1
|
Lützkendorf J, Matkovic-Rachid T, Liu S, Götz T, Gao L, Turrel O, Maglione M, Grieger M, Putignano S, Ramesh N, Ghelani T, Neumann A, Gimber N, Schmoranzer J, Stawrakakis A, Brence B, Baum D, Ludwig K, Heine M, Mielke T, Liu F, Walter AM, Wahl MC, Sigrist SJ. Blobby is a synaptic active zone assembly protein required for memory in Drosophila. Nat Commun 2025; 16:271. [PMID: 39747038 PMCID: PMC11696761 DOI: 10.1038/s41467-024-55382-9] [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: 03/27/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
At presynaptic active zones (AZs), scaffold proteins are critical for coordinating synaptic vesicle release and forming essential nanoarchitectures. However, regulatory principles steering AZ scaffold assembly, function, and plasticity remain insufficiently understood. We here identify an additional Drosophila AZ protein, "Blobby", essential for proper AZ nano-organization. Blobby biochemically associates with the ELKS family AZ scaffold protein Bruchpilot (BRP) and integrates into newly forming AZs. Loss of Blobby results in fewer AZs forming, ectopic AZ scaffold protein accumulations ("blobs") and disrupts nanoscale architecture of the BRP-AZ scaffold. Functionally, blobby mutants show diminished evoked synaptic currents due to reduced synaptic vesicle release probability and fewer functional release sites. Blobby is also present in adult brain synapses, and post-developmental knockdown of Blobby in the mushroom body impairs olfactory aversive memory consolidation. Thus, our analysis identifies an additional layer of AZ regulation critical for developmental AZ assembly but also for AZ-mediated plasticity controlling behavior.
Collapse
Affiliation(s)
- J Lützkendorf
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - T Matkovic-Rachid
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - S Liu
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry, Berlin, Germany
| | - T Götz
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - L Gao
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - O Turrel
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - M Maglione
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, SupraFAB, Berlin, Germany
| | - M Grieger
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - S Putignano
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - N Ramesh
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - T Ghelani
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
| | - A Neumann
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - N Gimber
- Charité- Universitätsmedizin, Advanced Medical Bioimaging Core Facility, Berlin, Germany
| | - J Schmoranzer
- Charité- Universitätsmedizin, Advanced Medical Bioimaging Core Facility, Berlin, Germany
| | - A Stawrakakis
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - B Brence
- Zuse Institute Berlin, Department of Visual and Data-Centric Computing, Berlin, Germany
| | - D Baum
- Zuse Institute Berlin, Department of Visual and Data-Centric Computing, Berlin, Germany
| | - Kai Ludwig
- Freie Universität Berlin, Institut für Chemie and Biochemie, Forschungszentrum für Elektronenmikroskopie, Berlin, Germany
| | - M Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - T Mielke
- Max Planck Institute for Molecular Genetics, Berlin, Microscopy and Cryo-Electron Microscopy Service Group, Berlin, Germany
| | - F Liu
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
| | - A M Walter
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
- University of Copenhagen, Department of Neuroscience, Copenhagen, Denmark
| | - M C Wahl
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry, Berlin, Germany
| | - S J Sigrist
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany.
- Charité Universitätsmedizin, NeuroCure Cluster of Excellence, Charitéplatz, Berlin, Germany.
| |
Collapse
|
2
|
Xu P, Xiao Y, Xiao Z, Li J. Exploitation and Application of a New Genetic Sex Marker Based on Intron Insertion Variation of erc2 Gene in Oplegnathus punctatus. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 26:1155-1164. [PMID: 39212851 DOI: 10.1007/s10126-024-10363-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Spotted knifejaw (Oplegnathus punctatus), one of the most valuable mariculture species, grows with significant sexual dimorphism, with males growing significantly faster than females. O. punctatus not only has excellent growth characteristics and high food value, but also shows high economic value in aquaculture, which has become a hotspot in the field of aquaculture. The current insufficiency of sex marker identification in O. punctatus restricts the process of its unisexual breeding. Rapid identification of sex will help to study the mechanisms of sex determination and accelerate the development of sex-controlled breeding. With the completion of the sequencing of the male and female genomes of O. punctatus, the efficient and precise development of genetic sex markers has been made possible. In this study, we used genome-wide information combined with molecular biology techniques from marker sequences to further establish a rapid method for DNA insertion variant detection in the intron of O. punctatus erc2 gene, which can be used to rapidly, accurately, and efficiently identify whether DNA insertion occurs in the intron of O. punctatus erc2 gene to be detected, and to identify the sex of O. punctatus to be detected. It could also be distinguished by agarose gel electrophoresis, which would shorten the time for accurate identification and improves the detection efficiency. Homozygous comparison of male and female individuals showed that the length of the DNA fragment of the erc2 gene was 239 bp on chromosome X1 and 1173 bp on chromosome Y. It can therefore be inferred that a 934 bp insertion fragment exists on the Y chromosome. The PCR amplification results showed that two DNA fragments of 1173 bp and 239 bp could be amplified in male O. punctatus, and the 1173 bp fragment was a marker fragment specific to the variant intron erc2 gene, while only a single DNA fragment of 239 bp was amplified in female O. punctatus. It has important significance and application value in the study of neurotransmitter transmission and environmental adaptability of female and male fish based on erc2 gene, as well as the identification of male and female sex, the preparation of high male fry, and family breeding.
Collapse
Affiliation(s)
- Pingrui Xu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS). Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Yongshuang Xiao
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS). Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
| | - Zhizhong Xiao
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS). Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Weihai Hao Huigan Marine Biotechnology Co, Weihai, China
| | - Jun Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS). Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
| |
Collapse
|
3
|
Ishchenko Y, Jeng AT, Feng S, Nottoli T, Manriquez-Rodriguez C, Nguyen KK, Carrizales MG, Vitarelli MJ, Corcoran EE, Greer CA, Myers SA, Koleske AJ. Heterozygosity for neurodevelopmental disorder-associated TRIO variants yields distinct deficits in behavior, neuronal development, and synaptic transmission in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574442. [PMID: 39131289 PMCID: PMC11312463 DOI: 10.1101/2024.01.05.574442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Genetic variants in TRIO are associated with neurodevelopmental disorders (NDDs) including schizophrenia (SCZ), autism spectrum disorder (ASD) and intellectual disability. TRIO uses its two guanine nucleotide exchange factor (GEF) domains to activate GTPases (GEF1: Rac1 and RhoG; GEF2: RhoA) that control neuronal development and connectivity. It remains unclear how discrete TRIO variants differentially impact these neurodevelopmental events. Here, we investigate how heterozygosity for NDD-associated Trio variants - +/K1431M (ASD), +/K1918X (SCZ), and +/M2145T (bipolar disorder, BPD) - impact mouse behavior, brain development, and synapse structure and function. Heterozygosity for different Trio variants impacts motor, social, and cognitive behaviors in distinct ways that align with clinical phenotypes in humans. Trio variants differentially impact head and brain size with corresponding changes in dendritic arbors of motor cortex layer 5 pyramidal neurons (M1 L5 PNs). Although neuronal structure was only modestly altered in the Trio variant heterozygotes, we observe significant changes in synaptic function and plasticity. We also identified distinct changes in glutamate synaptic release in +/K1431M and +/M2145T cortico-cortical synapses. The TRIO K1431M GEF1 domain has impaired ability to promote GTP exchange on Rac1, but +/K1431M mice exhibit increased Rac1 activity, associated with increased levels of the Rac1 GEF Tiam1. Acute Rac1 inhibition with NSC23766 rescued glutamate release deficits in +/K1431M variant cortex. Our work reveals that discrete NDD-associated Trio variants yield overlapping but distinct phenotypes in mice, demonstrates an essential role for Trio in presynaptic glutamate release, and underscores the importance of studying the impact of variant heterozygosity in vivo.
Collapse
Affiliation(s)
- Yevheniia Ishchenko
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Amanda T Jeng
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
| | - Shufang Feng
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Gerontology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Timothy Nottoli
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Khanh K Nguyen
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Melissa G Carrizales
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Matthew J Vitarelli
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Ellen E Corcoran
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Charles A Greer
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Samuel A Myers
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
4
|
Medeiros AT, Gratz SJ, Delgado A, Ritt JT, O'Connor-Giles KM. Ca 2+ channel and active zone protein abundance intersects with input-specific synapse organization to shape functional synaptic diversity. eLife 2024; 12:RP88412. [PMID: 39291956 PMCID: PMC11410372 DOI: 10.7554/elife.88412] [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] [Indexed: 09/19/2024] Open
Abstract
Synaptic heterogeneity is a hallmark of nervous systems that enables complex and adaptable communication in neural circuits. To understand circuit function, it is thus critical to determine the factors that contribute to the functional diversity of synapses. We investigated the contributions of voltage-gated calcium channel (VGCC) abundance, spatial organization, and subunit composition to synapse diversity among and between synapses formed by two closely related Drosophila glutamatergic motor neurons with distinct neurotransmitter release probabilities (Pr). Surprisingly, VGCC levels are highly predictive of heterogeneous Pr among individual synapses of either low- or high-Pr inputs, but not between inputs. We find that the same number of VGCCs are more densely organized at high-Pr synapses, consistent with tighter VGCC-synaptic vesicle coupling. We generated endogenously tagged lines to investigate VGCC subunits in vivo and found that the α2δ-3 subunit Straightjacket along with the CAST/ELKS active zone (AZ) protein Bruchpilot, both key regulators of VGCCs, are less abundant at high-Pr inputs, yet positively correlate with Pr among synapses formed by either input. Consistently, both Straightjacket and Bruchpilot levels are dynamically increased across AZs of both inputs when neurotransmitter release is potentiated to maintain stable communication following glutamate receptor inhibition. Together, these findings suggest a model in which VGCC and AZ protein abundance intersects with input-specific spatial and molecular organization to shape the functional diversity of synapses.
Collapse
Affiliation(s)
- Audrey T Medeiros
- Neuroscience Graduate Training Program, Brown University, Providence, United States
| | - Scott J Gratz
- Department of Neuroscience, Brown University, Providence, United States
| | - Ambar Delgado
- Department of Neuroscience, Brown University, Providence, United States
| | - Jason T Ritt
- Department of Neuroscience, Brown University, Providence, United States
- Carney Institute for Brain Science, Brown University, Providence, United States
| | - Kate M O'Connor-Giles
- Neuroscience Graduate Training Program, Brown University, Providence, United States
- Department of Neuroscience, Brown University, Providence, United States
- Carney Institute for Brain Science, Brown University, Providence, United States
| |
Collapse
|
5
|
Emperador-Melero J, Andersen JW, Metzbower SR, Levy AD, Dharmasri PA, de Nola G, Blanpied TA, Kaeser PS. Distinct active zone protein machineries mediate Ca 2+ channel clustering and vesicle priming at hippocampal synapses. Nat Neurosci 2024; 27:1680-1694. [PMID: 39160372 PMCID: PMC11682530 DOI: 10.1038/s41593-024-01720-5] [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: 10/27/2023] [Accepted: 06/28/2024] [Indexed: 08/21/2024]
Abstract
Action potentials trigger neurotransmitter release at the presynaptic active zone with spatiotemporal precision. This is supported by protein machinery that mediates synaptic vesicle priming and clustering of CaV2 Ca2+ channels nearby. One model posits that scaffolding proteins directly tether vesicles to CaV2s; however, here we find that at mouse hippocampal synapses, CaV2 clustering and vesicle priming are executed by separate machineries. CaV2 nanoclusters are positioned at variable distances from those of the priming protein Munc13. The active zone organizer RIM anchors both proteins but distinct interaction motifs independently execute these functions. In transfected cells, Liprin-α and RIM form co-assemblies that are separate from CaV2-organizing complexes. At synapses, Liprin-α1-Liprin-α4 knockout impairs vesicle priming but not CaV2 clustering. The cell adhesion protein PTPσ recruits Liprin-α, RIM and Munc13 into priming complexes without co-clustering CaV2s. We conclude that active zones consist of distinct machineries to organize CaV2s and prime vesicles, and Liprin-α and PTPσ specifically support priming site assembly.
