1
|
Wang F, Zhong W, Yang Q, Zhao W, Liu X, Rao B, Lin X, Zhang J. Distribution and synaptic organization of substance P-like immunoreactive neurons in the mouse retina. Brain Struct Funct 2023; 228:1703-1724. [PMID: 37481742 DOI: 10.1007/s00429-023-02688-x] [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: 01/17/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
Substance P (SP), a neuroprotective peptidergic neurotransmitter, is known to have immunoreactivity (IR) localized to amacrine and/or ganglion cells in a variety of species' retinas, but it has not yet been studied in the mouse retina. Thus, we investigated the distribution and synaptic organization of SP-IR by confocal and electron microscopy immunocytochemistry in the mouse retina. SP-IR was distributed in the inner nuclear layer (INL), inner plexiform layer (IPL), and ganglion cell layer (GCL). Most of the SP-IR somas belonged to amacrine cells (2.5% of all) in the INL and their processes stratified into the S1, S3, and S5 layers of the IPL, with the most intense band in the S5 layer. Some SP-IR somas can also be observed in the GCL, which were identified as displaced amacrine cells (82%, 1269/1550) and ganglion cells (18%, 281/1550) by antibodies against AP2α and RBPMS, respectively. Such SP-IR ganglion cells (1.2% of all RGCs) can be further divided into 3 subgroups expressing SP/α-Synuclein (α-Syn), SP/GAD67, and/or SP/GAD67/α-Syn. Possible physiological and pathological roles of these ganglion cells are discussed. Further, electron microscopy evidence demonstrates that SP-IR amacrine cells receive major inputs from other SP-IR amacrine cell processes (146/242 inputs) and output mostly to SP-negative amacrine cell processes (291/673 outputs), suggesting series inhibition among amacrine cells. These results reveal for the first time an explicit distribution, novel ganglion cell features, and synaptic organization of SP-IR in the mouse retina, which is important for the future use of mouse models to study the roles of SP in healthy and diseased (including Parkinson's disease) retinal states.
Collapse
Affiliation(s)
- Fenglan Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenhui Zhong
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qingwen Yang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenna Zhao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaoqing Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Bilin Rao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Xin Lin
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Jun Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
2
|
Liu Y, Savier EL, DePiero VJ, Chen C, Schwalbe DC, Abraham-Fan RJ, Chen H, Campbell JN, Cang J. Mapping visual functions onto molecular cell types in the mouse superior colliculus. Neuron 2023; 111:1876-1886.e5. [PMID: 37086721 PMCID: PMC10330256 DOI: 10.1016/j.neuron.2023.03.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 04/24/2023]
Abstract
The superficial superior colliculus (sSC) carries out diverse roles in visual processing and behaviors, but how these functions are delegated among collicular neurons remains unclear. Here, using single-cell transcriptomics, we identified 28 neuron subtypes and subtype-enriched marker genes from tens of thousands of adult mouse sSC neurons. We then asked whether the sSC's molecular subtypes are tuned to different visual stimuli. Specifically, we imaged calcium dynamics in single sSC neurons in vivo during visual stimulation and then mapped marker gene transcripts onto the same neurons ex vivo. Our results identify a molecular subtype of inhibitory neuron accounting for ∼50% of the sSC's direction-selective cells, suggesting a genetic logic for the functional organization of the sSC. In addition, our studies provide a comprehensive molecular atlas of sSC neuron subtypes and a multimodal mapping method that will facilitate investigation of their respective functions, connectivity, and development.
Collapse
Affiliation(s)
- Yuanming Liu
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Elise L Savier
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Victor J DePiero
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Chen Chen
- Department of Psychology, University of Virginia, Charlottesville, VA 22904, USA
| | - Dana C Schwalbe
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | | | - Hui Chen
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - John N Campbell
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA.
| | - Jianhua Cang
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA; Department of Psychology, University of Virginia, Charlottesville, VA 22904, USA.
| |
Collapse
|
3
|
Yang Q, Lin X, Xiao J, Zhong W, Wang F, Tan H, Rao B, Qu J, Zhang J. Expression of α‐Synuclein in the mouse retina is confined to inhibitory presynaptic elements. J Comp Neurol 2023; 531:1057-1079. [PMID: 37002599 DOI: 10.1002/cne.25481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/18/2023] [Accepted: 03/13/2023] [Indexed: 04/03/2023]
Abstract
α-Synuclein (α-Syn) is enriched in presynaptic terminals of the central nervous system including the retina and plays a role in the synaptic vesicle cycle and synaptic transmission. Abnormal aggregation of α-Syn is considered to be the main component of the Lewy bodies that are the pathological hallmarks of Parkinson's disease. Although expression pattern of α-Syn has been described in the retinas, its precise cellular and subcellular locations are poorly understood. We investigated the precise expression of α-Syn using light microscopy (LM) and electron microscopy (EM) with antibodies against α-Syn in the mouse retina. We found that the majority of α-Syn immunoreactivity (IR) is located in GABAergic, glycinergic, and dopaminergic amacrine cells, and their processes often make a direct synapse to other labeled or unlabeled amacrine profiles, bipolar cell terminals, or ganglion cell dendrites. Further, our LM and immuno-EM results confirm the absence of α-Syn in excitatory photoreceptors, bipolar cell bodies, and their ribbon synapses, providing evidence, for the first time, that ribbon synapses do not express α-Syn. Additionally, α-Syn IR is located in the ganglion cells, some of which are intrinsically photosensitive retinal ganglion cells. These results reveal a previously unappreciated inhibitory synapse-specific expression pattern of α-Syn in the retina, suggesting that α-Syn may play a distinct role in the modulation and integration of inhibitory synaptic transmission in the retina.
Collapse
Affiliation(s)
- Qingwen Yang
- Laboratory of Retinal Physiology and Disease School of Ophthalmology and Optometry and Eye Hospital Wenzhou Medical University Wenzhou China
| | - Xin Lin
- Laboratory of Retinal Physiology and Disease School of Ophthalmology and Optometry and Eye Hospital Wenzhou Medical University Wenzhou China
| | - Jiayi Xiao
- Laboratory of Retinal Physiology and Disease School of Ophthalmology and Optometry and Eye Hospital Wenzhou Medical University Wenzhou China
| | - Wenhui Zhong
- Laboratory of Retinal Physiology and Disease School of Ophthalmology and Optometry and Eye Hospital Wenzhou Medical University Wenzhou China
| | - Fenglan Wang
- Laboratory of Retinal Physiology and Disease School of Ophthalmology and Optometry and Eye Hospital Wenzhou Medical University Wenzhou China
| | - Hang Tan
- Laboratory of Retinal Physiology and Disease School of Ophthalmology and Optometry and Eye Hospital Wenzhou Medical University Wenzhou China
| | - Bilin Rao
- Laboratory of Retinal Physiology and Disease School of Ophthalmology and Optometry and Eye Hospital Wenzhou Medical University Wenzhou China
| | - Jia Qu
- State Key Laboratory of Ophthalmology Optometry and Vision Science, Eye Hospital, Wenzhou Medical University Wenzhou China
- National Clinical Research Center for Ocular Diseases Eye Hospital Wenzhou Medical University Wenzhou China
| | - Jun Zhang
- Laboratory of Retinal Physiology and Disease School of Ophthalmology and Optometry and Eye Hospital Wenzhou Medical University Wenzhou China
- State Key Laboratory of Ophthalmology Optometry and Vision Science, Eye Hospital, Wenzhou Medical University Wenzhou China
- National Clinical Research Center for Ocular Diseases Eye Hospital Wenzhou Medical University Wenzhou China
| |
Collapse
|
4
|
Chen AM, Azar SS, Harris A, Brecha NC, Pérez de Sevilla Müller L. PTEN Expression Regulates Gap Junction Connectivity in the Retina. Front Neuroanat 2021; 15:629244. [PMID: 34093139 PMCID: PMC8172595 DOI: 10.3389/fnana.2021.629244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/26/2021] [Indexed: 12/16/2022] Open
Abstract
Manipulation of the phosphatase and tensin homolog (PTEN) pathway has been suggested as a therapeutic approach to treat or prevent vision loss due to retinal disease. In this study, we investigated the effects of deleting one copy of Pten in a well-characterized class of retinal ganglion cells called α-ganglion cells in the mouse retina. In Pten +/- retinas, α-ganglion cells did not exhibit major changes in their dendritic structure, although most cells developed a few, unusual loop-forming dendrites. By contrast, α-ganglion cells exhibited a significant decrease in heterologous and homologous gap junction mediated cell coupling with other retinal ganglion and amacrine cells. Additionally, the majority of OFF α-ganglion cells (12/18 cells) formed novel coupling to displaced amacrine cells. The number of connexin36 puncta, the predominant connexin that mediates gap junction communication at electrical synapses, was decreased by at least 50% on OFF α-ganglion cells. Reduced and incorrect gap junction connectivity of α-ganglion cells will affect their functional properties and alter visual image processing in the retina. The anomalous connectivity of retinal ganglion cells would potentially limit future therapeutic approaches involving manipulation of the Pten pathway for treating ganglion cell degeneration in diseases like glaucoma, traumatic brain injury, Parkinson's, and Alzheimer's diseases.
Collapse
Affiliation(s)
- Ashley M. Chen
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shaghauyegh S. Azar
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alexander Harris
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nicholas C. Brecha
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
- Stein Eye Institute, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
- CURE Digestive Diseases Research Center, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Administration Greater Los Angeles Health System, Los Angeles, CA, United States
| | - Luis Pérez de Sevilla Müller
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
5
|
OTX2 Non-Cell Autonomous Activity Regulates Inner Retinal Function. eNeuro 2020; 7:ENEURO.0012-19.2020. [PMID: 32737182 PMCID: PMC7477954 DOI: 10.1523/eneuro.0012-19.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/19/2020] [Accepted: 05/05/2020] [Indexed: 12/11/2022] Open
Abstract
OTX2 is a homeoprotein transcription factor expressed in photoreceptors and bipolar cells in the retina. OTX2, like many other homeoproteins, transfers between cells and exerts non-cell autonomous effects such as promoting the survival of retinal ganglion cells that do not express the protein. Here we used a genetic approach to target extracellular OTX2 in the retina by conditional expression of a secreted single-chain anti-OTX2 antibody. Compared with control mice, the expression of this antibody by parvalbumin-expressing neurons in the retina is followed by a reduction in visual acuity in 1-month-old mice with no alteration of the retinal structure or cell type number or aspect. The a-waves and b-waves measured by electroretinogram were also indistinguishable from those of control mice, suggesting no functional deficit of photoreceptors and bipolar cells. Mice expressing the OTX2-neutralizing antibody did show a significant doubling in the flicker amplitude and a reduction in oscillatory potential, consistent with a change in inner retinal function. Our results show that interfering in vivo with OTX2 non-cell autonomous activity in the postnatal retina leads to an alteration in inner retinal cell functions and causes a deficit in visual acuity.
