1
|
Cohen N, Rabinowitch I. Resolving transitions between distinct phases of memory consolidation at high resolution in Caenorhabditis elegans. iScience 2024; 27:111147. [PMID: 39524366 PMCID: PMC11547966 DOI: 10.1016/j.isci.2024.111147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/26/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Memory consolidation following learning is a dynamic and complex process comprising several transitions between distinct memory phases. Although memory consolidation has been studied extensively, it remains difficult to draw an integral description that can delimit the transition points between specific memory phases at the behavioral, neuronal, and genetic levels. To this end, we have developed a rapid and robust aversive conditioning protocol for the nematode worm Caenorhabditis elegans, tracing memory consolidation within the first hour post conditioning and then up to 18 h post conditioning. This made it possible to uncover time-dependent involvement of primary sensory neurons, transcription and translation processes, and diverse gene populations in memory consolidation. The change in neuronal valence was strong enough to induce second order conditioning, and was amenable to considerable modulation in specific mutant strains. Together, our work lends memory consolidation to detailed temporal and spatial analysis, advancing system-wide understanding of learning and memory.
Collapse
Affiliation(s)
- Netanel Cohen
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| | - Ithai Rabinowitch
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| |
Collapse
|
2
|
Hayden AN, Brandel KL, Pietryk EW, Merlau PR, Vijayakumar P, Leptich EJ, Gaytan ES, Williams MI, Ni CW, Chao HT, Rosenfeld JA, Arey RN. Behavioral screening reveals a conserved residue in Y-Box RNA-binding protein required for associative learning and memory in C. elegans. PLoS Genet 2024; 20:e1011443. [PMID: 39423228 PMCID: PMC11524487 DOI: 10.1371/journal.pgen.1011443] [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: 05/17/2024] [Revised: 10/30/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
RNA-binding proteins (RBPs) regulate translation and plasticity which are required for memory. RBP dysfunction has been linked to a range of neurological disorders where cognitive impairments are a key symptom. However, of the 2,000 RBPs in the human genome, many are uncharacterized with regards to neurological phenotypes. To address this, we used the model organism C. elegans to assess the role of 20 conserved RBPs in memory. We identified eight previously uncharacterized memory regulators, three of which are in the C. elegans Y-Box (CEY) RBP family. Of these, we determined that cey-1 is the closest ortholog to the mammalian Y-Box (YBX) RBPs. We found that CEY-1 is both necessary in the nervous system for memory ability and sufficient to promote memory. Leveraging human datasets, we found both copy number variation losses and single nucleotide variants in YBX1 and YBX3 in individuals with neurological symptoms. We identified one predicted deleterious YBX3 variant of unknown significance, p.Asn127Tyr, in two individuals with neurological symptoms. Introducing this variant into endogenous cey-1 locus caused memory deficits in the worm. We further generated two humanized worm lines expressing human YBX3 or YBX1 at the cey-1 locus to test evolutionary conservation of YBXs in memory and the potential functional significance of the p.Asn127Tyr variant. Both YBX1/3 can functionally replace cey-1, and introduction of p.Asn127Tyr into the humanized YBX3 locus caused memory deficits. Our study highlights the worm as a model to reveal memory regulators and identifies YBX dysfunction as a potential new source of rare neurological disease.
Collapse
Affiliation(s)
- Ashley N. Hayden
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
| | - Katie L. Brandel
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
| | - Edward W. Pietryk
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Paul R. Merlau
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
| | - Priyadharshini Vijayakumar
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
| | - Emily J. Leptich
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
| | - Elizabeth S. Gaytan
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
- Postbaccalaureate Research Education Program, Baylor College of Medicine, Houston, Texas, United States of America
| | - Meredith I. Williams
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
| | - Connie W. Ni
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Neuroscience, Rice University, Houston, Texas, United States of America
| | - Hsiao-Tuan Chao
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, United States of America
- McNair Medical Institute, The Robert and Janice McNair Foundation, Houston, Texas, United States of America
| | - Jill A. Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Baylor Genetics Laboratories, Houston, Texas, United States of America
| | - Rachel N. Arey
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
3
|
Hayden AN, Brandel KL, Merlau PR, Vijayakumar P, Leptich EJ, Pietryk EW, Gaytan ES, Ni CW, Chao HT, Rosenfeld JA, Arey RN. Behavioral screening of conserved RNA-binding proteins reveals CEY-1/YBX RNA-binding protein dysfunction leads to impairments in memory and cognition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574402. [PMID: 38260399 PMCID: PMC10802296 DOI: 10.1101/2024.01.05.574402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
RNA-binding proteins (RBPs) regulate translation and plasticity which are required for memory. RBP dysfunction has been linked to a range of neurological disorders where cognitive impairments are a key symptom. However, of the 2,000 RBPs in the human genome, many are uncharacterized with regards to neurological phenotypes. To address this, we used the model organism C. elegans to assess the role of 20 conserved RBPs in memory. We identified eight previously uncharacterized memory regulators, three of which are in the C. elegans Y-Box (CEY) RBP family. Of these, we determined that cey-1 is the closest ortholog to the mammalian Y-Box (YBX) RBPs. We found that CEY-1 is both necessary in the nervous system for memory ability and sufficient to increase memory. Leveraging human datasets, we found both copy number variation losses and single nucleotide variants in YBX1 and YBX3 in individuals with neurological symptoms. We identified one predicted deleterious YBX3 variant of unknown significance, p.Asn127Tyr, in two individuals with neurological symptoms. Introducing this variant into endogenous cey-1 locus caused memory deficits in the worm. We further generated two humanized worm lines expressing human YBX3 or YBX1 at the cey-1 locus to test evolutionary conservation of YBXs in memory and the potential functional significance of the p.Asn127Tyr variant. Both YBX1/3 can functionally replace cey-1, and introduction of p.Asn127Tyr into the humanized YBX3 locus caused memory deficits. Our study highlights the worm as a model to reveal memory regulators and identifies YBX dysfunction as a potential new source of rare neurological disease.
Collapse
Affiliation(s)
- Ashley N Hayden
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Katie L Brandel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Paul R Merlau
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | | | - Emily J Leptich
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Edward W Pietryk
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
| | - Elizabeth S Gaytan
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Postbaccalaureate Research Education Program, Baylor College of Medicine, Houston, TX, 77030
| | - Connie W Ni
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Neuroscience, Rice University, Houston, TX 77005
| | - Hsiao-Tuan Chao
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
- Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, 77030
- Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, 77030
- McNair Medical Institute, The Robert and Janice McNair Foundation, Houston, TX, 77030
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
- Baylor Genetics Laboratories, Houston, TX 77021
| | - Rachel N Arey
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
| |
Collapse
|
4
|
Armada G, Roque S, Serre-Miranda C, Ferreira L, Vale A, Rodrigues AJ, Hong W, Correia-Neves M, Vieira N. SNX27: A trans-species cognitive modulator with implications for anxiety and stress susceptibility. Neurobiol Stress 2024; 30:100619. [PMID: 38500791 PMCID: PMC10945257 DOI: 10.1016/j.ynstr.2024.100619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/08/2024] [Accepted: 02/16/2024] [Indexed: 03/20/2024] Open
Abstract
Sorting Nexin 27 (SNX27) is a brain-enriched endosome-associated cargo adaptor that shapes excitatory control, being relevant for cognitive and reward processing, and for several neurological conditions. Despite this, SNX27's role in the nervous system remains poorly explored. To further understand SNX27 function, we performed an extensive behavioral characterization comprising motor, cognitive and emotional dimensions of SNX27+/- mice. Furthermore, attending on the recently described association between SNX27 function and cellular stress signaling mechanisms in vitro, we explored SNX27-stress interplay using a Caenorhabditis elegans Δsnx-27 mutant and wild-type (WT) rodents after stress exposure. SNX27+/- mice, as C. elegans Δsnx-27 mutants, present cognitive impairments, highlighting a conserved role for SNX27 in cognitive modulation across species. Interestingly, SNX27 downmodulation leads to anxiety-like behavior in mice evaluated in the Elevated Plus Maze (EPM). This anxious phenotype is associated with increased dendritic complexity of the bed nucleus of the stria terminalis (BNST) neurons, and increased complexity of the basolateral amygdala (BLA) pyramidal neurons. These findings highlight the still unknown role of SNX27 in anxiety regulation. Moreover, we uncovered a direct link between SNX27 dysfunction and stress susceptibility in C. elegans and found that stress-exposed rodents display decreased SNX27 levels in stress-susceptible brain regions. Altogether, we provided new insights on SNX27's relevance in anxiety-related behaviors and neuronal structure in stress-associated brain regions.
Collapse
Affiliation(s)
- Gisela Armada
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Susana Roque
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Cláudia Serre-Miranda
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Liliana Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Ana Vale
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Ana João Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Neide Vieira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
5
|
Gharat V, Peter F, de Quervain DJF, Papassotiropoulos A, Stetak A. Role of GLR-1 in Age-Dependent Short-Term Memory Decline. eNeuro 2024; 11:ENEURO.0420-23.2024. [PMID: 38519128 PMCID: PMC11005081 DOI: 10.1523/eneuro.0420-23.2024] [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: 10/18/2023] [Revised: 02/10/2024] [Accepted: 03/06/2024] [Indexed: 03/24/2024] Open
Abstract
As the global elderly population grows, age-related cognitive decline is becoming an increasingly significant healthcare issue, often leading to various neuropsychiatric disorders. Among the many molecular players involved in memory, AMPA-type glutamate receptors are known to regulate learning and memory, but how their dynamics change with age and affect memory decline is not well understood. Here, we examined the in vivo properties of the AMPA-type glutamate receptor GLR-1 in the AVA interneuron of the Caenorhabditis elegans nervous system during physiological aging. We found that both total and membrane-bound GLR-1 receptor levels decrease with age in wild-type worms, regardless of their location along the axon. Using fluorescence recovery after photobleaching, we also demonstrated that a reduction in GLR-1 abundance correlates with decreased local, synaptic GLR-1 receptor dynamics. Importantly, we found that reduced GLR-1 levels strongly correlate with the age-related decline in short-term associative memory. Genetic manipulation of GLR-1 stability, by either deleting msi-1 or expressing a ubiquitination-defective GLR-1 (4KR) variant, prevented this age-related reduction in receptor abundance and improved the short-term memory performance in older animals, which reached performance levels similar to those of young animals. Overall, our data indicate that AMPA-type glutamate receptor abundance and dynamics are key factors in maintaining memory function and that changes in these parameters are linked to age-dependent short-term memory decline.
