1
|
Ageta-Ishihara N, Mizukami M, Kinoshita I, Asami Y, Nishioka T, Bito H, Kaibuchi K, Kinoshita M. Phosphorylated septin 3 delocalizes from the spine base and facilitates endoplasmic reticulum extension into spines via myosin-Va. Mol Brain 2025; 18:43. [PMID: 40375097 DOI: 10.1186/s13041-025-01215-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 05/08/2025] [Indexed: 05/18/2025] Open
Abstract
Cytoskeletal remodeling drives morphological changes. Septin cytoskeleton assembles into hetero-oligomers. We previously demonstrated that late-phase long-term potentiation (L-LTP) induces smooth endoplasmic reticulum (sER) extension into dendritic spines via septin 3 (SEPT3), contributing to greater postsynaptic Ca2+ responses and enhanced activation of synaptically induced Ca2+ signaling. Sept3-/- mice exhibit a reduced number of sER-containing spines and show impaired long-term spatial/object memory despite normal short-term memory. Additionally, SEPT3 binds the motor protein myosin-Va (MYO5A) upon elevated Ca²⁺ concentrations, facilitating sER extension from the dendritic shaft into the spine. MYO5A localizes on the sER membrane, while SEPT3 remains at the spine base, accumulating on sER upon electroconvulsive stimulation (ECS). However, the mechanism underlying SEPT3's delocalization from the spine base and its cooperative role with MYO5A in sER extension remains unclear. In this study, we demonstrate that SEPT3 is phosphorylated in a stimulation-dependent manner. Phosphorylation at Thr211 releases SEPT3 from the spine base, enabling sER extension with constitutively active MYO5A mutant (MYO5A-CCtr). These findings provide molecular insight into the role of SEPT3 phosphorylation in regulating sER dynamics that sustain long-term spine activation.
Collapse
Grants
- JPMJPR21E1 Japan Science and Technology Agency
- 20K06852, 21H02579, 21K19427, 22H00432, 22H04922, 22H04926, 23K06394 Japan Society for the Promotion of Science
- 20K06852, 21H02579, 21K19427, 22H00432, 22H04922, 22H04926, 23K06394 Japan Society for the Promotion of Science
- 20K06852, 21H02579, 21K19427, 22H00432, 22H04922, 22H04926, 23K06394 Japan Society for the Promotion of Science
- JP21wm0525011, JP22jm0210098 Japan Agency for Medical Research and Development
- JP21wm0525011, JP22jm0210098 Japan Agency for Medical Research and Development
Collapse
Affiliation(s)
- Natsumi Ageta-Ishihara
- Department of Biomolecular Science, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, 274-8510, Chiba, Japan.
- Department of Molecular Biology, Division of Biological Sciences, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan.
| | - Masato Mizukami
- Department of Molecular Biology, Division of Biological Sciences, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Itsuki Kinoshita
- Department of Molecular Biology, Division of Biological Sciences, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Yurika Asami
- Department of Molecular Biology, Division of Biological Sciences, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Tomoki Nishioka
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, 470-1192, Aichi, Japan
- Open Facility Center, Research Promotion Headquarters, Fujita Health University, Toyoake, 470-1192, Aichi, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kozo Kaibuchi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, 470-1192, Aichi, Japan
| | - Makoto Kinoshita
- Department of Molecular Biology, Division of Biological Sciences, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan.
| |
Collapse
|
2
|
Ageta-Ishihara N, Fukazawa Y, Arima-Yoshida F, Okuno H, Ishii Y, Takao K, Konno K, Fujishima K, Ageta H, Hioki H, Tsuchida K, Sato Y, Kengaku M, Watanabe M, Watabe AM, Manabe T, Miyakawa T, Inokuchi K, Bito H, Kinoshita M. Septin 3 regulates memory and L-LTP-dependent extension of endoplasmic reticulum into spines. Cell Rep 2025; 44:115352. [PMID: 40023151 DOI: 10.1016/j.celrep.2025.115352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/11/2024] [Accepted: 02/05/2025] [Indexed: 03/04/2025] Open
Abstract
Transient memories are converted to persistent memories at the synapse and circuit/systems levels. The synapse-level consolidation parallels electrophysiological transition from early- to late-phase long-term potentiation of synaptic transmission (E-/L-LTP). While glutamate signaling upregulations coupled with dendritic spine enlargement are common underpinnings of E-LTP and L-LTP, synaptic mechanisms conferring persistence on L-LTP remain unclear. Here, we show that L-LTP induced at the perforant path-hippocampal dentate gyrus (DG) synapses accompanies cytoskeletal remodeling that involves actin and the septin subunit SEPT3. L-LTP in DG neurons causes fast spine enlargement, followed by SEPT3-dependent smooth endoplasmic reticulum (sER) extension into enlarged spines. Spines containing sER show greater Ca2+ responses upon synaptic input and local synaptic activity. Consistently, Sept3 knockout in mice (Sept3-/-) impairs memory consolidation and causes a scarcity of sER-containing spines. These findings indicate a concept that sER extension into active spines serves as a synaptic basis of memory consolidation.
Collapse
Affiliation(s)
- Natsumi Ageta-Ishihara
- Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Chiba 274-8510, Japan; Department of Molecular Biology, Division of Biological Sciences, Nagoya University Graduate School of Science, Chikusa-ku, Nagoya 464-8602, Japan.
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Faculty of Medical Science, University of Fukui, Yoshida-gun, Fukui 910-1193, Japan
| | - Fumiko Arima-Yoshida
- Division of Neuronal Network, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan; Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Kashiwa, Chiba 277-8567, Japan
| | - Hiroyuki Okuno
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Yuichiro Ishii
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Keizo Takao
- Department of Behavioral Physiology, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Kohtarou Konno
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Kazuto Fujishima
- Institute for Integrated Cell-Material Sciences, Kyoto University Institute for Advanced Study (KUIAS-iCeMS), Sakyo-ku, Kyoto 606-8501, Japan; Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569-8686, Japan
| | - Hiroshi Ageta
- Division for Therapies Against Intractable Diseases, Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Hiroyuki Hioki
- Department of Neuroanatomy, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Kunihiro Tsuchida
- Division for Therapies Against Intractable Diseases, Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Yoshikatsu Sato
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Chikusa-ku, Nagoya 464-8601, Japan
| | - Mineko Kengaku
- Institute for Integrated Cell-Material Sciences, Kyoto University Institute for Advanced Study (KUIAS-iCeMS), Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Ayako M Watabe
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Kashiwa, Chiba 277-8567, Japan
| | - Toshiya Manabe
- Division of Neuronal Network, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Kaoru Inokuchi
- Department of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Makoto Kinoshita
- Department of Molecular Biology, Division of Biological Sciences, Nagoya University Graduate School of Science, Chikusa-ku, Nagoya 464-8602, Japan.
| |
Collapse
|
3
|
Hasan S, Guo Y, Sandroni P, Dubey D, McKeon A. Anhidrosis in septin-7 autoimmunity. Clin Auton Res 2025:10.1007/s10286-025-01108-w. [PMID: 39815061 DOI: 10.1007/s10286-025-01108-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025]
Affiliation(s)
- Shemonti Hasan
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Yong Guo
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Paola Sandroni
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Divyanshu Dubey
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Andrew McKeon
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
4
|
Curtis BN, Gladfelter AS. Drivers of Morphogenesis: Curvature Sensor Self-Assembly at the Membrane. Cold Spring Harb Perspect Biol 2024; 16:a041528. [PMID: 38697653 PMCID: PMC11610757 DOI: 10.1101/cshperspect.a041528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
This review examines the relationships between membrane chemistry, curvature-sensing proteins, and cellular morphogenesis. Curvature-sensing proteins are often orders of magnitude smaller than the membrane curvatures they localize to. How are nanometer-scale proteins used to sense micrometer-scale membrane features? Here, we trace the journey of curvature-sensing proteins as they engage with lipid membranes through a combination of electrostatic and hydrophobic interactions. We discuss how curvature sensing hinges on membrane features like lipid charge, packing, and the directionality of membrane curvature. Once bound to the membrane, many curvature sensors undergo self-assembly (i.e., they oligomerize or form higher-order assemblies that are key for initiating and regulating cell shape transformations). Central to these discussions are the micrometer-scale curvature-sensing proteins' septins. By discussing recent literature surrounding septin membrane association, assembly, and their many functions in morphogenesis with support from other well-studied curvature sensors, we aim to synthesize possible mechanisms underlining cell shape sensing.
