1
|
Hanics J, Tretiakov EO, Romanov RA, Gáspárdy A, Hevesi Z, Schnell R, Harkany T, Alpár A. Neuronal activity modulates the expression of secretagogin, a Ca 2+ sensor protein, during mammalian forebrain development. Acta Physiol (Oxf) 2025; 241:e70031. [PMID: 40165367 PMCID: PMC11959173 DOI: 10.1111/apha.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 04/02/2025]
Abstract
AIM Because of their stable expression, some EF-hand Ca2+-binding proteins are broadly used as histochemical markers of neurons in the nervous system. Secretagogin is a member of "neuron-specific" Ca2+-sensor proteins, yet variations in its expression due, chiefly, to neuronal activity remain ambiguous. We aimed to fill this gap of knowledge both in its use as a cell identity marker and for mechanistic analysis. METHODS We mapped secretagogin distribution in human foetal forebrains. Then, Scgn-iCre::Ai9 mice in conjunction with single-cell RNA-seq were used to molecularly characterize cortical secretagogin-expressing neurons. Besides the in vitro manipulation of both SH-SY5Y neuroblastoma cells and primary cortical cultures from foetal mice, the activity dependence of secretagogin expression was also studied upon systemic kainate administration and dark rearing. RESULTS In the mammalian brain, including humans, both transient and stable secretagogin expression sites exist. In the cerebral cortex, we identified deep-layer pyramidal neurons with lifelong expression of secretagogin. Secretagogin expression was affected by neuronal activity: it was delayed in a cohort of human foetuses with Down's syndrome relative to age-matched controls. In mice, dark rearing reduced secretagogin expression in the superior colliculus, a midbrain structure whose development is dependent on topographic visual inputs. In contrast, excitation by both KCl exposure of SH-SY5Y cells and primary cortical neurons in vitro and through systemic kainate administration in mice increased secretagogin expression. CONCLUSION We suggest that secretagogin expression in neurons is developmentally regulated and activity dependent.
Collapse
Affiliation(s)
- János Hanics
- Department of AnatomySemmelweis UniversityBudapestHungary
- SE NAP Research Group of Experimental Neuroanatomy and Developmental BiologySemmelweis UniversityBudapestHungary
| | - Evgenii O. Tretiakov
- Department of Molecular NeurosciencesCenter for Brain Research, Medical University of ViennaViennaAustria
| | - Roman A. Romanov
- Department of Molecular NeurosciencesCenter for Brain Research, Medical University of ViennaViennaAustria
| | - Anna Gáspárdy
- Department of AnatomySemmelweis UniversityBudapestHungary
| | - Zsófia Hevesi
- Department of Molecular NeurosciencesCenter for Brain Research, Medical University of ViennaViennaAustria
| | - Robert Schnell
- Department of Molecular NeurosciencesCenter for Brain Research, Medical University of ViennaViennaAustria
- Department of NeuroscienceBiomedicum 7D, Karolinska InstitutetSolnaSweden
| | - Tibor Harkany
- Department of Molecular NeurosciencesCenter for Brain Research, Medical University of ViennaViennaAustria
- Department of NeuroscienceBiomedicum 7D, Karolinska InstitutetSolnaSweden
| | - Alán Alpár
- Department of AnatomySemmelweis UniversityBudapestHungary
- SE NAP Research Group of Experimental Neuroanatomy and Developmental BiologySemmelweis UniversityBudapestHungary
| |
Collapse
|
2
|
Procyk CA, Melati A, Ribeiro J, Liu J, Branch MJ, Delicata JD, Tariq M, Kalarygrou AA, Kapadia J, Khorsani MM, West EL, Smith AJ, Gonzalez-Cordero A, Ali RR, Pearson RA. Human cone photoreceptor transplantation stimulates remodeling and restores function in AIPL1 model of end-stage Leber congenital amaurosis. Stem Cell Reports 2025; 20:102470. [PMID: 40154478 PMCID: PMC12069896 DOI: 10.1016/j.stemcr.2025.102470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025] Open
Abstract
Photoreceptor degeneration is a leading cause of untreatable sight loss. Previously, we showed that human pluripotent stem cell-derived cone photoreceptors (hCones) can rescue retinal function in the Rd1 mouse model of rod-cone dystrophy. However, retinal degenerations display markedly different severities and concomitant remodeling of the remaining retina; for photoreceptor replacement therapy to be broadly effective, it must work for a variety of disease phenotypes. Here, we sought to rescue the Aipl1-/- model of Leber congenital amaurosis, a particularly fast, severe condition. After transplantation of hCones, host cone bipolar cells underwent extensive remodeling and formed nascent synaptic-like connections. Electrophysiological recordings showed robust rescue of light-evoked activity across visually relevant photopic intensities, and treated mice exhibited visually evoked optokinetic head-tracking behavior. Thus, human cone photoreceptor replacement therapy is feasible even in very severe cases of retinal dystrophy, offering promise as a disease-agnostic therapy in Leber congenital amaurosis (LCA) and in other advanced retinal degenerations.
Collapse
Affiliation(s)
- Christopher A Procyk
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Anna Melati
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Joana Ribeiro
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Jingshu Liu
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Matthew J Branch
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Jamie D Delicata
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Menahil Tariq
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Aikaterini A Kalarygrou
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Jessica Kapadia
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Majid Moshtagh Khorsani
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Emma L West
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Alexander J Smith
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Anai Gonzalez-Cordero
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Robin R Ali
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Rachael A Pearson
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London SE1 9RT, UK.
| |
Collapse
|
3
|
Ganzen L, Yadav SC, Wei M, Ma H, Nawy S, Kramer RH. Retinoic Acid-Dependent Loss of Synaptic Output from Bipolar Cells Impairs Visual Information Processing in Inherited Retinal Degeneration. J Neurosci 2024; 44:e0129242024. [PMID: 39060177 PMCID: PMC11358532 DOI: 10.1523/jneurosci.0129-24.2024] [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: 01/18/2024] [Revised: 07/09/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
In retinitis pigmentosa (RP), rod and cone photoreceptors degenerate, depriving downstream neurons of light-sensitive input, leading to vision impairment or blindness. Although downstream neurons survive, some undergo morphological and physiological remodeling. Bipolar cells (BCs) link photoreceptors, which sense light, to retinal ganglion cells (RGCs), which send information to the brain. While photoreceptor loss disrupts input synapses to BCs, whether BC output synapses remodel has remained unknown. Here we report that synaptic output from BCs plummets in RP mouse models of both sexes owing to loss of voltage-gated Ca2+ channels. Remodeling reduces the reliability of synaptic output to repeated optogenetic stimuli, causing RGC firing to fail at high-stimulus frequencies. Fortunately, functional remodeling of BCs can be reversed by inhibiting the retinoic acid receptor (RAR). RAR inhibitors targeted to BCs present a new therapeutic opportunity for mitigating detrimental effects of remodeling on signals initiated either by surviving photoreceptors or by vision-restoring tools.
Collapse
Affiliation(s)
- Logan Ganzen
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Shubhash Chandra Yadav
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Mingxiao Wei
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Hong Ma
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Scott Nawy
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Richard H Kramer
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| |
Collapse
|
4
|
Téllez de Meneses PG, Pérez-Revuelta L, Canal-Alonso Á, Hernández-Pérez C, Cocho T, Valero J, Weruaga E, Díaz D, Alonso JR. Immunohistochemical distribution of secretagogin in the mouse brain. Front Neuroanat 2023; 17:1224342. [PMID: 37711587 PMCID: PMC10498459 DOI: 10.3389/fnana.2023.1224342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/09/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Calcium is essential for the correct functioning of the central nervous system, and calcium-binding proteins help to finely regulate its concentration. Whereas some calcium-binding proteins such as calmodulin are ubiquitous and are present in many cell types, others such as calbindin, calretinin, and parvalbumin are expressed in specific neuronal populations. Secretagogin belongs to this latter group and its distribution throughout the brain is only partially known. In the present work, the distribution of secretagogin-immunopositive cells was studied in the entire brain of healthy adult mice. Methods Adult male C57BL/DBA mice aged between 5 and 7 months were used. Their whole brain was sectioned and used for immunohistochemistry. Specific neural populations were observed in different zones and nuclei identified according to Paxinos mouse brain atlas. Results Labelled cells were found with a Golgi-like staining, allowing an excellent characterization of their dendritic and axonal arborizations. Many secretagogin-positive cells were observed along different encephalic regions, especially in the olfactory bulb, basal ganglia, and hypothalamus. Immunostained populations were very heterogenous in both size and distribution, as some nuclei presented labelling in their entire extension, but in others, only scattered cells were present. Discussion Secretagogin can provide a more complete vision of calcium-buffering mechanisms in the brain, and can be a useful neuronal marker in different brain areas for specific populations.
Collapse
Affiliation(s)
- Pablo G. Téllez de Meneses
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Laura Pérez-Revuelta
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Ángel Canal-Alonso
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Bioinformatics, Intelligent Systems and Educational Technology (BISITE) Research Group, Universidad de Salamanca, Salamanca, Spain
| | - Carlos Hernández-Pérez
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Teresa Cocho
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Jorge Valero
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Eduardo Weruaga
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - David Díaz
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - José R. Alonso
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| |
Collapse
|
5
|
Balaji V, Haverkamp S, Seth PK, Günther A, Mendoza E, Schmidt J, Herrmann M, Pfeiffer LL, Němec P, Scharff C, Mouritsen H, Dedek K. Immunohistochemical characterization of bipolar cells in four distantly related avian species. J Comp Neurol 2023; 531:561-581. [PMID: 36550622 DOI: 10.1002/cne.25443] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
Visual (and probably also magnetic) signal processing starts at the first synapse, at which photoreceptors contact different types of bipolar cells, thereby feeding information into different processing channels. In the chicken retina, 15 and 22 different bipolar cell types have been identified based on serial electron microscopy and single-cell transcriptomics, respectively. However, immunohistochemical markers for avian bipolar cells were only anecdotally described so far. Here, we systematically tested 12 antibodies for their ability to label individual bipolar cells in the bird retina and compared the eight most suitable antibodies across distantly related species, namely domestic chicken, domestic pigeon, common buzzard, and European robin, and across retinal regions. While two markers (GNB3 and EGFR) labeled specifically ON bipolar cells, most markers labeled in addition to bipolar cells also other cell types in the avian retina. Staining pattern of four markers (CD15, PKCα, PKCβ, secretagogin) was species-specific. Two markers (calbindin and secretagogin) showed a different expression pattern in central and peripheral retina. For the chicken and European robin, we found slightly more ON bipolar cell somata in the inner nuclear layer than OFF bipolar cell somata. In contrast, OFF bipolar cells made more ribbon synapses than ON bipolar cells in the inner plexiform layer of these species. Finally, we also analyzed the photoreceptor connectivity of selected bipolar cell types in the European robin retina. In summary, we provide a catalog of bipolar cell markers for different bird species, which will greatly facilitate analyzing the retinal circuitry of birds on a larger scale.
Collapse
Affiliation(s)
- Vaishnavi Balaji
- Animal Navigation/Neurosensorics Group, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Silke Haverkamp
- Department Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior - caesar, Bonn, Germany
| | - Pranav Kumar Seth
- Animal Navigation/Neurosensorics Group, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Anja Günther
- Animal Navigation/Neurosensorics Group, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany.,Department Computational Neuroethology, Max Planck Institute for Neurobiology of Behavior - caesar, Bonn, Germany
| | - Ezequiel Mendoza
- Institut für Biologie, Freie Universität Berlin, Berlin, Germany
| | - Jessica Schmidt
- Animal Navigation/Neurosensorics Group, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Maike Herrmann
- Animal Navigation/Neurosensorics Group, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Leonie Lovis Pfeiffer
- Animal Navigation/Neurosensorics Group, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Pavel Němec
- Department of Zoology, Charles University, Prague, Czech Republic
| | | | - Henrik Mouritsen
- Animal Navigation/Neurosensorics Group, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| | - Karin Dedek
- Animal Navigation/Neurosensorics Group, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
6
|
Albrecht NE, Jiang D, Akhanov V, Hobson R, Speer CM, Robichaux MA, Samuel MA. Rapid 3D-STORM imaging of diverse molecular targets in tissue. CELL REPORTS METHODS 2022; 2:100253. [PMID: 35880013 PMCID: PMC9308169 DOI: 10.1016/j.crmeth.2022.100253] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/22/2022] [Accepted: 06/17/2022] [Indexed: 11/25/2022]
Abstract
Fine-scale molecular architecture is critical for nervous system and other biological functions. Methods to visualize these nanoscale structures would benefit from enhanced accessibility, throughput, and tissue compatibility. Here, we report RAIN-STORM, a rapid and scalable nanoscopic imaging optimization approach that improves three-dimensional visualization for subcellular targets in tissue at depth. RAIN-STORM uses conventional tissue samples and readily available reagents and is suitable for commercial instrumentation. To illustrate the efficacy of RAIN-STORM, we utilized the retina. We show that RAIN-STORM imaging is versatile and provide 3D nanoscopic data for over 20 synapse, neuron, glia, and vasculature targets. Sample preparation is also rapid, with a 1-day turnaround from tissue to image, and parameters are suitable for multiple tissue sources. Finally, we show that this method can be applied to clinical samples to reveal nanoscale features of human cells and synapses. RAIN-STORM thus paves the way for high-throughput studies of nanoscopic targets in tissue.