Collapse
Affiliation(s)
| | | | - Sarah R Metzbower
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aaron D Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Poorna A Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Giovanni de Nola
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Medeiros AT, Gratz S, Delgado A, Ritt J, O’Connor-Giles KM. Ca 2+ channel and active zone protein abundance intersects with input-specific synapse organization to shape functional synaptic diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.02.535290. [PMID: 37034654 PMCID: PMC10081318 DOI: 10.1101/2023.04.02.535290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Synaptic heterogeneity is a hallmark of nervous systems that enables complex and adaptable communication in neural circuits. To understand circuit function, it is thus critical to determine the factors that contribute to the functional diversity of synapses. We investigated the contributions of voltage-gated calcium channel (VGCC) abundance, spatial organization, and subunit composition to synapse diversity among and between synapses formed by two closely related Drosophila glutamatergic motor neurons with distinct neurotransmitter release probabilities (Pr). Surprisingly, VGCC levels are highly predictive of heterogeneous Pr among individual synapses of either low- or high-Pr inputs, but not between inputs. We find that the same number of VGCCs are more densely organized at high-Pr synapses, consistent with tighter VGCC-synaptic vesicle coupling. We generated endogenously tagged lines to investigate VGCC subunits in vivo and found that the α2δ-3 subunit Straightjacket along with the CAST/ELKS active zone (AZ) protein Bruchpilot, both key regulators of VGCCs, are less abundant at high-Pr inputs, yet positively correlate with Pr among synapses formed by either input. Consistently, both Straightjacket and Bruchpilot levels are dynamically increased across AZs of both inputs when neurotransmitter release is potentiated to maintain stable communication following glutamate receptor inhibition. Together, these findings suggest a model in which VGCC and AZ protein abundance intersects with input-specific spatial and molecular organization to shape the functional diversity of synapses.
Collapse
Affiliation(s)
- A. T. Medeiros
- Neuroscience Graduate Training Program, Brown University, Providence, RI
| | - S.J. Gratz
- Department of Neuroscience, Brown University, Providence, RI
| | - A. Delgado
- Department of Neuroscience, Brown University, Providence, RI
| | - J.T. Ritt
- Department of Neuroscience, Brown University, Providence, RI
- Carney Institute for Brain Science, Brown University, Providence, RI
| | - Kate M. O’Connor-Giles
- Neuroscience Graduate Training Program, Brown University, Providence, RI
- Department of Neuroscience, Brown University, Providence, RI
- Carney Institute for Brain Science, Brown University, Providence, RI
| |
Collapse
|
7
|
Li J, Veeraraghavan P, Young SM. Ca V 2.1 α 1 subunit motifs that control presynaptic Ca V 2.1 subtype abundance are distinct from Ca V 2.1 preference. J Physiol 2024; 602:485-506. [PMID: 38155373 PMCID: PMC10872416 DOI: 10.1113/jp284957] [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: 04/28/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023] Open
Abstract
Presynaptic voltage-gated Ca2+ channel (CaV ) subtype abundance at mammalian synapses regulates synaptic transmission in health and disease. In the mammalian central nervous system (CNS), most presynaptic terminals are CaV 2.1 dominant with a developmental reduction in CaV 2.2 and CaV 2.3 levels, and CaV 2 subtype levels are altered in various diseases. However, the molecular mechanisms controlling presynaptic CaV 2 subtype levels are largely unsolved. Because the CaV 2 α1 subunit cytoplasmic regions contain varying levels of sequence conservation, these regions are proposed to control presynaptic CaV 2 subtype preference and abundance. To investigate the potential role of these regions, we expressed chimeric CaV 2.1 α1 subunits containing swapped motifs with the CaV 2.2 and CaV 2.3 α1 subunit on a CaV 2.1/CaV 2.2 null background at the calyx of Held presynaptic terminals. We found that expression of CaV 2.1 α1 subunit chimeras containing the CaV 2.3 loop II-III region or cytoplasmic C-terminus (CT) resulted in a large reduction of presynaptic Ca2+ currents compared to the CaV 2.1 α1 subunit. However, the Ca2+ current sensitivity to the CaV 2.1 blocker agatoxin-IVA was the same between the chimeras and the CaV 2.1 α1 subunit. Additionally, we found no reduction in presynaptic Ca2+ currents with CaV 2.1/2.2 cytoplasmic CT chimeras. We conclude that the motifs in the CaV 2.1 loop II-III and CT do not individually regulate CaV 2.1 preference, although these motifs control CaV 2.1 levels and the CaV 2.3 CT contains motifs that negatively regulate presynaptic CaV 2.3 levels. We propose that the motifs controlling presynaptic CaV 2.1 preference are distinct from those regulating CaV 2.1 levels and may act synergistically to impact pathways regulating CaV 2.1 preference and abundance. KEY POINTS: Presynaptic CaV 2 subtype abundance regulates neuronal circuit properties, although the mechanisms regulating presynaptic CaV 2 subtype abundance and preference remain enigmatic. The CaV α1 subunit determines subtype and contains multiple motifs implicated in regulating presynaptic subtype abundance and preference. The CaV 2.1 α1 subunit domain II-III loop and cytoplasmic C-terminus are positive regulators of presynaptic CaV 2.1 abundance but do not regulate preference. The CaV 2.3 α1 subunit cytoplasmic C-terminus negatively regulates presynaptic CaV 2 subtype abundance but not preference, whereas the CaV 2.2 α1 subunit cytoplasmic C-terminus is not a key regulator of presynaptic CaV 2 subtype abundance or preference. The CaV 2 α1 subunit motifs determining the presynaptic CaV 2 preference are distinct from abundance.
Collapse
Affiliation(s)
- Jianing Li
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Cell Developmental Biology Graduate Program, University of Iowa, Iowa City, IA 52242, USA
| | | | - Samuel M. Young
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Department of Otolaryngology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
8
|
Emperador-Melero J, Andersen JW, Metzbower SR, Levy AD, Dharmasri PA, de Nola G, Blanpied TA, Kaeser PS. Molecular definition of distinct active zone protein machineries for Ca 2+ channel clustering and synaptic vesicle priming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.27.564439. [PMID: 37961089 PMCID: PMC10634917 DOI: 10.1101/2023.10.27.564439] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Action potentials trigger neurotransmitter release with minimal delay. Active zones mediate this temporal precision by co-organizing primed vesicles with CaV2 Ca2+ channels. The presumed model is that scaffolding proteins directly tether primed vesicles to CaV2s. We find that CaV2 clustering and vesicle priming are executed by separate machineries. At hippocampal synapses, CaV2 nanoclusters are positioned at variable distances from those of the priming protein Munc13. The active zone organizer RIM anchors both proteins, but distinct interaction motifs independently execute these functions. In heterologous cells, Liprin-α and RIM from co-assemblies that are separate from CaV2-organizing complexes upon co-transfection. At synapses, Liprin-α1-4 knockout impairs vesicle priming, but not CaV2 clustering. The cell adhesion protein PTPσ recruits Liprin-α, RIM and Munc13 into priming complexes without co-clustering of CaV2s. We conclude that active zones consist of distinct complexes to organize CaV2s and vesicle priming, and Liprin-α and PTPσ specifically support priming site assembly.
Collapse
Affiliation(s)
| | | | - Sarah R. Metzbower
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | - Aaron D. Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | - Poorna A. Dharmasri
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | | | - Thomas A. Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | | |
Collapse
|
9
|
Li J, Veeraraghavan P, Young SM. CaV2.1 α1 subunit motifs that control presynaptic CaV2.1 subtype abundance are distinct from CaV2.1 preference. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.28.538778. [PMID: 37162941 PMCID: PMC10168310 DOI: 10.1101/2023.04.28.538778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Presynaptic voltage-gated Ca2+ channels (CaV) subtype abundance at mammalian synapses regulates synaptic transmission in health and disease. In the mammalian central nervous system, most presynaptic terminals are CaV2.1 dominant with a developmental reduction in CaV2.2 and CaV2.3 levels, and CaV2 subtype levels are altered in various diseases. However, the molecular mechanisms controlling presynaptic CaV2 subtype levels are largely unsolved. Since the CaV2 α1 subunit cytoplasmic regions contain varying levels of sequence conservation, these regions are proposed to control presynaptic CaV2 subtype preference and abundance. To investigate the potential role of these regions, we expressed chimeric CaV2.1 α1subunits containing swapped motifs with the CaV2.2 and CaV2.3 α1 subunit on a CaV2.1/CaV2.2 null background at the calyx of Held presynaptic terminal. We found that expression of CaV2.1 α1 subunit chimeras containing the CaV2.3 loop II-III region or cytoplasmic C-terminus (CT) resulted in a large reduction of presynaptic Ca2+ currents compared to the CaV2.1 α1 subunit. However, the Ca2+ current sensitivity to the CaV2.1 blocker Agatoxin-IVA, was the same between the chimeras and the CaV2.1 α1 subunit. Additionally, we found no reduction in presynaptic Ca2+ currents with CaV2.1/2.2 cytoplasmic CT chimeras. We conclude that the motifs in the CaV2.1 loop II-III and CT do not individually regulate CaV2.1 preference, but these motifs control CaV2.1 levels and the CaV2.3 CT contains motifs that negatively regulate presynaptic CaV2.3 levels. We propose that the motifs controlling presynaptic CaV2.1 preference are distinct from those regulating CaV2.1 levels and may act synergistically to impact pathways regulating CaV2.1 preference and abundance.
Collapse
|
10
|
Keine C, Al-Yaari M, Radulovic T, Young SM. Stereotactic Delivery of Helper-dependent Adenoviral Viral Vectors at Distinct Developmental Time Points to Perform Age-dependent Molecular Manipulations of the Mouse Calyx of Held. Bio Protoc 2023; 13:e4793. [PMID: 37638292 PMCID: PMC10450731 DOI: 10.21769/bioprotoc.4793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/23/2023] [Accepted: 06/24/2023] [Indexed: 08/29/2023] Open
Abstract
Synapses are specialized structures that enable neuronal communication, which is essential for brain function and development. Alterations in synaptic proteins have been linked to various neurological and neuropsychiatric disorders. Therefore, manipulating synaptic proteins in vivo can provide insight into the molecular mechanisms underlying these disorders and aid in developing new therapeutic strategies. Previous methods such as constitutive knock-out animals are limited by developmental compensation and off-target effects. The current approach outlines procedures for age-dependent molecular manipulations in mice using helper-dependent adenovirus viral vectors (HdAd) at distinct developmental time points. Using stereotactic injection of HdAds in both newborn and juvenile mice, we demonstrate the versatility of this method to express Cre recombinase in globular bushy cells of juvenile Rac1fl/fl mice to ablate presynaptic Rac1 and study its role in synaptic transmission. Separately, we overexpress CaV2 α1 subunits at two distinct developmental time points to elucidate the mechanisms that determine presynaptic CaV2 channel abundance and preference. This method presents a reliable, cost-effective, and minimally invasive approach for controlling gene expression in specific regions of the mouse brain and will be a powerful tool to decipher brain function in health and disease. Key features Virus-mediated genetic perturbation in neonatal and young adult mice. Stereotaxic injection allows targeting of brain structures at different developmental stages to study the impact of genetic perturbation throughout the development.