Collapse
Affiliation(s)
- Vaibhav Gharat
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel 4055, Switzerland
| | - Fabian Peter
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel 4055, Switzerland
| | - Dominique J-F de Quervain
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
- Division of Cognitive Neuroscience, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
- University Psychiatric Clinics, University of Basel, Basel 4002, Switzerland
| | - Andreas Papassotiropoulos
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel 4055, Switzerland
- University Psychiatric Clinics, University of Basel, Basel 4002, Switzerland
| | - Attila Stetak
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel 4055, Switzerland
- University Psychiatric Clinics, University of Basel, Basel 4002, Switzerland
| |
Collapse
|
6
|
Stetak AL, Grenal T, Lenninger Z, Knight KM, Doser RL, Hoerndli FJ. A Necessary Role for PKC-2 and TPA-1 in Olfactory Memory and Synaptic AMPAR Trafficking in Caenorhabditis elegans. J Neurosci 2024; 44:e1120232024. [PMID: 38238075 PMCID: PMC10919255 DOI: 10.1523/jneurosci.1120-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/25/2024] Open
Abstract
Protein kinase C (PKC) functions are essential for synaptic plasticity, learning, and memory. However, the roles of specific members of the PKC family in synaptic function, learning, and memory are poorly understood. Here, we investigated the role of individual PKC homologs for synaptic plasticity in Caenorhabditis elegans and found a differential role for pkc-2 and tpa-1, but not pkc-1 and pkc-3 in associative olfactory learning and memory. More specifically we show that PKC-2 is essential for associative learning and TPA-1 for short-term associative memory (STAM). Using endogenous labeling and cell-specific rescues, we show that TPA-1 and PKC-2 are required in AVA for their functions. Previous studies demonstrated that olfactory learning and memory in C. elegans are tied to proper synaptic content and trafficking of AMPA-type ionotropic glutamate receptor homolog GLR-1 in the AVA command interneurons. Therefore, we quantified synaptic content, transport, and delivery of GLR-1 in AVA and showed that loss of pkc-2 and tpa-1 leads to decreased transport and delivery but only a subtle decrease in GLR-1 levels at synapses. AVA-specific expression of both PKC-2 and TPA-1 rescued these defects. Finally, genetic epistasis showed that PKC-2 and TPA-1 likely act in the same pathway to control GLR-1 transport and delivery, while regulating different aspects of olfactory learning and STAM. Thus, our data tie together cell-specific functions of 2 PKCs to neuronal and behavioral outcomes in C. elegans, enabling comparative approaches to understand the evolutionarily conserved role of PKC in synaptic plasticity, learning, and memory.
Collapse
Affiliation(s)
- Attila L Stetak
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, 4055 Basel, Switzerland
- University Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland
| | - Thomas Grenal
- Division of Molecular Neuroscience, Department of Biomedicine, University of Basel, 4055 Basel, Switzerland
| | - Zephyr Lenninger
- Departments of Biomedical Science, Colorado State University, Fort Collins, Colorado 80523
| | - Kaz M Knight
- Departments of Biomedical Science, Colorado State University, Fort Collins, Colorado 80523
| | - Rachel L Doser
- Departments of Biomedical Science, Colorado State University, Fort Collins, Colorado 80523
- Health and Exercise Sciences, Colorado State University, Fort Collins, Colorado 80523
| | - Frederic J Hoerndli
- Departments of Biomedical Science, Colorado State University, Fort Collins, Colorado 80523
| |
Collapse
|
7
|
Menzel R, Zhang X, Pietrucik T, Bathelt A, Ruess L. Omega-3 PUFA and the fitness and cognition of the nematode Caenorhabditis elegans under different environmental conditions. Comp Biochem Physiol B Biochem Mol Biol 2024; 270:110925. [PMID: 38040326 DOI: 10.1016/j.cbpb.2023.110925] [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: 10/05/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
Many invertebrate species possess the metabolic ability to synthesize long-chain ω3 polyunsaturated fatty acids (PUFA) de novo. Due to their diverse effects on membrane architecture, neuroplasticity, growth and reproduction, PUFA have a high potential to positively influence the fitness of an organism. But how and when do these supposed advantages actually come into play? Other species, that are often closely related, pass natural selection without this special metabolic ability. The ω3-PUFA rich model organism Caenorhabditis elegans (Nematoda) and its mutant fat-1(wa9), lacking these PUFA, are a suitable test system. We analyzed potential impairments in reproduction and growth in a soil assay. Further, chemotaxis after aversive olfactory, associative learning and integration of a second sensory signal were assessed on agar plates. Moreover, we analyzed the phospholipid pattern of both C. elegans strains and further free-living nematodes species at different temperatures. While the phenotypic effects were rather small under standard conditions, lowering the temperature to 15 or even 10 °C or reducing the soil moisture, led to significant limitations, with the investigated parameters for neuroplasticity being most impaired. The ω3-PUFA free C. elegans mutant strain fat-1 did not adapt the fatty acid composition of its phospholipids to a decreasing temperature, while ω3-PUFA containing nematodes proportionally increased this PUFA group. In contrats, other ω3-PUFA free nematode species produced significantly more ω6-PUFA. Thus, the ability to synthesize long-chain ω3-PUFA de novo likely is fundamental for an increase in neuroplasticity and an efficient way for regulating membrane fluidity to maintain their functionality.
Collapse
Affiliation(s)
- Ralph Menzel
- Humboldt-Universität zu Berlin, Institute of Biology, Ecology, Philippstr. 13, 10115 Berlin, Germany.
| | - Xuchao Zhang
- Humboldt-Universität zu Berlin, Institute of Biology, Ecology, Philippstr. 13, 10115 Berlin, Germany
| | - Tamara Pietrucik
- Humboldt-Universität zu Berlin, Institute of Biology, Ecology, Philippstr. 13, 10115 Berlin, Germany
| | - Antonia Bathelt
- Humboldt-Universität zu Berlin, Institute of Biology, Ecology, Philippstr. 13, 10115 Berlin, Germany
| | - Liliane Ruess
- Humboldt-Universität zu Berlin, Institute of Biology, Ecology, Philippstr. 13, 10115 Berlin, Germany
| |
Collapse
|
8
|
McMillen A, Chew Y. Neural mechanisms of dopamine function in learning and memory in Caenorhabditis elegans. Neuronal Signal 2024; 8:NS20230057. [PMID: 38572143 PMCID: PMC10987485 DOI: 10.1042/ns20230057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/03/2023] [Accepted: 12/11/2023] [Indexed: 04/05/2024] Open
Abstract
Research into learning and memory over the past decades has revealed key neurotransmitters that regulate these processes, many of which are evolutionarily conserved across diverse species. The monoamine neurotransmitter dopamine is one example of this, with countless studies demonstrating its importance in regulating behavioural plasticity. However, dopaminergic neural networks in the mammalian brain consist of hundreds or thousands of neurons, and thus cannot be studied at the level of single neurons acting within defined neural circuits. The nematode Caenorhabditis elegans (C. elegans) has an experimentally tractable nervous system with a completely characterized synaptic connectome. This makes it an advantageous system to undertake mechanistic studies into how dopamine encodes lasting yet flexible behavioural plasticity in the nervous system. In this review, we synthesize the research to date exploring the importance of dopaminergic signalling in learning, memory formation, and forgetting, focusing on research in C. elegans. We also explore the potential for dopamine-specific fluorescent biosensors in C. elegans to visualize dopaminergic neural circuits during learning and memory formation in real-time. We propose that the use of these sensors in C. elegans, in combination with optogenetic and other light-based approaches, will further illuminate the detailed spatiotemporal requirements for encoding behavioural plasticity in an accessible experimental system. Understanding the key molecules and circuit mechanisms that regulate learning and forgetting in more compact invertebrate nervous systems may reveal new druggable targets for enhancing memory storage and delaying memory loss in bigger brains.
Collapse
Affiliation(s)
- Anna McMillen
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, 5042, South Australia, Australia
| | - Yee Lian Chew
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, 5042, South Australia, Australia
| |
Collapse
|
9
|
Gomila Pelegri N, Stanczak AM, Bottomley AL, Milthorpe BK, Gorrie CA, Padula MP, Santos J. Adipose-Derived Stem Cells Spontaneously Express Neural Markers When Grown in a PEG-Based 3D Matrix. Int J Mol Sci 2023; 24:12139. [PMID: 37569515 PMCID: PMC10418654 DOI: 10.3390/ijms241512139] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Neurological diseases are among the leading causes of disability and death worldwide and remain difficult to treat. Tissue engineering offers avenues to test potential treatments; however, the development of biologically accurate models of brain tissues remains challenging. Given their neurogenic potential and availability, adipose-derived stem cells (ADSCs) are of interest for creating neural models. While progress has been made in differentiating ADSCs into neural cells, their differentiation in 3D environments, which are more representative of the in vivo physiological conditions of the nervous system, is crucial. This can be achieved by modulating the 3D matrix composition and stiffness. Human ADSCs were cultured for 14 days in a 1.1 kPa polyethylene glycol-based 3D hydrogel matrix to assess effects on cell morphology, cell viability, proteome changes and spontaneous neural differentiation. Results showed that cells continued to proliferate over the 14-day period and presented a different morphology to 2D cultures, with the cells elongating and aligning with one another. The proteome analysis revealed 439 proteins changed in abundance by >1.5 fold. Cyclic nucleotide 3'-phosphodiesterase (CNPase) markers were identified using immunocytochemistry and confirmed with proteomics. Findings indicate that ADSCs spontaneously increase neural marker expression when grown in an environment with similar mechanical properties to the central nervous system.
Collapse
Affiliation(s)
- Neus Gomila Pelegri
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (N.G.P.); (B.K.M.)