Collapse
|
6
|
Jiang D, Burger CA, Casasent A, Albrecht NE, Li F, Samuel MA. Spatiotemporal gene expression patterns reveal molecular relatedness between retinal laminae. J Comp Neurol 2020; 528:729-755. [PMID: 31609468 PMCID: PMC7147688 DOI: 10.1002/cne.24784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/28/2019] [Accepted: 09/13/2019] [Indexed: 12/16/2022]
Abstract
In several areas of the central nervous system, neurons are regionally organized into groups or layers that carry out specific activities. In this form of patterning, neurons of distinct types localize their cell bodies to just one or a few of the layers within a structure. However, little is known about whether diverse neuron types within a lamina share molecular features that coordinate their organization. To begin to identify such candidates, we used the laminated murine retina to screen 92 lacZ reporter lines available through the Knockout Mouse Project. Thirty-two of these displayed reporter expression in restricted subsets of inner retina neurons. We then identified the spatiotemporal expression patterns of these genes at key developmental stages. This uncovered several that were heavily enriched in development but reduced in adulthood, including the transcriptional regulator Hmga1. An additional set of genes displayed maturation associated laminar enrichment. Among these, we identified Bbox1 as a novel gene that specifically labels all neurons in the ganglion cell layer but is largely excluded from otherwise molecularly similar neurons in the inner retina. Finally, we established Dbn1 as a new marker enriched in amacrines and Fmnl3 as a marker for subsets of αRGCs. Together, these data provide a spatiotemporal map for laminae-specific molecules and suggest that diverse neuron types within a lamina share coordinating molecular features that may inform their fate or function.
Collapse
Affiliation(s)
- Danye Jiang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030
| | - Courtney A. Burger
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030
| | - Anna Casasent
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030
| | - Nicholas E. Albrecht
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030
| | - Fenge Li
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030
| | - Melanie A. Samuel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
7
|
Cui LJ, Chen WH, Liu AL, Han X, Jiang SX, Yuan F, Zhong YM, Yang XL, Weng SJ. nGnG Amacrine Cells and Brn3b-negative M1 ipRGCs are Specifically Labeled in the ChAT-ChR2-EYFP Mouse. Invest Ophthalmol Vis Sci 2020; 61:14. [PMID: 32049344 PMCID: PMC7326507 DOI: 10.1167/iovs.61.2.14] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Experimental access to specific cell subtypes is essential for deciphering the complexity of retinal networks. Here, we characterized the selective labeling, caused by ectopic transgene expression, of two atypical retinal neurons in the ChAT-Channelrhodopsin-2 (ChR2)-EYFP mouse. Methods Retinal sections and flat-mounts were prepared for double-staining immunohistochemistry with antibodies against EYFP and various neuronal markers. Sagittal/coronal brain slices were made to visualize EYFP signals in central nuclei. Whole-cell recordings were conducted to test the functionality of ChR2. Results Two populations of EYFP-positive retinal cells were observed. The inner nuclear layer (INL)-located one (type I cell) distributed regularly throughout the entire retina, whereas the ganglion cell layer (GCL)-residing one (type II cell) was restricted ventrally. None of them was cholinergic, as evidenced by the complete absence of ChAT immunoreactivity. Type I cells were immunolabeled by the amacrine marker syntaxin. However, the vast majority of them were neither positive to GABA/GAD65, nor to GlyT1/glycine, suggesting that they were non-GABAergic non-glycinergic amacrine cells (nGnG ACs), which was confirmed by double-labeling with the nGnG AC marker PPP1R17. Type II cells were immunopositive to melanopsin, but not to Brn3a or Brn3b. They possessed dendrites stratifying in the outermost inner plexiform layer (IPL) and axons projecting to the suprachiasmatic nucleus (SCN) rather than the olivary pretectal nucleus (OPN), suggesting that they belonged to a Brn3b-negative subset of M1-type intrinsically photosensitive retinal ganglion cells (ipRGCs). Glutamatergic transmission-independent photocurrents were elicited in EYFP-positive cells, indicating the functional expression of ChR2. Conclusions The ChAT-ChR2-EYFP retina exhibits ectopic, but functional, transgene expression in nGnG ACs and SCN-innervating M1 ipRGCs, thus providing an ideal tool to achieve efficient labeling and optogenetic manipulation of these cells.
Collapse
|
8
|
Bahr HI, Abdelghany AA, Galhom RA, Barakat BM, Arafa ESA, Fawzy MS. Duloxetine protects against experimental diabetic retinopathy in mice through retinal GFAP downregulation and modulation of neurotrophic factors. Exp Eye Res 2019; 186:107742. [PMID: 31344388 DOI: 10.1016/j.exer.2019.107742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 07/19/2019] [Accepted: 07/19/2019] [Indexed: 12/23/2022]
Abstract
Diabetic retinopathy (DR) is recognized as one of the leading causes of blindness worldwide. Searching and validation for a novel therapeutic strategy to prevent its progress are promising. This work aimed to assess the retinal protective effects of duloxetine (DLX) in Alloxan-induced diabetic mice model. Animals were equally and randomly divided to four groups (eight mice per group); group 1: is the control group, 2: diabetic group, 3&4: diabetic and after 9 weeks received DLX for 4 weeks (15 mg/kg and 30 mg/kg), respectively. Quantitative real-time PCR (qPCR) analysis revealed nerve growth factor (NGF), inducible nitric oxide synthase (iNOS) and transforming growth factor beta (TGF-β) genes upregulation in the diabetic group compared to controls. Also, increased retinal malondialdehyde (MDA) and the decline of reduced glutathione (GSH) levels were observed. The morphometric analysis of diabetic retina revealed a significant reduction in total retinal thickness compared to control. Diabetic retinal immunostaining and Western blot analyses displayed glial fibrillary acidic protein (GFAP) and vascular endothelial cell growth factor (VEGF) proteins expression upregulation as well as glucose transporter-1 (GLUT-1) downregulation comparing to controls. However, DLX-treated groups showed downregulated NGF, iNOS, and TGF-β that was more obviously seen in the DLX-30 mg/kg group than DLX-15 mg/kg group. Furthermore, these groups showed amelioration of the oxidative markers; MDA and GSH, retaining the total retinal thickness nearly to control, GFAP and VEGF downregulation, and GLUT-1 upregulation compared to diabetic group. Taken together, it could be summarized that duloxetine can attenuate DR via the anti-inflammatory and the anti-oxidative properties as well as modulating the angiogenic and the neurotrophic factors expressions. This could hopefully pave the road to be included in the novel list of the therapeutic regimen for DR after validation in the clinic.
Collapse
Affiliation(s)
- Hoda I Bahr
- Department of Biochemistry, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| | - Ahmed A Abdelghany
- Department of Ophthalmology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| | - Rania A Galhom
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| | - Bassant M Barakat
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt; Department of Clinical Pharmacy, College of Clinical Pharmacy, Al-Baha University, Al-Baha, Saudi Arabia.
| | - El-Shaimaa A Arafa
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, 346, United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Manal S Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia; Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
9
|
Ghinia MG, Novelli E, Sajgo S, Badea TC, Strettoi E. Brn3a and Brn3b knockout mice display unvaried retinal fine structure despite major morphological and numerical alterations of ganglion cells. J Comp Neurol 2019; 527:187-211. [PMID: 27391320 PMCID: PMC5219957 DOI: 10.1002/cne.24072] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/07/2016] [Accepted: 06/30/2016] [Indexed: 01/21/2023]
Abstract
Ganglion cells (GCs), the retinal output neurons, receive synaptic inputs from bipolar and amacrine cells in the inner plexiform layer (IPL) and send information to the brain nuclei via the optic nerve. Although GCs constitute less than 1% of the total retinal cells, they occur in numerous types and are the first neurons formed during retinal development. Using Brn3a and Brn3b mutant mice in which the alkaline phosphatase gene was knocked-in (Badea et al. [Neuron] 2009;61:852-864; Badea and Nathans [Vision Res] 2011;51:269-279), we studied the general effects after gene removal on the retinal neuropil together with the consequences of lack of development of large numbers of GCs onto the remaining retinal neurons of the same class. We analyzed the morphology, number, and general architecture of various neuronal types presynaptic to GCs, searching for changes secondary to the decrement in the number of their postsynaptic partners, as well as the morphology and distribution of retinal astrocytes, for their strong topographical relation to GCs. We found that, despite GC losses, retinal organization in Brn3 null mice is remarkably similar to that of wild-type controls. J. Comp. Neurol. 527:187-211, 2019. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Miruna Georgiana Ghinia
- Neuroscience Institute of the Italian National Research Council, Pisa Research Campus, 56124 Pisa, Italy
- Retinal CIrcuit Development & Genetics Unit, Neurobiology–Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892
- Babeş Bolyai University, 400084 Cluj Napoca, Romania
| | - Elena Novelli
- Neuroscience Institute of the Italian National Research Council, Pisa Research Campus, 56124 Pisa, Italy
| | - Szilard Sajgo
- Retinal CIrcuit Development & Genetics Unit, Neurobiology–Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Tudor Constantin Badea
- Retinal CIrcuit Development & Genetics Unit, Neurobiology–Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Enrica Strettoi
- Neuroscience Institute of the Italian National Research Council, Pisa Research Campus, 56124 Pisa, Italy
| |
Collapse
|
10
|
Abstract
The expression of serotonin (5-HT) in the retina was first reported in the sixties. The detection of vesicular monoamine transporter and serotonin receptors in several retinal cells confirm that 5-HT is playing a neuromodulatory role in this structure. Whereas signaling pathways activated by 5-HT receptor binding has been poorly investigated so far, numerous data demonstrated that 5-HT is involved in retinal physiology, retinal physiopathology and photoreceptor survival.
Collapse
|
11
|
Laboulaye MA, Duan X, Qiao M, Whitney IE, Sanes JR. Mapping Transgene Insertion Sites Reveals Complex Interactions Between Mouse Transgenes and Neighboring Endogenous Genes. Front Mol Neurosci 2018; 11:385. [PMID: 30405348 PMCID: PMC6206269 DOI: 10.3389/fnmol.2018.00385] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/25/2018] [Indexed: 11/13/2022] Open
Abstract
Transgenic mouse lines are routinely employed to label and manipulate distinct cell types. The transgene generally comprises cell-type specific regulatory elements linked to a cDNA encoding a reporter or other protein. However, off-target expression seemingly unrelated to the regulatory elements in the transgene is often observed, it is sometimes suspected to reflect influences related to the site of transgene integration in the genome. To test this hypothesis, we used a proximity ligation-based method, Targeted Locus Amplification (TLA), to map the insertion sites of three well-characterized transgenes that appeared to exhibit insertion site-dependent expression in retina. The nearest endogenous genes to transgenes HB9-GFP, Mito-P, and TYW3 are Cdh6, Fat4 and Khdrbs2, respectively. For two lines, we demonstrate that expression reflects that of the closest endogenous gene (Fat4 and Cdh6), even though the distance between transgene and endogenous gene is 550 and 680 kb, respectively. In all three lines, the transgenes decrease expression of the neighboring endogenous genes. In each case, the affected endogenous gene was expressed in at least some of the cell types that the transgenic line has been used to mark and study. These results provide insights into the effects of transgenes and endogenous genes on each other's expression, demonstrate that mapping insertion site is valuable for interpreting results obtained with transgenic lines, and indicate that TLA is a reliable method for integration site discovery.