Collapse
Affiliation(s)
- Christian Keine
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Oldenburg, Germany
| | - Mohammed Al-Yaari
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
| | - Tamara Radulovic
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Oldenburg, Germany
| | - Samuel M. Young
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Department of Otolaryngology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
11
|
Kim JK, Jha NN, Awano T, Caine C, Gollapalli K, Welby E, Kim SS, Fuentes-Moliz A, Wang X, Feng Z, Sera F, Takeda T, Homma S, Ko CP, Tabares L, Ebert AD, Rich MM, Monani UR. A spinal muscular atrophy modifier implicates the SMN protein in SNARE complex assembly at neuromuscular synapses. Neuron 2023; 111:1423-1439.e4. [PMID: 36863345 PMCID: PMC10164130 DOI: 10.1016/j.neuron.2023.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/11/2022] [Accepted: 02/02/2023] [Indexed: 03/04/2023]
Abstract
Reduced survival motor neuron (SMN) protein triggers the motor neuron disease, spinal muscular atrophy (SMA). Restoring SMN prevents disease, but it is not known how neuromuscular function is preserved. We used model mice to map and identify an Hspa8G470R synaptic chaperone variant, which suppressed SMA. Expression of the variant in the severely affected mutant mice increased lifespan >10-fold, improved motor performance, and mitigated neuromuscular pathology. Mechanistically, Hspa8G470R altered SMN2 splicing and simultaneously stimulated formation of a tripartite chaperone complex, critical for synaptic homeostasis, by augmenting its interaction with other complex members. Concomitantly, synaptic vesicular SNARE complex formation, which relies on chaperone activity for sustained neuromuscular synaptic transmission, was found perturbed in SMA mice and patient-derived motor neurons and was restored in modified mutants. Identification of the Hspa8G470R SMA modifier implicates SMN in SNARE complex assembly and casts new light on how deficiency of the ubiquitous protein causes motor neuron disease.
Collapse
Affiliation(s)
- Jeong-Ki Kim
- Department of Neurology, New York, NY, USA; Center for Motor Neuron Biology & Disease, New York, NY, USA
| | - Narendra N Jha
- Department of Neurology, New York, NY, USA; Center for Motor Neuron Biology & Disease, New York, NY, USA
| | - Tomoyuki Awano
- Department of Neurology, New York, NY, USA; Center for Motor Neuron Biology & Disease, New York, NY, USA
| | - Charlotte Caine
- Department of Neurology, New York, NY, USA; Center for Motor Neuron Biology & Disease, New York, NY, USA
| | - Kishore Gollapalli
- Department of Neurology, New York, NY, USA; Center for Motor Neuron Biology & Disease, New York, NY, USA
| | - Emily Welby
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Seung-Soo Kim
- Department of Obstetrics and Gynecology, New York, NY, USA
| | - Andrea Fuentes-Moliz
- Department of Medical Physiology and Biophysics, University of Seville School of Medicine, 41009, Seville, Spain
| | - Xueyong Wang
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435, USA
| | - Zhihua Feng
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Fusako Sera
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Taishi Takeda
- Department of Neurology, New York, NY, USA; Center for Motor Neuron Biology & Disease, New York, NY, USA
| | - Shunichi Homma
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Chien-Ping Ko
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Lucia Tabares
- Department of Medical Physiology and Biophysics, University of Seville School of Medicine, 41009, Seville, Spain
| | - Allison D Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mark M Rich
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435, USA
| | - Umrao R Monani
- Department of Neurology, New York, NY, USA; Department of Pathology & Cell Biology, New York, NY, USA; Center for Motor Neuron Biology & Disease, New York, NY, USA; Colleen Giblin Research Laboratory, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
12
|
Hagiwara A, Mizutani A, Kawamura S, Abe M, Hida Y, Sakimura K, Ohtsuka T. Critical Role of the Presynaptic Protein CAST in Maintaining the Photoreceptor Ribbon Synapse Triad. Int J Mol Sci 2023; 24:ijms24087251. [PMID: 37108413 PMCID: PMC10138387 DOI: 10.3390/ijms24087251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The cytomatrix at the active zone-associated structural protein (CAST) and its homologue, named ELKS, being rich in glutamate (E), leucine (L), lysine (K), and serine (S), belong to a family of proteins that organize presynaptic active zones at nerve terminals. These proteins interact with other active zone proteins, including RIMs, Munc13s, Bassoon, and the β subunit of Ca2+ channels, and have various roles in neurotransmitter release. A previous study showed that depletion of CAST/ELKS in the retina causes morphological changes and functional impairment of this structure. In this study, we investigated the roles of CAST and ELKS in ectopic synapse localization. We found that the involvement of these proteins in ribbon synapse distribution is complex. Unexpectedly, CAST and ELKS, in photoreceptors or in horizontal cells, did not play a major role in ribbon synapse ectopic localization. However, depletion of CAST and ELKS in the mature retina resulted in degeneration of the photoreceptors. These findings suggest that CAST and ELKS play critical roles in maintaining neural signal transduction in the retina, but the regulation of photoreceptor triad synapse distribution is not solely dependent on their actions within photoreceptors and horizontal cells.
Collapse
Affiliation(s)
- Akari Hagiwara
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba 278-8510, Japan
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Ayako Mizutani
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba 278-8510, Japan
| | - Saki Kawamura
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba 278-8510, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Yamato Hida
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| |
Collapse
|
13
|
Ghelani T, Escher M, Thomas U, Esch K, Lützkendorf J, Depner H, Maglione M, Parutto P, Gratz S, Matkovic-Rachid T, Ryglewski S, Walter AM, Holcman D, O‘Connor Giles K, Heine M, Sigrist SJ. Interactive nanocluster compaction of the ELKS scaffold and Cacophony Ca 2+ channels drives sustained active zone potentiation. SCIENCE ADVANCES 2023; 9:eade7804. [PMID: 36800417 PMCID: PMC9937578 DOI: 10.1126/sciadv.ade7804] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/17/2023] [Indexed: 06/01/2023]
Abstract
At presynaptic active zones (AZs), conserved scaffold protein architectures control synaptic vesicle (SV) release by defining the nanoscale distribution and density of voltage-gated Ca2+ channels (VGCCs). While AZs can potentiate SV release in the minutes range, we lack an understanding of how AZ scaffold components and VGCCs engage into potentiation. We here establish dynamic, intravital single-molecule imaging of endogenously tagged proteins at Drosophila AZs undergoing presynaptic homeostatic potentiation. During potentiation, the numbers of α1 VGCC subunit Cacophony (Cac) increased per AZ, while their mobility decreased and nanoscale distribution compacted. These dynamic Cac changes depended on the interaction between Cac channel's intracellular carboxyl terminus and the membrane-close amino-terminal region of the ELKS-family protein Bruchpilot, whose distribution compacted drastically. The Cac-ELKS/Bruchpilot interaction was also needed for sustained AZ potentiation. Our single-molecule analysis illustrates how the AZ scaffold couples to VGCC nanoscale distribution and dynamics to establish a state of sustained potentiation.
Collapse
Affiliation(s)
- Tina Ghelani
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
- Molecular and Theoretical Neuroscience Leibniz-Forschungs Institut für Molekulare Pharmakologie (FMP) im CharitéCrossOver (CCO) Charité–University Medicine Berlin Charité Campus Mitte, Charité Platz, 110117 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
| | - Marc Escher
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Ulrich Thomas
- Department of Cellular Neurobiology, Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Klara Esch
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Janine Lützkendorf
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Harald Depner
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Marta Maglione
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
- Institute for Chemistry and Biochemistry, SupraFAB, Freie Universität Berlin, Altensteinstr. 23a, 14195 Berlin, Germany
| | - Pierre Parutto
- Group of Applied Mathematics and Computational Biology, IBENS, Ecole Normale Superieure, Paris, France
- Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Cambridge CB2 0AH, UK
- Churchill College, University of Cambridge, Cambridge CB3 0DS, UK
| | - Scott Gratz
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Tanja Matkovic-Rachid
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Stefanie Ryglewski
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alexander M. Walter
- Molecular and Theoretical Neuroscience Leibniz-Forschungs Institut für Molekulare Pharmakologie (FMP) im CharitéCrossOver (CCO) Charité–University Medicine Berlin Charité Campus Mitte, Charité Platz, 110117 Berlin, Germany
- Department of Neuroscience, University of Copenhagen, Copenhagen 2200, Denmark
| | - David Holcman
- Group of Applied Mathematics and Computational Biology, IBENS, Ecole Normale Superieure, Paris, France
- Churchill College, University of Cambridge, Cambridge CB3 0DS, UK
| | - Kate O‘Connor Giles
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Martin Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
- Research Group Molecular Physiology, Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Stephan J. Sigrist
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
14
|
Mizumoto K, Jin Y, Bessereau JL. Synaptogenesis: unmasking molecular mechanisms using Caenorhabditis elegans. Genetics 2023; 223:iyac176. [PMID: 36630525 PMCID: PMC9910414 DOI: 10.1093/genetics/iyac176] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/22/2022] [Indexed: 01/13/2023] Open
Abstract
The nematode Caenorhabditis elegans is a research model organism particularly suited to the mechanistic understanding of synapse genesis in the nervous system. Armed with powerful genetics, knowledge of complete connectomics, and modern genomics, studies using C. elegans have unveiled multiple key regulators in the formation of a functional synapse. Importantly, many signaling networks display remarkable conservation throughout animals, underscoring the contributions of C. elegans research to advance the understanding of our brain. In this chapter, we will review up-to-date information of the contribution of C. elegans to the understanding of chemical synapses, from structure to molecules and to synaptic remodeling.
Collapse
Affiliation(s)
- Kota Mizumoto
- Department of Zoology, University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Yishi Jin
- Department of Neurobiology, University of California San Diego, La Jolla, CA 92093, USA
| | - Jean-Louis Bessereau
- Univ Lyon, University Claude Bernard Lyon 1, CNRS UMR 5284, INSERM U 1314, Melis, 69008 Lyon, France
| |
Collapse
|
15
|
A novel technique for large-fragment knock-in animal production without ex vivo handling of zygotes. Sci Rep 2023; 13:2245. [PMID: 36755180 PMCID: PMC9908863 DOI: 10.1038/s41598-023-29468-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
CRISPR/Cas-based genome editing has dramatically improved genetic modification technology. In situ electroporation called genome editing via oviductal nucleic acid delivery (GONAD), which eliminates the need for ex vivo embryo handling, is technically the simplest method for gene transfer and can be performed in laboratories without developmental engineering expertise including micromanipulation techniques. However, the use of this method remains challenging in the case of large-fragment knock-in, such as gene expression cassettes. Adeno-associated viruses (AAV) act as donor DNA for homologous recombination in infected cells, including rodent embryos. In this study, we demonstrated simultaneous electroporation of AAV donors and CRISPR/Cas9 components into embryos to create knock-in animals, and successfully generated knock-in rats carrying a gene cassette with a length of 3.0 kb using a small number of animals and in situ electroporation. These findings indicate that this technique is an efficient high-throughput strategy for producing genetically modified rodents and may be applicable to other animal species.
Collapse
|
16
|
Cunningham KL, Littleton JT. Mechanisms controlling the trafficking, localization, and abundance of presynaptic Ca 2+ channels. Front Mol Neurosci 2023; 15:1116729. [PMID: 36710932 PMCID: PMC9880069 DOI: 10.3389/fnmol.2022.1116729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/26/2022] [Indexed: 01/14/2023] Open
Abstract
Voltage-gated Ca2+ channels (VGCCs) mediate Ca2+ influx to trigger neurotransmitter release at specialized presynaptic sites termed active zones (AZs). The abundance of VGCCs at AZs regulates neurotransmitter release probability (Pr ), a key presynaptic determinant of synaptic strength. Given this functional significance, defining the processes that cooperate to establish AZ VGCC abundance is critical for understanding how these mechanisms set synaptic strength and how they might be regulated to control presynaptic plasticity. VGCC abundance at AZs involves multiple steps, including channel biosynthesis (transcription, translation, and trafficking through the endomembrane system), forward axonal trafficking and delivery to synaptic terminals, incorporation and retention at presynaptic sites, and protein recycling. Here we discuss mechanisms that control VGCC abundance at synapses, highlighting findings from invertebrate and vertebrate models.