- Neural Injury Research Unit, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Aleksandra M. Stanczak
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (A.M.S.); (M.P.P.)
| | - Amy L. Bottomley
- Microbial Imaging Facility, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Bruce K. Milthorpe
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (N.G.P.); (B.K.M.)
| | - Catherine A. Gorrie
- Neural Injury Research Unit, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Matthew P. Padula
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (A.M.S.); (M.P.P.)
| | - Jerran Santos
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (N.G.P.); (B.K.M.)
| |
Collapse
|
10
|
Brandel-Ankrapp KL, Arey RN. Uncovering novel regulators of memory using C. elegans genetic and genomic analysis. Biochem Soc Trans 2023; 51:161-171. [PMID: 36744642 PMCID: PMC10518207 DOI: 10.1042/bst20220455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/20/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023]
Abstract
How organisms learn and encode memory is an outstanding question in neuroscience research. Specifically, how memories are acquired and consolidated at the level of molecular and gene pathways remains unclear. In addition, memory is disrupted in a wide variety of neurological disorders; therefore, discovering molecular regulators of memory may reveal therapeutic targets for these disorders. C. elegans are an excellent model to uncover molecular and genetic regulators of memory. Indeed, the nematode's invariant neuronal lineage, fully mapped genome, and conserved associative behaviors have allowed the development of a breadth of genetic and genomic tools to examine learning and memory. In this mini-review, we discuss novel and exciting genetic and genomic techniques used to examine molecular and genetic underpinnings of memory from the level of the whole-worm to tissue-specific and cell-type specific approaches with high spatiotemporal resolution.
Collapse
Affiliation(s)
- Katie L. Brandel-Ankrapp
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, U.S.A
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, U.S.A
| | - Rachel N. Arey
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, U.S.A
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, U.S.A
| |
Collapse
|
11
|
(-)- Gossypol Inhibition of Musashi-Mediated Forgetting Improves Memory and Age-Dependent Memory Decline in Caenorhabditis elegans. Mol Neurobiol 2023; 60:820-835. [PMID: 36378468 PMCID: PMC9849318 DOI: 10.1007/s12035-022-03116-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
Abstract
Musashi RNA-binding proteins (MSIs) retain a pivotal role in stem cell maintenance, tumorigenesis, and nervous system development. Recently, we showed in C. elegans that Musashi (MSI-1) actively promotes forgetting upon associative learning via a 3'UTR-dependent translational expression of the Arp2/3 actin branching complex. Here, we investigated the evolutionary conserved role of MSI proteins and the effect of their pharmacological inhibition on memory. Expression of human Musashi 1 (MSI1) and Musashi 2 (MSI2) under the endogenous Musashi promoter fully rescued the phenotype of msi-1(lf) worms. Furthermore, pharmacological inhibition of human MSI1 and MSI2 activity using (-)- gossypol resulted in improved memory retention, without causing locomotor, chemotactic, or learning deficits. No drug effect was observed in msi-1(lf) treated worms. Using Western blotting and confocal microscopy, we found no changes in MSI-1 protein abundance following (-)- gossypol treatment, suggesting that Musashi gene expression remains unaltered and that the compound exerts its inhibitory effect post-translationally. Additionally, (-)- gossypol suppressed the previously seen rescue of the msi-1(lf) phenotype in worms expressing human MSI1 specifically in the AVA neuron, indicating that (-)- gossypol can regulate the Musashi pathway in a memory-related neuronal circuit in worms. Finally, treating aged worms with (-)- gossypol reversed physiological age-dependent memory decline. Taken together, our findings indicate that pharmacological inhibition of Musashi might represent a promising approach for memory modulation.
Collapse
|
12
|
Salmen F, De Jonghe J, Kaminski TS, Alemany A, Parada GE, Verity-Legg J, Yanagida A, Kohler TN, Battich N, van den Brekel F, Ellermann AL, Arias AM, Nichols J, Hemberg M, Hollfelder F, van Oudenaarden A. High-throughput total RNA sequencing in single cells using VASA-seq. Nat Biotechnol 2022; 40:1780-1793. [PMID: 35760914 PMCID: PMC9750877 DOI: 10.1038/s41587-022-01361-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 05/13/2022] [Indexed: 01/14/2023]
Abstract
Most methods for single-cell transcriptome sequencing amplify the termini of polyadenylated transcripts, capturing only a small fraction of the total cellular transcriptome. This precludes the detection of many long non-coding, short non-coding and non-polyadenylated protein-coding transcripts and hinders alternative splicing analysis. We, therefore, developed VASA-seq to detect the total transcriptome in single cells, which is enabled by fragmenting and tailing all RNA molecules subsequent to cell lysis. The method is compatible with both plate-based formats and droplet microfluidics. We applied VASA-seq to more than 30,000 single cells in the developing mouse embryo during gastrulation and early organogenesis. Analyzing the dynamics of the total single-cell transcriptome, we discovered cell type markers, many based on non-coding RNA, and performed in vivo cell cycle analysis via detection of non-polyadenylated histone genes. RNA velocity characterization was improved, accurately retracing blood maturation trajectories. Moreover, our VASA-seq data provide a comprehensive analysis of alternative splicing during mammalian development, which highlighted substantial rearrangements during blood development and heart morphogenesis.
Collapse
Affiliation(s)
- Fredrik Salmen
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Joachim De Jonghe
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Francis Crick Institute, London, UK
| | - Tomasz S Kaminski
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Anna Alemany
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | | | - Joe Verity-Legg
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Ayaka Yanagida
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Timo N Kohler
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Nicholas Battich
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Floris van den Brekel
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Anna L Ellermann
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Alfonso Martinez Arias
- Systems Bioengineering, DCEXS, Universidad Pompeu Fabra, Doctor Aiguader 88 ICREA (Institució Catalana de Recerca i Estudis Avançats), Barcelona, Spain
| | - Jennifer Nichols
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Martin Hemberg
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | | | - Alexander van Oudenaarden
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center, Utrecht, Netherlands.
- Oncode Institute, Utrecht, Netherlands.
| |
Collapse
|
13
|
Mastrandreas P, Boglari C, Arnold A, Peter F, de Quervain DJF, Papassotiropoulos A, Stetak A. Phosphorylation of MSI-1 is implicated in the regulation of associative memory in Caenorhabditis elegans. PLoS Genet 2022; 18:e1010420. [PMID: 36223338 PMCID: PMC9555661 DOI: 10.1371/journal.pgen.1010420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/09/2022] [Indexed: 11/07/2022] Open
Abstract
The Musashi family of RNA-binding proteins controls several biological processes including stem cell maintenance, cell division and neural function. Previously, we demonstrated that the C. elegans Musashi ortholog, msi-1, regulates forgetting via translational repression of the Arp2/3 actin-branching complex. However, the mechanisms controlling MSI-1 activity during the regulation of forgetting are currently unknown. Here we investigated the effects of protein phosphorylation on MSI-1 activity. We showed that MSI-1 function is likely controlled by alterations of its activity rather than its expression levels. Furthermore, we found that MSI-1 is phosphorylated and using mass spectrometry we identified MSI-1 phosphorylation at three residues (T18, S19 and S34). CRISPR-based manipulations of MSI-1 phosphorylation sites revealed that phosphorylation is necessary for MSI-1 function in both short- and long-term aversive olfactory associative memory. Thus, our study provides insight into the mechanisms regulating memory-related MSI-1 activity and may facilitate the development of novel therapeutic approaches. Understanding neural circuits and molecular mechanisms underlying learning and memory are the major challenges of neuroscience. It is a generally accepted model that a learning event causes modification of synapses; strengthening some within a circuit and weakening others (termed “synaptic plasticity”). A plastic nervous system requires not only the ability to acquire and store but also to forget new inputs. While learning and memory is widely investigated, clear-cut evidence for mechanisms involved in forgetting is still sparse. Previously, we demonstrated the role of the protein Musashi (MSI-1) in the active regulation of forgetting in the nematode C. elegans. Here we investigated the role of protein modification (phosphorylation) as a possible regulatory mechanism of the MSI-1 protein activity. We found that MSI-1 protein is modified at different positions and all of these modifications at the protein level contribute to the correct activity of the protein leading to active forgetting of short and long-term memories.
Collapse
Affiliation(s)
- Pavlina Mastrandreas
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
| | - Csaba Boglari
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
| | - Andreas Arnold
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
| | - Fabian Peter
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
| | - Dominique J.-F. de Quervain
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Cognitive Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- University Psychiatric Clinics, University of Basel, Basel, Switzerland
| | - Andreas Papassotiropoulos
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
- University Psychiatric Clinics, University of Basel, Basel, Switzerland
| | - Attila Stetak
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
- University Psychiatric Clinics, University of Basel, Basel, Switzerland
- * E-mail:
| |
Collapse
|
14
|
Invited review: Unearthing the mechanisms of age-related neurodegenerative disease using Caenorhabditis elegans. Comp Biochem Physiol A Mol Integr Physiol 2022; 267:111166. [PMID: 35176489 DOI: 10.1016/j.cbpa.2022.111166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022]
Abstract
As human life expectancy increases, neurodegenerative diseases present a growing public health threat, for which there are currently few effective treatments. There is an urgent need to understand the molecular and genetic underpinnings of these disorders so new therapeutic targets can be identified. Here we present the argument that the simple nematode worm Caenorhabditis elegans is a powerful tool to rapidly study neurodegenerative disorders due to their short lifespan and vast array of genetic tools, which can be combined with characterization of conserved neuronal processes and behavior orthologous to those disrupted in human disease. We review how pre-existing C. elegans models provide insight into human neurological disease as well as an overview of current tools available to study neurodegenerative diseases in the worm, with an emphasis on genetics and behavior. We also discuss open questions that C. elegans may be particularly well suited for in future studies and how worms will be a valuable preclinical model to better understand these devastating neurological disorders.
