251
|
Keeler AB, Molumby MJ, Weiner JA. Protocadherins branch out: Multiple roles in dendrite development. Cell Adh Migr 2015; 9:214-26. [PMID: 25869446 DOI: 10.1080/19336918.2014.1000069] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The proper formation of dendritic arbors is a critical step in neural circuit formation, and as such defects in arborization are associated with a variety of neurodevelopmental disorders. Among the best gene candidates are those encoding cell adhesion molecules, including members of the diverse cadherin superfamily characterized by distinctive, repeated adhesive domains in their extracellular regions. Protocadherins (Pcdhs) make up the largest group within this superfamily, encompassing over 80 genes, including the ∼60 genes of the α-, β-, and γ-Pcdh gene clusters and the non-clustered δ-Pcdh genes. An additional group includes the atypical cadherin genes encoding the giant Fat and Dachsous proteins and the 7-transmembrane cadherins. In this review we highlight the many roles that Pcdhs and atypical cadherins have been demonstrated to play in dendritogenesis, dendrite arborization, and dendritic spine regulation. Together, the published studies we discuss implicate these members of the cadherin superfamily as key regulators of dendrite development and function, and as potential therapeutic targets for future interventions in neurodevelopmental disorders.
Collapse
Key Words
- CNR, Cadherin related neuronal receptor
- CTCF, CCCTC-binding factor
- CaMKII, Ca2+/calmodulin-dependent protein kinase II.
- Celsr, Cadherin EGF LAG 7-pass G-type receptor 1
- DSCAM, Down syndrome cell adhesion molecule
- Dnmt3b, DNA (cytosine-5-)-methyltransferase 3 β
- Ds, Dachsous
- EC, extracellular cadherin
- EGF, Epidermal growth factor
- FAK, Focal adhesion kinase
- FMRP, Fragile X mental retardation protein
- Fj, Four jointed
- Fjx1, Four jointed box 1
- GPCR, G-protein-coupled receptor
- Gogo, Golden Goal
- LIM domain, Lin11, Isl-1 & Mec-3 domain
- MARCKS, Myristoylated alanine-rich C-kinase substrate
- MEF2, Myocyte enhancer factor 2
- MEK3, Mitogen-activated protein kinase kinase 3
- PCP, planar cell polarity
- PKC, Protein kinase C
- PSD, Post-synaptic density
- PYK2, Protein tyrosine kinase 2
- Pcdh
- Pcdh, Protocadherin
- RGC, Retinal ganglion cell
- RNAi, RNA interference
- Rac1, Ras-related C3 botulinum toxin substrate 1
- S2 cells, Schneider 2 cells
- SAC, starburst amacrine cell
- TAF1, Template-activating factor 1
- TAO2β, Thousand and one amino acid protein kinase 2 β
- TM, transmembrane
- arborization
- atypical cadherin
- branching
- cadherin superfamily
- cell adhesion
- da neuron, dendritic arborization neuron
- dendritic
- dendritic spine
- dendritogenesis
- fmi, Flamingo
- md neuron, multiple dendrite neuron
- neural circuit formation
- p38 MAPK, p38 mitogen-activated protein kinase
- self avoidance
- synaptogenesis
Collapse
Affiliation(s)
- Austin B Keeler
- a Department of Biology ; Neuroscience Graduate Program; University of Iowa ; Iowa City , IA USA
| | | | | |
Collapse
|
252
|
Abstract
During brain development, billions of neurons organize into highly specific circuits. To form specific circuits, neurons must build the appropriate types of synapses with appropriate types of synaptic partners while avoiding incorrect partners in a dense cellular environment. Defining the cellular and molecular rules that govern specific circuit formation has significant scientific and clinical relevance because fine scale connectivity defects are thought to underlie many cognitive and psychiatric disorders. Organizing specific neural circuits is an enormously complicated developmental process that requires the concerted action of many molecules, neural activity, and temporal events. This review focuses on one class of molecules postulated to play an important role in target selection and specific synapse formation: the classic cadherins. Cadherins have a well-established role in epithelial cell adhesion, and although it has long been appreciated that most cadherins are expressed in the brain, their role in synaptic specificity is just beginning to be unraveled. Here, we review past and present studies implicating cadherins as active participants in the formation, function, and dysfunction of specific neural circuits and pose some of the major remaining questions.
Collapse
Affiliation(s)
- Raunak Basu
- a Department of Neurobiology and Anatomy ; University of Utah ; Salt Lake City , UT USA
| | | | | |
Collapse
|
253
|
Soba P, Han C, Zheng Y, Perea D, Miguel-Aliaga I, Jan LY, Jan YN. The Ret receptor regulates sensory neuron dendrite growth and integrin mediated adhesion. eLife 2015; 4. [PMID: 25764303 PMCID: PMC4391025 DOI: 10.7554/elife.05491] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 03/11/2015] [Indexed: 12/11/2022] Open
Abstract
Neurons develop highly stereotyped receptive fields by coordinated growth of their dendrites. Although cell surface cues play a major role in this process, few dendrite specific signals have been identified to date. We conducted an in vivo RNAi screen in Drosophila class IV dendritic arborization (C4da) neurons and identified the conserved Ret receptor, known to play a role in axon guidance, as an important regulator of dendrite development. The loss of Ret results in severe dendrite defects due to loss of extracellular matrix adhesion, thus impairing growth within a 2D plane. We provide evidence that Ret interacts with integrins to regulate dendrite adhesion via rac1. In addition, Ret is required for dendrite stability and normal F-actin distribution suggesting it has an essential role in dendrite maintenance. We propose novel functions for Ret as a regulator in dendrite patterning and adhesion distinct from its role in axon guidance.
Collapse
Affiliation(s)
- Peter Soba
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf (UKE), University of Hamburg, Hamburg, Germany
| | - Chun Han
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Yi Zheng
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Daniel Perea
- Gut Signalling and Metabolism Group, MRC Clinical Sciences Centre, Imperial College London, London, United Kingdom
| | - Irene Miguel-Aliaga
- Gut Signalling and Metabolism Group, MRC Clinical Sciences Centre, Imperial College London, London, United Kingdom
| | - Lily Yeh Jan
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Yuh Nung Jan
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
254
|
Hayashi S, Takeichi M. Emerging roles of protocadherins: from self-avoidance to enhancement of motility. J Cell Sci 2015; 128:1455-64. [PMID: 25749861 DOI: 10.1242/jcs.166306] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Protocadherins are a group of transmembrane proteins belonging to the cadherin superfamily that are subgrouped into 'clustered' and 'non-clustered' protocadherins. Although cadherin superfamily members are known to regulate various forms of cell-cell interactions, including cell-cell adhesion, the functions of protocadherins have long been elusive. Recent studies are, however, uncovering their unique roles. The clustered protocadherins regulate neuronal survival, as well as dendrite self-avoidance. Combinatorial expression of clustered protocadherin isoforms creates a great diversity of adhesive specificity for cells, and this process is likely to underlie the dendritic self-avoidance. Non-clustered protocadherins promote cell motility rather than the stabilization of cell adhesion, unlike the classic cadherins, and mediate dynamic cellular processes, such as growth cone migration. Protocadherin dysfunction in humans is implicated in neurological disorders, such as epilepsy and mental retardation. This Commentary provides an overview of recent findings regarding protocadherin functions, as well as a discussion of the molecular basis underlying these functions.
Collapse
Affiliation(s)
- Shuichi Hayashi
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Masatoshi Takeichi
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| |
Collapse
|
255
|
Meguro R, Hishida R, Tsukano H, Yoshitake K, Imamura R, Tohmi M, Kitsukawa T, Hirabayashi T, Yagi T, Takebayashi H, Shibuki K. Impaired clustered protocadherin-α leads to aggregated retinogeniculate terminals and impaired visual acuity in mice. J Neurochem 2015; 133:66-72. [PMID: 25650227 DOI: 10.1111/jnc.13053] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 01/16/2015] [Accepted: 01/26/2015] [Indexed: 11/26/2022]
Abstract
Clustered protocadherins (cPcdhs) comprising cPcdh-α, -β, and -γ, encode a large family of cadherin-like cell-adhesion molecules specific to neurons. Impairment of cPcdh-α results in abnormal neuronal projection patterns in specific brain areas. To elucidate the role of cPcdh-α in retinogeniculate projections, we investigated the morphological patterns of retinogeniculate terminals in the lateral geniculate (LG) nucleus of mice with impaired cPcdh-α. We found huge aggregated retinogeniculate terminals in the dorsal LG nucleus, whereas no such aggregated terminals derived from the retina were observed in the olivary pretectal nucleus and the ventral LG nucleus. These aggregated terminals appeared between P10 and P14, just before eye opening and at the beginning of the refinement stage of the retinogeniculate projections. Reduced visual acuity was observed in adult mice with impaired cPcdh-α, whereas the orientation selectivity and direction selectivity of neurons in the primary visual cortex were apparently normal. These findings suggest that cPcdh-α is required for adequate spacing of retinogeniculate projections, which may be essential for normal development of visual acuity.