Collapse
Affiliation(s)
- Karen L. Cunningham
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | | |
Collapse
|
17
|
Colgren J, Burkhardt P. The premetazoan ancestry of the synaptic toolkit and appearance of first neurons. Essays Biochem 2022; 66:781-795. [PMID: 36205407 PMCID: PMC9750855 DOI: 10.1042/ebc20220042] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/31/2022] [Accepted: 09/13/2022] [Indexed: 12/13/2022]
Abstract
Neurons, especially when coupled with muscles, allow animals to interact with and navigate through their environment in ways unique to life on earth. Found in all major animal lineages except sponges and placozoans, nervous systems range widely in organization and complexity, with neurons possibly representing the most diverse cell-type. This diversity has led to much debate over the evolutionary origin of neurons as well as synapses, which allow for the directed transmission of information. The broad phylogenetic distribution of neurons and presence of many of the defining components outside of animals suggests an early origin of this cell type, potentially in the time between the first animal and the last common ancestor of extant animals. Here, we highlight the occurrence and function of key aspects of neurons outside of animals as well as recent findings from non-bilaterian animals in order to make predictions about when and how the first neuron(s) arose during animal evolution and their relationship to those found in extant lineages. With advancing technologies in single cell transcriptomics and proteomics as well as expanding functional techniques in non-bilaterian animals and the close relatives of animals, it is an exciting time to begin unraveling the complex evolutionary history of this fascinating animal cell type.
Collapse
Affiliation(s)
- Jeffrey Colgren
- Sars International Centre for Marine Molecular Biology, University of Bergen, Norway
| | - Pawel Burkhardt
- Sars International Centre for Marine Molecular Biology, University of Bergen, Norway
| |
Collapse
|
18
|
Midorikawa M. Developmental and activity-dependent modulation of coupling distance between release site and Ca2+ channel. Front Cell Neurosci 2022; 16:1037721. [DOI: 10.3389/fncel.2022.1037721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Synapses are junctions between a presynaptic neuron and a postsynaptic cell specialized for fast and precise information transfer. The presynaptic terminal secretes neurotransmitters via exocytosis of synaptic vesicles. Exocytosis is a tightly regulated reaction that occurs within a millisecond of the arrival of an action potential. One crucial parameter in determining the characteristics of the transmitter release kinetics is the coupling distance between the release site and the Ca2+ channel. Still, the technical limitations have hindered detailed analysis from addressing how the coupling distance is regulated depending on the development or activity of the synapse. However, recent technical advances in electrophysiology and imaging are unveiling their different configurations in different conditions. Here, I will summarize developmental- and activity-dependent changes in the coupling distances revealed by recent studies.
Collapse
|
19
|
Leung HW, Foo G, VanDongen A. Arc Regulates Transcription of Genes for Plasticity, Excitability and Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081946. [PMID: 36009494 PMCID: PMC9405677 DOI: 10.3390/biomedicines10081946] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
The immediate early gene Arc is a master regulator of synaptic function and a critical determinant of memory consolidation. Here, we show that Arc interacts with dynamic chromatin and closely associates with histone markers for active enhancers and transcription in cultured rat hippocampal neurons. Both these histone modifications, H3K27Ac and H3K9Ac, have recently been shown to be upregulated in late-onset Alzheimer’s disease (AD). When Arc induction by pharmacological network activation was prevented using a short hairpin RNA, the expression profile was altered for over 1900 genes, which included genes associated with synaptic function, neuronal plasticity, intrinsic excitability, and signalling pathways. Interestingly, about 100 Arc-dependent genes are associated with the pathophysiology of AD. When endogenous Arc expression was induced in HEK293T cells, the transcription of many neuronal genes was increased, suggesting that Arc can control expression in the absence of activated signalling pathways. Taken together, these data establish Arc as a master regulator of neuronal activity-dependent gene expression and suggest that it plays a significant role in the pathophysiology of AD.
Collapse
Affiliation(s)
| | - Gabriel Foo
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Antonius VanDongen
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Correspondence:
| |
Collapse
|
20
|
Mochida S. Mechanisms of Synaptic Vesicle Exo- and Endocytosis. Biomedicines 2022; 10:1593. [PMID: 35884898 PMCID: PMC9313035 DOI: 10.3390/biomedicines10071593] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 01/05/2023] Open
Abstract
Within 1 millisecond of action potential arrival at presynaptic terminals voltage-gated Ca2+ channels open. The Ca2+ channels are linked to synaptic vesicles which are tethered by active zone proteins. Ca2+ entrance into the active zone triggers: (1) the fusion of the vesicle and exocytosis, (2) the replenishment of the active zone with vesicles for incoming exocytosis, and (3) various types of endocytosis for vesicle reuse, dependent on the pattern of firing. These time-dependent vesicle dynamics are controlled by presynaptic Ca2+ sensor proteins, regulating active zone scaffold proteins, fusion machinery proteins, motor proteins, endocytic proteins, several enzymes, and even Ca2+ channels, following the decay of Ca2+ concentration after the action potential. Here, I summarize the Ca2+-dependent protein controls of synchronous and asynchronous vesicle release, rapid replenishment of the active zone, endocytosis, and short-term plasticity within 100 msec after the action potential. Furthermore, I discuss the contribution of active zone proteins to presynaptic plasticity and to homeostatic readjustment during and after intense activity, in addition to activity-dependent endocytosis.
Collapse
Affiliation(s)
- Sumiko Mochida
- Department of Physiology, Tokyo Medical University, Tokyo 160-8402, Japan
| |
Collapse
|
21
|
Tan C, Wang SSH, de Nola G, Kaeser PS. Rebuilding essential active zone functions within a synapse. Neuron 2022; 110:1498-1515.e8. [PMID: 35176221 PMCID: PMC9081183 DOI: 10.1016/j.neuron.2022.01.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 12/21/2021] [Accepted: 01/24/2022] [Indexed: 01/15/2023]
Abstract
Presynaptic active zones are molecular machines that control neurotransmitter secretion. They form sites for vesicle docking and priming and couple vesicles to Ca2+ entry for release triggering. The complexity of active zone machinery has made it challenging to determine its mechanisms in release. Simultaneous knockout of the active zone proteins RIM and ELKS disrupts active zone assembly, abolishes vesicle docking, and impairs release. We here rebuild docking, priming, and Ca2+ secretion coupling in these mutants without reinstating active zone networks. Re-expression of RIM zinc fingers recruited Munc13 to undocked vesicles and rendered the vesicles release competent. Action potential triggering of release was reconstituted by docking these primed vesicles to Ca2+ channels through attaching RIM zinc fingers to CaVβ4-subunits. Our work identifies an 80-kDa β4-Zn protein that bypasses the need for megadalton-sized secretory machines, establishes that fusion competence and docking are mechanistically separable, and defines RIM zinc finger-Munc13 complexes as hubs for active zone function.
Collapse
Affiliation(s)
- Chao Tan
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Shan Shan H Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni de Nola
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
22
|
Guerrero-Given D, Goldin SL, Thomas CI, Anthony SA, Jerez D, Kamasawa N. Gold In-and-Out: A Toolkit for Analyzing Subcellular Distribution of Immunogold-Labeled Membrane Proteins in Freeze-Fracture Replica Images. Front Neuroanat 2022; 16:855218. [PMID: 35444519 PMCID: PMC9014018 DOI: 10.3389/fnana.2022.855218] [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: 01/14/2022] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
Integral membrane proteins such as ion channels, transporters, and receptors shape cell activity and mediate cell-to-cell communication in the brain. The distribution, quantity, and clustering arrangement of those proteins contribute to the physiological properties of the cell; therefore, precise quantification of their state can be used to gain insight into cellular function. Using a highly sensitive immunoelectron microscopy technique called sodium dodecyl sulfate-digested freeze-fracture replica immunogold labeling (SDS-FRL), multiple membrane proteins can be tagged with different sizes of immunogold particles at once and visualized two-dimensionally. For quantification, gold particles in the images must be annotated, and then different mathematical and statistical methods must be applied to characterize the distribution states of proteins of interest. To perform such analyses in a user-friendly manner, we developed a program with a simple graphical user interface called Gold In-and-Out (GIO), which integrates several classical and novel analysis methods for immunogold labeled replicas into one self-contained package. GIO takes an input of particle coordinates, then allows users to implement analysis methods such as nearest neighbor distance (NND) and particle clustering. The program not only performs the selected analysis but also automatically compares the results of the real distribution to a random distribution of the same number of particles on the membrane region of interest. In addition to classical approaches for analyzing protein distribution, GIO includes new tools to analyze the positional bias of a target protein relative to a morphological landmark such as dendritic spines, and can also be applied for synaptic protein analysis. Gold Rippler provides a normalized metric of particle density that is resistant to differences in labeling efficiency among samples, while Gold Star is useful for quantifying distances between a protein and landmark. This package aims to help standardize analysis methods for subcellular and synaptic protein localization with a user-friendly interface while increasing the efficiency of these time-consuming analyses.
Collapse
Affiliation(s)
| | | | | | | | | | - Naomi Kamasawa
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Institute for Neuroscience, Jupiter, FL, United States
| |
Collapse
|
23
|
Phillips S, Ramos PV, Veeraraghavan P, Young SM. VikAD, a Vika site-specific recombinase-based system for efficient and scalable helper-dependent adenovirus production. Mol Ther Methods Clin Dev 2022; 24:117-126. [PMID: 35024378 PMCID: PMC8718833 DOI: 10.1016/j.omtm.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/04/2021] [Indexed: 11/09/2022]
Abstract
Recombinant viral vectors have become integral tools for basic in vivo research applications. Helper-dependent adenoviral (HdAd) vectors have a large packaging capacity of ∼36 kb of DNA that mediate long-term transgene expression in vitro and in vivo. The large carrying capacity of HdAd enables basic research or clinical applications requiring the delivery of large genes or multiple transgenes, which cannot be packaged into other widely used viral vectors. Currently, common HdAd production systems use an Ad helper virus (HV) with a packaging signal (Ψ) that is flanked by either loxP or FRT sites, which is excised in producer cells expressing Cre or Flp recombinases to prevent HV packaging. However, these production systems prevent the use of HdAd vectors for genetic strategies that rely on Cre or Flp recombination for cell-type-specific expression. To overcome these limitations, we developed the VikAD production system, which is based on producer cells expressing the Vika recombinase and an HV that contains a Ψ flanked by vox sites. The availability of this production system will greatly expand the utility and flexibility of HdAd vectors for use in research applications to monitor and manipulate cellular activity with increased specificity.
Collapse
Affiliation(s)
- Stacia Phillips
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, PBDB 5322, 169 Newton Road, Iowa City, IA 52242, USA
| | - Paula Valino Ramos
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, PBDB 5322, 169 Newton Road, Iowa City, IA 52242, USA
| | - Priyadharishini Veeraraghavan
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, PBDB 5322, 169 Newton Road, Iowa City, IA 52242, USA
| | - Samuel M. Young
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, PBDB 5322, 169 Newton Road, Iowa City, IA 52242, USA
- Department of Otolaryngology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| |
Collapse
|
24
|
Jackson RE, Compans B, Burrone J. Correlative Live-Cell and Super-Resolution Imaging to Link Presynaptic Molecular Organisation With Function. Front Synaptic Neurosci 2022; 14:830583. [PMID: 35242024 PMCID: PMC8885727 DOI: 10.3389/fnsyn.2022.830583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/12/2022] [Indexed: 11/13/2022] Open
Abstract
Information transfer at synapses occurs when vesicles fuse with the plasma membrane to release neurotransmitters, which then bind to receptors at the postsynaptic membrane. The process of neurotransmitter release varies dramatically between different synapses, but little is known about how this heterogeneity emerges. The development of super-resolution microscopy has revealed that synaptic proteins are precisely organised within and between the two parts of the synapse and that this precise spatiotemporal organisation fine-tunes neurotransmission. However, it remains unclear if variability in release probability could be attributed to the nanoscale organisation of one or several proteins of the release machinery. To begin to address this question, we have developed a pipeline for correlative functional and super-resolution microscopy, taking advantage of recent technological advancements enabling multicolour imaging. Here we demonstrate the combination of live imaging of SypHy-RGECO, a unique dual reporter that simultaneously measures presynaptic calcium influx and neurotransmitter release, with post hoc immunolabelling and multicolour single molecule localisation microscopy, to investigate the structure-function relationship at individual presynaptic boutons.
