1
|
Skelin L, Racetin A, Kelam N, Ogorevc M, Znaor L, Saraga-Babić M, Filipović N, Katsuyama Y, Pogorelić Z, Vukojević K. Connexin Expression Is Altered in the Eye Development of Yotari Mice: A Preliminary Study. Biomolecules 2024; 14:1174. [PMID: 39334940 PMCID: PMC11430515 DOI: 10.3390/biom14091174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
This study aimed to explore how Dab1 functional silencing influences the expression patterns of different connexins in the developing yotari (yot) mice eyes as potential determinants of retinogenesis. Using immunofluorescence staining, the protein expression of Dab1, Reelin, and connexin 37, 40, 43, and 45 (Cx37, Cx40, Cx43, and Cx45) in the wild-type (wt) and yot eyes at embryonic days 13.5 and 15.5 (E13.5 and E15.5) were analyzed. Different expression patterns of Cx37 were seen between the wt and yot groups. The highest fluorescence intensity of Cx37 was observed in the yot animals at E15.5. Cx40 had higher expression at the E13.5 when differentiation of retinal layers was still beginning, whereas it decreased at the E15.5 when differentiation was at the advanced stage. Higher expression of Cx43 was found in the yot group at both time points. Cx45 was predominantly expressed at E13.5 in both groups. Our results reveal the altered expression of connexins during retinogenesis in yot mice and their potential involvement in retinal pathology, where they might serve as prospective therapeutic targets.
Collapse
Affiliation(s)
- Ljubica Skelin
- Clinical Department of Ophthalmology, University Hospital of Split, 21000 Split, Croatia
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Marin Ogorevc
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Ljubo Znaor
- Clinical Department of Ophthalmology, University Hospital of Split, 21000 Split, Croatia
- Department of Ophthalmology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Mirna Saraga-Babić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Yu Katsuyama
- Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Zenon Pogorelić
- Department of Pediatric Surgery, Split University Hospital, 21000 Split, Croatia
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| |
Collapse
|
2
|
Voufo C, Chen AQ, Smith BE, Yan R, Feller MB, Tiriac A. Circuit mechanisms underlying embryonic retinal waves. eLife 2023; 12:e81983. [PMID: 36790167 PMCID: PMC9988258 DOI: 10.7554/elife.81983] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 02/13/2023] [Indexed: 02/16/2023] Open
Abstract
Spontaneous activity is a hallmark of developing neural systems. In the retina, spontaneous activity comes in the form of retinal waves, comprised of three stages persisting from embryonic day 16 (E16) to eye opening at postnatal day 14 (P14). Though postnatal retinal waves have been well characterized, little is known about the spatiotemporal properties or the mechanisms mediating embryonic retinal waves, designated stage 1 waves. Using a custom-built macroscope to record spontaneous calcium transients from whole embryonic retinas, we show that stage 1 waves are initiated at several locations across the retina and propagate across a broad range of areas. Blocking gap junctions reduced the frequency and size of stage 1 waves, nearly abolishing them. Global blockade of nAChRs similarly nearly abolished stage 1 waves. Thus, stage 1 waves are mediated by a complex circuitry involving subtypes of nAChRs and gap junctions. Stage 1 waves in mice lacking the β2 subunit of the nAChRs (β2-nAChR-KO) persisted with altered propagation properties and were abolished by a gap junction blocker. To assay the impact of stage 1 waves on retinal development, we compared the spatial distribution of a subtype of retinal ganglion cells, intrinsically photosensitive retinal ganglion cells (ipRGCs), which undergo a significant amount of cell death, in WT and β2-nAChR-KO mice. We found that the developmental decrease in ipRGC density is preserved between WT and β2-nAChR-KO mice, indicating that processes regulating ipRGC numbers and distributions are not influenced by spontaneous activity.
Collapse
Affiliation(s)
- Christiane Voufo
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
| | - Andy Quaen Chen
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Benjamin E Smith
- School of Optometry, University of California, BerkeleyBerkeleyUnited States
| | - Rongshan Yan
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Marla B Feller
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Alexandre Tiriac
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
3
|
Fitzpatrick MJ, Kerschensteiner D. Homeostatic plasticity in the retina. Prog Retin Eye Res 2022; 94:101131. [PMID: 36244950 DOI: 10.1016/j.preteyeres.2022.101131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 02/07/2023]
Abstract
Vision begins in the retina, whose intricate neural circuits extract salient features of the environment from the light entering our eyes. Neurodegenerative diseases of the retina (e.g., inherited retinal degenerations, age-related macular degeneration, and glaucoma) impair vision and cause blindness in a growing number of people worldwide. Increasing evidence indicates that homeostatic plasticity (i.e., the drive of a neural system to stabilize its function) can, in principle, preserve retinal function in the face of major perturbations, including neurodegeneration. Here, we review the circumstances and events that trigger homeostatic plasticity in the retina during development, sensory experience, and disease. We discuss the diverse mechanisms that cooperate to compensate and the set points and outcomes that homeostatic retinal plasticity stabilizes. Finally, we summarize the opportunities and challenges for unlocking the therapeutic potential of homeostatic plasticity. Homeostatic plasticity is fundamental to understanding retinal development and function and could be an important tool in the fight to preserve and restore vision.
Collapse
|
4
|
Chini M, Pfeffer T, Hanganu-Opatz I. An increase of inhibition drives the developmental decorrelation of neural activity. eLife 2022; 11:78811. [PMID: 35975980 PMCID: PMC9448324 DOI: 10.7554/elife.78811] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/16/2022] [Indexed: 11/23/2022] Open
Abstract
Throughout development, the brain transits from early highly synchronous activity patterns to a mature state with sparse and decorrelated neural activity, yet the mechanisms underlying this process are poorly understood. The developmental transition has important functional consequences, as the latter state is thought to allow for more efficient storage, retrieval, and processing of information. Here, we show that, in the mouse medial prefrontal cortex (mPFC), neural activity during the first two postnatal weeks decorrelates following specific spatial patterns. This process is accompanied by a concomitant tilting of excitation-inhibition (E-I) ratio toward inhibition. Using optogenetic manipulations and neural network modeling, we show that the two phenomena are mechanistically linked, and that a relative increase of inhibition drives the decorrelation of neural activity. Accordingly, in mice mimicking the etiology of neurodevelopmental disorders, subtle alterations in E-I ratio are associated with specific impairments in the correlational structure of spike trains. Finally, capitalizing on EEG data from newborn babies, we show that an analogous developmental transition takes place also in the human brain. Thus, changes in E-I ratio control the (de)correlation of neural activity and, by these means, its developmental imbalance might contribute to the pathogenesis of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Mattia Chini
- Institute of Developmental Neurophysiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Pfeffer
- Center for Brain and Cognition, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ileana Hanganu-Opatz
- Institute of Developmental Neurophysiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
5
|
Caval-Holme FS, Aranda ML, Chen AQ, Tiriac A, Zhang Y, Smith B, Birnbaumer L, Schmidt TM, Feller MB. The Retinal Basis of Light Aversion in Neonatal Mice. J Neurosci 2022; 42:4101-4115. [PMID: 35396331 PMCID: PMC9121827 DOI: 10.1523/jneurosci.0151-22.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 11/21/2022] Open
Abstract
Aversive responses to bright light (photoaversion) require signaling from the eye to the brain. Melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) encode absolute light intensity and are thought to provide the light signals for photoaversion. Consistent with this, neonatal mice exhibit photoaversion before the developmental onset of image vision, and melanopsin deletion abolishes photoaversion in neonates. It is not well understood how the population of ipRGCs, which constitutes multiple physiologically distinct types (denoted M1-M6 in mouse), encodes light stimuli to produce an aversive response. Here, we provide several lines of evidence that M1 ipRGCs that lack the Brn3b transcription factor drive photoaversion in neonatal mice. First, neonatal mice lacking TRPC6 and TRPC7 ion channels failed to turn away from bright light, while two photon Ca2+ imaging of their acutely isolated retinas revealed reduced photosensitivity in M1 ipRGCs, but not other ipRGC types. Second, mice in which all ipRGC types except for Brn3b-negative M1 ipRGCs are ablated exhibited normal photoaversion. Third, pharmacological blockade or genetic knockout of gap junction channels expressed by ipRGCs, which reduces the light sensitivity of M2-M6 ipRGCs in the neonatal retina, had small effects on photoaversion only at the brightest light intensities. Finally, M1s were not strongly depolarized by spontaneous retinal waves, a robust source of activity in the developing retina that depolarizes all other ipRGC types. M1s therefore constitute a separate information channel between the neonatal retina and brain that could ensure behavioral responses to light but not spontaneous retinal waves.SIGNIFICANCE STATEMENT At an early stage of development, before the maturation of photoreceptor input to the retina, neonatal mice exhibit photoaversion. On exposure to bright light, they turn away and emit ultrasonic vocalizations, a cue to their parents to return them to the nest. Neonatal photoaversion is mediated by intrinsically photosensitive retinal ganglion cells (ipRGCs), a small percentage of the retinal ganglion cell population that express the photopigment melanopsin and depolarize directly in response to light. This study shows that photoaversion is mediated by a subset of ipRGCs, called M1-ipRGCs. Moreover, M1-ipRGCs have reduced responses to retinal waves, providing a mechanism by which the mouse distinguishes light stimulation from developmental patterns of spontaneous activity.
Collapse
Affiliation(s)
- Franklin S Caval-Holme
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California 94720
| | - Marcos L Aranda
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208
| | - Andy Q Chen
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Alexandre Tiriac
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Yizhen Zhang
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Benjamin Smith
- School of Optometry, University of California Berkeley, Berkeley, California 94720
| | - Lutz Birnbaumer
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina 27709
- Institute of Biomedical Research, School of Medical Sciences, Catholic University of Argentina, Buenos Aires, Argentina C1107AFF
| | - Tiffany M Schmidt
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Marla B Feller
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California 94720
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| |
Collapse
|
6
|
Xie S, Li H, Yao F, Huang J, Yang X, Chen X, Liu Q, Zhuang M, He S. PUPIL enables mapping and stamping of transient electrical connectivity in developing nervous systems. Cell Rep 2021; 37:109853. [PMID: 34686323 DOI: 10.1016/j.celrep.2021.109853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/25/2021] [Accepted: 09/28/2021] [Indexed: 10/20/2022] Open
Abstract
Currently, many genetic methods are available for mapping chemical connectivity, but analogous methods for electrical synapses are lacking. Here, we present pupylation-based interaction labeling (PUPIL), a genetically encoded system for noninvasively mapping and stamping transient electrical synapses in the mouse brain. Upon fusion of connexin 26 (CX26) with the ligase PafA, pupylation yields tag puncta following conjugation of its substrate, a biotin- or fluorescent-protein-tagged PupE, to the neighboring proteins of electrical synapses containing CX26-PafA. Tag puncta are validated to correlate well with functional electrical synapses in immature neurons. Furthermore, puncta are retained in mature neurons when electrical synapses mostly disappear-suggesting successful stamping. We use PUPIL to uncover spatial subcellular localizations of electrical synapses and approach their physiological functions during development. Thus, PUPIL is a powerful tool for probing electrical connectivity patterns in complex nervous systems and has great potential for transient receptors and ion channels as well.
