1
|
Buyannemekh K, Villoutreix P, Bertrand V. Left/right asymmetrically expressed ephrin and Flamingo proteins regulate lateralized axon growth in C. elegans. Dev Biol 2025; 517:117-125. [PMID: 39341445 DOI: 10.1016/j.ydbio.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
While the nervous system of bilaterian animals is mainly left-right (L-R) symmetric at the anatomical level, some molecular and functional L-R asymmetries exist. However, the extent of these molecular asymmetries and their functional consequences remain poorly characterized. C. elegans allows to study L-R asymmetries in the nervous system with single-neuron resolution. We have previously shown that a neural bHLH transcription factor, HLH-16/Olig, is L-R asymmetrically expressed in the AIY neuron lineage and regulates AIY axon projections in a L-R asymmetric manner. Here, by combining a candidate approach and single-cell RNA sequencing data analysis, we identify the ephrin protein EFN-2 and the Flamingo protein FMI-1 as downstream targets of HLH-16 that are L-R asymmetrically expressed in the AIY lineage. We show that EFN-2 and FMI-1 collaborate in the L-R asymmetric regulation of axonal growth. EFN-2 may act via a non-canonical receptor of the L1CAM family, SAX-7. Our study reveals novel molecular L-R asymmetries in the C. elegans nervous system and their functional consequences.
Collapse
Affiliation(s)
- Khulganaa Buyannemekh
- Aix Marseille Univ, CNRS, IBDM, Turing Center for Living Systems, Marseille, France; Aix Marseille Univ, Université de Toulon, CNRS, LIS, Turing Centre for Living Systems, Marseille, France; Aix Marseille Univ, INSERM, MMG, Turing Centre for Living Systems, Marseille, France
| | - Paul Villoutreix
- Aix Marseille Univ, Université de Toulon, CNRS, LIS, Turing Centre for Living Systems, Marseille, France; Aix Marseille Univ, INSERM, MMG, Turing Centre for Living Systems, Marseille, France
| | - Vincent Bertrand
- Aix Marseille Univ, CNRS, IBDM, Turing Center for Living Systems, Marseille, France.
| |
Collapse
|
2
|
Moseley-Alldredge M, Aragón C, Vargus M, Alley D, Somia N, Chen L. The L1CAM SAX-7 is an antagonistic modulator of Erk Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.14.613091. [PMID: 39345534 PMCID: PMC11429911 DOI: 10.1101/2024.09.14.613091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
L1CAMs are immunoglobulin superfamily cell adhesion molecules that ensure proper nervous system development and function. In addition to being associated with the autism and schizophrenia spectrum disorders, mutations in the L1CAM family of genes also underlie distinct developmental syndromes with neurological conditions, such as intellectual disability, spastic paraplegia, hypotonia and congenital hydrocephalus. Studies in both vertebrate and invertebrate model organisms have established conserved neurodevelopmental roles for L1CAMs; these include axon guidance, dendrite morphogenesis, synaptogenesis, and maintenance of neural architecture, among others. In Caenorhabditis elegans , L1CAMs, encoded by the sax-7 gene, are required for coordinated locomotion. We previously uncovered a genetic interaction between sax-7 and components of synaptic vesicle cycle, revealing a non-developmental role for sax-7 in regulating synaptic activity. More recently, we determined that sax-7 also genetically interacts with extracellular signal-related kinase (ERK) signaling in controlling coordinated locomotion. C. elegans ERK, encoded by the mpk-1 gene, is a serine/threonine protein kinase belonging to the mitogen-activated protein kinase (MAPK) family that governs multiple aspects of animal development and cellular homeostasis. Here, we show this genetic interaction between sax-7 and mpk-1 occurs not only in cholinergic neurons for coordinated locomotion, but also extends outside the nervous system, revealing novel roles for SAX-7/L1CAM in non-neuronal processes, including vulval development. Our genetic findings in both the nervous system and developing vulva are consistent with SAX-7/L1CAM acting as an antagonistic modulator of ERK signaling.
Collapse
|
3
|
Cebul ER, Marivin A, Wexler LR, Perrat PN, Bénard CY, Garcia-Marcos M, Heiman MG. SAX-7/L1CAM acts with the adherens junction proteins MAGI-1, HMR-1/Cadherin, and AFD-1/Afadin to promote glial-mediated dendrite extension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.11.575259. [PMID: 38260503 PMCID: PMC10802611 DOI: 10.1101/2024.01.11.575259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Adherens junctions (AJs) are a fundamental organizing structure for multicellular life. Although AJs are studied mainly in epithelia, their core function - stabilizing cell contacts by coupling adhesion molecules to the cytoskeleton - is important in diverse tissues. We find that two C. elegans sensory neurons, URX and BAG, require conserved AJ proteins for dendrite morphogenesis. We previously showed that URX and BAG dendrites attach to the embryonic nose via the adhesion molecule SAX-7/L1CAM, acting both in neurons and glia, and then extend by stretch during embryo elongation. Here, we find that a PDZ-binding motif (PB) in the SAX-7 cytoplasmic tail acts with other interaction motifs to promote dendrite extension. Using pull-down assays, we find that the SAX-7 PB binds the multi-PDZ scaffolding protein MAGI-1, which bridges it to the cadherin-catenin complex protein HMP-2/β-catenin. Using cell-specific rescue and depletion, we find that both MAGI-1 and HMR-1/Cadherin act in glia to non-autonomously promote dendrite extension. Double mutant analysis indicates that each protein can act independently of SAX-7, suggesting a multivalent adhesion complex. The SAX-7 PB motif also binds AFD-1/Afadin, loss of which further enhances sax-7 BAG dendrite defects. As MAGI-1, HMR-1, and AFD-1 are all found in epithelial AJs, we propose that an AJ-like complex in glia promotes dendrite extension.
Collapse
Affiliation(s)
- Elizabeth R. Cebul
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- Present address: Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Arthur Marivin
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA, Boston University School of Medicine, Boston, MA 02118, USA
| | - Leland R. Wexler
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Paola N. Perrat
- Department of Biological Sciences, CERMO-FC Research Center, Universite du Québec à Montréal, Montreál, QC, Canada
| | - Claire Y. Bénard
- Department of Biological Sciences, CERMO-FC Research Center, Universite du Québec à Montréal, Montreál, QC, Canada
| | - Mikel Garcia-Marcos
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Biology, College of Arts & Sciences, Boston University, Boston, MA 02115, USA
| | - Maxwell G. Heiman
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
4
|
When is a neuron like an epithelial cell. Dev Biol 2022; 489:161-164. [DOI: 10.1016/j.ydbio.2022.06.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/18/2022] [Indexed: 11/30/2022]
|
5
|
Lillis M, Zaccardi NJ, Heiman MG. Axon-dendrite and apical-basolateral sorting in a single neuron. Genetics 2022; 221:iyac036. [PMID: 35244146 PMCID: PMC9071548 DOI: 10.1093/genetics/iyac036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Cells are highly organized machines with functionally specialized compartments. For example, membrane proteins are localized to axons or dendrites in neurons and to apical or basolateral surfaces in epithelial cells. Interestingly, many sensory cells-including vertebrate photoreceptors and olfactory neurons-exhibit both neuronal and epithelial features. Here, we show that Caenorhabditis elegans amphid neurons simultaneously exhibit axon-dendrite sorting like a neuron and apical-basolateral sorting like an epithelial cell. The distal ∼5-10 µm of the dendrite is apical, while the remainder of the dendrite, soma, and axon are basolateral. To determine how proteins are sorted among these compartments, we studied the localization of the conserved adhesion molecule SAX-7/L1CAM. Using minimal synthetic transmembrane proteins, we found that the 91-aa cytoplasmic tail of SAX-7 is necessary and sufficient to direct basolateral localization. Basolateral localization can be fully recapitulated using either of 2 short (10-aa or 19-aa) tail sequences that, respectively, resemble dileucine and Tyr-based motifs known to mediate sorting in mammalian epithelia. The Tyr-based motif is conserved in human L1CAM but had not previously been assigned a function. Disrupting key residues in either sequence leads to apical localization, while "improving" them to match epithelial sorting motifs leads to axon-only localization. Indeed, changing only 2 residues in a short motif is sufficient to redirect the protein between apical, basolateral, and axonal localization. Our results demonstrate that axon-dendrite and apical-basolateral sorting pathways can coexist in a single cell, and suggest that subtle changes to short sequence motifs are sufficient to redirect proteins between these pathways.
Collapse
Affiliation(s)
- Monique Lillis
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children’s Hospital, Boston, MA 02115, USA
| | - Nathan J Zaccardi
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children’s Hospital, Boston, MA 02115, USA
| | - Maxwell G Heiman
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
6
|
Moseley-Alldredge M, Sheoran S, Yoo H, O’Keefe C, Richmond JE, Chen L. A role for the Erk MAPK pathway in modulating SAX-7/L1CAM-dependent locomotion in Caenorhabditis elegans. Genetics 2022; 220:iyab215. [PMID: 34849872 PMCID: PMC9097276 DOI: 10.1093/genetics/iyab215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 11/11/2021] [Indexed: 01/13/2023] Open
Abstract
L1CAMs are immunoglobulin cell adhesion molecules that function in nervous system development and function. Besides being associated with autism and schizophrenia spectrum disorders, impaired L1CAM function also underlies the X-linked L1 syndrome, which encompasses a group of neurological conditions, including spastic paraplegia and congenital hydrocephalus. Studies on vertebrate and invertebrate L1CAMs established conserved roles that include axon guidance, dendrite morphogenesis, synapse development, and maintenance of neural architecture. We previously identified a genetic interaction between the Caenorhabditis elegans L1CAM encoded by the sax-7 gene and RAB-3, a GTPase that functions in synaptic neurotransmission; rab-3; sax-7 mutant animals exhibit synthetic locomotion abnormalities and neuronal dysfunction. Here, we show that this synergism also occurs when loss of SAX-7 is combined with mutants of other genes encoding key players of the synaptic vesicle (SV) cycle. In contrast, sax-7 does not interact with genes that function in synaptogenesis. These findings suggest a postdevelopmental role for sax-7 in the regulation of synaptic activity. To assess this possibility, we conducted electrophysiological recordings and ultrastructural analyses at neuromuscular junctions; these analyses did not reveal obvious synaptic abnormalities. Lastly, based on a forward genetic screen for suppressors of the rab-3; sax-7 synthetic phenotypes, we determined that mutants in the ERK Mitogen-activated Protein Kinase (MAPK) pathway can suppress the rab-3; sax-7 locomotion defects. Moreover, we established that Erk signaling acts in a subset of cholinergic neurons in the head to promote coordinated locomotion. In combination, these results suggest a modulatory role for Erk MAPK in L1CAM-dependent locomotion in C. elegans.