Collapse
Affiliation(s)
- Rachel E. Jackson
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Benjamin Compans
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Juan Burrone
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| |
Collapse
|
25
|
Takikawa K, Nishimune H. Similarity and Diversity of Presynaptic Molecules at Neuromuscular Junctions and Central Synapses. Biomolecules 2022; 12:biom12020179. [PMID: 35204679 PMCID: PMC8961632 DOI: 10.3390/biom12020179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/04/2022] Open
Abstract
Synaptic transmission is essential for controlling motor functions and maintaining brain functions such as walking, breathing, cognition, learning, and memory. Neurotransmitter release is regulated by presynaptic molecules assembled in active zones of presynaptic terminals. The size of presynaptic terminals varies, but the size of a single active zone and the types of presynaptic molecules are highly conserved among neuromuscular junctions (NMJs) and central synapses. Three parameters play an important role in the determination of neurotransmitter release properties at NMJs and central excitatory/inhibitory synapses: the number of presynaptic molecular clusters, the protein families of the presynaptic molecules, and the distance between presynaptic molecules and voltage-gated calcium channels. In addition, dysfunction of presynaptic molecules causes clinical symptoms such as motor and cognitive decline in patients with various neurological disorders and during aging. This review focuses on the molecular mechanisms responsible for the functional similarities and differences between excitatory and inhibitory synapses in the peripheral and central nervous systems, and summarizes recent findings regarding presynaptic molecules assembled in the active zone. Furthermore, we discuss the relationship between functional alterations of presynaptic molecules and dysfunction of NMJs or central synapses in diseases and during aging.
Collapse
Affiliation(s)
- Kenji Takikawa
- Laboratory of Neurobiology of Aging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan;
| | - Hiroshi Nishimune
- Laboratory of Neurobiology of Aging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan;
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu-shi, Tokyo 183-8538, Japan
- Correspondence: ; Tel.: +81-3-3964-3241
| |
Collapse
|
26
|
Skotte L, Fadista J, Bybjerg-Grauholm J, Appadurai V, Hildebrand MS, Hansen TF, Banasik K, Grove J, Albiñana C, Geller F, Bjurström CF, Vilhjálmsson BJ, Coleman M, Damiano JA, Burgess R, Scheffer IE, Pedersen OBV, Erikstrup C, Westergaard D, Nielsen KR, Sørensen E, Bruun MT, Liu X, Hjalgrim H, Pers TH, Mortensen PB, Mors O, Nordentoft M, Dreier JW, Børglum AD, Christensen J, Hougaard DM, Buil A, Hviid A, Melbye M, Ullum H, Berkovic SF, Werge T, Feenstra B. Genome-wide association study of febrile seizures implicates fever response and neuronal excitability genes. Brain 2022; 145:555-568. [PMID: 35022648 PMCID: PMC9128543 DOI: 10.1093/brain/awab260] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/09/2021] [Accepted: 06/07/2021] [Indexed: 12/19/2022] Open
Abstract
Febrile seizures represent the most common type of pathological brain activity in
young children and are influenced by genetic, environmental and developmental
factors. In a minority of cases, febrile seizures precede later development of
epilepsy. We conducted a genome-wide association study of febrile seizures in 7635 cases
and 83 966 controls identifying and replicating seven new loci, all with
P < 5 × 10−10. Variants at two loci were functionally related to altered expression of the fever
response genes PTGER3 and IL10, and four other
loci harboured genes (BSN, ERC2,
GABRG2, HERC1) influencing neuronal
excitability by regulating neurotransmitter release and binding, vesicular
transport or membrane trafficking at the synapse. Four previously reported loci
(SCN1A, SCN2A, ANO3 and
12q21.33) were all confirmed. Collectively, the seven novel and four previously
reported loci explained 2.8% of the variance in liability to febrile
seizures, and the single nucleotide polymorphism heritability based on all
common autosomal single nucleotide polymorphisms was 10.8%.
GABRG2, SCN1A and SCN2A
are well-established epilepsy genes and, overall, we found positive genetic
correlations with epilepsies (rg = 0.39,
P = 1.68 × 10−4). Further,
we found that higher polygenic risk scores for febrile seizures were associated
with epilepsy and with history of hospital admission for febrile seizures.
Finally, we found that polygenic risk of febrile seizures was lower in febrile
seizure patients with neuropsychiatric disease compared to febrile seizure
patients in a general population sample. In conclusion, this largest genetic investigation of febrile seizures to date
implicates central fever response genes as well as genes affecting neuronal
excitability, including several known epilepsy genes. Further functional and
genetic studies based on these findings will provide important insights into the
complex pathophysiological processes of seizures with and without fever.
Collapse
Affiliation(s)
- Line Skotte
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - João Fadista
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Sciences, Lund University Diabetes Centre, Malmö, Sweden
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Jonas Bybjerg-Grauholm
- Danish Centre for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
| | - Vivek Appadurai
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Mental Health Center Sct. Hans, Mental Health Services, Capital Region Denmark, Roskilde, Denmark
| | - Michael S Hildebrand
- Epilepsy Research Centre, Department of Medicine, University of Melbourne (Austin Health), Victoria, Australia
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
| | - Thomas F Hansen
- Danish Headache Center, Department of Neurology, Rigshospitalet-Glostrup, Denmark
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Grove
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine–Human Genetics, Aarhus University, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Clara Albiñana
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Carmen F Bjurström
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Bjarni J Vilhjálmsson
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Matthew Coleman
- Epilepsy Research Centre, Department of Medicine, University of Melbourne (Austin Health), Victoria, Australia
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
| | - John A Damiano
- Epilepsy Research Centre, Department of Medicine, University of Melbourne (Austin Health), Victoria, Australia
| | - Rosemary Burgess
- Epilepsy Research Centre, Department of Medicine, University of Melbourne (Austin Health), Victoria, Australia
| | - Ingrid E Scheffer
- Epilepsy Research Centre, Department of Medicine, University of Melbourne (Austin Health), Victoria, Australia
- Department of Paediatrics, Royal Children's Hospital, The University of Melbourne, Flemington, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, Heidelberg, Victoria, Australia
| | | | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Kaspar René Nielsen
- Department of Clinical Immunology, Aalborg University Hospital North, Aalborg, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Mie Topholm Bruun
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | - Xueping Liu
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Henrik Hjalgrim
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Haematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Preben Bo Mortensen
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Ole Mors
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
| | - Merete Nordentoft
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Mental Health Center Copenhagen, Mental Health Services in the Capital Region of Denmark, Copenhagen, Denmark
| | - Julie W Dreier
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Anders D Børglum
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine–Human Genetics, Aarhus University, Aarhus, Denmark
| | - Jakob Christensen
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - David M Hougaard
- Danish Centre for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
| | - Alfonso Buil
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Mental Health Center Sct. Hans, Mental Health Services, Capital Region Denmark, Roskilde, Denmark
| | - Anders Hviid
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Drug Design and Pharmacology, Pharmacovigilance Research Center, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Henrik Ullum
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Statens Serum Institut, Copenhagen, Denmark
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, University of Melbourne (Austin Health), Victoria, Australia
| | - Thomas Werge
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Mental Health Center Sct. Hans, Mental Health Services, Capital Region Denmark, Roskilde, Denmark
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
27
|
Cheng P, Chen K, Zhang S, Mu KT, Liang S, Zhang Y. IDH1 R132C and ERC2 L309I Mutations Contribute to the Development of Maffucci's Syndrome. Front Endocrinol (Lausanne) 2021; 12:763349. [PMID: 34790172 PMCID: PMC8591216 DOI: 10.3389/fendo.2021.763349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Background Maffucci's syndrome is characterized by the coexistence of multiple enchondromas and soft-tissue hemangiomas. It has been clear that somatic mosaic isocitrate dehydrogenase type 1 (IDH1) or isocitrate dehydrogenase type 2 (IDH2) mutations are associated with Maffucci's syndrome and Ollier disease, but the mechanisms underlying hemangiomas of the Maffucci's syndrome is still obscure. This study aimed to determine the mechanism of hemangiomas in Maffucci's syndrome. Methods We received a 26-year-old female patient with typical Maffucci's syndrome, and exome sequencing was conducted using DNA from her peripheral blood and enchondroma tissues. Somatic mutations were characterized by a comparative analysis of exome sequences and further confirmed by the sequencing of PCR products derived from original blood and tissue samples. The mutations of an additional 69 patients with Ollier disease were further tested. The functional impacts of these somatic mutations on Maffucci's syndrome, especially the development of hemangiomas, were evaluated. Results We reported a typical case of Maffucci's syndrome, which was confirmed by both imaging findings and pathology. Through exome sequencing of this patient's DNA samples, we identified an R132C mutation in the isocitrate dehydrogenase type 1 (IDH1) gene and an L309I mutation in the ELKS/RAB6-interacting/CAST family member 2 (ERC2) gene in this patient. Approximately 33.3% of the clones were positive for the IDH1 R132C mutation, and 19.0% of the clones were positive for the ECR2 L309I mutation. The IDH1 R132C mutation was detected in most of the patients with Ollier disease (51/69 patients), and the mean frequency of this mutation was 63.3% in total sequence readouts, but the ECR2 L309I mutation was absent in all of the patients with Ollier disease. In vitro experiments confirmed that the IDH1 R132C mutation promotes chondrocyte proliferation, and the ERC2 L309I mutation enhances angiogenesis. Conclusions Our results suggest that while IDH1 is a known pathogenic gene in enchondromatosis, ERC2 is a novel gene identified in Maffucci's syndrome. The somatic L309I mutation of ERC2 contributes to the pathogenesis of hypervascularization to facilitate the development of hemangiomas in Maffucci's syndrome. The combination of the IDH1 R132C and ERC2 L309I mutations contributes to the development of Maffucci's syndrome, and these results may enable further research on the pathogenesis of Maffucci's syndrome.
Collapse
Affiliation(s)
- Peng Cheng
- Department of Orthopedics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Chen
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Shu Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Huazhong University of Science and Technology, Wuhan, China
| | - Ke-tao Mu
- Department of Radiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Liang
- Department of Orthopedics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Zhang
- Department of Nephrology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Mochida S. Stable and Flexible Synaptic Transmission Controlled by the Active Zone Protein Interactions. Int J Mol Sci 2021; 22:ijms222111775. [PMID: 34769208 PMCID: PMC8583982 DOI: 10.3390/ijms222111775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 12/28/2022] Open
Abstract
An action potential triggers neurotransmitter release from synaptic vesicles docking to a specialized release site of the presynaptic plasma membrane, the active zone. The active zone is a highly organized structure with proteins that serves as a platform for synaptic vesicle exocytosis, mediated by SNAREs complex and Ca2+ sensor proteins, within a sub-millisecond opening of nearby Ca2+ channels with the membrane depolarization. In response to incoming neuronal signals, each active zone protein plays a role in the release-ready site replenishment with synaptic vesicles for sustainable synaptic transmission. The active zone release apparatus provides a possible link between neuronal activity and plasticity. This review summarizes the mostly physiological role of active zone protein interactions that control synaptic strength, presynaptic short-term plasticity, and homeostatic synaptic plasticity.