Collapse
Affiliation(s)
- Shu Xie
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China; 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; University of Chinese Academy of Sciences, Beijing, China
| | - Haixiang Li
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China; 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; University of Chinese Academy of Sciences, Beijing, China
| | - Fenyong Yao
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Jiechang Huang
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China; 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; University of Chinese Academy of Sciences, Beijing, China
| | - Xiaomei Yang
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Xin Chen
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China; 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; University of Chinese Academy of Sciences, Beijing, China
| | - Qiang Liu
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China; 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; University of Chinese Academy of Sciences, Beijing, China
| | - Min Zhuang
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China
| | - Shuijin He
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong New District, Shanghai 201210, China.
| |
Collapse
|
7
|
Linden NJ, Tabuena DR, Steinmetz NA, Moody WJ, Brunton SL, Brunton BW. Go with the FLOW: visualizing spatiotemporal dynamics in optical widefield calcium imaging. J R Soc Interface 2021; 18:20210523. [PMID: 34428947 PMCID: PMC8385384 DOI: 10.1098/rsif.2021.0523] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/02/2021] [Indexed: 12/11/2022] Open
Abstract
Widefield calcium imaging has recently emerged as a powerful experimental technique to record coordinated large-scale brain activity. These measurements present a unique opportunity to characterize spatiotemporally coherent structures that underlie neural activity across many regions of the brain. In this work, we leverage analytic techniques from fluid dynamics to develop a visualization framework that highlights features of flow across the cortex, mapping wavefronts that may be correlated with behavioural events. First, we transform the time series of widefield calcium images into time-varying vector fields using optic flow. Next, we extract concise diagrams summarizing the dynamics, which we refer to as FLOW (flow lines in optical widefield imaging) portraits. These FLOW portraits provide an intuitive map of dynamic calcium activity, including regions of initiation and termination, as well as the direction and extent of activity spread. To extract these structures, we use the finite-time Lyapunov exponent technique developed to analyse time-varying manifolds in unsteady fluids. Importantly, our approach captures coherent structures that are poorly represented by traditional modal decomposition techniques. We demonstrate the application of FLOW portraits on three simple synthetic datasets and two widefield calcium imaging datasets, including cortical waves in the developing mouse and spontaneous cortical activity in an adult mouse.
Collapse
Affiliation(s)
- Nathaniel J. Linden
- Department of Bioengineering, University of Washington, Seattle, WA 98195-0005, USA
- Department of Biology, University of Washington, Seattle, WA 98195-0005, USA
| | - Dennis R. Tabuena
- Department of Biology, University of Washington, Seattle, WA 98195-0005, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195-0005, USA
| | - Nicholas A. Steinmetz
- Department of Biological Structure, University of Washington, Seattle, WA 98195-0005, USA
| | - William J. Moody
- Department of Biology, University of Washington, Seattle, WA 98195-0005, USA
| | - Steven L. Brunton
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195-0005, USA
| | - Bingni W. Brunton
- Department of Biology, University of Washington, Seattle, WA 98195-0005, USA
| |
Collapse
|
8
|
Choi BJ, Chen YCD, Desplan C. Building a circuit through correlated spontaneous neuronal activity in the developing vertebrate and invertebrate visual systems. Genes Dev 2021; 35:677-691. [PMID: 33888564 PMCID: PMC8091978 DOI: 10.1101/gad.348241.121] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
During the development of the vertebrate nervous systems, genetic programs assemble an immature circuit that is subsequently refined by neuronal activity evoked by external stimuli. However, prior to sensory experience, the intrinsic property of the developing nervous system also triggers correlated network-level neuronal activity, with retinal waves in the developing vertebrate retina being the best documented example. Spontaneous activity has also been found in the visual system of Drosophila Here, we compare the spontaneous activity of the developing visual system between mammalian and Drosophila and suggest that Drosophila is an emerging model for mechanistic and functional studies of correlated spontaneous activity.
Collapse
Affiliation(s)
- Ben Jiwon Choi
- Department of Biology, New York University, New York, New York 10003, USA
| | | | - Claude Desplan
- Department of Biology, New York University, New York, New York 10003, USA
| |
Collapse
|
9
|
Abstract
Retinal ganglion cells (RGCs) serve as a crucial communication channel from the retina to the brain. In the adult, these cells receive input from defined sets of presynaptic partners and communicate with postsynaptic brain regions to convey features of the visual scene. However, in the developing visual system, RGC interactions extend beyond their synaptic partners such that they guide development before the onset of vision. In this Review, we summarize our current understanding of how interactions between RGCs and their environment influence cellular targeting, migration and circuit maturation during visual system development. We describe the roles of RGC subclasses in shaping unique developmental responses within the retina and at central targets. Finally, we highlight the utility of RNA sequencing and genetic tools in uncovering RGC type-specific roles during the development of the visual system.
Collapse
Affiliation(s)
- Shane D'Souza
- The Visual Systems Group, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
- Center for Chronobiology, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| | - Richard A Lang
- The Visual Systems Group, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
- Center for Chronobiology, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
- Division of Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
- Department of Ophthalmology, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
10
|
Del Pino I, Tocco C, Magrinelli E, Marcantoni A, Ferraguto C, Tomagra G, Bertacchi M, Alfano C, Leinekugel X, Frick A, Studer M. COUP-TFI/Nr2f1 Orchestrates Intrinsic Neuronal Activity during Development of the Somatosensory Cortex. Cereb Cortex 2020; 30:5667-5685. [DOI: 10.1093/cercor/bhaa137] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/17/2020] [Accepted: 04/29/2020] [Indexed: 01/19/2023] Open
Abstract
Abstract
The formation of functional cortical maps in the cerebral cortex results from a timely regulated interaction between intrinsic genetic mechanisms and electrical activity. To understand how transcriptional regulation influences network activity and neuronal excitability within the neocortex, we used mice deficient for Nr2f1 (also known as COUP-TFI), a key determinant of primary somatosensory (S1) area specification during development. We found that the cortical loss of Nr2f1 impacts on spontaneous network activity and synchronization of S1 cortex at perinatal stages. In addition, we observed alterations in the intrinsic excitability and morphological features of layer V pyramidal neurons. Accordingly, we identified distinct voltage-gated ion channels regulated by Nr2f1 that might directly influence intrinsic bioelectrical properties during critical time windows of S1 cortex specification. Altogether, our data suggest a tight link between Nr2f1 and neuronal excitability in the developmental sequence that ultimately sculpts the emergence of cortical network activity within the immature neocortex.
Collapse
Affiliation(s)
- Isabel Del Pino
- Université de Bordeaux, Inserm U1215, Neurocentre Magendie, 33077 Bordeaux, France
- Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
| | - Chiara Tocco
- Université Côte d’Azur, CNRS, Inserm, iBV, 06108 Nice, France
| | - Elia Magrinelli
- Université Côte d’Azur, CNRS, Inserm, iBV, 06108 Nice, France
- Département des Neurosciences Fondamentales, Université de Lausanne, CH-1005 Lausanne, Switzerland
| | - Andrea Marcantoni
- Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, 10125 Torino, Italy
| | | | - Giulia Tomagra
- Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, 10125 Torino, Italy
| | | | | | - Xavier Leinekugel
- Université de Bordeaux, Inserm U1215, Neurocentre Magendie, 33077 Bordeaux, France
| | - Andreas Frick
- Université de Bordeaux, Inserm U1215, Neurocentre Magendie, 33077 Bordeaux, France
| | - Michèle Studer
- Université Côte d’Azur, CNRS, Inserm, iBV, 06108 Nice, France
| |
Collapse
|
11
|
dos Santos ÍGD, de Oliveira Mendes TA, Silva GAB, Reis AMS, Monteiro-Vitorello CB, Schaker PDC, Herai RH, Fabotti ABC, Coutinho LL, Jorge EC. Didelphis albiventris: an overview of unprecedented transcriptome sequencing of the white-eared opossum. BMC Genomics 2019; 20:866. [PMID: 31730444 PMCID: PMC6858782 DOI: 10.1186/s12864-019-6240-x] [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: 07/20/2019] [Accepted: 10/29/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The white-eared opossum (Didelphis albiventris) is widely distributed throughout Brazil and South America. It has been used as an animal model for studying different scientific questions ranging from the restoration of degraded green areas to medical aspects of Chagas disease, leishmaniasis and resistance against snake venom. As a marsupial, D. albiventris can also contribute to the understanding of the molecular mechanisms that govern the different stages of organogenesis. Opossum joeys are born after only 13 days, and the final stages of organogenesis occur when the neonates are inside the pouch, depending on lactation. As neither the genome of this opossum species nor its transcriptome has been completely sequenced, the use of D. albiventris as an animal model is limited. In this work, we sequenced the D. albiventris transcriptome by RNA-seq to obtain the first catalogue of differentially expressed (DE) genes and gene ontology (GO) annotations during the neonatal stages of marsupial development. RESULTS The D. albiventris transcriptome was obtained from whole neonates harvested at birth (P0), at 5 days of age (P5) and at 10 days of age (P10). The de novo assembly of these transcripts generated 85,338 transcripts. Approximately 30% of these transcripts could be mapped against the amino acid sequences of M. domestica, the evolutionarily closest relative of D. albiventris to be sequenced thus far. Among the expressed transcripts, 2077 were found to be DE between P0 and P5, 13,780 between P0 and P10, and 1453 between P5 and P10. The enriched GO terms were mainly related to the immune system, blood tissue development and differentiation, vision, hearing, digestion, the CNS and limb development. CONCLUSIONS The elucidation of opossum transcriptomes provides an out-group for better understanding the distinct characteristics associated with the evolution of mammalian species. This study provides the first transcriptome sequences and catalogue of genes for a marsupial species at different neonatal stages, allowing the study of the mechanisms involved in organogenesis.