Collapse
Affiliation(s)
- Brandy N Curtis
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
- Department of Cell Biology, Duke University, Durham, North Carolina 27708, USA
| | - Amy S Gladfelter
- Department of Cell Biology, Duke University, Durham, North Carolina 27708, USA
| |
Collapse
|
5
|
Verma H, Kaur S, Kaur S, Gangwar P, Dhiman M, Mantha AK. Role of Cytoskeletal Elements in Regulation of Synaptic Functions: Implications Toward Alzheimer's Disease and Phytochemicals-Based Interventions. Mol Neurobiol 2024; 61:8320-8343. [PMID: 38491338 DOI: 10.1007/s12035-024-04053-3] [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: 07/03/2023] [Accepted: 02/13/2024] [Indexed: 03/18/2024]
Abstract
Alzheimer's disease (AD), a multifactorial disease, is characterized by the accumulation of neurofibrillary tangles (NFTs) and amyloid beta (Aβ) plaques. AD is triggered via several factors like alteration in cytoskeletal proteins, a mutation in presenilin 1 (PSEN1), presenilin 2 (PSEN2), amyloid precursor protein (APP), and post-translational modifications (PTMs) in the cytoskeletal elements. Owing to the major structural and functional role of cytoskeletal elements, like the organization of axon initial segmentation, dendritic spines, synaptic regulation, and delivery of cargo at the synapse; modulation of these elements plays an important role in AD pathogenesis; like Tau is a microtubule-associated protein that stabilizes the microtubules, and it also causes inhibition of nucleo-cytoplasmic transportation by disrupting the integrity of nuclear pore complex. One of the major cytoskeletal elements, actin and its dynamics, regulate the dendritic spine structure and functions; impairments have been documented towards learning and memory defects. The second major constituent of these cytoskeletal elements, microtubules, are necessary for the delivery of the cargo, like ion channels and receptors at the synaptic membranes, whereas actin-binding protein, i.e., Cofilin's activation form rod-like structures, is involved in the formation of paired helical filaments (PHFs) observed in AD. Also, the glial cells rely on their cytoskeleton to maintain synaptic functionality. Thus, making cytoskeletal elements and their regulation in synaptic structure and function as an important aspect to be focused for better management and targeting AD pathology. This review advocates exploring phytochemicals and Ayurvedic plant extracts against AD by elucidating their neuroprotective mechanisms involving cytoskeletal modulation and enhancing synaptic plasticity. However, challenges include their limited bioavailability due to the poor solubility and the limited potential to cross the blood-brain barrier (BBB), emphasizing the need for targeted strategies to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Harkomal Verma
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, VPO - Ghudda, Bathinda, 151 401, Punjab, India
| | - Sharanjot Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, India
| | - Sukhchain Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, India
| | - Prabhakar Gangwar
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, VPO - Ghudda, Bathinda, 151 401, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, India
| | - Anil Kumar Mantha
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, VPO - Ghudda, Bathinda, 151 401, Punjab, India.
| |
Collapse
|
6
|
Varela Salgado M, Piatti S. Septin Organization and Dynamics for Budding Yeast Cytokinesis. J Fungi (Basel) 2024; 10:642. [PMID: 39330402 PMCID: PMC11433133 DOI: 10.3390/jof10090642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/28/2024] Open
Abstract
Cytokinesis, the process by which the cytoplasm divides to generate two daughter cells after mitosis, is a crucial stage of the cell cycle. Successful cytokinesis must be coordinated with chromosome segregation and requires the fine orchestration of several processes, such as constriction of the actomyosin ring, membrane reorganization, and, in fungi, cell wall deposition. In Saccharomyces cerevisiae, commonly known as budding yeast, septins play a pivotal role in the control of cytokinesis by assisting the assembly of the cytokinetic machinery at the division site and controlling its activity. Yeast septins form a collar at the division site that undergoes major dynamic transitions during the cell cycle. This review discusses the functions of septins in yeast cytokinesis, their regulation and the implications of their dynamic remodelling for cell division.
Collapse
Affiliation(s)
- Maritzaida Varela Salgado
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293 Montpellier, France
| | - Simonetta Piatti
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293 Montpellier, France
| |
Collapse
|
7
|
Mendonça DC, Morais STB, Ciol H, Pinto APA, Leonardo DA, Pereira HD, Valadares NF, Portugal RV, Klaholz BP, Garratt RC, Araujo APU. Structural Insights into Ciona intestinalis Septins: Complexes Suggest a Mechanism for Nucleotide-dependent Interfacial Cross-talk. J Mol Biol 2024; 436:168693. [PMID: 38960133 DOI: 10.1016/j.jmb.2024.168693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024]
Abstract
Septins are filamentous nucleotide-binding proteins which can associate with membranes in a curvature-dependent manner leading to structural remodelling and barrier formation. Ciona intestinalis, a model for exploring the development and evolution of the chordate lineage, has only four septin-coding genes within its genome. These represent orthologues of the four classical mammalian subgroups, making it a minimalist non-redundant model for studying the modular assembly of septins into linear oligomers and thereby filamentous polymers. Here, we show that C. intestinalis septins present a similar biochemistry to their human orthologues and also provide the cryo-EM structures of an octamer, a hexamer and a tetrameric sub-complex. The octamer, which has the canonical arrangement (2-6-7-9-9-7-6-2) clearly shows an exposed NC-interface at its termini enabling copolymerization with hexamers into mixed filaments. Indeed, only combinations of septins which had CiSEPT2 occupying the terminal position were able to assemble into filaments via NC-interface association. The CiSEPT7-CiSEPT9 tetramer is the smallest septin particle to be solved by Cryo-EM to date and its good resolution (2.7 Å) provides a well-defined view of the central NC-interface. On the other hand, the CiSEPT7-CiSEPT9 G-interface shows signs of fragility permitting toggling between hexamers and octamers, similar to that seen in human septins but not in yeast. The new structures provide insights concerning the molecular mechanism for cross-talk between adjacent interfaces. This indicates that C. intestinalis may represent a valuable tool for future studies, fulfilling the requirements of a complete but simpler system to understand the mechanisms behind the assembly and dynamics of septin filaments.
Collapse
Affiliation(s)
| | | | - Heloísa Ciol
- São Carlos Institute of Physics, USP, São Carlos, SP, Brazil
| | | | | | | | | | - Rodrigo V Portugal
- Brazilian Nanotechnology National Laboratory, CNPEM, Campinas, SP, Brazil; Biotechnosciency Program, Federal University of ABC, Santo André, SP, Brazil
| | - Bruno P Klaholz
- Centre for Integrative Biology (CBI), Department of Integrated Structural Biology, IGBMC (Institute of Genetics and of Molecular and Cellular Biology), 67404 Illkirch, France; Centre National de la Recherche Scientifique (CNRS) UMR 7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U964, 67404 Illkirch, France; Université de Strasbourg, 67081 Strasbourg, France
| | | | - Ana P U Araujo
- São Carlos Institute of Physics, USP, São Carlos, SP, Brazil.
| |
Collapse
|
8
|
Scott-Hewitt N, Mahoney M, Huang Y, Korte N, Yvanka de Soysa T, Wilton DK, Knorr E, Mastro K, Chang A, Zhang A, Melville D, Schenone M, Hartigan C, Stevens B. Microglial-derived C1q integrates into neuronal ribonucleoprotein complexes and impacts protein homeostasis in the aging brain. Cell 2024; 187:4193-4212.e24. [PMID: 38942014 DOI: 10.1016/j.cell.2024.05.058] [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: 05/31/2023] [Revised: 01/08/2024] [Accepted: 05/31/2024] [Indexed: 06/30/2024]
Abstract
Neuroimmune interactions mediate intercellular communication and underlie critical brain functions. Microglia, CNS-resident macrophages, modulate the brain through direct physical interactions and the secretion of molecules. One such secreted factor, the complement protein C1q, contributes to complement-mediated synapse elimination in both developmental and disease models, yet brain C1q protein levels increase significantly throughout aging. Here, we report that C1q interacts with neuronal ribonucleoprotein (RNP) complexes in an age-dependent manner. Purified C1q protein undergoes RNA-dependent liquid-liquid phase separation (LLPS) in vitro, and the interaction of C1q with neuronal RNP complexes in vivo is dependent on RNA and endocytosis. Mice lacking C1q have age-specific alterations in neuronal protein synthesis in vivo and impaired fear memory extinction. Together, our findings reveal a biophysical property of C1q that underlies RNA- and age-dependent neuronal interactions and demonstrate a role of C1q in critical intracellular neuronal processes.
Collapse
Affiliation(s)
- Nicole Scott-Hewitt
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Matthew Mahoney
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Youtong Huang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nils Korte
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - T Yvanka de Soysa
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniel K Wilton
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Emily Knorr
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kevin Mastro
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Allison Chang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Allison Zhang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - David Melville
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Monica Schenone
- The Broad Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Christina Hartigan
- The Broad Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Investigator, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Ayanoğlu M, Çevik Ö, Erdoğan Ö, Tosun AF. TARC and Septin 7 can be better monitoring biomarkers than CX3CL1, sICAM5, and IRF5 in children with seizure-free epilepsy with monotherapy and drug-resistant epilepsy. Int J Neurosci 2024; 134:243-252. [PMID: 35822432 DOI: 10.1080/00207454.2022.2100773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 06/04/2022] [Accepted: 06/23/2022] [Indexed: 10/17/2022]
Abstract
Aim: To evaluate i) the relationship between epilepsy and inflammation by analyzing the levels of thymus activation-regulated chemokine (TARC), and interferon regulatory factor 5 (IRF5) in healthy controls, patients with epilepsy on monotherapy and polytherapy, ii) the levels of sICAM5, chemokine (c-x3-c motif) ligand 1 (CX3CL1), and septin 7 (SEPT7) which are important in both inflammation and synaptic formation. Methods: Patients who were seizure-free with monotherapy (epilepsy group-1), patients with drug-resistant epilepsy (epilepsy group-2), and healthy controls were included. Demographical data, disease durations, and medications were noted. Measurements were made by commercial ELISA kits. Results: The numbers of epilepsy group-1, epilepsy group-2, and healthy controls were 23, 20, and 21, respectively. TARC levels were significantly lower in healthy controls than in both epilepsy groups. Higher TARC levels than 0.58 pg/ml indicated epilepsy with a sensitivity of 81.8% and specificity of 84.0%. SEPT7 levels were significantly higher in epilepsy group-1 than in those epilepsy group-2. A negative correlation was found between SEPT7 levels and disease duration as is the case for the correlation between SEPT7 and average seizure duration. A positive correlation was found between IRF5 and CX3CL1 levels, SEPT7 and IRF5 levels, and IRF5 and sICAM5 levels. Conclusions: We suggest that TARC is a promising biomarker, even in a heterogeneous epilepsy group not only for drug-resistance epilepsy but also for seizure-free epilepsy with monotherapy. Additionally, drug resistance, longer disease, and longer seizure durations are related to lower levels of SEPT7, which has an essential role in immunological functions and dendritic morphology.