Collapse
Affiliation(s)
| | | | | | | | - Joshua R. Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, United States
| |
Collapse
|
12
|
Jo A, Xu J, Deniz S, Cherian S, DeVries SH, Zhu Y. Intersectional Strategies for Targeting Amacrine and Ganglion Cell Types in the Mouse Retina. Front Neural Circuits 2018; 12:66. [PMID: 30186122 PMCID: PMC6113359 DOI: 10.3389/fncir.2018.00066] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/02/2018] [Indexed: 12/02/2022] Open
Abstract
The mammalian retina harbors over 100 different cell types. To understand how retinal circuits work, it is essential to systematically access each type. A widely used approach for achieving targeted transgene expression exploits promoter-driven Cre lines. However, Cre expression in a given transgenic line in the retina and elsewhere in the brain is rarely confined to a single cell type, contributing ambiguity to the interpretation of results from broadly applied manipulations. To obtain unambiguous information about retinal processing, it is desirable to have strategies for further restricting transgene expression to a few or even to a single cell type. We employed an intersectional strategy based on a Cre/Flp double recombinase system to target amacrine and ganglion cell types in the inner retina. We analyzed expression patterns in seven Flp drivers and then created combinational mouse lines by selective cross breeding with Cre drivers. Breeding with Flp drivers can routinely remove labeling from more than 90% of the cells in Cre drivers, leading to only a handful cell types, typically 2–3, remaining in the intersection. Cre/Flp combinatorial mouse lines enabled us to identify and anatomically characterize retinal cell types with greater ease and demonstrated the feasibility of intersectional strategies in retinal research. In addition to the retina, we examined Flp expression in the lateral geniculate nucleus and superior colliculus. Our results establish a foundation for future application of intersectional strategies in the retina and retino-recipient regions.
Collapse
Affiliation(s)
- Andrew Jo
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jian Xu
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Sercan Deniz
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Suraj Cherian
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Steven H DeVries
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Yongling Zhu
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
13
|
Abstract
Recent technological advances have extended the range of analytic tools to very small samples. It is now possible to assay the transcriptome, and in some cases even the proteome, of single cells reliably. This allows addressing novel questions, such as the genotype/phenotype relationships of single neurons, heterogeneity within individual cells of the same type, or the basis of differential vulnerability to injury. An important prerequisite for these kinds of studies is the ability to isolate well-defined individual cells without contamination by adjacent tissue. In the retina and optic nerve, cells of different types and functions are closely intermingled, limiting the use of standard methods such as laser capture microdissection. Here, we describe a simple method to isolate morphologically intact cells from the retina and the optic nerve and discuss considerations in recognizing and isolating different cell types after dissociation.
Collapse
|
14
|
Pérez de Sevilla Müller L, Solomon A, Sheets K, Hapukino H, Rodriguez AR, Brecha NC. Multiple cell types form the VIP amacrine cell population. J Comp Neurol 2017; 527:133-158. [PMID: 28472856 DOI: 10.1002/cne.24234] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 04/21/2017] [Accepted: 04/27/2017] [Indexed: 12/21/2022]
Abstract
Amacrine cells are a heterogeneous group of interneurons that form microcircuits with bipolar, amacrine and ganglion cells to process visual information in the inner retina. This study has characterized the morphology, neurochemistry and major cell types of a VIP-ires-Cre amacrine cell population. VIP-tdTomato and -Confetti (Brainbow2.1) mouse lines were generated by crossing a VIP-ires-Cre line with either a Cre-dependent tdTomato or Brainbow2.1 reporter line. Retinal sections and whole-mounts were evaluated by quantitative, immunohistochemical, and intracellular labeling approaches. The majority of tdTomato and Confetti fluorescent cell bodies were in the inner nuclear layer (INL) and a few cell bodies were in the ganglion cell layer (GCL). Fluorescent processes ramified in strata 1, 3, 4, and 5 of the inner plexiform layer (IPL). All tdTomato fluorescent cells expressed syntaxin 1A and GABA-immunoreactivity indicating they were amacrine cells. The average VIP-tdTomato fluorescent cell density in the INL and GCL was 535 and 24 cells/mm2 , respectively. TdTomato fluorescent cells in the INL and GCL contained VIP-immunoreactivity. The VIP-ires-Cre amacrine cell types were identified in VIP-Brainbow2.1 retinas or by intracellular labeling in VIP-tdTomato retinas. VIP-1 amacrine cells are bistratified, wide-field cells that ramify in strata 1, 4, and 5, VIP-2A and 2B amacrine cells are medium-field cells that mainly ramify in strata 3 and 4, and VIP-3 displaced amacrine cells are medium-field cells that ramify in strata 4 and 5 of the IPL. VIP-ires-Cre amacrine cells form a neuropeptide-expressing cell population with multiple cell types, which are likely to have distinct roles in visual processing.
Collapse
Affiliation(s)
- Luis Pérez de Sevilla Müller
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Alexander Solomon
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Kristopher Sheets
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Hinekura Hapukino
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Allen R Rodriguez
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Nicholas C Brecha
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763.,Department of Medicine, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763.,Department of Ophthalmology and the Stein Eye Institute, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763.,CURE Digestive Diseases Research Center, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763.,Veterans Administration Greater Los Angeles Health System, Los Angeles, California, 90073
| |
Collapse
|
15
|
Popova E, Kupenova P. Interaction between the serotoninergic and GABAergic systems in frog retina as revealed by electroretinogram. Acta Neurobiol Exp (Wars) 2017. [DOI: 10.21307/ane-2017-067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
16
|
Shekhar K, Lapan SW, Whitney IE, Tran NM, Macosko EZ, Kowalczyk M, Adiconis X, Levin JZ, Nemesh J, Goldman M, McCarroll SA, Cepko CL, Regev A, Sanes JR. Comprehensive Classification of Retinal Bipolar Neurons by Single-Cell Transcriptomics. Cell 2016; 166:1308-1323.e30. [PMID: 27565351 DOI: 10.1016/j.cell.2016.07.054] [Citation(s) in RCA: 726] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/10/2016] [Accepted: 07/28/2016] [Indexed: 12/15/2022]
Abstract
Patterns of gene expression can be used to characterize and classify neuronal types. It is challenging, however, to generate taxonomies that fulfill the essential criteria of being comprehensive, harmonizing with conventional classification schemes, and lacking superfluous subdivisions of genuine types. To address these challenges, we used massively parallel single-cell RNA profiling and optimized computational methods on a heterogeneous class of neurons, mouse retinal bipolar cells (BCs). From a population of ∼25,000 BCs, we derived a molecular classification that identified 15 types, including all types observed previously and two novel types, one of which has a non-canonical morphology and position. We validated the classification scheme and identified dozens of novel markers using methods that match molecular expression to cell morphology. This work provides a systematic methodology for achieving comprehensive molecular classification of neurons, identifies novel neuronal types, and uncovers transcriptional differences that distinguish types within a class.
Collapse
Affiliation(s)
- Karthik Shekhar
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Sylvain W Lapan
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Irene E Whitney
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Nicholas M Tran
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Evan Z Macosko
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | | | - Xian Adiconis
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Joshua Z Levin
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - James Nemesh
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Melissa Goldman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Steven A McCarroll
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Constance L Cepko
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Aviv Regev
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Department of Biology and Koch Institute, MIT, Cambridge, MA 02139, USA.
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA.
| |
Collapse
|
17
|
Struebing FL, Lee RK, Williams RW, Geisert EE. Genetic Networks in Mouse Retinal Ganglion Cells. Front Genet 2016; 7:169. [PMID: 27733864 PMCID: PMC5039302 DOI: 10.3389/fgene.2016.00169] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/06/2016] [Indexed: 01/17/2023] Open
Abstract
Retinal ganglion cells (RGCs) are the output neuron of the eye, transmitting visual information from the retina through the optic nerve to the brain. The importance of RGCs for vision is demonstrated in blinding diseases where RGCs are lost, such as in glaucoma or after optic nerve injury. In the present study, we hypothesize that normal RGC function is transcriptionally regulated. To test our hypothesis, we examine large retinal expression microarray datasets from recombinant inbred mouse strains in GeneNetwork and define transcriptional networks of RGCs and their subtypes. Two major and functionally distinct transcriptional networks centering around Thy1 and Tubb3 (Class III beta-tubulin) were identified. Each network is independently regulated and modulated by unique genomic loci. Meta-analysis of publically available data confirms that RGC subtypes are differentially susceptible to death, with alpha-RGCs and intrinsically photosensitive RGCs (ipRGCs) being less sensitive to cell death than other RGC subtypes in a mouse model of glaucoma.
Collapse
Affiliation(s)
- Felix L Struebing
- Department of Ophthalmology, Emory University School of Medicine Atlanta, GA, USA
| | - Richard K Lee
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine Miami, FL, USA
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center Memphis, TN, USA
| | - Eldon E Geisert
- Department of Ophthalmology, Emory University School of Medicine Atlanta, GA, USA
| |
Collapse
|
18
|
Retinal Neuroprotective Effects of Flibanserin, an FDA-Approved Dual Serotonin Receptor Agonist-Antagonist. PLoS One 2016; 11:e0159776. [PMID: 27447833 PMCID: PMC4957778 DOI: 10.1371/journal.pone.0159776] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 06/07/2016] [Indexed: 12/30/2022] Open
Abstract
PURPOSE To assess the neuroprotective effects of flibanserin (formerly BIMT-17), a dual 5-HT1A agonist and 5-HT2A antagonist, in a light-induced retinopathy model. METHODS Albino BALB/c mice were injected intraperitoneally with either vehicle or increasing doses of flibanserin ranging from 0.75 to 15 mg/kg flibanserin. To assess 5-HT1A-mediated effects, BALB/c mice were injected with 10 mg/kg WAY 100635, a 5-HT1A antagonist, prior to 6 mg/kg flibanserin and 5-HT1A knockout mice were injected with 6 mg/kg flibanserin. Injections were administered once immediately prior to light exposure or over the course of five days. Light exposure lasted for one hour at an intensity of 10,000 lux. Retinal structure was assessed using spectral domain optical coherence tomography and retinal function was assessed using electroretinography. To investigate the mechanisms of flibanserin-mediated neuroprotection, gene expression, measured by RT-qPCR, was assessed following five days of daily 15 mg/kg flibanserin injections. RESULTS A five-day treatment regimen of 3 to 15 mg/kg of flibanserin significantly preserved outer retinal structure and function in a dose-dependent manner. Additionally, a single-day treatment regimen of 6 to 15 mg/kg of flibanserin still provided significant protection. The action of flibanserin was hindered by the 5-HT1A antagonist, WAY 100635, and was not effective in 5-HT1A knockout mice. Creb, c-Jun, c-Fos, Bcl-2, Cast1, Nqo1, Sod1, and Cat were significantly increased in flibanserin-injected mice versus vehicle-injected mice. CONCLUSIONS Intraperitoneal delivery of flibanserin in a light-induced retinopathy mouse model provides retinal neuroprotection. Mechanistic data suggests that this effect is mediated through 5-HT1A receptors and that flibanserin augments the expression of genes capable of reducing mitochondrial dysfunction and oxidative stress. Since flibanserin is already FDA-approved for other indications, the potential to repurpose this drug for treating retinal degenerations merits further investigation.