Collapse
|
15
|
Nguyen NM, Vellichirammal NN, Guda C, Pendyala G. Decoding the Synaptic Proteome with Long-Term Exposure to Midazolam during Early Development. Int J Mol Sci 2022; 23:ijms23084137. [PMID: 35456952 PMCID: PMC9027542 DOI: 10.3390/ijms23084137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 12/05/2022] Open
Abstract
The intensive use of anesthetic and sedative agents in the neonatal intensive care unit (NICU) has raised controversial concerns about the potential neurodevelopmental risks. This study focused on midazolam (MDZ), a common benzodiazepine regularly used as a sedative on neonates in the NICU. Mounting evidence suggests a single exposure to MDZ during the neonatal period leads to learning disturbances. However, a knowledge gap that remains is how long-term exposure to MDZ during very early stages of life impacts synaptic alterations. Using a preclinical rodent model system, we mimicked a dose-escalation regimen on postnatal day 3 (P3) pups until day 21. Next, purified synaptosomes from P21 control and MDZ animals were subjected to quantitative mass-spectrometry-based proteomics, to identify potential proteomic signatures. Further analysis by ClueGO identified enrichment of proteins associated with actin-binding and protein depolymerization process. One potential hit identified was alpha adducin (ADD1), belonging to the family of cytoskeleton proteins, which was upregulated in the MDZ group and whose expression was further validated by Western blot. In summary, this study sheds new information on the long-term exposure of MDZ during the early stages of development impacts synaptic function, which could subsequently perturb neurobehavioral outcomes at later stages of life.
Collapse
Affiliation(s)
- Nghi M. Nguyen
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.N.V.); (C.G.)
| | - Neetha N. Vellichirammal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.N.V.); (C.G.)
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.N.V.); (C.G.)
| | - Gurudutt Pendyala
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.N.V.); (C.G.)
- Child Health Research Institute, Omaha, NE 68198, USA
- Correspondence: ; Tel.: +1-402-559-8690
| |
Collapse
|
16
|
Variants in ADD1 cause intellectual disability, corpus callosum dysgenesis, and ventriculomegaly in humans. Genet Med 2022; 24:319-331. [PMID: 34906466 PMCID: PMC8802223 DOI: 10.1016/j.gim.2021.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/24/2021] [Accepted: 09/21/2021] [Indexed: 02/03/2023] Open
Abstract
PURPOSE Adducins interconnect spectrin and actin filaments to form polygonal scaffolds beneath the cell membranes and form ring-like structures in neuronal axons. Adducins regulate mouse neural development, but their function in the human brain is unknown. METHODS We used exome sequencing to uncover ADD1 variants associated with intellectual disability (ID) and brain malformations. We studied ADD1 splice isoforms in mouse and human neocortex development with RNA sequencing, super resolution imaging, and immunoblotting. We investigated 4 variant ADD1 proteins and heterozygous ADD1 cells for protein expression and ADD1-ADD2 dimerization. We studied Add1 functions in vivo using Add1 knockout mice. RESULTS We uncovered loss-of-function ADD1 variants in 4 unrelated individuals affected by ID and/or structural brain defects. Three additional de novo copy number variations covering the ADD1 locus were associated with ID and brain malformations. ADD1 is highly expressed in the neocortex and the corpus callosum, whereas ADD1 splice isoforms are dynamically expressed between cortical progenitors and postmitotic neurons. Human variants impair ADD1 protein expression and/or dimerization with ADD2. Add1 knockout mice recapitulate corpus callosum dysgenesis and ventriculomegaly phenotypes. CONCLUSION Our human and mouse genetics results indicate that pathogenic ADD1 variants cause corpus callosum dysgenesis, ventriculomegaly, and/or ID.
Collapse
|
17
|
Rahmani A, Chew YL. Investigating the molecular mechanisms of learning and memory using Caenorhabditis elegans. J Neurochem 2021; 159:417-451. [PMID: 34528252 DOI: 10.1111/jnc.15510] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/15/2021] [Accepted: 09/08/2021] [Indexed: 11/30/2022]
Abstract
Learning is an essential biological process for survival since it facilitates behavioural plasticity in response to environmental changes. This process is mediated by a wide variety of genes, mostly expressed in the nervous system. Many studies have extensively explored the molecular and cellular mechanisms underlying learning and memory. This review will focus on the advances gained through the study of the nematode Caenorhabditis elegans. C. elegans provides an excellent system to study learning because of its genetic tractability, in addition to its invariant, compact nervous system (~300 neurons) that is well-characterised at the structural level. Importantly, despite its compact nature, the nematode nervous system possesses a high level of conservation with mammalian systems. These features allow the study of genes within specific sensory-, inter- and motor neurons, facilitating the interrogation of signalling pathways that mediate learning via defined neural circuits. This review will detail how learning and memory can be studied in C. elegans through behavioural paradigms that target distinct sensory modalities. We will also summarise recent studies describing mechanisms through which key molecular and cellular pathways are proposed to affect associative and non-associative forms of learning.
Collapse
Affiliation(s)
- Aelon Rahmani
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Yee Lian Chew
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
18
|
Fenyves BG, Arnold A, Gharat VG, Haab C, Tishinov K, Peter F, de Quervain D, Papassotiropoulos A, Stetak A. Dual Role of an mps-2/KCNE-Dependent Pathway in Long-Term Memory and Age-Dependent Memory Decline. Curr Biol 2020; 31:527-539.e7. [PMID: 33259792 DOI: 10.1016/j.cub.2020.10.069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/14/2020] [Accepted: 10/21/2020] [Indexed: 01/24/2023]
Abstract
Activity-dependent persistent changes in neuronal intrinsic excitability and synaptic strength are underlying learning and memory. Voltage-gated potassium (Kv) channels are potential regulators of memory and may be linked to age-dependent neuronal disfunction. MinK-related peptides (MiRPs) are conserved transmembrane proteins modulating Kv channels; however, their possible role in the regulation of memory and age-dependent memory decline are unknown. Here, we show that, in C. elegans, mps-2 is the sole member of the MiRP family that controls exclusively long-term associative memory (LTAM) in AVA neuron. In addition, we demonstrate that mps-2 also plays a critical role in age-dependent memory decline. In young adult worms, mps-2 is transcriptionally upregulated by CRH-1/cyclic AMP (cAMP)-response-binding protein (CREB) during LTAM, although the mps-2 baseline expression is CREB independent and instead, during aging, relies on nhr-66, which acts as an age-dependent repressor. Deletion of nhr-66 or its binding element in the mps-2 promoter prevents age-dependent transcriptional repression of mps-2 and memory decline. Finally, MPS-2 acts through the modulation of the Kv2.1/KVS-3 and Kv2.2/KVS-4 heteromeric potassium channels. Altogether, we describe a conserved MPS-2/KVS-3/KVS-4 pathway essential for LTAM and also for a programmed control of physiological age-dependent memory decline.
Collapse
Affiliation(s)
- Bank G Fenyves
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, 1094 Budapest, Hungary
| | - Andreas Arnold
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland
| | - Vaibhav G Gharat
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland
| | - Carmen Haab
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland
| | - Kiril Tishinov
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Fabian Peter
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland
| | - Dominique de Quervain
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Cognitive Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; University Psychiatric Clinics, University of Basel, Wilhelm Klein-Strasse 27, 4055 Basel, Switzerland
| | - Andreas Papassotiropoulos
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Biozentrum, Life Sciences Training Facility, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland; University Psychiatric Clinics, University of Basel, Wilhelm Klein-Strasse 27, 4055 Basel, Switzerland
| | - Attila Stetak
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; Division of Molecular Neuroscience, Department of Psychology, University of Basel, Birmannsgasse 8, 4055 Basel, Switzerland; University Psychiatric Clinics, University of Basel, Wilhelm Klein-Strasse 27, 4055 Basel, Switzerland.
| |
Collapse
|
19
|
Fadda M, De Fruyt N, Borghgraef C, Watteyne J, Peymen K, Vandewyer E, Naranjo Galindo FJ, Kieswetter A, Mirabeau O, Chew YL, Beets I, Schoofs L. NPY/NPF-Related Neuropeptide FLP-34 Signals from Serotonergic Neurons to Modulate Aversive Olfactory Learning in Caenorhabditis elegans. J Neurosci 2020; 40:6018-6034. [PMID: 32576621 PMCID: PMC7392509 DOI: 10.1523/jneurosci.2674-19.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/26/2020] [Accepted: 06/12/2020] [Indexed: 02/03/2023] Open
Abstract
Aversive learning is fundamental for animals to increase chances of survival. In addition to classical neurotransmitters, neuropeptides have emerged to modulate such complex behaviors. Among them, neuropeptide Y (NPY) is well known to promote aversive memory acquisition in mammals. Here we identify an NPY/neuropeptide F (NPF)-related neuropeptide system in Caenorhabditis elegans and show that this FLP-34/NPR-11 system is required for learning negative associations, a process that is reminiscent of NPY signaling in mammals. The Caenorhabditis elegans NPY/NPF ortholog FLP-34 displays conserved structural hallmarks of bilaterian-wide NPY/NPF neuropeptides. We show that it is required for aversive olfactory learning after pairing diacetyl with the absence of food, but not for appetitive olfactory learning in response to butanone. To mediate diacetyl learning and thus integrate the aversive food context with the diacetyl odor, FLP-34 is released from serotonergic neurons and signals through its evolutionarily conserved NPY/NPF GPCR, NPR-11, in downstream AIA interneurons. NPR-11 activation in the AIA integration center results in avoidance of a previously attractive stimulus. This study opens perspectives for a deeper understanding of stress conditions in which aversive learning results in excessive avoidance.SIGNIFICANCE STATEMENT Aversive learning evolved early in evolution to promote avoidance of dangerous and stressful situations. In addition to classical neurotransmitters, neuropeptides are emerging as modulators of complex behaviors, including learning and memory. Here, we identified the evolutionary ortholog of neuropeptide Y/neuropeptide F in the nematode Caenorhabditis elegans, and we discovered that it is required for olfactory aversive learning. In addition, we elucidated the neural circuit underlying this avoidance behavior, and we discovered a novel coordinated action of Caenorhabditis elegans neuropeptide Y/neuropeptide F and serotonin that could aid in our understanding of the molecular mechanisms underlying stress disorders in which excessive avoidance results in maladaptive behaviors.
