syntaxin 1A and syntaxin 3B with other synaptic proteins was examined. We found that both proteins bind Munc18/N-sec1 with similar affinity. In contrast, syntaxin 3B had a much lower binding affinity for the t-SNARE SNAP25 compared with syntaxin 1A. By using an in vitro fusion assay, we could demonstrate that vesicles containing syntaxin 3B and SNAP25 could fuse with vesicles containing synaptobrevin2/VAMP2, demonstrating that syntaxin 3B can function as a t-SNARE.
Collapse
Affiliation(s)
- Leigh B Curtis
- Department of Neurobiology and Anatomy, The University of Texas Medical School, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
47
|
Witkovsky P, Gábriel R, Krizaj D. Anatomical and neurochemical characterization of dopaminergic interplexiform processes in mouse and rat retinas. J Comp Neurol 2008; 510:158-74. [PMID: 18615559 DOI: 10.1002/cne.21784] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Dopaminergic (DA) neurons of mouse and rat retinas are of the interplexiform subtype (DA-IPC), i.e., they send processes distally toward the outer retina, exhibiting numerous varicosities along their course. The primary question we addressed was whether distally located DA-IPC varicosities, identified by tyrosine hydroxylase (TH) immunoreactivity, had the characteristic presynaptic proteins associated with calcium-dependent vesicular release of neurotransmitter. We found that TH immunoreactive varicosities in the outer retina possessed vesicular monoamine transporter 2 and vesicular GABA transporter, but they lacked immunostaining for any of nine subtypes of voltage-dependent calcium channel. Immunoreactivity for other channels that may permit calcium influx such as certain ionotropic glutamate receptors and canonical transient receptor potential channels (TRPCs) was similarly absent, although DA-IPC varicosities did show ryanodine receptor immunoreactivity, indicating the presence of intracellular calcium stores. The synaptic vesicle proteins sv2a and sv2b and certain other proteins associated with the presynaptic membrane were absent from DA-IPC varicosities, but the vesicular SNARE protein, vamp2, was present in a fraction of those varicosities. We identified a presumed second class of IPC that is GABAergic but not dopaminergic. Outer retinal varicosities of this putative GABAergic IPC did colocalize synaptic vesicle protein 2a, suggesting they possessed a conventional vesicular release mechanism.
Collapse
Affiliation(s)
- Paul Witkovsky
- Department of Ophthalmology, New York University School of Medicine, New York, New York 10016, USA.
| | | | | |
Collapse
|
48
|
Cohen R, Marom M, Atlas D. Depolarization-evoked secretion requires two vicinal transmembrane cysteines of syntaxin 1A. PLoS One 2007; 2:e1273. [PMID: 18060067 PMCID: PMC2094736 DOI: 10.1371/journal.pone.0001273] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2007] [Accepted: 11/14/2007] [Indexed: 11/24/2022] Open
Abstract
Background The interactions of the voltage-gated Ca2+ channel (VGCC) with syntaxin 1A (Sx 1A), Synaptosome-associated protein of 25 kD (SNAP-25), and synaptotagmin, couple electrical excitation to evoked secretion. Two vicinal Cys residues, Cys 271 and Cys 272 in the Sx 1A transmembrane domain, are highly conserved and participate in modulating channel kinetics. Each of the Sx1A Cys mutants, differently modify the kinetics of Cav1.2, and neuronal Cav2.2 calcium channel. Methodology/Principle Findings We examined the effects of various Sx1A Cys mutants and the syntaxin isoforms 2, 3, and 4 each of which lack vicinal Cys residues, on evoked secretion, monitoring capacitance transients in a functional release assay. Membrane capacitance in Xenopus oocytes co-expressing Cav1.2, Sx1A, SNAP-25 and synaptotagmin, which is Bot C- and Bot A-sensitive, was elicited by a double 500 ms depolarizing pulse to 0 mV. The evoked-release was obliterated when a single Cys Sx1A mutant or either one of the Sx isoforms were substituted for Sx 1A, demonstrating the essential role of vicinal Cys residues in the depolarization mediated process. Protein expression and confocal imaging established the level of the mutated proteins in the cell and their targeting to the plasma membrane. Conclusions/Significance We propose a model whereby the two adjacent transmembranal Cys residues of Sx 1A, lash two calcium channels. Consistent with the necessity of a minimal fusion complex termed the excitosome, each Sx1A is in a complex with SNAP-25, Syt1, and the Ca2+ channel. A Hill coefficient >2 imply that at least three excitosome complexes are required for generating a secreting hetero-oligomer protein complex. This working model suggests that a fusion pore that opens during membrane depolarization could be lined by alternating transmembrane segments of Sx1A and VGCC. The functional coupling of distinct amino acids of Sx 1A with VGCC appears to be essential for depolarization-evoked secretion.