Collapse
Affiliation(s)
- Íria Gabriela Dias dos Santos
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | | | - Gerluza Aparecida Borges Silva
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | - Amanda Maria Sena Reis
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| | | | - Patricia Dayane Carvalho Schaker
- Departamento de Genética, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, São Paulo Brazil
| | - Roberto Hirochi Herai
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Paraná, Brazil
| | | | - Luiz Lehmann Coutinho
- Departamento de Zootecnia, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, São Paulo Brazil
| | - Erika Cristina Jorge
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais Brazil
| |
Collapse
|
12
|
Lin CI, Chiao CC. Blue Light Promotes Neurite Outgrowth of Retinal Explants in Postnatal ChR2 Mice. eNeuro 2019; 6:ENEURO.0391-18.2019. [PMID: 31362954 PMCID: PMC6712202 DOI: 10.1523/eneuro.0391-18.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 07/05/2019] [Accepted: 07/19/2019] [Indexed: 11/29/2022] Open
Abstract
Neurons in the adult mammalian CNS fails to regenerate after severe injury. However, it is known that an increase in neural activity occurs in mouse retinal ganglion cells (RGCs) after extrinsic stimulation and this can induce axon growth. In the present study, we applied an optogenetic approach using a mouse model, specifically involving channelrhodopsin-2 (ChR2) expression in RGCs. We investigated whether modulation of RGC neural activity exclusively by blue light stimulation is able to promote neurite outgrowth of postnatal retinal explants. The results showed that activation of RGCs expressing ChR2 by 20 Hz blue light for 1 h is a most effective way of enhancing neurite outgrowth in postnatal retinas. This is achieved via gap junctions that spread neural activity across the whole retina. Moreover, we found that activation of intrinsic photosensitive RGCs (ipRGCs) by blue light also contributes significantly to the promotion of neurite outgrowth in the same postnatal retinal explants. Our findings not only demonstrate that a short-term increase in RGC neural activity is sufficient to facilitate the neurite outgrowth of retinal explants, but also highlight the fact that the temporal pattern of neural activity in RGCs is a critical factor in regulating axon regeneration.
Collapse
Affiliation(s)
- Chin-I Lin
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chuan-Chin Chiao
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu 30013, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
13
|
Kähne M, Rüdiger S, Kihara AH, Lindner B. Gap junctions set the speed and nucleation rate of stage I retinal waves. PLoS Comput Biol 2019; 15:e1006355. [PMID: 31034472 PMCID: PMC6508742 DOI: 10.1371/journal.pcbi.1006355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 05/09/2019] [Accepted: 11/27/2018] [Indexed: 11/18/2022] Open
Abstract
Spontaneous waves in the developing retina are essential in the formation of the retinotopic mapping in the visual system. From experiments in rabbits, it is known that the earliest type of retinal waves (stage I) is nucleated spontaneously, propagates at a speed of 451±91 μm/sec and relies on gap junction coupling between ganglion cells. Because gap junctions (electrical synapses) have short integration times, it has been argued that they cannot set the low speed of stage I retinal waves. Here, we present a theoretical study of a two-dimensional neural network of the ganglion cell layer with gap junction coupling and intrinsic noise. We demonstrate that this model can explain observed nucleation rates as well as the comparatively slow propagation speed of the waves. From the interaction between two coupled neurons, we estimate the wave speed in the model network. Furthermore, using simulations of small networks of neurons (N≤260), we estimate the nucleation rate in the form of an Arrhenius escape rate. These results allow for informed simulations of a realistically sized network, yielding values of the gap junction coupling and the intrinsic noise level that are in a physiologically plausible range.
Collapse
Affiliation(s)
- Malte Kähne
- Institut für Physik, Humboldt-Universität zu Berlin, Berlin, Germany
- * E-mail:
| | - Sten Rüdiger
- Institut für Physik, Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Benjamin Lindner
- Institut für Physik, Humboldt-Universität zu Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| |
Collapse
|
14
|
Abudara V, Retamal MA, Del Rio R, Orellana JA. Synaptic Functions of Hemichannels and Pannexons: A Double-Edged Sword. Front Mol Neurosci 2018; 11:435. [PMID: 30564096 PMCID: PMC6288452 DOI: 10.3389/fnmol.2018.00435] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/08/2018] [Indexed: 01/18/2023] Open
Abstract
The classical view of synapses as the functional contact between presynaptic and postsynaptic neurons has been challenged in recent years by the emerging regulatory role of glial cells. Astrocytes, traditionally considered merely supportive elements are now recognized as active modulators of synaptic transmission and plasticity at the now so-called "tripartite synapse." In addition, an increasing body of evidence indicates that beyond immune functions microglia also participate in various processes aimed to shape synaptic plasticity. Release of neuroactive compounds of glial origin, -process known as gliotransmission-, constitute a widespread mechanism through which glial cells can either potentiate or reduce the synaptic strength. The prevailing vision states that gliotransmission depends on an intracellular Ca2+/exocytotic-mediated release; notwithstanding, growing evidence is pointing at hemichannels (connexons) and pannexin channels (pannexons) as alternative non-vesicular routes for gliotransmitters efflux. In concurrence with this novel concept, both hemichannels and pannexons are known to mediate the transfer of ions and signaling molecules -such as ATP and glutamate- between the cytoplasm and the extracellular milieu. Importantly, recent reports show that glial hemichannels and pannexons are capable to perceive synaptic activity and to respond to it through changes in their functional state. In this article, we will review the current information supporting the "double edge sword" role of hemichannels and pannexons in the function of central and peripheral synapses. At one end, available data support the idea that these channels are chief components of a feedback control mechanism through which gliotransmitters adjust the synaptic gain in either resting or stimulated conditions. At the other end, we will discuss how the excitotoxic release of gliotransmitters and [Ca2+]i overload linked to the opening of hemichannels/pannexons might impact cell function and survival in the nervous system.
Collapse
Affiliation(s)
- Verónica Abudara
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile.,Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Santiago, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Envejecimiento y Regeneración, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes, Universidad de Magallanes, Punta Arenas, Chile
| | - Juan A Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes, Santiago, Chile
| |
Collapse
|
15
|
Tiriac A, Smith BE, Feller MB. Light Prior to Eye Opening Promotes Retinal Waves and Eye-Specific Segregation. Neuron 2018; 100:1059-1065.e4. [PMID: 30392793 DOI: 10.1016/j.neuron.2018.10.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 08/24/2018] [Accepted: 10/05/2018] [Indexed: 11/16/2022]
Abstract
Retinal waves are bursts of correlated activity that occur prior to eye opening and provide a critical source of activity that drives the refinement of retinofugal projections. Retinal waves are thought to be initiated spontaneously with their spatiotemporal features dictated by immature neural circuits. Here we demonstrate that, during the second postnatal week in mice, changes in light intensity dictate where and when a subset of retinal waves are triggered via activation of conventional photoreceptors. Propagation properties of triggered waves are indistinguishable from spontaneous waves, indicating that they are activating the same retinal circuits. Using whole-brain imaging techniques, we demonstrate that light deprivation prior to eye opening diminishes eye-specific segregation of the retinal projections to the dorsolateral geniculate nucleus of the thalamus, but not other retinal targets. These data indicate that light that passes through the closed eyelids plays a critical role in the development of the image-forming visual system.
Collapse
Affiliation(s)
- Alexandre Tiriac
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Benjamin E Smith
- School of Optometry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Marla B Feller
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
16
|
O'Brien J, Bloomfield SA. Plasticity of Retinal Gap Junctions: Roles in Synaptic Physiology and Disease. Annu Rev Vis Sci 2018; 4:79-100. [DOI: 10.1146/annurev-vision-091517-034133] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Electrical synaptic transmission via gap junctions underlies direct and rapid neuronal communication in the central nervous system. The diversity of functional roles played by electrical synapses is perhaps best exemplified in the vertebrate retina, in which gap junctions are expressed by each of the five major neuronal types. These junctions are highly plastic; they are dynamically regulated by ambient illumination and circadian rhythms acting through light-activated neuromodulators. The networks formed by electrically coupled neurons provide plastic, reconfigurable circuits positioned to play key and diverse roles in the transmission and processing of visual information at every retinal level. Recent work indicates gap junctions also play a role in the progressive cell death and aberrant activity seen in various pathological conditions of the retina. Gap junctions thus form potential targets for novel neuroprotective therapies in the treatment of neurodegenerative retinal diseases such as glaucoma and ischemic retinopathies.
Collapse
Affiliation(s)
- John O'Brien
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Stewart A. Bloomfield
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY 10036, USA
| |
Collapse
|
17
|
Akopian A, Kumar S, Ramakrishnan H, Roy K, Viswanathan S, Bloomfield SA. Targeting neuronal gap junctions in mouse retina offers neuroprotection in glaucoma. J Clin Invest 2017; 127:2647-2661. [PMID: 28604388 PMCID: PMC5490768 DOI: 10.1172/jci91948] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/27/2017] [Indexed: 01/26/2023] Open
Abstract
The progressive death of retinal ganglion cells and resulting visual deficits are hallmarks of glaucoma, but the underlying mechanisms remain unclear. In many neurodegenerative diseases, cell death induced by primary insult is followed by a wave of secondary loss. Gap junctions (GJs), intercellular channels composed of subunit connexins, can play a major role in secondary cell death by forming conduits through which toxic molecules from dying cells pass to and injure coupled neighbors. Here we have shown that pharmacological blockade of GJs or genetic ablation of connexin 36 (Cx36) subunits, which are highly expressed by retinal neurons, markedly reduced loss of neurons and optic nerve axons in a mouse model of glaucoma. Further, functional parameters that are negatively affected in glaucoma, including the electroretinogram, visual evoked potential, visual spatial acuity, and contrast sensitivity, were maintained at control levels when Cx36 was ablated. Neuronal GJs may thus represent potential therapeutic targets to prevent the progressive neurodegeneration and visual impairment associated with glaucoma.