Collapse
|
19
|
Sanges D, Simonte G, Di Vicino U, Romo N, Pinilla I, Nicolás M, Cosma MP. Reprogramming Müller glia via in vivo cell fusion regenerates murine photoreceptors. J Clin Invest 2016; 126:3104-16. [PMID: 27427986 DOI: 10.1172/jci85193] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 05/24/2016] [Indexed: 12/17/2022] Open
Abstract
Vision impairments and blindness caused by retinitis pigmentosa result from severe neurodegeneration that leads to a loss of photoreceptors, the specialized light-sensitive neurons that enable vision. Although the mammalian nervous system is unable to replace neurons lost due to degeneration, therapeutic approaches to reprogram resident glial cells to replace retinal neurons have been proposed. Here, we demonstrate that retinal Müller glia can be reprogrammed in vivo into retinal precursors that then differentiate into photoreceptors. We transplanted hematopoietic stem and progenitor cells (HSPCs) into retinas affected by photoreceptor degeneration and observed spontaneous cell fusion events between Müller glia and the transplanted cells. Activation of Wnt signaling in the transplanted HSPCs enhanced survival and proliferation of Müller-HSPC hybrids as well as their reprogramming into intermediate photoreceptor precursors. This suggests that Wnt signaling drives the reprogrammed cells toward a photoreceptor progenitor fate. Finally, Müller-HSPC hybrids differentiated into photoreceptors. Transplantation of HSPCs with activated Wnt functionally rescued the retinal degeneration phenotype in rd10 mice, a model for inherited retinitis pigmentosa. Together, these results suggest that photoreceptors can be generated by reprogramming Müller glia and that this approach may have potential as a strategy for reversing retinal degeneration.
Collapse
|
20
|
Analogous Convergence of Sustained and Transient Inputs in Parallel On and Off Pathways for Retinal Motion Computation. Cell Rep 2016; 14:1892-900. [PMID: 26904938 PMCID: PMC6404534 DOI: 10.1016/j.celrep.2016.02.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/20/2015] [Accepted: 01/23/2016] [Indexed: 02/03/2023] Open
Abstract
Visual motion information is computed by parallel On and Off pathways in the retina, which lead to On and Off types of starburst amacrine cells (SACs). The approximate mirror symmetry between this pair of cell types suggests that On and Off pathways might compute motion using analogous mechanisms. To test this idea, we reconstructed On SACs and On bipolar cells (BCs) from serial electron microscopic images of a mouse retina. We defined a new On BC type in the course of classifying On BCs. Through quantitative contact analysis, we found evidence that sustained and transient On BC types are wired to On SAC dendrites at different distances from the SAC soma, mirroring our previous wiring diagram for the Off BC-SAC circuit. Our finding is consistent with the hypothesis that On and Off pathways contain parallel correlation-type motion detectors. Greene et al. find that in the mouse retina, sustained and transient On bipolar cell (BC) types are wired to dendrites of On starburst amacrine cells (SACs) at different distances from the SAC soma. This wiring specificity may support retinal computation of the direction of a moving stimulus.
Collapse
|
21
|
Gábriel R, Erdélyi F, Szabó G, Lawrence JJ, Wilhelm M. Ectopic transgene expression in the retina of four transgenic mouse lines. Brain Struct Funct 2015; 221:3729-41. [PMID: 26563404 DOI: 10.1007/s00429-015-1128-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 10/03/2015] [Indexed: 12/11/2022]
Abstract
Retinal expression of transgenes was examined in four mouse lines. Two constructs were driven by the choline acetyltransferase (ChAT) promoter: green fluorescent protein conjugated to tau protein (tau-GFP) or cytosolic yellow fluorescent protein (YFP) generated through CRE recombinase-induced expression of Rosa26 (ChAT-CRE/Rosa26YFP). Two other constructs targeted inhibitory interneurons: GABAergic horizontal and amacrine cells identified by glutamic acid decarboxylase (GAD65-GFP) or parvalbumin (PV) cells (PV-CRE/Rosa26YFP). Animals were transcardially perfused and retinal sections prepared. Antibodies against PV, calretinin (CALR), calbindin (CALB), and tyrosine hydroxylase (TH) were used to counterstain transgene-expressing cells. In PVxRosa and ChAT-tauGFP constructs, staining appeared in vertically oriented row of processes resembling Müller cells. In the ChATxRosa construct, populations of amacrine cells and neurons in the ganglion cell layer were labeled. Some cones also exhibited GFP fluorescence. CALR, PV and TH were found in none of these cells. Occasionally, we found GFP/CALR and GFP/PV double-stained cells in the ganglion cell layer (GCL). In the GAD65-GFP construct, all layers of the neuroretina were labeled, except photoreceptors. Not all horizontal cells expressed GFP. We did not find GFP/TH double-labeled cells and GFP was rarely present in CALR- and CALB-containing cells. Many PV-positive neurons were also labeled for GFP, including small diameter amacrines. In the GCL, single labeling for GFP and PV was ascertained, as well as several CALR/PV double-stained neurons. In the GCL, cells triple labeled with GFP/CALR/CALB were sparse. In conclusion, only one of the four transgenic constructs exhibited an expression pattern consistent with endogenous retinal protein expression, while the others strongly suggested ectopic gene expression.
Collapse
Affiliation(s)
- Robert Gábriel
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Ferenc Erdélyi
- Laboratory of Molecular Biology and Genetics, Institute of Experimental Medicine, Hungarian Academy of Sciences, 1450, Budapest, Hungary
| | - Gábor Szabó
- Laboratory of Molecular Biology and Genetics, Institute of Experimental Medicine, Hungarian Academy of Sciences, 1450, Budapest, Hungary
| | - J Josh Lawrence
- Center for Structural and Functional Neuroscience, University of Montana, Missoula, MT, USA.,Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA.,Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, 79430, USA
| | - Márta Wilhelm
- Institute of Sport Sciences and Physical Education, University of Pécs, Ifjúság u. 6., 7624, Pécs, Hungary.
| |
Collapse
|
22
|
Vuong HE, Pérez de Sevilla Müller L, Hardi CN, McMahon DG, Brecha NC. Heterogeneous transgene expression in the retinas of the TH-RFP, TH-Cre, TH-BAC-Cre and DAT-Cre mouse lines. Neuroscience 2015; 307:319-37. [PMID: 26335381 PMCID: PMC4603663 DOI: 10.1016/j.neuroscience.2015.08.060] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 08/21/2015] [Accepted: 08/24/2015] [Indexed: 11/29/2022]
Abstract
Transgenic mouse lines are essential tools for understanding the connectivity, physiology and function of neuronal circuits, including those in the retina. This report compares transgene expression in the retina of a tyrosine hydroxylase (TH)-red fluorescent protein (RFP) mouse line with three catecholamine-related Cre recombinase mouse lines [TH-bacterial artificial chromosome (BAC)-, TH-, and dopamine transporter (DAT)-Cre] that were crossed with a ROSA26-tdTomato reporter line. Retinas were evaluated and immunostained with commonly used antibodies including those directed to TH, GABA and glycine to characterize the RFP or tdTomato fluorescent-labeled amacrine cells, and an antibody directed to RNA-binding protein with multiple splicing to identify ganglion cells. In TH-RFP retinas, types 1 and 2 dopamine (DA) amacrine cells were identified by their characteristic cellular morphology and type 1 DA cells by their expression of TH immunoreactivity. In the TH-BAC-, TH-, and DAT-tdTomato retinas, less than 1%, ∼ 6%, and 0%, respectively, of the fluorescent cells were the expected type 1 DA amacrine cells. Instead, in the TH-BAC-tdTomato retinas, fluorescently labeled AII amacrine cells were predominant, with some medium diameter ganglion cells. In TH-tdTomato retinas, fluorescence was in multiple neurochemical amacrine cell types, including four types of polyaxonal amacrine cells. In DAT-tdTomato retinas, fluorescence was in GABA immunoreactive amacrine cells, including two types of bistratified and two types of monostratified amacrine cells. Although each of the Cre lines was generated with the intent to specifically label DA cells, our findings show a cellular diversity in Cre expression in the adult retina and indicate the importance of careful characterization of transgene labeling patterns. These mouse lines with their distinctive cellular labeling patterns will be useful tools for future studies of retinal function and visual processing.
Collapse
Affiliation(s)
- H E Vuong
- Molecular, Cellular, and Integrative Physiology Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - L Pérez de Sevilla Müller
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - C N Hardi
- Department of Psychology, College of Letters and Science, UCLA, Los Angeles, CA 90095, United States
| | - D G McMahon
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, United States
| | - N C Brecha
- Molecular, Cellular, and Integrative Physiology Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; CURE-Digestive Diseases Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90095, United States.
| |
Collapse
|
23
|
Liu X, Yang C, Liu J, Liu J, Hu R, Lian H, Lin G, Liu L, Yong KT, Ye L. In Vitro evaluation and monitoring of the expression level and localization of aldose reductase using functionalized quantum dots and EGFP. BIOTECHNOL BIOPROC E 2015. [DOI: 10.1007/s12257-015-0022-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
24
|
Sanes JR, Masland RH. The types of retinal ganglion cells: current status and implications for neuronal classification. Annu Rev Neurosci 2015; 38:221-46. [PMID: 25897874 DOI: 10.1146/annurev-neuro-071714-034120] [Citation(s) in RCA: 499] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In the retina, photoreceptors pass visual information to interneurons, which process it and pass it to retinal ganglion cells (RGCs). Axons of RGCs then travel through the optic nerve, telling the rest of the brain all it will ever know about the visual world. Research over the past several decades has made clear that most RGCs are not merely light detectors, but rather feature detectors, which send a diverse set of parallel, highly processed images of the world on to higher centers. Here, we review progress in classification of RGCs by physiological, morphological, and molecular criteria, making a particular effort to distinguish those cell types that are definitive from those for which information is partial. We focus on the mouse, in which molecular and genetic methods are most advanced. We argue that there are around 30 RGC types and that we can now account for well over half of all RGCs. We also use RGCs to examine the general problem of neuronal classification, arguing that insights and methods from the retina can guide the classification enterprise in other brain regions.