Collapse
Affiliation(s)
- Melissa Fadda
- Department of Biology, KU Leuven, Leuven, 3000, Belgium
| | | | | | - Jan Watteyne
- Department of Biology, KU Leuven, Leuven, 3000, Belgium
| | | | | | | | | | - Olivier Mirabeau
- Genetics and Biology of Cancers Unit, Institut Curie, Institut National de la Santé et de la Recherche Médicale U830, Paris Sciences et Lettres Research University, Paris, 75005, France
| | - Yee Lian Chew
- Illawarra Health & Medical Research Institute School of Chemistry & Molecular Bioscience, University of Wollongong, Wollongong, 2522 New South Wales, Australia
| | - Isabel Beets
- Department of Biology, KU Leuven, Leuven, 3000, Belgium
| | | |
Collapse
|
20
|
Vukojevic V, Mastrandreas P, Arnold A, Peter F, Kolassa IT, Wilker S, Elbert T, de Quervain DJF, Papassotiropoulos A, Stetak A. Evolutionary conserved role of neural cell adhesion molecule-1 in memory. Transl Psychiatry 2020; 10:217. [PMID: 32632143 PMCID: PMC7338365 DOI: 10.1038/s41398-020-00899-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 06/11/2020] [Accepted: 06/16/2020] [Indexed: 01/17/2023] Open
Abstract
The neural cell adhesion molecule 1 (NCAM-1) has been implicated in several brain-related biological processes, including neuronal migration, axonal branching, fasciculation, and synaptogenesis, with a pivotal role in synaptic plasticity. Here, we investigated the evolutionary conserved role of NCAM-1 in learning and memory. First, we investigated sustained changes in ncam-1 expression following aversive olfactory conditioning in C. elegans using molecular genetic methods. Furthermore, we examined the link between epigenetic signatures of the NCAM1 gene and memory in two human samples of healthy individuals (N = 568 and N = 319) and in two samples of traumatized individuals (N = 350 and N = 463). We found that olfactory conditioning in C. elegans induced ncam-1 expression and that loss of ncam-1 function selectively impaired associative long-term memory, without causing acquisition, sensory, or short-term memory deficits. Reintroduction of the C. elegans or human NCAM1 fully rescued memory impairment, suggesting a conserved role of NCAM1 for memory. In parallel, DNA methylation of the NCAM1 promoter in two independent healthy Swiss cohorts was associated with memory performance. In two independent Sub-Saharan populations of conflict zone survivors who had faced severe trauma, DNA methylation at an alternative promoter of the NCAM1 gene was associated with traumatic memories. Our results support a role of NCAM1 in associative memory in nematodes and humans, and might, ultimately, be helpful in elucidating diagnostic markers or suggest novel therapy targets for memory-related disorders, like PTSD.
Collapse
Affiliation(s)
- Vanja Vukojevic
- University of Basel, Department of Psychology, Division of Molecular Neuroscience, Birmannsgasse 8, CH-4055, Basel, Switzerland.
- University of Basel, Department Biozentrum, Life Sciences Training Facility, Klingelbergstrasse 50-70, CH-4056, Basel, Switzerland.
- University of Basel, Psychiatric University Clinics, Wilhelm Klein-Strasse 27, CH-4012, Basel, Switzerland.
- University of Basel, Transfaculty Research Platform, Birmannsgasse 8, CH-4055, Basel, Switzerland.
| | - Pavlina Mastrandreas
- University of Basel, Department of Psychology, Division of Molecular Neuroscience, Birmannsgasse 8, CH-4055, Basel, Switzerland.
- University of Basel, Department Biozentrum, Life Sciences Training Facility, Klingelbergstrasse 50-70, CH-4056, Basel, Switzerland.
- University of Basel, Transfaculty Research Platform, Birmannsgasse 8, CH-4055, Basel, Switzerland.
| | - Andreas Arnold
- University of Basel, Department of Psychology, Division of Molecular Neuroscience, Birmannsgasse 8, CH-4055, Basel, Switzerland
- University of Basel, Department Biozentrum, Life Sciences Training Facility, Klingelbergstrasse 50-70, CH-4056, Basel, Switzerland
- University of Basel, Transfaculty Research Platform, Birmannsgasse 8, CH-4055, Basel, Switzerland
| | - Fabian Peter
- University of Basel, Department of Psychology, Division of Molecular Neuroscience, Birmannsgasse 8, CH-4055, Basel, Switzerland
- University of Basel, Department Biozentrum, Life Sciences Training Facility, Klingelbergstrasse 50-70, CH-4056, Basel, Switzerland
- University of Basel, Transfaculty Research Platform, Birmannsgasse 8, CH-4055, Basel, Switzerland
| | - Iris-T Kolassa
- Ulm University, Clinical & Biological Psychology, Institute for Psychology & Education, Albert-Einstein-Allee 47, D-89069, Ulm, Germany
| | - Sarah Wilker
- Ulm University, Clinical & Biological Psychology, Institute for Psychology & Education, Albert-Einstein-Allee 47, D-89069, Ulm, Germany
- University Bielefeld, Department for Psychology and Sports Science, P.O. Box 100131, D-33501, Bielefeld, Germany
| | - Thomas Elbert
- University of Konstanz, Clinical Psychology & Behavioural Neuroscience, D-78457, Konstanz, Germany
| | - Dominique J-F de Quervain
- University of Basel, Department of Psychology, Division of Molecular Neuroscience, Birmannsgasse 8, CH-4055, Basel, Switzerland
- University of Basel, Transfaculty Research Platform, Birmannsgasse 8, CH-4055, Basel, Switzerland
- University of Basel, Department of Psychology, Division of Cognitive Neuroscience, Birmannsgasse 8, CH-4055, Basel, Switzerland
| | - Andreas Papassotiropoulos
- University of Basel, Department of Psychology, Division of Molecular Neuroscience, Birmannsgasse 8, CH-4055, Basel, Switzerland.
- University of Basel, Department Biozentrum, Life Sciences Training Facility, Klingelbergstrasse 50-70, CH-4056, Basel, Switzerland.
- University of Basel, Psychiatric University Clinics, Wilhelm Klein-Strasse 27, CH-4012, Basel, Switzerland.
- University of Basel, Transfaculty Research Platform, Birmannsgasse 8, CH-4055, Basel, Switzerland.
| | - Attila Stetak
- University of Basel, Department of Psychology, Division of Molecular Neuroscience, Birmannsgasse 8, CH-4055, Basel, Switzerland
- University of Basel, Department Biozentrum, Life Sciences Training Facility, Klingelbergstrasse 50-70, CH-4056, Basel, Switzerland
- University of Basel, Psychiatric University Clinics, Wilhelm Klein-Strasse 27, CH-4012, Basel, Switzerland
- University of Basel, Transfaculty Research Platform, Birmannsgasse 8, CH-4055, Basel, Switzerland
| |
Collapse
|
21
|
Zhu J, Lee KY, Jong TT, Tsai NP. C2-lacking isoform of Nedd4-2 regulates excitatory synaptic strength through GluA1 ubiquitination-independent mechanisms. J Neurochem 2019; 151:289-300. [PMID: 31357244 DOI: 10.1111/jnc.14840] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 01/06/2023]
Abstract
Neural precursor cell expressed developmentally downregulated gene 4-like (Nedd4-2) is an epilepsy-associated gene, which encodes a ubiquitin E3 ligase that is highly expressed in the brain. Nedd4-2's substrates include many ion channels and receptors because its N-terminal C2 domain guides Nedd4-2 to the cell membrane. We previously found that Nedd4-2 ubiquitinates the glutamate receptor subunit 1 (GluA1) subunit of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor, which leads to reduction of neuronal excitability and seizures in mice. However, despite awareness of a Nedd4-2 isoform with no C2 domain, the functions of this isoform remain elusive. In this study, we showed that the C2-lacking Nedd4-2 has reduced membrane distribution and exhibits reduced affinity toward ubiquitinating GluA1. However, when expressed in primary cortical neurons, we found that the C2-lacking Nedd4-2 exhibits a similar activity toward reducing excitatory synaptic strength as does the C2-containing Nedd4-2. In an attempt to identify novel Nedd4-2 substrates that could mediate excitatory synaptic strength, we used unbiased proteomic screening and found multiple synaptic regulators that were up-regulated in the brain of conditional Nedd4-2 knockout mice, including protein phosphatase 3 catalytic subunit-α (PPP3CA; alternately called calcineurin A-α). We confirmed PPP3CA as a substrate of the C2-lacking Nedd4-2 and showed that all three epilepsy-associated missense mutations of Nedd4-2 disrupted PPP3CA ubiquitination. Altogether, our results revealed novel potential Nedd4-2 substrates and suggest that the C2-lacking Nedd4-2 represses excitatory synaptic strength most likely through GluA1 ubiquitination-independent mechanisms. These findings provide novel information to further our knowledge about Nedd4-2-dependent neuronal excitability homeostasis and pathological hyperexcitability when Nedd4-2 is compromised.
Collapse
Affiliation(s)
- Jiuhe Zhu
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Tiffany T Jong
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
22
|
Goswami AM. α-Adducin nsSNPs affect mRNA secondary structure, protein modification and stability. Meta Gene 2018. [DOI: 10.1016/j.mgene.2018.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
23
|
Aoki I, Tateyama M, Shimomura T, Ihara K, Kubo Y, Nakano S, Mori I. SLO potassium channels antagonize premature decision making in C. elegans. Commun Biol 2018; 1:123. [PMID: 30272003 PMCID: PMC6123717 DOI: 10.1038/s42003-018-0124-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Abstract
Animals must modify their behavior with appropriate timing to respond to environmental changes. Yet, the molecular and neural mechanisms regulating the timing of behavioral transition remain largely unknown. By performing forward genetics to reveal mechanisms that underlie the plasticity of thermotaxis behavior in C. elegans, we demonstrated that SLO potassium channels and a cyclic nucleotide-gated channel, CNG-3, determine the timing of transition of temperature preference after a shift in cultivation temperature. We further revealed that SLO and CNG-3 channels act in thermosensory neurons and decelerate alteration in the responsiveness of these neurons, which occurs prior to the preference transition after a temperature shift. Our results suggest that regulation of sensory adaptation is a major determinant of latency before animals make decisions to change their behavior.