Collapse
Affiliation(s)
- Melinda Moseley-Alldredge
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Seema Sheoran
- Department of Biological Sciences, University of Illinois, Chicago, IL 60607, USA
| | - Hayoung Yoo
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Calvin O’Keefe
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Janet E Richmond
- Department of Biological Sciences, University of Illinois, Chicago, IL 60607, USA
| | - Lihsia Chen
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
7
|
Desse VE, Blanchette CR, Nadour M, Perrat P, Rivollet L, Khandekar A, Bénard CY. Neuronal post-developmentally acting SAX-7S/L1CAM can function as cleaved fragments to maintain neuronal architecture in C. elegans. Genetics 2021; 218:6296841. [PMID: 34115111 DOI: 10.1093/genetics/iyab086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 05/24/2021] [Indexed: 01/09/2023] Open
Abstract
Whereas remarkable advances have uncovered mechanisms that drive nervous system assembly, the processes responsible for the lifelong maintenance of nervous system architecture remain poorly understood. Subsequent to its establishment during embryogenesis, neuronal architecture is maintained throughout life in the face of the animal's growth, maturation processes, the addition of new neurons, body movements, and aging. The C. elegans protein SAX-7, homologous to the vertebrate L1 protein family of neural adhesion molecules, is required for maintaining the organization of neuronal ganglia and fascicles after their successful initial embryonic development. To dissect the function of sax-7 in neuronal maintenance, we generated a null allele and sax-7S-isoform-specific alleles. We find that the null sax-7(qv30) is, in some contexts, more severe than previously described mutant alleles, and that the loss of sax-7S largely phenocopies the null, consistent with sax-7S being the key isoform in neuronal maintenance. Using a sfGFP::SAX-7S knock-in, we observe sax-7S to be predominantly expressed across the nervous system, from embryogenesis to adulthood. Yet, its role in maintaining neuronal organization is ensured by post-developmentally acting SAX-7S, as larval transgenic sax-7S(+) expression alone is sufficient to profoundly rescue the null mutants' neuronal maintenance defects. Moreover, the majority of the protein SAX-7 appears to be cleaved, and we show that these cleaved SAX-7S fragments together, not individually, can fully support neuronal maintenance. These findings contribute to our understanding of the role of the conserved protein SAX-7/L1CAM in long-term neuronal maintenance, and may help decipher processes that go awry in some neurodegenerative conditions.
Collapse
Affiliation(s)
- Virginie E Desse
- Department of Biological Sciences, CERMO-FC Research Center, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
| | - Cassandra R Blanchette
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Malika Nadour
- Department of Biological Sciences, CERMO-FC Research Center, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
| | - Paola Perrat
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lise Rivollet
- Department of Biological Sciences, CERMO-FC Research Center, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
| | - Anagha Khandekar
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Claire Y Bénard
- Department of Biological Sciences, CERMO-FC Research Center, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
8
|
Sherry T, Handley A, Nicholas HR, Pocock R. Harmonization of L1CAM expression facilitates axon outgrowth and guidance of a motor neuron. Development 2020; 147:dev.193805. [PMID: 32994172 DOI: 10.1242/dev.193805] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/18/2020] [Indexed: 12/28/2022]
Abstract
Brain development requires precise regulation of axon outgrowth, guidance and termination by multiple signaling and adhesion molecules. How the expression of these neurodevelopmental regulators is transcriptionally controlled is poorly understood. The Caenorhabditis elegans SMD motor neurons terminate axon outgrowth upon sexual maturity and partially retract their axons during early adulthood. Here we show that C-terminal binding protein 1 (CTBP-1), a transcriptional corepressor, is required for correct SMD axonal development. Loss of CTBP-1 causes multiple defects in SMD axon development: premature outgrowth, defective guidance, delayed termination and absence of retraction. CTBP-1 controls SMD axon guidance by repressing the expression of SAX-7, an L1 cell adhesion molecule (L1CAM). CTBP-1-regulated repression is crucial because deregulated SAX-7/L1CAM causes severely aberrant SMD axons. We found that axonal defects caused by deregulated SAX-7/L1CAM are dependent on a distinct L1CAM, called LAD-2, which itself plays a parallel role in SMD axon guidance. Our results reveal that harmonization of L1CAM expression controls the development and maturation of a single neuron.
Collapse
Affiliation(s)
- Tessa Sherry
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Ava Handley
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Hannah R Nicholas
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Roger Pocock
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| |
Collapse
|
9
|
Cebul ER, McLachlan IG, Heiman MG. Dendrites with specialized glial attachments develop by retrograde extension using SAX-7 and GRDN-1. Development 2020; 147:dev.180448. [PMID: 31988188 DOI: 10.1242/dev.180448] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 01/07/2020] [Indexed: 12/18/2022]
Abstract
Dendrites develop elaborate morphologies in concert with surrounding glia, but the molecules that coordinate dendrite and glial morphogenesis are mostly unknown. C. elegans offers a powerful model for identifying such factors. Previous work in this system examined dendrites and glia that develop within epithelia, similar to mammalian sense organs. Here, we focus on the neurons BAG and URX, which are not part of an epithelium but instead form membranous attachments to a single glial cell at the nose, reminiscent of dendrite-glia contacts in the mammalian brain. We show that these dendrites develop by retrograde extension, in which the nascent dendrite endings anchor to the presumptive nose and then extend by stretching during embryo elongation. Using forward genetic screens, we find that dendrite development requires the adhesion protein SAX-7/L1CAM and the cytoplasmic protein GRDN-1/CCDC88C to anchor dendrite endings at the nose. SAX-7 acts in neurons and glia, while GRDN-1 acts in glia to non-autonomously promote dendrite extension. Thus, this work shows how glial factors can help to shape dendrites, and identifies a novel molecular mechanism for dendrite growth by retrograde extension.
Collapse
Affiliation(s)
- Elizabeth R Cebul
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Ian G McLachlan
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Maxwell G Heiman
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| |
Collapse
|
10
|
Zheleva A, Gómez-Orte E, Sáenz-Narciso B, Ezcurra B, Kassahun H, de Toro M, Miranda-Vizuete A, Schnabel R, Nilsen H, Cabello J. Reduction of mRNA export unmasks different tissue sensitivities to low mRNA levels during Caenorhabditis elegans development. PLoS Genet 2019; 15:e1008338. [PMID: 31525188 PMCID: PMC6762213 DOI: 10.1371/journal.pgen.1008338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 09/26/2019] [Accepted: 07/31/2019] [Indexed: 12/25/2022] Open
Abstract
Animal development requires the execution of specific transcriptional programs in different sets of cells to build tissues and functional organs. Transcripts are exported from the nucleus to the cytoplasm where they are translated into proteins that, ultimately, carry out the cellular functions. Here we show that in Caenorhabditis elegans, reduction of mRNA export strongly affects epithelial morphogenesis and germline proliferation while other tissues remain relatively unaffected. Epithelialization and gamete formation demand a large number of transcripts in the cytoplasm for the duration of these processes. In addition, our findings highlight the existence of a regulatory feedback mechanism that activates gene expression in response to low levels of cytoplasmic mRNA. We expand the genetic characterization of nuclear export factor NXF-1 to other members of the mRNA export pathway to model mRNA export and recycling of NXF-1 back to the nucleus. Our model explains how mutations in genes involved in general processes, such as mRNA export, may result in tissue-specific developmental phenotypes.
Collapse
Affiliation(s)
- Angelina Zheleva
- CIBIR (Center for Biomedical Research of La Rioja), Logroño, La Rioja, Spain
| | - Eva Gómez-Orte
- CIBIR (Center for Biomedical Research of La Rioja), Logroño, La Rioja, Spain
| | | | - Begoña Ezcurra
- CIBIR (Center for Biomedical Research of La Rioja), Logroño, La Rioja, Spain
| | - Henok Kassahun
- Department of Clinical Molecular Biology, Institute of Clinical Medicine, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - María de Toro
- CIBIR (Center for Biomedical Research of La Rioja), Logroño, La Rioja, Spain
| | - Antonio Miranda-Vizuete
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Ralf Schnabel
- Institute of Genetics, Technische Universität Braunschweig, Germany
| | - Hilde Nilsen
- Department of Clinical Molecular Biology, Institute of Clinical Medicine, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Juan Cabello
- CIBIR (Center for Biomedical Research of La Rioja), Logroño, La Rioja, Spain
| |
Collapse
|
11
|
Weber T, Stephan R, Moreno E, Pielage J. The Ankyrin Repeat Domain Controls Presynaptic Localization of Drosophila Ankyrin2 and Is Essential for Synaptic Stability. Front Cell Dev Biol 2019; 7:148. [PMID: 31475145 PMCID: PMC6703079 DOI: 10.3389/fcell.2019.00148] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/16/2019] [Indexed: 01/24/2023] Open
Abstract
The structural integrity of synaptic connections critically depends on the interaction between synaptic cell adhesion molecules (CAMs) and the underlying actin and microtubule cytoskeleton. This interaction is mediated by giant Ankyrins, that act as specialized adaptors to establish and maintain axonal and synaptic compartments. In Drosophila, two giant isoforms of Ankyrin2 (Ank2) control synapse stability and organization at the larval neuromuscular junction (NMJ). Both Ank2-L and Ank2-XL are highly abundant in motoneuron axons and within the presynaptic terminal, where they control synaptic CAMs distribution and organization of microtubules. Here, we address the role of the conserved N-terminal ankyrin repeat domain (ARD) for subcellular localization and function of these giant Ankyrins in vivo. We used a P[acman] based rescue approach to generate deletions of ARD subdomains, that contain putative binding sites of interacting transmembrane proteins. We show that specific subdomains control synaptic but not axonal localization of Ank2-L. These domains contain binding sites to L1-family member CAMs, and we demonstrate that these regions are necessary for the organization of synaptic CAMs and for the control of synaptic stability. In contrast, presynaptic Ank2-XL localization only partially depends on the ARD but strictly requires the presynaptic presence of Ank2-L demonstrating a critical co-dependence of the two isoforms at the NMJ. Ank2-XL dependent control of microtubule organization correlates with presynaptic abundance of the protein and is thus only partially affected by ARD deletions. Together, our data provides novel insights into the synaptic targeting of giant Ankyrins with relevance for the control of synaptic plasticity and maintenance.
Collapse
Affiliation(s)
- Tobias Weber
- Department of Zoology and Neurobiology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Raiko Stephan
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Eliza Moreno
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Jan Pielage
- Department of Zoology and Neurobiology, University of Kaiserslautern, Kaiserslautern, Germany
| |
Collapse
|
12
|
Rahe D, Carrera I, Cosmanescu F, Hobert O. An isoform-specific allele of the sax-7 locus. MICROPUBLICATION BIOLOGY 2019; 2019. [PMID: 32550459 PMCID: PMC7252388 DOI: 10.17912/micropub.biology.000092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Dylan Rahe
- Department of Biological Sciences, Columbia University, New York, NY, USA, Howard Hughes Medical Institute
| | - Ines Carrera
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, NY, USA.,Current Address: School of Chemistry, University of the Republic, Montevideo, Uruguay
| | - Filip Cosmanescu
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, NY, USA
| | - Oliver Hobert
- Department of Biological Sciences, Columbia University, New York, NY, USA, Howard Hughes Medical Institute.,Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
13
|
Gordon KL, Payne SG, Linden-High LM, Pani AM, Goldstein B, Hubbard EJA, Sherwood DR. Ectopic Germ Cells Can Induce Niche-like Enwrapment by Neighboring Body Wall Muscle. Curr Biol 2019; 29:823-833.e5. [PMID: 30799241 DOI: 10.1016/j.cub.2019.01.056] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 12/20/2022]
Abstract
Niche cell enwrapment of stem cells and their differentiating progeny is common and provides a specialized signaling and protective environment. Elucidating the mechanisms underlying enwrapment behavior has important basic and clinical significance in not only understanding how niches are formed and maintained but also how they can be engineered and how they are misregulated in human pathologies, such as cancer. Previous work in C. elegans found that, when germ cells, which are enwrapped by somatic gonadal niche cells, are freed into the body cavity, they embed into other tissues. We investigated this phenomenon using live-cell imaging and discovered that ectopic germ cells preferentially induce body-wall muscle to extend cellular processes that enwrap the germ cells, the extent of which was strikingly similar to the distal tip cell (DTC)-germ stem cell niche. Enwrapment was specific for escaped germ cells, and genetic analysis revealed it did not depend on pathways that control cell death and engulfment or muscle arm extension. Instead, using a large-scale RNAi screen and GFP knockin strains, we discovered that the enwrapping behavior of muscle relied upon the same suite of cell-cell adhesion molecules that functioned in the endogenous niche: the C. elegans E-cadherin HMR-1, its intracellular associates α-catenin (HMP-1) and β-catenin (HMP-2), and the L1CAM protein SAX-7. This ectopic niche-like behavior resembles the seed-and-soil model of cancer metastasis and offers a new model to understand factors regulating ectopic niche formation.
Collapse
Affiliation(s)
- Kacy L Gordon
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Sara G Payne
- Department of Biology, Duke University, Durham, NC 27708, USA
| | | | - Ariel M Pani
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bob Goldstein
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - E Jane Albert Hubbard
- Skirball Institute of Biomolecular Medicine, Departments of Cell Biology and Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - David R Sherwood
- Department of Biology, Duke University, Durham, NC 27708, USA; Regeneration Next, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
14
|
Philbrook A, Ramachandran S, Lambert CM, Oliver D, Florman J, Alkema MJ, Lemons M, Francis MM. Neurexin directs partner-specific synaptic connectivity in C. elegans. eLife 2018; 7:35692. [PMID: 30039797 PMCID: PMC6057746 DOI: 10.7554/elife.35692] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/21/2018] [Indexed: 01/14/2023] Open
Abstract
In neural circuits, individual neurons often make projections onto multiple postsynaptic partners. Here, we investigate molecular mechanisms by which these divergent connections are generated, using dyadic synapses in C. elegans as a model. We report that C. elegans nrx-1/neurexin directs divergent connectivity through differential actions at synapses with partnering neurons and muscles. We show that cholinergic outputs onto neurons are, unexpectedly, located at previously undefined spine-like protrusions from GABAergic dendrites. Both these spine-like features and cholinergic receptor clustering are strikingly disrupted in the absence of nrx-1. Excitatory transmission onto GABAergic neurons, but not neuromuscular transmission, is also disrupted. Our data indicate that NRX-1 located at presynaptic sites specifically directs postsynaptic development in GABAergic neurons. Our findings provide evidence that individual neurons can direct differential patterns of connectivity with their post-synaptic partners through partner-specific utilization of synaptic organizers, offering a novel view into molecular control of divergent connectivity. Nervous systems are complex networks of interconnected cells called neurons. These networks vary in size from a few hundred cells in worms, to tens of billions in the human brain. Within these networks, each individual neuron forms connections – called synapses – with many others. But these partner neurons are not necessarily alike. In fact, they may be different cell types. How neurons form distinct connections with different partner cells remains unclear. Part of the answer may lie in specialized proteins called cell adhesion molecules. These proteins occur on the cell surface and enable neurons to recognize one another. This helps ensure that the cells form appropriate connections via synapses. Cell adhesion molecules are therefore also known as synaptic organizers. Philbrook et al. have now examined the role of synaptic organizers in wiring up the nervous system of the nematode worm and model organism Caenorhabditis elegans. Motor neurons form connections with two types of partner cell: muscle cells and neurons. Philbrook et al. screened C. elegans that have mutations in genes encoding various synaptic organizers. This revealed that a protein called neurexin must be present for motor neurons to form synapses with other neurons. By contrast, neurexin is not required for the same neurons to establish synapses with muscles. Philbrook et al. found that neuron-to-neuron synapses arise at specialized finger-like projections. These resemble the dendritic spines at which synapses form in the brains of mammals, and had not been previously identified in C. elegans. In worms that lack neurexin, these spine-like structures do not form correctly, disrupting the formation of neuron-to-neuron connections. Previous work has implicated neurexin in synapse formation in the mammalian brain. But this is the first study to reveal a role for neurexin in establishing partner-specific synaptic connections. Mutations in synaptic organizers, including neurexin, contribute to disorders of brain development. These include schizophrenia and autism spectrum disorders. Learning more about how neurexin helps establish specific synaptic connections may help us understand how these disorders arise.
Collapse
Affiliation(s)
- Alison Philbrook
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Shankar Ramachandran
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Christopher M Lambert
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Devyn Oliver
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Jeremy Florman
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Mark J Alkema
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Michele Lemons
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States.,Department of Natural Sciences, Assumption College, Worcester, United States
| | - Michael M Francis
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| |
Collapse
|
15
|
Ma YC, Zhang L, Dai LL, Khan RU, Zou CG. mir-67 regulates P. aeruginosa avoidance behavior in C. elegans. Biochem Biophys Res Commun 2017; 494:120-125. [PMID: 29050943 DOI: 10.1016/j.bbrc.2017.10.069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 10/14/2017] [Indexed: 01/27/2023]
Abstract
Pathogen avoidance behaviors are found throughout the animal kingdom and are important for animal's survival in nature. As a free-living nematode, C. elegans is exposed to a variety of microorganisms, including toxic or pathogenic bacteria, in soil. C. elegans can develop efficient avoidance responses to pathogenic bacteria to minimize the infection risk. However, the role of microRNAs (miRNAs) in pathogen avoidance in C. elegans remains unclear. In this report, we showed that the miRNA mir-67 was involved in a behavioral avoidance response to P. aeruginosa PA14. Exposure to P. aeruginosa PA14 induced the expression of mir-67 in worms. mir-67(n4899) mutants exhibited a reduced ability to avoid P. aeruginosa PA14. By combining quantitative proteomic analysis with miRNA target prediction algorithms, we identified SAX-7/L1CAM, which is transmembrane cell adhesion receptor molecule, as the target of mir-67. Silencing of sax-7 by RNAi on mir-67 mutants rescued avoidance behavioral. Our data demonstrate that the mir-67-SAX-7 pathway modulate the behavioral avoidance response to pathogens, thus providing a new perspective in the role of miRNAs in host-microbe interactions.
Collapse
Affiliation(s)
- Yi-Cheng Ma
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan 650091, China
| | - Lu Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan 650091, China
| | - Li-Li Dai
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan 650091, China
| | - Rahat Ullah Khan
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan 650091, China
| | - Cheng-Gang Zou
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan 650091, China.
| |
Collapse
|
16
|
Schmeisser K, Parker JA. Worms on the spectrum - C. elegans models in autism research. Exp Neurol 2017; 299:199-206. [PMID: 28434869 DOI: 10.1016/j.expneurol.2017.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/10/2017] [Accepted: 04/18/2017] [Indexed: 12/13/2022]
Abstract
The small non-parasitic nematode Caenorhabditis elegans is widely used in neuroscience thanks to its well-understood development and lineage of the nervous system. Furthermore, C. elegans has been used to model many human developmental and neurological conditions to better understand disease mechanisms and identify potential therapeutic strategies. Autism spectrum disorder (ASD) is the most prevalent of all neurodevelopmental disorders, and the C. elegans system may provide opportunities to learn more about this complex disorder. Since basic cell biology and biochemistry of the C. elegans nervous system is generally very similar to mammals, cellular or molecular phenotypes can be investigated, along with a repertoire of behaviours. For instance, worms have contributed greatly to the understanding of mechanisms underlying mutations in genes coding for synaptic proteins such as neuroligin and neurexin. Using worms to model neurodevelopmental disorders like ASD is an emerging topic that harbours great, untapped potential. This review summarizes the numerous contributions of C. elegans to the field of neurodevelopment and introduces the nematode system as a potential research tool to study essential roles of genes associated with ASD.
Collapse
Affiliation(s)
- Kathrin Schmeisser
- Centre de Recherche du Centre Hospitalier de l'Université de Montreál (CRCHUM), 900 St-Denis Street, Montreál, Queb́ec H2X 0A9, Canada
| | - J Alex Parker
- Centre de Recherche du Centre Hospitalier de l'Université de Montreál (CRCHUM), 900 St-Denis Street, Montreál, Queb́ec H2X 0A9, Canada; Department of Neuroscience, Université de Montreál, 2960 Chemin de la Tour, Montreál, Queb́ec H3T 1J4, Canada.
| |
Collapse
|
17
|
Schmeisser K, Fardghassemi Y, Parker JA. A rapid chemical-genetic screen utilizing impaired movement phenotypes in C. elegans: Input into genetics of neurodevelopmental disorders. Exp Neurol 2017; 293:101-114. [PMID: 28373024 DOI: 10.1016/j.expneurol.2017.03.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 11/17/2022]
Abstract
Autism spectrum disorder (ASD) is the most common neurodevelopmental disorder with a constantly increasing prevalence. Model organisms may be tools to identify underlying cellular and molecular mechanisms, as well as aid the discovery and development of novel therapeutic approaches. A simple animal such as the nematode Caenorhabditis elegans may provide insights into the extreme complexity of ASD genetics. Despite its potential, using C. elegans in ASD research is a controversial approach and has not yet been used extensively in this context. In this study, we present a screening approach of potential C. elegans mutants as potential ASD models. We screened these mutants for motor-deficiency phenotypes, which can be exploited to study underlying mechanisms of the disorder. Selected motor-deficient mutants were then used in a comprehensive drug screen of over 3900 compounds, including many FDA-approved and natural molecules, that were analyzed for their ability to suppress motility defects caused by ASD-associated gene orthologues. This genetic-chemical approach, i.e. establishing C. elegans models for ASD and screening of a well-characterized compound library, might be a promising first step to understand the mechanisms of how gene variations cause neuronal dysfunction, leading to ASD and other neurological disorders. Positively acting compounds could also be promising candidates for preclinical studies.
Collapse
Affiliation(s)
- Kathrin Schmeisser
- Centre de Recherche du Centre Hospitalier de l'Université de Montreál (CRCHUM), 900 St-Denis Street, Montreál, Québec H2X 0A9, Canada
| | - Yasmin Fardghassemi
- Centre de Recherche du Centre Hospitalier de l'Université de Montreál (CRCHUM), 900 St-Denis Street, Montreál, Québec H2X 0A9, Canada; Department of Biochemistry and Molecular Medicine, Université de Montreál, 2960 Chemin de la Tour, Montreál, Québec H3T 1J4, Canada
| | - J Alex Parker
- Centre de Recherche du Centre Hospitalier de l'Université de Montreál (CRCHUM), 900 St-Denis Street, Montreál, Québec H2X 0A9, Canada; Department of Neuroscience, Université de Montreál, 2960 Chemin de la Tour, Montreál, Québec H3T 1J4, Canada.
| |
Collapse
|
18
|
Zou W, Shen A, Dong X, Tugizova M, Xiang YK, Shen K. A multi-protein receptor-ligand complex underlies combinatorial dendrite guidance choices in C. elegans. eLife 2016; 5. [PMID: 27705746 PMCID: PMC5079751 DOI: 10.7554/elife.18345] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 10/03/2016] [Indexed: 11/24/2022] Open
Abstract
Ligand receptor interactions instruct axon guidance during development. How dendrites are guided to specific targets is less understood. The C. elegans PVD sensory neuron innervates muscle-skin interface with its elaborate dendritic branches. Here, we found that LECT-2, the ortholog of leukocyte cell-derived chemotaxin-2 (LECT2), is secreted from the muscles and required for muscle innervation by PVD. Mosaic analyses showed that LECT-2 acted locally to guide the growth of terminal branches. Ectopic expression of LECT-2 from seam cells is sufficient to redirect the PVD dendrites onto seam cells. LECT-2 functions in a multi-protein receptor-ligand complex that also contains two transmembrane ligands on the skin, SAX-7/L1CAM and MNR-1, and the neuronal transmembrane receptor DMA-1. LECT-2 greatly enhances the binding between SAX-7, MNR-1 and DMA-1. The activation of DMA-1 strictly requires all three ligands, which establishes a combinatorial code to precisely target and pattern dendritic arbors. DOI:http://dx.doi.org/10.7554/eLife.18345.001
Collapse
Affiliation(s)
- Wei Zou
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Ao Shen
- Department of Pharmacology, University of California, Davis, Davis, United States
| | - Xintong Dong
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Madina Tugizova
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Yang K Xiang
- Department of Pharmacology, University of California, Davis, Davis, United States
| | - Kang Shen
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| |
Collapse
|
19
|
Dong X, Chiu H, Park YJ, Zou W, Zou Y, Özkan E, Chang C, Shen K. Precise regulation of the guidance receptor DMA-1 by KPC-1/Furin instructs dendritic branching decisions. eLife 2016; 5. [PMID: 26974341 PMCID: PMC4811766 DOI: 10.7554/elife.11008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 02/26/2016] [Indexed: 01/15/2023] Open
Abstract
Extracellular adhesion molecules and their neuronal receptors guide the growth and branching of axons and dendrites. Growth cones are attracted to intermediate targets, but they must switch their response upon arrival so that they can move away and complete the next stage of growth. Here, we show that KPC-1, a C. elegans Furin homolog, regulates the level of the branching receptor DMA-1 on dendrites by targeting it to late endosomes. In kpc-1 mutants, the level of DMA-1 is abnormally high on dendrites, resulting in trapping of dendrites at locations where a high level of the cognate ligand, the adhesion molecule SAX-7/L1, is present. The misregulation of DMA-1 also causes dendritic self-avoidance defects. Thus, precise regulation of guidance receptors creates flexibility of responses to guidance signals and is critical for neuronal morphogenesis. DOI:http://dx.doi.org/10.7554/eLife.11008.001 Neurons are the principal cells in the nervous system that send and receive information. A vast network of neurons helps transmit information throughout the brain and body. The end of the neuron that receives messages forms branched structures called dendrites, the shapes of which determine the signals the neuron receives. Therefore, establishing the correct shape of the dendrites is critical for the neurons to work correctly. As dendrites grow during development, signals from the environment tell them where to branch and where to stop. For example, the neurons that transmit information about touch respond to signals from skin cells to guide the growth of their dendrites. These signals bind to receptor proteins on the surface of the neuron. However, the environment around the neurons also contains many guidance signals that the neurons must ignore. Dong et al. now show that touch neurons control how they respond to signals by adjusting the abundance of the receptors on their surface. First, genetic mutations were identified that distort the shape of the dendrites of touch-sensing neurons in a simple worm called Caenorhabditis elegans. These neurons lacked the equivalent of an enzyme called Furin and had abnormally high amounts of a receptor protein called DMA-1 on their surfaces. This suggests that controlling the receptor level on dendrites creates flexibility in the guidance choices of dendrites. Furin usually cuts up proteins. However, Dong et al. found that Furin prevents DMA-1 from inserting into the membrane of neurons by binding to the receptors and sending them to the lysosomes, cellular compartments where proteins are destroyed. Reducing the number of receptors at the surface of the cell in this way prevents the neuron from responding to the guidance signals at wrong locations. In the future, more studies are needed to understand how the neuron checks and balances this process and how it eventually is turned off. DOI:http://dx.doi.org/10.7554/eLife.11008.002
Collapse
Affiliation(s)
- Xintong Dong
- Department of Biology, Stanford University, Stanford, United States.,Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Hui Chiu
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, United States
| | - Yeonhee Jenny Park
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, United States
| | - Wei Zou
- Department of Biology, Stanford University, Stanford, United States.,Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Yan Zou
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, United States
| | - Engin Özkan
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, United States
| | - Chieh Chang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, United States
| | - Kang Shen
- Department of Biology, Stanford University, Stanford, United States.,Howard Hughes Medical Institute, Stanford University, Stanford, United States
| |
Collapse
|
20
|
Dong B, Moseley-Alldredge M, Schwieterman AA, Donelson CJ, McMurry JL, Hudson ML, Chen L. EFN-4 functions in LAD-2-mediated axon guidance in Caenorhabditis elegans. Development 2016; 143:1182-91. [PMID: 26903502 DOI: 10.1242/dev.128934] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 02/12/2016] [Indexed: 11/20/2022]
Abstract
During development of the nervous system, growing axons rely on guidance molecules to direct axon pathfinding. A well-characterized family of guidance molecules are the membrane-associated ephrins, which together with their cognate Eph receptors, direct axon navigation in a contact-mediated fashion. InC. elegans, the ephrin-Eph signaling system is conserved and is best characterized for their roles in neuroblast migration during early embryogenesis. This study demonstrates a role for the C. elegans ephrin EFN-4 in axon guidance. We provide both genetic and biochemical evidence that is consistent with the C. elegans divergent L1 cell adhesion molecule LAD-2 acting as a non-canonical ephrin receptor to EFN-4 to promote axon guidance. We also show that EFN-4 probably functions as a diffusible factor because EFN-4 engineered to be soluble can promote LAD-2-mediated axon guidance. This study thus reveals a potential additional mechanism for ephrins in regulating axon guidance and expands the repertoire of receptors by which ephrins can signal.
Collapse
Affiliation(s)
- Bingyun Dong
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Melinda Moseley-Alldredge
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alicia A Schwieterman
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA
| | - Cory J Donelson
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA
| | - Jonathan L McMurry
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA
| | - Martin L Hudson
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA
| | - Lihsia Chen
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
21
|
An Adaptable Spectrin/Ankyrin-Based Mechanism for Long-Range Organization of Plasma Membranes in Vertebrate Tissues. CURRENT TOPICS IN MEMBRANES 2015; 77:143-84. [PMID: 26781832 DOI: 10.1016/bs.ctm.2015.10.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Ankyrins are membrane-associated proteins that together with their spectrin partners are responsible for micron-scale organization of vertebrate plasma membranes, including those of erythrocytes, excitable membranes of neurons and heart, lateral membrane domains of columnar epithelial cells, and striated muscle. Ankyrins coordinate functionally related membrane transporters and cell adhesion proteins (15 protein families identified so far) within plasma membrane compartments through independently evolved interactions of intrinsically disordered sequences with a highly conserved peptide-binding groove formed by the ANK repeat solenoid. Ankyrins are coupled to spectrins, which are elongated organelle-sized proteins that form mechanically resilient arrays through cross-linking by specialized actin filaments. In addition to protein interactions, cellular targeting and assembly of spectrin/ankyrin domains also critically depend on palmitoylation of ankyrin-G by aspartate-histidine-histidine-cysteine 5/8 palmitoyltransferases, as well as interaction of beta-2 spectrin with phosphoinositide lipids. These lipid-dependent spectrin/ankyrin domains are not static but are locally dynamic and determine membrane identity through opposing endocytosis of bulk lipids as well as specific proteins. A partnership between spectrin, ankyrin, and cell adhesion molecules first emerged in bilaterians over 500 million years ago. Ankyrin and spectrin may have been recruited to plasma membranes from more ancient roles in organelle transport. The basic bilaterian spectrin-ankyrin toolkit markedly expanded in vertebrates through gene duplications combined with variation in unstructured intramolecular regulatory sequences as well as independent evolution of ankyrin-binding activity by ion transporters involved in action potentials and calcium homeostasis. In addition, giant vertebrate ankyrins with specialized roles in axons acquired new coding sequences by exon shuffling. We speculate that early axon initial segments and epithelial lateral membranes initially were based on spectrin-ankyrin-cell adhesion molecule assemblies and subsequently served as "incubators," where ion transporters independently acquired ankyrin-binding activity through positive selection.
Collapse
|
22
|
Luck AN, Anderson KG, McClung CM, VerBerkmoes NC, Foster JM, Michalski ML, Slatko BE. Tissue-specific transcriptomics and proteomics of a filarial nematode and its Wolbachia endosymbiont. BMC Genomics 2015; 16:920. [PMID: 26559510 PMCID: PMC4642636 DOI: 10.1186/s12864-015-2083-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 10/15/2015] [Indexed: 11/12/2022] Open
Abstract
Background Filarial nematodes cause debilitating human diseases. While treatable, recent evidence suggests drug resistance is developing, necessitating the development of novel targets and new treatment options. Although transcriptomic and proteomic studies around the nematode life cycle have greatly enhanced our knowledge, whole organism approaches have not provided spatial resolution of gene expression, which can be gained by examining individual tissues. Generally, due to their small size, tissue dissection of human-infecting filarial nematodes remains extremely challenging. However, canine heartworm disease is caused by a closely related and much larger filarial nematode, Dirofilaria immitis. As with many other filarial nematodes, D. immitis contains Wolbachia, an obligate bacterial endosymbiont present in the hypodermis and developing oocytes within the uterus. Here, we describe the first concurrent tissue-specific transcriptomic and proteomic profiling of a filarial nematode (D. immitis) and its Wolbachia (wDi) in order to better understand tissue functions and identify tissue-specific antigens that may be used for the development of new diagnostic and therapeutic tools. Methods Adult D. immitis worms were dissected into female body wall (FBW), female uterus (FU), female intestine (FI), female head (FH), male body wall (MBW), male testis (MT), male intestine (MI), male head (MH) and 10.1186/s12864-015-2083-2 male spicule (MS) and used to prepare transcriptomic and proteomic libraries. Results Transcriptomic and proteomic analysis of several D. immitis tissues identified many biological functions enriched within certain tissues. Hierarchical clustering of the D. immitis tissue transcriptomes, along with the recently published whole-worm adult male and female D. immitis transcriptomes, revealed that the whole-worm transcriptome is typically dominated by transcripts originating from reproductive tissue. The uterus appeared to have the most variable transcriptome, possibly due to age. Although many functions are shared between the reproductive tissues, the most significant differences in gene expression were observed between the uterus and testis. Interestingly, wDi gene expression in the male and female body wall is fairly similar, yet slightly different to that of Wolbachia gene expression in the uterus. Proteomic methods verified 32 % of the predicted D. immitis proteome, including over 700 hypothetical proteins of D. immitis. Of note, hypothetical proteins were among some of the most abundant Wolbachia proteins identified, which may fulfill some important yet still uncharacterized biological function. Conclusions The spatial resolution gained from this parallel transcriptomic and proteomic analysis adds to our understanding of filarial biology and serves as a resource with which to develop future therapeutic strategies against filarial nematodes and their Wolbachia endosymbionts. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2083-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ashley N Luck
- Genome Biology Division, New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | - Kathryn G Anderson
- Department of Biology and Microbiology, University of Wisconsin Oshkosh, Oshkosh, WI, 54901, USA
| | - Colleen M McClung
- Chemical Biology Division, New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | - Nathan C VerBerkmoes
- Chemical Biology Division, New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | - Jeremy M Foster
- Genome Biology Division, New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | - Michelle L Michalski
- Department of Biology and Microbiology, University of Wisconsin Oshkosh, Oshkosh, WI, 54901, USA
| | - Barton E Slatko
- Genome Biology Division, New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA.
| |
Collapse
|
23
|
Lemonidis K, Sanchez-Perez MC, Chamberlain LH. Identification of a Novel Sequence Motif Recognized by the Ankyrin Repeat Domain of zDHHC17/13 S-Acyltransferases. J Biol Chem 2015; 290:21939-50. [PMID: 26198635 PMCID: PMC4571948 DOI: 10.1074/jbc.m115.657668] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 07/20/2015] [Indexed: 11/06/2022] Open
Abstract
S-Acylation is a major post-translational modification affecting several cellular processes. It is particularly important for neuronal functions. This modification is catalyzed by a family of transmembrane S-acyltransferases that contain a conserved zinc finger DHHC (zDHHC) domain. Typically, eukaryote genomes encode for 7-24 distinct zDHHC enzymes, with two members also harboring an ankyrin repeat (AR) domain at their cytosolic N termini. The AR domain of zDHHC enzymes is predicted to engage in numerous interactions and facilitates both substrate recruitment and S-acylation-independent functions; however, the sequence/structural features recognized by this module remain unknown. The two mammalian AR-containing S-acyltransferases are the Golgi-localized zDHHC17 and zDHHC13, also known as Huntingtin-interacting proteins 14 and 14-like, respectively; they are highly expressed in brain, and their loss in mice leads to neuropathological deficits that are reminiscent of Huntington's disease. Here, we report that zDHHC17 and zDHHC13 recognize, via their AR domain, evolutionary conserved and closely related sequences of a [VIAP][VIT]XXQP consensus in SNAP25, SNAP23, cysteine string protein, Huntingtin, cytoplasmic linker protein 3, and microtubule-associated protein 6. This novel AR-binding sequence motif is found in regions predicted to be unstructured and is present in a number of zDHHC17 substrates and zDHHC17/13-interacting S-acylated proteins. This is the first study to identify a motif recognized by AR-containing zDHHCs.
Collapse
Affiliation(s)
- Kimon Lemonidis
- From the Strathclyde Institute of Pharmacy and Biomedical Sciences, Univesity of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Maria C Sanchez-Perez
- From the Strathclyde Institute of Pharmacy and Biomedical Sciences, Univesity of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Luke H Chamberlain
- From the Strathclyde Institute of Pharmacy and Biomedical Sciences, Univesity of Strathclyde, Glasgow G4 0RE, United Kingdom
| |
Collapse
|
24
|
Díaz-Balzac CA, Lázaro-Peña MI, Ramos-Ortiz GA, Bülow HE. The Adhesion Molecule KAL-1/anosmin-1 Regulates Neurite Branching through a SAX-7/L1CAM-EGL-15/FGFR Receptor Complex. Cell Rep 2015; 11:1377-84. [PMID: 26004184 PMCID: PMC4464948 DOI: 10.1016/j.celrep.2015.04.057] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 03/20/2015] [Accepted: 04/27/2015] [Indexed: 01/10/2023] Open
Abstract
Neurite branching is essential for correct assembly of neural circuits, yet it remains a poorly understood process. For example, the neural cell adhesion molecule KAL-1/anosmin-1, which is mutated in Kallmann syndrome, regulates neurite branching through mechanisms largely unknown. Here, we show that KAL-1/anosmin-1 mediates neurite branching as an autocrine co-factor with EGL-17/FGF through a receptor complex consisting of the conserved cell adhesion molecule SAX-7/L1CAM and the fibroblast growth factor receptor EGL-15/FGFR. This protein complex, which appears conserved in humans, requires the immunoglobulin (Ig) domains of SAX-7/L1CAM and the FN(III) domains of KAL-1/anosmin-1 for formation in vitro as well as function in vivo. The kinase domain of the EGL-15/FGFR is required for branching, and genetic evidence suggests that ras-mediated signaling downstream of EGL-15/FGFR is necessary to effect branching. Our studies establish a molecular pathway that regulates neurite branching during development of the nervous system.
Collapse
Affiliation(s)
- Carlos A Díaz-Balzac
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - María I Lázaro-Peña
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Gibram A Ramos-Ortiz
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Biology, University of Puerto Rico-Río Piedras, San Juan 00931, Puerto Rico
| | - Hannes E Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
25
|
Hartin SN, Hudson ML, Yingling C, Ackley BD. A Synthetic Lethal Screen Identifies a Role for Lin-44/Wnt in C. elegans Embryogenesis. PLoS One 2015; 10:e0121397. [PMID: 25938228 PMCID: PMC4418752 DOI: 10.1371/journal.pone.0121397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/31/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The C. elegans proteins PTP-3/LAR-RPTP and SDN-1/Syndecan are conserved cell adhesion molecules. Loss-of-function (LOF) mutations in either ptp-3 or sdn-1 result in low penetrance embryonic developmental defects. Work from other systems has shown that syndecans can function as ligands for LAR receptors in vivo. We used double mutant analysis to test whether ptp-3 and sdn-1 function in a linear genetic pathway during C. elegans embryogenesis. RESULTS We found animals with LOF in both sdn-1 and ptp-3 exhibited a highly penetrant synthetic lethality (SynLet), with only a small percentage of animals surviving to adulthood. Analysis of the survivors demonstrated that these animals had a synergistic increase in the penetrance of embryonic developmental defects. Together, these data strongly suggested PTP-3 and SDN-1 function in parallel during embryogenesis. We subsequently used RNAi to knockdown ~3,600 genes predicted to encode secreted and/or transmembrane molecules to identify genes that interacted with ptp-3 or sdn-1. We found that the Wnt ligand, lin-44, was SynLet with sdn-1, but not ptp-3. We used 4-dimensional time-lapse analysis to characterize the interaction between lin-44 and sdn-1. We found evidence that loss of lin-44 caused defects in the polarization and migration of endodermal precursors during gastrulation, a previously undescribed role for lin-44 that is strongly enhanced by the loss of sdn-1. CONCLUSIONS PTP-3 and SDN-1 function in compensatory pathways during C. elegans embryonic and larval development, as simultaneous loss of both genes has dire consequences for organismal survival. The Wnt ligand lin-44 contributes to the early stages of gastrulation in parallel to sdn-1, but in a genetic pathway with ptp-3. Overall, the SynLet phenotype provides a robust platform to identify ptp-3 and sdn-1 interacting genes, as well as other genes that function in development, yet might be missed in traditional forward genetic screens.
Collapse
Affiliation(s)
- Samantha N. Hartin
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, United States of America
| | - Martin L. Hudson
- Department of Biology and Physics, Kennesaw State University, Kennesaw, GA, United States of America
| | - Curtis Yingling
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, United States of America
| | - Brian D. Ackley
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, United States of America
- * E-mail:
| |
Collapse
|
26
|
Opperman K, Moseley-Alldredge M, Yochem J, Bell L, Kanayinkal T, Chen L. A novel nondevelopmental role of the sax-7/L1CAM cell adhesion molecule in synaptic regulation in Caenorhabditis elegans. Genetics 2015; 199:497-509. [PMID: 25488979 PMCID: PMC4317657 DOI: 10.1534/genetics.114.169581] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 12/02/2014] [Indexed: 01/25/2023] Open
Abstract
The L1CAM family of cell adhesion molecules is a conserved set of single-pass transmembrane proteins that play diverse roles required for proper nervous system development and function. Mutations in L1CAMs can cause the neurological L1 syndrome and are associated with autism and neuropsychiatric disorders. L1CAM expression in the mature nervous system suggests additional functions besides the well-characterized developmental roles. In this study, we demonstrate that the gene encoding the Caenorhabditis elegans L1CAM, sax-7, genetically interacts with gtl-2, as well as with unc-13 and rab-3, genes that function in neurotransmission. These sax-7 genetic interactions result in synthetic phenotypes that are consistent with abnormal synaptic function. Using an inducible sax-7 expression system and pharmacological reagents that interfere with cholinergic transmission, we uncovered a previously uncharacterized nondevelopmental role for sax-7 that impinges on synaptic function.
Collapse
Affiliation(s)
- Karla Opperman
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Melinda Moseley-Alldredge
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455 Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota 55455
| | - John Yochem
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Leslie Bell
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Tony Kanayinkal
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Lihsia Chen
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455 Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
27
|
Abstract
PHA-1 encodes a cytoplasmic protein that is required for embryonic morphogenesis and attachment of the foregut (pharynx) to the mouth (buccal capsule). Previous reports have in some cases suggested that PHA-1 is essential for the differentiation of most or all pharyngeal cell types. By performing mosaic analysis with a recently acquired pha-1 null mutation (tm3671), we found that PHA-1 is not required within most or all pharyngeal cells for their proper specification, differentiation, or function. Rather, our evidence suggests that PHA-1 acts in the arcade or anterior epithelial cells of the pharynx to promote attachment of the pharynx to the future buccal capsule. In addition, PHA-1 appears to be required in the epidermis for embryonic morphogenesis, in the excretory system for osmoregulation, and in the somatic gonad for normal ovulation and fertility. PHA-1 activity is also required within at least a subset of intestinal cells for viability. To better understand the role of PHA-1 in the epidermis, we analyzed several apical junction markers in pha-1(tm3671) homozygous embryos. PHA-1 regulates the expression of several components of two apical junction complexes including AJM-1–DLG-1/discs large complex and the classical cadherin–catenin complex, which may account for the role of PHA-1 in embryonic morphogenesis.
Collapse
|
28
|
Nagaraj K, Mualla R, Hortsch M. The L1 Family of Cell Adhesion Molecules: A Sickening Number of Mutations and Protein Functions. ADVANCES IN NEUROBIOLOGY 2014; 8:195-229. [DOI: 10.1007/978-1-4614-8090-7_9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
29
|
McLachlan IG, Heiman MG. Shaping dendrites with machinery borrowed from epithelia. Curr Opin Neurobiol 2013; 23:1005-10. [DOI: 10.1016/j.conb.2013.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 06/27/2013] [Indexed: 10/26/2022]
|
30
|
Abstract
Receptor Tyrosine Kinase (RTK)-Ras-Extracellular signal-regulated kinase (ERK) signaling pathways control many aspects of C. elegans development and behavior. Studies in C. elegans helped elucidate the basic framework of the RTK-Ras-ERK pathway and continue to provide insights into its complex regulation, its biological roles, how it elicits cell-type appropriate responses, and how it interacts with other signaling pathways to do so. C. elegans studies have also revealed biological contexts in which alternative RTK- or Ras-dependent pathways are used instead of the canonical pathway.
Collapse
Affiliation(s)
- Meera V Sundaram
- Dept. of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6145, USA.
| |
Collapse
|
31
|
Bennett V, Lorenzo DN. Spectrin- and Ankyrin-Based Membrane Domains and the Evolution of Vertebrates. CURRENT TOPICS IN MEMBRANES 2013; 72:1-37. [DOI: 10.1016/b978-0-12-417027-8.00001-5] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
32
|
Cadherins and their partners in the nematode worm Caenorhabditis elegans. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 116:239-62. [PMID: 23481198 DOI: 10.1016/b978-0-12-394311-8.00011-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The extreme simplicity of Caenorhabditis elegans makes it an ideal system to study the basic principles of cadherin function at the level of single cells within the physiologically relevant context of a developing animal. The genetic tractability of C. elegans also means that components of cadherin complexes can be identified through genetic modifier screens, allowing a comprehensive in vivo characterization of the macromolecular assemblies involved in cadherin function during tissue formation and maintenance in C. elegans. This work shows that a single cadherin system, the classical cadherin-catenin complex, is essential for diverse morphogenetic events during embryogenesis through its interactions with a range of mostly conserved proteins that act to modulate its function. The role of other members of the cadherin family in C. elegans, including members of the Fat-like, Flamingo/CELSR and calsyntenin families is less well characterized, but they have clear roles in neuronal development and function.
Collapse
|
33
|
Katidou M, Tavernarakis N, Karagogeos D. The contactin RIG-6 mediates neuronal and non-neuronal cell migration in Caenorhabditis elegans. Dev Biol 2012; 373:184-95. [PMID: 23123963 DOI: 10.1016/j.ydbio.2012.10.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 10/22/2012] [Accepted: 10/25/2012] [Indexed: 12/20/2022]
Abstract
Cell adhesion molecules of the Immunoglobulin Superfamily (IgCAMs) are key factors in nervous system formation. The contactin subgroup of IgCAMs consists of GPI-anchored glycoproteins implicated in axon outgrowth, guidance, fasciculation and neuronal differentiation. The mechanism by which contactins facilitate neuronal development is not understood. To gain insight into the function of contactins, we characterized RIG-6, the sole contactin of Caenorhabditis elegans. We show that the contactin RIG-6 is involved in excretory cell (EC) tubular elongation. We also show that RIG-6 mediates axon outgrowth and guidance along both the anterior-posterior and dorso-ventral axis, during C. elegans development. We find that optimal RIG-6 expression is critical for accurate mechanosensory neuron axon elongation and ventral nerve cord architecture. In addition, our data suggest that the cytoplasmic UNC-53/NAV2 proteins may contribute to relay signaling via contactins.
Collapse
Affiliation(s)
- Markella Katidou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | | | | |
Collapse
|
34
|
Mualla R, Nagaraj K, Hortsch M. A phylogenetic analysis of the L1 family of neural cell adhesion molecules. Neurochem Res 2012; 38:1196-207. [PMID: 23011207 DOI: 10.1007/s11064-012-0892-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 09/10/2012] [Accepted: 09/12/2012] [Indexed: 11/25/2022]
Abstract
L1-type genes form one of several distinct gene families that encode adhesive proteins, which are predominantly expressed in developing and mature metazoan nervous systems. These proteins have a multitude of different important cellular functions in neuronal and glial cells. L1-type gene products are transmembrane proteins with a characteristic extracellular domain structure consisting of six immunoglobulin and three to five fibronectin type III protein folds. As reported here, L1-type proteins can be identified in most metazoan phyla with the notable exception of Porifera (sponges). This puts the origin of L1-type genes at a point in time when primitive cellular neural networks emerged, approximately 1,200 to 1,500 million years ago. Subsequently, several independent gene duplication events generated multiple paralogous L1-type genes in some phyla, allowing for a considerable diversification of L1 structures and the emergence of new functional features and molecular interactions. One such evolutionary newer feature is the appearance of RGD integrin-binding motifs in some vertebrate L1 family members.
Collapse
Affiliation(s)
- Rula Mualla
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
35
|
Carberry K, Wiesenfahrt T, Geisler F, Stöcker S, Gerhardus H, Überbach D, Davis W, Jorgensen E, Leube RE, Bossinger O. The novel intestinal filament organizer IFO-1 contributes to epithelial integrity in concert with ERM-1 and DLG-1. Development 2012; 139:1851-62. [PMID: 22510987 DOI: 10.1242/dev.075788] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The nematode Caenorhabditis elegans is an excellent model system in which to study in vivo organization and function of the intermediate filament (IF) system for epithelial development and function. Using a transgenic ifb-2::cfp reporter strain, a mutagenesis screen was performed to identify mutants with aberrant expression patterns of the IF protein IFB-2, which is expressed in a dense network at the subapical endotube just below the microvillar brush border of intestinal cells. Two of the isolated alleles (kc2 and kc3) were mapped to the same gene, which we refer to as ifo-1 (intestinal filament organizer). The encoded polypeptide colocalizes with IF proteins and F-actin in the intestine. The apical localization of IFO-1 does not rely on IFB-2 but is dependent on LET-413, a basolateral protein involved in apical junction assembly and maintenance of cell polarity. In mutant worms, IFB-2 and IFC-2 are mislocalized in cytoplasmic granules and accumulate in large aggregates at the C. elegans apical junction (CeAJ) in a DLG-1-dependent fashion. Electron microscopy reveals loss of the prominent endotube and disordered but still intact microvilli. Semiquantitative fluorescence microscopy revealed a significant decrease of F-actin, suggesting a general role of IFO-1 in cytoskeletal organization. Furthermore, downregulation of the cytoskeletal organizer ERM-1 and the adherens junction component DLG-1, each of which leads to F-actin reduction on its own, induces a novel synthetic phenotype in ifo-1 mutants resulting in disruption of the lumen. We conclude that IFO-1 is a multipurpose linker between different cytoskeletal components of the C. elegans intestinal terminal web and contributes to proper epithelial tube formation.
Collapse
Affiliation(s)
- Katrin Carberry
- Institute of Molecular and Cellular Anatomy (MOCA), RWTH Aachen University, D-52074 Aachen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Chihara D, Nance J. An E-cadherin-mediated hitchhiking mechanism for C. elegans germ cell internalization during gastrulation. Development 2012; 139:2547-56. [PMID: 22675206 DOI: 10.1242/dev.079863] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Gastrulation movements place endodermal precursors, mesodermal precursors and primordial germ cells (PGCs) into the interior of the embryo. Somatic cell gastrulation movements are regulated by transcription factors that also control cell fate, coupling cell identity and position. By contrast, PGCs in many species are transcriptionally quiescent, suggesting that they might use alternative gastrulation strategies. Here, we show that C. elegans PGCs internalize by attaching to internal endodermal cells, which undergo morphogenetic movements that pull the PGCs into the embryo. We show that PGCs enrich HMR-1/E-cadherin at their surfaces to stick to endoderm. HMR-1 expression in PGCs is necessary and sufficient to ensure internalization, suggesting that HMR-1 can promote PGC-endoderm adhesion through a mechanism other than homotypic trans interactions between the two cell groups. Finally, we demonstrate that the hmr-1 3' untranslated region promotes increased HMR-1 translation in PGCs. Our findings reveal that quiescent PGCs employ a post-transcriptionally regulated hitchhiking mechanism to internalize during gastrulation, and demonstrate a morphogenetic role for the conserved association of PGCs with the endoderm.
Collapse
Affiliation(s)
- Daisuke Chihara
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
| | | |
Collapse
|
37
|
|
38
|
Chen L, Wang Z, Ghosh-Roy A, Hubert T, Yan D, O'Rourke S, Bowerman B, Wu Z, Jin Y, Chisholm AD. Axon regeneration pathways identified by systematic genetic screening in C. elegans. Neuron 2011; 71:1043-57. [PMID: 21943602 PMCID: PMC3183436 DOI: 10.1016/j.neuron.2011.07.009] [Citation(s) in RCA: 160] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2011] [Indexed: 12/18/2022]
Abstract
The mechanisms underlying the ability of axons to regrow after injury remain poorly explored at the molecular genetic level. We used a laser injury model in Caenorhabditis elegans mechanosensory neurons to screen 654 conserved genes for regulators of axonal regrowth. We uncover several functional clusters of genes that promote or repress regrowth, including genes classically known to affect axon guidance, membrane excitability, neurotransmission, and synaptic vesicle endocytosis. The conserved Arf Guanine nucleotide Exchange Factor (GEF), EFA-6, acts as an intrinsic inhibitor of regrowth. By combining genetics and in vivo imaging, we show that EFA-6 inhibits regrowth via microtubule dynamics, independent of its Arf GEF activity. Among newly identified regrowth inhibitors, only loss of function in EFA-6 partially bypasses the requirement for DLK-1 kinase. Identification of these pathways significantly expands our understanding of the genetic basis of axonal injury responses and repair.
Collapse
Affiliation(s)
- Lizhen Chen
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Bernadskaya YY, Patel FB, Hsu HT, Soto MC. Arp2/3 promotes junction formation and maintenance in the Caenorhabditis elegans intestine by regulating membrane association of apical proteins. Mol Biol Cell 2011; 22:2886-99. [PMID: 21697505 PMCID: PMC3154884 DOI: 10.1091/mbc.e10-10-0862] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
It has been proposed that Arp2/3, which promotes nucleation of branched actin, is needed for epithelial junction initiation but is less important as junctions mature. We focus here on how Arp2/3 contributes to the Caenorhabditis elegans intestinal epithelium and find important roles for Arp2/3 in the maturation and maintenance of junctions in embryos and adults. Electron microscope studies show that embryos depleted of Arp2/3 form apical actin-rich microvilli and electron-dense apical junctions. However, whereas apical/basal polarity initiates, apical maturation is defective, including decreased apical F-actin enrichment, aberrant lumen morphology, and reduced accumulation of some apical junctional proteins, including DLG-1. Depletion of Arp2/3 in adult animals leads to similar intestinal defects. The DLG-1/AJM-1 apical junction proteins, and the ezrin-radixin-moesin homologue ERM-1, a protein that connects F-actin to membranes, are required along with Arp2/3 for apical F-actin enrichment in embryos, whereas cadherin junction proteins are not. Arp2/3 affects the subcellular distribution of DLG-1 and ERM-1. Loss of Arp2/3 shifts both ERM-1 and DLG-1 from pellet fractions to supernatant fractions, suggesting a role for Arp2/3 in the distribution of membrane-associated proteins. Thus, Arp2/3 is required as junctions mature to maintain apical proteins associated with the correct membranes.
Collapse
Affiliation(s)
- Yelena Y Bernadskaya
- Department of Pathology and Laboratory Medicine, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | | | |
Collapse
|
40
|
Abstract
The dystrophin protein complex, an important regulator of muscle membrane integrity, also maintains neural organization through interactions with the L1CAM family member SAX-7. The dystrophin protein complex (DPC), composed of dystrophin and associated proteins, is essential for maintaining muscle membrane integrity. The link between mutations in dystrophin and the devastating muscle failure of Duchenne’s muscular dystrophy (DMD) has been well established. Less well appreciated are the accompanying cognitive impairment and neuropsychiatric disorders also presented in many DMD patients, which suggest a wider role for dystrophin in membrane–cytoskeleton function. This study provides genetic evidence of a novel role for DYS-1/dystrophin in maintaining neural organization in Caenorhabditis elegans. This neuronal function is distinct from the established role of DYS-1/dystrophin in maintaining muscle integrity and regulating locomotion. SAX-7, an L1 cell adhesion molecule (CAM) homologue, and STN-2/γ-syntrophin also function to maintain neural integrity in C. elegans. This study provides biochemical data that show that SAX-7 associates with DYS-1 in an STN-2/γ-syntrophin–dependent manner. These results reveal a recruitment of L1CAMs to the DPC to ensure neural integrity is maintained.
Collapse
Affiliation(s)
- Shan Zhou
- Department of Genetics, Cell Biology, and Development, Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | | |
Collapse
|
41
|
Vansteenhouse HC, Horton ZA, O'Hagan R, Tai MH, Zipser B. Phylogenetic conservation of the cell-type-specific Lan3-2 glycoepitope in Caenorhabditis elegans. Dev Genes Evol 2010; 220:77-87. [PMID: 20563596 DOI: 10.1007/s00427-010-0330-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 06/01/2010] [Indexed: 10/19/2022]
Abstract
The biological function of a cell-type-specific glycosylation of an adhesion molecule belonging to the L1CAM immunoglobulin superfamily was previously determined in the nervous system of the embryonic leech, Hirudo medicinalis. The Lan3-2 glycoepitope is a surface marker of sensory afferent neurons and is required for their appropriate developmental collateral branching and synaptogenesis in the CNS. The chemical structure of the Lan3-2 glycoepitope consists of beta-(1,4)-linked mannopyranose. Here, we show the conservation of the cell-type-specific expression of this mannose polymer in Caenorhabditis elegans. The Lan3-2 glycoepitope is expressed on the cell surface of a subset of dissociated embryonic neurons and, in the adult worm, by the pharyngeal motor neuron, M5, and the chemosensory afferents, the amphids. Additionally, the vulval epithelium expresses the Lan3-2 glycoepitope in late L4 larvae and in adult hermaphrodites. To investigate proteins carrying this restrictively expressed glycoepitope, worm extract was immunoaffinity purified with Lan3-2 monoclonal antibody and Western blotted. A polyclonal antibody reactive with the cytoplasmic tail of LAD-1/SAX-7, a C. elegans member of the L1CAM family, recognizes a 270 kDa protein band while Lan3-2 antibody also recognizes a 190 kDa glycoform, its putative Lan3-2 ectodomain. Thus, in C. elegans, as in leech, the Lan3-2 epitope is located on a L1CAM homologue. The cell-type-specific expression of the Lan3-2 glycoepitope shared by leech and C. elegans will be useful for understanding how cell-type-specific glycoepitopes mediate cell-cell interactions during development.
Collapse
|
42
|
Tee JM, Peppelenbosch MP. Anchoring skeletal muscle development and disease: the role of ankyrin repeat domain containing proteins in muscle physiology. Crit Rev Biochem Mol Biol 2010; 45:318-30. [PMID: 20515317 PMCID: PMC2942773 DOI: 10.3109/10409238.2010.488217] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The ankyrin repeat is a protein module with high affinity for other ankyrin repeats based on strong Van der Waals forces. The resulting dimerization is unusually resistant to both mechanical forces and alkanization, making this module exceedingly useful for meeting the extraordinary demands of muscle physiology. Many aspects of muscle function are controlled by the superfamily ankyrin repeat domain containing proteins, including structural fixation of the contractile apparatus to the muscle membrane by ankyrins, the archetypical member of the family. Additionally, other ankyrin repeat domain containing proteins critically control the various differentiation steps during muscle development, with Notch and developmental stage-specific expression of the members of the Ankyrin repeat and SOCS box (ASB) containing family of proteins controlling compartment size and guiding the various steps of muscle specification. Also, adaptive responses in fully formed muscle require ankyrin repeat containing proteins, with Myotrophin/V-1 ankyrin repeat containing proteins controlling the induction of hypertrophic responses following excessive mechanical load, and muscle ankyrin repeat proteins (MARPs) acting as protective mechanisms of last resort following extreme demands on muscle tissue. Knowledge on mechanisms governing the ordered expression of the various members of superfamily of ankyrin repeat domain containing proteins may prove exceedingly useful for developing novel rational therapy for cardiac disease and muscle dystrophies.
Collapse
Affiliation(s)
- Jin-Ming Tee
- Hubrecht Institute for Developmental Biology and Stem Cell Research-University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | | |
Collapse
|
43
|
Grana TM, Cox EA, Lynch AM, Hardin J. SAX-7/L1CAM and HMR-1/cadherin function redundantly in blastomere compaction and non-muscle myosin accumulation during Caenorhabditis elegans gastrulation. Dev Biol 2010; 344:731-44. [PMID: 20515680 PMCID: PMC2914123 DOI: 10.1016/j.ydbio.2010.05.507] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2010] [Accepted: 05/24/2010] [Indexed: 01/13/2023]
Abstract
Gastrulation is the first major morphogenetic movement in development and requires dynamic regulation of cell adhesion and the cytoskeleton. Caenorhabditis elegans gastrulation begins with the migration of the two endodermal precursors, Ea and Ep, from the surface of the embryo into the interior. Ea/Ep migration provides a relatively simple system to examine the intersection of cell adhesion, cell signaling, and cell movement. Ea/Ep ingression depends on correct cell fate specification and polarization, apical myosin accumulation, and Wnt activated actomyosin contraction that drives apical constriction and ingression (Lee et al., 2006; Nance et al., 2005). Here, we show that Ea/Ep ingression also requires the function of either HMR-1/cadherin or SAX-7/L1CAM. Both cadherin complex components and L1CAM are localized at all sites of cell-cell contact during gastrulation. Either system is sufficient for Ea/Ep ingression, but loss of both together leads to a failure of apical constriction and ingression. Similar results are seen with isolated blastomeres. Ea/Ep are properly specified and appear to display correct apical-basal polarity in sax-7(eq1);hmr-1(RNAi) embryos. Significantly, in sax-7(eq1);hmr-1(RNAi) embryos, Ea and Ep fail to accumulate myosin (NMY-2Colon, two colonsGFP) at their apical surfaces, but in either sax-7(eq1) or hmr-1(RNAi) embryos, apical myosin accumulation is comparable to wild type. Thus, the cadherin and L1CAM adhesion systems are redundantly required for localized myosin accumulation and hence for actomyosin contractility during gastrulation. We also show that sax-7 and hmr-1 function are redundantly required for Wnt-dependent spindle polarization during division of the ABar blastomere, indicating that these cell surface proteins redundantly regulate multiple developmental events in early embryos.
Collapse
Affiliation(s)
- Theresa M. Grana
- Department of Biological Sciences, University of Mary Washington, 1301 College Ave., Fredericksburg, VA 22401
| | - Elisabeth A. Cox
- Department of Biology, SUNY College at Geneseo, 1 College Cir., Geneseo, NY 14454
| | - Allison M. Lynch
- Program in Genetics, University of Wisconsin, 1117 W. Johnson St., Madison, WI 53706
| | - Jeff Hardin
- Program in Genetics, University of Wisconsin, 1117 W. Johnson St., Madison, WI 53706
- Department of Zoology, University of Wisconsin, 1117 W. Johnson St., Madison, WI 53706
| |
Collapse
|
44
|
Baines AJ. The spectrin-ankyrin-4.1-adducin membrane skeleton: adapting eukaryotic cells to the demands of animal life. PROTOPLASMA 2010; 244:99-131. [PMID: 20668894 DOI: 10.1007/s00709-010-0181-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 07/05/2010] [Indexed: 05/29/2023]
Abstract
The cells in animals face unique demands beyond those encountered by their unicellular eukaryotic ancestors. For example, the forces engendered by the movement of animals places stresses on membranes of a different nature than those confronting free-living cells. The integration of cells into tissues, as well as the integration of tissue function into whole animal physiology, requires specialisation of membrane domains and the formation of signalling complexes. With the evolution of mammals, the specialisation of cell types has been taken to an extreme with the advent of the non-nucleated mammalian red blood cell. These and other adaptations to animal life seem to require four proteins--spectrin, ankyrin, 4.1 and adducin--which emerged during eumetazoan evolution. Spectrin, an actin cross-linking protein, was probably the earliest of these, with ankyrin, adducin and 4.1 only appearing as tissues evolved. The interaction of spectrin with ankyrin is probably a prerequisite for the formation of tissues; only with the advent of vertebrates did 4.1 acquires the ability to bind spectrin and actin. The latter activity seems to allow the spectrin complex to regulate the cell surface accumulation of a wide variety of proteins. Functionally, the spectrin-ankyrin-4.1-adducin complex is implicated in the formation of apical and basolateral domains, in aspects of membrane trafficking, in assembly of certain signalling and cell adhesion complexes and in providing stability to otherwise mechanically fragile cell membranes. Defects in this complex are manifest in a variety of hereditary diseases, including deafness, cardiac arrhythmia, spinocerebellar ataxia, as well as hereditary haemolytic anaemias. Some of these proteins also function as tumor suppressors. The spectrin-ankyrin-4.1-adducin complex represents a remarkable system that underpins animal life; it has been adapted to many different functions at different times during animal evolution.
Collapse
Affiliation(s)
- Anthony J Baines
- School of Biosciences and Centre for Biomedical Informatics, University of Kent, Canterbury, CT2 7NJ, UK.
| |
Collapse
|
45
|
Zhu H, Duchesne L, Rudland PS, Fernig DG. The heparan sulfate co-receptor and the concentration of fibroblast growth factor-2 independently elicit different signalling patterns from the fibroblast growth factor receptor. Cell Commun Signal 2010; 8:14. [PMID: 20576134 PMCID: PMC2912315 DOI: 10.1186/1478-811x-8-14] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 06/24/2010] [Indexed: 01/12/2023] Open
Abstract
Background The fibroblast growth factor receptor (FGFR) interprets concentration gradients of FGF ligands and structural changes in the heparan sulfate (HS) co-receptor to generate different cellular responses. However, whether the FGFR generates different signals is not known. Results We have previously shown in rat mammary fibroblasts that in cells deficient in sulfation, and so in HS co-receptor, FGF-2 can only stimulate a transient phosphorylation of p42/44 MAPK and so cannot stimulate DNA synthesis. Here we demonstrate that this is because in the absence of HS, FGF-2 fails to stimulate the phosphorylation of the adaptor FGFR substrate 2 (FRS2). In cells possessing the HS co-receptor, FGF-2 elicits a bell-shaped dose response: optimal concentrations stimulate DNA synthesis, but supramaximal concentrations (≥ 100 ng/mL) have little effect. At optimal concentrations (300 pg/mL) FGF-2 stimulates a sustained dual phosphorylation of p42/44 MAPK and tyrosine phosphorylation of FRS2. In contrast, 100 ng/mL FGF-2 only stimulates a transient early peak of p42/44 MAPK phosphorylation and fails to stimulate appreciably the phosphorylation of FRS2 on tyrosine. Conclusions These results suggest that the nature of the FGFR signal produced is determined by a combination of the HS co-receptor and the concentration of FGF ligand. Both the phosphorylation of the adaptor FRS2, the kinetics (sustained or transient) of phosphorylation of p42/44(MAPK) are varied, and so differing cellular responses are produced.
Collapse
Affiliation(s)
- Hongyan Zhu
- School of Biological Sciences, Biosciences Building, Crown Street, University of Liverpool, Liverpool, L69 7ZB, UK.
| | | | | | | |
Collapse
|
46
|
Nakamura Y, Lee S, Haddox CL, Weaver EJ, Lemmon VP. Role of the cytoplasmic domain of the L1 cell adhesion molecule in brain development. J Comp Neurol 2010; 518:1113-32. [PMID: 20127821 DOI: 10.1002/cne.22267] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Mutations in the human L1CAM gene cause X-linked hydrocephalus and MASA (Mental retardation, Aphasia, Shuffling gait, Adducted thumbs) syndrome. In vitro studies have shown that the L1 cytoplasmic domain (L1CD) is involved in L1 trafficking, neurite branching, signaling, and interactions with the cytoskeleton. L1cam knockout (L1(KO)) mice have hydrocephalus, a small cerebellum, hyperfasciculation of corticothalamic tracts, and abnormal peripheral nerves. To explore the function of the L1CD, we made three new mice lines in which different parts of the L1CD have been altered. In all mutant lines L1 protein is expressed and transported into the axon. Interestingly, these new L1CD mutant lines display normal brain morphology. However, the expression of L1 protein in the adult is dramatically reduced in the two L1CD mutant lines that lack the ankyrin-binding region and they show defects in motor function. Therefore, the L1CD is not responsible for the major defects observed in L1(KO) mice, yet it is required for continued L1 protein expression and motor function in the adult.
Collapse
Affiliation(s)
- Yukiko Nakamura
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | | | | | | | | |
Collapse
|
47
|
Abstract
The L1 family of cell adhesion molecules (L1CAMs) in vertebrates has long been studied for its roles in nervous system development and function. Members of this family have been associated with distinct neurological disorders that include CRASH, autism, 3p syndrome, and schizophrenia. The conservation of L1CAMs in Drosophila and Caenorhabditis elegans allows the opportunity to take advantage of these simple model organisms and their accessible genetic manipulations to dissect L1CAM functions and mechanisms of action. This review summarizes the discoveries of L1CAMs made in C. elegans, showcasing this simple model organism as a powerful system to uncover L1CAM mechanisms and roles in healthy and diseased states.
Collapse
Affiliation(s)
- Lihsia Chen
- Department of Genetics, Cell Biology, and Development, Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | |
Collapse
|
48
|
Hadwiger G, Dour S, Arur S, Fox P, Nonet ML. A monoclonal antibody toolkit for C. elegans. PLoS One 2010; 5:e10161. [PMID: 20405020 PMCID: PMC2854156 DOI: 10.1371/journal.pone.0010161] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 03/22/2010] [Indexed: 01/12/2023] Open
Abstract
Background Antibodies are critical tools in many avenues of biological research. Though antibodies can be produced in the research laboratory setting, most research labs working with vertebrates avail themselves of the wide array of commercially available reagents. By contrast, few such reagents are available for work with model organisms. Methodology/Principal Findings We report the production of monoclonal antibodies directed against a wide range of proteins that label specific subcellular and cellular components, and macromolecular complexes. Antibodies were made to synaptobrevin (SNB-1), a component of synaptic vesicles; to Rim (UNC-10), a protein localized to synaptic active zones; to transforming acidic coiled-coil protein (TAC-1), a component of centrosomes; to CENP-C (HCP-4), which in worms labels the entire length of their holocentric chromosomes; to ORC2 (ORC-2), a subunit of the DNA origin replication complex; to the nucleolar phosphoprotein NOPP140 (DAO-5); to the nuclear envelope protein lamin (LMN-1); to EHD1 (RME-1) a marker for recycling endosomes; to caveolin (CAV-1), a marker for caveolae; to the cytochrome P450 (CYP-33E1), a resident of the endoplasmic reticulum; to β-1,3-glucuronyltransferase (SQV-8) that labels the Golgi; to a chaperonin (HSP-60) targeted to mitochondria; to LAMP (LMP-1), a resident protein of lysosomes; to the alpha subunit of the 20S subcomplex (PAS-7) of the 26S proteasome; to dynamin (DYN-1) and to the α-subunit of the adaptor complex 2 (APA-2) as markers for sites of clathrin-mediated endocytosis; to the MAGUK, protein disks large (DLG-1) and cadherin (HMR-1), both of which label adherens junctions; to a cytoskeletal linker of the ezrin-radixin-moesin family (ERM-1), which localized to apical membranes; to an ERBIN family protein (LET-413) which localizes to the basolateral membrane of epithelial cells and to an adhesion molecule (SAX-7) which localizes to the plasma membrane at cell-cell contacts. In addition to working in whole mount immunocytochemistry, most of these antibodies work on western blots and thus should be of use for biochemical fractionation studies. Conclusions/Significance We have produced a set of monoclonal antibodies to subcellular components of the nematode C. elegans for the research community. These reagents are being made available through the Developmental Studies Hybridoma Bank (DSHB).
Collapse
Affiliation(s)
- Gayla Hadwiger
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Scott Dour
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Swathi Arur
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Paul Fox
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael L. Nonet
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
49
|
Hoffmann M, Segbert C, Helbig G, Bossinger O. Intestinal tube formation in Caenorhabditis elegans requires vang-1 and egl-15 signaling. Dev Biol 2010; 339:268-79. [DOI: 10.1016/j.ydbio.2009.12.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 11/26/2009] [Accepted: 12/01/2009] [Indexed: 10/20/2022]
|
50
|
Guan H, Maness PF. Perisomatic GABAergic innervation in prefrontal cortex is regulated by ankyrin interaction with the L1 cell adhesion molecule. Cereb Cortex 2010; 20:2684-93. [PMID: 20156840 DOI: 10.1093/cercor/bhq016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The L1 adhesion molecule functions in axon growth and guidance, but a role in synaptic development of cortical inhibitory interneurons is largely unexplored. L1 mediates adhesion by engaging the actin cytoskeleton through binding the actin/spectrin adapter protein ankyrin. Loss of L1-ankyrin interaction impaired process elaboration/branching by GABAergic interneurons, including basket cells, and reduced the number of perisomatic synapses in the cingulate cortex as shown in L1 mutant mice (L1YH) with a mutation in the ankyrin-binding site, either alone or intercrossed with GAD67-enhanced green fluorescence protein reporter mice. Electron microscopy revealed that perisomatic inhibitory synapses but not excitatory synapses in the neuropil were specifically affected. In wild-type cingulate cortex, L1 colocalized with perisomatic synaptic markers, whereas L1 phosphorylation on Tyr(1229) decreased postnatally, correlating with increased ankyrin binding and synaptic development. These results suggest a novel role for L1 engagement with the actin cytoskeleton in development of inhibitory connectivity within the cingulate cortex.
Collapse
Affiliation(s)
- Hanjun Guan
- Department of Biochemistry, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | |
Collapse
|