Collapse
Affiliation(s)
- Joshua R Sanes
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138;
| | | |
Collapse
|
25
|
Wilhelm M, Lawrence JJ, Gábriel R. Enteric plexuses of two choline-acetyltransferase transgenic mouse lines: chemical neuroanatomy of the fluorescent protein-expressing nerve cells. Brain Res Bull 2015; 111:76-83. [PMID: 25592616 DOI: 10.1016/j.brainresbull.2015.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 12/23/2014] [Accepted: 01/05/2015] [Indexed: 12/31/2022]
Abstract
We studied cholinergic circuit elements in the enteric nervous system (ENS) of two distinct transgenic mouse lines in which fluorescent protein expression was driven by the choline-acetyltransferase (ChAT) promoter. In the first mouse line, green fluorescent protein was fused to the tau gene. This construct allowed the visualization of the fiber tracts and ganglia, however the nerve cells were poorly resolved. In the second mouse line (ChATcre-YFP), CRE/loxP recombination yielded cytosolic expression of yellow fluorescent protein (YFP). In these preparations the morphology of enteric neurons could be well studied. We also determined the neurochemical identity of ENS neurons in muscular and submucous layers using antibodies against YFP, calretinin (CALR), calbindin (CALB), and vasoactive intestinal peptide (VIP). Confocal microscopic imaging was used to visualize fluorescently-conjugated secondary antibodies. In ChATcre-YFP preparations, YFP was readily apparent in somatodendritic regions of ENS neurons. In the myenteric plexus, YFP/CALR/VIP staining revealed that 34% of cholinergic cells co-labeled with CALR. Few single-stained CR-positive cells were observed. Neither YFP nor CALR co-localized with VIP. In GFP/CALB/CALR staining, all co-localization combinations were represented. In the submucosal plexus, YFP/CALR/VIP staining revealed discrete neuronal populations. However, in separate preparations, double labeling was observed for YFP/CALR and CALR/VIP. In YFP/CALR/CALB staining, all combinations of double staining and triple labeling were verified. In conclusion, the neurochemical coding of ENS neurons in these mouse lines is consistent with many observations in non-transgenic animals. Thus, they provide useful tools for physiological and pharmacological studies on distinct neurochemical subtypes of ENS neurons.
Collapse
Affiliation(s)
- Márta Wilhelm
- Institute of Sport Sciences and Physical Education, University of Pécs, Pécs, Hungary
| | - J Josh Lawrence
- COBRE Center for Structural and Functional Neuroscience; Department of Biomedical Sciences, University of Montana, Missoula, Montana, USA
| | - Robert Gábriel
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary.
| |
Collapse
|
26
|
Duan X, Krishnaswamy A, De la Huerta I, Sanes JR. Type II cadherins guide assembly of a direction-selective retinal circuit. Cell 2014; 158:793-807. [PMID: 25126785 DOI: 10.1016/j.cell.2014.06.047] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 05/10/2014] [Accepted: 06/20/2014] [Indexed: 01/02/2023]
Abstract
Complex retinal circuits process visual information and deliver it to the brain. Few molecular determinants of synaptic specificity in this system are known. Using genetic and optogenetic methods, we identified two types of bipolar interneurons that convey visual input from photoreceptors to a circuit that computes the direction in which objects are moving. We then sought recognition molecules that promote selective connections of these cells with previously characterized components of the circuit. We found that the type II cadherins, cdh8 and cdh9, are each expressed selectively by one of the two bipolar cell types. Using loss- and gain-of-function methods, we showed that they are critical determinants of connectivity in this circuit and that perturbation of their expression leads to distinct defects in visually evoked responses. Our results reveal cellular components of a retinal circuit and demonstrate roles of type II cadherins in synaptic choice and circuit function.
Collapse
Affiliation(s)
- Xin Duan
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Arjun Krishnaswamy
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Irina De la Huerta
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joshua R Sanes
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
27
|
Rodriguez AR, de Sevilla Müller LP, Brecha NC. The RNA binding protein RBPMS is a selective marker of ganglion cells in the mammalian retina. J Comp Neurol 2014; 522:1411-43. [PMID: 24318667 DOI: 10.1002/cne.23521] [Citation(s) in RCA: 324] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 11/27/2013] [Accepted: 12/03/2013] [Indexed: 12/12/2022]
Abstract
There are few neurochemical markers that reliably identify retinal ganglion cells (RGCs), which are a heterogeneous population of cells that integrate and transmit the visual signal from the retina to the central visual nuclei. We have developed and characterized a new set of affinity-purified guinea pig and rabbit antibodies against RNA-binding protein with multiple splicing (RBPMS). On western blots these antibodies recognize a single band at 〜24 kDa, corresponding to RBPMS, and they strongly label RGC and displaced RGC (dRGC) somata in mouse, rat, guinea pig, rabbit, and monkey retina. RBPMS-immunoreactive cells and RGCs identified by other techniques have a similar range of somal diameters and areas. The density of RBPMS cells in mouse and rat retina is comparable to earlier semiquantitative estimates of RGCs. RBPMS is mainly expressed in medium and large DAPI-, DRAQ5-, NeuroTrace- and NeuN-stained cells in the ganglion cell layer (GCL), and RBPMS is not expressed in syntaxin (HPC-1)-immunoreactive cells in the inner nuclear layer (INL) and GCL, consistent with their identity as RGCs, and not displaced amacrine cells. In mouse and rat retina, most RBPMS cells are lost following optic nerve crush or transection at 3 weeks, and all Brn3a-, SMI-32-, and melanopsin-immunoreactive RGCs also express RBPMS immunoreactivity. RBPMS immunoreactivity is localized to cyan fluorescent protein (CFP)-fluorescent RGCs in the B6.Cg-Tg(Thy1-CFP)23Jrs/J mouse line. These findings show that antibodies against RBPMS are robust reagents that exclusively identify RGCs and dRGCs in multiple mammalian species, and they will be especially useful for quantification of RGCs.
Collapse
Affiliation(s)
- Allen R Rodriguez
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | | | | |
Collapse
|
28
|
Sharma TP, McDowell CM, Liu Y, Wagner AH, Thole D, Faga BP, Wordinger RJ, Braun TA, Clark AF. Optic nerve crush induces spatial and temporal gene expression patterns in retina and optic nerve of BALB/cJ mice. Mol Neurodegener 2014; 9:14. [PMID: 24767545 PMCID: PMC4113182 DOI: 10.1186/1750-1326-9-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 04/18/2014] [Indexed: 12/18/2022] Open
Abstract
Background Central nervous system (CNS) trauma and neurodegenerative disorders trigger a cascade of cellular and molecular events resulting in neuronal apoptosis and regenerative failure. The pathogenic mechanisms and gene expression changes associated with these detrimental events can be effectively studied using a rodent optic nerve crush (ONC) model. The purpose of this study was to use a mouse ONC model to: (a) evaluate changes in retina and optic nerve (ON) gene expression, (b) identify neurodegenerative pathogenic pathways and (c) discover potential new therapeutic targets. Results Only 54% of total neurons survived in the ganglion cell layer (GCL) 28 days post crush. Using Bayesian Estimation of Temporal Regulation (BETR) gene expression analysis, we identified significantly altered expression of 1,723 and 2,110 genes in the retina and ON, respectively. Meta-analysis of altered gene expression (≥1.5, ≤-1.5, p < 0.05) using Partek and DAVID demonstrated 28 up and 20 down-regulated retinal gene clusters and 57 up and 41 down-regulated optic nerve clusters. Regulated gene clusters included regenerative change, synaptic plasticity, axonogenesis, neuron projection, and neuron differentiation. Expression of selected genes (Vsnl1, Syt1, Synpr and Nrn1) from retinal and ON neuronal clusters were quantitatively and qualitatively examined for their relation to axonal neurodegeneration by immunohistochemistry and qRT-PCR. Conclusion A number of detrimental gene expression changes occur that contribute to trauma-induced neurodegeneration after injury to ON axons. Nrn1 (synaptic plasticity gene), Synpr and Syt1 (synaptic vesicle fusion genes), and Vsnl1 (neuron differentiation associated gene) were a few of the potentially unique genes identified that were down-regulated spatially and temporally in our rodent ONC model. Bioinformatic meta-analysis identified significant tissue-specific and time-dependent gene clusters associated with regenerative changes, synaptic plasticity, axonogenesis, neuron projection, and neuron differentiation. These ONC induced neuronal loss and regenerative failure associated clusters can be extrapolated to changes occurring in other forms of CNS trauma or in clinical neurodegenerative pathological settings. In conclusion, this study identified potential therapeutic targets to address two key mechanisms of CNS trauma and neurodegeneration: neuronal loss and regenerative failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Abbot F Clark
- North Texas Eye Research Institute, Ft, Worth, TX USA.
| |
Collapse
|
29
|
De Sevilla Müller LP, Liu J, Solomon A, Rodriguez A, Brecha NC. Expression of voltage-gated calcium channel α(2)δ(4) subunits in the mouse and rat retina. J Comp Neurol 2014; 521:2486-501. [PMID: 23296739 DOI: 10.1002/cne.23294] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 12/12/2012] [Accepted: 12/27/2012] [Indexed: 01/18/2023]
Abstract
High-voltage activated Ca channels participate in multiple cellular functions, including transmitter release, excitation, and gene transcription. Ca channels are heteromeric proteins consisting of a pore-forming α(1) subunit and auxiliary α(2)δ and β subunits. Although there are reports of α(2)δ(4) subunit mRNA in the mouse retina and localization of the α(2)δ(4) subunit immunoreactivity to salamander photoreceptor terminals, there is a limited overall understanding of its expression and localization in the retina. α(2)δ(4) subunit expression and distribution in the mouse and rat retina were evaluated by using reverse transcriptase polymerase chain reaction, western blot, and immunohistochemistry with specific primers and a well-characterized antibody to the α(2)δ(4) subunit. α(2)δ(4) subunit mRNA and protein are present in mouse and rat retina, brain, and liver homogenates. Immunostaining for the α(2)δ(4) subunit is mainly localized to Müller cell processes and endfeet, photoreceptor terminals, and photoreceptor outer segments. This subunit is also expressed in a few displaced ganglion cells and bipolar cell dendrites. These findings suggest that the α(2)δ(4) subunit participates in the modulation of L-type Ca(2+) current regulating neurotransmitter release from photoreceptor terminals and Ca(2+)-dependent signaling pathways in bipolar and Müller cells.