Collapse
Affiliation(s)
- Roy Cohen
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Merav Marom
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Daphne Atlas
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
49
|
Smith JD, Greenlee JJ, Hamir AN, West Greenlee MH. Retinal cell types are differentially affected in sheep with scrapie. J Comp Pathol 2007; 138:12-22. [PMID: 18061608 DOI: 10.1016/j.jcpa.2007.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2006] [Accepted: 09/15/2007] [Indexed: 10/22/2022]
Abstract
Transmissible spongiform encephalopathies (TSEs) are a group of fatal neurodegenerative diseases characterized microscopically by spongiform lesions (vacuolation) in the neuropil, neuronal loss, and gliosis. Accumulation of the abnormal form of the prion protein (PrP(Sc)) has been demonstrated in the retina of natural and non-natural TSE-affected hosts, with or without evidence of microscopically detectable retinal pathology. This study was conducted to investigate the effect of PrP(Sc) accumulation on retinal neurons in a natural host lacking overt microscopical evidence of retinal degeneration by comparing the distribution of retinal cell type-specific markers in control and scrapie-affected sheep. In retinas with PrP(Sc)-immunoreactivity, there was disruption of the normal immunoreactivity patterns of the alpha isoform of protein kinase C (PKCalpha) and vesicular glutamate transporter 1 (VGLUT1), markers of retinal bipolar cells. Altered immunoreactivity was also observed for microtubule-associated protein 2 (MAP2), a marker of a subset of retinal ganglion cells, and glutamine synthetase (GS), a marker of Müller glia. These results demonstrate alterations of immunoreactivity patterns for proteins associated with specific cell types in retinas with PrP(Sc) accumulation, despite an absence of microscopical evidence of retinal degeneration.
Collapse
Affiliation(s)
- J D Smith
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50010, USA
| | | | | | | |
Collapse
|
50
|
Thoreson WB. Kinetics of synaptic transmission at ribbon synapses of rods and cones. Mol Neurobiol 2007; 36:205-23. [PMID: 17955196 PMCID: PMC2474471 DOI: 10.1007/s12035-007-0019-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2007] [Accepted: 05/18/2007] [Indexed: 11/24/2022]
Abstract
The ribbon synapse is a specialized structure that allows photoreceptors to sustain the continuous release of vesicles for hours upon hours and years upon years but also respond rapidly to momentary changes in illumination. Light responses of cones are faster than those of rods and, mirroring this difference, synaptic transmission from cones is also faster than transmission from rods. This review evaluates the various factors that regulate synaptic kinetics and contribute to kinetic differences between rod and cone synapses. Presynaptically, the release of glutamate-laden synaptic vesicles is regulated by properties of the synaptic proteins involved in exocytosis, influx of calcium through calcium channels, calcium release from intracellular stores, diffusion of calcium to the release site, calcium buffering, and extrusion of calcium from the cytoplasm. The rate of vesicle replenishment also limits the ability of the synapse to follow changes in release. Post-synaptic factors include properties of glutamate receptors, dynamics of glutamate diffusion through the cleft, and glutamate uptake by glutamate transporters. Thus, multiple synaptic mechanisms help to shape the responses of second-order horizontal and bipolar cells.
Collapse
Affiliation(s)
- Wallace B Thoreson
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, 4th floor, Durham Research Center, 985840 Nebraska Medical Center, Omaha, NE 68198-5840, USA.
| |
Collapse
|