Collapse
|
18
|
Chew KS, Renna JM, McNeill DS, Fernandez DC, Keenan WT, Thomsen MB, Ecker JL, Loevinsohn GS, VanDunk C, Vicarel DC, Tufford A, Weng S, Gray PA, Cayouette M, Herzog ED, Zhao H, Berson DM, Hattar S. A subset of ipRGCs regulates both maturation of the circadian clock and segregation of retinogeniculate projections in mice. eLife 2017; 6:e22861. [PMID: 28617242 PMCID: PMC5513697 DOI: 10.7554/elife.22861] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 06/14/2017] [Indexed: 12/25/2022] Open
Abstract
The visual system consists of two major subsystems, image-forming circuits that drive conscious vision and non-image-forming circuits for behaviors such as circadian photoentrainment. While historically considered non-overlapping, recent evidence has uncovered crosstalk between these subsystems. Here, we investigated shared developmental mechanisms. We revealed an unprecedented role for light in the maturation of the circadian clock and discovered that intrinsically photosensitive retinal ganglion cells (ipRGCs) are critical for this refinement process. In addition, ipRGCs regulate retinal waves independent of light, and developmental ablation of a subset of ipRGCs disrupts eye-specific segregation of retinogeniculate projections. Specifically, a subset of ipRGCs, comprising ~200 cells and which project intraretinally and to circadian centers in the brain, are sufficient to mediate both of these developmental processes. Thus, this subset of ipRGCs constitute a shared node in the neural networks that mediate light-dependent maturation of the circadian clock and light-independent refinement of retinogeniculate projections.
Collapse
Affiliation(s)
- Kylie S Chew
- Department of Biology, Johns Hopkins University, Baltimore, United States
- Department of Biology, Stanford University, Stanford, United States
| | - Jordan M Renna
- Department of Biology, Program in Integrated Bioscience, The University of Akron, Akron, United States
| | - David S McNeill
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Diego C Fernandez
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - William T Keenan
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Michael B Thomsen
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Jennifer L Ecker
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | | | - Cassandra VanDunk
- Department of Anatomy and Neurobiology, Washington University, St. Louis, United States
- Department of Neurobiology, Northwestern University, Evanston, United States
| | - Daniel C Vicarel
- Department of Biology, Program in Integrated Bioscience, The University of Akron, Akron, United States
| | - Adele Tufford
- Cellular Neurobiology Research Unit, Institut De Recherches Cliniques De Montréal, Montreal, Canada
| | - Shijun Weng
- Department of Neuroscience, Brown University, Providence, United States
| | - Paul A Gray
- Department of Anatomy and Neurobiology, Washington University, St. Louis, United States
- Indigo Agriculture, Charlestown, United States
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Institut De Recherches Cliniques De Montréal, Montreal, Canada
- Faculty of Medicine, Université De Montréal, Montreal, Canada
| | - Erik D Herzog
- Department of Biology, Washington University, St. Louis, United States
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - David M Berson
- Department of Neuroscience, Brown University, Providence, United States
| | - Samer Hattar
- Department of Biology, Johns Hopkins University, Baltimore, United States
| |
Collapse
|
19
|
Bosma MM. Regulation of Spontaneous Propagating Waves in the Embryonic Mouse Brainstem. Front Neural Circuits 2017; 10:110. [PMID: 28101007 PMCID: PMC5209361 DOI: 10.3389/fncir.2016.00110] [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: 10/10/2016] [Accepted: 12/15/2016] [Indexed: 12/03/2022] Open
Abstract
Spontaneous activity (SA) modulates many aspects of neural development, including neuronal phenotype, axon path-finding and synaptic connectivity. In the embryonic mouse brainstem, SA initially is recorded in isolated cells at embryonic day (E) 9.5, and 48 h later takes the form of propagating waves. The majority of these waves originate from one midline initiation zone (InZ), which is situated within the developing serotonergic raphe. InZ cells express a t-type calcium channel, are depolarized, and have high membrane resistance, the combination of which allows spontaneous depolarization. Propagating events require signaling at metabotropic 5-HT receptors; a possible source could be 5-HT released by newly differentiating 5-HT neurons. At E11.5, waves propagate throughout the hindbrain, with some events crossing into the midbrain. At E12.5, lateral cells (further than 150 μm from the midline) up-regulate expression of a K channel that increases resting conductance and hyperpolarizes them, preventing the propagation of waves laterally. At the same stage, cells in the isthmus up-regulate t-type calcium channels, permitting more events to cross into the midbrain, some of which form recurring loops of activity that are able to keep intracellular calcium levels high for many minutes. At E13.5, caudal hindbrain cells hyperpolarize utilizing the same K conductance, and 24 h later, at E14.5, the InZ hyperpolarizes and no longer undergoes spontaneous events. Thus, 5-HT receptor-dependent propagating waves in the embryonic brainstem are generated and propagated by regulation of membrane conductance. We discuss these mechanisms, and the possible role of this SA in neuronal development.
Collapse
Affiliation(s)
- Martha M Bosma
- Department of Biology, University of Washington Seattle, WA, USA
| |
Collapse
|
20
|
Ivanova E, Yee CW, Baldoni R, Sagdullaev BT. Aberrant activity in retinal degeneration impairs central visual processing and relies on Cx36-containing gap junctions. Exp Eye Res 2016; 150:81-9. [PMID: 26005040 PMCID: PMC4655183 DOI: 10.1016/j.exer.2015.05.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/13/2015] [Accepted: 05/19/2015] [Indexed: 11/20/2022]
Abstract
In retinal degenerative disease (RD), the diminished light signal from dying photoreceptors has been considered the sole cause of visual impairment. Recent studies show a 10-fold increase in spontaneous activity in the RD network, challenging this paradigm. This aberrant activity forms a new barrier for the light signal, and not only exacerbates the loss of vision, but also may stand in the way of visual restoration. This activity originates in AII amacrine cells and relies on excessive activation of gap junctions. However, it remains unclear whether aberrant activity affects central visual processing and what mechanisms lead to this excessive activation of gap junctions. By combining genetic manipulation with electrophysiological recordings of light-induced activity in both living mice and isolated wholemount retina, we demonstrate that aberrant activity extends along retinotectal projections to alter activity in higher brain centers. Next, to selectively eliminate Cx36-containing gap junctions, which are the primary type expressed by AII amacrine cells, we crossed rd10 mice, a slow-degenerating model of RD, with Cx36 knockout mice. We found that retinal aberrant activity was reduced in the rd10/Cx36KO mice compared to rd10 controls, a direct evidence for involvement of Cx36-containing gap junctions in generating aberrant activity in RD. These data provide an essential support for future experiments to determine if selectively targeting these gap junctions could be a valid strategy for reducing aberrant activity and restoring light responses in RD.
Collapse
Affiliation(s)
- Elena Ivanova
- Departments of Ophthalmology and Neurology, Weill Medical College of Cornell University, Burke Medical Research Institute, White Plains, NY 10605, USA
| | - Christopher W Yee
- Departments of Ophthalmology and Neurology, Weill Medical College of Cornell University, Burke Medical Research Institute, White Plains, NY 10605, USA
| | - Robert Baldoni
- Departments of Ophthalmology and Neurology, Weill Medical College of Cornell University, Burke Medical Research Institute, White Plains, NY 10605, USA
| | - Botir T Sagdullaev
- Departments of Ophthalmology and Neurology, Weill Medical College of Cornell University, Burke Medical Research Institute, White Plains, NY 10605, USA.
| |
Collapse
|
21
|
Zolnik TA, Connors BW. Electrical synapses and the development of inhibitory circuits in the thalamus. J Physiol 2016; 594:2579-92. [PMID: 26864476 PMCID: PMC4865577 DOI: 10.1113/jp271880] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/05/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The thalamus is a structure critical for information processing and transfer to the cortex. Thalamic reticular neurons are inhibitory cells interconnected by electrical synapses, most of which require the gap junction protein connexin36 (Cx36). We investigated whether electrical synapses play a role in the maturation of thalamic networks by studying neurons in mice with and without Cx36. When Cx36 was deleted, inhibitory synapses were more numerous, although both divergent inhibitory connectivity and dendritic complexity were reduced. Surprisingly, we observed non-Cx36-dependent electrical synapses with unusual biophysical properties interconnecting some reticular neurons in mice lacking Cx36. The results of the present study suggest an important role for Cx36-dependent electrical synapses in the development of thalamic circuits. ABSTRACT Neurons within the mature thalamic reticular nucleus (TRN) powerfully inhibit ventrobasal (VB) thalamic relay neurons via GABAergic synapses. TRN neurons are also coupled to one another by electrical synapses that depend strongly on the gap junction protein connexin36 (Cx36). Electrical synapses in the TRN precede the postnatal development of TRN-to-VB inhibition. We investigated how the deletion of Cx36 affects the maturation of TRN and VB neurons, electrical coupling and GABAergic synapses by studying wild-type (WT) and Cx36 knockout (KO) mice. The incidence and strength of electrical coupling in TRN was sharply reduced, but not abolished, in KO mice. Surprisingly, electrical synapses between Cx36-KO neurons had faster voltage-dependent decay kinetics and conductance asymmetry (rectification) than did electrical synapses between WT neurons. The properties of TRN-mediated inhibition in VB also depended on the Cx36 genotype. Deletion of Cx36 increased the frequency and shifted the amplitude distributions of miniature IPSCs, whereas the paired-pulse ratio of evoked IPSCs was unaffected, suggesting that the absence of Cx36 led to an increase in GABAergic synaptic contacts. VB neurons from Cx36-KO mice also tended to have simpler dendritic trees and fewer divergent inputs from the TRN compared to WT cells. The findings obtained in the present study suggest that proper development of thalamic inhibitory circuitry, neuronal morphology, TRN cell function and electrical coupling requires Cx36. In the absence of Cx36, some TRN neurons express asymmetric electrical coupling mediated by other unidentified connexin subtypes.