Collapse
Affiliation(s)
- Reiko Meguro
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
256
|
Missaire M, Hindges R. The role of cell adhesion molecules in visual circuit formation: from neurite outgrowth to maps and synaptic specificity. Dev Neurobiol 2015; 75:569-83. [PMID: 25649254 PMCID: PMC4855686 DOI: 10.1002/dneu.22267] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 01/08/2015] [Accepted: 01/09/2015] [Indexed: 11/08/2022]
Abstract
The formation of visual circuitry is a multistep process that involves cell–cell interactions based on a range of molecular mechanisms. The correct implementation of individual events, including axon outgrowth and guidance, the formation of the topographic map, or the synaptic targeting of specific cellular subtypes, are prerequisites for a fully functional visual system that is able to appropriately process the information captured by the eyes. Cell adhesion molecules (CAMs) with their adhesive properties and their high functional diversity have been identified as key actors in several of these fundamental processes. Because of their growth‐promoting properties, CAMs play an important role in neuritogenesis. Furthermore, they are necessary to control additional neurite development, regulating dendritic spacing and axon pathfinding. Finally, trans‐synaptic interactions of CAMs ensure cell type‐specific connectivity as a basis for the establishment of circuits processing distinct visual features. Recent discoveries implicating CAMs in novel mechanisms have led to a better general understanding of neural circuit formation, but also revealed an increasing complexity of their function. This review aims at describing the different levels of action for CAMs to shape neural connectivity, with a special focus on the visual system. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 75: 569–583, 2015
Collapse
Affiliation(s)
- Mégane Missaire
- MRC Centre for Developmental Neurobiology, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, United Kingdom
| | - Robert Hindges
- MRC Centre for Developmental Neurobiology, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, United Kingdom
| |
Collapse
|
257
|
Friedman LG, Benson DL, Huntley GW. Cadherin-based transsynaptic networks in establishing and modifying neural connectivity. Curr Top Dev Biol 2015; 112:415-65. [PMID: 25733148 DOI: 10.1016/bs.ctdb.2014.11.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It is tacitly understood that cell adhesion molecules (CAMs) are critically important for the development of cells, circuits, and synapses in the brain. What is less clear is what CAMs continue to contribute to brain structure and function after the early period of development. Here, we focus on the cadherin family of CAMs to first briefly recap their multidimensional roles in neural development and then to highlight emerging data showing that with maturity, cadherins become largely dispensible for maintaining neuronal and synaptic structure, instead displaying new and narrower roles at mature synapses where they critically regulate dynamic aspects of synaptic signaling, structural plasticity, and cognitive function. At mature synapses, cadherins are an integral component of multiprotein networks, modifying synaptic signaling, morphology, and plasticity through collaborative interactions with other CAM family members as well as a variety of neurotransmitter receptors, scaffolding proteins, and other effector molecules. Such recognition of the ever-evolving functions of synaptic cadherins may yield insight into the pathophysiology of brain disorders in which cadherins have been implicated and that manifest at different times of life.
Collapse
Affiliation(s)
- Lauren G Friedman
- Fishberg Department of Neuroscience, Friedman Brain Institute and the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Deanna L Benson
- Fishberg Department of Neuroscience, Friedman Brain Institute and the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - George W Huntley
- Fishberg Department of Neuroscience, Friedman Brain Institute and the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
258
|
Thu CA, Chen WV, Rubinstein R, Chevee M, Wolcott HN, Felsovalyi KO, Tapia JC, Shapiro L, Honig B, Maniatis T. Single-cell identity generated by combinatorial homophilic interactions between α, β, and γ protocadherins. Cell 2015; 158:1045-1059. [PMID: 25171406 DOI: 10.1016/j.cell.2014.07.012] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 06/18/2014] [Accepted: 07/07/2014] [Indexed: 02/07/2023]
Abstract
Individual mammalian neurons stochastically express distinct repertoires of α, β, and γ protocadherin (Pcdh) proteins, which function in neural circuit assembly. We report that all three subfamilies of clustered Pcdhs can engage in specific homophilic interactions, that cell surface delivery of Pcdhα isoforms requires cis interactions with other Pcdhs, and that the extracellular cadherin domain EC6 plays a critical role in this process. Examination of homophilic interactions between specific combinations of multiple Pcdh isoforms revealed that Pcdh combinatorial recognition specificities depend on the identity of all of the expressed isoforms. A single mismatched Pcdh isoform can interfere with these combinatorial homophilic interactions. A theoretical analysis reveals that assembly of Pcdh isoforms into multimeric recognition units and the observed tolerance for mismatched isoforms can generate cell surface diversity sufficient for single-cell identity. However, the competing demands of nonself discrimination and self-recognition place limitations on the mechanisms by which homophilic recognition units can function.
Collapse
Affiliation(s)
- Chan Aye Thu
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, 701 W 168th Street, New York, NY 10032, USA
| | - Weisheng V Chen
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, 701 W 168th Street, New York, NY 10032, USA
| | - Rotem Rubinstein
- Center for Computational Biology and Bioinformatics, Columbia University Medical Center, 1130 St. Nicholas Avenue, New York, NY 10032, USA; Department of System Biology, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA; Howard Hughes Medical Institute
| | - Maxime Chevee
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, 701 W 168th Street, New York, NY 10032, USA
| | - Holly N Wolcott
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, 1150 St. Nicholas Avenue, Russ Berrie Pavilion, New York, NY 10032, USA
| | - Klara O Felsovalyi
- Center for Computational Biology and Bioinformatics, Columbia University Medical Center, 1130 St. Nicholas Avenue, New York, NY 10032, USA; Department of System Biology, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA; Howard Hughes Medical Institute
| | - Juan Carlos Tapia
- Department of Neuroscience, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Lawrence Shapiro
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, 1150 St. Nicholas Avenue, Russ Berrie Pavilion, New York, NY 10032, USA; Department of System Biology, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Barry Honig
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, 1150 St. Nicholas Avenue, Russ Berrie Pavilion, New York, NY 10032, USA; Center for Computational Biology and Bioinformatics, Columbia University Medical Center, 1130 St. Nicholas Avenue, New York, NY 10032, USA; Department of System Biology, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA; Howard Hughes Medical Institute.
| | - Tom Maniatis
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, 701 W 168th Street, New York, NY 10032, USA.
| |
Collapse
|
259
|
Abstract
We describe recent progress toward defining neuronal cell types in the mouse retina and attempt to extract lessons that may be generally useful in the mammalian brain. Achieving a comprehensive catalog of retinal cell types now appears within reach, because researchers have achieved consensus concerning two fundamental challenges. The first is accuracy-defining pure cell types rather than settling for neuronal classes that are mixtures of types. The second is completeness-developing methods guaranteed to eventually identify all cell types, as well as criteria for determining when all types have been found. Case studies illustrate how these two challenges are handled by combining state-of-the-art molecular, anatomical, and physiological techniques. Progress is also being made in observing and modeling connectivity between cell types. Scaling up to larger brain regions, such as the cortex, will require not only technical advances but also careful consideration of the challenges of accuracy and completeness.
Collapse
|
260
|
Li JSS, Shin GJE, Millard SS. Neuronal cell-type-specific alternative splicing: A mechanism for specifying connections in the brain? NEUROGENESIS 2015; 2:e1122699. [PMID: 27606331 PMCID: PMC4973604 DOI: 10.1080/23262133.2015.1122699] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/16/2015] [Indexed: 02/05/2023]
Abstract
Alternative splicing (AS) allows a single gene to generate multiple protein isoforms. It has been hypothesized that AS plays a role in brain wiring by increasing the number of cell recognition molecules necessary for forming connections between neurons. Many studies have characterized isoform expression patterns of various genes in the brain, but very few have addressed whether specific isoforms play a functional role in neuronal wiring. In our recent work, we reported the cell-type-specific AS of the cell recognition molecule Dscam2. Exclusive expression of Dscam2 isoforms allows tightly associated neurons to signal repulsion selectively within the same cell-types, without interfering with one another. We show that preventing cell-specific isoform expression in 2 closely associated neurons disrupts their axon terminal morphology. We propose that the requirement for isoform specificity extends to synapses and discuss experiments that can test this directly. Factors that regulate Dscam2 cell-type-specific AS likely regulate the splicing of many genes involved in neurodevelopment. These regulators of alternative splicing may act broadly to control many genes involved in the development of specific neuron types. Identifying these factors is a key step in understanding how AS contributes to the brain connectome.
Collapse
Affiliation(s)
- Joshua Shing Shun Li
- School of Biomedical Sciences; The University of Queensland ; Brisbane, QLD, Australia
| | - Grace Ji-Eun Shin
- School of Biomedical Sciences; The University of Queensland ; Brisbane, QLD, Australia
| | - S Sean Millard
- School of Biomedical Sciences; The University of Queensland ; Brisbane, QLD, Australia
| |
Collapse
|
261
|
Dumas L, Heitz-Marchaland C, Fouquet S, Suter U, Livet J, Moreau-Fauvarque C, Chédotal A. Multicolor analysis of oligodendrocyte morphology, interactions, and development with Brainbow. Glia 2014; 63:699-717. [PMID: 25530205 DOI: 10.1002/glia.22779] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 12/08/2014] [Indexed: 11/12/2022]
Abstract
Oligodendrocytes are the myelinating cells of the central nervous system. Multiple markers are available to analyze the populations of oligodendroglial cells and their precursors during development and in pathological conditions. However, the behavior of oligodendrocytes remains poorly characterized in vivo, especially at the level of individual cells. Studying this aspect has been impaired so far by the lack of suitable methods for visualizing single oligodendrocytes, their processes, and their interactions during myelination. Here, we have used multicolor labeling technology to single-out simultaneously many individual oligodendrocytes in the postnatal mouse optic nerve. This method is based on Brainbow, a transgenic system for stochastic expression of multiple fluorescent protein genes through Cre-lox recombination, previously used for visualizing axons and neurons. We used tamoxifen-inducible recombination in myelinating cells of Brainbow transgenic mice to obtain multicolor labeling of oligodendrocytes. We show that the palette of colors expressed by labeled oligodendrocytes is tamoxifen dependent, with the highest doses producing the densest and most colorful labeling. At low doses of tamoxifen, the morphology of single or small clusters of fluorescent oligodendrocytes can be studied during postnatal development and in adult. Internodes are labeled to their extremities, revealing nodes of Ranvier. The new mouse model presented here opens new possibilities to explore the organization and development of the oligodendrocyte network with single-cell resolution.
Collapse
Affiliation(s)
- Laura Dumas
- INSERM, UMRS_U968, Institut de la Vision, Paris, F-75012, France; Sorbonne Universités, UPMC Univ Paris 06, Institut de la Vision, Paris, F-75012, France; CNRS, UMR_7210, Paris, F-75012, France
| | | | | | | | | | | | | |
Collapse
|
262
|
Duan X, Krishnaswamy A, De la Huerta I, Sanes JR. Type II cadherins guide assembly of a direction-selective retinal circuit. Cell 2014; 158:793-807. [PMID: 25126785 DOI: 10.1016/j.cell.2014.06.047] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 05/10/2014] [Accepted: 06/20/2014] [Indexed: 01/02/2023]
Abstract
Complex retinal circuits process visual information and deliver it to the brain. Few molecular determinants of synaptic specificity in this system are known. Using genetic and optogenetic methods, we identified two types of bipolar interneurons that convey visual input from photoreceptors to a circuit that computes the direction in which objects are moving. We then sought recognition molecules that promote selective connections of these cells with previously characterized components of the circuit. We found that the type II cadherins, cdh8 and cdh9, are each expressed selectively by one of the two bipolar cell types. Using loss- and gain-of-function methods, we showed that they are critical determinants of connectivity in this circuit and that perturbation of their expression leads to distinct defects in visually evoked responses. Our results reveal cellular components of a retinal circuit and demonstrate roles of type II cadherins in synaptic choice and circuit function.
Collapse
Affiliation(s)
- Xin Duan
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Arjun Krishnaswamy
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Irina De la Huerta
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joshua R Sanes
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
263
|
Özlü N, Qureshi MH, Toyoda Y, Renard BY, Mollaoglu G, Özkan NE, Bulbul S, Poser I, Timm W, Hyman AA, Mitchison TJ, Steen JA. Quantitative comparison of a human cancer cell surface proteome between interphase and mitosis. EMBO J 2014; 34:251-65. [PMID: 25476450 DOI: 10.15252/embj.201385162] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The cell surface is the cellular compartment responsible for communication with the environment. The interior of mammalian cells undergoes dramatic reorganization when cells enter mitosis. These changes are triggered by activation of the CDK1 kinase and have been studied extensively. In contrast, very little is known of the cell surface changes during cell division. We undertook a quantitative proteomic comparison of cell surface-exposed proteins in human cancer cells that were tightly synchronized in mitosis or interphase. Six hundred and twenty-eight surface and surface-associated proteins in HeLa cells were identified; of these, 27 were significantly enriched at the cell surface in mitosis and 37 in interphase. Using imaging techniques, we confirmed the mitosis-selective cell surface localization of protocadherin PCDH7, a member of a family with anti-adhesive roles in embryos. We show that PCDH7 is required for development of full mitotic rounding pressure at the onset of mitosis. Our analysis provided basic information on how cell cycle progression affects the cell surface. It also provides potential pharmacodynamic biomarkers for anti-mitotic cancer chemotherapy.
Collapse
Affiliation(s)
- Nurhan Özlü
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey Proteomics Center at Children's Hospital Boston, Boston, MA, USA Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Mohammad H Qureshi
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Yusuke Toyoda
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | - Bernhard Y Renard
- Research Group Bioinformatics (NG 4), Robert Koch-Institute, Berlin, Germany
| | - Gürkan Mollaoglu
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Nazlı E Özkan
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Selda Bulbul
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Ina Poser
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | - Wiebke Timm
- Proteomics Center at Children's Hospital Boston, Boston, MA, USA
| | - Anthony A Hyman
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Judith A Steen
- Proteomics Center at Children's Hospital Boston, Boston, MA, USA Department of Neurobiology, Harvard Medical School and Children's Hospital Boston, Boston, MA, USA
| |
Collapse
|
264
|
Wernet MF, Huberman AD, Desplan C. So many pieces, one puzzle: cell type specification and visual circuitry in flies and mice. Genes Dev 2014; 28:2565-84. [PMID: 25452270 PMCID: PMC4248288 DOI: 10.1101/gad.248245.114] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The visual system is a powerful model for probing the development, connectivity, and function of neural circuits. Two genetically tractable species, mice and flies, are together providing a great deal of understanding of these processes. Current efforts focus on integrating knowledge gained from three cross-fostering fields of research: (1) understanding how the fates of different cell types are specified during development, (2) revealing the synaptic connections between identified cell types ("connectomics") by high-resolution three-dimensional circuit anatomy, and (3) causal testing of how identified circuit elements contribute to visual perception and behavior. Here we discuss representative examples from fly and mouse models to illustrate the ongoing success of this tripartite strategy, focusing on the ways it is enhancing our understanding of visual processing and other sensory systems.
Collapse
Affiliation(s)
- Mathias F Wernet
- Department of Neurobiology, Stanford University, Stanford, California 94305, USA; New York University Abu Dhabi, Saadiyat Island, Abu Dhabi 129188, United Arab Emirates; Department of Biology, New York University, New York, New York 10003, USA
| | - Andrew D Huberman
- Department of Neurosciences, Neurobiology Section, Division of Biological Sciences, University of California at San Diego, La Jolla, California 92093, USA
| | - Claude Desplan
- New York University Abu Dhabi, Saadiyat Island, Abu Dhabi 129188, United Arab Emirates; Department of Biology, New York University, New York, New York 10003, USA
| |
Collapse
|
265
|
Abstract
The recent descriptions of widespread random monoallelic expression (RMAE) of genes distributed throughout the autosomal genome indicate that there are more genes subject to RMAE on autosomes than the number of genes on the X chromosome where X-inactivation dictates RMAE of X-linked genes. Several of the autosomal genes that undergo RMAE have independently been implicated in human Mendelian disorders. Thus, parsing the relationship between allele-specific expression of these genes and disease is of interest. Mutations in the human forkhead box P2 gene, FOXP2, cause developmental verbal dyspraxia with profound speech and language deficits. Here, we show that the human FOXP2 gene undergoes RMAE. Studying an individual with developmental verbal dyspraxia, we identify a deletion 3 Mb away from the FOXP2 gene, which impacts FOXP2 gene expression in cis. Together these data suggest the intriguing possibility that RMAE impacts the haploinsufficiency phenotypes observed for FOXP2 mutations.
Collapse
|
266
|
Izuta Y, Taira T, Asayama A, Machigashira M, Kinoshita T, Fujiwara M, Suzuki ST. Protocadherin-9 involvement in retinal development in Xenopus laevis. J Biochem 2014; 157:235-49. [PMID: 25414271 DOI: 10.1093/jb/mvu070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Biological roles of most protocadherins (Pcdhs) are a largely unsolved problem. Therefore, we cloned cDNA for Xenopus laevis protocadherin-9 and characterized its properties to elucidate the role. The deduced amino acid sequence was highly homologous to those of mammalian protocadherin-9 s. X. laevis protocadherin-9 expressed from the cDNA in L cells showed basic properties similar to those of mammalian Pcdhs. Expression of X. laevis protocadherin-9 was first detected in stage-31 embryos and increased as the development proceeded. In the later stage embryos and the adults, the retina strongly expressed protocadherin-9, which was mainly localized at the plexiform layers. Injection of morpholino anti-sense oligonucleotide against protocadherin-9 into the fertilized eggs inhibited eye development; and eye growth and formation of the retinal laminar structure were hindered. Moreover, affected retina showed abnormal extension of neurites into the ganglion cell layer. Co-injection of protocadherin-9 mRNA with the morpholino anti-sense oligonucleotide rescued the embryos from the defects. These results suggest that X. laevis protocadherin-9 was involved in the development of retina structure possibly through survival of neurons, formation of the lamina structure and neurite localization.
Collapse
Affiliation(s)
- Yusuke Izuta
- Department of Bioscience, School of Science and Technology, Kwansei Gakuin University, 2-1 Gakuen, Sanda-Shi, Hyogo-Ken 669-1337, Japan and Rikkyo College of Science, Rikkyo University, 3-34-1 Nishishinjyuku, Toshima-ku, Tokyo 171-8501, Japan
| | - Tetsuro Taira
- Department of Bioscience, School of Science and Technology, Kwansei Gakuin University, 2-1 Gakuen, Sanda-Shi, Hyogo-Ken 669-1337, Japan and Rikkyo College of Science, Rikkyo University, 3-34-1 Nishishinjyuku, Toshima-ku, Tokyo 171-8501, Japan
| | - Ayako Asayama
- Department of Bioscience, School of Science and Technology, Kwansei Gakuin University, 2-1 Gakuen, Sanda-Shi, Hyogo-Ken 669-1337, Japan and Rikkyo College of Science, Rikkyo University, 3-34-1 Nishishinjyuku, Toshima-ku, Tokyo 171-8501, Japan
| | - Mika Machigashira
- Department of Bioscience, School of Science and Technology, Kwansei Gakuin University, 2-1 Gakuen, Sanda-Shi, Hyogo-Ken 669-1337, Japan and Rikkyo College of Science, Rikkyo University, 3-34-1 Nishishinjyuku, Toshima-ku, Tokyo 171-8501, Japan
| | - Tsutomu Kinoshita
- Department of Bioscience, School of Science and Technology, Kwansei Gakuin University, 2-1 Gakuen, Sanda-Shi, Hyogo-Ken 669-1337, Japan and Rikkyo College of Science, Rikkyo University, 3-34-1 Nishishinjyuku, Toshima-ku, Tokyo 171-8501, Japan
| | - Miwako Fujiwara
- Department of Bioscience, School of Science and Technology, Kwansei Gakuin University, 2-1 Gakuen, Sanda-Shi, Hyogo-Ken 669-1337, Japan and Rikkyo College of Science, Rikkyo University, 3-34-1 Nishishinjyuku, Toshima-ku, Tokyo 171-8501, Japan
| | - Shintaro T Suzuki
- Department of Bioscience, School of Science and Technology, Kwansei Gakuin University, 2-1 Gakuen, Sanda-Shi, Hyogo-Ken 669-1337, Japan and Rikkyo College of Science, Rikkyo University, 3-34-1 Nishishinjyuku, Toshima-ku, Tokyo 171-8501, Japan
| |
Collapse
|
267
|
Mondal A, Black B, Kim YT, Mohanty S. Loop formation and self-fasciculation of cortical axon using photonic guidance at long working distance. Sci Rep 2014; 4:6902. [PMID: 25376602 PMCID: PMC4223660 DOI: 10.1038/srep06902] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 10/10/2014] [Indexed: 11/14/2022] Open
Abstract
The accuracy of axonal pathfinding and the formation of functional neural circuitry are crucial for an organism to process, store, and retrieve information from internal networks as well as from the environment. Aberrations in axonal migration is believed to lead to loop formation and self-fasciculation, which can lead to highly dysfunctional neural circuitry and therefore self-avoidance of axons is proposed to be the regulatory mechanism for control of synaptogenesis. Here, we report the application of a newly developed non-contact optical method using a weakly-focused, near infrared laser beam for highly efficient axonal guidance, and demonstrate the formation of axonal loops in cortical neurons, which demonstrate that cortical neurons can self-fasciculate in contrast to self-avoidance. The ability of light for axonal nano-loop formation opens up new avenues for the construction of complex neural circuitry, and non-invasive guidance of neurons at long working distances for restoration of impaired neural connections and functions.
Collapse
Affiliation(s)
- Argha Mondal
- Biophysics and Physiology Lab, Department of Physics
| | - Bryan Black
- Biophysics and Physiology Lab, Department of Physics
| | - Young-tae Kim
- Department of Bioengineering, The University of Texas at Arlington, TX 76019
| | | |
Collapse
|
268
|
Lyons DB, Magklara A, Goh T, Sampath SC, Schaefer A, Schotta G, Lomvardas S. Heterochromatin-mediated gene silencing facilitates the diversification of olfactory neurons. Cell Rep 2014; 9:884-92. [PMID: 25437545 PMCID: PMC4251488 DOI: 10.1016/j.celrep.2014.10.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 08/29/2014] [Accepted: 09/27/2014] [Indexed: 01/26/2023] Open
Abstract
An astounding property of the nervous system is its cellular diversity. This diversity, which was initially realized by morphological and electrophysiological differences, is ultimately produced by variations in gene-expression programs. In most cases, these variations are determined by external cues. However, a growing number of neuronal types have been identified in which inductive signals cannot explain the few but decisive transcriptional differences that cause cell diversification. Here, we show that heterochromatic silencing, which we find is governed by histone methyltransferases G9a (KMT1C) and GLP (KMT1D), is essential for stochastic and singular olfactory receptor (OR) expression. Deletion of G9a and GLP dramatically reduces the complexity of the OR transcriptome, resulting in transcriptional domination by a few ORs and loss of singularity in OR expression. Thus, our data suggest that, in addition to its previously known functions, heterochromatin creates an epigenetic platform that affords stochastic, mutually exclusive gene choices and promotes cellular diversity.
Collapse
Affiliation(s)
- David B Lyons
- Tetrad Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Angeliki Magklara
- Division of Biomedical Research, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 45110 Ioannina, Greece
| | - Tracie Goh
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94920, USA
| | - Srihari C Sampath
- Laboratory of Immune Cell Epigenetics and Signaling, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Anne Schaefer
- Laboratory of Immune Cell Epigenetics and Signaling, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Gunnar Schotta
- Munich Center for Integrated Protein Science, Adolf-Butenandt-Institute, Ludwig Maximilian University, Schillerstrasse 44, 80336 Munich, Germany
| | - Stavros Lomvardas
- Tetrad Program, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA 94920, USA.
| |
Collapse
|
269
|
Abstract
The complex, branched morphology of dendrites is a cardinal feature of neurons and has been used as a criterion for cell type identification since the beginning of neurobiology. Regulated dendritic outgrowth and branching during development form the basis of receptive fields for neurons and are essential for the wiring of the nervous system. The cellular and molecular mechanisms of dendritic morphogenesis have been an intensely studied area. In this review, we summarize the major experimental systems that have contributed to our understandings of dendritic development as well as the intrinsic and extrinsic mechanisms that instruct the neurons to form cell type-specific dendritic arbors.
Collapse
|
270
|
Roles for DSCAM and DSCAML1 in central nervous system development and disease. ADVANCES IN NEUROBIOLOGY 2014; 8:249-70. [PMID: 25300140 DOI: 10.1007/978-1-4614-8090-7_11] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
DSCAMs (Down syndrome cell adhesion molecules) are a group of immunoglobulin-like transmembrane proteins that contain fibronectin III domains. The founding member of the family was isolated in a positional cloning study that sought to identify genes located on chromosome 21 at the locus 21q22.2-q22.3 that is implicated in the neurological and cardiac phenotypes associated with Down's syndrome. In Drosophila, Dscam proteins are involved in neuronal wiring, while in vertebrates, the role of these cell adhesion molecules in neurogenesis, dendritogenesis, axonal outgrowth, synaptogenesis, and synaptic plasticity is only just beginning to be understood. In this chapter, we will review the functions ascribed to the two paralogous proteins found in humans, DSCAM and DSCAML1 (DSCAM-like 1), based on findings in knockout mice. The signaling pathways downstream of DSCAM activation and the role of DSCAM miss-expression in disease will be also discussed, particularly with regard to the intellectual disability in Down's syndrome.
Collapse
|
271
|
Sox2 regulates cholinergic amacrine cell positioning and dendritic stratification in the retina. J Neurosci 2014; 34:10109-21. [PMID: 25057212 DOI: 10.1523/jneurosci.0415-14.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The retina contains two populations of cholinergic amacrine cells, one positioned in the ganglion cell layer (GCL) and the other in the inner nuclear layer (INL), that together comprise ∼1/2 of a percent of all retinal neurons. The present study examined the genetic control of cholinergic amacrine cell number and distribution between these two layers. The total number of cholinergic amacrine cells was quantified in the C57BL/6J and A/J inbred mouse strains, and in 25 recombinant inbred strains derived from them, and variations in their number and ratio (GCL/INL) across these strains were mapped to genomic loci. The total cholinergic amacrine cell number was found to vary across the strains, from 27,000 to 40,000 cells, despite little variation within individual strains. The number of cells was always lower within the GCL relative to the INL, and the sizes of the two populations were strongly correlated, yet there was variation in their ratio between the strains. Approximately 1/3 of that variation in cell ratio was mapped to a locus on chromosome 3, where Sex determining region Y box 2 (Sox2) was identified as a candidate gene due to the presence of a 6-nucleotide insertion in the protein-coding sequence in C57BL/6J and because of robust and selective expression in cholinergic amacrine cells. Conditionally deleting Sox2 from the population of nascent cholinergic amacrine cells perturbed the normal ratio of cells situated in the GCL versus the INL and induced a bistratifying morphology, with dendrites distributed to both ON and OFF strata within the inner plexiform layer.
Collapse
|
272
|
Friedman LG, Riemslagh FW, Sullivan JM, Mesias R, Williams FM, Huntley GW, Benson DL. Cadherin-8 expression, synaptic localization, and molecular control of neuronal form in prefrontal corticostriatal circuits. J Comp Neurol 2014; 523:75-92. [PMID: 25158904 DOI: 10.1002/cne.23666] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/25/2014] [Accepted: 08/25/2014] [Indexed: 02/02/2023]
Abstract
Neocortical interactions with the dorsal striatum support many motor and executive functions, and such underlying functional networks are particularly vulnerable to a variety of developmental, neurological, and psychiatric brain disorders, including autism spectrum disorders, Parkinson's disease, and Huntington's disease. Relatively little is known about the development of functional corticostriatal interactions, and in particular, virtually nothing is known of the molecular mechanisms that control generation of prefrontal cortex-striatal circuits. Here, we used regional and cellular in situ hybridization techniques coupled with neuronal tract tracing to show that Cadherin-8 (Cdh8), a homophilic adhesion protein encoded by a gene associated with autism spectrum disorders and learning disability susceptibility, is enriched within striatal projection neurons in the medial prefrontal cortex and in striatal medium spiny neurons forming the direct or indirect pathways. Developmental analysis of quantitative real-time polymerase chain reaction and western blot data show that Cdh8 expression peaks in the prefrontal cortex and striatum at P10, when cortical projections start to form synapses in the striatum. High-resolution immunoelectron microscopy shows that Cdh8 is concentrated at excitatory synapses in the dorsal striatum, and Cdh8 knockdown in cortical neurons impairs dendritic arborization and dendrite self-avoidance. Taken together, our findings indicate that Cdh8 delineates developing corticostriatal circuits where it is a strong candidate for regulating the generation of normal cortical projections, neuronal morphology, and corticostriatal synapses.
Collapse
Affiliation(s)
- Lauren G Friedman
- Fishberg Department of Neuroscience, Friedman Brain Institute and The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029
| | | | | | | | | | | | | |
Collapse
|
273
|
Hayashi S, Inoue Y, Kiyonari H, Abe T, Misaki K, Moriguchi H, Tanaka Y, Takeichi M. Protocadherin-17 mediates collective axon extension by recruiting actin regulator complexes to interaxonal contacts. Dev Cell 2014; 30:673-87. [PMID: 25199687 DOI: 10.1016/j.devcel.2014.07.015] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 06/07/2014] [Accepted: 07/16/2014] [Indexed: 11/17/2022]
Abstract
In the process of neuronal wiring, axons derived from the same functional group typically extend together, resulting in fascicle formation. How these axons communicate with one another remains largely unknown. Here, we show that protocadherin-17 (Pcdh17) supports this group extension by recruiting actin polymerization regulators to interaxonal contact sites. Pcdh17 is expressed by a subset of amygdala neurons, and it accumulates at axon-axon boundaries because of homophilic binding. Pcdh17 knockout in mice suppressed the extension of these axons. Ectopically expressed Pcdh17 altered the pattern of axon extension. In in-vitro cultures, wild-type growth cones normally migrate along other axons, whereas Pcdh17 null growth cones do not. Pcdh17 recruits the WAVE complex, Lamellipodin, and Ena/VASP to cell-cell contacts, converting these sites into motile structures. We propose that, through these mechanisms, Pcdh17 maintains the migration of growth cones that are in contact with other axons, thereby supporting their collective extension.
Collapse
Affiliation(s)
- Shuichi Hayashi
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Yoko Inoue
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Kazuyo Misaki
- Laboratory for Electron Microscope, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Hiroyuki Moriguchi
- Laboratory for Integrated Biodevice, RIKEN Quantitative Biology Center, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Yo Tanaka
- Laboratory for Integrated Biodevice, RIKEN Quantitative Biology Center, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Masatoshi Takeichi
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan.
| |
Collapse
|
274
|
Expansion of stochastic expression repertoire by tandem duplication in mouse Protocadherin-α cluster. Sci Rep 2014; 4:6263. [PMID: 25179445 PMCID: PMC4151104 DOI: 10.1038/srep06263] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 08/13/2014] [Indexed: 11/08/2022] Open
Abstract
Tandem duplications are concentrated within the Pcdh cluster throughout vertebrate evolution and as copy number variations (CNVs) in human populations, but the effects of tandem duplication in the Pcdh cluster remain elusive. To investigate the effects of tandem duplication in the Pcdh cluster, here we generated and analyzed a new line of the Pcdh cluster mutant mice. In the mutant allele, a 218-kb region containing the Pcdh-α2 to Pcdh-αc2 variable exons with their promoters was duplicated and the individual duplicated Pcdh isoforms can be disctinguished. The individual duplicated Pcdh-α isoforms showed diverse expression level with stochastic expression manner, even though those have an identical promoter sequence. Interestingly, the 5'-located duplicated Pcdh-αc2, which is constitutively expressed in the wild-type brain, shifted to stochastic expression accompanied by increased DNA methylation. These results demonstrate that tandem duplication in the Pcdh cluster expands the stochastic expression repertoire irrespective of sequence divergence.
Collapse
|
275
|
Developmental localization of adhesion and scaffolding proteins at the cone synapse. Gene Expr Patterns 2014; 16:36-50. [PMID: 25176525 DOI: 10.1016/j.gep.2014.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/30/2014] [Accepted: 07/07/2014] [Indexed: 11/22/2022]
Abstract
The cone synapse is a complex signaling hub composed of the cone photoreceptor terminal and the dendrites of bipolar and horizontal cells converging around multiple ribbon synapses. Factors that promote organization of this structure are largely unexplored. In this study we characterize the localization of adhesion and scaffolding proteins that are localized to the cone synapse, including alpha-n-catenin, beta-catenin, gamma-protocadherin, cadherin-8, MAGI2 and CASK. We describe the localization of these proteins during development of the mouse retina and in the adult macaque retina and find that these proteins are concentrated at the cone synapse. The localization of these proteins was then characterized at the cellular and subcellular levels. Alpha-n-catenin, gamma-protocadherin and cadherin-8 were concentrated in the dendrites of bipolar cells that project to the cone synapse but were not detected or stained very dimly in the dendrites of cells projecting to rod synapses. This study adds to our knowledge of cone synapse development by characterizing the developmental localization of these factors and identifies these factors as candidates for functional analysis of cone synapse formation.
Collapse
|
276
|
Regulation of the protocadherin Celsr3 gene and its role in globus pallidus development and connectivity. Mol Cell Biol 2014; 34:3895-910. [PMID: 25113559 DOI: 10.1128/mcb.00760-14] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The globus pallidus (GP) is a central component of basal ganglia whose malfunctions cause a variety of neuropsychiatric disorders as well as cognitive impairments in neurodegenerative diseases such as Parkinson's disease. Here we report that the protocadherin gene Celsr3 is regulated by the insulator CCCTC-binding factor (CTCF) and the repressor neuron-restrictive silencer factor (NRSF, also known as REST) and is required for the development and connectivity of GP. Specifically, CTCF/cohesin and NRSF inhibit the expression of Celsr3 through specific binding to its promoter. In addition, we found that the Celsr3 promoter interacts with CTCF/cohesin-occupied neighboring promoters. In Celsr3 knockout mice, we found that the ventral GP is occupied by aberrant calbindin-positive cholinergic neurons ectopic from the nucleus basalis of Meynert. Furthermore, the guidepost cells for thalamocortical axonal development are missing in the caudal GP. Finally, axonal connections of GP with striatum, subthalamic nucleus, substantia nigra, and raphe are compromised. These data reveal the essential role of Celsr3 in GP development in the basal forebrain and shed light on the mechanisms of the axonal defects caused by the Celsr3 deletion.
Collapse
|
277
|
Reese BE, Keeley PW. Design principles and developmental mechanisms underlying retinal mosaics. Biol Rev Camb Philos Soc 2014; 90:854-76. [PMID: 25109780 DOI: 10.1111/brv.12139] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/24/2014] [Accepted: 07/15/2014] [Indexed: 01/26/2023]
Abstract
Most structures within the central nervous system (CNS) are composed of different types of neuron that vary in both number and morphology, but relatively little is known about the interplay between these two features, i.e. about the population dynamics of a given cell type. How such arrays of neurons are distributed within a structure, and how they differentiate their dendrites relative to each other, are issues that have recently drawn attention in the invertebrate nervous system, where the genetic and molecular underpinnings of these organizing principles are being revealed in exquisite detail. The retina is one of the few locations where these principles have been extensively studied in the vertebrate CNS, indeed, where the design principles of 'mosaic regularity' and 'uniformity of coverage' were first explicitly defined, quantified, and related to each other. Recent studies have revealed a number of genes that influence the formation of these histotypical features in the retina, including homologues of those invertebrate genes, although close inspection reveals that they do not always mediate comparable developmental processes nor elucidate fundamental design principles. The present review considers just how pervasive these features of 'mosaic regularity' and 'uniform dendritic coverage' are within the mammalian retina, discussing the means by which such features can be assessed in the mature and developing nervous system and examining the limitations associated with those assessments. We then address the extent to which these two design principles co-exist within different populations of neurons, and how they are achieved during development. Finally, we consider the neural phenotypes obtained in mutant nervous systems, to address whether a prospective gene of interest underlies those very design principles.
Collapse
Affiliation(s)
- Benjamin E Reese
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106-5060, U.S.A.,Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA 93106-9660, U.S.A
| | - Patrick W Keeley
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106-5060, U.S.A.,Department of Molecular, Cellular & Developmental Biology, University of California, Santa Barbara, CA 93106-9625, U.S.A
| |
Collapse
|
278
|
Samuel MA, Voinescu PE, Lilley BN, de Cabo R, Foretz M, Viollet B, Pawlyk B, Sandberg MA, Vavvas DG, Sanes JR. LKB1 and AMPK regulate synaptic remodeling in old age. Nat Neurosci 2014; 17:1190-7. [PMID: 25086610 PMCID: PMC5369022 DOI: 10.1038/nn.3772] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 07/01/2014] [Indexed: 02/07/2023]
Abstract
Age-related decreases in neural function result in part from alterations in synapses. To identify molecular defects that lead to such changes, we focused on the outer retina, in which synapses are markedly altered in old rodents and humans. We found that the serine/threonine kinase LKB1 and one of its substrates, AMPK, regulate this process. In old mice, synaptic remodeling was accompanied by specific decreases in the levels of total LKB1 and active (phosphorylated) AMPK. In the absence of either kinase, young adult mice developed retinal defects similar to those that occurred in old wild-type animals. LKB1 and AMPK function in rod photoreceptors where their loss leads to aberrant axonal retraction, the extension of postsynaptic dendrites and the formation of ectopic synapses. Conversely, increasing AMPK activity genetically or pharmacologically attenuates and may reverse age-related synaptic alterations. Together, these results identify molecular determinants of age-related synaptic remodeling and suggest strategies for attenuating these changes.
Collapse
Affiliation(s)
- Melanie A Samuel
- 1] Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA. [2]
| | - P Emanuela Voinescu
- 1] Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA. [2]
| | - Brendan N Lilley
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA
| | - Rafa de Cabo
- Laboratory of Experimental Gerontology, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, Baltimore, Maryland, USA
| | - Marc Foretz
- 1] Inserm, U1016, Institut Cochin, Paris, France. [2] CNRS, UMR8104, Paris, France. [3] Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Benoit Viollet
- 1] Inserm, U1016, Institut Cochin, Paris, France. [2] CNRS, UMR8104, Paris, France. [3] Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Basil Pawlyk
- The Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael A Sandberg
- The Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, USA
| | - Demetrios G Vavvas
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
279
|
Polymorphic receptors: neuronal functions and molecular mechanisms of diversification. Curr Opin Neurobiol 2014; 27:25-30. [DOI: 10.1016/j.conb.2014.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/20/2014] [Accepted: 02/06/2014] [Indexed: 01/10/2023]
|
280
|
Hoon M, Okawa H, Della Santina L, Wong ROL. Functional architecture of the retina: development and disease. Prog Retin Eye Res 2014; 42:44-84. [PMID: 24984227 DOI: 10.1016/j.preteyeres.2014.06.003] [Citation(s) in RCA: 348] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/08/2014] [Accepted: 06/22/2014] [Indexed: 12/22/2022]
Abstract
Structure and function are highly correlated in the vertebrate retina, a sensory tissue that is organized into cell layers with microcircuits working in parallel and together to encode visual information. All vertebrate retinas share a fundamental plan, comprising five major neuronal cell classes with cell body distributions and connectivity arranged in stereotypic patterns. Conserved features in retinal design have enabled detailed analysis and comparisons of structure, connectivity and function across species. Each species, however, can adopt structural and/or functional retinal specializations, implementing variations to the basic design in order to satisfy unique requirements in visual function. Recent advances in molecular tools, imaging and electrophysiological approaches have greatly facilitated identification of the cellular and molecular mechanisms that establish the fundamental organization of the retina and the specializations of its microcircuits during development. Here, we review advances in our understanding of how these mechanisms act to shape structure and function at the single cell level, to coordinate the assembly of cell populations, and to define their specific circuitry. We also highlight how structure is rearranged and function is disrupted in disease, and discuss current approaches to re-establish the intricate functional architecture of the retina.
Collapse
Affiliation(s)
- Mrinalini Hoon
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Haruhisa Okawa
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Luca Della Santina
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Rachel O L Wong
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA.
| |
Collapse
|
281
|
He H, Kise Y, Izadifar A, Urwyler O, Ayaz D, Parthasarthy A, Yan B, Erfurth ML, Dascenco D, Schmucker D. Cell-intrinsic requirement of Dscam1 isoform diversity for axon collateral formation. Science 2014; 344:1182-6. [DOI: 10.1126/science.1251852] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
282
|
Gibson DA, Tymanskyj S, Yuan RC, Leung HC, Lefebvre JL, Sanes JR, Chédotal A, Ma L. Dendrite self-avoidance requires cell-autonomous slit/robo signaling in cerebellar purkinje cells. Neuron 2014; 81:1040-1056. [PMID: 24607227 DOI: 10.1016/j.neuron.2014.01.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2013] [Indexed: 10/25/2022]
Abstract
Dendrites from the same neuron usually develop nonoverlapping patterns by self-avoidance, a process requiring contact-dependent recognition and repulsion. Recent studies have implicated homophilic interactions of cell surface molecules, including Dscams and Pcdhgs, in self-recognition, but repulsive molecular mechanisms remain obscure. Here, we report a role for the secreted molecule Slit2 and its receptor Robo2 in self-avoidance of cerebellar Purkinje cells (PCs). Both molecules are highly expressed by PCs, and their deletion leads to excessive dendrite self-crossing without affecting arbor size and shape. This cell-autonomous function is supported by the boundary-establishing activity of Slit in culture and the phenotype rescue by membrane-associated Slit2 activities. Furthermore, genetic studies show that they act independently from Pcdhg-mediated recognition. Finally, PC-specific deletion of Robo2 is associated with motor behavior alterations. Thus, our study uncovers a local repulsive mechanism required for self-avoidance and demonstrates the molecular complexity at the cell surface in dendritic patterning.
Collapse
Affiliation(s)
- Daniel A Gibson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Stephen Tymanskyj
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Rachel C Yuan
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Haiwen C Leung
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Julie L Lefebvre
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Alain Chédotal
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMR_S968, CNRS_UMR7210, Institut de la Vision, 750012, Paris, France
| | - Le Ma
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
283
|
Yang J, Harte-Hargrove LC, Siao CJ, Marinic T, Clarke R, Ma Q, Jing D, Lafrancois JJ, Bath KG, Mark W, Ballon D, Lee FS, Scharfman HE, Hempstead BL. proBDNF negatively regulates neuronal remodeling, synaptic transmission, and synaptic plasticity in hippocampus. Cell Rep 2014; 7:796-806. [PMID: 24746813 PMCID: PMC4118923 DOI: 10.1016/j.celrep.2014.03.040] [Citation(s) in RCA: 211] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 01/30/2014] [Accepted: 03/12/2014] [Indexed: 02/08/2023] Open
Abstract
Experience-dependent plasticity shapes postnatal development of neural circuits, but the mechanisms that refine dendritic arbors, remodel spines, and impair synaptic activity are poorly understood. Mature brain-derived neurotrophic factor (BDNF) modulates neuronal morphology and synaptic plasticity, including long-term potentiation (LTP) via TrkB activation. BDNF is initially translated as proBDNF, which binds p75(NTR). In vitro, recombinant proBDNF modulates neuronal structure and alters hippocampal long-term plasticity, but the actions of endogenously expressed proBDNF are unclear. Therefore, we generated a cleavage-resistant probdnf knockin mouse. Our results demonstrate that proBDNF negatively regulates hippocampal dendritic complexity and spine density through p75(NTR). Hippocampal slices from probdnf mice exhibit depressed synaptic transmission, impaired LTP, and enhanced long-term depression (LTD) in area CA1. These results suggest that proBDNF acts in vivo as a biologically active factor that regulates hippocampal structure, synaptic transmission, and plasticity, effects that are distinct from those of mature BDNF.
Collapse
Affiliation(s)
- Jianmin Yang
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | | - Chia-Jen Siao
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Tina Marinic
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Roshelle Clarke
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Qian Ma
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Deqiang Jing
- Department of Psychiatry, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | - Willie Mark
- Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Douglas Ballon
- Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Helen E Scharfman
- The Nathan Kline Institute, Orangeburg, NY 10962, USA; New York University Langone Medical Center, New York, NY 10016, USA.
| | - Barbara L Hempstead
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
284
|
Roy P, Bandyopadhyay A. Spatio-temporally restricted expression of cell adhesion molecules during chicken embryonic development. PLoS One 2014; 9:e96837. [PMID: 24806091 PMCID: PMC4013082 DOI: 10.1371/journal.pone.0096837] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 04/10/2014] [Indexed: 11/23/2022] Open
Abstract
Differential cell adhesive properties are known to regulate important developmental events like cell sorting and cell migration. Cadherins and protocadherins are known to mediate these cellular properties. Though a large number of such molecules have been predicted, their characterization in terms of interactive properties and cellular roles is far from being comprehensive. To narrow down the tissue context and collect correlative evidence for tissue specific roles of these molecules, we have carried out whole-mount in situ hybridization based RNA expression study for seven cadherins and four protocadherins. In developing chicken embryos (HH stages 18, 22, 26 and 28) cadherins and protocadherins are expressed in tissue restricted manner. This expression study elucidates precise expression domains of cell adhesion molecules in the context of developing embryos. These expression domains provide spatio-temporal context in which the function of these genes can be further explored.
Collapse
Affiliation(s)
- Priti Roy
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, U.P., India
- * E-mail: (PR); (AB)
| | - Amitabha Bandyopadhyay
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, U.P., India
- * E-mail: (PR); (AB)
| |
Collapse
|
285
|
Sotomayor M, Gaudet R, Corey DP. Sorting out a promiscuous superfamily: towards cadherin connectomics. Trends Cell Biol 2014; 24:524-36. [PMID: 24794279 DOI: 10.1016/j.tcb.2014.03.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 03/23/2014] [Accepted: 03/25/2014] [Indexed: 12/21/2022]
Abstract
Members of the cadherin superfamily of proteins are involved in diverse biological processes such as morphogenesis, sound transduction, and neuronal connectivity. Key to cadherin function is their extracellular domain containing cadherin repeats, which can mediate interactions involved in adhesion and cell signaling. Recent cellular, biochemical, and structural studies have revealed that physical interaction among cadherins is more complex than originally thought. Here we review work on new cadherin complexes and discuss how the classification of the mammalian family can be used to search for additional cadherin-interacting partners. We also highlight some of the challenges in cadherin research; namely, the characterization of a cadherin connectome in biochemical and structural terms, as well as the elucidation of molecular mechanisms underlying the functional diversity of nonclassical cadherins in vivo.
Collapse
Affiliation(s)
- Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus OH 43210, USA.
| | - Rachelle Gaudet
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA.
| | - David P Corey
- Howard Hughes Medical Institute, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
286
|
Tsai L, Barnea G. A critical period defined by axon-targeting mechanisms in the murine olfactory bulb. Science 2014; 344:197-200. [PMID: 24723611 DOI: 10.1126/science.1248806] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The olfactory system remains plastic throughout life because of continuous neurogenesis of sensory neurons in the nose and inhibitory interneurons in the olfactory bulb. Here, we reveal that transgenic expression of an odorant receptor has non-cell autonomous effects on axons expressing this receptor from the endogenous gene. Perinatal expression of transgenic odorant receptor causes rerouting of like axons to new glomeruli, whereas expression after the sensory map is established does not lead to rerouting. Further, chemical ablation of the map after rerouting does not restore the normal map, even when the transgenic receptor is no longer expressed. Our results reveal that glomeruli are designated as targets for sensory neurons expressing specific odorant receptors during a critical period in the formation of the olfactory sensory map.
Collapse
Affiliation(s)
- Lulu Tsai
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | | |
Collapse
|
287
|
Singhania A, Grueber WB. Development of the embryonic and larval peripheral nervous system of Drosophila. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 3:193-210. [PMID: 24896657 DOI: 10.1002/wdev.135] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 02/19/2014] [Accepted: 03/05/2014] [Indexed: 01/01/2023]
Abstract
The peripheral nervous system (PNS) of embryonic and larval stage Drosophila consists of diverse types of sensory neurons positioned along the body wall. Sensory neurons, and associated end organs, show highly stereotyped locations and morphologies. Many powerful genetic tools for gene manipulation available in Drosophila make the PNS an advantageous system for elucidating basic principles of neural development. Studies of the Drosophila PNS have provided key insights into molecular mechanisms of cell fate specification, asymmetric cell division, and dendritic morphogenesis. A canonical lineage gives rise to sensory neurons and associated organs, and cells within this lineage are diversified through asymmetric cell divisions. Newly specified sensory neurons develop specific dendritic patterns, which are controlled by numerous factors including transcriptional regulators, interactions with neighboring neurons, and intracellular trafficking systems. In addition, sensory axons show modality specific terminations in the central nervous system, which are patterned by secreted ligands and their receptors expressed by sensory axons. Modality-specific axon projections are critical for coordinated larval behaviors. We review the molecular basis for PNS development and address some of the instances in which the mechanisms and molecules identified are conserved in vertebrate development.
Collapse
Affiliation(s)
- Aditi Singhania
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | | |
Collapse
|
288
|
Satija R, Shalek AK. Heterogeneity in immune responses: from populations to single cells. Trends Immunol 2014; 35:219-29. [PMID: 24746883 DOI: 10.1016/j.it.2014.03.004] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 03/05/2014] [Accepted: 03/07/2014] [Indexed: 12/18/2022]
Abstract
The mammalian immune system is tasked with protecting the host against a broad range of threats. Understanding how immune populations leverage cellular diversity to achieve this breadth and flexibility, particularly during dynamic processes such as differentiation and antigenic response, is a core challenge that is well suited for single cell analysis. Recent years have witnessed transformative and intersecting advances in nanofabrication and genomics that enable deep profiling of individual cells, affording exciting opportunities to study heterogeneity in the immune response at an unprecedented scope. In light of these advances, here we review recent work exploring how immune populations generate and leverage cellular heterogeneity at multiple molecular and phenotypic levels. Additionally, we highlight opportunities for single cell technologies to shed light on the causes and consequences of heterogeneity in the immune system.
Collapse
Affiliation(s)
- Rahul Satija
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA 02142, USA.
| | - Alex K Shalek
- Department of Chemistry and Chemical Biology and Department of Physics, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA.
| |
Collapse
|
289
|
Alexander JM, Lomvardas S. Nuclear architecture as an epigenetic regulator of neural development and function. Neuroscience 2014; 264:39-50. [PMID: 24486963 PMCID: PMC4006947 DOI: 10.1016/j.neuroscience.2014.01.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/11/2014] [Accepted: 01/22/2014] [Indexed: 12/23/2022]
Abstract
The nervous system of higher organisms is characterized by an enormous diversity of cell types that function in concert to carry out a myriad of neuronal functions. Differences in connectivity, and subsequent physiology of the connected neurons, are a result of differences in transcriptional programs. The extraordinary complexity of the nervous system requires an equally complex regulatory system. It is well established that transcription factor combinations and the organization of cis-regulatory sequences control commitment to differentiation programs and preserve a nuclear plasticity required for neuronal functions. However, an additional level of regulation is provided by epigenetic controls. Among various epigenetic processes, nuclear organization and the control of genome architecture emerge as an efficient and powerful form of gene regulation that meets the unique needs of the post-mitotic neuron. Here, we present an outline of how nuclear architecture affects transcription and provide examples from the recent literature where these principles are used by the nervous system.
Collapse
Affiliation(s)
- J M Alexander
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - S Lomvardas
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
290
|
Toyoda S, Kawaguchi M, Kobayashi T, Tarusawa E, Toyama T, Okano M, Oda M, Nakauchi H, Yoshimura Y, Sanbo M, Hirabayashi M, Hirayama T, Hirabayashi T, Yagi T. Developmental epigenetic modification regulates stochastic expression of clustered protocadherin genes, generating single neuron diversity. Neuron 2014; 82:94-108. [PMID: 24698270 DOI: 10.1016/j.neuron.2014.02.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2014] [Indexed: 01/08/2023]
Abstract
In the brain, enormous numbers of neurons have functional individuality and distinct circuit specificities. Clustered Protocadherins (Pcdhs), diversified cell-surface proteins, are stochastically expressed by alternative promoter choice and affect dendritic arborization in individual neurons. Here we found that the Pcdh promoters are differentially methylated by the de novo DNA methyltransferase Dnmt3b during early embryogenesis. To determine this methylation's role in neurons, we produced chimeric mice from Dnmt3b-deficient induced pluripotent stem cells (iPSCs). Single-cell expression analysis revealed that individual Dnmt3b-deficient Purkinje cells expressed increased numbers of Pcdh isoforms; in vivo, they exhibited abnormal dendritic arborization. These results indicate that DNA methylation by Dnmt3b at early embryonic stages regulates the probability of expression for the stochastically expressed Pcdh isoforms. They also suggest a mechanism for a rare human recessive disease, the ICF (Immunodeficiency, Centromere instability, and Facial anomalies) syndrome, which is caused by Dnmt3b mutations.
Collapse
Affiliation(s)
- Shunsuke Toyoda
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masahumi Kawaguchi
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihiro Kobayashi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Japan Science Technology Agency, ERATO, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Etsuko Tarusawa
- Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Division of Developmental Neurophysiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Tomoko Toyama
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masaki Okano
- Laboratory for Mammalian Epigenetic Studies, RIKEN Center for Developmental Biology, Minatojima-minamimachi 2-2-3, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Masaaki Oda
- Laboratory for Mammalian Epigenetic Studies, RIKEN Center for Developmental Biology, Minatojima-minamimachi 2-2-3, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Japan Science Technology Agency, ERATO, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yumiko Yoshimura
- Division of Developmental Neurophysiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, The Graduate University for Advanced Studies, Okazaki, Aichi 444-8585, Japan
| | - Makoto Sanbo
- Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
| | - Masumi Hirabayashi
- Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Physiological Sciences, The Graduate University for Advanced Studies, Okazaki, Aichi 444-8585, Japan; Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
| | - Teruyoshi Hirayama
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takahiro Hirabayashi
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takeshi Yagi
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
291
|
High frequency of rare variants with a moderate-to-high predicted biological effect in protocadherin genes of extremely obese. GENES AND NUTRITION 2014; 9:399. [PMID: 24682882 DOI: 10.1007/s12263-014-0399-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 03/17/2014] [Indexed: 10/25/2022]
Abstract
Relatively rare variants with a moderate-to-high biological effect may contribute to the genetic predisposition of common disorders. To investigate this for obesity, we performed exome sequencing for 30 young (mean age: 29.7 years) extremely obese Caucasian subjects (mean body mass index: 51.1 kg/m(2); m/f = 11/29). Rare variants with a moderate-to-high predicted biological effect were assembled and subjected to functional clustering analysis. It showed that the 55 clustered protocadherin genes on chromosome 5q31 have a significantly (P = 0.002) higher frequency of rare variants than a set of 325 reference genes. Since the protocadherin genes are expressed in the hypothalamus, we tested another 167 genes related to the function of the hypothalamus, but in those genes, the frequency of rare variants was not different from that of the reference genes. To verify the relation of variation in the protocadherin genes with extreme obesity, we analyzed data from more than 4,000 European Americans present on the Exome Variant Server, representing a sample of the general population. The significant enrichment of rare variants in the protocadherin genes was only observed with the group of extremely obese individuals but not in the "general population", indicating an association between rare variants in the protocadherin cluster genes and extreme obesity.
Collapse
|
292
|
Abstract
Injuries to peripheral nerves can cause paralysis and sensory disturbances, but such functional impairments are often short lived because of efficient regeneration of damaged axons. The time required for functional recovery, however, increases with advancing age (Verdú et al., 2000; Kawabuchi et al., 2011). Incomplete or delayed recovery after peripheral nerve damage is a major health concern in the aging population because it can severely restrict a person's mobility and independence. A variety of possible causes have been suggested to explain why nervous systems in aged individuals recover more slowly from nerve damage. Potential causes include age-related declines in the regenerative potential of peripheral axons and decreases in the supply or responsivity to trophic and/or tropic factors. However, there have been few direct analyses of age-related axon regeneration. Our aim here was to observe axons directly in young and old mice as they regenerate and ultimately reoccupy denervated neuromuscular synaptic sites to learn what changes in this process are age related. We find that damaged nerves in aged animals clear debris more slowly than nerves in young animals and that the greater number of obstructions regenerating axons encounter in the endoneurial tubes of old animals give rise to slower regeneration. Surprisingly, however, axons from aged animals regenerate quickly when not confronted by debris and reoccupy neuromuscular junction sites efficiently. These results imply that facilitating clearance of axon debris might be a good target for the treatment of nerve injury in the aged.
Collapse
|
293
|
Nguyen-Ba-Charvet KT, Chédotal A. Development of retinal layers. C R Biol 2014; 337:153-9. [PMID: 24702841 DOI: 10.1016/j.crvi.2013.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 11/28/2013] [Indexed: 11/26/2022]
Abstract
A noticeable characteristic of nervous systems is the arrangement of synapses into distinct layers. Such laminae are fundamental for the spatial organisation of synaptic connections transmitting different kinds of information. A major example of this is the inner plexiform layer (IPL) of the vertebrate retina, which is subdivided into at least ten sublayers. Another noticeable characteristic of these retina layers is that neurons are displayed in the horizontal plane in a non-random array termed as mosaic patterning. Recent studies of vertebrate and invertebrate systems have identified molecules that mediate these interactions. Here, we review the last mechanisms and molecules mediating retinal layering.
Collapse
Affiliation(s)
- Kim Tuyen Nguyen-Ba-Charvet
- Institut national de la santé et de la recherche médicale, UMR S968, CNRS UMR 7210, Université Pierre et Marie Curie (Paris-6), Institut de la vision, 17, rue Moreau, 75012 Paris, France
| | - Alain Chédotal
- Institut national de la santé et de la recherche médicale, UMR S968, CNRS UMR 7210, Université Pierre et Marie Curie (Paris-6), Institut de la vision, 17, rue Moreau, 75012 Paris, France.
| |
Collapse
|
294
|
Miura SK, Martins A, Zhang KX, Graveley BR, Zipursky SL. Probabilistic splicing of Dscam1 establishes identity at the level of single neurons. Cell 2014; 155:1166-77. [PMID: 24267895 DOI: 10.1016/j.cell.2013.10.018] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/27/2013] [Accepted: 10/09/2013] [Indexed: 11/17/2022]
Abstract
The Drosophila Dscam1 gene encodes a vast number of cell recognition molecules through alternative splicing. These exhibit isoform-specific homophilic binding and regulate self-avoidance, the tendency of neurites from the same cell to repel one another. Genetic experiments indicate that different cells must express different isoforms. How this is achieved is unknown, as expression of alternative exons in vivo has not been shown. Here, we modified the endogenous Dscam1 locus to generate splicing reporters for all variants of exon 4. We demonstrate that splicing does not occur in a cell-type-specific fashion, that cells sharing the same anatomical location in different individuals express different exon 4 variants, and that the splicing pattern in a given neuron can change over time. We conclude that splicing is probabilistic. This is compatible with a widespread role in neural circuit assembly through self-avoidance and is incompatible with models in which specific isoforms of Dscam1 mediate homophilic recognition between processes of different cells.
Collapse
Affiliation(s)
- Satoru K Miura
- Department of Biological Chemistry, Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | |
Collapse
|
295
|
Ting CY, McQueen PG, Pandya N, Lin TY, Yang M, Reddy OV, O'Connor MB, McAuliffe M, Lee CH. Photoreceptor-derived activin promotes dendritic termination and restricts the receptive fields of first-order interneurons in Drosophila. Neuron 2014; 81:830-846. [PMID: 24462039 DOI: 10.1016/j.neuron.2013.12.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2013] [Indexed: 01/10/2023]
Abstract
How neurons form appropriately sized dendritic fields to encounter their presynaptic partners is poorly understood. The Drosophila medulla is organized in layers and columns and innervated by medulla neuron dendrites and photoreceptor axons. Here, we show that three types of medulla projection (Tm) neurons extend their dendrites in stereotyped directions and to distinct layers within a single column for processing retinotopic information. In contrast, the Dm8 amacrine neurons form a wide dendritic field to receive ∼16 R7 photoreceptor inputs. R7- and R8-derived Activin selectively restricts the dendritic fields of their respective postsynaptic partners, Dm8 and Tm20, to the size appropriate for their functions. Canonical Activin signaling promotes dendritic termination without affecting dendritic routing direction or layer. Tm20 neurons lacking Activin signaling expanded their dendritic fields and aberrantly synapsed with neighboring photoreceptors. We suggest that afferent-derived Activin regulates the dendritic field size of their postsynaptic partners to ensure appropriate synaptic partnership.
Collapse
Affiliation(s)
- Chun-Yuan Ting
- Section on Neuronal Connectivity, Laboratory of Gene Regulation and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Philip G McQueen
- Mathematical and Statistical Computing Laboratory, Division of Computational Bioscience, Center for Information Technology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nishith Pandya
- Biomedical Imaging Research Services Section, Center for Information Technology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tzu-Yang Lin
- Section on Neuronal Connectivity, Laboratory of Gene Regulation and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Meiluen Yang
- Section on Neuronal Connectivity, Laboratory of Gene Regulation and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - O Venkateswara Reddy
- National Center for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bangalore 560065, India
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, MN 55455, USA
| | - Matthew McAuliffe
- Biomedical Imaging Research Services Section, Center for Information Technology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chi-Hon Lee
- Section on Neuronal Connectivity, Laboratory of Gene Regulation and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
296
|
Tucker RP, Adams JC. Adhesion networks of cnidarians: a postgenomic view. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 308:323-77. [PMID: 24411175 DOI: 10.1016/b978-0-12-800097-7.00008-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell-extracellular matrix (ECM) and cell-cell adhesion systems are fundamental to the multicellularity of metazoans. Members of phylum Cnidaria were classified historically by their radial symmetry as an outgroup to bilaterian animals. Experimental study of Hydra and jellyfish has fascinated zoologists for many years. Laboratory studies, based on dissection, biochemical isolations, or perturbations of the living organism, have identified the ECM layer of cnidarians (mesoglea) and its components as important determinants of stem cell properties, cell migration and differentiation, tissue morphogenesis, repair, and regeneration. Studies of the ultrastructure and functions of intercellular gap and septate junctions identified parallel roles for these structures in intercellular communication and morphogenesis. More recently, the sequenced genomes of sea anemone Nematostella vectensis, Hydra magnipapillata, and coral Acropora digitifera have opened up a new frame of reference for analyzing the cell-ECM and cell-cell adhesion molecules of cnidarians and examining their conservation with bilaterians. This chapter integrates a review of literature on the structure and functions of cell-ECM and cell-cell adhesion systems in cnidarians with current analyses of genome-encoded repertoires of adhesion molecules. The postgenomic perspective provides a fresh view on fundamental similarities between cnidarian and bilaterian animals and is impelling wider adoption of species from phylum Cnidaria as model organisms.
Collapse
Affiliation(s)
- Richard P Tucker
- Department of Cell Biology and Human Anatomy, University of California, Davis, California, USA.
| | - Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
297
|
Abstract
A complete understanding of nervous system function cannot be achieved without the identification of its component cell types. In this Perspective, we explore a series of related issues surrounding cell identity and how revolutionary methods for labeling and probing specific neuronal types have clarified this question. Specifically, we ask the following questions: what is the purpose of such diversity, how is it generated, how is it maintained, and, ultimately, how can one unambiguously identity one cell type from another? We suggest that each cell type can be defined by a unique and conserved molecular ground state that determines its capabilities. We believe that gaining an understanding of these molecular barcodes will advance our ability to explore brain function, enhance our understanding of the biochemical basis of CNS disorders, and aid in the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Gord Fishell
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University, New York, NY 10016, USA.
| | | |
Collapse
|
298
|
Visual circuit assembly requires fine tuning of the novel Ig transmembrane protein Borderless. J Neurosci 2013; 33:17413-21. [PMID: 24174674 DOI: 10.1523/jneurosci.1878-13.2013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Establishment of synaptic connections in the neuropils of the developing nervous system requires the coordination of specific neurite-neurite interactions (i.e., axon-axon, dendrite-dendrite and axon-dendrite interactions). The molecular mechanisms underlying coordination of neurite-neurite interactions for circuit assembly are incompletely understood. In this report, we identify a novel Ig superfamily transmembrane protein that we named Borderless (Bdl), as a novel regulator of neurite-neurite interactions in Drosophila. Bdl induces homotypic cell-cell adhesion in vitro and mediates neurite-neurite interactions in the developing visual system. Bdl interacts physically and genetically with the Ig transmembrane protein Turtle, a key regulator of axonal tiling. Our results also show that the receptor tyrosine phosphatase leukocyte common antigen-related protein (LAR) negatively regulates Bdl to control synaptic-layer selection. We propose that precise regulation of Bdl action coordinates neurite-neurite interactions for circuit formation in Drosophila.
Collapse
|
299
|
Dong X, Liu OW, Howell AS, Shen K. An extracellular adhesion molecule complex patterns dendritic branching and morphogenesis. Cell 2013; 155:296-307. [PMID: 24120131 PMCID: PMC3927720 DOI: 10.1016/j.cell.2013.08.059] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 07/29/2013] [Accepted: 08/26/2013] [Indexed: 12/21/2022]
Abstract
Robust dendrite morphogenesis is a critical step in the development of reproducible neural circuits. However, little is known about the extracellular cues that pattern complex dendrite morphologies. In the model nematode Caenorhabditis elegans, the sensory neuron PVD establishes stereotypical, highly branched dendrite morphology. Here, we report the identification of a tripartite ligand-receptor complex of membrane adhesion molecules that is both necessary and sufficient to instruct spatially restricted growth and branching of PVD dendrites. The ligand complex SAX-7/L1CAM and MNR-1 function at defined locations in the surrounding hypodermal tissue, whereas DMA-1 acts as the cognate receptor on PVD. Mutations in this complex lead to dramatic defects in the formation, stabilization, and organization of the dendritic arbor. Ectopic expression of SAX-7 and MNR-1 generates a predictable, unnaturally patterned dendritic tree in a DMA-1-dependent manner. Both in vivo and in vitro experiments indicate that all three molecules are needed for interaction.
Collapse
Affiliation(s)
- Xintong Dong
- Howard Hughes Medical Institute, Department of Biology, Stanford University, 385 Serra Mall, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
300
|
Baier H. Synaptic laminae in the visual system: molecular mechanisms forming layers of perception. Annu Rev Cell Dev Biol 2013; 29:385-416. [PMID: 24099086 DOI: 10.1146/annurev-cellbio-101011-155748] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Synaptic connections between neurons form the basis for perception and behavior. Synapses are often clustered in space, forming stereotyped layers. In the retina and optic tectum, multiple such synaptic laminae are stacked on top of each other, giving rise to stratified neuropil regions in which each layer combines synapses responsive to a particular sensory feature. Recently, several cellular and molecular mechanisms that underlie the development of multilaminar arrays of synapses have been discovered. These mechanisms include neurite guidance and cell-cell recognition. Molecules of the Slit, Semaphorin, Netrin, and Hedgehog families, binding to their matching receptors, bring axons and dendrites into spatial register. These guidance cues may diffuse over short distances or bind to sheets of extracellular matrix, thus conditioning the local extracellular milieu, or are presented on the surface of cells bordering the future neuropil. In addition, mutual recognition of axons and dendrites through adhesion molecules with immunoglobulin domains ensures cell type-specific connections within a given layer. Thus, an elaborate genetic program assembles the parallel processing channels that underlie visual perception.
Collapse
Affiliation(s)
- Herwig Baier
- Genes - Circuits - Behavior, Max Planck Institute of Neurobiology, 82152 Martinsried near Munich, Germany;
| |
Collapse
|