Collapse
Affiliation(s)
- Sumiko Mochida
- Department of Physiology, Tokyo Medical University, Tokyo 160-8402, Japan
| |
Collapse
|
29
|
Mrestani A, Pauli M, Kollmannsberger P, Repp F, Kittel RJ, Eilers J, Doose S, Sauer M, Sirén AL, Heckmann M, Paul MM. Active zone compaction correlates with presynaptic homeostatic potentiation. Cell Rep 2021; 37:109770. [PMID: 34610300 DOI: 10.1016/j.celrep.2021.109770] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 05/14/2021] [Accepted: 09/07/2021] [Indexed: 12/30/2022] Open
Abstract
Neurotransmitter release is stabilized by homeostatic plasticity. Presynaptic homeostatic potentiation (PHP) operates on timescales ranging from minute- to life-long adaptations and likely involves reorganization of presynaptic active zones (AZs). At Drosophila melanogaster neuromuscular junctions, earlier work ascribed AZ enlargement by incorporating more Bruchpilot (Brp) scaffold protein a role in PHP. We use localization microscopy (direct stochastic optical reconstruction microscopy [dSTORM]) and hierarchical density-based spatial clustering of applications with noise (HDBSCAN) to study AZ plasticity during PHP at the synaptic mesoscale. We find compaction of individual AZs in acute philanthotoxin-induced and chronic genetically induced PHP but unchanged copy numbers of AZ proteins. Compaction even occurs at the level of Brp subclusters, which move toward AZ centers, and in Rab3 interacting molecule (RIM)-binding protein (RBP) subclusters. Furthermore, correlative confocal and dSTORM imaging reveals how AZ compaction in PHP translates into apparent increases in AZ area and Brp protein content, as implied earlier.
Collapse
Affiliation(s)
- Achmed Mrestani
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany; Department of Neurology, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Martin Pauli
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany; Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Philip Kollmannsberger
- Center for Computational and Theoretical Biology, Julius Maximilians University Würzburg, 97074 Würzburg, Germany
| | - Felix Repp
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany; Center for Computational and Theoretical Biology, Julius Maximilians University Würzburg, 97074 Würzburg, Germany; Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Robert J Kittel
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany; Institute of Biology, Department of Animal Physiology, Leipzig University, 04103 Leipzig, Germany; Carl-Ludwig-Institute for Physiology, Leipzig University, 04103 Leipzig, Germany
| | - Jens Eilers
- Carl-Ludwig-Institute for Physiology, Leipzig University, 04103 Leipzig, Germany
| | - Sören Doose
- Department of Biotechnology and Biophysics, Julius Maximilians University Würzburg, 97074 Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Julius Maximilians University Würzburg, 97074 Würzburg, Germany
| | - Anna-Leena Sirén
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany; Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Manfred Heckmann
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany.
| | - Mila M Paul
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany; Department of Orthopaedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany.
| |
Collapse
|
30
|
Oh KH, Krout MD, Richmond JE, Kim H. UNC-2 CaV2 Channel Localization at Presynaptic Active Zones Depends on UNC-10/RIM and SYD-2/Liprin-α in Caenorhabditis elegans. J Neurosci 2021; 41:4782-4794. [PMID: 33975919 PMCID: PMC8260173 DOI: 10.1523/jneurosci.0076-21.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/07/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022] Open
Abstract
Presynaptic active zone proteins couple calcium influx with synaptic vesicle exocytosis. However, the control of presynaptic calcium channel localization by active zone proteins is not completely understood. In a Caenorhabditis elegans (C. elegans) forward genetic screen, we find that UNC-10/RIM (Rab3-interacting molecule) and SYD-2/Liprin-α regulate presynaptic localization of UNC-2, the CaV2 channel ortholog. We further quantitatively analyzed live animals using endogenously GFP-tagged UNC-2 and active zone components. Consistent with the interaction between RIM and CaV2 in mammals, the intensity and number of UNC-2 channel puncta at presynaptic terminals were greatly reduced in unc-10 mutant animals. To understand how SYD-2 regulates presynaptic UNC-2 channel localization, we analyzed presynaptic localization of endogenous SYD-2, UNC-10, RIMB-1/RIM-BP (RIM binding protein), and ELKS-1. Our analysis revealed that although SYD-2 is the most critical for active zone assembly, loss of SYD-2 function does not completely abolish presynaptic localization of UNC-10, RIMB-1, and ELKS-1, suggesting an existence of SYD-2-independent active zone assembly. UNC-2 localization analysis in double and triple mutants of active zone components show that SYD-2 promotes UNC-2 localization by partially controlling UNC-10 localization, and ELKS-1 and RIMB-1 also contribute to UNC-2 channel localization. In addition, we find that core active zone proteins are unequal in their abundance. Although the abundance of UNC-10 at the active zone is comparable to UNC-2, SYD-2 and ELKS-1 are twice more and RIMB-1 four times more abundant than UNC-2. Together our data show that UNC-10, SYD-2, RIMB-1, and ELKS-1 control presynaptic UNC-2 channel localization in redundant yet distinct manners.SIGNIFICANCE STATEMENT Precise control of neurotransmission is dependent on the tight coupling of the calcium influx through voltage-gated calcium channels (VGCCs) to the exocytosis machinery at the presynaptic active zones. However, how these VGCCs are tethered to the active zone is incompletely understood. To understand the mechanism of presynaptic VGCC localization, we performed a C. elegans forward genetic screen and quantitatively analyzed endogenous active zones and presynaptic VGCCs. In addition to RIM, our study finds that SYD-2/Liprin-α is critical for presynaptic localization of VGCCs. Yet, the loss of SYD-2, a core active zone scaffolding protein, does not completely abolish the presynaptic localization of the VGCC, showing that the active zone is a resilient structure assembled by redundant mechanisms.
Collapse
Affiliation(s)
- Kelly H Oh
- Center for Cancer Cell Biology, Immunology, and Infection, Department of Cell Biology and Anatomy, Chicago Medical School, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| | - Mia D Krout
- Department of Biological Science, University of Illinois at Chicago, Chicago, Illinois 60607
| | - Janet E Richmond
- Department of Biological Science, University of Illinois at Chicago, Chicago, Illinois 60607
| | - Hongkyun Kim
- Center for Cancer Cell Biology, Immunology, and Infection, Department of Cell Biology and Anatomy, Chicago Medical School, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| |
Collapse
|
31
|
Nyitrai H, Wang SSH, Kaeser PS. ELKS1 Captures Rab6-Marked Vesicular Cargo in Presynaptic Nerve Terminals. Cell Rep 2021; 31:107712. [PMID: 32521280 PMCID: PMC7360120 DOI: 10.1016/j.celrep.2020.107712] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/07/2020] [Accepted: 05/06/2020] [Indexed: 11/28/2022] Open
Abstract
Neurons face unique transport challenges. They need to deliver cargo over long axonal distances and to many presynaptic nerve terminals. Rab GTPases are master regulators of vesicular traffic, but essential presynaptic Rabs have not been identified. Here, we find that Rab6, a Golgi-derived GTPase for constitutive secretion, associates with mobile axonal cargo and localizes to nerve terminals. ELKS1 is a stationary presynaptic protein with Golgin homology that binds to Rab6. Knockout and rescue experiments for ELKS1 and Rab6 establish that ELKS1 captures Rab6 cargo. The ELKS1-Rab6-capturing mechanism can be transferred to mitochondria by mistargeting ELKS1 or Rab6 to them. We conclude that nerve terminals have repurposed mechanisms from constitutive exocytosis for their highly regulated secretion. By employing Golgin-like mechanisms with anchored ELKS extending its coiled-coils to capture Rab6 cargo, they have spatially separated cargo capture from fusion. ELKS complexes connect to active zones and may mediate vesicle progression toward release sites.
Collapse
Affiliation(s)
- Hajnalka Nyitrai
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Shan Shan H Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
32
|
A deep learning approach to identifying immunogold particles in electron microscopy images. Sci Rep 2021; 11:7771. [PMID: 33833289 PMCID: PMC8032809 DOI: 10.1038/s41598-021-87015-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
Electron microscopy (EM) enables high-resolution visualization of protein distributions in biological tissues. For detection, gold nanoparticles are typically used as an electron-dense marker for immunohistochemically labeled proteins. Manual annotation of gold particle labels is laborious and time consuming, as gold particle counts can exceed 100,000 across hundreds of image segments to obtain conclusive data sets. To automate this process, we developed Gold Digger, a software tool that uses a modified pix2pix deep learning network capable of detecting and annotating colloidal gold particles in biological EM images obtained from both freeze-fracture replicas and plastic sections prepared with the post-embedding method. Gold Digger performs at near-human-level accuracy, can handle large images, and includes a user-friendly tool with a graphical interface for proof reading outputs by users. Manual error correction also helps for continued re-training of the network to improve annotation accuracy over time. Gold Digger thus enables rapid high-throughput analysis of immunogold-labeled EM data and is freely available to the research community.
Collapse
|
33
|
Xie X, Liang M, Yu C, Wei Z. Liprin-α-Mediated Assemblies and Their Roles in Synapse Formation. Front Cell Dev Biol 2021; 9:653381. [PMID: 33869211 PMCID: PMC8044993 DOI: 10.3389/fcell.2021.653381] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/25/2021] [Indexed: 01/20/2023] Open
Abstract
Brain's functions, such as memory and learning, rely on synapses that are highly specialized cellular junctions connecting neurons. Functional synapses orchestrate the assembly of ion channels, receptors, enzymes, and scaffold proteins in both pre- and post-synapse. Liprin-α proteins are master scaffolds in synapses and coordinate various synaptic proteins to assemble large protein complexes. The functions of liprin-αs in synapse formation have been largely uncovered by genetic studies in diverse model systems. Recently, emerging structural and biochemical studies on liprin-α proteins and their binding partners begin to unveil the molecular basis of the synaptic assembly. This review summarizes the recent structural findings on liprin-αs, proposes the assembly mechanism of liprin-α-mediated complexes, and discusses the liprin-α-organized assemblies in the regulation of synapse formation and function.
Collapse
Affiliation(s)
- Xingqiao Xie
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
- Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, China
| | - Mingfu Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Cong Yu
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, China
| | - Zhiyi Wei
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
- Brain Research Center, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
34
|
Young SM, Veeraraghavan P. Presynaptic voltage-gated calcium channels in the auditory brainstem. Mol Cell Neurosci 2021; 112:103609. [PMID: 33662542 DOI: 10.1016/j.mcn.2021.103609] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/06/2021] [Accepted: 02/17/2021] [Indexed: 10/22/2022] Open
Abstract
Sound information encoding within the initial synapses in the auditory brainstem requires reliable and precise synaptic transmission in response to rapid and large fluctuations in action potential (AP) firing rates. The magnitude and location of Ca2+ entry through voltage-gated Ca2+ channels (CaV) in the presynaptic terminal are key determinants in triggering AP-mediated release. In the mammalian central nervous system (CNS), the CaV2.1 subtype is the critical subtype for CNS function, since it is the most efficient CaV2 subtype in triggering AP-mediated synaptic vesicle (SV) release. Auditory brainstem synapses utilize CaV2.1 to sustain fast and repetitive SV release to encode sound information. Therefore, understanding the presynaptic mechanisms that control CaV2.1 localization, organization and biophysical properties are integral to understanding auditory processing. Here, we review our current knowledge about the control of presynaptic CaV2 abundance and organization in the auditory brainstem and impact on the regulation of auditory processing.