Collapse
Affiliation(s)
- Luis Pérez De Sevilla Müller
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California 90095, USA.
| | | | | | | | | |
Collapse
|
30
|
Knop GC, Pottek M, Monyer H, Weiler R, Dedek K. Morphological and physiological properties of enhanced green fluorescent protein (EGFP)-expressing wide-field amacrine cells in the ChAT-EGFP mouse line. Eur J Neurosci 2013; 39:800-10. [PMID: 24299612 DOI: 10.1111/ejn.12443] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 11/01/2013] [Accepted: 11/06/2013] [Indexed: 01/25/2023]
Abstract
Mammalian retinas comprise a variety of interneurons, among which amacrine cells represent the largest group, with more than 30 different cell types each exhibiting a rather distinctive morphology and carrying out a unique function in retinal processing. However, many amacrine types have not been studied systematically because, in particular, amacrine cells with large dendritic fields, i.e. wide-field amacrine cells, have a low abundance and are therefore difficult to target. Here, we used a transgenic mouse line expressing the coding sequence of enhanced green fluorescent protein under the promoter for choline acetyltransferase (ChAT-EGFP mouse) and characterized a single wide-field amacrine cell population monostratifying in layer 2/3 of the inner plexiform layer (WA-S2/3 cell). Somata of WA-S2/3 cells are located either in the inner nuclear layer or are displaced to the ganglion cell layer and exhibit a low cell density. Using immunohistochemistry, we show that WA-S2/3 cells are presumably GABAergic but may also release acetylcholine as their somata are weakly positive for ChAT. Two-photon-guided patch-clamp recordings from intact retinas revealed WA-S2/3 cells to be ON-OFF cells with a homogenous receptive field even larger than the dendritic field. The large spatial extent of the receptive field is most likely due to the extensive homologous and heterologous coupling among WA-S2/3 cells and to other amacrine cells, respectively, as indicated by tracer injections. In summary, we have characterized a novel type of GABAergic ON-OFF wide-field amacrine cell which is ideally suited to providing long-range inhibition to ganglion cells due to its strong coupling.
Collapse
Affiliation(s)
- Gabriel C Knop
- Neurobiology, University of Oldenburg, 26111, Oldenburg, Germany
| | | | | | | | | |
Collapse
|
31
|
Kunzevitzky NJ, Willeford KT, Feuer WJ, Almeida MV, Goldberg JL. Amacrine cell subtypes differ in their intrinsic neurite growth capacity. Invest Ophthalmol Vis Sci 2013; 54:7603-13. [PMID: 24130183 DOI: 10.1167/iovs.13-12691] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
PURPOSE Amacrine cell neurite patterning has been extensively studied in vivo, and more than 30 subpopulations with varied morphologies have been identified in the mammalian retina. It is not known, however, whether the complex amacrine cell morphology is determined intrinsically, is signaled by extrinsic cues, or both. METHODS Here we purified rat amacrine cell subpopulations away from their retinal neighbors and glial-derived factors to ask questions about their intrinsic neurite growth ability. In defined medium strongly trophic for amacrine cells in vitro, we characterized survival and neurite growth of amacrine cell subpopulations defined by expression of specific markers. RESULTS We found that a series of amacrine cell subtype markers are developmentally regulated, turning on through early postnatal development. Subtype marker expression was observed in similar fractions of cultured amacrine cells as was observed in vivo, and was maintained with time in culture. Overall, amacrine cell neurite growth followed principles very similar to those in postnatal retinal ganglion cells, but embryonic retinal ganglion cells demonstrated different features, relating to their rapid axon growth. Surprisingly, the three subpopulations of amacrine cells studied in vitro recapitulated quantitatively and qualitatively the varied morphologies they have in vivo. CONCLUSIONS Our data suggest that cultured amacrine cells maintain intrinsic fidelity to their identified in vivo subtypes, and furthermore, that cell-autonomous, intrinsic factors contribute to the regulation of neurite patterning.
Collapse
|
32
|
Ivanova E, Lee P, Pan ZH. Characterization of multiple bistratified retinal ganglion cells in a purkinje cell protein 2-Cre transgenic mouse line. J Comp Neurol 2013; 521:2165-80. [PMID: 23224947 DOI: 10.1002/cne.23279] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 07/28/2012] [Accepted: 11/28/2012] [Indexed: 01/08/2023]
Abstract
Retinal ganglion cells are categorized into multiple classes, including multiple types of bistratified ganglion cells (BGCs). The recent use of transgenic mouse lines with specific type(s) of ganglion cells that are labeled by fluorescent markers has facilitated the morphological and physiological studies of BGCs, particularly the directional-selective BGCs. The most important benefit from using transgenic animals is the capability to perform in vivo gene manipulation. In particular, the Cre/LoxP recombination system has become a powerful tool, allowing gene deletion, overexpression, and ectopic expression in a cell type-specific and temporally controlled fashion. The key to this tool is the availability of Cre mouse lines with cell or tissue type-specific expression of Cre recombinase. In this study we characterized the Cre-positive retinal ganglion cells in a PCP2 (Purkinje cell protein 2)-cre mouse line. We found that all of the Cre-positive retinal ganglion cells were BGCs. Based on morphological criteria, we determined that they can be grouped into five types. The On- and Off-dendrites of three of these types stratified outside of the cholinergic bands and differed from directional selective ganglion cells (DSGCs) morphologically. These cells were negative for Brn-3b and positive for both calretinin and CART retina markers. The remaining two types were identified as putative On-Off and On-DSGCs. This Cre mouse line could be useful for further studies of the molecular and functional properties of BGCs in mice.
Collapse
Affiliation(s)
- Elena Ivanova
- Department of Ophthalmology and Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | |
Collapse
|
33
|
Toychiev AH, Sagdullaev B, Yee CW, Ivanova E, Sagdullaev BT. A time and cost efficient approach to functional and structural assessment of living neuronal tissue. J Neurosci Methods 2013; 214:105-12. [PMID: 23370309 DOI: 10.1016/j.jneumeth.2013.01.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 01/16/2013] [Accepted: 01/18/2013] [Indexed: 11/18/2022]
Abstract
In this manuscript, we describe a protocol for capturing both physiological and structural properties of living neuronal tissue. An essential aspect of this method is its flexibility and fast turnaround time. It is a streamlined process that includes recording of electrophysiological neuronal activity, calcium imaging, and structural analysis. This is accomplished by placing intact tissue on a modified Millicell Biopore insert. The Biopore membrane suspends the tissue in the perfusion solution, allowing for complete access to nutrients, oxygen, and pharmacological agents. The ring that holds the membrane ensures its structural stability; forceps can be used to grip the ring without contacting the filter or the tissue, for easy transfer between multiple setups. We show that tissue readily adheres to the surface of the membrane, its entire surface is visible in transmitted light and accessible for recording and imaging, and remains responsive to physiological stimuli for extended periods of time.
Collapse
Affiliation(s)
- Abduqodir H Toychiev
- Department of Ophthalmology, Weill Medical College of Cornell University, Burke Medical Research Institute, White Plains, NY 10605, USA
| | | | | | | | | |
Collapse
|
34
|
Herranz-Martín S, Jimeno D, Paniagua AE, Velasco A, Lara JM, Aijón J, Lillo C. Immunocytochemical evidence of the localization of the Crumbs homologue 3 protein (CRB3) in the developing and mature mouse retina. PLoS One 2012; 7:e50511. [PMID: 23226298 PMCID: PMC3511585 DOI: 10.1371/journal.pone.0050511] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 10/22/2012] [Indexed: 12/28/2022] Open
Abstract
CRB3 (Crumbs homologue 3), a member of the CRB protein family (homologous to the Drosophila Crumbs), is expressed in different epithelium-derived cell types in mammals, where it seems to be involved in regulating the establishment and stability of tight junctions and in ciliogenesis. This protein has been also detected in the retina, but little is known about its localization and function in this tissue. Our goal here was to perform an in-depth study of the presence of CRB3 protein in the mouse retina and to analyze its expression during photoreceptor ciliogenesis and the establishment of the plexiform retinal layers. Double immunofluorescence experiments for CRB3 and well-known markers for the different retinal cell types were performed to study the localization of the CRB3 protein. According to our results, CRB3 is present from postnatal day 0 (P0) until adulthood in the mouse retina. It is localized in the inner segments (IS) of photoreceptor cells, especially concentrated in the area where the connecting cilium is located, in their synaptic terminals in the outer plexiform layer (OPL), and in sub-populations of amacrine and bipolar cells in the inner plexiform layer (IPL).
Collapse
Affiliation(s)
- Saúl Herranz-Martín
- Institute of Neurosciences of Castilla y León (INCyL), Institute of Biomedical Research of Salamanca (IBSAL), Cell Biology and Pathology, University of Salamanca, Salamanca, Spain
| | - David Jimeno
- Centro de Investigación del Cáncer-IBMCC (CSIC-USAL), University of Salamanca, Campus Unamuno, Salamanca, Spain
| | - Antonio E. Paniagua
- Institute of Neurosciences of Castilla y León (INCyL), Institute of Biomedical Research of Salamanca (IBSAL), Cell Biology and Pathology, University of Salamanca, Salamanca, Spain
| | - Almudena Velasco
- Institute of Neurosciences of Castilla y León (INCyL), Institute of Biomedical Research of Salamanca (IBSAL), Cell Biology and Pathology, University of Salamanca, Salamanca, Spain
| | - Juan M. Lara
- Institute of Neurosciences of Castilla y León (INCyL), Institute of Biomedical Research of Salamanca (IBSAL), Cell Biology and Pathology, University of Salamanca, Salamanca, Spain
| | - José Aijón
- Institute of Neurosciences of Castilla y León (INCyL), Institute of Biomedical Research of Salamanca (IBSAL), Cell Biology and Pathology, University of Salamanca, Salamanca, Spain
| | - Concepción Lillo
- Institute of Neurosciences of Castilla y León (INCyL), Institute of Biomedical Research of Salamanca (IBSAL), Cell Biology and Pathology, University of Salamanca, Salamanca, Spain
- * E-mail:
| |
Collapse
|
35
|
Puthussery T, Gayet-Primo J, Taylor WR. Carbonic anhydrase-related protein VIII is expressed in rod bipolar cells and alters signaling at the rod bipolar to AII-amacrine cell synapse in the mammalian retina. Eur J Neurosci 2011; 34:1419-31. [PMID: 22004450 DOI: 10.1111/j.1460-9568.2011.07861.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mutation of the gene encoding carbonic anhydrase-related protein VIII (CAVIII) results in motor coordination deficits in mice and humans, due to loss of this protein in Purkinje cells of the cerebellum. Recent studies have indicated that the CAVIII gene, Car8, is also expressed in rod bipolar cells (RBCs), a critical glutamatergic neuron for scotopic vision. We investigated the localization of CAVIII in the mouse and macaque retina, and utilized the wdl mouse, which has a null mutation in the Car8 gene, to determine how the loss of CAVIII affects retinal signaling. CAVIII immunoreactivity was observed in RBCs, with particularly high staining intensity in the axon terminals. In addition, weaker staining was observed in a subset of cone bipolar cells and γ-aminobutyric acid (GABA)ergic amacrine cells. Light-evoked current and voltage responses of RBCs were not altered in the wdl mutant. However, light-evoked current responses from the AII-amacrine cell, a postsynaptic partner at the RBC ribbon synapse, were significantly larger, and more prolonged than in control mice. These changes could not be attributed to alterations in calcium current activation or inactivation, or to changes in the density of RBCs. Furthermore, no gross synaptic alterations were evident in the wdl mutant at the light or ultrastructural level. These data provide evidence that the CAVIII protein, which is highly conserved in vertebrates, is selectively expressed within neural circuits, and may be important for modulating retinal neurotransmission.