Collapse
Affiliation(s)
- Ichiro Aoki
- Neuroscience Institute of the Graduate School of Science, Nagoya University, Nagoya, 464-8602, Japan
- Group of Molecular Neurobiology, Graduate School of Science, Nnagoya University, Nagoya, 464-8602, Japan
| | - Michihiro Tateyama
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Takushi Shimomura
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Kunio Ihara
- Center for Gene Research, Nagoya University, Nagoya, 464-8602, Japan
| | - Yoshihiro Kubo
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Shunji Nakano
- Neuroscience Institute of the Graduate School of Science, Nagoya University, Nagoya, 464-8602, Japan
- Group of Molecular Neurobiology, Graduate School of Science, Nnagoya University, Nagoya, 464-8602, Japan
| | - Ikue Mori
- Neuroscience Institute of the Graduate School of Science, Nagoya University, Nagoya, 464-8602, Japan.
- Group of Molecular Neurobiology, Graduate School of Science, Nnagoya University, Nagoya, 464-8602, Japan.
| |
Collapse
|
24
|
Arey RN, Stein GM, Kaletsky R, Kauffman A, Murphy CT. Activation of G αq Signaling Enhances Memory Consolidation and Slows Cognitive Decline. Neuron 2018; 98:562-574.e5. [PMID: 29656871 DOI: 10.1016/j.neuron.2018.03.039] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/06/2017] [Accepted: 03/21/2018] [Indexed: 01/25/2023]
Abstract
Perhaps the most devastating decline with age is the loss of memory. Therefore, identifying mechanisms to restore memory function with age is critical. Using C. elegans associative learning and memory assays, we identified a gain-of-function Gαq signaling pathway mutant that forms a long-term (cAMP response element binding protein [CREB]-dependent) memory following one conditioned stimulus-unconditioned stimulus (CS-US) pairing, which usually requires seven CS-US pairings. Increased CREB activity in AIM interneurons reduces the threshold for memory consolidation through transcription of a set of previously identified "long-term memory" genes. Enhanced Gαq signaling in the AWC sensory neuron is both necessary and sufficient for improved memory and increased AIM CREB activity, and activation of Gαq specifically in aged animals rescues the ability to form memory. Activation of Gαq in AWC sensory neurons non-cell autonomously induces consolidation after one CS-US pairing, enabling both cognitive function maintenance with age and restoration of memory function in animals with impaired memory performance without decreased longevity.
Collapse
Affiliation(s)
- Rachel N Arey
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Geneva M Stein
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Rachel Kaletsky
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Amanda Kauffman
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Coleen T Murphy
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
25
|
Yaguchi H, Yabe I, Takahashi H, Watanabe M, Nomura T, Kano T, Matsumoto M, Nakayama KI, Watanabe M, Hatakeyama S. Sez6l2 regulates phosphorylation of ADD and neuritogenesis. Biochem Biophys Res Commun 2017; 494:234-241. [PMID: 29032200 DOI: 10.1016/j.bbrc.2017.10.047] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 10/10/2017] [Indexed: 12/11/2022]
Abstract
Increasing evidence shows that immune-mediated mechanisms may contribute to the pathogenesis of central nervous system disorders including cerebellar ataxias, as indicated by the aberrant production of neuronal surface antibodies. We previously reported a patient with cerebellar ataxia associated with production of a new anti-neuronal antibody, anti-seizure-related 6 homolog like 2 (Sez6l2). Sez6l2 is a type 1 membrane protein that is highly expressed in the hippocampus and cerebellar cortex and mice lacking Sez6l2 protein family members develop ataxia. Here we used a proteomics-based approach to show that serum derived from this patient recognizes the extracellular domain of Sez6l2 and that Sez6l2 protein binds to both adducin (ADD) and glutamate receptor 1 (GluR1). Our results indicate that Sez6l2 is one of the auxiliary subunits of the AMPA receptor and acts as a scaffolding protein to link GluR1 to ADD. Furthermore, Sez6l2 overexpression upregulates ADD phosphorylation, whereas siRNA-mediated downregulation of Sez612 prevents ADD phosphorylation, suggesting that Sez6l2 modulates AMPA-ADD signal transduction.
Collapse
Affiliation(s)
- Hiroaki Yaguchi
- Department of Neurology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan; Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan; Department of Neurology, Brain Science Center, Sapporo City General Hospital, Sapporo, Hokkaido, Japan.
| | - Ichiro Yabe
- Department of Neurology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hidehisa Takahashi
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masashi Watanabe
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Taichi Nomura
- Department of Neurology, Brain Science Center, Sapporo City General Hospital, Sapporo, Hokkaido, Japan
| | - Takahiro Kano
- Department of Neurology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masaki Matsumoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Shigetsugu Hatakeyama
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan.
| |
Collapse
|
26
|
Multiple Signaling Pathways Coordinately Regulate Forgetting of Olfactory Adaptation through Control of Sensory Responses in Caenorhabditis elegans. J Neurosci 2017; 37:10240-10251. [PMID: 28924007 DOI: 10.1523/jneurosci.0031-17.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 09/04/2017] [Indexed: 01/08/2023] Open
Abstract
Forgetting memories is important for animals to properly respond to continuously changing environments. To elucidate the mechanisms of forgetting, we used one of the behavioral plasticities of Caenorhabditis elegans hermaphrodite, olfactory adaptation to an attractive odorant, diacetyl, as a simple model of learning. In C. elegans, the TIR-1/JNK-1 pathway accelerates forgetting of olfactory adaptation by facilitating neural secretion from AWC sensory neurons. In this study, to identify the downstream effectors of the TIR-1/JNK-1 pathway, we conducted a genetic screen for suppressors of the gain-of-function mutant of tir-1 (ok1052), which shows excessive forgetting. Our screening showed that three proteins-a membrane protein, MACO-1; a receptor tyrosine kinase, SCD-2; and its putative ligand, HEN-1-regulated forgetting downstream of the TIR-1/JNK-1 pathway. We further demonstrated that MACO-1 and SCD-2/HEN-1 functioned in parallel genetic pathways, and only MACO-1 regulated forgetting of olfactory adaptation to isoamyl alcohol, which is an attractive odorant sensed by different types of sensory neurons. In olfactory adaptation, odor-evoked Ca2+ responses in olfactory neurons are attenuated by conditioning and recovered thereafter. A Ca2+ imaging study revealed that this attenuation is sustained longer in maco-1 and scd-2 mutant animals than in wild-type animals like the TIR-1/JNK-1 pathway mutants. Furthermore, temporal silencing by histamine-gated chloride channels revealed that the neuronal activity of AWC neurons after conditioning is important for proper forgetting. We propose that distinct signaling pathways, each of which has a specific function, may coordinately and temporally regulate forgetting by controlling sensory responses.SIGNIFICANCE STATEMENT Active forgetting is an important process to understand the whole mechanisms of memories. Recent papers have reported that the noncell autonomous regulations are required for proper forgetting in invertebrates. We found that in Caenorhabditis elegans hermaphrodite, the noncell autonomous regulations of forgetting of olfactory adaptation is regulated by three conserved proteins: a membrane protein, MACO-1; a receptor tyrosine kinase, SCD-2: and its ligand, HEN-1. MACO-1 and SCD-2/HEN-1, working in coordination, accelerate forgetting by controlling sensory responses in parallel. Furthermore, temporal regulation of neuronal activity is important for proper forgetting. We suggest that multiple pathways may coordinately and temporally regulate forgetting through control of sensory responses. This study should lead to a better understanding of forgetting in higher organisms.
Collapse
|
27
|
Genome-Wide Temporal Expression Profiling in Caenorhabditis elegans Identifies a Core Gene Set Related to Long-Term Memory. J Neurosci 2017; 37:6661-6672. [PMID: 28592692 DOI: 10.1523/jneurosci.3298-16.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 04/09/2017] [Accepted: 05/10/2017] [Indexed: 11/21/2022] Open
Abstract
The identification of genes related to encoding, storage, and retrieval of memories is a major interest in neuroscience. In the current study, we analyzed the temporal gene expression changes in a neuronal mRNA pool during an olfactory long-term associative memory (LTAM) in Caenorhabditis elegans hermaphrodites. Here, we identified a core set of 712 (538 upregulated and 174 downregulated) genes that follows three distinct temporal peaks demonstrating multiple gene regulation waves in LTAM. Compared with the previously published positive LTAM gene set (Lakhina et al., 2015), 50% of the identified upregulated genes here overlap with the previous dataset, possibly representing stimulus-independent memory-related genes. On the other hand, the remaining genes were not previously identified in positive associative memory and may specifically regulate aversive LTAM. Our results suggest a multistep gene activation process during the formation and retrieval of long-term memory and define general memory-implicated genes as well as conditioning-type-dependent gene sets.SIGNIFICANCE STATEMENT The identification of genes regulating different steps of memory is of major interest in neuroscience. Identification of common memory genes across different learning paradigms and the temporal activation of the genes are poorly studied. Here, we investigated the temporal aspects of Caenorhabditis elegans gene expression changes using aversive olfactory associative long-term memory (LTAM) and identified three major gene activation waves. Like in previous studies, aversive LTAM is also CREB dependent, and CREB activity is necessary immediately after training. Finally, we define a list of memory paradigm-independent core gene sets as well as conditioning-dependent genes.