Collapse
Affiliation(s)
- Müge Ayanoğlu
- Department of Pediatric Neurology, Adnan Menderes University School of Medicine, Aydın, Turkey
| | - Özge Çevik
- Department of Biochemistry, Adnan Menderes University School of Medicine, Aydın, Turkey
| | - Ömer Erdoğan
- Department of Biochemistry, Adnan Menderes University School of Medicine, Aydın, Turkey
| | - Ayşe Fahriye Tosun
- Department of Pediatric Neurology, Adnan Menderes University School of Medicine, Aydın, Turkey
| |
Collapse
|
10
|
Zhang Y, Lin C. Lipid osmosis, membrane tension, and other mechanochemical driving forces of lipid flow. Curr Opin Cell Biol 2024; 88:102377. [PMID: 38823338 PMCID: PMC11193448 DOI: 10.1016/j.ceb.2024.102377] [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: 01/10/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 06/03/2024]
Abstract
Nonvesicular lipid transport among different membranes or membrane domains plays crucial roles in lipid homeostasis and organelle biogenesis. However, the forces that drive such lipid transport are not well understood. We propose that lipids tend to flow towards the membrane area with a higher membrane protein density in a process termed lipid osmosis. This process lowers the membrane tension in the area, resulting in a membrane tension difference called osmotic membrane tension. We examine the thermodynamic basis and experimental evidence of lipid osmosis and osmotic membrane tension. We predict that lipid osmosis can drive bulk lipid flows between different membrane regions through lipid transfer proteins, scramblases, or similar barriers that selectively pass lipids but not membrane proteins. We also speculate on the biological functions of lipid osmosis. Finally, we explore other driving forces for lipid transfer and describe potential methods and systems to further test our theory.
Collapse
Affiliation(s)
- Yongli Zhang
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA.
| | - Chenxiang Lin
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA; Nanobiology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
11
|
Zhang Y, Lin C. Lipid osmosis, membrane tension, and other mechanochemical driving forces of lipid flow. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574656. [PMID: 38260424 PMCID: PMC10802412 DOI: 10.1101/2024.01.08.574656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Nonvesicular lipid transport among different membranes or membrane domains plays crucial roles in lipid homeostasis and organelle biogenesis. However, the forces that drive such lipid transport are not well understood. We propose that lipids tend to flow towards the membrane area with a higher membrane protein density in a process termed lipid osmosis. This process lowers the membrane tension in the area, resulting in a membrane tension difference called osmotic membrane tension. We examine the thermodynamic basis and experimental evidence of lipid osmosis and osmotic membrane tension. We predict that lipid osmosis can drive bulk lipid flows between different membrane regions through lipid transfer proteins, scramblases, or other similar barriers that selectively pass lipids but not membrane proteins. We also speculate on the biological functions of lipid osmosis. Finally, we explore other driving forces for lipid transfer and describe potential methods and systems to further test our theory.
Collapse
Affiliation(s)
- Yongli Zhang
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Chenxiang Lin
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Nanobiology Institute, Yale University, West Haven, CT 06516, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
12
|
Varela Salgado M, Adriaans IE, Touati SA, Ibanes S, Lai-Kee-Him J, Ancelin A, Cipelletti L, Picas L, Piatti S. Phosphorylation of the F-BAR protein Hof1 drives septin ring splitting in budding yeast. Nat Commun 2024; 15:3383. [PMID: 38649354 PMCID: PMC11035697 DOI: 10.1038/s41467-024-47709-3] [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: 07/12/2023] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
A double septin ring accompanies cytokinesis in yeasts and mammalian cells. In budding yeast, reorganisation of the septin collar at the bud neck into a dynamic double ring is essential for actomyosin ring constriction and cytokinesis. Septin reorganisation requires the Mitotic Exit Network (MEN), a kinase cascade essential for cytokinesis. However, the effectors of MEN in this process are unknown. Here we identify the F-BAR protein Hof1 as a critical target of MEN in septin remodelling. Phospho-mimicking HOF1 mutant alleles overcome the inability of MEN mutants to undergo septin reorganisation by decreasing Hof1 binding to septins and facilitating its translocation to the actomyosin ring. Hof1-mediated septin rearrangement requires its F-BAR domain, suggesting that it may involve a local membrane remodelling that leads to septin reorganisation. In vitro Hof1 can induce the formation of intertwined septin bundles, while a phosphomimetic Hof1 protein has impaired septin-bundling activity. Altogether, our data indicate that Hof1 modulates septin architecture in distinct ways depending on its phosphorylation status.
Collapse
Affiliation(s)
- Maritzaida Varela Salgado
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293, Montpellier, France
| | - Ingrid E Adriaans
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293, Montpellier, France
| | - Sandra A Touati
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013, Paris, France
| | - Sandy Ibanes
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293, Montpellier, France
| | - Joséphine Lai-Kee-Him
- CBS (Centre de Biologie Structurale), University of Montpellier, CNRS UMR 5048, INSERM U 1054, 34090, Montpellier, France
| | - Aurélie Ancelin
- CBS (Centre de Biologie Structurale), University of Montpellier, CNRS UMR 5048, INSERM U 1054, 34090, Montpellier, France
| | - Luca Cipelletti
- L2C (Laboratoire Charles Coulomb), University of Montpellier, CNRS 34095, Montpellier, France
- IUF (Institut Universitaire de France, 75231, Paris, France
| | - Laura Picas
- IRIM (Institut de Recherche en Infectiologie de Montpellier), University of Montpellier, CNRS UMR 9004, 34293, Montpellier, France
| | - Simonetta Piatti
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293, Montpellier, France.
| |
Collapse
|
13
|
Nakazawa K, Chauvin B, Mangenot S, Bertin A. Reconstituted in vitro systems to reveal the roles and functions of septins. J Cell Sci 2023; 136:jcs259448. [PMID: 37815088 DOI: 10.1242/jcs.259448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023] Open
Abstract
Septins are essential cytoskeletal proteins involved in key cellular processes and have also been implicated in diseases from cancers to neurodegenerative pathologies. However, they have not been as thoroughly studied as other cytoskeletal proteins. In vivo, septins interact with other cytoskeletal proteins and with the inner plasma membrane. Hence, bottom-up in vitro cell-free assays are well suited to dissect the roles and behavior of septins in a controlled environment. Specifically, in vitro studies have been invaluable in describing the self-assembly of septins into a large diversity of ultrastructures. Given that septins interact specifically with membrane, the details of these septin-membrane interactions have been analyzed using reconstituted lipid systems. In particular, at a membrane, septins are often localized at curvatures of micrometer scale. In that context, in vitro assays have been performed with substrates of varying curvatures (spheres, cylinders or undulated substrates) to probe the sensitivity of septins to membrane curvature. This Review will first present the structural properties of septins in solution and describe the interplay of septins with cytoskeletal partners. We will then discuss how septins interact with biomimetic membranes and induce their reshaping. Finally, we will highlight the curvature sensitivity of septins and how they alter the mechanical properties of membranes.
Collapse
Affiliation(s)
- Koyomi Nakazawa
- Physico Chimie Curie , Institut Curie, CNRS UMR 168, Sorbonne Université, 11 Rue Pierre et Paris Curie, 75005 Paris, France
| | - Brieuc Chauvin
- Physico Chimie Curie , Institut Curie, CNRS UMR 168, Sorbonne Université, 11 Rue Pierre et Paris Curie, 75005 Paris, France
| | - Stéphanie Mangenot
- Laboratoire Matière et Systèmes Complexes , Université de Paris Cité, CNRS UMR 7057, 45 Rue des Saint Pères, 75006 Paris, France
| | - Aurélie Bertin
- Physico Chimie Curie , Institut Curie, CNRS UMR 168, Sorbonne Université, 11 Rue Pierre et Paris Curie, 75005 Paris, France
| |
Collapse
|
14
|
Sharma K, Menon MB. Decoding post-translational modifications of mammalian septins. Cytoskeleton (Hoboken) 2023; 80:169-181. [PMID: 36797225 DOI: 10.1002/cm.21747] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/21/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023]
Abstract
Septins are cytoskeletal GTPases that form nonpolar filaments and higher-ordered structures and they take part in a wide range of cellular processes. Septins are conserved from yeast to mammals but absent from higher plants. The number of septin genes vary between organisms and they usually form complex heteropolymeric networks. Most septins are known to be capable of GTP hydrolysis which may regulate septin dynamics. Knowledge on regulation of septin function by post-translational modifications is still in its infancy. In this review article, we highlight the post-translational modifications reported for the 13 human septins and discuss their implications on septin functions. In addition to the functionally investigated modifications, we also try to make sense of the complex septin post-translational modification code revealed from large-scale phospho-proteomic datasets. Future studies may determine how these isoform-specific and homology group specific modifications affect septin structure and function.