Collapse
Affiliation(s)
- T Puthussery
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Sciences University, Portland, OR, USA
| | | | | |
Collapse
|
36
|
Müller LPDS, Do MTH, Yau KW, He S, Baldridge WH. Tracer coupling of intrinsically photosensitive retinal ganglion cells to amacrine cells in the mouse retina. J Comp Neurol 2011; 518:4813-24. [PMID: 20963830 DOI: 10.1002/cne.22490] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) are a subtype of ganglion cell in the mammalian retina that expresses the photopigment melanopsin and drives non-image-forming visual functions. Three morphological subtypes of ipRGCs (M1, M2, and M3) have been described based on their dendritic stratifications in the inner plexiform layer (IPL), but the question of their potential interactions via electrical coupling remains unsettled. In this study, we have addressed this question in the mouse retina by, injecting the tracer Neurobiotin into ipRGCs that had been genetically labelled with the fluorescent protein, tdTomato. We confirmed the presence of the M1-M3 subtypes of ipRGCs based on their distinct dendritic stratifications. All three subtypes were tracer coupled to putative amacrine cells situated within the ganglion cell layer (GCL) but not the inner nuclear layer (INL). The cells tracer coupled to the M1 and M2 cells were shown to be widefield GABA-immunoreactive amacrine cells. We found no evidence of homologous tracer coupling of ipRGCs or heterologous coupling to other types of ganglion cells.
Collapse
|
37
|
Retinal ganglion cells with distinct directional preferences differ in molecular identity, structure, and central projections. J Neurosci 2011; 31:7753-62. [PMID: 21613488 DOI: 10.1523/jneurosci.0907-11.2011] [Citation(s) in RCA: 257] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The retina contains ganglion cells (RGCs) that respond selectively to objects moving in particular directions. Individual members of a group of ON-OFF direction-selective RGCs (ooDSGCs) detect stimuli moving in one of four directions: ventral, dorsal, nasal, or temporal. Despite this physiological diversity, little is known about subtype-specific differences in structure, molecular identity, and projections. To seek such differences, we characterized mouse transgenic lines that selectively mark ooDSGCs preferring ventral or nasal motion as well as a line that marks both ventral- and dorsal-preferring subsets. We then used the lines to identify cell surface molecules, including Cadherin 6, CollagenXXVα1, and Matrix metalloprotease 17, that are selectively expressed by distinct subsets of ooDSGCs. We also identify a neuropeptide, CART (cocaine- and amphetamine-regulated transcript), that distinguishes all ooDSGCs from other RGCs. Together, this panel of endogenous and transgenic markers distinguishes the four ooDSGC subsets. Patterns of molecular diversification occur before eye opening and are therefore experience independent. They may help to explain how the four subsets obtain distinct inputs. We also demonstrate differences among subsets in their dendritic patterns within the retina and their axonal projections to the brain. Differences in projections indicate that information about motion in different directions is sent to different destinations.
Collapse
|
38
|
Azeredo da Silveira R, Roska B. Cell types, circuits, computation. Curr Opin Neurobiol 2011; 21:664-71. [PMID: 21641794 DOI: 10.1016/j.conb.2011.05.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2011] [Revised: 05/10/2011] [Accepted: 05/10/2011] [Indexed: 12/25/2022]
Abstract
How does the connectivity of a neuronal circuit, together with the individual properties of the cell types that take part in it, result in a given computation? We examine this question in the context of retinal circuits. We suggest that the retina can be viewed as a parallel assemblage of many small computational devices, highly stereotypical and task-specific circuits afferent to a given ganglion cell type, and we discuss some rules that govern computation in these devices. Multi-device processing in retina poses conceptual problems when it is contrasted with cortical processing. We lay out open questions both on processing in retinal circuits and on implications for cortical processing of retinal inputs.
Collapse
Affiliation(s)
- Rava Azeredo da Silveira
- Department of Physics and Department of Cognitive Studies, École Normale Supérieure, Paris, France.
| | | |
Collapse
|
39
|
Bernstein SL, Guo Y. Changes in cholinergic amacrine cells after rodent anterior ischemic optic neuropathy (rAION). Invest Ophthalmol Vis Sci 2011; 52:904-10. [PMID: 20847114 DOI: 10.1167/iovs.10-5247] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Displaced cholinergic amacrine cell neurons comprise a significant fraction of the retinal ganglion cell (RGC) layer. Rodent anterior ischemic optic neuropathy (rAION) is an optic nerve infarct, which results in RGC loss in mice. The goal was to determine whether rAION produces changes in amacrine cell neurons. METHODS rAION was generated in transgenic mice carrying a cyan fluorescent reporter protein (CFP) gene linked to the Thy-1 promoter, which expresses CFP in RGCs. rAION was induced with standard parameters. Retinas were examined pre-and post-induction by retinal fundus microscopy. rAION induction severity was scored by changes in retinal transparency and RGC loss. Cholinergic amacrine cells were identified via choline acetyltransferase (ChAT) immunohistochemistry. ChAT and CFP expression was evaluated in flat-mounted retinas examined by confocal microscopy and western analysis. RESULTS Moderate rAION induction levels (defined as early retention of retinal transparency and <70% RGC loss) did not alter amacrine cell numbers in the RGC layer, but changed the relative levels of ChAT expression by immunohistochemistry. No changes in total ChAT protein were seen. Severe rAION induction (defined as loss of retinal transparency and >70% RGC loss) resulted in a trend toward amacrine cell loss and decreased ChAT protein levels. CONCLUSIONS There is wide disparity in mouse rAION induction levels using standardized parameters. Moderate rAION induction levels without direct retinal compromise produces isolated RGC loss, with displaced amacrine cell changes likely due to changes in RGC-amacrine communication. Severe rAION induction results in both RGC and amacrine cell loss, possibly due to intra-retinal ischemic changes.
Collapse
Affiliation(s)
- Steven L Bernstein
- Departments of Ophthalmology, University of Maryland at Baltimore, School of Medicine, Baltimore, Maryland 21201, USA.
| | | |
Collapse
|
40
|
Schmidt TM, Kofuji P. An isolated retinal preparation to record light response from genetically labeled retinal ganglion cells. J Vis Exp 2011:2367. [PMID: 21307827 DOI: 10.3791/2367] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The first steps in vertebrate vision take place when light stimulates the rod and cone photoreceptors of the retina. This information is then segregated into what are known as the ON and OFF pathways. The photoreceptors signal light information to the bipolar cells (BCs), which depolarize in response to increases (On BCs) or decreases (Off BCs) in light intensity. This segregation of light information is maintained at the level of the retinal ganglion cells (RGCs), which have dendrites stratifying in either the Off sublamina of the inner plexiform layer (IPL), where they receive direct excitatory input from Off BCs, or stratifying in the On sublamina of the IPL, where they receive direct excitatory input from On BCs. This segregation of information regarding increases or decreases in illumination (the On and Off pathways) is conserved and signaled to the brain in parallel. The RGCs are the output cells of the retina, and are thus an important cell to study in order to understand how light information is signaled to visual nuclei in the brain. Advances in mouse genetics over recent decades have resulted in a variety of fluorescent reporter mouse lines where specific RGC populations are labeled with a fluorescent protein to allow for identification of RGC subtypes and specific targeting for electrophysiological recording. Here, we present a method for recording light responses from fluorescently labeled ganglion cells in an intact, isolated retinal preparation. This isolated retinal preparation allows for recordings from RGCs where the dendritic arbor is intact and the inputs across the entire RGC dendritic arbor are preserved. This method is applicable across a variety of ganglion cell subtypes and is amenable to a wide variety of single-cell physiological techniques.
Collapse
|
41
|
Gallagher SK, Witkovsky P, Roux MJ, Low MJ, Otero-Corchon V, Hentges ST, Vigh J. beta-Endorphin expression in the mouse retina. J Comp Neurol 2010; 518:3130-48. [PMID: 20533364 PMCID: PMC3095846 DOI: 10.1002/cne.22387] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Evidence showing expression of endogenous opioids in the mammalian retina is sparse. In the present study we examined a transgenic mouse line expressing an obligate dimerized form of Discosoma red fluorescent protein (DsRed) under the control of the pro-opiomelanocortin promoter and distal upstream regulatory elements to assess whether pro-opiomelanocortin peptide (POMC), and its opioid cleavage product, beta-endorphin, are expressed in the mouse retina. Using double label immunohistochemistry we found that DsRed fluorescence was restricted to a subset of GAD-67-positive cholinergic amacrine cells of both orthotopic and displaced subtypes. About 50% of cholinergic amacrine cells colocalized DsRed and a large fraction of DsRed-expressing amacrine cells was positive for beta-endorphin immunostaining, whereas beta-endorphin-immunoreactive neurons were absent in retinas of POMC null mice. Our findings contribute to a growing body of evidence demonstrating that opioid peptides are an integral component of vertebrate retinas, including those of mammals.
Collapse
Affiliation(s)
- Shannon K. Gallagher
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Paul Witkovsky
- Department of Ophthalmology, New York University School of Medicine, New York, NY 10016, USA
| | - Michel J. Roux
- Department of Neurobiology and Genetics, IGBMC, CNRS UMR 7104, Inserm U 964, Université de Strasbourg, F-67404 Illkirch, France
| | - Malcolm J. Low
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Veronica Otero-Corchon
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shane T. Hentges
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Jozsef Vigh
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
42
|
Laminar restriction of retinal ganglion cell dendrites and axons: subtype-specific developmental patterns revealed with transgenic markers. J Neurosci 2010; 30:1452-62. [PMID: 20107072 DOI: 10.1523/jneurosci.4779-09.2010] [Citation(s) in RCA: 217] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Retinal ganglion cells (RGCs), which transfer information from the eye to the brain, are heterogeneous in structure and function, but developmental studies have generally treated them as a single group. Here, we investigate the development of RGC axonal and dendritic arbors using four mouse transgenic lines in which nonoverlapping subsets of RGCs are indelibly labeled with a fluorescent protein. Each subset has a distinct functional signature, size, and morphology. Dendrites of each subset are restricted to specific sublaminae within the inner plexiform layer in adulthood, but acquire their restriction in different ways: one subset has lamina-restricted dendrites from an early postnatal stage, a second remodels an initially diffuse pattern, and two others develop stepwise. Axons of each subset arborize in discrete laminar zones within the lateral geniculate nucleus or superior colliculus, demonstrating previously unrecognized subdivisions of retinorecipient layers. As is the case for dendrites, lamina-restricted axonal projections of RGC subsets develop in different ways. For example, while axons of two RGC subsets arborize in definite zones of the superior colliculus from an early postnatal stage, axons of another subset initially occupy a deep layer, then translocate to a narrow subpial zone. Together, these results show that RGC subsets use a variety of strategies to construct lamina-restricted dendritic and axonal arbors. Taking account of these subtype-specific features will facilitate identification of the molecules and cells that regulate arbor formation.