Collapse
Affiliation(s)
- Nicholas E. Albrecht
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Danye Jiang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Viktor Akhanov
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Robert Hobson
- Bruker Nano Surfaces Division, Salt Lake City, UT 84108, USA
| | - Colenso M. Speer
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Michael A. Robichaux
- Departments of Ophthalmology and Biochemistry, West Virginia University, Morgantown, WV 26506, USA
| | - Melanie A. Samuel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
7
|
Sinha T, Ikelle L, Makia MS, Crane R, Zhao X, Kakakhel M, Al-Ubaidi MR, Naash MI. Riboflavin deficiency leads to irreversible cellular changes in the RPE and disrupts retinal function through alterations in cellular metabolic homeostasis. Redox Biol 2022; 54:102375. [PMID: 35738087 PMCID: PMC9233280 DOI: 10.1016/j.redox.2022.102375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 10/25/2022] Open
Abstract
Ariboflavinosis is a pathological condition occurring as a result of riboflavin deficiency. This condition is treatable if detected early enough, but it lacks timely diagnosis. Critical symptoms of ariboflavinosis include neurological and visual manifestations, yet the effects of flavin deficiency on the retina are not well investigated. Here, using a diet induced mouse model of riboflavin deficiency, we provide the first evidence of how retinal function and metabolism are closely intertwined with riboflavin homeostasis. We find that diet induced riboflavin deficiency causes severe decreases in retinal function accompanied by structural changes in the neural retina and retinal pigment epithelium (RPE). This is preceded by increased signs of cellular oxidative stress and metabolic disorder, in particular dysregulation in lipid metabolism, which is essential for both photoreceptors and the RPE. Though many of these deleterious phenotypes can be ameliorated by riboflavin supplementation, our data suggests that some patients may continue to suffer from multiple pathologies at later ages. These studies provide an essential cellular and mechanistic foundation linking defects in cellular flavin levels with the manifestation of functional deficiencies in the visual system and paves the way for a more in-depth understanding of the cellular consequences of ariboflavinosis.
Collapse
Affiliation(s)
- Tirthankar Sinha
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Larissa Ikelle
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Mustafa S Makia
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Ryan Crane
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Xue Zhao
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Mashal Kakakhel
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Muayyad R Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
8
|
Basu S, Mitra S, Singh O, Chandramohan B, Singru PS. Secretagogin in the brain and pituitary of the catfish, Clarias batrachus: Molecular characterization and regulation by insulin. J Comp Neurol 2022; 530:1743-1772. [PMID: 35322425 DOI: 10.1002/cne.25311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 11/12/2022]
Abstract
Secretagogin (scgn), is a novel hexa EF-hand, phylogenetically conserved calcium-binding protein. It serves as Ca2+ sensor and participates in Ca2+ -signaling and neuroendocrine regulation in mammals. However, its relevance in the brain of non-mammalian vertebrates has largely remained unexplored. To address this issue, we studied the cDNA encoding scgn, scgn mRNA expression, and distribution of scgn-equipped elements in the brain and pituitary of a teleost, Clarias batrachus (cb). The cbscgn cDNA consists of three transcripts (T) variants: T1 (2185 bp), T2 (2151 bp) and T3 (2060 bp). While 816 bp ORF in T1 and T2 encodes highly conserved six EF-hand 272 aa protein fully capable of Ca2+ -binding, 726-bp ORF in T3 encodes 242 aa protein. The T1 showed >90% and >70% identity with scgn of catfishes, and other teleosts and mammals, respectively. The T1-mRNA was widely expressed in the brain and pituitary, while the expression of T3 was restricted to the telencephalon. Application of the anti-scgn antiserum revealed a ∼32 kDa scgn-immunoreactive (scgn-i) band (known molecular weight of scgn) in the forebrain tissue, and immunohistochemically labeled neurons in the olfactory epithelium and bulb, telencephalon, preoptic area, hypothalamus, thalamus, and hindbrain. In the pituitary, scgn-i cells were seen in the pars distalis and intermedia. Insulin is reported to regulate scgn mRNA in the mammalian hippocampus, and feeding-related neuropeptides in the telencephalon of teleost. Intracranial injection of insulin significantly increased T1-mRNA expression and scgn-immunoreactivity in the telencephalon. We suggest that scgn may be an important player in the regulation of olfactory, neuroendocrine system, and energy balance functions in C. batrachus.
Collapse
Affiliation(s)
- Sumela Basu
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Saptarsi Mitra
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Omprakash Singh
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Bathrachalam Chandramohan
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India
| | - Praful S Singru
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
9
|
Kajtna J, Tsang SH, Koch SF. Late-stage rescue of visually guided behavior in the context of a significantly remodeled retinitis pigmentosa mouse model. Cell Mol Life Sci 2022; 79:148. [PMID: 35195763 PMCID: PMC8866266 DOI: 10.1007/s00018-022-04161-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/13/2022] [Accepted: 01/19/2022] [Indexed: 11/25/2022]
Abstract
Patients with progressive neurodegenerative disorder retinitis pigmentosa (RP) are diagnosed in the midst of ongoing retinal degeneration and remodeling. Here, we used a Pde6b-deficient RP gene therapy mouse model to test whether treatment at late disease stages can halt photoreceptor degeneration and degradative remodeling, while sustaining constructive remodeling and restoring function. We demonstrated that when fewer than 13% of rods remain, our genetic rescue halts photoreceptor degeneration, electroretinography (ERG) functional decline and inner retinal remodeling. In addition, in a water maze test, the performance of mice treated at 16 weeks of age or earlier was indistinguishable from wild type. In contrast, no efficacy was apparent in mice treated at 24 weeks of age, suggesting the photoreceptors had reached a point of no return. Further, remodeling in the retinal pigment epithelium (RPE) and retinal vasculature was not halted at 16 or 24 weeks of age, although there appeared to be some slowing of blood vessel degradation. These data suggest a novel working model in which restoration of clinically significant visual function requires only modest threshold numbers of resilient photoreceptors, halting of destructive remodeling and sustained constructive remodeling. These novel findings define the potential and limitations of RP treatment and suggest possible nonphotoreceptor targets for gene therapy optimization.
Collapse
Affiliation(s)
- Jacqueline Kajtna
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
- Physiological Genomics, BioMedical Center, Ludwig-Maximilians-Universität München, Planegg/Martinsried, Germany
| | - Stephen H Tsang
- Jonas Children's Vision Care, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, 10032, USA
| | - Susanne F Koch
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany.
- Physiological Genomics, BioMedical Center, Ludwig-Maximilians-Universität München, Planegg/Martinsried, Germany.
| |
Collapse
|
10
|
Behrens C, Yadav SC, Korympidou MM, Zhang Y, Haverkamp S, Irsen S, Schaedler A, Lu X, Liu Z, Lause J, St-Pierre F, Franke K, Vlasits A, Dedek K, Smith RG, Euler T, Berens P, Schubert T. Retinal horizontal cells use different synaptic sites for global feedforward and local feedback signaling. Curr Biol 2022; 32:545-558.e5. [PMID: 34910950 PMCID: PMC8886496 DOI: 10.1016/j.cub.2021.11.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 10/19/2021] [Accepted: 11/23/2021] [Indexed: 11/19/2022]
Abstract
In the outer plexiform layer (OPL) of the mammalian retina, cone photoreceptors (cones) provide input to more than a dozen types of cone bipolar cells (CBCs). In the mouse, this transmission is modulated by a single horizontal cell (HC) type. HCs perform global signaling within their laterally coupled network but also provide local, cone-specific feedback. However, it is unknown how HCs provide local feedback to cones at the same time as global forward signaling to CBCs and where the underlying synapses are located. To assess how HCs simultaneously perform different modes of signaling, we reconstructed the dendritic trees of five HCs as well as cone axon terminals and CBC dendrites in a serial block-face electron microscopy volume and analyzed their connectivity. In addition to the fine HC dendritic tips invaginating cone axon terminals, we also identified "bulbs," short segments of increased dendritic diameter on the primary dendrites of HCs. These bulbs are in an OPL stratum well below the cone axon terminal base and make contacts with other HCs and CBCs. Our results from immunolabeling, electron microscopy, and glutamate imaging suggest that HC bulbs represent GABAergic synapses that do not receive any direct photoreceptor input. Together, our data suggest the existence of two synaptic strata in the mouse OPL, spatially separating cone-specific feedback and feedforward signaling to CBCs. A biophysical model of a HC dendritic branch and voltage imaging support the hypothesis that this spatial arrangement of synaptic contacts allows for simultaneous local feedback and global feedforward signaling by HCs.
Collapse
Affiliation(s)
- Christian Behrens
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Bernstein Center for Computational Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, Otfried-Müller-Str. 27, 72076 Tübingen, Germany
| | - Shubhash Chandra Yadav
- Neurosensorics/Animal Navigation, Institute for Biology and Environmental Sciences, University of Oldenburg, Carl-von-Ossietzky-Str. 9-11, 26111 Oldenburg, Germany
| | - Maria M Korympidou
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, Otfried-Müller-Str. 27, 72076 Tübingen, Germany
| | - Yue Zhang
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, Otfried-Müller-Str. 27, 72076 Tübingen, Germany
| | - Silke Haverkamp
- Department of Computational Neuroethology, Center of Advanced European Studies and Research (caesar), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Stephan Irsen
- Electron Microscopy and Analytics, Center of Advanced European Studies and Research (caesar), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Anna Schaedler
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, Otfried-Müller-Str. 27, 72076 Tübingen, Germany
| | - Xiaoyu Lu
- Systems, Synthetic, and Physical Biology Program, Rice University, 6500 Main St., Houston, TX 77005, USA
| | - Zhuohe Liu
- Department of Electrical and Computer Engineering, Rice University, 6100 Main St., Houston, TX 77005, USA
| | - Jan Lause
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Bernstein Center for Computational Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, Otfried-Müller-Str. 27, 72076 Tübingen, Germany
| | - François St-Pierre
- Systems, Synthetic, and Physical Biology Program, Rice University, 6500 Main St., Houston, TX 77005, USA; Department of Electrical and Computer Engineering, Rice University, 6100 Main St., Houston, TX 77005, USA; Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Katrin Franke
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Bernstein Center for Computational Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany
| | - Anna Vlasits
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Bernstein Center for Computational Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany
| | - Karin Dedek
- Neurosensorics/Animal Navigation, Institute for Biology and Environmental Sciences, University of Oldenburg, Carl-von-Ossietzky-Str. 9-11, 26111 Oldenburg, Germany
| | - Robert G Smith
- Department of Neuroscience, University of Pennsylvania, 422 Curie Blvd, Philadelphia, PA 19104, USA
| | - Thomas Euler
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Bernstein Center for Computational Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany
| | - Philipp Berens
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Bernstein Center for Computational Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany; Tübingen AI Center, University of Tübingen, Maria-von-Linden-Straße 6, 72076 Tübingen, Germany
| | - Timm Schubert
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Str. 7, 72076 Tübingen, Germany; Center for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Str. 25, 72076 Tübingen, Germany.
| |
Collapse
|
11
|
Nemitz L, Dedek K, Janssen-Bienhold U. Synaptic Remodeling in the Cone Pathway After Early Postnatal Horizontal Cell Ablation. Front Cell Neurosci 2021; 15:657594. [PMID: 34122012 PMCID: PMC8187617 DOI: 10.3389/fncel.2021.657594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/03/2021] [Indexed: 11/30/2022] Open
Abstract
The first synapse of the visual pathway is formed by photoreceptors, horizontal cells and bipolar cells. While ON bipolar cells invaginate into the photoreceptor terminal and form synaptic triads together with invaginating horizontal cell processes, OFF bipolar cells make flat contacts at the base of the terminal. When horizontal cells are ablated during retina development, no invaginating synapses are formed in rod photoreceptors. However, how cone photoreceptors and their synaptic connections with bipolar cells react to this insult, is unclear so far. To answer this question, we specifically ablated horizontal cells from the developing mouse retina. Following ablation around postnatal day 4 (P4)/P5, cones initially exhibited a normal morphology and formed flat contacts with OFF bipolar cells, but only few invaginating contacts with ON bipolar cells. From P15 on, synaptic remodeling became obvious with clustering of cone terminals and mislocalized cone somata in the OPL. Adult cones (P56) finally displayed highly branched axons with numerous terminals which contained ribbons and vesicular glutamate transporters. Furthermore, type 3a, 3b, and 4 OFF bipolar cell dendrites sprouted into the outer nuclear layer and even expressed glutamate receptors at the base of newly formed cone terminals. These results indicate that cones may be able to form new synapses with OFF bipolar cells in adult mice. In contrast, cone terminals lost their invaginating contacts with ON bipolar cells, highlighting the importance of horizontal cells for synapse maintenance. Taken together, our data demonstrate that early postnatal horizontal cell ablation leads to differential remodeling in the cone pathway: whereas synapses between cones and ON bipolar cells were lost, new putative synapses were established between cones and OFF bipolar cells. These results suggest that synapse formation and maintenance are regulated very differently between flat and invaginating contacts at cone terminals.
Collapse
Affiliation(s)
- Lena Nemitz
- Visual Neuroscience, Department of Neuroscience, University of Oldenburg, Oldenburg, Germany
| | - Karin Dedek
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| | - Ulrike Janssen-Bienhold
- Visual Neuroscience, Department of Neuroscience, University of Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
12
|
Kelemen K, Szilágyi T. New Approach for Untangling the Role of Uncommon Calcium-Binding Proteins in the Central Nervous System. Brain Sci 2021. [PMID: 34069107 DOI: 10.3390/brainsci11050634ht] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
Although Ca2+ ion plays an essential role in cellular physiology, calcium-binding proteins (CaBPs) were long used for mainly as immunohistochemical markers of specific cell types in different regions of the central nervous system. They are a heterogeneous and wide-ranging group of proteins. Their function was studied intensively in the last two decades and a tremendous amount of information was gathered about them. Girard et al. compiled a comprehensive list of the gene-expression profiles of the entire EF-hand gene superfamily in the murine brain. We selected from this database those CaBPs which are related to information processing and/or neuronal signalling, have a Ca2+-buffer activity, Ca2+-sensor activity, modulator of Ca2+-channel activity, or a yet unknown function. In this way we created a gene function-based selection of the CaBPs. We cross-referenced these findings with publicly available, high-quality RNA-sequencing and in situ hybridization databases (Human Protein Atlas (HPA), Brain RNA-seq database and Allen Brain Atlas integrated into the HPA) and created gene expression heat maps of the regional and cell type-specific expression levels of the selected CaBPs. This represents a useful tool to predict and investigate different expression patterns and functions of the less-known CaBPs of the central nervous system.
Collapse
Affiliation(s)
- Krisztina Kelemen
- Department of Physiology, Doctoral School, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania
| | - Tibor Szilágyi
- Department of Physiology, Doctoral School, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania
| |
Collapse
|
13
|
Kelemen K, Szilágyi T. New Approach for Untangling the Role of Uncommon Calcium-Binding Proteins in the Central Nervous System. Brain Sci 2021; 11:brainsci11050634. [PMID: 34069107 PMCID: PMC8156796 DOI: 10.3390/brainsci11050634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/09/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
Although Ca2+ ion plays an essential role in cellular physiology, calcium-binding proteins (CaBPs) were long used for mainly as immunohistochemical markers of specific cell types in different regions of the central nervous system. They are a heterogeneous and wide-ranging group of proteins. Their function was studied intensively in the last two decades and a tremendous amount of information was gathered about them. Girard et al. compiled a comprehensive list of the gene-expression profiles of the entire EF-hand gene superfamily in the murine brain. We selected from this database those CaBPs which are related to information processing and/or neuronal signalling, have a Ca2+-buffer activity, Ca2+-sensor activity, modulator of Ca2+-channel activity, or a yet unknown function. In this way we created a gene function-based selection of the CaBPs. We cross-referenced these findings with publicly available, high-quality RNA-sequencing and in situ hybridization databases (Human Protein Atlas (HPA), Brain RNA-seq database and Allen Brain Atlas integrated into the HPA) and created gene expression heat maps of the regional and cell type-specific expression levels of the selected CaBPs. This represents a useful tool to predict and investigate different expression patterns and functions of the less-known CaBPs of the central nervous system.
Collapse
|
14
|
Vassalli QA, Colantuono C, Nittoli V, Ferraioli A, Fasano G, Berruto F, Chiusano ML, Kelsh RN, Sordino P, Locascio A. Onecut Regulates Core Components of the Molecular Machinery for Neurotransmission in Photoreceptor Differentiation. Front Cell Dev Biol 2021; 9:602450. [PMID: 33816460 PMCID: PMC8012850 DOI: 10.3389/fcell.2021.602450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/11/2021] [Indexed: 11/13/2022] Open
Abstract
Photoreceptor cells (PRC) are neurons highly specialized for sensing light stimuli and have considerably diversified during evolution. The genetic mechanisms that underlie photoreceptor differentiation and accompanied the progressive increase in complexity and diversification of this sensory cell type are a matter of great interest in the field. A role of the homeodomain transcription factor Onecut (Oc) in photoreceptor cell formation is proposed throughout multicellular organisms. However, knowledge of the identity of the Oc downstream-acting factors that mediate specific tasks in the differentiation of the PRC remains limited. Here, we used transgenic perturbation of the Ciona robusta Oc protein to show its requirement for ciliary PRC differentiation. Then, transcriptome profiling between the trans-activation and trans-repression Oc phenotypes identified differentially expressed genes that are enriched in exocytosis, calcium homeostasis, and neurotransmission. Finally, comparison of RNA-Seq datasets in Ciona and mouse identifies a set of Oc downstream genes conserved between tunicates and vertebrates. The transcription factor Oc emerges as a key regulator of neurotransmission in retinal cell types.
Collapse
Affiliation(s)
- Quirino Attilio Vassalli
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Chiara Colantuono
- Department of Research Infrastructures for Marine Biological Resources, Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Valeria Nittoli
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Anna Ferraioli
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Giulia Fasano
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Federica Berruto
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Maria Luisa Chiusano
- Department of Research Infrastructures for Marine Biological Resources, Stazione Zoologica Anton Dohrn, Naples, Italy
- Department of Agriculture, Università degli Studi di Napoli Federico II, Portici, Italy
| | - Robert Neil Kelsh
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, University of Bath, London, United Kingdom
| | - Paolo Sordino
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Annamaria Locascio
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy
| |
Collapse
|
15
|
Striebel JF, Race B, Leung JM, Schwartz C, Chesebro B. Prion-induced photoreceptor degeneration begins with misfolded prion protein accumulation in cones at two distinct sites: cilia and ribbon synapses. Acta Neuropathol Commun 2021; 9:17. [PMID: 33509294 PMCID: PMC7845122 DOI: 10.1186/s40478-021-01120-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/09/2021] [Indexed: 12/15/2022] Open
Abstract
Accumulation of misfolded host proteins is central to neuropathogenesis of numerous human brain diseases including prion and prion-like diseases. Neurons of retina are also affected by these diseases. Previously, our group and others found that prion-induced retinal damage to photoreceptor cells in mice and humans resembled pathology of human retinitis pigmentosa caused by mutations in retinal proteins. Here, using confocal, epifluorescent and electron microscopy we followed deposition of disease-associated prion protein (PrPSc) and its association with damage to critical retinal structures following intracerebral prion inoculation. The earliest time and place of retinal PrPSc deposition was 67 days post-inoculation (dpi) on the inner segment (IS) of cone photoreceptors. At 104 and 118 dpi, PrPSc was associated with the base of cilia and swollen cone inner segments, suggesting ciliopathy as a pathogenic mechanism. By 118 dpi, PrPSc was deposited in both rods and cones which showed rootlet damage in the IS, and photoreceptor cell death was indicated by thinning of the outer nuclear layer. In the outer plexiform layer (OPL) in uninfected mice, normal host PrP (PrPC) was mainly associated with cone bipolar cell processes, but in infected mice, at 118 dpi, PrPSc was detected on cone and rod bipolar cell dendrites extending into ribbon synapses. Loss of ribbon synapses in cone pedicles and rod spherules in the OPL was observed to precede destruction of most rods and cones over the next 2–3 weeks. However, bipolar cells and horizontal cells were less damaged, indicating high selectivity among neurons for injury by prions. PrPSc deposition in cone and rod inner segments and on the bipolar cell processes participating in ribbon synapses appear to be critical early events leading to damage and death of photoreceptors after prion infection. These mechanisms may also occur in human retinitis pigmentosa and prion-like diseases, such as AD.
Collapse
|
16
|
Tsuboi A. LRR-Containing Oncofetal Trophoblast Glycoprotein 5T4 Shapes Neural Circuits in Olfactory and Visual Systems. Front Mol Neurosci 2020; 13:581018. [PMID: 33192298 PMCID: PMC7655536 DOI: 10.3389/fnmol.2020.581018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/22/2020] [Indexed: 01/19/2023] Open
Abstract
In mammals, the sensory experience can regulate the development of various brain structures, including the cortex, hippocampus, retina, and olfactory bulb (OB). Odor experience-evoked neural activity drives the development of dendrites on excitatory projection neurons in the OB, such as mitral and tufted cells, as well as inhibitory interneurons. OB interneurons are generated continuously in the subventricular zone and differentiate into granule cells (GCs) and periglomerular cells (PGCs). However, it remains unknown what role each type of OB interneuron plays in controlling olfactory behaviors. Recent studies showed that among the various types of OB interneurons, a subtype of GCs expressing oncofetal trophoblast glycoprotein 5T4 is required for simple odor detection and discrimination behaviors. Mouse 5T4 (also known as Tpbg) is a type I membrane glycoprotein whose extracellular domain contains seven leucine-rich repeats (LRRs) sandwiched between characteristic LRR-N and LRR-C regions. Recently, it was found that the developmental expression of 5T4 increases dramatically in the retina just before eye-opening. Single-cell transcriptomics further suggests that 5T4 is involved in the development and maintenance of functional synapses in a subset of retinal interneurons, including rod bipolar cells (RBCs) and amacrine cells (ACs). Collectively, 5T4, expressed in interneurons of the OB and retina, plays a key role in sensory processing in the olfactory and visual systems.
Collapse
Affiliation(s)
- Akio Tsuboi
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
17
|
Lozoya OA, McClelland KS, Papas BN, Li JL, Yao HHC. Patterns, Profiles, and Parsimony: Dissecting Transcriptional Signatures From Minimal Single-Cell RNA-Seq Output With SALSA. Front Genet 2020; 11:511286. [PMID: 33193599 PMCID: PMC7586319 DOI: 10.3389/fgene.2020.511286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 09/18/2020] [Indexed: 11/23/2022] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) technologies have precipitated the development of bioinformatic tools to reconstruct cell lineage specification and differentiation processes with single-cell precision. However, current start-up costs and recommended data volumes for statistical analysis remain prohibitively expensive, preventing scRNA-seq technologies from becoming mainstream. Here, we introduce single-cell amalgamation by latent semantic analysis (SALSA), a versatile workflow that combines measurement reliability metrics with latent variable extraction to infer robust expression profiles from ultra-sparse sc-RNAseq data. SALSA uses a matrix focusing approach that starts by identifying facultative genes with expression levels greater than experimental measurement precision and ends with cell clustering based on a minimal set of Profiler genes, each one a putative biomarker of cluster-specific expression profiles. To benchmark how SALSA performs in experimental settings, we used the publicly available 10X Genomics PBMC 3K dataset, a pre-curated silver standard from human frozen peripheral blood comprising 2,700 single-cell barcodes, and identified 7 major cell groups matching transcriptional profiles of peripheral blood cell types and driven agnostically by < 500 Profiler genes. Finally, we demonstrate successful implementation of SALSA in a replicative scRNA-seq scenario by using previously published DropSeq data from a multi-batch mouse retina experimental design, thereby identifying 10 transcriptionally distinct cell types from > 64,000 single cells across 7 independent biological replicates based on < 630 Profiler genes. With these results, SALSA demonstrates that robust pattern detection from scRNA-seq expression matrices only requires a fraction of the accrued data, suggesting that single-cell sequencing technologies can become affordable and widespread if meant as hypothesis-generation tools to extract large-scale differential expression effects.
Collapse
Affiliation(s)
- Oswaldo A. Lozoya
- Genomic Integrity & Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Kathryn S. McClelland
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Brian N. Papas
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Jian-Liang Li
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Humphrey H.-C. Yao
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| |
Collapse
|
18
|
Goodson NB, Kaufman MA, Park KU, Brzezinski JA. Simultaneous deletion of Prdm1 and Vsx2 enhancers in the retina alters photoreceptor and bipolar cell fate specification, yet differs from deleting both genes. Development 2020; 147:dev190272. [PMID: 32541005 PMCID: PMC10666920 DOI: 10.1242/dev.190272] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022]
Abstract
The transcription factor OTX2 is required for photoreceptor and bipolar cell formation in the retina. It directly activates the transcription factors Prdm1 and Vsx2 through cell type-specific enhancers. PRDM1 and VSX2 work in opposition, such that PRDM1 promotes photoreceptor fate and VSX2 bipolar cell fate. To determine how OTX2+ cell fates are regulated in mice, we deleted Prdm1 and Vsx2 or their cell type-specific enhancers simultaneously using a CRISPR/Cas9 in vivo retina electroporation strategy. Double gene or enhancer targeting effectively removed PRDM1 and VSX2 protein expression. However, double enhancer targeting favored bipolar fate outcomes, whereas double gene targeting favored photoreceptor fate. Both conditions generated excess amacrine cells. Combined, these fate changes suggest that photoreceptors are a default fate outcome in OTX2+ cells and that VSX2 must be present in a narrow temporal window to drive bipolar cell formation. Prdm1 and Vsx2 also appear to redundantly restrict the competence of OTX2+ cells, preventing amacrine cell formation. By taking a combinatorial deletion approach of both coding sequences and enhancers, our work provides new insights into the complex regulatory mechanisms that control cell fate choice.
Collapse
Affiliation(s)
- Noah B Goodson
- Sue Anschutz Rodgers Eye Center, Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael A Kaufman
- Sue Anschutz Rodgers Eye Center, Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Cell Biology, Stem Cells, and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ko U Park
- Sue Anschutz Rodgers Eye Center, Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joseph A Brzezinski
- Sue Anschutz Rodgers Eye Center, Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
19
|
Prdm1 overexpression causes a photoreceptor fate-shift in nascent, but not mature, bipolar cells. Dev Biol 2020; 464:111-123. [PMID: 32562755 DOI: 10.1016/j.ydbio.2020.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/14/2022]
Abstract
The transcription factors Prdm1 (Blimp1) and Vsx2 (Chx10) work downstream of Otx2 to regulate photoreceptor and bipolar cell fates in the developing retina. Mice that lack Vsx2 fail to form bipolar cells while Prdm1 mutants form excess bipolars at the direct expense of photoreceptors. Excess bipolars in Prdm1 mutants appear to derive from rods, suggesting that photoreceptor fate remains mutable for some time after cells become specified. Here we tested whether bipolar cell fate is also plastic during development. To do this, we created a system to conditionally misexpress Prdm1 at different stages of bipolar cell development. We found that Prdm1 blocks bipolar cell formation if expressed before the fate choice decision occurred. When we misexpressed Prdm1 just after the decision to become a bipolar cell was made, some cells were reprogrammed into photoreceptors. In contrast, Prdm1 misexpression in mature bipolar cells did not affect cell fate. We also provide evidence that sustained misexpression of Prdm1 was selectively toxic to photoreceptors. Our data show that bipolar fate is malleable, but only for a short temporal window following fate specification. Prdm1 and Vsx2 act by stabilizing photoreceptor and bipolar fates in developing OTX2+ cells of the retina.
Collapse
|
20
|
Grünert U, Martin PR. Cell types and cell circuits in human and non-human primate retina. Prog Retin Eye Res 2020; 78:100844. [PMID: 32032773 DOI: 10.1016/j.preteyeres.2020.100844] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 12/12/2022]
Abstract
This review summarizes our current knowledge of primate including human retina focusing on bipolar, amacrine and ganglion cells and their connectivity. We have two main motivations in writing. Firstly, recent progress in non-invasive imaging methods to study retinal diseases mean that better understanding of the primate retina is becoming an important goal both for basic and for clinical sciences. Secondly, genetically modified mice are increasingly used as animal models for human retinal diseases. Thus, it is important to understand to which extent the retinas of primates and rodents are comparable. We first compare cell populations in primate and rodent retinas, with emphasis on how the fovea (despite its small size) dominates the neural landscape of primate retina. We next summarise what is known, and what is not known, about the postreceptoral neurone populations in primate retina. The inventories of bipolar and ganglion cells in primates are now nearing completion, comprising ~12 types of bipolar cell and at least 17 types of ganglion cell. Primate ganglion cells show clear differences in dendritic field size across the retina, and their morphology differs clearly from that of mouse retinal ganglion cells. Compared to bipolar and ganglion cells, amacrine cells show even higher morphological diversity: they could comprise over 40 types. Many amacrine types appear conserved between primates and mice, but functions of only a few types are understood in any primate or non-primate retina. Amacrine cells appear as the final frontier for retinal research in monkeys and mice alike.
Collapse
Affiliation(s)
- Ulrike Grünert
- The University of Sydney, Save Sight Institute, Faculty of Medicine and Health, Sydney, NSW, 2000, Australia; Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, NSW, 2000, Australia.
| | - Paul R Martin
- The University of Sydney, Save Sight Institute, Faculty of Medicine and Health, Sydney, NSW, 2000, Australia; Australian Research Council Centre of Excellence for Integrative Brain Function, Sydney Node, The University of Sydney, Sydney, NSW, 2000, Australia
| |
Collapse
|
21
|
Stefanov A, Novelli E, Strettoi E. Inner retinal preservation in the photoinducible I307N rhodopsin mutant mouse, a model of autosomal dominant retinitis pigmentosa. J Comp Neurol 2019; 528:1502-1522. [PMID: 31811649 PMCID: PMC7187456 DOI: 10.1002/cne.24838] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 12/20/2022]
Abstract
Rod‐cone degenerations, for example, retinitis pigmentosa are leading causes of blindness worldwide. Despite slow disease progression in humans, vision loss is inevitable; therefore, development of vision restoration strategies is crucial. Among others, promising approaches include optogenetics and prosthetic implants, which aim to bypass lost photoreceptors (PRs). Naturally, the efficacy of these therapeutic strategies will depend on inner retinal structural and functional preservation. The present study shows that in photoinducible I307N rhodopsin mice (Translational Vision Research Model 4 [Tvrm4]), a 12k lux light exposure eliminates PRs in the central retina in 1 week, but interneurons and their synapses are maintained for as long as 9 weeks postinduction. Despite bipolar cell dendritic retraction and moderate loss of horizontal cells, the survival rate of various cell types is very high. Significant preservation of conventional synapses and gap junctions in the inner plexiform layer is also observed. We found the number of synaptic ribbons to gradually decline and their ultrastructure to become transiently abnormal, although based on our findings intrinsic retinal architecture is maintained despite complete loss of PRs. Unlike common rodent models of PR degeneration, where the disease phenotype often interferes with retinal development, in Tvrm4 mice, the degenerative process can be induced after retinal development is complete. This time course more closely mimics the timing of disease onset in affected patients. Stability of the inner retina found in these mutants 2 months after PR degeneration suggests moderate, stereotyped remodeling in the early stages of the human disease and represents a promising finding for prompt approaches of vision restoration.
Collapse
Affiliation(s)
- Antonia Stefanov
- Institute of Neuroscience, Italian National Research Council - CNR, Pisa, Italy.,Regional Doctoral School of Neuroscience, University of Florence, Florence, Italy
| | - Elena Novelli
- Institute of Neuroscience, Italian National Research Council - CNR, Pisa, Italy
| | - Enrica Strettoi
- Institute of Neuroscience, Italian National Research Council - CNR, Pisa, Italy
| |
Collapse
|
22
|
Falasconi A, Biagioni M, Novelli E, Piano I, Gargini C, Strettoi E. Retinal Phenotype in the rd9 Mutant Mouse, a Model of X-Linked RP. Front Neurosci 2019; 13:991. [PMID: 31607844 PMCID: PMC6761883 DOI: 10.3389/fnins.2019.00991] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/03/2019] [Indexed: 12/25/2022] Open
Abstract
Retinal degeneration 9 (rd9) mice carry a mutation in the retina specific “Retinitis Pigmentosa GTPase Regulator (RPGR)” Open Reading Frame (ORF) 15 gene, located on the X chromosome and represent a rare model of X-linked Retinitis Pigmentosa (XLRP), a common and severe form of retinal degeneration (Wright et al., 2010; Tsang and Sharma, 2018). The rd9 RPGR-ORF15 mutation in mice causes lack of the protein in photoreceptors and a slow degeneration of these cells with consequent decrease in Outer Nuclear Layer (ONL) thickness and amplitude of ERG responses, as previously described (Thompson et al., 2012). However, relative rates of rod and cone photoreceptor loss, as well as secondary alterations occurring in neuronal and non-neuronal retinal cell types of rd9 mutants remain to be assessed. Aim of this study is to extend phenotype analysis of the rd9 mouse retina focusing on changes occurring in cells directly interacting with photoreceptors. To this purpose, first we estimated rod and cone survival and its degree of intraretinal variation over time; then, we studied the morphology of horizontal and bipolar cells and of the retinal pigment epithelium (RPE), extending our observations to glial cell reactivity. We found that in rd9 retinas rod (but not cone) death is the main cause of decrease in ONL thickness and that degeneration shows a high degree of intraretinal variation. Rod loss drives remodeling in the outer retina, with sprouting of second-order neurons of the rod-pathway and relative sparing of cone pathway elements. Remarkably, despite cone survival, functional defects can be clearly detected in ERG recordings in both scotopic and photopic conditions. Moderate levels of Muller cells and microglial reactivity are sided by striking attenuation of staining for RPE tight junctions, suggesting altered integrity of the outer Blood Retina Barrier (BRB). Because of many features resembling slowly progressing photoreceptor degeneration paradigms or early stages of more aggressive forms of RP, the rd9 mouse model can be considered a rare and useful tool to investigate retinal changes associated to a process of photoreceptor death sustained throughout life and to reveal disease biomarkers (e.g., BRB alterations) of human XLRP.
Collapse
Affiliation(s)
- Antonio Falasconi
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy.,Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Martina Biagioni
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Elena Novelli
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Enrica Strettoi
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| |
Collapse
|
23
|
Tetenborg S, Yadav SC, Brüggen B, Zoidl GR, Hormuzdi SG, Monyer H, van Woerden GM, Janssen-Bienhold U, Dedek K. Localization of Retinal Ca 2+/Calmodulin-Dependent Kinase II-β (CaMKII-β) at Bipolar Cell Gap Junctions and Cross-Reactivity of a Monoclonal Anti-CaMKII-β Antibody With Connexin36. Front Mol Neurosci 2019; 12:206. [PMID: 31555090 PMCID: PMC6724749 DOI: 10.3389/fnmol.2019.00206] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/07/2019] [Indexed: 11/13/2022] Open
Abstract
Neuronal gap junctions formed by connexin36 (Cx36) and chemical synapses share striking similarities in terms of plasticity. Ca2+/calmodulin-dependent protein kinase II (CaMKII), an enzyme known to induce memory formation at chemical synapses, has recently been described to potentiate electrical coupling in the retina and several other brain areas via phosphorylation of Cx36. The contribution of individual CaMKII isoforms to this process, however, remains unknown. We recently identified CaMKII-β at electrical synapses in the mouse retina. Now, we set out to identify cell types containing Cx36 gap junctions that also express CaMKII-β. To ensure precise description, we first tested the specificity of two commercially available antibodies on CaMKII-β-deficient retinas. We found that a polyclonal antibody was highly specific for CaMKII-β. However, a monoclonal antibody (CB-β-1) recognized CaMKII-β but also cross-reacted with the C-terminal tail of Cx36, making localization analyses with this antibody inaccurate. Using the polyclonal antibody, we identified strong CaMKII-β expression in bipolar cell terminals that were secretagogin- and HCN1-positive and thus represent terminals of type 5 bipolar cells. In these terminals, a small fraction of CaMKII-β also colocalized with Cx36. A similar pattern was observed in putative type 6 bipolar cells although there, CaMKII expression seemed less pronounced. Next, we tested whether CaMKII-β influenced the Cx36 expression in bipolar cell terminals by quantifying the number and size of Cx36-immunoreactive puncta in CaMKII-β-deficient retinas. However, we found no significant differences between the genotypes, indicating that CaMKII-β is not necessary for the formation and maintenance of Cx36-containing gap junctions in the retina. In addition, in wild-type retinas, we observed frequent association of Cx36 and CaMKII-β with synaptic ribbons, i.e., chemical synapses, in bipolar cell terminals. This arrangement resembled the composition of mixed synapses found for example in Mauthner cells, in which electrical coupling is regulated by glutamatergic activity. Taken together, our data imply that CaMKII-β may fulfill several functions in bipolar cell terminals, regulating both Cx36-containing gap junctions and ribbon synapses and potentially also mediating cross-talk between these two types of bipolar cell outputs.
Collapse
Affiliation(s)
- Stephan Tetenborg
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Shubhash Chandra Yadav
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Bianca Brüggen
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Georg R Zoidl
- Department of Biology & Center for Vision Research, York University, Toronto, ON, Canada
| | - Sheriar G Hormuzdi
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | | | - Geeske M van Woerden
- Department of Neuroscience, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Ulrike Janssen-Bienhold
- Department of Neuroscience, Visual Neuroscience, University of Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| | - Karin Dedek
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
24
|
Grassmeyer JJ, Cahill AL, Hays CL, Barta C, Quadros RM, Gurumurthy CB, Thoreson WB. Ca 2+ sensor synaptotagmin-1 mediates exocytosis in mammalian photoreceptors. eLife 2019; 8:e45946. [PMID: 31172949 PMCID: PMC6588344 DOI: 10.7554/elife.45946] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 06/06/2019] [Indexed: 11/24/2022] Open
Abstract
To encode light-dependent changes in membrane potential, rod and cone photoreceptors utilize synaptic ribbons to sustain continuous exocytosis while making rapid, fine adjustments to release rate. Release kinetics are shaped by vesicle delivery down ribbons and by properties of exocytotic Ca2+ sensors. We tested the role for synaptotagmin-1 (Syt1) in photoreceptor exocytosis by using novel mouse lines in which Syt1 was conditionally removed from rods or cones. Photoreceptors lacking Syt1 exhibited marked reductions in exocytosis as measured by electroretinography and single-cell recordings. Syt1 mediated all evoked release in cones, whereas rods appeared capable of some slow Syt1-independent release. Spontaneous release frequency was unchanged in cones but increased in rods lacking Syt1. Loss of Syt1 did not alter synaptic anatomy or reduce Ca2+ currents. These results suggest that Syt1 mediates both phasic and tonic release at photoreceptor synapses, revealing unexpected flexibility in the ability of Syt1 to regulate Ca2+-dependent synaptic transmission.
Collapse
Affiliation(s)
- Justin J Grassmeyer
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaUnited States
| | - Asia L Cahill
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
| | - Cassandra L Hays
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical CenterOmahaUnited States
| | - Cody Barta
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
| | - Rolen M Quadros
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research OfficeUniversity of Nebraska Medical CenterOmahaUnited States
| | - Channabasavaiah B Gurumurthy
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research OfficeUniversity of Nebraska Medical CenterOmahaUnited States
- Developmental Neuroscience, Munroe Meyer Institute for Genetics and RehabilitationUniversity of Nebraska Medical CenterOmahaUnited States
| | - Wallace B Thoreson
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaUnited States
| |
Collapse
|
25
|
Kovács-Öller T, Szarka G, Ganczer A, Tengölics Á, Balogh B, Völgyi B. Expression of Ca 2+-Binding Buffer Proteins in the Human and Mouse Retinal Neurons. Int J Mol Sci 2019; 20:E2229. [PMID: 31067641 PMCID: PMC6539911 DOI: 10.3390/ijms20092229] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/31/2022] Open
Abstract
Ca2+-binding buffer proteins (CaBPs) are widely expressed by various neurons throughout the central nervous system (CNS), including the retina. While the expression of CaBPs by photoreceptors, retinal interneurons and the output ganglion cells in the mammalian retina has been extensively studied, a general description is still missing due to the differences between species, developmental expression patterns and study-to-study discrepancies. Furthermore, CaBPs are occasionally located in a compartment-specific manner and two or more CaBPs can be expressed by the same neuron, thereby sharing the labor of Ca2+ buffering in the intracellular milieu. This article reviews this topic by providing a framework on CaBP functional expression by neurons of the mammalian retina with an emphasis on human and mouse retinas and the three most abundant and extensively studied buffer proteins: parvalbumin, calretinin and calbindin.
Collapse
Affiliation(s)
- Tamás Kovács-Öller
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
- Medical School, University of Pécs, 7624 Pécs, Hungary.
| | - Gergely Szarka
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Alma Ganczer
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Ádám Tengölics
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Boglárka Balogh
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Béla Völgyi
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
- Medical School, University of Pécs, 7624 Pécs, Hungary.
| |
Collapse
|
26
|
Yadav SC, Tetenborg S, Dedek K. Gap Junctions in A8 Amacrine Cells Are Made of Connexin36 but Are Differently Regulated Than Gap Junctions in AII Amacrine Cells. Front Mol Neurosci 2019; 12:99. [PMID: 31065239 PMCID: PMC6489437 DOI: 10.3389/fnmol.2019.00099] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/03/2019] [Indexed: 01/01/2023] Open
Abstract
In the mammalian retina, amacrine cells represent the most diverse cell class and are involved in the spatio-temporal processing of visual signals in the inner plexiform layer. They are connected to bipolar, other amacrine and ganglion cells, forming complex networks via electrical and chemical synapses. The small-field A8 amacrine cell was shown to receive non-selective glutamatergic input from OFF and ON cone bipolar cells at its bistratified dendrites in sublamina 1 and 4 of the inner plexiform layer. Interestingly, it was also shown to form electrical synapses with ON cone bipolar cells, thus resembling the rod pathway-specific AII amacrine cell. In contrast to the AII cell, however, the electrical synapses of A8 cells are poorly understood. Therefore, we made use of the Ier5-GFP mouse line, in which A8 cells are labeled by GFP, to study the gap junction composition and frequency in A8 cells. We found that A8 cells form <20 gap junctions per cell and these gap junctions consist of connexin36. Connexin36 is present at both OFF and ON dendrites of A8 cells, preferentially connecting A8 cells to type 1 OFF and type 6 and 7 ON bipolar cells and presumably other amacrine cells. Additionally, we show that the OFF dendrites of A8 cells co-stratify with the processes of dopaminergic amacrine cells from which they may receive GABAergic input via GABAA receptor subunit α3. As we found A8 cells to express dopamine receptor D1 (but not D2), we also tested whether A8 cell coupling is modulated by D1 receptor agonists and antagonists as was shown for the coupling of AII cells. However, this was not the case. In summary, our data suggests that A8 coupling is differently regulated than AII cells and may even be independent of ambient light levels and serve signal facilitation rather than providing a separate neuronal pathway.
Collapse
Affiliation(s)
- Shubhash C Yadav
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Stephan Tetenborg
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Karin Dedek
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
27
|
Maj M, Wagner L, Tretter V. 20 Years of Secretagogin: Exocytosis and Beyond. Front Mol Neurosci 2019; 12:29. [PMID: 30853888 PMCID: PMC6396707 DOI: 10.3389/fnmol.2019.00029] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 01/23/2019] [Indexed: 01/04/2023] Open
Abstract
Calcium is one of the most important signaling factors in mammalian cells. Specific temporal and spatial calcium signals underlie fundamental processes such as cell growth, development, circadian rhythms, neurotransmission, hormonal actions and apoptosis. In order to translate calcium signals into cellular processes a vast number of proteins bind this ion with affinities from the nanomolar to millimolar range. Using classical biochemical methods an impressing number of calcium binding proteins (CBPs) have been discovered since the late 1960s, some of which are expressed ubiquitously, others are more restricted to specific cell types. In the nervous system expression patterns of different CBPs have been used to discern different neuronal cell populations, especially before advanced methods like single-cell transcriptomics and activity recording were available to define neuronal identity. However, understanding CBPs and their interacting proteins is still of central interest. The post-genomic era has coined the term “calciomics,” to describe a whole new research field, that engages in the identification and characterization of CBPs and their interactome. Secretagogin is a CBP, that was discovered 20 years ago in the pancreas. Consecutively it was found also in other organs including the nervous system, with characteristic expression patterns mostly forming cell clusters. Its regional expression and subcellular location together with the identification of protein interaction partners implicated, that secretagogin has a central role in hormone secretion. Meanwhile, with the help of modern proteomics a large number of actual and putative interacting proteins has been identified, that allow to anticipate a much more complex role of secretagogin in developing and adult neuronal cells. Here, we review recent findings that appear like puzzle stones of a greater picture.
Collapse
Affiliation(s)
- Magdalena Maj
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA, United States
| | - Ludwig Wagner
- Department of Internal Medicine III, Division of Nephrology and Dialysis, Medizinische Universität Wien, Vienna, Austria
| | - Verena Tretter
- Department of Anesthesia and General Intensive Care, Clinical Department of Anesthesia, Medizinische Universität Wien, Vienna, Austria
| |
Collapse
|
28
|
Kerov V, Laird JG, Joiner ML, Knecht S, Soh D, Hagen J, Gardner SH, Gutierrez W, Yoshimatsu T, Bhattarai S, Puthussery T, Artemyev NO, Drack AV, Wong RO, Baker SA, Lee A. α 2δ-4 Is Required for the Molecular and Structural Organization of Rod and Cone Photoreceptor Synapses. J Neurosci 2018; 38:6145-6160. [PMID: 29875267 PMCID: PMC6031576 DOI: 10.1523/jneurosci.3818-16.2018] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 05/10/2018] [Accepted: 05/31/2018] [Indexed: 11/21/2022] Open
Abstract
α2δ-4 is an auxiliary subunit of voltage-gated Cav1.4 L-type channels that regulate the development and mature exocytotic function of the photoreceptor ribbon synapse. In humans, mutations in the CACNA2D4 gene encoding α2δ-4 cause heterogeneous forms of vision impairment in humans, the underlying pathogenic mechanisms of which remain unclear. To investigate the retinal function of α2δ-4, we used genome editing to generate an α2δ-4 knock-out (α2δ-4 KO) mouse. In male and female α2δ-4 KO mice, rod spherules lack ribbons and other synaptic hallmarks early in development. Although the molecular organization of cone synapses is less affected than rod synapses, horizontal and cone bipolar processes extend abnormally in the outer nuclear layer in α2δ-4 KO retina. In reconstructions of α2δ-4 KO cone pedicles by serial block face scanning electron microscopy, ribbons appear normal, except that less than one-third show the expected triadic organization of processes at ribbon sites. The severity of the synaptic defects in α2δ-4 KO mice correlates with a progressive loss of Cav1.4 channels, first in terminals of rods and later cones. Despite the absence of b-waves in electroretinograms, visually guided behavior is evident in α2δ-4 KO mice and better under photopic than scotopic conditions. We conclude that α2δ-4 plays an essential role in maintaining the structural and functional integrity of rod and cone synapses, the disruption of which may contribute to visual impairment in humans with CACNA2D4 mutations.SIGNIFICANCE STATEMENT In the retina, visual information is first communicated by the synapse formed between photoreceptors and second-order neurons. The mechanisms that regulate the structural integrity of this synapse are poorly understood. Here we demonstrate a role for α2δ-4, a subunit of voltage-gated Ca2+ channels, in organizing the structure and function of photoreceptor synapses. We find that presynaptic Ca2+ channels are progressively lost and that rod and cone synapses are disrupted in mice that lack α2δ-4. Our results suggest that alterations in presynaptic Ca2+ signaling and photoreceptor synapse structure may contribute to vision impairment in humans with mutations in the CACNA2D4 gene encoding α2δ-4.
Collapse
Affiliation(s)
- Vasily Kerov
- Department of Molecular Physiology and Biophysics
| | | | | | - Sharmon Knecht
- Department of Biological Structure, University of Washington, Seattle, Washington 98195, and
| | - Daniel Soh
- Department of Molecular Physiology and Biophysics
| | | | | | | | - Takeshi Yoshimatsu
- Department of Biological Structure, University of Washington, Seattle, Washington 98195, and
| | - Sajag Bhattarai
- Department of Ophthalmology and Institute for Vision Research
| | - Teresa Puthussery
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97239
| | | | - Arlene V Drack
- Department of Ophthalmology and Institute for Vision Research
| | - Rachel O Wong
- Department of Biological Structure, University of Washington, Seattle, Washington 98195, and
| | - Sheila A Baker
- Department of Biochemistry,
- Department of Ophthalmology and Institute for Vision Research
| | - Amy Lee
- Department of Molecular Physiology and Biophysics,
- Department of Otolaryngology-Head and Neck Surgery
- Department of Neurology, University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
29
|
Sarin S, Zuniga-Sanchez E, Kurmangaliyev YZ, Cousins H, Patel M, Hernandez J, Zhang KX, Samuel MA, Morey M, Sanes JR, Zipursky SL. Role for Wnt Signaling in Retinal Neuropil Development: Analysis via RNA-Seq and In Vivo Somatic CRISPR Mutagenesis. Neuron 2018; 98:109-126.e8. [PMID: 29576390 DOI: 10.1016/j.neuron.2018.03.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 01/16/2018] [Accepted: 03/02/2018] [Indexed: 12/22/2022]
Abstract
Screens for genes that orchestrate neural circuit formation in mammals have been hindered by practical constraints of germline mutagenesis. To overcome these limitations, we combined RNA-seq with somatic CRISPR mutagenesis to study synapse development in the mouse retina. Here synapses occur between cellular layers, forming two multilayered neuropils. The outer neuropil, the outer plexiform layer (OPL), contains synapses made by rod and cone photoreceptor axons on rod and cone bipolar dendrites, respectively. We used RNA-seq to identify selectively expressed genes encoding cell surface and secreted proteins and CRISPR-Cas9 electroporation with cell-specific promoters to assess their roles in OPL development. Among the genes identified in this way are Wnt5a and Wnt5b. They are produced by rod bipolars and activate a non-canonical signaling pathway in rods to regulate early OPL patterning. The approach we use here can be applied to other parts of the brain.
Collapse
Affiliation(s)
- Sumeet Sarin
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Elizabeth Zuniga-Sanchez
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yerbol Z Kurmangaliyev
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Henry Cousins
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Mili Patel
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Jeanette Hernandez
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kelvin X Zhang
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Melanie A Samuel
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Marta Morey
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA.
| | - S Lawrence Zipursky
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
30
|
Kosaka T, Kosaka K. Calcium-binding protein, secretagogin, specifies the microcellular tegmental nucleus and intermediate and ventral parts of the cuneiform nucleus of the mouse and rat. Neurosci Res 2018; 134:30-38. [PMID: 29366872 DOI: 10.1016/j.neures.2018.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 11/30/2022]
Abstract
Secretagogin (SCGN) is a recently discovered calcium binding protein of the EF hand family, cloned from β cells of pancreatic island of Langerhans and endocrine cells of the gastrointestinal gland. SCGN characterizes some particular neuron groups in various regions of the nervous system and is considered as one of the useful neuron subpopulation markers. In the present study we reported that SCGN specifically labelled a particular neuronal cluster in the brainstem of the mice and rats. The comparison of the SCGN immunostaining with the choline acetyltransferase immunostaining and acetylcholinesterase staining clearly indicated that the particular cluster of SCGN positive neurons corresponded to the microcellular tegmental nucleus (MiTg) and the ventral portion of the cuneiform nucleus (CnF), both of which are components of the isthmus. The analyses in mice indicated that SCGN positive neurons in the MiTg and CnF were homogeneous in size and shape, appearing to compose a single complex: their somata were small comparing with the adjacent cholinergic neurons in the pedunculotegmantal nucleus, 10.5 vs 16.0 μm in diameter, and extended 2-3 slender smooth processes. SCGN might be one of significant markers to reconsider the delineations of the structures of the mouse, and presumably rat, brainstem.