Collapse
|
28
|
The intestinal TORC2 signaling pathway contributes to associative learning in Caenorhabditis elegans. PLoS One 2017; 12:e0177900. [PMID: 28542414 PMCID: PMC5444632 DOI: 10.1371/journal.pone.0177900] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 05/04/2017] [Indexed: 12/25/2022] Open
Abstract
Several types of associative learning are dependent upon the presence or absence of food, and are crucial for the survival of most animals. Target of rapamycin (TOR), a kinase which exists as a component of two complexes, TOR complex 1 (TORC1) and TOR complex 2 (TORC2), is known to act as a nutrient sensor in numerous organisms. However, the in vivo roles of TOR signaling in the nervous system remain largely unclear, partly because its multifunctionality and requirement for survival make it difficult to investigate. Here, using pharmacological inhibitors and genetic analyses, we show that TORC1 and TORC2 contribute to associative learning between salt and food availability in the nematode Caenorhabditis elegans in a process called taste associative learning. Worms migrate to salt concentrations experienced previously during feeding, but they avoid salt concentrations experienced under starvation conditions. Administration of the TOR inhibitor rapamycin causes a behavioral defect after starvation conditioning. Worms lacking either RICT-1 or SINH-1, two TORC2 components, show defects in migration to high salt levels after learning under both fed and starved conditions. We also analyzed the behavioral phenotypes of mutants of the putative TORC1 substrate RSKS-1 (the C. elegans homolog of the mammalian S6 kinase S6K) and the putative TORC2 substrates SGK-1 and PKC-2 (homologs of the serum and glucocorticoid-induced kinase 1, SGK1, and protein kinase C-α, PKC-α, respectively) and found that neuronal RSKS-1 and PKC-2, as well as intestinal SGK-1, are involved in taste associative learning. Our findings shed light on the functions of TOR signaling in behavioral plasticity and provide insight into the mechanisms by which information sensed in the intestine affects the nervous system to modulate food-searching behaviors.
Collapse
|
29
|
Arey RN, Murphy CT. Conserved regulators of cognitive aging: From worms to humans. Behav Brain Res 2016; 322:299-310. [PMID: 27329151 DOI: 10.1016/j.bbr.2016.06.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/27/2016] [Accepted: 06/17/2016] [Indexed: 01/25/2023]
Abstract
Cognitive decline is a major deficit that arises with age in humans. While some research on the underlying causes of these problems can be done in humans, harnessing the strengths of small model systems, particularly those with well-studied longevity mutants, such as the nematode C. elegans, will accelerate progress. Here we review the approaches being used to study cognitive decline in model organisms and show how simple model systems allow the rapid discovery of conserved molecular mechanisms, which will eventually enable the development of therapeutics to slow cognitive aging.
Collapse
Affiliation(s)
- Rachel N Arey
- Department of Molecular Biology & LSI Genomics, Princeton University, Princeton, NJ 08544, United States
| | - Coleen T Murphy
- Department of Molecular Biology & LSI Genomics, Princeton University, Princeton, NJ 08544, United States.
| |
Collapse
|
30
|
Krieger C, Wang SJH, Yoo SH, Harden N. Adducin at the Neuromuscular Junction in Amyotrophic Lateral Sclerosis: Hanging on for Dear Life. Front Cell Neurosci 2016; 10:11. [PMID: 26858605 PMCID: PMC4731495 DOI: 10.3389/fncel.2016.00011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/13/2016] [Indexed: 12/11/2022] Open
Abstract
The neurological dysfunction in amyotrophic lateral sclerosis (ALS)/motor neurone disease (MND) is associated with defective nerve-muscle contacts early in the disease suggesting that perturbations of cell adhesion molecules (CAMs) linking the pre- and post-synaptic components of the neuromuscular junction (NMJ) are involved. To search for candidate proteins implicated in this degenerative process, researchers have studied the Drosophila larval NMJ and find that the cytoskeleton-associated protein, adducin, is ideally placed to regulate synaptic contacts. By controlling the levels of synaptic proteins, adducin can de-stabilize synaptic contacts. Interestingly, elevated levels of phosphorylated adducin have been reported in ALS patients and in a mouse model of the disease. Adducin is regulated by phosphorylation through protein kinase C (PKC), some isoforms of which exhibit Ca2+-dependence, raising the possibility that changes in intracellular Ca2+ might alter PKC activation and secondarily influence adducin phosphorylation. Furthermore, adducin has interactions with the alpha subunit of the Na+/K+-ATPase. Thus, the phosphorylation of adducin may secondarily influence synaptic stability at the NMJ and so influence pre- and post-synaptic integrity at the NMJ in ALS.
Collapse
Affiliation(s)
- Charles Krieger
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University Burnaby, BC, Canada
| | - Simon Ji Hau Wang
- Department of Biomedical Physiology and Kinesiology, Simon Fraser UniversityBurnaby, BC, Canada; Department of Molecular Biology and Biochemistry, Simon Fraser UniversityBurnaby, BC, Canada
| | - Soo Hyun Yoo
- Department of Biomedical Physiology and Kinesiology, Simon Fraser UniversityBurnaby, BC, Canada; Department of Molecular Biology and Biochemistry, Simon Fraser UniversityBurnaby, BC, Canada
| | - Nicholas Harden
- Department of Molecular Biology and Biochemistry, Simon Fraser University Burnaby, BC, Canada
| |
Collapse
|
31
|
Bosia M, Pigoni A, Zagato L, Merlino L, Casamassima N, Lorenzi C, Pirovano A, Smeraldi E, Manunta P, Cavallaro R. ADDing a piece to the puzzle of cognition in schizophrenia. Eur J Med Genet 2016; 59:26-31. [DOI: 10.1016/j.ejmg.2015.12.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 12/02/2015] [Accepted: 12/21/2015] [Indexed: 12/18/2022]
|
32
|
Gyurkó MD, Csermely P, Sőti C, Steták A. Distinct roles of the RasGAP family proteins in C. elegans associative learning and memory. Sci Rep 2015; 5:15084. [PMID: 26469632 PMCID: PMC4606830 DOI: 10.1038/srep15084] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/09/2015] [Indexed: 12/14/2022] Open
Abstract
The Ras GTPase activating proteins (RasGAPs) are regulators of the conserved Ras/MAPK pathway. Various roles of some of the RasGAPs in learning and memory have been reported in different model systems, yet, there is no comprehensive study to characterize all gap genes in any organism. Here, using reverse genetics and neurobehavioural tests, we studied the role of all known genes of the rasgap family in C. elegans in associative learning and memory. We demonstrated that their proteins are implicated in different parts of the learning and memory processes. We show that gap-1 contribute redundantly with gap-3 to the chemosensation of volatile compounds, gap-1 plays a major role in associative learning, while gap-2 and gap-3 are predominantly required for short- and long-term associative memory. Our results also suggest that the C. elegans Ras orthologue let-60 is involved in multiple processes during learning and memory. Thus, we show that the different classes of RasGAP proteins are all involved in cognitive function and their complex interplay ensures the proper formation and storage of novel information in C. elegans.
Collapse
Affiliation(s)
- M Dávid Gyurkó
- Semmelweis University, Department of Medical Chemistry, Budapest, Üllői út 26 1085, Hungary
| | - Péter Csermely
- Semmelweis University, Department of Medical Chemistry, Budapest, Üllői út 26 1085, Hungary
| | - Csaba Sőti
- Semmelweis University, Department of Medical Chemistry, Budapest, Üllői út 26 1085, Hungary
| | - Attila Steták
- University of Basel, Transfaculty Research Platform Molecular and Cognitive Neurosciences, Birmannsgasse 8, 4055 Basel, Switzerland.,University of Basel, University Psychiatric Clinics, Wilhelm Klein-Strasse 27, 4055 Basel, Switzerland
| |
Collapse
|
33
|
Gorrepati L, Krause MW, Chen W, Brodigan TM, Correa-Mendez M, Eisenmann DM. Identification of Wnt Pathway Target Genes Regulating the Division and Differentiation of Larval Seam Cells and Vulval Precursor Cells in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2015; 5:1551-66. [PMID: 26048561 PMCID: PMC4528312 DOI: 10.1534/g3.115.017715] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/18/2015] [Indexed: 12/29/2022]
Abstract
The evolutionarily conserved Wnt/β-catenin signaling pathway plays a fundamental role during metazoan development, regulating numerous processes including cell fate specification, cell migration, and stem cell renewal. Wnt ligand binding leads to stabilization of the transcriptional effector β-catenin and upregulation of target gene expression to mediate a cellular response. During larval development of the nematode Caenorhabditis elegans, Wnt/β-catenin pathways act in fate specification of two hypodermal cell types, the ventral vulval precursor cells (VPCs) and the lateral seam cells. Because little is known about targets of the Wnt signaling pathways acting during larval VPC and seam cell differentiation, we sought to identify genes regulated by Wnt signaling in these two hypodermal cell types. We conditionally activated Wnt signaling in larval animals and performed cell type-specific "mRNA tagging" to enrich for VPC and seam cell-specific mRNAs, and then used microarray analysis to examine gene expression compared to control animals. Two hundred thirty-nine genes activated in response to Wnt signaling were identified, and we characterized 50 genes further. The majority of these genes are expressed in seam and/or vulval lineages during normal development, and reduction of function for nine genes caused defects in the proper division, fate specification, fate execution, or differentiation of seam cells and vulval cells. Therefore, the combination of these techniques was successful at identifying potential cell type-specific Wnt pathway target genes from a small number of cells and at increasing our knowledge of the specification and behavior of these C. elegans larval hypodermal cells.
Collapse
Affiliation(s)
- Lakshmi Gorrepati
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| | | | - Weiping Chen
- Intramural Research Program, NIDDK, Bethesda, Maryland 20814
| | | | - Margarita Correa-Mendez
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| | - David M Eisenmann
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| |
Collapse
|
34
|
Lakhina V, Arey RN, Kaletsky R, Kauffman A, Stein G, Keyes W, Xu D, Murphy CT. Genome-wide functional analysis of CREB/long-term memory-dependent transcription reveals distinct basal and memory gene expression programs. Neuron 2015; 85:330-45. [PMID: 25611510 DOI: 10.1016/j.neuron.2014.12.029] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2014] [Indexed: 12/30/2022]
Abstract
Induced CREB activity is a hallmark of long-term memory, but the full repertoire of CREB transcriptional targets required specifically for memory is not known in any system. To obtain a more complete picture of the mechanisms involved in memory, we combined memory training with genome-wide transcriptional analysis of C. elegans CREB mutants. This approach identified 757 significant CREB/memory-induced targets and confirmed the involvement of known memory genes from other organisms, but also suggested new mechanisms and novel components that may be conserved through mammals. CREB mediates distinct basal and memory transcriptional programs at least partially through spatial restriction of CREB activity: basal targets are regulated primarily in nonneuronal tissues, while memory targets are enriched for neuronal expression, emanating from CREB activity in AIM neurons. This suite of novel memory-associated genes will provide a platform for the discovery of orthologous mammalian long-term memory components.