Collapse
Affiliation(s)
- Khushboo Sharma
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Manoj B Menon
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
15
|
Tomasso MR, Padrick SB. BORG family proteins in physiology and human disease. Cytoskeleton (Hoboken) 2023; 80:182-198. [PMID: 37403807 DOI: 10.1002/cm.21768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/06/2023]
Abstract
The binder of rho GTPases (BORG)/Cdc42 effector proteins (Cdc42EP) family is composed of five Rho GTPase binding proteins whose functions and mechanism of actions are of emerging interest. Here, we review recent findings pertaining to the family as a whole and consider how these change our understanding of cellular organization. Recent studies have implicated BORGs in both fundamental physiology and in human diseases, mainly cancers. An emerging pattern suggests that BORG family members cancer-promoting properties are related to their ability to regulate the cytoskeleton, with many impacting the organization of acto-myosin stress fibers. This is consistent with the broader literature indicating that BORG family members are regulators of both the septin and actin cytoskeleton networks. The exact mechanism through which BORGs modify the cytoskeleton is not clear, but we consider here a few data-supported and speculative possibilities. Finally, we delve into how the Rho GTPase Cdc42 modifies BORG function in cells. This remains open-ended as Cdc42's effects on BORGs appear cell type- and cell state-dependent. Collectively, these data point to the importance of the BORG family and suggest broader themes in their function and regulation.
Collapse
Affiliation(s)
- Meagan R Tomasso
- Department of Biochemistry and Molecular Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Shae B Padrick
- Department of Biochemistry and Molecular Biology, Drexel University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
16
|
Minegishi T, Kastian RF, Inagaki N. Mechanical regulation of synapse formation and plasticity. Semin Cell Dev Biol 2023; 140:82-89. [PMID: 35659473 DOI: 10.1016/j.semcdb.2022.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 01/28/2023]
Abstract
Dendritic spines are small protrusions arising from dendrites and constitute the major compartment of excitatory post-synapses. They change in number, shape, and size throughout life; these changes are thought to be associated with formation and reorganization of neuronal networks underlying learning and memory. As spines in the brain are surrounded by the microenvironment including neighboring cells and the extracellular matrix, their protrusion requires generation of force to push against these structures. In turn, neighboring cells receive force from protruding spines. Recent studies have identified BAR-domain proteins as being involved in membrane deformation to initiate spine formation. In addition, forces for dendritic filopodium extension and activity-induced spine expansion are generated through cooperation between actin polymerization and clutch coupling. On the other hand, force from expanding spines affects neurotransmitter release from presynaptic terminals. Here, we review recent advances in our understanding of the physical aspects of synapse formation and plasticity, mainly focusing on spine dynamics.
Collapse
Affiliation(s)
- Takunori Minegishi
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Ria Fajarwati Kastian
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan; Research Center for Genetic Engineering, National Research and Innovation Agency Republic of Indonesia, Cibinong, Bogor, Indonesia
| | - Naoyuki Inagaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan.
| |
Collapse
|
17
|
Martins CS, Taveneau C, Castro-Linares G, Baibakov M, Buzhinsky N, Eroles M, Milanović V, Omi S, Pedelacq JD, Iv F, Bouillard L, Llewellyn A, Gomes M, Belhabib M, Kuzmić M, Verdier-Pinard P, Lee S, Badache A, Kumar S, Chandre C, Brasselet S, Rico F, Rossier O, Koenderink GH, Wenger J, Cabantous S, Mavrakis M. Human septins organize as octamer-based filaments and mediate actin-membrane anchoring in cells. J Cell Biol 2023; 222:213778. [PMID: 36562751 PMCID: PMC9802686 DOI: 10.1083/jcb.202203016] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 10/20/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Septins are cytoskeletal proteins conserved from algae and protists to mammals. A unique feature of septins is their presence as heteromeric complexes that polymerize into filaments in solution and on lipid membranes. Although animal septins associate extensively with actin-based structures in cells, whether septins organize as filaments in cells and if septin organization impacts septin function is not known. Customizing a tripartite split-GFP complementation assay, we show that all septins decorating actin stress fibers are octamer-containing filaments. Depleting octamers or preventing septins from polymerizing leads to a loss of stress fibers and reduced cell stiffness. Super-resolution microscopy revealed septin fibers with widths compatible with their organization as paired septin filaments. Nanometer-resolved distance measurements and single-protein tracking further showed that septin filaments are membrane bound and largely immobilized. Finally, reconstitution assays showed that septin filaments mediate actin-membrane anchoring. We propose that septin organization as octamer-based filaments is essential for septin function in anchoring and stabilizing actin filaments at the plasma membrane.
Collapse
Affiliation(s)
- Carla Silva Martins
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, Marseille, France.,Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM, Université de Toulouse, UPS, CNRS, Toulouse, France
| | - Cyntia Taveneau
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
| | - Gerard Castro-Linares
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, The Netherlands
| | - Mikhail Baibakov
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, Marseille, France
| | - Nicolas Buzhinsky
- CNRS, INSERM, LAI, Turing Centre for Living Systems, Aix-Marseille Univ, Marseille, France>
| | - Mar Eroles
- CNRS, INSERM, LAI, Turing Centre for Living Systems, Aix-Marseille Univ, Marseille, France>
| | - Violeta Milanović
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR, Bordeaux, France
| | - Shizue Omi
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, Marseille, France
| | - Jean-Denis Pedelacq
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Francois Iv
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, Marseille, France
| | - Léa Bouillard
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, Marseille, France
| | - Alexander Llewellyn
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, Marseille, France
| | - Maxime Gomes
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, Marseille, France
| | - Mayssa Belhabib
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, Marseille, France
| | - Mira Kuzmić
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, Marseille, France
| | - Pascal Verdier-Pinard
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, Marseille, France
| | - Stacey Lee
- Department of Bioengineering, University of California, Berkeley, CA, USA
| | - Ali Badache
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, Marseille, France
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, CA, USA
| | | | - Sophie Brasselet
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, Marseille, France
| | - Felix Rico
- CNRS, INSERM, LAI, Turing Centre for Living Systems, Aix-Marseille Univ, Marseille, France>
| | - Olivier Rossier
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR, Bordeaux, France
| | - Gijsje H Koenderink
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, The Netherlands
| | - Jerome Wenger
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, Marseille, France
| | - Stéphanie Cabantous
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM, Université de Toulouse, UPS, CNRS, Toulouse, France
| | - Manos Mavrakis
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, Marseille, France
| |
Collapse
|
18
|
Shi W, Cannon KS, Curtis BN, Edelmaier C, Gladfelter AS, Nazockdast E. Curvature sensing as an emergent property of multiscale assembly of septins. Proc Natl Acad Sci U S A 2023; 120:e2208253120. [PMID: 36716363 PMCID: PMC9963131 DOI: 10.1073/pnas.2208253120] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 12/15/2022] [Indexed: 02/01/2023] Open
Abstract
The ability of cells to sense and communicate their shape is central to many of their functions. Much is known about how cells generate complex shapes, yet how they sense and respond to geometric cues remains poorly understood. Septins are GTP-binding proteins that localize to sites of micrometer-scale membrane curvature. Assembly of septins is a multistep and multiscale process, but it is unknown how these discrete steps lead to curvature sensing. Here, we experimentally examine the time-dependent binding of septins at different curvatures and septin bulk concentrations. These experiments unexpectedly indicated that septins' curvature preference is not absolute but rather is sensitive to the combinations of membrane curvatures present in a reaction, suggesting that there is competition between different curvatures for septin binding. To understand the physical underpinning of this result, we developed a kinetic model that connects septins' self-assembly and curvature-sensing properties. Our experimental and modeling results are consistent with curvature-sensitive assembly being driven by cooperative associations of septin oligomers in solution with the bound septins. When combined, the work indicates that septin curvature sensing is an emergent property of the multistep, multiscale assembly of membrane-bound septins. As a result, curvature preference is not absolute and can be modulated by changing the physicochemical and geometric parameters involved in septin assembly, including bulk concentration, and the available membrane curvatures. While much geometry-sensitive assembly in biology is thought to be guided by intrinsic material properties of molecules, this is an important example of how curvature sensing can arise from multiscale assembly of polymers.
Collapse
Affiliation(s)
- Wenzheng Shi
- Department of Applied Physical Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Kevin S. Cannon
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Brandy N. Curtis
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Christopher Edelmaier
- Department of Applied Physical Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Amy S. Gladfelter
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Marine Biology Laboratory, Woods Hole, MA02543
| | - Ehssan Nazockdast
- Department of Applied Physical Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| |
Collapse
|
19
|
Ho CT, Gupton SL. Cytoskeleton: Septin wreaths regulate actin in neuritogenesis. Curr Biol 2023; 33:R98-R100. [PMID: 36750031 DOI: 10.1016/j.cub.2022.12.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The shape of a neuron changes dramatically during development. New work reports a novel septin cytoskeleton network that is important in establishing proper neuronal morphology.
Collapse
Affiliation(s)
- Chris T Ho
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Medical Biomolecular Research Building 4332, Campus Box 7090, Chapel Hill, NC 27599-7545, USA
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Medical Biomolecular Research Building 4332, Campus Box 7090, Chapel Hill, NC 27599-7545, USA; UNC Neuroscience Center, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA.
| |
Collapse
|
20
|
Radler MR, Liu X, Peng M, Doyle B, Toyo-Oka K, Spiliotis ET. Pyramidal neuron morphogenesis requires a septin network that stabilizes filopodia and suppresses lamellipodia during neurite initiation. Curr Biol 2023; 33:434-448.e8. [PMID: 36538929 PMCID: PMC9905282 DOI: 10.1016/j.cub.2022.11.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/31/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022]
Abstract
Pyramidal neurons are a major cell type of the forebrain, consisting of a pyramidally shaped soma with axonal and apicobasal dendritic processes. It is poorly understood how the neuronal soma develops its pyramidal morphology, while generating neurites of the proper shape and orientation. Here, we discovered that the spherical somata of immature neurite-less neurons possess a circumferential wreath-like network of septin filaments, which promotes neuritogenesis by balancing the protrusive activity of lamellipodia and filopodia. In embryonic rat hippocampal and mouse cortical neurons, the septin wreath network consists of curvilinear filaments that contain septins 5, 7, and 11 (Sept5/7/11). The Sept5/7/11 wreath network demarcates a zone of myosin II enrichment and Arp2/3 diminution at the base of filopodial actin bundles. In Sept7-depleted neurons, cell bodies are enlarged with hyperextended lamellae and abnormally shaped neurites that originate from lamellipodia. This phenotype is accompanied by diminished myosin II and filopodia lifetimes and increased Arp2/3 and lamellipodial activity. Inhibition of Arp2/3 rescues soma and neurite phenotypes, indicating that the septin wreath network suppresses the extension of lamellipodia, facilitating the formation of neurites from the filopodia of a consolidated soma. We show that this septin function is critical for developing a pyramidally shaped soma with properly distributed and oriented dendrites in cultured rat hippocampal neurons and in vivo in mouse perinatal cortical neurons. Therefore, the somatic septin cytoskeleton provides a key morphogenetic mechanism for neuritogenesis and the development of pyramidal neurons.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Xiaonan Liu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Megan Peng
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Brenna Doyle
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
21
|
Hinson SR, Honorat JA, Grund EM, Clarkson BD, Miske R, Scharf M, Zivelonghi C, Al-Lozi MT, Bucelli RC, Budhram A, Cho T, Choi E, Grell J, Lopez-Chiriboga AS, Levin M, Merati M, Montalvo M, Pittock SJ, Wilson MR, Howe CL, McKeon A. Septin-5 and -7-IgGs: Neurologic, Serologic, and Pathophysiologic Characteristics. Ann Neurol 2022; 92:1090-1101. [PMID: 36053822 PMCID: PMC9672904 DOI: 10.1002/ana.26482] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND AND OBJECTIVES We sought to determine clinical significance of neuronal septin autoimmunity and evaluate for potential IgG effects. METHODS Septin-IgGs were detected by indirect immunofluorescence assays (IFAs; mouse tissue and cell based) or Western blot. IgG binding to (and internalization of) extracellular septin epitopes were evaluated for by live rat hippocampal neuron assay. The impact of purified patient IgGs on murine cortical neuron function was determined by recording extracellular field potentials in a multielectrode array platform. RESULTS Septin-IgGs were identified in 23 patients. All 8 patients with septin-5-IgG detected had cerebellar ataxia, and 7 had prominent eye movement disorders. One of 2 patients with co-existing septin-7-IgG had additional psychiatric phenotype (apathy, emotional blunting, and poor insight). Fifteen patients had septin-7 autoimmunity, without septin-5-IgG detected. Disorders included encephalopathy (11; 2 patients with accompanying myelopathy, and 2 were relapsing), myelopathy (3), and episodic ataxia (1). Psychiatric symptoms (≥1 of agitation, apathy, catatonia, disorganized thinking, and paranoia) were prominent in 6 of 11 patients with encephalopathic symptoms. Eight of 10 patients with data available (from 23 total) improved after immunotherapy, and a further 2 patients improved spontaneously. Staining of plasma membranes of live hippocampal neurons produced by patient IgGs (subclasses 1 and 2) colocalized with pre- and post-synaptic markers. Decreased spiking and bursting behavior in mixed cultures of murine glutamatergic and GABAergic cortical neurons produced by patient IgGs were attributable to neither antigenic crosslinking and internalization nor complement activation. INTERPRETATION Septin-IgGs are predictive of distinct treatment-responsive autoimmune central nervous system (CNS) disorders. Live neuron binding and induced electrophysiologic effects by patient IgGs may support septin-specific pathophysiology. ANN NEUROL 2022;92:1090-1101.
Collapse
Affiliation(s)
- Shannon R. Hinson
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
| | - Josephe A. Honorat
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
| | - Ethan M. Grund
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
| | | | - Ramona Miske
- Institute for Experimental Immunology, affiliated to
EUROIMMUN Medizinische Labordiagnostika, Lubeck, Germany
| | - Madeleine Scharf
- Institute for Experimental Immunology, affiliated to
EUROIMMUN Medizinische Labordiagnostika, Lubeck, Germany
| | - Cecilia Zivelonghi
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
| | | | | | - Adrian Budhram
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
| | - Tracey Cho
- Department of Neurology, University of Iowa, Iowa,
USA
| | - Ellie Choi
- Overlake Hospital, Bellevue, Washington, USA
| | - Jacquelyn Grell
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
| | | | - Marc Levin
- Department of Ophthalmology, Palo Alto Medical Foundation,
Palo Alto, CA, USA
| | - Melody Merati
- Department of Neurology, Michigan State University,
Lansing, MI, USA
| | - Mayra Montalvo
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
| | - Sean J. Pittock
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
| | - Michael R. Wilson
- Weill Institute for Neurosciences, Department of
Neurology, University of California, San Francisco, USA
| | | | - Andrew McKeon
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
| |
Collapse
|
22
|
Gönczi M, Ráduly Z, Szabó L, Fodor J, Telek A, Dobrosi N, Balogh N, Szentesi P, Kis G, Antal M, Trencsenyi G, Dienes B, Csernoch L. Septin7 is indispensable for proper skeletal muscle architecture and function. eLife 2022; 11:e75863. [PMID: 35929607 PMCID: PMC9355566 DOI: 10.7554/elife.75863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 07/03/2022] [Indexed: 11/13/2022] Open
Abstract
Today septins are considered as the fourth component of the cytoskeleton, with the Septin7 isoform playing a critical role in the formation of higher-order structures. While its importance has already been confirmed in several intracellular processes of different organs, very little is known about its role in skeletal muscle. Here, using Septin7 conditional knockdown (KD) mouse model, the C2C12 cell line, and enzymatically isolated adult muscle fibers, the organization and localization of septin filaments are revealed, and an ontogenesis-dependent expression of Septin7 is demonstrated. KD mice displayed a characteristic hunchback phenotype with skeletal deformities, reduction in in vivo and in vitro force generation, and disorganized mitochondrial networks. Furthermore, knockout of Septin7 in C2C12 cells resulted in complete loss of cell division while KD cells provided evidence that Septin7 is essential for proper myotube differentiation. These and the transient increase in Septin7 expression following muscle injury suggest that it may be involved in muscle regeneration and development.
Collapse
Affiliation(s)
- Mónika Gönczi
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Zsolt Ráduly
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
- Doctoral School of Molecular Medicine, University of DebrecenDebrecenHungary
| | - László Szabó
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
- Doctoral School of Molecular Medicine, University of DebrecenDebrecenHungary
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Andrea Telek
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Nóra Dobrosi
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Norbert Balogh
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
- Doctoral School of Molecular Medicine, University of DebrecenDebrecenHungary
| | - Péter Szentesi
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Gréta Kis
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Miklós Antal
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - György Trencsenyi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of DebrecenDebrecenHungary
| |
Collapse
|
23
|
Voglewede MM, Zhang H. Polarity proteins: Shaping dendritic spines and memory. Dev Biol 2022; 488:68-73. [PMID: 35580729 PMCID: PMC9953585 DOI: 10.1016/j.ydbio.2022.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023]
Abstract
The morphogenesis and plasticity of dendritic spines are associated with synaptic strength, learning, and memory. Dendritic spines are highly compartmentalized structures, which makes proteins involved in cellular polarization and membrane compartmentalization likely candidates regulating their formation and maintenance. Indeed, recent studies suggest polarity proteins help form and maintain dendritic spines by compartmentalizing the spine neck and head. Here, we review emerging evidence that polarity proteins regulate dendritic spine plasticity and stability through the cytoskeleton, scaffolding molecules, and signaling molecules. We specifically analyze various polarity complexes known to contribute to different forms of cell polarization processes and examine the essential conceptual context linking these groups of polarity proteins to dendritic spine morphogenesis, plasticity, and cognitive functions.
Collapse
Affiliation(s)
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
| |
Collapse
|
24
|
Viard J, Loe-Mie Y, Daudin R, Khelfaoui M, Plancon C, Boland A, Tejedor F, Huganir RL, Kim E, Kinoshita M, Liu G, Haucke V, Moncion T, Yu E, Hindie V, Bléhaut H, Mircher C, Herault Y, Deleuze JF, Rain JC, Simonneau M, Lepagnol-Bestel AM. Chr21 protein-protein interactions: enrichment in proteins involved in intellectual disability, autism, and late-onset Alzheimer's disease. Life Sci Alliance 2022; 5:e202101205. [PMID: 35914814 PMCID: PMC9348576 DOI: 10.26508/lsa.202101205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Down syndrome (DS) is caused by human chromosome 21 (HSA21) trisomy. It is characterized by a poorly understood intellectual disability (ID). We studied two mouse models of DS, one with an extra copy of the <i>Dyrk1A</i> gene (189N3) and the other with an extra copy of the mouse Chr16 syntenic region (Dp(16)1Yey). RNA-seq analysis of the transcripts deregulated in the embryonic hippocampus revealed an enrichment in genes associated with chromatin for the 189N3 model, and synapses for the Dp(16)1Yey model. A large-scale yeast two-hybrid screen (82 different screens, including 72 HSA21 baits and 10 rebounds) of a human brain library containing at least 10<sup>7</sup> independent fragments identified 1,949 novel protein-protein interactions. The direct interactors of HSA21 baits and rebounds were significantly enriched in ID-related genes (<i>P</i>-value < 2.29 × 10<sup>-8</sup>). Proximity ligation assays showed that some of the proteins encoded by HSA21 were located at the dendritic spine postsynaptic density, in a protein network at the dendritic spine postsynapse. We located HSA21 DYRK1A and DSCAM, mutations of which increase the risk of autism spectrum disorder (ASD) 20-fold, in this postsynaptic network. We found that an intracellular domain of DSCAM bound either DLGs, which are multimeric scaffolds comprising receptors, ion channels and associated signaling proteins, or DYRK1A. The DYRK1A-DSCAM interaction domain is conserved in <i>Drosophila</i> and humans. The postsynaptic network was found to be enriched in proteins associated with ARC-related synaptic plasticity, ASD, and late-onset Alzheimer's disease. These results highlight links between DS and brain diseases with a complex genetic basis.
Collapse
Affiliation(s)
- Julia Viard
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | - Yann Loe-Mie
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
| | - Rachel Daudin
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
| | - Malik Khelfaoui
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
| | - Christine Plancon
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | - Anne Boland
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | - Francisco Tejedor
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Universidad Miguel Hernandez-Campus de San Juan, San Juan, Spain
| | - Richard L Huganir
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Makoto Kinoshita
- Department of Molecular Biology, Division of Biological Science, Nagoya University Graduate School of Science, Nagoya, Japan
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Volker Haucke
- Department of Molecular Pharmacology and Cell Biology, Leibniz Institut für Molekulare Pharmakologie (FMP) and Freie Universität Berlin, Berlin, Germany
| | | | - Eugene Yu
- Department of Cellular and Molecular Biology, Roswell Park Division of Graduate School, State University of New York at Buffalo, Buffalo, NY, USA
| | | | | | | | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- INSERM, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
- PHENOMIN, Institut Clinique de la Souris, ICS, GIE CERBM, CNRS, INSERM, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Jean-François Deleuze
- Laboratoire de Génomique Fonctionnelle, CNG, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Evry, France
| | | | - Michel Simonneau
- Centre Psychiatrie and Neurosciences, INSERM U894, Paris, France
- Université Paris-Saclay, CNRS, ENS Paris-Saclay, CentraleSupélec, LuMIn, Gif sur Yvette, France
- Department of Biology, Ecole Normale Supérieure Paris-Saclay Université Paris-Saclay, Gif sur Yvette, France
| | | |
Collapse
|
25
|
Radler MR, Spiliotis ET. Right place, right time - Spatial guidance of neuronal morphogenesis by septin GTPases. Curr Opin Neurobiol 2022; 75:102557. [PMID: 35609489 PMCID: PMC9968515 DOI: 10.1016/j.conb.2022.102557] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/07/2022] [Accepted: 04/20/2022] [Indexed: 12/21/2022]
Abstract
Neuronal morphogenesis is guided by outside-in signals and inside-out mechanisms, which require spatiotemporal precision. How the intracellular mechanisms of neuronal morphogenesis are spatiotemporally controlled is not well understood. Septins comprise a unique GTPase module, which consists of complexes with differential localizations and functions. Septins demarcate distinct membrane domains in neural precursor cells, orienting the axis of cell division and the sites of neurite formation. By controlling the localization of membrane and cytoskeletal proteins, septins promote axon-dendrite formation and polarity. Furthermore, septins modulate vesicle exocytosis at pre-synaptic terminals, and stabilize dendritic spines and post-synaptic densities in a phospho-regulatable manner. We posit that neuronal septins are topologically and functionally specialized for the spatiotemporal regulation of neuronal morphogenesis and plasticity.
Collapse
Affiliation(s)
- Megan R. Radler
- Department of Biology, Drexel University, Papadakis Integrated Sciences Building 423, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Elias T. Spiliotis
- Department of Biology, Drexel University, Papadakis Integrated Sciences Building 423, 3245 Chestnut St, Philadelphia, PA 19104, USA
| |
Collapse
|
26
|
Kim OV, Litvinov RI, Mordakhanova ER, Bi E, Vagin O, Weisel JW. Contribution of septins to human platelet structure and function. iScience 2022; 25:104654. [PMID: 35832887 PMCID: PMC9272382 DOI: 10.1016/j.isci.2022.104654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 04/23/2022] [Accepted: 06/17/2022] [Indexed: 11/29/2022] Open
Abstract
Although septins have been well-studied in nucleated cells, their role in anucleate blood platelets remains obscure. Here, we elucidate the contribution of septins to human platelet structure and functionality. We show that Septin-2 and Septin-9 are predominantly distributed at the periphery of resting platelets and co-localize strongly with microtubules. Activation of platelets by thrombin causes clustering of septins and impairs their association with microtubules. Inhibition of septin dynamics with forchlorfenuron (FCF) reduces thrombin-induced densification of septins and lessens their colocalization with microtubules in resting and activated platelets. Exposure to FCF alters platelet shape, suggesting that septins stabilize platelet cytoskeleton. FCF suppresses platelet integrin αIIbβ3 activation, promotes phosphatidylserine exposure on activated platelets, and induces P-selectin expression on resting platelets, suggesting septin involvement in these processes. Inhibition of septin dynamics substantially reduces platelet contractility and abrogates their spreading on fibrinogen-coated surfaces. Overall, septins strongly contribute to platelet structure, activation and biomechanics.
Collapse
Affiliation(s)
- Oleg V. Kim
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rustem I. Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elmira R. Mordakhanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Erfei Bi
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olga Vagin
- Department of Pediatrics, Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - John W. Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
27
|
Menon MB, Gaestel M. Editorial: Emerging Functions of Septins—Volume II. Front Cell Dev Biol 2022; 10:949824. [PMID: 35784463 PMCID: PMC9246257 DOI: 10.3389/fcell.2022.949824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 05/31/2022] [Indexed: 11/26/2022] Open
Affiliation(s)
- Manoj B. Menon
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
- *Correspondence: Manoj B. Menon, ; Matthias Gaestel,
| | - Matthias Gaestel
- Institute for Cell Biochemistry, Hannover Medical School, Hannover, Germany
- *Correspondence: Manoj B. Menon, ; Matthias Gaestel,
| |
Collapse
|
28
|
Byeon S, Werner B, Falter R, Davidsen K, Snyder C, Ong SE, Yadav S. Proteomic Identification of Phosphorylation-Dependent Septin 7 Interactors that Drive Dendritic Spine Formation. Front Cell Dev Biol 2022; 10:836746. [PMID: 35602601 PMCID: PMC9114808 DOI: 10.3389/fcell.2022.836746] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
Septins are a family of cytoskeletal proteins that regulate several important aspects of neuronal development. Septin 7 (Sept7) is enriched at the base of dendritic spines in excitatory neurons and mediates both spine formation and spine and synapse maturation. Phosphorylation at a conserved C-terminal tail residue of Sept7 mediates its translocation into the dendritic spine head to allow spine and synapse maturation. The mechanistic basis for postsynaptic stability and compartmentalization conferred by phosphorylated Sept7, however, is unclear. We report herein the proteomic identification of Sept7 phosphorylation-dependent neuronal interactors. Using Sept7 C-terminal phosphopeptide pulldown and biochemical assays, we show that the 14-3-3 family of proteins specifically interacts with Sept7 when phosphorylated at the T426 residue. Biochemically, we validate the interaction between Sept7 and 14-3-3 isoform gamma and show that 14-3-3 gamma is also enriched in the mature dendritic spine head. Furthermore, we demonstrate that interaction of phosphorylated Sept7 with 14-3-3 protects it from dephosphorylation, as expression of a 14-3-3 antagonist significantly decreases phosphorylated Sept7 in neurons. This study identifies 14-3-3 proteins as an important physiological regulator of Sept7 function in neuronal development.
Collapse
Affiliation(s)
- Sujin Byeon
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
| | - Bailey Werner
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Reilly Falter
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Kristian Davidsen
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Calvin Snyder
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Shao-En Ong
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
29
|
Chen KR, Wang HY, Liao YH, Sun LH, Huang YH, Yu L, Kuo PL. Effects of Septin-14 Gene Deletion on Adult Cognitive/Emotional Behavior. Front Mol Neurosci 2022; 15:880858. [PMID: 35571367 PMCID: PMC9100402 DOI: 10.3389/fnmol.2022.880858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
While various septin GTPases have been reported for their physiological functions, their roles in orchestrating complex cognitive/emotional functions in adult mammals remained scarcely explored. A comprehensive behavioral test battery was administered to two sexes of 12-week-old Septin-14 (SEPT14) knockout (KO) and wild-type (WT) mice. The sexually dimorphic effects of brain SEPT14 KO on inhibitory avoidance (IA) and hippocampal mGluR5 expression were noticed with greater IA latency and elevated mGluR5 level exclusively in male KO mice. Moreover, SEPT14 KO appeared to be associated with stress-provoked anxiety increase in a stress-related navigation task regardless of animals’ sexes. While male and female WT mice demonstrated comparable cell proliferation in the dorsal and ventral hippocampal dentate gyrus (DG), both sexes of SEPT14 KO mice had increased cell proliferation in the ventral DG. Finally, male and female SEPT14 KO mice displayed dampened observational fear conditioning magnitude and learning-provoked corticosterone secretion as compared to their same-sex WT mice. These results, taken together, prompt us to conclude that male, but not female, mice lacking the Septin-14 gene may exhibit increased aversive emotion-related learning and dorsal/ventral hippocampal mGluR5 expressions. Moreover, deletion of SEPT14 may be associated with elevated ventral hippocampal DG cell proliferation and stress-provoked anxiety-like behavior, while dampening vicarious fear conditioning magnitudes.
Collapse
Affiliation(s)
- Kuan-Ru Chen
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
- Department of Obstetrics and Gynecology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Han-Yu Wang
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Yi-Han Liao
- Department of Physiology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Li-Han Sun
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan, Taiwan
- Department of Physiology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Yu-Han Huang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
- Department of Obstetrics and Gynecology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Lung Yu
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan, Taiwan
- Department of Physiology, National Cheng Kung University College of Medicine, Tainan, Taiwan
- Lung Yu,
| | - Pao-Lin Kuo
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
- Department of Obstetrics and Gynecology, National Cheng Kung University College of Medicine, Tainan, Taiwan
- *Correspondence: Pao-Lin Kuo,
| |
Collapse
|
30
|
Lau Y, Oamen HP, Grogg M, Parfenova I, Saarikangas J, Hannay R, Nichols RA, Hilvert D, Barral Y, Caudron F. Whi3 mnemon association with endoplasmic reticulum membranes confines the memory of deceptive courtship to the yeast mother cell. Curr Biol 2022; 32:963-974.e7. [PMID: 35085498 PMCID: PMC8938615 DOI: 10.1016/j.cub.2022.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/19/2021] [Accepted: 01/04/2022] [Indexed: 12/17/2022]
Abstract
Prion-like proteins are involved in many aspects of cellular physiology, including cellular memory. In response to deceptive courtship, budding yeast escapes pheromone-induced cell-cycle arrest through the coalescence of the G1/S inhibitor Whi3 into a dominant, inactive super-assembly. Whi3 is a mnemon (Whi3mnem), a protein that conformational change maintains as a trait in the mother cell but is not inherited by the daughter cells. How the maintenance and asymmetric inheritance of Whi3mnem are achieved is unknown. Here, we report that Whi3mnem is closely associated with endoplasmic reticulum (ER) membranes and is retained in the mother cell by the lateral diffusion barriers present at the bud neck. Strikingly, barrier defects made Whi3mnem propagate in a mitotically stable, prion-like manner. The amyloid-forming glutamine-rich domain of Whi3 was required for both mnemon and prion-like behaviors. Thus, we propose that Whi3mnem is in a self-templating state, lending temporal maintenance of memory, whereas its association with the compartmentalized membranes of the ER prevents infectious propagation to the daughter cells. These results suggest that confined self-templating super-assembly is a powerful mechanism for the long-term encoding of information in a spatially defined manner. Yeast courtship may provide insights on how individual synapses become potentiated in neuronal memory.
Collapse
Affiliation(s)
- Yasmin Lau
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Henry Patrick Oamen
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Marcel Grogg
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg, 8093 Zürich, Switzerland
| | - Iuliia Parfenova
- Institute of Biochemistry, ETH Zürich, Otto-Stern-Weg, 8093 Zürich, Switzerland
| | - Juha Saarikangas
- Helsinki Institute of Life Science HiLIFE, Viikinkaari 5, 00790 Helsinki, Finland; Faculty of Biological and Environmental Sciences, Viikinkaari 5, 00790 Helsinki, Finland; Neuroscience Center, University of Helsinki, Viikinkaari 5, 00790 Helsinki, Finland
| | - Robin Hannay
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Richard Alan Nichols
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Donald Hilvert
- Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg, 8093 Zürich, Switzerland
| | - Yves Barral
- Institute of Biochemistry, ETH Zürich, Otto-Stern-Weg, 8093 Zürich, Switzerland
| | - Fabrice Caudron
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK; IGMM, Univ Montpellier, CNRS, Route de Mende, 34293 Montpellier, France.
| |
Collapse
|
31
|
Dhanya SK, Hasan G. Deficits Associated With Loss of STIM1 in Purkinje Neurons Including Motor Coordination Can Be Rescued by Loss of Septin 7. Front Cell Dev Biol 2021; 9:794807. [PMID: 34993201 PMCID: PMC8724567 DOI: 10.3389/fcell.2021.794807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/15/2021] [Indexed: 12/26/2022] Open
Abstract
Septins are cytoskeletal proteins that can assemble to form heteromeric filamentous complexes and regulate a range of membrane-associated cellular functions. SEPT7, a member of the septin family, functions as a negative regulator of the plasma membrane–localized store-operated Ca2+ entry (SOCE) channel, Orai in Drosophila neurons, and in human neural progenitor cells. Knockdown of STIM, a Ca2+ sensor in the endoplasmic reticulum (ER) and an integral component of SOCE, leads to flight deficits in Drosophila that can be rescued by partial loss of SEPT7 in neurons. Here, we tested the effect of reducing and removing SEPT7 in mouse Purkinje neurons (PNs) with the loss of STIM1. Mice with the complete knockout of STIM1 in PNs exhibit several age-dependent changes. These include altered gene expression in PNs, which correlates with increased synapses between climbing fiber (CF) axons and Purkinje neuron (PN) dendrites and a reduced ability to learn a motor coordination task. Removal of either one or two copies of the SEPT7 gene in STIM1KO PNs restored the expression of a subset of genes, including several in the category of neuron projection development. Importantly, the rescue of gene expression in these animals is accompanied by normal CF-PN innervation and an improved ability to learn a motor coordination task in aging mice. Thus, the loss of SEPT7 in PNs further modulates cerebellar circuit function in STIM1KO animals. Our findings are relevant in the context of identifying SEPT7 as a putative therapeutic target for various neurodegenerative diseases caused by reduced intracellular Ca2+ signaling.
Collapse
Affiliation(s)
- Sreeja Kumari Dhanya
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
- SASTRA University, Thanjavur, India
| | - Gaiti Hasan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
- *Correspondence: Gaiti Hasan,
| |
Collapse
|
32
|
Marquardt J, Chen X, Bi E. Septin Assembly and Remodeling at the Cell Division Site During the Cell Cycle. Front Cell Dev Biol 2021; 9:793920. [PMID: 34901034 PMCID: PMC8656427 DOI: 10.3389/fcell.2021.793920] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/08/2021] [Indexed: 11/17/2022] Open
Abstract
The septin family of proteins can assemble into filaments that further organize into different higher order structures to perform a variety of different functions in different cell types and organisms. In the budding yeast Saccharomyces cerevisiae, the septins localize to the presumptive bud site as a cortical ring prior to bud emergence, expand into an hourglass at the bud neck (cell division site) during bud growth, and finally “split” into a double ring sandwiching the cell division machinery during cytokinesis. While much work has been done to understand the functions and molecular makeups of these structures, the mechanisms underlying the transitions from one structure to another have largely remained elusive. Recent studies involving advanced imaging and in vitro reconstitution have begun to reveal the vast complexity involved in the regulation of these structural transitions, which defines the focus of discussion in this mini-review.
Collapse
Affiliation(s)
- Joseph Marquardt
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Xi Chen
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Erfei Bi
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
33
|
Schuster T, Geiger H. Septins in Stem Cells. Front Cell Dev Biol 2021; 9:801507. [PMID: 34957123 PMCID: PMC8695968 DOI: 10.3389/fcell.2021.801507] [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: 10/25/2021] [Accepted: 11/24/2021] [Indexed: 12/01/2022] Open
Abstract
Septins were first described in yeast. Due to extensive research in non-yeast cells, Septins are now recognized across all species as important players in the regulation of the cytoskeleton, in the establishment of polarity, for migration, vesicular trafficking and scaffolding. Stem cells are primarily quiescent cells, and this actively maintained quiescent state is critical for proper stem cell function. Equally important though, stem cells undergo symmetric or asymmetric division, which is likely linked to the level of symmetry found in the mother stem cell. Due to the ability to organize barriers and be able to break symmetry in cells, Septins are thought to have a significant impact on organizing quiescence as well as the mode (symmetric vs asymmetric) of stem cell division to affect self-renewal versus differentiation. Mechanisms of regulating mammalian quiescence and symmetry breaking by Septins are though still somewhat elusive. Within this overview article, we summarize current knowledge on the role of Septins in stem cells ranging from yeast to mice especially with respect to quiescence and asymmetric division, with a special focus on hematopoietic stem cells.
Collapse
Affiliation(s)
| | - Hartmut Geiger
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| |
Collapse
|
34
|
Kuzmić M, Linares GC, Fialová JL, Iv F, Salaün D, Llewellyn A, Gomes M, Belhabib M, Liu Y, Asano K, Rodrigues M, Isnardon D, Tachibana T, Koenderink GH, Badache A, Mavrakis M, Verdier-Pinard P. Septin-microtubule association via a motif unique to the isoform 1 of septin 9 tunes stress fibers. J Cell Sci 2021; 135:273936. [PMID: 34854883 DOI: 10.1242/jcs.258850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/19/2021] [Indexed: 10/19/2022] Open
Abstract
Septins, a family of GTP-binding proteins assembling into higher order structures, interface with the membrane, actin filaments and microtubules, which positions them as important regulators of cytoarchitecture. Septin 9 (SEPT9), which is frequently overexpressed in tumors and mutated in hereditary neuralgic amyotrophy (HNA), mediates the binding of septins to microtubules, but the molecular determinants of this interaction remained uncertain. We demonstrate that a short MAP-like motif unique to SEPT9 isoform 1 (SEPT9_i1) drives septin octamer-microtubule interaction in cells and in vitro reconstitutions. Septin-microtubule association requires polymerizable septin octamers harboring SEPT9_i1. Although outside of the MAP-like motif, HNA mutations abrogates this association, identifying a putative regulatory domain. Removal of this domain from SEPT9_i1 sequesters septins on microtubules, promotes microtubule stability and alters actomyosin fiber distribution and tension. Thus, we identify key molecular determinants and potential regulatory roles of septin-microtubule interaction, paving the way to deciphering the mechanisms underlying septin-associated pathologies.
Collapse
Affiliation(s)
- Mira Kuzmić
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| | - Gerard Castro Linares
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Jindřiška Leischner Fialová
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France.,Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - François Iv
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Danièle Salaün
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| | - Alex Llewellyn
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Maxime Gomes
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Mayssa Belhabib
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Yuxiang Liu
- Department of Bioengineering, Graduate School of Engineering, Osaka City University, Osaka, Japan
| | - Keisuke Asano
- Department of Bioengineering, Graduate School of Engineering, Osaka City University, Osaka, Japan
| | - Magda Rodrigues
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| | - Daniel Isnardon
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| | - Taro Tachibana
- Department of Bioengineering, Graduate School of Engineering, Osaka City University, Osaka, Japan.,Cell Engineering Corporation, Osaka, Japan
| | - Gijsje H Koenderink
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Ali Badache
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| | - Manos Mavrakis
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Pascal Verdier-Pinard
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| |
Collapse
|
35
|
PIAS1 Regulates Hepatitis C Virus-Induced Lipid Droplet Accumulation by Controlling Septin 9 and Microtubule Filament Assembly. Pathogens 2021; 10:pathogens10101327. [PMID: 34684276 PMCID: PMC8537804 DOI: 10.3390/pathogens10101327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 01/22/2023] Open
Abstract
Chronic hepatitis C virus (HCV) infection often leads to fibrosis and chronic hepatitis, then cirrhosis and ultimately hepatocellular carcinoma (HCC). The processes of the HVC life cycle involve intimate interactions between viral and host cell proteins and lipid metabolism. However, the molecules and mechanisms involved in this tripartite interaction remain poorly understood. Herein, we show that the infection of HCC-derived Huh7.5 cells with HCV promotes upregulation of the protein inhibitor of activated STAT1 (PIAS1). Reciprocally, PIAS1 regulated the expression of HCV core protein and HCV-induced LD accumulation and impaired HCV replication. Furthermore, PIAS1 controlled HCV-promoted septin 9 filament formation and microtubule polymerization. Subsequently, we found that PIAS1 interacted with septin 9 and controlled its assembly on filaments, which thus affected septin 9-induced lipid droplet accumulation. Taken together, these data reveal that PIAS1 regulates the accumulation of lipid droplets and offer a meaningful insight into how HCV interacts with host proteins.
Collapse
|
36
|
Lo LHY, Dong R, Lyu Q, Lai KO. The Protein Arginine Methyltransferase PRMT8 and Substrate G3BP1 Control Rac1-PAK1 Signaling and Actin Cytoskeleton for Dendritic Spine Maturation. Cell Rep 2021; 31:107744. [PMID: 32521269 DOI: 10.1016/j.celrep.2020.107744] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/01/2020] [Accepted: 05/18/2020] [Indexed: 01/25/2023] Open
Abstract
Excitatory synapses of neurons are located on dendritic spines. Spine maturation is essential for the stability of synapses and memory consolidation, and overproduction of the immature filopodia is associated with brain disorders. The structure and function of synapses can be modulated by protein post-translational modification (PTM). Arginine methylation is a major PTM that regulates chromatin structure, transcription, and splicing within the nucleus. Here we find that the protein arginine methyltransferase PRMT8 is present at neuronal synapses and its expression is upregulated in the hippocampus when dendritic spine maturation occurs. Depletion of PRMT8 leads to overabundance of filopodia and mis-localization of excitatory synapses. Mechanistically, PRMT8 promotes dendritic spine morphology through methylation of the dendritic RNA-binding protein G3BP1 and suppression of the Rac1-PAK1 signaling pathway to control synaptic actin dynamics. Our findings unravel arginine methylation as a crucial regulatory mechanism for actin cytoskeleton during synapse development.
Collapse
Affiliation(s)
- Louisa Hoi-Ying Lo
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Rui Dong
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Quanwei Lyu
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Kwok-On Lai
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
37
|
Abstract
Septins are an integral component of the cytoskeleton, assembling into higher-order oligomers and filamentous polymers that associate with actin filaments, microtubules and membranes. Here, we review septin interactions with actin and microtubules, and septin-mediated regulation of the organization and dynamics of these cytoskeletal networks, which is critical for cellular morphogenesis. We discuss how actomyosin-associated septins function in cytokinesis, cell migration and host defense against pathogens. We highlight newly emerged roles of septins at the interface of microtubules and membranes with molecular motors, which point to a 'septin code' for the regulation of membrane traffic. Additionally, we revisit the functions of microtubule-associated septins in mitosis and meiosis. In sum, septins comprise a unique module of cytoskeletal regulators that are spatially and functionally specialized and have properties of bona fide actin-binding and microtubule-associated proteins. With many questions still outstanding, the study of septins will continue to provide new insights into fundamental problems of cytoskeletal organization and function.
Collapse
|
38
|
Szuba A, Bano F, Castro-Linares G, Iv F, Mavrakis M, Richter RP, Bertin A, Koenderink GH. Membrane binding controls ordered self-assembly of animal septins. eLife 2021; 10:63349. [PMID: 33847563 PMCID: PMC8099429 DOI: 10.7554/elife.63349] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 04/12/2021] [Indexed: 12/23/2022] Open
Abstract
Septins are conserved cytoskeletal proteins that regulate cell cortex mechanics. The mechanisms of their interactions with the plasma membrane remain poorly understood. Here, we show by cell-free reconstitution that binding to flat lipid membranes requires electrostatic interactions of septins with anionic lipids and promotes the ordered self-assembly of fly septins into filamentous meshworks. Transmission electron microscopy reveals that both fly and mammalian septin hexamers form arrays of single and paired filaments. Atomic force microscopy and quartz crystal microbalance demonstrate that the fly filaments form mechanically rigid, 12- to 18-nm thick, double layers of septins. By contrast, C-terminally truncated septin mutants form 4-nm thin monolayers, indicating that stacking requires the C-terminal coiled coils on DSep2 and Pnut subunits. Our work shows that membrane binding is required for fly septins to form ordered arrays of single and paired filaments and provides new insights into the mechanisms by which septins may regulate cell surface mechanics.
Collapse
Affiliation(s)
- Agata Szuba
- AMOLF, Department of Living Matter, Biological Soft Matter group, Amsterdam, Netherlands
| | - Fouzia Bano
- School of Biomedical Sciences, Faculty of Biological Sciences, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom.,School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, United Kingdom.,Bragg Centre for Materials Research, University of Leeds, Leeds, United Kingdom
| | - Gerard Castro-Linares
- AMOLF, Department of Living Matter, Biological Soft Matter group, Amsterdam, Netherlands.,Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, Netherlands
| | - Francois Iv
- Institut Fresnel, CNRS, Aix-Marseille Univ, Centrale Marseille, Marseille, France
| | - Manos Mavrakis
- Institut Fresnel, CNRS, Aix-Marseille Univ, Centrale Marseille, Marseille, France
| | - Ralf P Richter
- School of Biomedical Sciences, Faculty of Biological Sciences, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom.,School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, United Kingdom.,Bragg Centre for Materials Research, University of Leeds, Leeds, United Kingdom
| | - Aurélie Bertin
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, Paris, France.,Sorbonne Université, Paris, France
| | - Gijsje H Koenderink
- AMOLF, Department of Living Matter, Biological Soft Matter group, Amsterdam, Netherlands.,Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, Netherlands
| |
Collapse
|
39
|
Biophysical Analysis of Schistosoma mansoni Septins. Methods Mol Biol 2021; 2151:197-210. [PMID: 32452006 DOI: 10.1007/978-1-0716-0635-3_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Septins are dynamic filament-forming proteins that are recognized as important components of the cytoskeleton and are involved in numerous functions inside the cells, such as cytokinesis, exocytosis, and ciliogenesis and even in defense against pathogenic bacteria. Despite being highly conserved in eukaryotes, there is scarce literature on the role of septins in organisms other than humans and yeast. Therefore, septins from Schistosoma mansoni represent an interesting model to study an unexplored branch of this protein family. Here we described standard protocols for recombinant production and initial characterization of septins from S. mansoni. Septins are notably difficult to purify, mostly due to their tendency to assemble into filaments. Therefore, specific protocols to stabilize these proteins have been developed. In this chapter, we systematically describe protocols to clone, express, and purify schistosome septins. We also describe the use of circular dichroism to assess the folding and stability of septins and use of chromatography to characterize their oligomeric state, bound guanine nucleotide, and GTP hydrolysis. We expect that these protocols may help researchers involved in the study of schistosome septins as well as assist to establish protocols for septins from other organisms.
Collapse
|
40
|
Nourbakhsh K, Yadav S. Kinase Signaling in Dendritic Development and Disease. Front Cell Neurosci 2021; 15:624648. [PMID: 33642997 PMCID: PMC7902504 DOI: 10.3389/fncel.2021.624648] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/06/2021] [Indexed: 01/19/2023] Open
Abstract
Dendrites undergo extensive growth and remodeling during their lifetime. Specification of neurites into dendrites is followed by their arborization, maturation, and functional integration into synaptic networks. Each of these distinct developmental processes is spatially and temporally controlled in an exquisite fashion. Protein kinases through their highly specific substrate phosphorylation regulate dendritic growth and plasticity. Perturbation of kinase function results in aberrant dendritic growth and synaptic function. Not surprisingly, kinase dysfunction is strongly associated with neurodevelopmental and psychiatric disorders. Herein, we review, (a) key kinase pathways that regulate dendrite structure, function and plasticity, (b) how aberrant kinase signaling contributes to dendritic dysfunction in neurological disorders and (c) emergent technologies that can be applied to dissect the role of protein kinases in dendritic structure and function.
Collapse
Affiliation(s)
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
41
|
Kushnireva L, Korkotian E, