Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
J Neurosci ; 43(6): 918-935, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36604170

ABSTRACT

The establishment of a functional cerebral cortex depends on the proper execution of multiple developmental steps, culminating in dendritic and axonal outgrowth and the formation and maturation of synaptic connections. Dysregulation of these processes can result in improper neuronal connectivity, including that associated with various neurodevelopmental disorders. The γ-Protocadherins (γ-Pcdhs), a family of 22 distinct cell adhesion molecules that share a C-terminal cytoplasmic domain, are involved in multiple aspects of neurodevelopment including neuronal survival, dendrite arborization, and synapse development. The extent to which individual γ-Pcdh family members play unique versus common roles remains unclear. We demonstrated previously that the γ-Pcdh-C3 isoform (γC3), via its unique "variable" cytoplasmic domain (VCD), interacts in cultured cells with Axin1, a Wnt-pathway scaffold protein that regulates the differentiation and morphology of neurons. Here, we confirm that γC3 and Axin1 interact in the cortex in vivo and show that both male and female mice specifically lacking γC3 exhibit disrupted Axin1 localization to synaptic fractions, without obvious changes in dendritic spine density or morphology. However, both male and female γC3 knock-out mice exhibit severely decreased dendritic complexity of cortical pyramidal neurons that is not observed in mouse lines lacking several other γ-Pcdh isoforms. Combining knock-out with rescue constructs in cultured cortical neurons pooled from both male and female mice, we show that γC3 promotes dendritic arborization through an Axin1-dependent mechanism mediated through its VCD. Together, these data identify a novel mechanism through which γC3 uniquely regulates the formation of cortical circuitry.SIGNIFICANCE STATEMENT The complexity of a neuron's dendritic arbor is critical for its function. We showed previously that the γ-Protocadherin (γ-Pcdh) family of 22 cell adhesion molecules promotes arborization during development; it remained unclear whether individual family members played unique roles. Here, we show that one γ-Pcdh isoform, γC3, interacts in the brain with Axin1, a scaffolding protein known to influence dendrite development. A CRISPR/Cas9-generated mutant mouse line lacking γC3 (but not lines lacking other γ-Pcdhs) exhibits severely reduced dendritic complexity of cerebral cortex neurons. Using cultured γC3 knock-out neurons and a variety of rescue constructs, we confirm that the γC3 cytoplasmic domain promotes arborization through an Axin1-dependent mechanism. Thus, γ-Pcdh isoforms are not interchangeable, but rather can play unique neurodevelopmental roles.


Subject(s)
Dendrites , Protocadherins , Animals , Female , Male , Mice , Axin Protein/metabolism , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Dendrites/physiology , Mice, Knockout , Neuronal Plasticity , Protein Isoforms/genetics , Protein Isoforms/metabolism
2.
PLoS Genet ; 15(12): e1008554, 2019 12.
Article in English | MEDLINE | ID: mdl-31877124

ABSTRACT

The mammalian Pcdhg gene cluster encodes a family of 22 cell adhesion molecules, the gamma-Protocadherins (γ-Pcdhs), critical for neuronal survival and neural circuit formation. The extent to which isoform diversity-a γ-Pcdh hallmark-is required for their functions remains unclear. We used a CRISPR/Cas9 approach to reduce isoform diversity, targeting each Pcdhg variable exon with pooled sgRNAs to generate an allelic series of 26 mouse lines with 1 to 21 isoforms disrupted via discrete indels at guide sites and/or larger deletions/rearrangements. Analysis of 5 mutant lines indicates that postnatal viability and neuronal survival do not require isoform diversity. Surprisingly, given reports that it might not independently engage in trans-interactions, we find that γC4, encoded by Pcdhgc4, is the only critical isoform. Because the human orthologue is the only PCDHG gene constrained in humans, our results indicate a conserved γC4 function that likely involves distinct molecular mechanisms.


Subject(s)
Alternative Splicing , Cadherins/genetics , Mutation , Neurons/metabolism , Animals , CRISPR-Cas Systems , Cadherin Related Proteins , Cadherins/metabolism , Embryonic Development , Exons , Female , Humans , INDEL Mutation , Male , Mice , Multigene Family , Neurons/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Deletion , Whole Genome Sequencing
3.
Proc Natl Acad Sci U S A ; 115(43): E10216-E10224, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30297418

ABSTRACT

During neural development, self-avoidance ensures that a neuron's processes arborize to evenly fill a particular spatial domain. At the individual cell level, self-avoidance is promoted by genes encoding cell-surface molecules capable of generating thousands of diverse isoforms, such as Dscam1 (Down syndrome cell adhesion molecule 1) in Drosophila Isoform choice differs between neighboring cells, allowing neurons to distinguish "self" from "nonself". In the mouse retina, Dscam promotes self-avoidance at the level of cell types, but without extreme isoform diversity. Therefore, we hypothesize that DSCAM is a general self-avoidance cue that "masks" other cell type-specific adhesion systems to prevent overly exuberant adhesion. Here, we provide in vivo and in vitro evidence that DSCAM masks the functions of members of the cadherin superfamily, supporting this hypothesis. Thus, unlike the isoform-rich molecules tasked with self-avoidance at the individual cell level, here the diversity resides on the adhesive side, positioning DSCAM as a generalized modulator of cell adhesion during neural development.


Subject(s)
Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Retina/metabolism , Animals , Cell Adhesion/physiology , Cell Line , Cell Membrane/metabolism , Choice Behavior/physiology , Drosophila/metabolism , Drosophila Proteins/metabolism , HEK293 Cells , Humans , Mice , Neural Cell Adhesion Molecules/metabolism , Neurites/metabolism , Neurogenesis/physiology , Neurons/metabolism
4.
bioRxiv ; 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38328061

ABSTRACT

The Pcdhg gene cluster encodes 22 γ-Protocadherin (γ-Pcdh) cell adhesion molecules that critically regulate multiple aspects of neural development, including neuronal survival, dendritic and axonal arborization, and synapse formation and maturation. Each γ-Pcdh isoform has unique protein domains-a homophilically-interacting extracellular domain and a juxtamembrane cytoplasmic domain-as well as a C-terminal cytoplasmic domain shared by all isoforms. The extent to which isoform-specific vs. shared domains regulate distinct γ-Pcdh functions remains incompletely understood. Our previous in vitro studies identified PKC phosphorylation of a serine residue within a shared C-terminal motif as a mechanism through which γ-Pcdh promotion of dendrite arborization via MARCKS is abrogated. Here, we used CRISPR/Cas9 genome editing to generate two new mouse lines expressing only non-phosphorylatable γ-Pcdhs, due either to a serine-to-alanine mutation (PcdhgS/A) or to a 15-amino acid C-terminal deletion resulting from insertion of an early stop codon (PcdhgCTD). Both lines are viable and fertile, and the density and maturation of dendritic spines remains unchanged in both PcdhgS/A and PcdhgCTD cortex. Dendrite arborization of cortical pyramidal neurons, however, is significantly increased in both lines, as are levels of active MARCKS. Intriguingly, despite having significantly reduced levels of γ-Pcdh proteins, the PcdhgCTD mutation yields the strongest phenotype, with even heterozygous mutants exhibiting increased arborization. The present study confirms that phosphorylation of a shared C-terminal motif is a key γ-Pcdh negative regulation point, and contributes to a converging understanding of γ-Pcdh family function in which distinct roles are played by both individual isoforms and discrete protein domains.

5.
Dev Neurobiol ; 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38837880

ABSTRACT

The Pcdhg gene cluster encodes 22 γ-Protocadherin (γ-Pcdh) cell adhesion molecules that critically regulate multiple aspects of neural development, including neuronal survival, dendritic and axonal arborization, and synapse formation and maturation. Each γ-Pcdh isoform has unique protein domains-a homophilically interacting extracellular domain and a juxtamembrane cytoplasmic domain-as well as a C-terminal cytoplasmic domain shared by all isoforms. The extent to which isoform-specific versus shared domains regulate distinct γ-Pcdh functions remains incompletely understood. Our previous in vitro studies identified protein kinase C (PKC) phosphorylation of a serine residue within a shared C-terminal motif as a mechanism through which γ-Pcdh promotion of dendrite arborization via myristoylated alanine-rich C-kinase substrate (MARCKS) is abrogated. Here, we used CRISPR/Cas9 genome editing to generate two new mouse lines expressing only non-phosphorylatable γ-Pcdhs, due either to a serine-to-alanine mutation (PcdhgS/A) or to a 15-amino acid C-terminal deletion resulting from insertion of an early stop codon (PcdhgCTD). Both lines are viable and fertile, and the density and maturation of dendritic spines remain unchanged in both PcdhgS/A and PcdhgCTD cortex. Dendrite arborization of cortical pyramidal neurons, however, is significantly increased in both lines, as are levels of active MARCKS. Intriguingly, despite having significantly reduced levels of γ-Pcdh proteins, the PcdhgCTD mutation yields the strongest phenotype, with even heterozygous mutants exhibiting increased arborization. The present study confirms that phosphorylation of a shared C-terminal motif is a key γ-Pcdh negative regulation point and contributes to a converging understanding of γ-Pcdh family function in which distinct roles are played by both individual isoforms and discrete protein domains.

6.
Neuron ; 111(11): 1776-1794.e10, 2023 06 07.
Article in English | MEDLINE | ID: mdl-37028432

ABSTRACT

Light touch sensation begins with activation of low-threshold mechanoreceptor (LTMR) endings in the skin and propagation of their signals to the spinal cord and brainstem. We found that the clustered protocadherin gamma (Pcdhg) gene locus, which encodes 22 cell-surface homophilic binding proteins, is required in somatosensory neurons for normal behavioral reactivity to a range of tactile stimuli. Developmentally, distinct Pcdhg isoforms mediate LTMR synapse formation through neuron-neuron interactions and peripheral axonal branching through neuron-glia interactions. The Pcdhgc3 isoform mediates homophilic interactions between sensory axons and spinal cord neurons to promote synapse formation in vivo and is sufficient to induce postsynaptic specializations in vitro. Moreover, loss of Pcdhgs and somatosensory synaptic inputs to the dorsal horn leads to fewer corticospinal synapses on dorsal horn neurons. These findings reveal essential roles for Pcdhg isoform diversity in somatosensory neuron synapse formation, peripheral axonal branching, and stepwise assembly of central mechanosensory circuitry.


Subject(s)
Sensory Receptor Cells , Spinal Cord , Sensory Receptor Cells/physiology , Spinal Cord/physiology , Cadherins/genetics , Cadherins/metabolism , Synapses , Spinal Cord Dorsal Horn , Protein Isoforms/genetics , Protein Isoforms/metabolism
7.
Curr Top Dev Biol ; 148: 115-137, 2022.
Article in English | MEDLINE | ID: mdl-35461562

ABSTRACT

Since their first description, the clustered protocadherins (cPcdhs) have sparked interest for their potential to generate diverse cell-surface recognition cues and their widespread expression in the nervous system. Through the use of mouse models, we have learned a great deal about the functions served by cPcdhs, and how their molecular diversity is regulated. cPcdhs are essential contributors to a host of processes during neural circuit formation, including neuronal survival, dendritic and axonal branching, self-avoidance and targeting, and synapse formation. Their expression is controlled by the interplay of epigenetic marks with proximal and distal elements involving high order DNA looping, regulating transcription factor binding. Here, we will review various mouse models targeting the cPcdh locus and how they have been instructive in uncovering the regulation and function of the cPcdhs.


Subject(s)
Cadherins , Protocadherins , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Communication , Cell Survival , Mice , Neurogenesis
8.
Prog Retin Eye Res ; 90: 101038, 2022 09.
Article in English | MEDLINE | ID: mdl-35066146

ABSTRACT

Cadherins are a superfamily of calcium-dependent intercellular adhesion molecules that are widely expressed in living tissues. Within the retina and retinal pigment epithelium (RPE), cadherins contribute to tissue morphogenesis, neural circuit formation, adherens junctions of the outer blood-retinal barrier, photoreceptor disc morphogenesis, maintenance and survival. Four monogenic disorders involving genes which encode cadherins have been identified as causes of inherited retinal degeneration: the retinal cadherinopathies (CDHR1, CDH23, PCDH15, CDH3). Biallelic variants in CDHR1 result in cone-rod dystrophy, rod-cone dystrophy or late-onset macular dystrophy which may be misclassified as dry age-related macular degeneration. Biallelic variants in CDH23 and PCDH15 underlie Usher Syndrome type 1D and 1F. Hypotrichosis with juvenile macular dystrophy results from biallelic variants in CDH3, which contributes to adherens tight junctions between RPE cells. In this review, we summarise the classification of cadherins, and the role of cadherins in the physiology and morphogenesis of the inner and outer retina. Cadherins expressed in primate photoreceptors (CDHR1, CDH23 and PCDH15) have evolved complex roles in outer segment disc morphogenesis and maintenance involving intracellular heterophilic interactions which are as yet incompletely characterised. We highlight what is currently unknown about the molecular function of these cadherins, and review the pathogenesis, clinical phenotype and molecular genetics of each monogenic retinal cadherinopathy. Genes regulating the expression and post-translational modification of retinal cadherins, or those coding for as yet unidentified interacting partners, are candidates for unsolved cases of retinal degeneration. This group of disorders is potentially treatable; we summarise the likely molecular therapeutic approaches and future directions for each retinal cadherinopathy.


Subject(s)
Macular Degeneration , Retinal Degeneration , Animals , Cadherins/genetics , Cadherins/metabolism , Macular Degeneration/genetics , Retina/metabolism , Retinal Degeneration/genetics
9.
J Neurosci ; 29(38): 11723-31, 2009 Sep 23.
Article in English | MEDLINE | ID: mdl-19776259

ABSTRACT

Recent studies indicate that astrocytes, whose processes enwrap synaptic terminals, promote synapse formation both by releasing soluble factors and through contact-dependent mechanisms. Although astrocyte-secreted synaptogenic factors have been identified, the molecules underlying perisynaptic astroctye-neuron contacts are unknown. Here we show that the gamma-protocadherins (gamma-Pcdhs), a family of 22 neuronal adhesion molecules encoded by a single gene cluster, are also expressed by astrocytes and localize to their perisynaptic processes. Using cocultures in which either astrocytes or neurons are Pcdh-gamma-null, we find that astrocyte-neuron gamma-Pcdh contacts are critical for synaptogenesis in developing cultures. Synaptogenesis can eventually proceed among neurons cocultured with Pcdh-gamma-null astrocytes, but only if these neurons themselves express the gamma-Pcdhs. Consistent with this, restricted mutation of the Pcdh-gamma cluster in astrocytes in vivo significantly delays both excitatory and inhibitory synapse formation. Together, these results identify the first known contact-dependent mechanism by which perisynaptic astrocyte processes promote synaptogenesis.


Subject(s)
Astrocytes/physiology , Cadherins/metabolism , Hippocampus/physiology , Neurons/physiology , Spinal Cord/physiology , Synapses/physiology , Animals , Animals, Newborn , Cadherin Related Proteins , Cadherins/genetics , Coculture Techniques , Culture Media, Conditioned , Fluorescent Antibody Technique , In Situ Hybridization, Fluorescence , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/embryology , Spinal Cord/growth & development
10.
Cell Rep ; 18(11): 2702-2714, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28297673

ABSTRACT

The 22 γ-Protocadherin (γ-Pcdh) cell adhesion molecules are critical for the elaboration of complex dendritic arbors in the cerebral cortex. Here, we provide evidence that the γ-Pcdhs negatively regulate synapse development by inhibiting the postsynaptic cell adhesion molecule, neuroligin-1 (Nlg1). Mice lacking all γ-Pcdhs in the forebrain exhibit significantly increased dendritic spine density in vivo, while spine density is significantly decreased in mice overexpressing one of the 22 γ-Pcdh isoforms. Co-expression of γ-Pcdhs inhibits the ability of Nlg1 to increase spine density and to induce presynaptic differentiation in hippocampal neurons in vitro. The γ-Pcdhs physically interact in cis with Nlg1 both in vitro and in vivo, and we present evidence that this disrupts Nlg1 binding to its presynaptic partner neurexin1ß. Together with prior work, these data identify a mechanism through which γ-Pcdhs could coordinate dendrite arbor growth and complexity with spine maturation in the developing brain.


Subject(s)
Cadherins/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Dendritic Spines/metabolism , Neurogenesis , Animals , COS Cells , Cadherin Related Proteins , Cell Membrane/metabolism , Cells, Cultured , Chlorocebus aethiops , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Presynaptic Terminals/metabolism , Protein Binding
11.
Elife ; 52016 09 16.
Article in English | MEDLINE | ID: mdl-27637097

ABSTRACT

Different types of neurons in the retina are organized vertically into layers and horizontally in a mosaic pattern that helps ensure proper neural network formation and information processing throughout the visual field. The vertebrate Dscams (DSCAM and DSCAML1) are cell adhesion molecules that support the development of this organization by promoting self-avoidance at the level of cell types, promoting normal developmental cell death, and directing vertical neurite stratification. To understand the molecular interactions required for these activities, we tested the functional significance of the interaction between the C-terminus of the Dscams and multi-PDZ domain-containing scaffolding proteins in mouse. We hypothesized that this PDZ-interacting domain would mediate a subset of the Dscams' functions. Instead, we found that in the absence of these interactions, some cell types developed almost normally, while others resembled complete loss of function. Thus, we show differential dependence on this domain for Dscams' functions in different cell types.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Epitopes/genetics , Epitopes/metabolism , Protein Interaction Domains and Motifs , Animals , Mice , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
Front Mol Neurosci ; 9: 3, 2016.
Article in English | MEDLINE | ID: mdl-26858598

ABSTRACT

Boundary cap cells (BCC) are a transient, neural-crest-derived population found at the motor exit point (MEP) and dorsal root entry zone (DREZ) of the embryonic spinal cord. These cells contribute to the central/peripheral nervous system (CNS/PNS) boundary, and in their absence neurons and glia from the CNS migrate into the PNS. We found Netrin5 (Ntn5), a previously unstudied member of the netrin gene family, to be robustly expressed in BCC. We generated Ntn5 knockout mice and examined neurodevelopmental and BCC-related phenotypes. No abnormalities in cranial nerve guidance, dorsal root organization, or sensory projections were found. However, Ntn5 mutant embryos did have ectopic motor neurons (MNs) that migrated out of the ventral horn and into the motor roots. Previous studies have implicated semaphorin6A (Sema6A) in BCC signaling to plexinA2 (PlxnA2)/neuropilin2 (Nrp2) in MNs in restricting MN cell bodies to the ventral horn, particularly in the caudal spinal cord. In Ntn5 mutants, ectopic MNs are likely to be a different population, as more ectopias were found rostrally. Furthermore, ectopic MNs in Ntn5 mutants were not immunoreactive for NRP2. The netrin receptor deleted in colorectal cancer (DCC) is a potential receptor for NTN5 in MNs, as similar ectopic neurons were found in Dcc mutant mice, but not in mice deficient for other netrin receptors. Thus, Ntn5 is a novel netrin family member that is expressed in BCC, functioning to prevent MN migration out of the CNS.

13.
Elife ; 42015 Aug 25.
Article in English | MEDLINE | ID: mdl-26305501

ABSTRACT

Proteins called gamma-protocadherins are essential for the establishment of working circuits of neurons in the retina.


Subject(s)
Amacrine Cells/drug effects , Amacrine Cells/physiology , Cadherins/metabolism , Dendrites/drug effects , Dendrites/physiology , Animals
14.
Front Mol Neurosci ; 5: 86, 2012.
Article in English | MEDLINE | ID: mdl-22912601

ABSTRACT

Many of the models of neurodevelopmental processes such as cell migration, axon outgrowth, and dendrite arborization involve cell adhesion and chemoattraction as critical physical or mechanical aspects of the mechanism. However, the prevention of adhesion or attraction is under-appreciated as a necessary, active process that balances these forces, insuring that the correct cells are present and adhering in the correct place at the correct time. The phenomenon of not adhering is often viewed as the passive alternative to adhesion, and in some cases this may be true. However, it is becoming increasingly clear that active signaling pathways are involved in preventing adhesion. These provide a balancing force during development that prevents overly exuberant adhesion, which would otherwise disrupt normal cellular and tissue morphogenesis. The strength of chemoattractive signals may be similarly modulated. Recent studies, described here, suggest that Down Syndrome Cell Adhesion Molecule (DSCAM), and closely related proteins such as DSCAML1, may play an important developmental role as such balancers in multiple systems.

15.
Dev Cell ; 22(1): 5-6, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22264725

ABSTRACT

Previous models of neuronal dendrite arborization suggested that contact-dependent self-avoidance between dendrite branches prevents self-crossings within the arbor. Two papers in Neuron show how integrin-mediated adhesion to the extracellular matrix restricts dendrites to a two-dimensional space to optimize this mechanism (Han et al., 2012; Kim et al., 2012).

16.
Neuron ; 74(2): 269-76, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22542181

ABSTRACT

The 22 γ-protocadherins (γ-Pcdhs) potentially specify thousands of distinct homophilic adhesive interactions in the brain. Neonatal lethality of mice lacking the Pcdh-γ gene cluster has, however, precluded analysis of many brain regions. Here, we use a conditional Pcdh-γ allele to restrict mutation to the cerebral cortex and find that, in contrast to other central nervous system phenotypes, loss of γ-Pcdhs in cortical neurons does not affect their survival or result in reduced synaptic density. Instead, mutant cortical neurons exhibit severely reduced dendritic arborization. Mutant cortices have aberrantly high levels of protein kinase C (PKC) activity and of phosphorylated (inactive) myristoylated alanine-rich C-kinase substrate, a PKC target that promotes arborization. Dendrite complexity can be rescued in Pcdh-γ mutant neurons by inhibiting PKC, its upstream activator phospholipase C, or the γ-Pcdh binding partner focal adhesion kinase. Our results reveal a distinct role for the γ-Pcdhs in cortical development and identify a signaling pathway through which they play this role.


Subject(s)
Cadherins/metabolism , Cerebral Cortex/cytology , Dendrites/genetics , Gene Expression Regulation, Developmental/genetics , Neurons/cytology , Signal Transduction/physiology , Age Factors , Animals , Animals, Newborn , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cadherin Related Proteins , Cadherins/genetics , Cells, Cultured , Cerebral Cortex/abnormalities , Focal Adhesion Kinase 1/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Myristoylated Alanine-Rich C Kinase Substrate , Protein Kinase C/metabolism , Sequence Deletion/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection/methods
17.
Dev Neurobiol ; 71(12): 1258-72, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21630473

ABSTRACT

In the developing nervous system, individual neurons must occupy appropriate positions within circuits. This requires that these neurons recognize and form connections with specific pre- and postsynaptic partners. Cellular recognition is also required for the spacing of cell bodies and the arborization of dendrites, factors that determine the inputs onto a given neuron. These issues are particularly evident in the retina, where different types of neurons are evenly spaced relative to other cells of the same type. This establishes a reiterated columnar circuitry resembling the insect retina. Establishing these mosaic patterns requires that cells of a given type (homotypic cells) be able to sense their neighbors. Therefore, both synaptic specificity and mosaic spacing require cellular identifiers. In synaptic specificity, recognition often occurs between different types of cells in a pre- and postsynaptic pairing. In mosaic spacing, recognition is often occurring between different cells of the same type, orhomotypic self-recognition. Dendritic arborization can require recognition of different neurites of the same cell, or isoneuronal self-recognition. The retina is an extremely amenable system for studying the molecular identifiers that drive these various forms of recognition. The different neuronal types in the retina are well defined, and the genetic tools for marking cell types are increasingly available. In this review we will summarize retinal anatomy and describe cell types in the retina and how they are defined. We will then describe the requirements of a recognition code and discuss newly emerging candidate molecular mechanisms for recognition that may meet these requirements.


Subject(s)
Neurons/classification , Neurons/physiology , Retina , Animals , Dendrites/physiology , Humans , Neurons/cytology , Retina/cytology , Retina/embryology , Retina/growth & development , Synapses/physiology , Visual Pathways/physiology
SELECTION OF CITATIONS
SEARCH DETAIL