Collapse
Affiliation(s)
- Vanisha Lakhina
- Department of Molecular Biology & LSI Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Rachel N Arey
- Department of Molecular Biology & LSI Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Rachel Kaletsky
- Department of Molecular Biology & LSI Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Amanda Kauffman
- Department of Molecular Biology & LSI Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Geneva Stein
- Department of Molecular Biology & LSI Genomics, Princeton University, Princeton, NJ 08544, USA
| | - William Keyes
- Department of Molecular Biology & LSI Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Daniel Xu
- Department of Molecular Biology & LSI Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Coleen T Murphy
- Department of Molecular Biology & LSI Genomics, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
35
|
Hamakawa M, Uozumi T, Ueda N, Iino Y, Hirotsu T. A role for Ras in inhibiting circular foraging behavior as revealed by a new method for time and cell-specific RNAi. BMC Biol 2015; 13:6. [PMID: 25603799 PMCID: PMC4321700 DOI: 10.1186/s12915-015-0114-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/09/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The nematode worm Caenorhabditis elegans, in which loss-of-function mutants and RNA interference (RNAi) models are available, is a model organism useful for analyzing effects of genes on various life phenomena, including behavior. In particular, RNAi is a powerful tool that enables time- or cell-specific knockdown via heat shock-inducible RNAi or cell-specific RNAi. However, conventional RNAi is insufficient for investigating pleiotropic genes with various sites of action and life stage-dependent functions. RESULTS Here, we investigated the Ras gene for its role in exploratory behavior in C. elegans. We found that, under poor environmental conditions, mutations in the Ras-MAPK signaling pathway lead to circular locomotion instead of normal exploratory foraging. Spontaneous foraging is regulated by a neural circuit composed of three classes of neurons: IL1, OLQ, and RMD, and we found that Ras functions in this neural circuit to modulate the direction of locomotion. We further observed that Ras plays an essential role in the regulation of GLR-1 glutamate receptor localization in RMD neurons. To investigate the temporal- and cell-specific profiles of the functions of Ras, we developed a new RNAi method that enables simultaneous time- and cell-specific knockdown. In this method, one RNA strand is expressed by a cell-specific promoter and the other by a heat shock promoter, resulting in only expression of double-stranded RNA in the target cell when heat shock is induced. This technique revealed that control of GLR-1 localization in RMD neurons requires Ras at the adult stage. Further, we demonstrated the application of this method to other genes. CONCLUSIONS We have established a new RNAi method that performs simultaneous time- and cell-specific knockdown and have applied this to reveal temporal profiles of the Ras-MAPK pathway in the control of exploratory behavior under poor environmental conditions.
Collapse
Affiliation(s)
- Masayuki Hamakawa
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 812-8581, Japan.
| | - Takayuki Uozumi
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 812-8581, Japan.
| | - Naoko Ueda
- Department of Biology, Faculty of Sciences, Kyushu University, Fukuoka, 812-8581, Japan.
| | - Yuichi Iino
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Tokyo, 113-0033, Japan.
| | - Takaaki Hirotsu
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 812-8581, Japan. .,Department of Biology, Faculty of Sciences, Kyushu University, Fukuoka, 812-8581, Japan. .,Department of Biology, Graduate School of Sciences, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka, 812-8581, Japan. .,Division of Applied Medical Sensing, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, 819-0395, Japan.
| |
Collapse
|
36
|
Bernardinelli Y, Nikonenko I, Muller D. Structural plasticity: mechanisms and contribution to developmental psychiatric disorders. Front Neuroanat 2014; 8:123. [PMID: 25404897 PMCID: PMC4217507 DOI: 10.3389/fnana.2014.00123] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/14/2014] [Indexed: 01/31/2023] Open
Abstract
Synaptic plasticity mechanisms are usually discussed in terms of changes in synaptic strength. The capacity of excitatory synapses to rapidly modify the membrane expression of glutamate receptors in an activity-dependent manner plays a critical role in learning and memory processes by re-distributing activity within neuronal networks. Recent work has however also shown that functional plasticity properties are associated with a rewiring of synaptic connections and a selective stabilization of activated synapses. These structural aspects of plasticity have the potential to continuously modify the organization of synaptic networks and thereby introduce specificity in the wiring diagram of cortical circuits. Recent work has started to unravel some of the molecular mechanisms that underlie these properties of structural plasticity, highlighting an important role of signaling pathways that are also major candidates for contributing to developmental psychiatric disorders. We review here some of these recent advances and discuss the hypothesis that alterations of structural plasticity could represent a common mechanism contributing to the cognitive and functional defects observed in diseases such as intellectual disability, autism spectrum disorders and schizophrenia.
Collapse
Affiliation(s)
- Yann Bernardinelli
- Department of Basic Neurosciences, University of Geneva Medical School Geneva, Switzerland
| | - Irina Nikonenko
- Department of Basic Neurosciences, University of Geneva Medical School Geneva, Switzerland
| | - Dominique Muller
- Department of Basic Neurosciences, University of Geneva Medical School Geneva, Switzerland
| |
Collapse
|
37
|
Stein GM, Murphy CT. C. elegans positive olfactory associative memory is a molecularly conserved behavioral paradigm. Neurobiol Learn Mem 2014; 115:86-94. [PMID: 25108196 DOI: 10.1016/j.nlm.2014.07.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 07/17/2014] [Accepted: 07/30/2014] [Indexed: 11/17/2022]
Abstract
While it is thought that short-term memory arises from changes in protein dynamics that increase the strength of synaptic signaling, many of the underlying fundamental molecular mechanisms remain unknown.Our lab developed a Caenorhabditis elegans assay of positive olfactory short-term associative memory (STAM), in which worms learn to associate food with an odor and can remember this association for over 1h. Here we use this massed olfactory associative assay to identify regulators of C. elegans short-term and intermediate-term associative memory (ITAM) processes. We show that there are unique molecular characteristics for different temporal phases of STAM, which include: learning, which is tested immediately after training, short-term memory, tested 30min after training, intermediate-term memory, tested 1h after training, and forgetting, tested 2h after training. We find that, as in higher organisms, C. elegans STAM requires calcium and cAMP signaling, and ITAM requires protein translation. Additionally, we found that STAM and ITAM are distinct from olfactory adaptation, an associative paradigm in which worms learn to disregard an inherently attractive odor after starvation in the presence of that odor. Adaptation mutants show variable responses to short-term associative memory training. Our data distinguish between shorter forms of a positive associative memory in C. elegans that require canonical memory pathways. Study of STAM and ITAM in C. elegans could lead to a more general understanding of the distinctions between these important processes and also to the discovery of novel conserved memory regulators.
Collapse
Affiliation(s)
- Geneva M Stein
- Lewis-Sigler Institute for Integrative Genomics, Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Coleen T Murphy
- Lewis-Sigler Institute for Integrative Genomics, Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
38
|
Sugi T, Ohtani Y. Simplified method for cell-specific gene expression analysis in Caenorhabditis elegans. Biochem Biophys Res Commun 2014; 450:330-4. [PMID: 24942876 DOI: 10.1016/j.bbrc.2014.05.124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 05/25/2014] [Indexed: 10/25/2022]
Abstract
In the neural circuit functional identities of individual neurons are mainly specified by their differential gene expression patterns. Unveiling functional roles of each neuron requires cell-specific interrogation of neural circuitry in the context of gene expressions. The mRNA tagging strategy in Caenorhabditis elegans is a powerful technique, in which cell-specific transcripts can be isolated by co-immunoprecipitating the complexes of mRNAs and epitope-tagged poly(A) binding protein (3× FLAG-PAB-1), expressed in target neurons. However, the conventional protocol requires laborious and time-consuming procedures; chromosomal integration of gene encoding 3× FLAG-PAB-1 and bleaching of obtained integrant animals for the isolation of huge amounts of synchronized animals. In this paper, we have presented a simplified methodology for cell-specific mRNA tagging analysis in C. elegans. We show that mRNA tagging was achieved using transgenic animals expressing 3× FLAG-PAB-1 as an extrachromosomal array under the control of the flp-18 promoter, without the chromosomal integration procedure. Furthermore, we successfully isolated cell-specific mRNAs from adult transgenic animals synchronously grown from eggs laid by gravid adults during a time window of 3h. This simplification facilitates the implementation of cell-specific gene expression analysis of C. elegans, which contributes to the understanding of neural circuitry at a cell-specific resolution.
Collapse
Affiliation(s)
- Takuma Sugi
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyo-ku, Kyoto 606-8501, Japan; PRESTO, Japanese Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Japan.
| | - Yasuko Ohtani
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
39
|
Lamprecht R. The actin cytoskeleton in memory formation. Prog Neurobiol 2014; 117:1-19. [DOI: 10.1016/j.pneurobio.2014.02.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 02/02/2014] [Accepted: 02/03/2014] [Indexed: 01/21/2023]
|
40
|
Hadziselimovic N, Vukojevic V, Peter F, Milnik A, Fastenrath M, Fenyves B, Hieber P, Demougin P, Vogler C, de Quervain DF, Papassotiropoulos A, Stetak A. Forgetting Is Regulated via Musashi-Mediated Translational Control of the Arp2/3 Complex. Cell 2014; 156:1153-1166. [DOI: 10.1016/j.cell.2014.01.054] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 11/27/2013] [Accepted: 01/17/2014] [Indexed: 01/21/2023]
|
41
|
Lau HL, Timbers TA, Mahmoud R, Rankin CH. Genetic dissection of memory for associative and non-associative learning inCaenorhabditis elegans. GENES BRAIN AND BEHAVIOR 2012; 12:210-23. [DOI: 10.1111/j.1601-183x.2012.00863.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 08/30/2012] [Accepted: 09/22/2012] [Indexed: 01/20/2023]
|