Collapse
Affiliation(s)
- Toshio Kosaka
- Department of Medical Science Technology, Faculty of Health and Welfare Sciences in Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa City, Fukuoka 831-8501, Japan.
| | - Katsuko Kosaka
- Department of Medical Science Technology, Faculty of Health and Welfare Sciences in Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa City, Fukuoka 831-8501, Japan
| |
Collapse
|
31
|
Llonch S, Carido M, Ader M. Organoid technology for retinal repair. Dev Biol 2017; 433:132-143. [PMID: 29291970 DOI: 10.1016/j.ydbio.2017.09.028] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/05/2017] [Accepted: 09/21/2017] [Indexed: 02/07/2023]
Abstract
A major cause for vision impairment and blindness in industrialized countries is the loss of the light-sensing retinal tissue in the eye. Photoreceptor damage is one of the main characteristics found in retinal degeneration diseases, such as Retinitis Pigmentosa or age-related macular degeneration. The lack of effective therapies to stop photoreceptor loss together with the absence of significant intrinsic regeneration in the human retina converts such degenerative diseases into permanent conditions that are currently irreversible. Cell replacement by means of photoreceptor transplantation has been proposed as a potential approach to tackle cell loss in the retina. Since the first attempt of photoreceptor transplantation in humans, about twenty years ago, several research groups have focused in the development and improvement of technologies necessary to bring cell transplantation for retinal degeneration diseases to reality. Progress in recent years in the generation of human tissue derived from pluripotent stem cells (PSCs) has significantly improved our tools to study human development and disease in the dish. Particularly the availability of 3D culture systems for the generation of PSC-derived organoids, including the human retina, has dramatically increased access to human material for basic and medical research. In this review, we focus on important milestones towards the generation of transplantable photoreceptor precursors from PSC-derived retinal organoids and discuss recent pre-clinical transplantation studies using organoid-derived photoreceptors in context to related in vivo work using primary photoreceptors as donor material. Additionally, we summarize remaining challenges for developing photoreceptor transplantation towards clinical application.
Collapse
Affiliation(s)
- Sílvia Llonch
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Madalena Carido
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany; German Center for Neurodegenerative Diseases Dresden (DZNE), Arnoldstraße 18, 01307 Dresden, Germany
| | - Marius Ader
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany.
| |
Collapse
|
32
|
Dudczig S, Currie PD, Jusuf PR. Developmental and adult characterization of secretagogin expressing amacrine cells in zebrafish retina. PLoS One 2017; 12:e0185107. [PMID: 28949993 PMCID: PMC5614429 DOI: 10.1371/journal.pone.0185107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 09/06/2017] [Indexed: 12/19/2022] Open
Abstract
Calcium binding proteins show stereotypical expression patterns within diverse neuron types across the central nervous system. Here, we provide a characterization of developmental and adult secretagogin-immunolabelled neurons in the zebrafish retina with an emphasis on co-expression of multiple calcium binding proteins. Secretagogin is a recently identified and cloned member of the F-hand family of calcium binding proteins, which labels distinct neuron populations in the retinas of mammalian vertebrates. Both the adult distribution of secretagogin labeled retinal neurons as well as the developmental expression indicative of the stage of neurogenesis during which this calcium binding protein is expressed was quantified. Secretagogin expression was confined to an amacrine interneuron population in the inner nuclear layer, with monostratified neurites in the center of the inner plexiform layer and a relatively regular soma distribution (regularity index > 2.5 across central–peripheral areas). However, only a subpopulation (~60%) co-labeled with gamma-aminobutyric acid as their neurotransmitter, suggesting that possibly two amacrine subtypes are secretagogin immunoreactive. Quantitative co-labeling analysis with other known amacrine subtype markers including the three main calcium binding proteins parvalbumin, calbindin and calretinin identifies secretagogin immunoreactive neurons as a distinct neuron population. The highest density of secretagogin cells of ~1800 cells / mm2 remained relatively evenly along the horizontal meridian, whilst the density dropped of to 125 cells / mm2 towards the dorsal and ventral periphery. Thus, secretagogin represents a new amacrine label within the zebrafish retina. The developmental expression suggests a possible role in late stage differentiation. This characterization forms the basis of functional studies assessing how the expression of distinct calcium binding proteins might be regulated to compensate for the loss of one of the others.
Collapse
Affiliation(s)
- Stefanie Dudczig
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
- School of Biosciences, University of Melbourne, Parkville, VIC, Australia
| | - Peter David Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Patricia Regina Jusuf
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
- School of Biosciences, University of Melbourne, Parkville, VIC, Australia
- * E-mail:
| |
Collapse
|
33
|
Kántor O, Varga A, Nitschke R, Naumann A, Énzsöly A, Lukáts Á, Szabó A, Németh J, Völgyi B. Bipolar cell gap junctions serve major signaling pathways in the human retina. Brain Struct Funct 2017; 222:2603-2624. [PMID: 28070649 DOI: 10.1007/s00429-016-1360-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/22/2016] [Indexed: 11/26/2022]
Abstract
Connexin36 (Cx36) constituent gap junctions (GJ) throughout the brain connect neurons into functional syncytia. In the retina they underlie the transmission, averaging and correlation of signals prior conveying visual information to the brain. This is the first study that describes retinal bipolar cell (BC) GJs in the human inner retina, whose function is enigmatic even in the examined animal models. Furthermore, a number of unique features (e.g. fovea, trichromacy, midget system) necessitate a reexamination of the animal model results in the human retina. Well-preserved postmortem human samples of this study are allowed to identify Cx36 expressing BCs neurochemically. Results reveal that both rod and cone pathway interneurons display strong Cx36 expression. Rod BC inputs to AII amacrine cells (AC) appear in juxtaposition to AII GJs, thus suggesting a strategic AII cell targeting by rod BCs. Cone BCs serving midget, parasol or koniocellular signaling pathways display a wealth of Cx36 expression to form homologously coupled arrays. In addition, they also establish heterologous GJ contacts to serve an exchange of information between parallel signaling streams. Interestingly, a prominent Cx36 expression was exhibited by midget system BCs that appear to maintain intimate contacts with bistratified BCs serving other pathways. These findings suggest that BC GJs in parallel signaling streams serve both an intra- and inter-pathway exchange of signals in the human retina.
Collapse
Affiliation(s)
- Orsolya Kántor
- Department of Neuroanatomy, Faculty of Medicine, Institute for Anatomy and Cell Biology, University of Freiburg, 79104, Freiburg, Germany
- MTA-PTE NAP B Retinal Electrical Synapses Research Group, Pécs, 7624, Hungary
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Alexandra Varga
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Roland Nitschke
- Life Imaging Center, Center for Biological Systems Analysis, University of Freiburg, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Angela Naumann
- Life Imaging Center, Center for Biological Systems Analysis, University of Freiburg, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Anna Énzsöly
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
- Department of Ophthalmology, Semmelweis University, Budapest, 1085, Hungary
| | - Ákos Lukáts
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Arnold Szabó
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - János Németh
- Department of Ophthalmology, Semmelweis University, Budapest, 1085, Hungary
| | - Béla Völgyi
- MTA-PTE NAP B Retinal Electrical Synapses Research Group, Pécs, 7624, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, 7624, Hungary.
- János Szentágothai Research Center, University of Pécs, Ifjúság street 20, Pécs, 7624, Hungary.
- Department of Ophthalmology, New York University Langone Medical Center, New York, NY, 10016, USA.
| |
Collapse
|
34
|
Park KU, Randazzo G, Jones KL, Brzezinski JA. Gsg1, Trnp1, and Tmem215 Mark Subpopulations of Bipolar Interneurons in the Mouse Retina. Invest Ophthalmol Vis Sci 2017; 58:1137-1150. [PMID: 28199486 PMCID: PMC5317276 DOI: 10.1167/iovs.16-19767] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose How retinal bipolar cell interneurons are specified and assigned to specialized subtypes is only partially understood. In part, this is due to a lack of early pan- and subtype-specific bipolar cell markers. To discover these factors, we identified genes that were upregulated in Blimp1 (Prdm1) mutant retinas, which exhibit precocious bipolar cell development. Methods Postnatal day (P)2 retinas from Blimp1 conditional knock-out (CKO) mice and controls were processed for RNA sequencing. Genes that increased at least 45% and were statistically different between conditions were considered candidate bipolar-specific factors. Candidates were further evaluated by RT-PCR, in situ hybridization, and immunohistochemistry. Knock-in Tmem215-LacZ mice were used to better trace retinal expression. Results A comparison between Blimp1 CKO and control RNA-seq datasets revealed approximately 40 significantly upregulated genes. We characterized the expression of three genes that have no known function in the retina, Gsg1 (germ cell associated gene), Trnp1 (TMF-regulated nuclear protein), and Tmem215 (a predicted transmembrane protein). Germ cell associated gene appeared restricted to a small subset of cone bipolars while Trnp1 was seen in all ON type bipolar cells. Using Tmem215-LacZ heterozygous knock-in mice, we observed that β-galactosidase expression started early in bipolar cell development. In adults, Tmem215 was expressed by a subset of ON and OFF cone bipolar cells. Conclusions We have identified Gsg1, Tmem215, and Trnp1 as novel bipolar subtype-specific genes. The spatial and temporal pattern of their expression is consistent with a role in controlling bipolar subtype fate choice, differentiation, or physiology.
Collapse
Affiliation(s)
- Ko Uoon Park
- Department of Ophthalmology, University of Colorado Denver, Aurora, Colorado, United States
| | - Grace Randazzo
- Department of Ophthalmology, University of Colorado Denver, Aurora, Colorado, United States
| | - Kenneth L Jones
- Department of Pediatrics, Section Hematology/Oncology, University of Colorado Denver, Aurora, Colorado, United States
| | - Joseph A Brzezinski
- Department of Ophthalmology, University of Colorado Denver, Aurora, Colorado, United States
| |
Collapse
|
35
|
Deafferented Adult Rod Bipolar Cells Create New Synapses with Photoreceptors to Restore Vision. J Neurosci 2017; 37:4635-4644. [PMID: 28373392 DOI: 10.1523/jneurosci.2570-16.2017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 11/21/2022] Open
Abstract
Upon degeneration of photoreceptors in the adult retina, interneurons, including bipolar cells, exhibit a plastic response leading to their aberrant rewiring. Photoreceptor reintroduction has been suggested as a potential approach to sight restoration, but the ability of deafferented bipolar cells to establish functional synapses with photoreceptors is poorly understood. Here we use photocoagulation to selectively destroy photoreceptors in adult rabbits while preserving the inner retina. We find that rods and cones shift into the ablation zone over several weeks, reducing the blind spot at scotopic and photopic luminances. During recovery, rod and cone bipolar cells exhibit markedly different responses to deafferentation. Rod bipolar cells extend their dendrites to form new synapses with healthy photoreceptors outside the lesion, thereby restoring visual function in the deafferented retina. Secretagogin-positive cone bipolar cells did not exhibit such obvious dendritic restructuring. These findings are encouraging to the idea of photoreceptor reintroduction for vision restoration in patients blinded by retinal degeneration. At the same time, they draw attention to the postsynaptic side of photoreceptor reintroduction; various bipolar cell types, representing different visual pathways, vary in their response to the photoreceptor loss and in their consequent dendritic restructuring.SIGNIFICANCE STATEMENT Loss of photoreceptors during retinal degeneration results in permanent visual impairment. Strategies for vision restoration based on the reintroduction of photoreceptors inherently rely on the ability of the remaining retinal neurons to correctly synapse with new photoreceptors. We show that deafferented bipolar cells in the adult mammalian retina can reconnect to rods and cones and restore retinal sensitivity at scotopic and photopic luminances. Rod bipolar cells extend their dendrites to form new synapses with healthy rod photoreceptors. These findings support the idea that bipolar cells might be able to synapse with reintroduced photoreceptors, thereby restoring vision in patients blinded by retinal degeneration.
Collapse
|
36
|
Goodings L, He J, Wood AJ, Harris WA, Currie PD, Jusuf PR. In vivo expression of Nurr1/Nr4a2a in developing retinal amacrine subtypes in zebrafish Tg(nr4a2a:eGFP) transgenics. J Comp Neurol 2017; 525:1962-1979. [PMID: 28177524 DOI: 10.1002/cne.24185] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 01/29/2017] [Accepted: 01/30/2017] [Indexed: 12/12/2022]
Abstract
The Nuclear receptor subfamily 4 group A member 2 (Nr4a2) is crucial for the formation or maintenance of dopaminergic neurons in the central nervous system including the retina, where dopaminergic amacrine cells contribute to visual function. Little is known about which cells express Nr4a2 at which developmental stage. Furthermore, whether Nr4a2 functions in combination with other genes is poorly understood. Thus, we generated a novel transgenic to visualize Nr4a2 expression in vivo during zebrafish retinogenesis. A 4.1 kb fragment of the nr4a2a promoter was used to drive green fluorescent protein expression in this Tg(nr4a2a:eGFP) line. In situ hybridization showed that transgene expression follows endogenous RNA expression at a cellular level. Temporal expression and lineages were quantified using in vivo time-lapse imaging in embryos. Nr4a2 expressing retinal subtypes were characterized immunohistochemically. Nr4a2a:eGFP labeled multiple neuron subtypes including 24.5% of all amacrine interneurons. Nr4a2a:eGFP labels all tyrosine hydroxylase labeled dopaminergic amacrine cells, and other nondopaminergic GABAergic amacrine populations. Nr4a2a:eGFP is confined to a specific progenitor lineage identified by sequential expression of the bhlh transcription factor Atonal7 (Atoh7) and Pancreas transcription factor 1a (Ptf1a), and labels postmitotic postmigratory amacrine cells. Thus, developmental Nr4a2a expression indicates a role during late differentiation of specific amacrine interneurons. Tg(nr4a2a:eGFP) is an early marker of distinct neurons including dopaminergic amacrine cells. It can be utilized to assess consequences of gene manipulations and understand whether Nr4a2 only carries out its role in the presence of specific coexpressed genes. This will allow Nr4a2 use to be refined for regenerative approaches.
Collapse
Affiliation(s)
- Liana Goodings
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Jie He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Alasdair J Wood
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - William A Harris
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Patricia R Jusuf
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,School of Biosciences, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
37
|
Kosaka T, Yasuda S, Kosaka K. Calcium-binding protein, secretagogin, characterizes novel groups of interneurons in the rat striatum. Neurosci Res 2017; 119:53-60. [PMID: 28193530 DOI: 10.1016/j.neures.2017.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/28/2016] [Accepted: 01/18/2017] [Indexed: 11/25/2022]
Abstract
In the rat striatum numerous secretagogin (SCGN) positive neurons were scattered. They were heterogeneous in their morphological and chemical properties. We examined the colocalization of SCGN with known four interneuron markers, parvalbumin (PV), calretinin (CR), nitric oxide synthase (NOS) and choline acetyl transferase (ChAT). 60-70% of SCGN positive striatal neurons contained either PV or CR or ChAT, but none contained NOS. On the other hand the remaining 30-40% expressed none of these markers, most of which were GAD positive. The present study indicates that there are hitherto unknown groups of striatal interneurons in the rat striatum.
Collapse
Affiliation(s)
- Toshio Kosaka
- Department of Medical Science Technology, Faculty of Health and Welfare Sciences in Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa City, Fukuoka 831-8501, Japan.
| | - Seiko Yasuda
- Department of Medical Science Technology, Faculty of Health and Welfare Sciences in Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa City, Fukuoka 831-8501, Japan
| | - Katsuko Kosaka
- Department of Medical Science Technology, Faculty of Health and Welfare Sciences in Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa City, Fukuoka 831-8501, Japan
| |
Collapse
|
38
|
Kim E, Lee I. Network-Based Gene Function Prediction in Mouse and Other Model Vertebrates Using MouseNet Server. Methods Mol Biol 2017; 1611:183-198. [PMID: 28451980 DOI: 10.1007/978-1-4939-7015-5_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The mouse, Mus musculus, is a popular model organism for the study of human genes involved in development, immunology, and disease phenotypes. Despite recent revolutions in gene-knockout technologies in mouse, identification of candidate genes for functions of interest can further accelerate the discovery of novel gene functions. The collaborative nature of genetic functions allows for the inference of gene functions based on the principle of guilt-by-association. Genome-scale co-functional networks could therefore provide functional predictions for genes via network analysis. We recently constructed such a network for mouse (MouseNet), which interconnects over 88% of protein-coding genes with 788,080 functional relationships. The companion web server ( www.inetbio.org/mousenet ) enables researchers with no bioinformatics expertise to generate predictions that facilitate discovery of novel gene functions. In this chapter, we present the theoretical framework for MouseNet, as well as step-by-step instructions and technical tips for functional prediction of genes and pathways in mouse and other model vertebrates.
Collapse
Affiliation(s)
- Eiru Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| |
Collapse
|
39
|
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: 771] [Impact Index Per Article: 85.7] [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
|
40
|
Chaney SY, Mukherjee S, Giddabasappa A, Rueda EM, Hamilton WR, Johnson JE, Fox DA. Increased proliferation of late-born retinal progenitor cells by gestational lead exposure delays rod and bipolar cell differentiation. Mol Vis 2016; 22:1468-1489. [PMID: 28050121 PMCID: PMC5204453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 12/22/2016] [Indexed: 10/26/2022] Open
Abstract
PURPOSE Studies of neuronal development in the retina often examine the stages of proliferation, differentiation, and synaptic development, albeit independently. Our goal was to determine if a known neurotoxicant insult to a population of retinal progenitor cells (RPCs) would affect their eventual differentiation and synaptic development. To that end, we used our previously published human equivalent murine model of low-level gestational lead exposure (GLE). Children and animals with GLE exhibit increased scotopic electroretinogram a- and b-waves. Adult mice with GLE exhibit an increased number of late-born RPCs, a prolonged period of RPC proliferation, and an increased number of late-born rod photoreceptors and rod and cone bipolar cells (BCs), with no change in the number of late-born Müller glial cells or early-born neurons. The specific aims of this study were to determine whether increased and prolonged RPC proliferation alters the spatiotemporal differentiation and synaptic development of rods and BCs in early postnatal GLE retinas compared to control retinas. METHODS C57BL/6N mouse pups were exposed to lead acetate via drinking water throughout gestation and until postnatal day 10, which is equivalent to the human gestation period for retinal neurogenesis. RT-qPCR, immunohistochemical analysis, and western blots of well-characterized, cell-specific genes and proteins were performed at embryonic and early postnatal ages to assess rod and cone photoreceptor differentiation, rod and BC differentiation and synaptic development, and Müller glial cell differentiation. RESULTS Real-time quantitative PCR (RT-qPCR) with the rod-specific transcription factors Nrl, Nr2e3, and Crx and the rod-specific functional gene Rho, along with central retinal confocal studies with anti-recoverin and anti-rhodopsin antibodies, revealed a two-day delay in the differentiation of rod photoreceptors in GLE retinas. Rhodopsin immunoblots supported this conclusion. No changes in glutamine synthetase gene or protein expression, a marker for late-born Müller glial cells, were observed in the developing retinas. In the retinas from the GLE mice, anti-PKCα, -Chx10 (Vsx2) and -secretagogin antibodies revealed a two- to three-day delay in the differentiation of rod and cone BCs, whereas the expression of the proneural and BC genes Otx2 and Chx10, respectively, increased. In addition, confocal studies of proteins associated with functional synapses (e.g., vesicular glutamate transporter 1 [VGluT1], plasma membrane calcium ATPase [PMCA], transient receptor potential channel M1 [TRPM1], and synaptic vesicle glycoprotein 2B [SV2B]) revealed a two-day delay in the formation of the outer and inner plexiform layers of the GLE retinas. Moreover, several markers revealed that the initiation of the differentiation and intensity of the labeling of early-born cells in the retinal ganglion cell and inner plexiform layers were not different in the control retinas. CONCLUSIONS Our combined gene, confocal, and immunoblot findings revealed that the onset of rod and BC differentiation and their subsequent synaptic development is delayed by two to three days in GLE retinas. These results suggest that perturbations during the early proliferative stages of late-born RPCs fated to be rods and BCs ultimately alter the coordinated time-dependent progression of rod and BC differentiation and synaptic development. These GLE effects were selective for late-born neurons. Although the molecular mechanisms are unknown, alterations in soluble neurotrophic factors and/or their receptors are likely to play a role. Since neurodevelopmental delays and altered synaptic connectivity are associated with neuropsychiatric and behavioral disorders as well as cognitive deficits, future work is needed to determine if similar effects occur in the brains of GLE mice and whether children with GLE experience similar delays in retinal and brain neuronal differentiation and synaptic development.
Collapse
Affiliation(s)
- Shawnta Y. Chaney
- College of Optometry, University of Houston, Houston, TX,Department of Biology and Biochemistry, University of Houston, Houston, TX
| | - Shradha Mukherjee
- College of Optometry, University of Houston, Houston, TX,Department of Biology and Biochemistry, University of Houston, Houston, TX
| | - Anand Giddabasappa
- College of Optometry, University of Houston, Houston, TX,Department of Biology and Biochemistry, University of Houston, Houston, TX
| | - Elda M. Rueda
- College of Optometry, University of Houston, Houston, TX
| | - W. Ryan Hamilton
- College of Optometry, University of Houston, Houston, TX,Department of Biology and Biochemistry, University of Houston, Houston, TX
| | - Jerry E. Johnson
- College of Optometry, University of Houston, Houston, TX,Department of Natural Sciences, University of Houston-Downtown; Houston, TX
| | - Donald A. Fox
- College of Optometry, University of Houston, Houston, TX,Department of Biology and Biochemistry, University of Houston, Houston, TX,Department of Pharmacology and Pharmaceutical Science, University of Houston, Houston, TX
| |
Collapse
|
41
|
Lagali PS, Medina CF, Zhao BYH, Yan K, Baker AN, Coupland SG, Tsilfidis C, Wallace VA, Picketts DJ. Retinal interneuron survival requires non-cell-autonomous Atrx activity. Hum Mol Genet 2016; 25:4787-4803. [PMID: 28173139 DOI: 10.1093/hmg/ddw306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 08/25/2016] [Accepted: 08/30/2016] [Indexed: 01/13/2023] Open
Abstract
ATRX is a chromatin remodeling protein that is mutated in several intellectual disability disorders including alpha-thalassemia/mental retardation, X-linked (ATR-X) syndrome. We previously reported the prevalence of ophthalmological defects in ATR-X syndrome patients, and accordingly we find morphological and functional visual abnormalities in a mouse model harboring a mutation occurring in ATR-X patients. The visual system abnormalities observed in these mice parallels the Atrx-null retinal phenotype characterized by interneuron defects and selective loss of amacrine and horizontal cells. The mechanisms that underlie selective neuronal vulnerability and neurodegeneration in the central nervous system upon Atrx mutation or deletion are unknown. To interrogate the cellular specificity of Atrx for its retinal neuroprotective functions, we employed a combination of temporal and lineage-restricted conditional ablation strategies to generate five different conditional knockout mouse models, and subsequently identified a non-cell-autonomous requirement for Atrx in bipolar cells for inhibitory interneuron survival in the retina. Atrx-deficient retinal bipolar cells exhibit functional, structural and molecular alterations consistent with impairments in neuronal activity and connectivity. Gene expression changes in the Atrx-null retina indicate defective synaptic structure and neuronal circuitry, suggest excitotoxic mechanisms of neurodegeneration, and demonstrate that common targets of ATRX in the forebrain and retina may contribute to similar neuropathological processes underlying cognitive impairment and visual dysfunction in ATR-X syndrome.
Collapse
Affiliation(s)
- Pamela S Lagali
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Chantal F Medina
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Brandon Y H Zhao
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Keqin Yan
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Adam N Baker
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Stuart G Coupland
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada.,Department of Ophthalmology, University of Ottawa, Ottawa, ON K1H 8M5, Canada,,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Catherine Tsilfidis
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada.,Department of Ophthalmology, University of Ottawa, Ottawa, ON K1H 8M5, Canada,,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Valerie A Wallace
- Vision Research Division, Krembil Research Institute, Toronto, Ontario, Canada M5T 2S8,,Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON M5T 3A9, Canada
| | - David J Picketts
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada,,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
42
|
Calcium buffer proteins are specific markers of human retinal neurons. Cell Tissue Res 2016; 365:29-50. [PMID: 26899253 DOI: 10.1007/s00441-016-2376-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 02/04/2016] [Indexed: 10/22/2022]
Abstract
Ca(2+)-buffer proteins (CaBPs) modulate the temporal and spatial characteristics of transient intracellular Ca(2+)-concentration changes in neurons in order to fine-tune the strength and duration of the output signal. CaBPs have been used as neurochemical markers to identify and trace neurons of several brain loci including the mammalian retina. The CaBP content of retinal neurons, however, varies between species and, thus, the results inferred from animal models cannot be utilised directly by clinical ophthalmologists. Moreover, the shortage of well-preserved human samples greatly impedes human retina studies at the cellular and network level. Our purpose has therefore been to examine the distribution of major CaBPs, including calretinin, calbindin-D28, parvalbumin and the recently discovered secretagogin in exceptionally well-preserved human retinal samples. Based on a combination of immunohistochemistry, Neurolucida tracing and Lucifer yellow injections, we have established a database in which the CaBP marker composition can be defined for morphologically identified cell types of the human retina. Hence, we describe the full CaBP make-up for a number of human retinal neurons, including HII horizontal cells, AII amacrine cells, type-1 tyrosine-hydroxylase-expressing amacrine cells and other lesser known neurons. We have also found a number of unidentified cells whose morphology remains to be characterised. We present several examples of the colocalisation of two or three CaBPs with slightly different subcellular distributions in the same cell strongly suggesting a compartment-specific division of labour of Ca(2+)-buffering by CaBPs. Our work thus provides a neurochemical framework for future ophthalmological studies and renders new information concerning the cellular and subcellular distribution of CaBPs for experimental neuroscience.
Collapse
|
43
|
Kim E, Hwang S, Kim H, Shim H, Kang B, Yang S, Shim JH, Shin SY, Marcotte EM, Lee I. MouseNet v2: a database of gene networks for studying the laboratory mouse and eight other model vertebrates. Nucleic Acids Res 2016; 44:D848-54. [PMID: 26527726 PMCID: PMC4702832 DOI: 10.1093/nar/gkv1155] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/05/2015] [Accepted: 10/19/2015] [Indexed: 12/26/2022] Open
Abstract
Laboratory mouse, Mus musculus, is one of the most important animal tools in biomedical research. Functional characterization of the mouse genes, hence, has been a long-standing goal in mammalian and human genetics. Although large-scale knockout phenotyping is under progress by international collaborative efforts, a large portion of mouse genome is still poorly characterized for cellular functions and associations with disease phenotypes. A genome-scale functional network of mouse genes, MouseNet, was previously developed in context of MouseFunc competition, which allowed only limited input data for network inferences. Here, we present an improved mouse co-functional network, MouseNet v2 (available at http://www.inetbio.org/mousenet), which covers 17 714 genes (>88% of coding genome) with 788 080 links, along with a companion web server for network-assisted functional hypothesis generation. The network database has been substantially improved by large expansion of genomics data. For example, MouseNet v2 database contains 183 co-expression networks inferred from 8154 public microarray samples. We demonstrated that MouseNet v2 is predictive for mammalian phenotypes as well as human diseases, which suggests its usefulness in discovery of novel disease genes and dissection of disease pathways. Furthermore, MouseNet v2 database provides functional networks for eight other vertebrate models used in various research fields.
Collapse
Affiliation(s)
- Eiru Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Sohyun Hwang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea Center for Systems and Synthetic Biology, Institute for Cellular and Molecular Biology, University of Texas at Austin, TX 78712, USA
| | - Hyojin Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Hongseok Shim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Byunghee Kang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Sunmo Yang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Jae Ho Shim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Seung Yeon Shin
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Edward M Marcotte
- Center for Systems and Synthetic Biology, Institute for Cellular and Molecular Biology, University of Texas at Austin, TX 78712, USA
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| |
Collapse
|
44
|
Gayet-Primo J, Puthussery T. Alterations in Kainate Receptor and TRPM1 Localization in Bipolar Cells after Retinal Photoreceptor Degeneration. Front Cell Neurosci 2015; 9:486. [PMID: 26733812 PMCID: PMC4686838 DOI: 10.3389/fncel.2015.00486] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 11/30/2015] [Indexed: 11/21/2022] Open
Abstract
Photoreceptor degeneration differentially impacts glutamatergic signaling in downstream On and Off bipolar cells. In rodent models, photoreceptor degeneration leads to loss of glutamatergic signaling in On bipolar cells, whereas Off bipolar cells appear to retain glutamate sensitivity, even after extensive photoreceptor loss. The localization and identity of the receptors that mediate these residual glutamate responses in Off bipolar cells have not been determined. Recent studies show that macaque and mouse Off bipolar cells receive glutamatergic input primarily through kainate-type glutamate receptors. Here, we studied the impact of photoreceptor degeneration on glutamate receptor and their associated proteins in Off and On bipolar cells. We show that the kainate receptor subunit, GluK1, persists in remodeled Off bipolar cell dendrites of the rd10 mouse retina. However, the pattern of expression is altered and the intensity of staining is reduced compared to wild-type retina. The kainate receptor auxiliary subunit, Neto1, also remains in Off bipolar cell dendrites after extensive photoreceptor degeneration. Similar preservation of kainate receptor subunits was evident in human retina in which photoreceptors had degenerated due to serous retinal detachment. In contrast, photoreceptor degeneration leads to loss of synaptic expression of TRPM1 in mouse and human On bipolar cells, but strong somatic expression remains. These findings demonstrate that Off bipolar cells retain dendritic glutamate receptors during retinal degeneration and could thus serve as a conduit for signal transmission from transplanted or optogenetically restored photoreceptors.
Collapse
Affiliation(s)
- Jacqueline Gayet-Primo
- Casey Eye Institute, Department of Ophthalmology, Oregon Health and Science University, Portland OR, USA
| | - Theresa Puthussery
- Casey Eye Institute, Department of Ophthalmology, Oregon Health and Science University, Portland OR, USA
| |
Collapse
|
45
|
Lee SCS, Meyer A, Schubert T, Hüser L, Dedek K, Haverkamp S. Morphology and connectivity of the small bistratified A8 amacrine cell in the mouse retina. J Comp Neurol 2015; 523:1529-47. [PMID: 25630271 DOI: 10.1002/cne.23752] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 01/22/2015] [Accepted: 01/24/2015] [Indexed: 01/23/2023]
Abstract
Amacrine cells comprise ∼ 30 morphological types in the mammalian retina. The synaptic connectivity and function of a few γ-aminobutyric acid (GABA)ergic wide-field amacrine cells have recently been studied; however, with the exception of the rod pathway-specific AII amacrine cell, the connectivity of glycinergic small-field amacrine cells has not been investigated in the mouse retina. Here, we studied the morphology and connectivity pattern of the small-field A8 amacrine cell. A8 cells in mouse retina are bistratified with lobular processes in the ON sublamina and arboreal dendrites in the OFF sublamina of the inner plexiform layer. The distinct bistratified morphology was first visible at postnatal day 8, reaching the adult shape at P13, around eye opening. The connectivity of A8 cells to bipolar cells and ganglion cells was studied by double and triple immunolabeling experiments by using various cell markers combined with synaptic markers. Our data suggest that A8 amacrine cells receive glutamatergic input from both OFF and ON cone bipolar cells. Furthermore, A8 cells are coupled to ON cone bipolar cells by gap junctions, and provide inhibitory input via glycine receptor (GlyR) subunit α1 to OFF cone bipolar cells and to ON A-type ganglion cells. Measurements of spontaneous glycinergic postsynaptic currents and GlyR immunolabeling revealed that A8 cells express GlyRs containing the α2 subunit. The results show that the bistratified A8 cell makes very similar synaptic contacts with cone bipolar cells as the rod pathway-specific AII amacrine cell. However, unlike AII cells, A8 amacrine cells provide glycinergic input to ON A-type ganglion cells.
Collapse
Affiliation(s)
- Sammy C S Lee
- Max Planck Institute for Brain Research, 60438, Frankfurt am Main, Germany.,University of Sydney-Save Sight Institute, Sydney, New South Wales, 2000, Australia
| | - Arndt Meyer
- Department of Neurobiology, University of Oldenburg, 26129, Oldenburg, Germany
| | - Timm Schubert
- Werner Reichardt Center for Integrative Neuroscience (CIN)/Institute for Ophthalmic Research, University of Tübingen, 72076, Tübingen, Germany
| | - Laura Hüser
- Max Planck Institute for Brain Research, 60438, Frankfurt am Main, Germany
| | - Karin Dedek
- Department of Neurobiology, University of Oldenburg, 26129, Oldenburg, Germany.,Research Center for Neurosensory Science, University of Oldenburg, 26129, Oldenburg, Germany
| | - Silke Haverkamp
- Max Planck Institute for Brain Research, 60438, Frankfurt am Main, Germany
| |
Collapse
|
46
|
Weltzien F, Percival KA, Martin PR, Grünert U. Analysis of bipolar and amacrine populations in marmoset retina. J Comp Neurol 2014; 523:313-34. [DOI: 10.1002/cne.23683] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 09/22/2014] [Accepted: 09/22/2014] [Indexed: 12/29/2022]
Affiliation(s)
- Felix Weltzien
- Department of Ophthalmology and Save Sight Institute; The University of Sydney; Sydney New South Wales 2000 Australia
| | - Kumiko A. Percival
- Department of Ophthalmology and Save Sight Institute; The University of Sydney; Sydney New South Wales 2000 Australia
| | - Paul R. Martin
- Department of Ophthalmology and Save Sight Institute; The University of Sydney; Sydney New South Wales 2000 Australia
- Australian Research Council Centre of Excellence for Integrative Brain Function; The University of Sydney; Sydney New South Wales 2000 Australia
- School of Medical Sciences, The University of Sydney; Sydney New South Wales 2000 Australia
| | - Ulrike Grünert
- Department of Ophthalmology and Save Sight Institute; The University of Sydney; Sydney New South Wales 2000 Australia
- Australian Research Council Centre of Excellence for Integrative Brain Function; The University of Sydney; Sydney New South Wales 2000 Australia
- School of Medical Sciences, The University of Sydney; Sydney New South Wales 2000 Australia
| |
Collapse
|
47
|
Suzuki Y, Kiyokage E, Sohn J, Hioki H, Toida K. Structural basis for serotonergic regulation of neural circuits in the mouse olfactory bulb. J Comp Neurol 2014; 523:262-80. [PMID: 25234191 DOI: 10.1002/cne.23680] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 09/17/2014] [Accepted: 09/17/2014] [Indexed: 11/05/2022]
Abstract
Olfactory processing is well known to be regulated by centrifugal afferents from other brain regions, such as noradrenergic, acetylcholinergic, and serotonergic neurons. Serotonergic neurons widely innervate and regulate the functions of various brain regions. In the present study, we focused on serotonergic regulation of the olfactory bulb (OB), one of the most structurally and functionally well-defined brain regions. Visualization of a single neuron among abundant and dense fibers is essential to characterize and understand neuronal circuits. We accomplished this visualization by successfully labeling and reconstructing serotonin (5-hydroxytryptamine: 5-HT) neurons by infection with sindbis and adeno-associated virus into dorsal raphe nuclei (DRN) of mice. 5-HT synapses were analyzed by correlative confocal laser microscopy and serial-electron microscopy (EM) study. To further characterize 5-HT neuronal and network function, we analyzed whether glutamate was released from 5-HT synaptic terminals using immuno-EM. Our results are the first visualizations of complete 5-HT neurons and fibers projecting from DRN to the OB with bifurcations. We found that a single 5-HT axon can form synaptic contacts to both type 1 and 2 periglomerular cells within a single glomerulus. Through immunolabeling, we also identified vesicular glutamate transporter 3 in 5-HT neurons terminals, indicating possible glutamatergic transmission. Our present study strongly implicates the involvement of brain regions such as the DRN in regulation of the elaborate mechanisms of olfactory processing. We further provide a structure basis of the network for coordinating or linking olfactory encoding with other neural systems, with special attention to serotonergic regulation.
Collapse
Affiliation(s)
- Yoshinori Suzuki
- Department of Anatomy, Kawasaki Medical School, Okayama, 701-0192, Japan
| | | | | | | | | |
Collapse
|
48
|
Immunolocalization of the P2X4 receptor on neurons and glia in the mammalian retina. Neuroscience 2014; 277:55-71. [DOI: 10.1016/j.neuroscience.2014.06.055] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 06/23/2014] [Accepted: 06/24/2014] [Indexed: 02/07/2023]
|
49
|
Singh RK, Kolandaivelu S, Ramamurthy V. Early alteration of retinal neurons in Aipl1-/- animals. Invest Ophthalmol Vis Sci 2014; 55:3081-92. [PMID: 24736053 DOI: 10.1167/iovs.13-13728] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Mutations in the photoreceptor cell-specific gene encoding aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) lead to Leber congenital amaurosis (LCA4), retinitis pigmentosa, and cone-rod dystrophy. Gene therapy appears to be promising in the treatment for AIPL1-mediated vision loss in humans. Prior to initiating these treatments, however, it is crucial to understand how the retinal neurons remodel themselves in response to photoreceptor cell degeneration. In this study, using an animal model for AIPL1-LCA, Aipl1(-/-) mice, we investigate the changes in postreceptoral retinal neurons during the course of photoreceptor cell loss. METHODS Morphology of the Aipl1(-/-) retina from postnatal day 8 to 150 was compared to that of age-matched, wild-type C57Bl6/J retina (WT) by immunocytochemistry using cell-specific markers. RESULTS Expression of postsynaptic proteins in bipolar cells is reduced prior to photoreceptor cell degeneration at postnatal day 8. Bipolar and horizontal cells retract their dendrites. Cell bodies and axons of bipolar and horizontal cells are disorganized during the course of degeneration. Müller cell processes become hypertrophic and form a dense fibrotic layer outside the inner nuclear layer. CONCLUSIONS An early defect in photoreceptor cells in the AIPL1-LCA mouse model affects the expression of postsynaptic markers, suggesting abnormal development of bipolar synapses. Once degeneration of photoreceptor cells is initiated, remodeling of retinal neurons in the Aipl1(-/-) animal is rapid.
Collapse
Affiliation(s)
- Ratnesh Kumar Singh
- Departments of Ophthalmology and Biochemistry, Center for Neuroscience, West Virginia University, Morgantown, West Virginia, United States
| | - Saravanan Kolandaivelu
- Departments of Ophthalmology and Biochemistry, Center for Neuroscience, West Virginia University, Morgantown, West Virginia, United States
| | - Visvanathan Ramamurthy
- Departments of Ophthalmology and Biochemistry, Center for Neuroscience, West Virginia University, Morgantown, West Virginia, United States
| |
Collapse
|
50
|
Nivison-Smith L, Sun D, Fletcher EL, Marc RE, Kalloniatis M. Mapping kainate activation of inner neurons in the rat retina. J Comp Neurol 2014; 521:2416-38. [PMID: 23348566 DOI: 10.1002/cne.23305] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 12/06/2012] [Accepted: 01/17/2013] [Indexed: 11/10/2022]
Abstract
Kainate receptors mediate fast, excitatory synaptic transmission for a range of inner neurons in the mammalian retina. However, allocation of functional kainate receptors to known cell types and their sensitivity remains unresolved. Using the cation channel probe 1-amino-4-guanidobutane agmatine (AGB), we investigated kainate sensitivity of neurochemically identified cell populations within the structurally intact rat retina. Most inner retinal neuron populations responded to kainate in a concentration-dependent manner. OFF cone bipolar cells demonstrated the highest sensitivity of all inner neurons to kainate. Immunocytochemical localization of AGB and macromolecular markers confirmed that type 2 bipolar cells were part of this kainate-sensitive population. The majority of amacrine (ACs) and ganglion cells (GCs) showed kainate responses with different sensitivities between major neurochemical classes (γ-aminobutyric acid [GABA]/glycine ACs > glycine ACs > GABA ACs; glutamate [Glu]/weakly GABA GCs > Glu GCs). Conventional and displaced cholinergic ACs were highly responsive to kainate, whereas dopaminergic ACs do not appear to express functional kainate receptors. These findings further contribute to our understanding of neuronal networks in complex multicellular tissues.
Collapse
Affiliation(s)
- Lisa Nivison-Smith
- School of Optometry and Vision Science, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | | | | | | | | |
Collapse
|