Collapse
Affiliation(s)
- Samuel M Young
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Otolaryngology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | | |
Collapse
|
35
|
Mochida S. Neurotransmitter Release Site Replenishment and Presynaptic Plasticity. Int J Mol Sci 2020; 22:ijms22010327. [PMID: 33396919 PMCID: PMC7794938 DOI: 10.3390/ijms22010327] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/23/2020] [Accepted: 12/27/2020] [Indexed: 12/19/2022] Open
Abstract
An action potential (AP) triggers neurotransmitter release from synaptic vesicles (SVs) docking to a specialized release site of presynaptic plasma membrane, the active zone (AZ). The AP simultaneously controls the release site replenishment with SV for sustainable synaptic transmission in response to incoming neuronal signals. Although many studies have suggested that the replenishment time is relatively slow, recent studies exploring high speed resolution have revealed SV dynamics with milliseconds timescale after an AP. Accurate regulation is conferred by proteins sensing Ca2+ entering through voltage-gated Ca2+ channels opened by an AP. This review summarizes how millisecond Ca2+ dynamics activate multiple protein cascades for control of the release site replenishment with release-ready SVs that underlie presynaptic short-term plasticity.
Collapse
Affiliation(s)
- Sumiko Mochida
- Department of Physiology, Tokyo Medical University, Tokyo 160-8402, Japan
| |
Collapse
|
36
|
Cong S, Yao X, Huang Z, Risacher SL, Nho K, Saykin AJ, Shen L. Volumetric GWAS of medial temporal lobe structures identifies an ERC1 locus using ADNI high-resolution T2-weighted MRI data. Neurobiol Aging 2020; 95:81-93. [PMID: 32768867 PMCID: PMC7609616 DOI: 10.1016/j.neurobiolaging.2020.07.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/09/2020] [Accepted: 07/04/2020] [Indexed: 12/18/2022]
Abstract
Medial temporal lobe (MTL) consists of hippocampal subfields and neighboring cortices. These heterogeneous structures are differentially involved in memory, cognitive and emotional functions, and present nonuniformly distributed atrophy contributing to cognitive disorders. This study aims to examine how genetics influences Alzheimer's disease (AD) pathogenesis via MTL substructures by analyzing high-resolution magnetic resonance imaging (MRI) data. We performed genome-wide association study to examine the associations between 565,373 single nucleotide polymorphisms (SNPs) and 14 MTL substructure volumes. A novel association with right Brodmann area 36 volume was discovered in an ERC1 SNP (i.e., rs2968869). Further analyses on larger samples found rs2968869 to be associated with gray matter density and glucose metabolism measures in the right hippocampus, and disease status. Tissue-specific transcriptomic analysis identified the minor allele of rs2968869 (rs2968869-C) to be associated with reduced ERC1 expression in the hippocampus. All the findings indicated a protective role of rs2968869-C in AD. We demonstrated the power of high-resolution MRI and the promise of fine-grained MTL substructures for revealing the genetic basis of AD biomarkers.
Collapse
Affiliation(s)
- Shan Cong
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, USA
| | - Xiaohui Yao
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhi Huang
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, USA
| | - Shannon L Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Rizalar FS, Roosen DA, Haucke V. A Presynaptic Perspective on Transport and Assembly Mechanisms for Synapse Formation. Neuron 2020; 109:27-41. [PMID: 33098763 DOI: 10.1016/j.neuron.2020.09.038] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/26/2020] [Accepted: 09/25/2020] [Indexed: 01/01/2023]
Abstract
Neurons are highly polarized cells with a single axon and multiple dendrites derived from the cell body to form tightly associated pre- and postsynaptic compartments. As the biosynthetic machinery is largely restricted to the somatodendritic domain, the vast majority of presynaptic components are synthesized in the neuronal soma, packaged into synaptic precursor vesicles, and actively transported along the axon to sites of presynaptic biogenesis. In contrast with the significant progress that has been made in understanding synaptic transmission and processing of information at the post-synapse, comparably little is known about the formation and dynamic remodeling of the presynaptic compartment. We review here our current understanding of the mechanisms that govern the biogenesis, transport, and assembly of the key components for presynaptic neurotransmission, discuss how alterations in presynaptic assembly may impact nervous system function or lead to disease, and outline key open questions for future research.
Collapse
Affiliation(s)
- Filiz Sila Rizalar
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Dorien A Roosen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany.
| |
Collapse
|
38
|
Abstract
Background Insulin is stored within large dense-core granules in pancreatic beta (β)-cells and is released by Ca2+-triggered exocytosis with increasing blood glucose levels. Polarized and targeted secretion of insulin from β-cells in pancreatic islets into the vasculature has been proposed; however, the mechanisms related to cellular and molecular localization remain largely unknown. Within nerve terminals, the Ca2+-dependent release of a polarized transmitter is limited to the active zone, a highly specialized area of the presynaptic membrane. Several active zone-specific proteins have been characterized; among them, the CAST/ELKS protein family members have the ability to form large protein complexes with other active zone proteins to control the structure and function of the active zone for tight regulation of neurotransmitter release. Notably, ELKS but not CAST is also expressed in β-cells, implying that ELKS may be involved in polarized insulin secretion from β-cells. Scope of review This review provides an overview of the current findings regarding the role(s) of ELKS and other active zone proteins in β-cells and focuses on the molecular mechanism underlying ELKS regulation within polarized insulin secretion from islets. Major conclusions ELKS localizes at the vascular-facing plasma membrane of β-cells in mouse pancreatic islets. ELKS forms a potent insulin secretion complex with L-type voltage-dependent Ca2+ channels on the vascular-facing plasma membrane of β-cells, enabling polarized Ca2+ influx and first-phase insulin secretion from islets. This model provides novel insights into the functional polarity observed during insulin secretion from β-cells within islets at the molecular level. This active zone-like region formed by ELKS at the vascular side of the plasma membrane is essential for coordinating physiological insulin secretion and may be disrupted in diabetes.
Collapse
Affiliation(s)
- Mica Ohara-Imaizumi
- Department of Cellular Biochemistry, Kyorin University School of Medicine, Tokyo 181-8611, Japan.
| | - Kyota Aoyagi
- Department of Cellular Biochemistry, Kyorin University School of Medicine, Tokyo 181-8611, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| |
Collapse
|
39
|
Radulovic T, Dong W, Goral RO, Thomas CI, Veeraraghavan P, Montesinos MS, Guerrero-Given D, Goff K, Lübbert M, Kamasawa N, Ohtsuka T, Young SM. Presynaptic development is controlled by the core active zone proteins CAST/ELKS. J Physiol 2020; 598:2431-2452. [PMID: 32304329 DOI: 10.1113/jp279736] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/14/2020] [Indexed: 12/24/2022] Open
Abstract
KEY POINTS CAST/ELKS are positive regulators of presynaptic growth and are suppressors of active zone expansion at the developing mouse calyx of Held. CAST/ELKS regulate all three CaV 2 subtype channel levels in the presynaptic terminal and not just CaV 2.1. The half-life of ELKS is on the timescale of days and not weeks. Synaptic transmission was not impacted by the loss of CAST/ELKS. CAST/ELKS are involved in pathways regulating morphological properties of presynaptic terminals during an early stage of circuit maturation. ABSTRACT Many presynaptic active zone (AZ) proteins have multiple regulatory roles that vary during distinct stages of neuronal circuit development. The CAST/ELKS protein family are evolutionarily conserved presynaptic AZ molecules that regulate presynaptic calcium channels, synaptic transmission and plasticity in the mammalian CNS. However, how these proteins regulate synapse development and presynaptic function in a developing neuronal circuit in its native environment is unclear. To unravel the roles of CAST/ELKS in glutamatergic synapse development and in presynaptic function, we used CAST knockout (KO) and ELKS conditional KO (CKO) mice to examine how their loss during the early stages of circuit maturation impacted the calyx of Held presynaptic terminal development and function. Morphological analysis from confocal z-stacks revealed that combined deletion of CAST/ELKS resulted in a reduction in the surface area and volume of the calyx. Analysis of AZ ultrastructure showed that AZ size was increased in the absence of CAST/ELKS. Patch clamp recordings demonstrated a reduction of all presynaptic CaV 2 channel subtype currents that correlated with a loss in presynaptic CaV 2 channel numbers. However, these changes did not impair synaptic transmission and plasticity and synaptic vesicle release kinetics. We conclude that CAST/ELKS proteins are positive regulators of presynaptic growth and are suppressors of AZ expansion and CaV 2 subtype currents and levels during calyx of Held development. We propose that CAST/ELKS are involved in pathways regulating presynaptic morphological properties and CaV 2 channel subtypes and suggest there is developmental compensation to preserve synaptic transmission during early stages of neuronal circuit maturation.
Collapse
Affiliation(s)
- Tamara Radulovic
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research of Southwest Medical University, Luzhou, 646000, China
| | - R Oliver Goral
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Connon I Thomas
- Electron Microscopy Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | | | - Monica Suarez Montesinos
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Debbie Guerrero-Given
- Electron Microscopy Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Kevin Goff
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Matthias Lübbert
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Naomi Kamasawa
- Electron Microscopy Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Toshihisa Ohtsuka
- Department of Biochemistry , Graduate School of Medicine/Faculty of Medicine, University of Yamanashi, 1110 Shimokato Chuo, Yamanashi, 409-3898, Japan
| | - Samuel M Young
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.,Department of Otolaryngology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
40
|
Dolphin AC, Lee A. Presynaptic calcium channels: specialized control of synaptic neurotransmitter release. Nat Rev Neurosci 2020; 21:213-229. [PMID: 32161339 PMCID: PMC7873717 DOI: 10.1038/s41583-020-0278-2] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2020] [Indexed: 11/09/2022]
Abstract
Chemical synapses are heterogeneous junctions formed between neurons that are specialized for the conversion of electrical impulses into the exocytotic release of neurotransmitters. Voltage-gated Ca2+ channels play a pivotal role in this process as they are the major conduits for the Ca2+ ions that trigger the fusion of neurotransmitter-containing vesicles with the presynaptic membrane. Alterations in the intrinsic function of these channels and their positioning within the active zone can profoundly alter the timing and strength of synaptic output. Advances in optical and electron microscopic imaging, structural biology and molecular techniques have facilitated recent breakthroughs in our understanding of the properties of voltage-gated Ca2+ channels that support their presynaptic functions. Here we examine the nature of these channels, how they are trafficked to and anchored within presynaptic boutons, and the mechanisms that allow them to function optimally in shaping the flow of information through neural circuits.
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
41
|
ELKS/Voltage-Dependent Ca 2+ Channel-β Subunit Module Regulates Polarized Ca 2+ Influx in Pancreatic β Cells. Cell Rep 2020; 26:1213-1226.e7. [PMID: 30699350 DOI: 10.1016/j.celrep.2018.12.106] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 10/29/2018] [Accepted: 12/27/2018] [Indexed: 12/14/2022] Open
Abstract
Pancreatic β cells secrete insulin by Ca2+-triggered exocytosis. However, there is no apparent secretory site similar to the neuronal active zones, and the cellular and molecular localization mechanism underlying polarized exocytosis remains elusive. Here, we report that ELKS, a vertebrate active zone protein, is used in β cells to regulate Ca2+ influx for insulin secretion. β cell-specific ELKS-knockout (KO) mice showed impaired glucose-stimulated first-phase insulin secretion and reduced L-type voltage-dependent Ca2+ channel (VDCC) current density. In situ Ca2+ imaging of β cells within islets expressing a membrane-bound G-CaMP8b Ca2+ sensor demonstrated initial local Ca2+ signals at the ELKS-localized vascular side of the β cell plasma membrane, which were markedly decreased in ELKS-KO β cells. Mechanistically, ELKS directly interacted with the VDCC-β subunit via the GK domain. These findings suggest that ELKS and VDCCs form a potent insulin secretion complex at the vascular side of the β cell plasma membrane for polarized Ca2+ influx and first-phase insulin secretion from pancreatic islets.