Collapse
Affiliation(s)
- Timothy A Zolnik
- Department of Neuroscience, Division of Biology & Medicine, Brown University, Providence, RI, USA
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Barry W Connors
- Department of Neuroscience, Division of Biology & Medicine, Brown University, Providence, RI, USA
| |
Collapse
|
22
|
Abstract
Spontaneous activity patterns propagate through many parts of the developing nervous system and shape the wiring of emerging circuits. Prior to vision, waves of activity originating in the retina propagate through the lateral geniculate nucleus (LGN) of the thalamus to primary visual cortex (V1). Retinal waves have been shown to instruct the wiring of ganglion cell axons in LGN and of thalamocortical axons in V1 via correlation-based plasticity rules. Across species, retinal waves mature in three stereotypic stages (I-III), in which distinct circuit mechanisms give rise to unique activity patterns that serve specific functions in visual system refinement. Here, I review insights into the patterns, mechanisms, and functions of stage III retinal waves, which rely on glutamatergic signaling. As glutamatergic waves spread across the retina, neighboring ganglion cells with opposite light responses (ON vs. OFF) are activated sequentially. Recent studies identified lateral excitatory networks in the inner retina that generate and propagate glutamatergic waves, and vertical inhibitory networks that desynchronize the activity of ON and OFF cells in the wavefront. Stage III wave activity patterns may help segregate axons of ON and OFF ganglion cells in the LGN, and could contribute to the emergence of orientation selectivity in V1.
Collapse
Affiliation(s)
- Daniel Kerschensteiner
- Departments of Ophthalmology and Visual Sciences, Neuroscience, and Biomedical Engineering, Hope Center for Neurological Diseases, Washington University School of Medicine Saint Louis, MO, USA
| |
Collapse
|
23
|
Abstract
Early in development, before the onset of vision, the retina establishes direction-selective responses. During this time period, the retina spontaneously generates bursts of action potentials that propagate across its extent. The precise spatial and temporal properties of these "retinal waves" have been implicated in the formation of retinal projections to the brain. However, their role in the development of direction selective circuits within the retina has not yet been determined. We addressed this issue by combining multielectrode array and cell-attached recordings to examine mice that lack the CaV3.2 subunit of T-type Ca2+ channels (CaV3.2 KO) because these mice exhibit disrupted waves during the period that direction selective circuits are established. We found that the spontaneous activity of these mice displays wave-associated bursts of action potentials that are altered from that of control mice: the frequency of these bursts is significantly decreased and the firing rate within each burst is reduced. Moreover, the projection patterns of the retina demonstrate decreased eye-specific segregation in the dorsal lateral geniculate nucleus (dLGN). However, after eye-opening, the direction selective responses of CaV3.2 KO direction selective ganglion cells (DSGCs) are indistinguishable from those of wild-type DSGCs. Our data indicate that although the temporal properties of the action potential bursts associated with retinal waves are important for activity-dependent refining of retinal projections to central targets, they are not critical for establishing direction selectivity in the retina.
Collapse
|
24
|
Pinto MCX, Kihara AH, Goulart VAM, Tonelli FMP, Gomes KN, Ulrich H, Resende RR. Calcium signaling and cell proliferation. Cell Signal 2015; 27:2139-49. [PMID: 26275497 DOI: 10.1016/j.cellsig.2015.08.006] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/04/2015] [Accepted: 08/10/2015] [Indexed: 12/17/2022]
Abstract
Cell proliferation is orchestrated through diverse proteins related to calcium (Ca(2+)) signaling inside the cell. Cellular Ca(2+) influx that occurs first by various mechanisms at the plasma membrane, is then followed by absorption of Ca(2+) ions by mitochondria and endoplasmic reticulum, and, finally, there is a connection of calcium stores to the nucleus. Experimental evidence indicates that the fluctuation of Ca(2+) from the endoplasmic reticulum provides a pivotal and physiological role for cell proliferation. Ca(2+) depletion in the endoplasmatic reticulum triggers Ca(2+) influx across the plasma membrane in an phenomenon called store-operated calcium entries (SOCEs). SOCE is activated through a complex interplay between a Ca(2+) sensor, denominated STIM, localized in the endoplasmic reticulum and a Ca(2+) channel at the cell membrane, denominated Orai. The interplay between STIM and Orai proteins with cell membrane receptors and their role in cell proliferation is discussed in this review.
Collapse
Affiliation(s)
- Mauro Cunha Xavier Pinto
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Presyes 748, 05508-000 São Paulo, SP, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - Alexandre Hiroaki Kihara
- Universidade Federal do ABC, Centro de Matemática, Computação e Cognição, Rua Arcturus (Jd Antares), 09606-070, São Bernardo do Campo, SP, Brazil
| | - Vânia A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - Fernanda M P Tonelli
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - Katia N Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Presyes 748, 05508-000 São Paulo, SP, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Univtreersidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil.
| |
Collapse
|
25
|
Tao Y, Chen T, Liu B, Yang GQ, Peng G, Zhang H, Huang YF. The neurotoxic effects of N-methyl-N-nitrosourea on the electrophysiological property and visual signal transmission of rat's retina. Toxicol Appl Pharmacol 2015; 286:44-52. [DOI: 10.1016/j.taap.2015.03.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 03/10/2015] [Accepted: 03/10/2015] [Indexed: 10/23/2022]
|
26
|
Fern R. Ischemic tolerance in pre-myelinated white matter: the role of astrocyte glycogen in brain pathology. J Cereb Blood Flow Metab 2015; 35:951-8. [PMID: 25669910 PMCID: PMC4640254 DOI: 10.1038/jcbfm.2015.3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/15/2014] [Accepted: 12/17/2014] [Indexed: 11/09/2022]
Abstract
In isolated white matter, ischemic tolerance changes dramatically in the period immediately before the onset of myelination. In the absence of an extrinsic energy source, postnatal day 0 to 2 (P0 to P2) white matter axons are here shown to maintain excitability for over twice as long as axons >P2, a differential that was dependent on glycogen metabolism. Prolonged withdrawal of extrinsic energy supply tended to spare axons in zones around astrocytes, which are shown to be the sole repository for glycogen particles in developing white matter. Analysis of mitochondrial volume fraction revealed that neither axons nor astrocytes had a low metabolic rate in neonatal white matter, while oligodendroglia at older ages had an elevated metabolism. The astrocyte population is established early in neural development, and exhibits reduced cell density as maturation progresses and white matter expands. The findings show that this event establishes the necessary conditions for ischemia sensitivity in white matter and indicates that astrocyte proximity may be significant for the survival of neuronal elements in conditions associated with compromised energy supply.
Collapse
Affiliation(s)
- Robert Fern
- Peninsula School of Medicine and Dentistry, University of Plymouth, Plymouth, UK
| |
Collapse
|
27
|
Abstract
Spontaneous retinal activity mediated by glutamatergic neurotransmission-so-called "Stage 3" retinal waves-drives anti-correlated spiking in ON and OFF RGCs during the second week of postnatal development of the mouse. In the mature retina, the activity of a retinal interneuron called the AII amacrine cell is responsible for anti-correlated spiking in ON and OFF α-RGCs. In mature AIIs, membrane hyperpolarization elicits bursting behavior. Here, we postulated that bursting in AIIs underlies the initiation of glutamatergic retinal waves. We tested this hypothesis by using two-photon calcium imaging of spontaneous activity in populations of retinal neurons and by making whole-cell recordings from individual AIIs and α-RGCs in in vitro preparations of mouse retina. We found that AIIs participated in retinal waves, and that their activity was correlated with that of ON α-RGCs and anti-correlated with that of OFF α-RGCs. Though immature AIIs lacked the complement of membrane conductances necessary to generate bursting, pharmacological activation of the M-current, a conductance that modulates bursting in mature AIIs, blocked retinal wave generation. Interestingly, blockade of the pacemaker conductance Ih, a conductance absent in AIIs but present in both ON and OFF cone bipolar cells, caused a dramatic loss of spatial coherence of spontaneous activity. We conclude that during glutamatergic waves, AIIs act to coordinate and propagate activity generated by BCs rather than to initiate spontaneous activity.
Collapse
|
28
|
Detecting pairwise correlations in spike trains: an objective comparison of methods and application to the study of retinal waves. J Neurosci 2015; 34:14288-303. [PMID: 25339742 DOI: 10.1523/jneurosci.2767-14.2014] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Correlations in neuronal spike times are thought to be key to processing in many neural systems. Many measures have been proposed to summarize these correlations and of these the correlation index is widely used and is the standard in studies of spontaneous retinal activity. We show that this measure has two undesirable properties: it is unbounded above and confounded by firing rate. We list properties needed for a measure to fairly quantify and compare correlations and we propose a novel measure of correlation-the spike time tiling coefficient. This coefficient, the correlation index, and 33 other measures of correlation of spike times are blindly tested for the required properties on synthetic and experimental data. Based on this, we propose a measure (the spike time tiling coefficient) to replace the correlation index. To demonstrate the benefits of this measure, we reanalyze data from seven key studies, which previously used the correlation index to investigate the nature of spontaneous activity. We reanalyze data from β2(KO) and β2(TG) mutants, mutants lacking connexin isoforms, and also the age-dependent changes in wild-type and β2(KO) correlations. Reanalysis of the data using the proposed measure can significantly change the conclusions. It leads to better quantification of correlations and therefore better inference from the data. We hope that the proposed measure will have wide applications, and will help clarify the role of activity in retinotopic map formation.
Collapse
|
29
|
Nishii K, Shibata Y, Kobayashi Y. Connexin mutant embryonic stem cells and human diseases. World J Stem Cells 2014; 6:571-578. [PMID: 25426253 PMCID: PMC4178256 DOI: 10.4252/wjsc.v6.i5.571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 09/11/2014] [Accepted: 09/17/2014] [Indexed: 02/06/2023] Open
Abstract
Intercellular communication via gap junctions allows cells within multicellular organisms to share small molecules. The effect of such interactions has been elucidated using mouse gene knockout strategies. Although several mutations in human gap junction-encoding connexin (Cx) have been described, Cx mutants in mice do not always recapitulate the human disease. Among the 20 mouse Cxs, Cx26, Cx43, and Cx45 play roles in early cardiac or placental development, and disruption of the genes results in lethality that hampers further analyses. Embryonic stem cells (ESCs) that lack Cx43 or Cx45 have made analysis feasible in both in vitro differentiated cell cultures and in vivo chimeric tissues. The success of mouse ESCs studies is leading to the use of induced pluripotent stem cells to learn more about the pathogenesis of human Cx diseases. This review summarizes the current status of mouse Cx disruption models and ESC differentiation studies, and discusses their implication for understanding human Cx diseases.
Collapse
|
30
|
Burbridge TJ, Xu HP, Ackman JB, Ge X, Zhang Y, Ye MJ, Zhou ZJ, Xu J, Contractor A, Crair MC. Visual circuit development requires patterned activity mediated by retinal acetylcholine receptors. Neuron 2014; 84:1049-64. [PMID: 25466916 DOI: 10.1016/j.neuron.2014.10.051] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2014] [Indexed: 01/17/2023]
Abstract
The elaboration of nascent synaptic connections into highly ordered neural circuits is an integral feature of the developing vertebrate nervous system. In sensory systems, patterned spontaneous activity before the onset of sensation is thought to influence this process, but this conclusion remains controversial, largely due to the inherent difficulty recording neural activity in early development. Here, we describe genetic and pharmacological manipulations of spontaneous retinal activity, assayed in vivo, that demonstrate a causal link between retinal waves and visual circuit refinement. We also report a decoupling of downstream activity in retinorecipient regions of the developing brain after retinal wave disruption. Significantly, we show that the spatiotemporal characteristics of retinal waves affect the development of specific visual circuits. These results conclusively establish retinal waves as necessary and instructive for circuit refinement in the developing nervous system and reveal how neural circuits adjust to altered patterns of activity prior to experience.
Collapse
Affiliation(s)
- Timothy J Burbridge
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Hong-Ping Xu
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - James B Ackman
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Xinxin Ge
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yueyi Zhang
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Mei-Jun Ye
- Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Z Jimmy Zhou
- Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Jian Xu
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Anis Contractor
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Michael C Crair
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT 06510, USA; Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06510, USA; Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
31
|
Speer CM, Sun C, Liets LC, Stafford BK, Chapman B, Cheng HJ. Eye-specific retinogeniculate segregation proceeds normally following disruption of patterned spontaneous retinal activity. Neural Dev 2014; 9:25. [PMID: 25377639 PMCID: PMC4289266 DOI: 10.1186/1749-8104-9-25] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 10/20/2014] [Indexed: 11/10/2022] Open
Abstract
Background Spontaneous retinal activity (SRA) is important during eye-specific segregation within the dorsal lateral geniculate nucleus (dLGN), but the feature(s) of activity critical for retinogeniculate refinement are controversial. Pharmacologically or genetically manipulating cholinergic signaling during SRA perturbs correlated retinal ganglion cell (RGC) spiking and disrupts eye-specific retinofugal refinement in vivo, consistent with an instructive role for SRA during visual system development. Paradoxically, ablating the starburst amacrine cells (SACs) that generate cholinergic spontaneous activity disrupts correlated RGC firing without impacting retinal activity levels or eye-specific segregation in the dLGN. Such experiments suggest that patterned SRA during retinal waves is not critical for eye-specific refinement and instead, normal activity levels are permissive for retinogeniculate development. Here we revisit the effects of ablating the cholinergic network during eye-specific segregation and show that SAC ablation disrupts, but does not eliminate, retinal waves with no concomitant impact on normal eye-specific segregation in the dLGN. Results We induced SAC ablation in postnatal ferret pups beginning at birth by intraocular injection of a novel immunotoxin selective for the ferret vesicular acetylcholine transporter (Ferret VAChT-Sap). Through dual-patch whole-cell and multi-electrode array recording we found that SAC ablation altered SRA patterns and led to significantly smaller retinal waves compared with controls. Despite these defects, eye-specific segregation was normal. Further, interocular competition for target territory in the dLGN proceeded in cases where SAC ablation was asymmetric in the two eyes. Conclusions Our data demonstrate normal eye-specific retinogeniculate development despite significant abnormalities in patterned SRA. Comparing our current results with earlier studies suggests that defects in retinal wave size, absolute levels of SRA, correlations between RGC pairs, RGC burst frequency, high frequency RGC firing during bursts, and the number of spikes per RGC burst are each uncorrelated with abnormalities in eye-specific segregation in the dLGN. An increase in the fraction of asynchronous spikes occurring outside of bursts and waves correlates with eye-specific segregation defects in studies reported to date. These findings highlight the relative importance of different features of SRA while providing additional constraints for computational models of Hebbian plasticity mechanisms in the developing visual system. Electronic supplementary material The online version of this article (doi:10.1186/1749-8104-9-25) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | - Hwai-Jong Cheng
- Center for Neuroscience, University of California, Davis, 1544 Newton Court, Davis, CA 95618, USA.
| |
Collapse
|
32
|
Kovács-Öller T, Raics K, Orbán J, Nyitrai M, Völgyi B. Developmental changes in the expression level of connexin36 in the rat retina. Cell Tissue Res 2014; 358:289-302. [PMID: 25110193 DOI: 10.1007/s00441-014-1967-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 07/09/2014] [Indexed: 02/03/2023]
Abstract
Connexin36 (Cx36) is the major gap junction forming protein in the brain and the retina; thus, alterations in its expression indicate changes in the corresponding circuitry. Many structural changes occur in the early postnatal retina before functional neuronal circuits are finalized, including those that incorporate gap junctions. To reveal the time-lapse formation of inner retinal gap junctions, we examine the developing postnatal rat retina from birth (P0) to young adult age (P20) and follow the expression of Cx36 in the mRNA and protein levels. We found a continuous elevation in the expression of both the Cx36 transcript and protein between P0 and P20 and a somewhat delayed Cx36 plaque formation throughout the inner plexiform layer (IPL) starting at P10. By using tristratificated calretinin positive (CaR(+)) fibers in the IPL as a guide, we detected a clear preference of Cx36 plaques for the ON sublamina from the earliest time of detection. This distributional preference became more pronounced at P15 and P20 due to the emergence and widespread expression of large (>0.1 μm(2)) Cx36 plaques in the ON sublamina. Finally, we showed that parvalbumin-positive (PV(+)) AII amacrine cell dendrites colocalize with Cx36 plaques as early as P10 in strata 3 and 4, whereas colocalizations in stratum 5 became characteristic only around P20. We conclude that Cx36 expression in the rat IPL displays a characteristic succession of changes during retinogenesis reflecting the formation of the underlying electrical synaptic circuitry. In particular, AII cell gap junctions, first formed with ON cone bipolar cells and later with other AII amacrine cells, accounted for the observed Cx36 expressional changes.
Collapse
Affiliation(s)
- Tamás Kovács-Öller
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Ifjúság street 6, Hungary
| | | | | | | | | |
Collapse
|
33
|
Carlisle TC, Ribera AB. Connexin 35b expression in the spinal cord of Danio rerio embryos and larvae. J Comp Neurol 2014; 522:861-75. [PMID: 23939687 DOI: 10.1002/cne.23449] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 07/10/2013] [Accepted: 08/02/2013] [Indexed: 11/11/2022]
Abstract
Electrical synapses are expressed prominently in the developing and mature nervous systems. Unlike chemical synapses, little is known about the developmental role of electrical synapses, reflecting the limitations imposed by the lack of selective pharmacological blockers. At a molecular level, the building blocks of electrical synapses are connexin proteins. In this study, we report the expression pattern for neuronally expressed connexin 35b (cx35b), the zebrafish orthologue of mammalian connexin (Cx) 36. We find that cx35b is expressed at the time of neural induction, indicating a possible early role in neural progenitor cells. Additionally, cx35b localizes to the ventral spinal cord during embryonic and early larval stages. We detect cx35b mRNA in secondary motor neurons (SMNs) and interneurons. We identified the premotor circumferential descending (CiD) interneuron as one interneuron subtype expressing cx35b. In addition, cx35b is present in other ventral interneurons of unknown subtype(s). This early expression of cx35b in SMNs and CiDs suggests a possible role in motor network function during embryonic and larval stages.
Collapse
Affiliation(s)
- Tara C Carlisle
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045; Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045; Colorado Clinical and Translational Sciences Institute, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045; Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045
| | | |
Collapse
|
34
|
Akopian A, Atlasz T, Pan F, Wong S, Zhang Y, Völgyi B, Paul DL, Bloomfield SA. Gap junction-mediated death of retinal neurons is connexin and insult specific: a potential target for neuroprotection. J Neurosci 2014; 34:10582-91. [PMID: 25100592 PMCID: PMC4200109 DOI: 10.1523/jneurosci.1912-14.2014] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 06/06/2014] [Accepted: 06/27/2014] [Indexed: 01/02/2023] Open
Abstract
Secondary cell death via gap junctions (GJs) plays a role in the propagation of neuronal loss under a number of degenerative disorders. Here, we examined the role of GJs in neuronal death in the retina, which has arguably the most diverse expression of GJs in the CNS. Initially, we induced apoptotic death by injecting single retinal ganglion cells and glia with cytochrome C and found that this resulted in the loss of neighboring cells to which they were coupled via GJs. We next found that pharmacological blockade of GJs eradicated nearly all amacrine cell loss and reduced retinal ganglion cell loss by ∼70% after induction of either excitotoxic or ischemic insult conditions. These data indicate that the GJ-mediated secondary cell death was responsible for the death of most cells. Whereas genetic deletion of the GJ subunit Cx36 increased cell survivability by ∼50% under excitotoxic condition, cell loss in Cx45 knock-out mouse retinas was similar to that seen in wild-type mice. In contrast, ablation of Cx45 reduced neuronal loss by ∼50% under ischemic insult, but ablation of Cx36 offered no protection. Immunolabeling of the connexins showed differential changes in protein expression consistent with their differing roles in propagating death signals under the two insults. These data indicate that secondary cell death is mediated by different cohorts of GJs dependent on the connexins they express and the type of initial insult. Our results suggest that targeting specific connexins offers a novel therapeutic strategy to reduce progressive cell loss under different neurodegenerative conditions.
Collapse
Affiliation(s)
- Abram Akopian
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, New York 10036, Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016
| | - Tamas Atlasz
- Department of Ophthalmology, New York University School of Medicine, New York, New York 10016, and
| | - Feng Pan
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, New York 10036, Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016
| | - Sze Wong
- Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016
| | - Yi Zhang
- Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016
| | - Béla Völgyi
- Department of Ophthalmology, New York University School of Medicine, New York, New York 10016, and
| | - David L Paul
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115
| | - Stewart A Bloomfield
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, New York 10036, Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016, Department of Ophthalmology, New York University School of Medicine, New York, New York 10016, and
| |
Collapse
|
35
|
Eglen SJ, Weeks M, Jessop M, Simonotto J, Jackson T, Sernagor E. A data repository and analysis framework for spontaneous neural activity recordings in developing retina. Gigascience 2014; 3:3. [PMID: 24666584 PMCID: PMC4076503 DOI: 10.1186/2047-217x-3-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 03/13/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND During early development, neural circuits fire spontaneously, generating activity episodes with complex spatiotemporal patterns. Recordings of spontaneous activity have been made in many parts of the nervous system over the last 25 years, reporting developmental changes in activity patterns and the effects of various genetic perturbations. RESULTS We present a curated repository of multielectrode array recordings of spontaneous activity in developing mouse and ferret retina. The data have been annotated with minimal metadata and converted into HDF5. This paper describes the structure of the data, along with examples of reproducible research using these data files. We also demonstrate how these data can be analysed in the CARMEN workflow system. This article is written as a literate programming document; all programs and data described here are freely available. CONCLUSIONS 1. We hope this repository will lead to novel analysis of spontaneous activity recorded in different laboratories. 2. We encourage published data to be added to the repository. 3. This repository serves as an example of how multielectrode array recordings can be stored for long-term reuse.
Collapse
Affiliation(s)
- Stephen John Eglen
- Cambridge Computational Biology Institute, University of Cambridge, Wilberforce Road, CB3 0WA Cambridge, UK.
| | | | | | | | | | | |
Collapse
|
36
|
Meyrand P, Bem T. Variety of alternative stable phase-locking in networks of electrically coupled relaxation oscillators. PLoS One 2014; 9:e86572. [PMID: 24520321 PMCID: PMC3919711 DOI: 10.1371/journal.pone.0086572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/12/2013] [Indexed: 11/22/2022] Open
Abstract
We studied the dynamics of a large-scale model network comprised of oscillating electrically coupled neurons. Cells are modeled as relaxation oscillators with short duty cycle, so they can be considered either as models of pacemaker cells, spiking cells with fast regenerative and slow recovery variables or firing rate models of excitatory cells with synaptic depression or cellular adaptation. It was already shown that electrically coupled relaxation oscillators exhibit not only synchrony but also anti-phase behavior if electrical coupling is weak. We show that a much wider spectrum of spatiotemporal patterns of activity can emerge in a network of electrically coupled cells as a result of switching from synchrony, produced by short external signals of different spatial profiles. The variety of patterns increases with decreasing rate of neuronal firing (or duty cycle) and with decreasing strength of electrical coupling. We study also the effect of network topology - from all-to-all – to pure ring connectivity, where only the closest neighbors are coupled. We show that the ring topology promotes anti-phase behavior as compared to all-to-all coupling. It also gives rise to a hierarchical organization of activity: during each of the main phases of a given pattern cells fire in a particular sequence determined by the local connectivity. We have analyzed the behavior of the network using geometric phase plane methods and we give heuristic explanations of our findings. Our results show that complex spatiotemporal activity patterns can emerge due to the action of stochastic or sensory stimuli in neural networks without chemical synapses, where each cell is equally coupled to others via gap junctions. This suggests that in developing nervous systems where only electrical coupling is present such a mechanism can lead to the establishment of proto-networks generating premature multiphase oscillations whereas the subsequent emergence of chemical synapses would later stabilize generated patterns.
Collapse
Affiliation(s)
- Pierre Meyrand
- Univ. Bordeaux, IMN, UMR 5293, Bordeaux, France
- CNRS, IMN, UMR 5293, Bordeaux, France
- * E-mail:
| | - Tiaza Bem
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
37
|
Kirkby LA, Sack GS, Firl A, Feller MB. A role for correlated spontaneous activity in the assembly of neural circuits. Neuron 2014; 80:1129-44. [PMID: 24314725 DOI: 10.1016/j.neuron.2013.10.030] [Citation(s) in RCA: 217] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2013] [Indexed: 11/28/2022]
Abstract
Before the onset of sensory transduction, developing neural circuits spontaneously generate correlated activity in distinct spatial and temporal patterns. During this period of patterned activity, sensory maps develop and initial coarse connections are refined, which are critical steps in the establishment of adult neural circuits. Over the last decade, there has been substantial evidence that altering the pattern of spontaneous activity disrupts refinement, but the mechanistic understanding of this process remains incomplete. In this review, we discuss recent experimental and theoretical progress toward the process of activity-dependent refinement, focusing on circuits in the visual, auditory, and motor systems. Although many outstanding questions remain, the combination of several novel approaches has brought us closer to a comprehensive understanding of how complex neural circuits are established by patterned spontaneous activity during development.
Collapse
Affiliation(s)
- Lowry A Kirkby
- Biophysics Graduate Group, UC Berkeley, Berkeley, CA 94720, USA
| | | | | | | |
Collapse
|
38
|
Peiris H, Bonder CS, Coates PTH, Keating DJ, Jessup CF. The β-cell/EC axis: how do islet cells talk to each other? Diabetes 2014; 63:3-11. [PMID: 24357688 DOI: 10.2337/db13-0617] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Within the pancreatic islet, the β-cell represents the ultimate biosensor. Its central function is to accurately sense glucose levels in the blood and consequently release appropriate amounts of insulin. As the only cell type capable of insulin production, the β-cell must balance this crucial workload with self-preservation and, when required, regeneration. Evidence suggests that the β-cell has an important ally in intraislet endothelial cells (ECs). As well as providing a conduit for delivery of the primary input stimulus (glucose) and dissemination of its most important effector (insulin), intraislet blood vessels deliver oxygen to these dense clusters of metabolically active cells. Furthermore, it appears that ECs directly impact insulin gene expression and secretion and β-cell survival. This review discusses the molecules and pathways involved in the crosstalk between β-cells and intraislet ECs. The evidence supporting the intraislet EC as an important partner for β-cell function is examined to highlight the relevance of this axis in the context of type 1 and type 2 diabetes. Recent work that has established the potential of ECs or their progenitors to enhance the re-establishment of glycemic control following pancreatic islet transplantation in animal models is discussed.
Collapse
Affiliation(s)
- Heshan Peiris
- Department of Human Physiology, Centre for Neuroscience, Flinders University of South Australia, Adelaide, Australia
| | | | | | | | | |
Collapse
|
39
|
Toychiev AH, Yee CW, Sagdullaev BT. Correlated spontaneous activity persists in adult retina and is suppressed by inhibitory inputs. PLoS One 2013; 8:e77658. [PMID: 24204906 PMCID: PMC3812233 DOI: 10.1371/journal.pone.0077658] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 09/04/2013] [Indexed: 11/29/2022] Open
Abstract
Spontaneous rhythmic activity is a hallmark feature of the developing retina, where propagating retinal waves instruct axonal targeting and synapse formation. Retinal waves cease around the time of eye-opening; however, the fate of the underlying synaptic circuitry is unknown. Whether retinal waves are unique to the developing retina or if they can be induced in adulthood is not known. Combining patch-clamp techniques with calcium imaging, we demonstrate that propagative events persist in adult mouse retina when it is deprived of inhibitory input. This activity originates in bipolar cells, resembling glutamatergic stage III retinal waves. We find that, as it develops, the network interactions progressively curtail this activity. Together, this provides evidence that the correlated propagative neuronal activity can be induced in adult retina following the blockade of inhibitory interactions.
Collapse
Affiliation(s)
- Abduqodir H Toychiev
- Department of Neurology, Weill Medical College of Cornell University, New York, New York, United States of America ; Department of Ophthalmology, Weill Medical College of Cornell University, New York, New York, United States of America
| | | | | |
Collapse
|
40
|
Pang JJ, Paul DL, Wu SM. Survey on amacrine cells coupling to retrograde-identified ganglion cells in the mouse retina. Invest Ophthalmol Vis Sci 2013; 54:5151-62. [PMID: 23821205 DOI: 10.1167/iovs.13-11774] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Retinal amacrine cells (ACs) may make inhibitory chemical synapses and potentially excitatory gap junctions on ganglion cells (GCs). The total number and subtypes of ACs coupled to the entire GC population were investigated in wild-type and three lines of transgenic mice. METHODS GCs and GC-coupled ACs were identified by the previously established LY-NB (Lucifer yellow-Neurobiotin) retrograde double-labeling technique, in conjunction with specific antibodies and confocal microscopy. RESULTS GC-coupled ACs (NB-positive and LY-negative) comprised nearly 11% of displaced ACs and 4% of conventional ACs in wild-type mice, and were 9% and 4% of displaced ACs in Cx45(-/-) and Cx36/45(-/-) mice, respectively. Their somas were small in Cx36/45(-/-) mice, but variable in other strains. They were mostly γ-aminobutyric acid (GABA)-immunoreactive (IR) and located in the GC layer. They comprised only a small portion in the AC subpopulations, including GABA-IR, glycine-IR, calretinin-IR, 5-HT-accumulating, and ON-type choline acetyltransferase (ChAT) ACs in wild-type and ChAT transgenic mice (ChAT- tdTomato). In the distal 80% of the inner plexiform layer (IPL), dense GC dendrites coexisted with rich glycine-IR and GABA-IR. In the inner 20% of the IPL, sparse GC dendrites presented with a major GABA band and sparse glycine-IR. CONCLUSIONS Various subtypes of ACs may couple to GCs. ACs of the same immunoreactivity may either couple or not couple to GCs. Cx36 and Cx45 dominate GC-AC coupling except for small ACs. The overall potency of GC-AC coupling is moderate, especially in the proximal 20% of the IPL, where inhibitory chemical signals are dominated by GABA ACs.
Collapse
Affiliation(s)
- Ji-Jie Pang
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
41
|
Kirkby LA, Feller MB. Intrinsically photosensitive ganglion cells contribute to plasticity in retinal wave circuits. Proc Natl Acad Sci U S A 2013; 110:12090-5. [PMID: 23821744 PMCID: PMC3718101 DOI: 10.1073/pnas.1222150110] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Correlated spontaneous activity in the developing nervous system is robust to perturbations in the circuits that generate it, suggesting that mechanisms exist to ensure its maintenance. We examine this phenomenon in the developing retina, where blockade of cholinergic circuits that mediate retinal waves during the first postnatal week leads to the generation of "recovered" waves through a distinct, gap junction-mediated circuit. Unlike cholinergic waves, these recovered waves were modulated by dopaminergic and glutamatergic signaling, and required the presence of the gap junction protein connexin 36. Moreover, in contrast to cholinergic waves, recovered waves were stimulated by ambient light via activation of melanopsin-expressing intrinsically photosensitive retinal ganglion cells. The involvement of intrinsically photosensitive retinal ganglion cells in this reconfiguration of wave-generating circuits offers an avenue of retinal circuit plasticity during development that was previously unknown.
Collapse
Affiliation(s)
| | - Marla B. Feller
- Department of Molecular and Cell Biology and the Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720
| |
Collapse
|
42
|
Akrouh A, Kerschensteiner D. Intersecting circuits generate precisely patterned retinal waves. Neuron 2013; 79:322-34. [PMID: 23830830 DOI: 10.1016/j.neuron.2013.05.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2013] [Indexed: 11/29/2022]
Abstract
The developing retina generates spontaneous glutamatergic (stage III) waves of activity that sequentially recruit neighboring ganglion cells with opposite light responses (ON and OFF RGCs). This activity pattern is thought to help establish parallel ON and OFF pathways in downstream visual areas. The circuits that produce stage III waves and desynchronize ON and OFF RGC firing remain obscure. Using dual patch-clamp recordings, we find that ON and OFF RGCs receive sequential excitatory input from ON and OFF cone bipolar cells (CBCs), respectively. This input sequence is generated by crossover circuits, in which ON CBCs control glutamate release from OFF CBCs via diffusely stratified inhibitory amacrine cells. In addition, neighboring ON CBCs communicate directly and indirectly through lateral glutamatergic transmission and gap junctions, both of which are required for wave initiation and propagation. Thus, intersecting lateral excitatory and vertical inhibitory circuits give rise to precisely patterned stage III retinal waves.
Collapse
Affiliation(s)
- Alejandro Akrouh
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, Saint Louis, MO 63110, USA
| | | |
Collapse
|
43
|
Nelson CS, Fuller CK, Fordyce PM, Greninger AL, Li H, DeRisi JL. Microfluidic affinity and ChIP-seq analyses converge on a conserved FOXP2-binding motif in chimp and human, which enables the detection of evolutionarily novel targets. Nucleic Acids Res 2013; 41:5991-6004. [PMID: 23625967 PMCID: PMC3695516 DOI: 10.1093/nar/gkt259] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The transcription factor forkhead box P2 (FOXP2) is believed to be important in the evolution of human speech. A mutation in its DNA-binding domain causes severe speech impairment. Humans have acquired two coding changes relative to the conserved mammalian sequence. Despite intense interest in FOXP2, it has remained an open question whether the human protein’s DNA-binding specificity and chromatin localization are conserved. Previous in vitro and ChIP-chip studies have provided conflicting consensus sequences for the FOXP2-binding site. Using MITOMI 2.0 microfluidic affinity assays, we describe the binding site of FOXP2 and its affinity profile in base-specific detail for all substitutions of the strongest binding site. We find that human and chimp FOXP2 have similar binding sites that are distinct from previously suggested consensus binding sites. Additionally, through analysis of FOXP2 ChIP-seq data from cultured neurons, we find strong overrepresentation of a motif that matches our in vitro results and identifies a set of genes with FOXP2 binding sites. The FOXP2-binding sites tend to be conserved, yet we identified 38 instances of evolutionarily novel sites in humans. Combined, these data present a comprehensive portrait of FOXP2’s-binding properties and imply that although its sequence specificity has been conserved, some of its genomic binding sites are newly evolved.
Collapse
Affiliation(s)
- Christopher S Nelson
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94131, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Yamamoto N, López-Bendito G. Shaping brain connections through spontaneous neural activity. Eur J Neurosci 2012; 35:1595-604. [PMID: 22607005 DOI: 10.1111/j.1460-9568.2012.08101.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An overwhelming number of observations demonstrate that neural activity and genetic programs interact to specify the composition and organization of neural circuits during all stages of development. Spontaneous neuronal activities have been documented in several developing neural regions in both invertebrates and vertebrates, and their roles are mostly conserved among species. Among these roles, Ca(2+) spikes and levels of electrical activity have been shown to regulate neurite growth, axon extension and axon branching. Here, we review selected findings concerning the role of spontaneous activity on circuit development.
Collapse
Affiliation(s)
- Nobuhiko Yamamoto
- Laboratory of Cellular and Molecular Neurobiology, Graduate School of Frontier Biosciences, Osaka University, Yamadaoka, Suita, Osaka, Japan.
| | | |
Collapse
|
45
|
Paschon V, Higa GSV, Resende RR, Britto LRG, Kihara AH. Blocking of connexin-mediated communication promotes neuroprotection during acute degeneration induced by mechanical trauma. PLoS One 2012; 7:e45449. [PMID: 23029016 PMCID: PMC3447938 DOI: 10.1371/journal.pone.0045449] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 08/22/2012] [Indexed: 01/27/2023] Open
Abstract
Accruing evidence indicates that connexin (Cx) channels in the gap junctions (GJ) are involved in neurodegeneration after injury. However, studies using KO animal models endowed apparently contradictory results in relation to the role of coupling in neuroprotection. We analyzed the role of Cx-mediated communication in a focal lesion induced by mechanical trauma of the retina, a model that allows spatial and temporal definition of the lesion with high reproducibility, permitting visualization of the focus, penumbra and adjacent areas. Cx36 and Cx43 exhibited distinct gene expression and protein levels throughout the neurodegeneration progress. Cx36 was observed close to TUNEL-positive nuclei, revealing the presence of this protein surrounding apoptotic cells. The functional role of cell coupling was assessed employing GJ blockers and openers combined with lactate dehydrogenase (LDH) assay, a direct method for evaluating cell death/viability. Carbenoxolone (CBX), a broad-spectrum GJ blocker, reduced LDH release after 4 hours, whereas quinine, a Cx36-channel specific blocker, decreased LDH release as early as 1 hour after lesion. Furthermore, analysis of dying cell distribution confirmed that the use of GJ blockers reduced apoptosis spread. Accordingly, blockade of GJ communication during neurodegeneration with quinine, but not CBX, caused downregulation of initial and effector caspases. To summarize, we observed specific changes in Cx gene expression and protein distribution during the progress of retinal degeneration, indicating the participation of these elements in acute neurodegeneration processes. More importantly, our results revealed that direct control of GJ channels permeability may take part in reliable neuroprotection strategies aimed to rapid, fast treatment of mechanical trauma in the retina.
Collapse
Affiliation(s)
- Vera Paschon
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Santo André, São Paulo, Brazil
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Guilherme Shigueto Vilar Higa
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Santo André, São Paulo, Brazil
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Rodrigo Ribeiro Resende
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiz Roberto G. Britto
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Alexandre Hiroaki Kihara
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Santo André, São Paulo, Brazil
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
46
|
Eugenin EA, Basilio D, Sáez JC, Orellana JA, Raine CS, Bukauskas F, Bennett MVL, Berman JW. The role of gap junction channels during physiologic and pathologic conditions of the human central nervous system. J Neuroimmune Pharmacol 2012; 7:499-518. [PMID: 22438035 PMCID: PMC3638201 DOI: 10.1007/s11481-012-9352-5] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 02/28/2012] [Indexed: 12/15/2022]
Abstract
Gap junctions (GJs) are expressed in most cell types of the nervous system, including neuronal stem cells, neurons, astrocytes, oligodendrocytes, cells of the blood brain barrier (endothelial cells and astrocytes) and under inflammatory conditions in microglia/macrophages. GJs connect cells by the docking of two hemichannels, one from each cell with each hemichannel being formed by 6 proteins named connexins (Cx). Unapposed hemichannels (uHC) also can be open on the surface of the cells allowing the release of different intracellular factors to the extracellular space. GJs provide a mechanism of cell-to-cell communication between adjacent cells that enables the direct exchange of intracellular messengers, such as calcium, nucleotides, IP(3), and diverse metabolites, as well as electrical signals that ultimately coordinate tissue homeostasis, proliferation, differentiation, metabolism, cell survival and death. Despite their essential functions in physiological conditions, relatively little is known about the role of GJs and uHC in human diseases, especially within the nervous system. The focus of this review is to summarize recent findings related to the role of GJs and uHC in physiologic and pathologic conditions of the central nervous system.
Collapse
Affiliation(s)
- Eliseo A Eugenin
- Department of Pathology, F727, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Paul A, Cai Y, Atwal GS, Huang ZJ. Developmental Coordination of Gene Expression between Synaptic Partners During GABAergic Circuit Assembly in Cerebellar Cortex. Front Neural Circuits 2012; 6:37. [PMID: 22754500 PMCID: PMC3385560 DOI: 10.3389/fncir.2012.00037] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 06/01/2012] [Indexed: 01/14/2023] Open
Abstract
The assembly of neural circuits involves multiple sequential steps such as the specification of cell-types, their migration to proper brain locations, morphological and physiological differentiation, and the formation and maturation of synaptic connections. This intricate and often prolonged process is guided by elaborate genetic mechanisms that regulate each step. Evidence from numerous systems suggests that each cell-type, once specified, is endowed with a genetic program that unfolds in response to, and is regulated by, extrinsic signals, including cell–cell and synaptic interactions. To a large extent, the execution of this intrinsic program is achieved by the expression of specific sets of genes that support distinct developmental processes. Therefore, a comprehensive analysis of the developmental progression of gene expression in synaptic partners of neurons may provide a basis for exploring the genetic mechanisms regulating circuit assembly. Here we examined the developmental gene expression profiles of well-defined cell-types in a stereotyped microcircuit of the cerebellar cortex. We found that the transcriptomes of Purkinje cell and stellate/basket cells are highly dynamic throughout postnatal development. We revealed “phasic expression” of transcription factors, ion channels, receptors, cell adhesion molecules, gap junction proteins, and identified distinct molecular pathways that might contribute to sequential steps of cerebellar inhibitory circuit formation. We further revealed a correlation between genomic clustering and developmental co-expression of hundreds of transcripts, suggesting the involvement of chromatin level gene regulation during circuit formation.
Collapse
Affiliation(s)
- Anirban Paul
- Cold Spring Harbor Laboratory, Neuroscience Cold Spring Harbor, New York, NY, USA
| | | | | | | |
Collapse
|