Collapse
|
43
|
Sanes JR, Zipursky SL. Design principles of insect and vertebrate visual systems. Neuron 2010; 66:15-36. [PMID: 20399726 DOI: 10.1016/j.neuron.2010.01.018] [Citation(s) in RCA: 385] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2010] [Indexed: 01/26/2023]
Abstract
A century ago, Cajal noted striking similarities between the neural circuits that underlie vision in vertebrates and flies. Over the past few decades, structural and functional studies have provided strong support for Cajal's view. In parallel, genetic studies have revealed some common molecular mechanisms controlling development of vertebrate and fly visual systems and suggested that they share a common evolutionary origin. Here, we review these shared features, focusing on the first several layers-retina, optic tectum (superior colliculus), and lateral geniculate nucleus in vertebrates; and retina, lamina, and medulla in fly. We argue that vertebrate and fly visual circuits utilize common design principles and that taking advantage of this phylogenetic conservation will speed progress in elucidating both functional strategies and developmental mechanisms, as has already occurred in other areas of neurobiology ranging from electrical signaling and synaptic plasticity to neurogenesis and axon guidance.
Collapse
Affiliation(s)
- Joshua R Sanes
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA.
| | | |
Collapse
|
44
|
Puthussery T, Gayet-Primo J, Taylor WR. Localization of the calcium-binding protein secretagogin in cone bipolar cells of the mammalian retina. J Comp Neurol 2010; 518:513-25. [PMID: 20020539 DOI: 10.1002/cne.22234] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Secretagogin, a recently cloned member of the EF-hand family of calcium binding proteins, was localized in the mouse, rat, and rabbit retina using immunofluorescence immunohistochemistry. Secretagogin is expressed in subpopulations of ON and OFF cone bipolar cells; however, no immunoreactivity was observed in rod bipolar cells in any of these species. Using subtype-specific markers and mice expressing green fluorescent protein (GFP) within specific cell classes, we found that secretagogin is expressed in Types 2, 3, 4, 5, 6 and possibly Type 8 cone bipolar cells in the mouse retina. The expression pattern in the rat retina differs slightly with expression in cone bipolar cell Types 2, 5, 6, 7, and 8. Evaluation of secretagogin in the developing mouse retina revealed expression as early as postnatal day 6, with OFF cone bipolar cells showing secretagogin expression prior to the ON cone bipolar cells. Secretagogin is a useful marker of cone bipolar cells for studying alterations in bipolar cell morphology during development and degeneration. Further work will be necessary to elucidate the functional role of this protein in bipolar cells.
Collapse
Affiliation(s)
- Theresa Puthussery
- Casey Eye Institute, Department of Ophthalmology, Oregon Health and Sciences University, Portland, Oregon 97239, USA.
| | | | | |
Collapse
|
45
|
Ivanova E, Hwang GS, Pan ZH. Characterization of transgenic mouse lines expressing Cre recombinase in the retina. Neuroscience 2009; 165:233-43. [PMID: 19837136 DOI: 10.1016/j.neuroscience.2009.10.021] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Revised: 10/09/2009] [Accepted: 10/13/2009] [Indexed: 11/25/2022]
Abstract
The mammalian retina consists of five major classes of neuronal cells, as well as glial cells, and it contains more than 50 cell types. The ability to manipulate gene expression in specific cell type(s) in the retina is important for understanding the molecular mechanisms of retinal function and diseases. The Cre/LoxP recombination system has become a powerful tool, allowing gene deletion, over-expression, and ectopic expression in vivo in a cell- and tissue-specific fashion. The key to this tool is the availability of Cre mouse lines with cell- or tissue-type specific expression of Cre recombinase. To date, a large number of Cre-transgenic mouse lines have been generated to target Cre recombinase expression to specific neuronal and glial cell populations in the central nervous system; however, information about the expression patterns of Cre recombinase lines in the retina is largely lacking. In this study, we examined and characterized the expression patterns of Cre recombinase in the retinas of 15 Cre-transgenic mouse lines. Significant Cre-induced recombination or expression of Cre recombinase was observed in the majority of these lines. In particular, we found several Cre lines in which the Cre-induced recombination was found to target exclusively or predominantly a single type or class of retinal cells, including bistratified retinal ganglion cells, starburst amacrine cells, rod bipolar cells, and Müller glial cells. In other lines, the Cre-induced recombination was found in several retinal cell types. These Cre lines provide a valuable resource for retinal research.
Collapse
Affiliation(s)
- E Ivanova
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | |
Collapse
|
46
|
Lu Q, Ivanova E, Pan ZH. Characterization of green fluorescent protein-expressing retinal cone bipolar cells in a 5-hydroxytryptamine receptor 2a transgenic mouse line. Neuroscience 2009; 163:662-8. [PMID: 19589372 PMCID: PMC2769501 DOI: 10.1016/j.neuroscience.2009.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 07/01/2009] [Accepted: 07/02/2009] [Indexed: 10/20/2022]
Abstract
Retinal bipolar cells relay visual information from photoreceptors to third-order retinal neurons. Bipolar cells, comprising multiple types, play an essential role in segregating visual information into multiple parallel pathways in the retina. The identification of molecular markers that can label specific retinal bipolar cells could facilitate the investigation of bipolar cell functions in the retina. Transgenic mice with specific cell type(s) labeled with green fluorescent protein (GFP) have become a powerful tool for morphological and functional studies of neurons in the CNS, including the retina. In this study, we report a 5-hydroxytryptamine receptor 2a (5-HTR2a) transgenic mouse line in which expression of GFP was observed in two populations of bipolar cells in the retina. Based on the terminal stratification and immunostaining, all the strongly GFP-labeled bipolar cells were found to be type 4 cone bipolar cells. A small population of weakly labeled bipolar cells was also observed, which may represent type 8 or 9 cone bipolar cells. GFP expression in retinal cone bipolar cells was seen as early as postnatal day 5. In addition, despite severe retinal degeneration due to the presence of the rd1 mutation in this transgenic line, the density of GFP-labeled cone bipolar cells remained stable up to at least 6 months of age. This transgenic mouse line will be a useful tool for the study of type 4 cone bipolar cells in the retina under both normal and disease conditions.
Collapse
Affiliation(s)
- Q Lu
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, 540 East Canfield Avenue, Detroit, MI 48201, USA
| | | | | |
Collapse
|
47
|
Templeton JP, Nassr M, Vazquez-Chona F, Freeman-Anderson NE, Orr WE, Williams RW, Geisert EE. Differential response of C57BL/6J mouse and DBA/2J mouse to optic nerve crush. BMC Neurosci 2009; 10:90. [PMID: 19643015 PMCID: PMC2727955 DOI: 10.1186/1471-2202-10-90] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Accepted: 07/30/2009] [Indexed: 01/02/2023] Open
Abstract
Background Retinal ganglion cell (RGC) death is the final consequence of many blinding diseases, where there is considerable variation in the time course and severity of RGC loss. Indeed, this process appears to be influenced by a wide variety of genetic and environmental factors. In this study we explored the genetic basis for differences in ganglion cell death in two inbred strains of mice. Results We found that RGCs are more susceptible to death following optic nerve crush in C57BL/6J mice (54% survival) than in DBA/2J mice (62% survival). Using the Illumina Mouse-6 microarray, we identified 1,580 genes with significant change in expression following optic nerve crush in these two strains of mice. Our analysis of the changes occurring after optic nerve crush demonstrated that the greatest amount of change (44% of the variance) was due to the injury itself. This included changes associated with ganglion cell death, reactive gliosis, and abortive regeneration. The second pattern of gene changes (23% of the variance) was primarily related to differences in gene expressions observed between the C57BL/6J and DBA/2J mouse strains. The remaining changes in gene expression represent interactions between the effects of optic nerve crush and the genetic background of the mouse. We extracted one genetic network from this dataset that appears to be related to tissue remodeling. One of the most intriguing sets of changes included members of the crystallin family of genes, which may represent a signature of pathways modulating the susceptibility of cells to death. Conclusion Differential responses to optic nerve crush between two widely used strains of mice were used to define molecular networks associated with ganglion cell death and reactive gliosis. These results form the basis for our continuing interest in the modifiers of retinal injury.
Collapse
Affiliation(s)
- Justin P Templeton
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis TN, 38163, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Wässle H, Heinze L, Ivanova E, Majumdar S, Weiss J, Harvey RJ, Haverkamp S. Glycinergic transmission in the Mammalian retina. Front Mol Neurosci 2009; 2:6. [PMID: 19924257 PMCID: PMC2777502 DOI: 10.3389/neuro.02.006.2009] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Accepted: 06/21/2009] [Indexed: 11/24/2022] Open
Abstract
Glycine and γ-aminobutyric acid (GABA) are the major inhibitory neurotransmitters in the retina. Approximately half of the amacrine cells release glycine at their synapses with bipolar, other amacrine, and ganglion cells. Glycinergic amacrine cells are small-field amacrine cells with vertically oriented dendrites and comprise more than 10 different morphological types. The retinal distributions of glycine receptor (GlyR) α1, α2, α3 and α4 subtypes have been mapped with subunit-specific antibodies. GlyRs were clustered at postsynaptic hot spots which showed selective distributions for the different subunits. As a rule, only one α subunit was expressed at a given postsynaptic site. The kinetic properties of GlyRs were measured by recording spontaneous inhibitory postsynaptic currents (sIPSCs) from identified retinal neurons in wild-type, Glra1spd-ot, Glra2 and Glra3 knockout mice. From observed differences of sIPSCs in wild-type and mutant mice, the cell-type specific subunit composition of GlyRs could be defined. OFF-cone bipolar cells and A-type ganglion cells receive prominent glycinergic input with fast kinetics that is mainly mediated by α1β GlyRs (decay time constant τ ∼ 5 ms). By contrast, AII amacrine cells express α3β GlyRs with medium fast kinetics (τ ∼ 11 ms). Narrow-field (NF) and wide-field amacrine cells contain predominantly α2β GlyRs with slow kinetics (τ ∼ 27 ms). Lastly, ON-starburst, narrow-field and wide-field amacrine cells in Glra2 knockout mice express α4β GlyRs with very slow kinetics (τ ∼ 70 ms).
Collapse
Affiliation(s)
- Heinz Wässle
- Department of Neuroanatomy, Max-Planck-Institute for Brain Research Frankfurt, Germany
| | | | | | | | | | | | | |
Collapse
|