Collapse
|
42
|
Hagiwara A, Hamada S, Hida Y, Ohtsuka T. Double deletion of the active zone proteins CAST/ELKS in the mouse forebrain causes high mortality of newborn pups. Mol Brain 2020; 13:13. [PMID: 31996256 PMCID: PMC6988216 DOI: 10.1186/s13041-020-0557-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/21/2020] [Indexed: 11/10/2022] Open
Abstract
Presynaptic active zone cytomatrix proteins are essential elements of neurotransmitter release machinery that govern neural transmission. Among active zone proteins, cytomatrix at the active zone-associated structural protein (CAST) is known to regulate active zone size in retinal photoreceptors and neurotransmitter release by recruiting Ca2+ channels at various synapses. However, the role of ELKS-a protein from the same family as CAST-and the synergistic roles of CAST/ELKS have not been thoroughly investigated, particularly with regard to mouse behavior. Here, we generated ELKS conditional KO in mouse forebrain synapses by crossing ELKS flox mice with a CaMKII promoter-induced Cre line. Results showed that CAST is dominant at these synapses and that ELKS can support CAST function, but is less effective in the ELKS single KO. Pups of CAST/ELKS double KO in the forebrain were born in Mendelian rations but resulted in eventual death right after the birth. Anatomically, the forebrain neuronal compositions of CAST KO and CAST/ELKS double KO mice were indistinguishable, and the sensory neural network from whiskers on the face was identified as barrelette-like patches in the spinal trigeminal nucleus. Therefore, depletion of CAST and ELKS disrupts neurotransmission from sensory to motor networks, which can lead to deficits in exploration and failure to suckle.
Collapse
Affiliation(s)
- Akari Hagiwara
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Shun Hamada
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Yamato Hida
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan.
| |
Collapse
|
43
|
Ge D, Noakes PG, Lavidis NA. What are Neurotransmitter Release Sites and Do They Interact? Neuroscience 2020; 425:157-168. [DOI: 10.1016/j.neuroscience.2019.11.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 11/10/2019] [Accepted: 11/11/2019] [Indexed: 12/22/2022]
|
44
|
Presynaptic Mitochondria Volume and Abundance Increase during Development of a High-Fidelity Synapse. J Neurosci 2019; 39:7994-8012. [PMID: 31455662 DOI: 10.1523/jneurosci.0363-19.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 08/19/2019] [Accepted: 08/22/2019] [Indexed: 12/16/2022] Open
Abstract
The calyx of Held, a large glutamatergic presynaptic terminal in the auditory brainstem undergoes developmental changes to support the high action-potential firing rates required for auditory information encoding. In addition, calyx terminals are morphologically diverse, which impacts vesicle release properties and synaptic plasticity. Mitochondria influence synaptic plasticity through calcium buffering and are crucial for providing the energy required for synaptic transmission. Therefore, it has been postulated that mitochondrial levels increase during development and contribute to the morphological-functional diversity in the mature calyx. However, the developmental profile of mitochondrial volumes and subsynaptic distribution at the calyx of Held remains unclear. To provide insight on this, we developed a helper-dependent adenoviral vector that expresses the genetically encoded peroxidase marker for mitochondria, mito-APEX2, at the mouse calyx of Held. We developed protocols to detect labeled mitochondria for use with serial block face scanning electron microscopy to carry out semiautomated segmentation of mitochondria, high-throughput whole-terminal reconstruction, and presynaptic ultrastructure in mice of either sex. Subsequently, we measured mitochondrial volumes and subsynaptic distributions at the immature postnatal day (P)7 and the mature (P21) calyx. We found an increase of mitochondria volumes in terminals and axons from P7 to P21 but did not observe differences between stalk and swelling subcompartments in the mature calyx. Based on these findings, we propose that mitochondrial volumes and synaptic localization developmentally increase to support high firing rates required in the initial stages of auditory information processing.SIGNIFICANCE STATEMENT Elucidating the developmental processes of auditory brainstem presynaptic terminals is critical to understanding auditory information encoding. Additionally, morphological-functional diversity at these terminals is proposed to enhance coding capacity. Mitochondria provide energy for synaptic transmission and can buffer calcium, impacting synaptic plasticity; however, their developmental profile to ultimately support the energetic demands of synapses following the onset of hearing remains unknown. Therefore, we created a helper-dependent adenoviral vector with the mitochondria-targeting peroxidase mito-APEX2 and expressed it at the mouse calyx of Held. Volumetric reconstructions of serial block face electron microscopy data of immature and mature labeled calyces reveal that mitochondrial volumes are increased to support high firing rates upon maturity.
Collapse
|
45
|
Hendi A, Kurashina M, Mizumoto K. Intrinsic and extrinsic mechanisms of synapse formation and specificity in C. elegans. Cell Mol Life Sci 2019; 76:2719-2738. [PMID: 31037336 PMCID: PMC11105629 DOI: 10.1007/s00018-019-03109-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 12/18/2022]
Abstract
Precise neuronal wiring is critical for the function of the nervous system and is ultimately determined at the level of individual synapses. Neurons integrate various intrinsic and extrinsic cues to form synapses onto their correct targets in a stereotyped manner. In the past decades, the nervous system of nematode (Caenorhabditis elegans) has provided the genetic platform to reveal the genetic and molecular mechanisms of synapse formation and specificity. In this review, we will summarize the recent discoveries in synapse formation and specificity in C. elegans.
Collapse
Affiliation(s)
- Ardalan Hendi
- Department of Zoology, The University of British Columbia, 2406-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Mizuki Kurashina
- Department of Zoology, The University of British Columbia, 2406-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Kota Mizumoto
- Department of Zoology, The University of British Columbia, 2406-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
46
|
Goel P, Dufour Bergeron D, Böhme MA, Nunnelly L, Lehmann M, Buser C, Walter AM, Sigrist SJ, Dickman D. Homeostatic scaling of active zone scaffolds maintains global synaptic strength. J Cell Biol 2019; 218:1706-1724. [PMID: 30914419 PMCID: PMC6504899 DOI: 10.1083/jcb.201807165] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/14/2018] [Accepted: 03/06/2019] [Indexed: 12/23/2022] Open
Abstract
Synaptic terminals grow and retract throughout life, yet synaptic strength is maintained within stable physiological ranges. To study this process, we investigated Drosophila endophilin (endo) mutants. Although active zone (AZ) number is doubled in endo mutants, a compensatory reduction in their size homeostatically adjusts global neurotransmitter output to maintain synaptic strength. We find an inverse adaptation in rab3 mutants. Additional analyses using confocal, STED, and electron microscopy reveal a stoichiometric tuning of AZ scaffolds and nanoarchitecture. Axonal transport of synaptic cargo via the lysosomal kinesin adapter Arl8 regulates AZ abundance to modulate global synaptic output and sustain the homeostatic potentiation of neurotransmission. Finally, we find that this AZ scaling can interface with two independent homeostats, depression and potentiation, to remodel AZ structure and function, demonstrating a robust balancing of separate homeostatic adaptations. Thus, AZs are pliable substrates with elastic and modular nanostructures that can be dynamically sculpted to stabilize and tune both local and global synaptic strength.
Collapse
Affiliation(s)
- Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, CA
| | | | - Mathias A Böhme
- Neurocure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Luke Nunnelly
- Department of Neurobiology, University of Southern California, Los Angeles, CA
| | - Martin Lehmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | | | - Alexander M Walter
- Neurocure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | | | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA
| |
Collapse
|
47
|
Lübbert M, Goral RO, Keine C, Thomas C, Guerrero-Given D, Putzke T, Satterfield R, Kamasawa N, Young SM. Ca V2.1 α 1 Subunit Expression Regulates Presynaptic Ca V2.1 Abundance and Synaptic Strength at a Central Synapse. Neuron 2018; 101:260-273.e6. [PMID: 30545599 DOI: 10.1016/j.neuron.2018.11.028] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/22/2018] [Accepted: 11/15/2018] [Indexed: 11/28/2022]
Abstract
The abundance of presynaptic CaV2 voltage-gated Ca2+ channels (CaV2) at mammalian active zones (AZs) regulates the efficacy of synaptic transmission. It is proposed that presynaptic CaV2 levels are saturated in AZs due to a finite number of slots that set CaV2 subtype abundance and that CaV2.1 cannot compete for CaV2.2 slots. However, at most AZs, CaV2.1 levels are highest and CaV2.2 levels are developmentally reduced. To investigate CaV2.1 saturation states and preference in AZs, we overexpressed the CaV2.1 and CaV2.2 α1 subunits at the calyx of Held at immature and mature developmental stages. We found that AZs prefer CaV2.1 to CaV2.2. Remarkably, CaV2.1 α1 subunit overexpression drove increased CaV2.1 currents and channel numbers and increased synaptic strength at both developmental stages examined. Therefore, we propose that CaV2.1 levels in the AZ are not saturated and that synaptic strength can be modulated by increasing CaV2.1 levels to regulate neuronal circuit output. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Matthias Lübbert
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - R Oliver Goral
- Department of Anatomy and Cell Biology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Christian Keine
- Department of Anatomy and Cell Biology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Connon Thomas
- Max Planck Florida Electron Microscopy Core, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Debbie Guerrero-Given
- Max Planck Florida Electron Microscopy Core, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Travis Putzke
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Rachel Satterfield
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Naomi Kamasawa
- Max Planck Florida Electron Microscopy Core, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Samuel M Young
- Department of Anatomy and Cell Biology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA; Department of Otolaryngology, Iowa Neuroscience Institute, Aging Mind Brain Initiative, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
48
|
Hagiwara A, Kitahara Y, Grabner CP, Vogl C, Abe M, Kitta R, Ohta K, Nakamura K, Sakimura K, Moser T, Nishi A, Ohtsuka T. Cytomatrix proteins CAST and ELKS regulate retinal photoreceptor development and maintenance. J Cell Biol 2018; 217:3993-4006. [PMID: 30190286 PMCID: PMC6219712 DOI: 10.1083/jcb.201704076] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/18/2018] [Accepted: 08/03/2018] [Indexed: 01/06/2023] Open
Abstract
The retinal ribbon synapse is important for the processing of visual information. Hagiwara et al. show that the active zone proteins CAST and ELKS perform both redundant and unique functions in photoreceptors to promote the maturation, maintenance, and activity of ribbon synapses. At the presynaptic active zone (AZ), the related cytomatrix proteins CAST and ELKS organize the presynaptic release machinery. While CAST is known to regulate AZ size and neurotransmitter release, the role of ELKS and the integral system of CAST/ELKS together is poorly understood. Here, we show that CAST and ELKS have both redundant and unique roles in coordinating synaptic development, function, and maintenance of retinal photoreceptor ribbon synapses. A CAST/ELKS double knockout (dKO) mouse showed high levels of ectopic synapses and reduced responses to visual stimulation. Ectopic formation was not observed in ELKS conditional KO but progressively increased with age in CAST KO mice with higher rates in the dKO. Presynaptic calcium influx was strongly reduced in rod photoreceptors of CAST KO and dKO mice. Three-dimensional scanning EM reconstructions showed structural abnormalities in rod triads of CAST KO and dKO. Remarkably, AAV-mediated acute ELKS deletion after synapse maturation induced neurodegeneration and loss of ribbon synapses. These results suggest that CAST and ELKS work in concert to promote retinal synapse formation, transmission, and maintenance.
Collapse
Affiliation(s)
- Akari Hagiwara
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yosuke Kitahara
- Department of Pharmacology, Kurume University School of Medicine, Fukuoka, Japan
| | - Chad Paul Grabner
- Institute for Auditory Neuroscience and Inner Ear Lab, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Christian Vogl
- Institute for Auditory Neuroscience and Inner Ear Lab, University Medical Center Göttingen, Göttingen, Germany
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Ryo Kitta
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Keisuke Ohta
- Department of Anatomy, Kurume University School of Medicine, Fukuoka, Japan
| | - Keiichiro Nakamura
- Department of Anatomy, Kurume University School of Medicine, Fukuoka, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tobias Moser
- Institute for Auditory Neuroscience and Inner Ear Lab, University Medical Center Göttingen, Göttingen, Germany .,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Akinori Nishi
- Department of Pharmacology, Kurume University School of Medicine, Fukuoka, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|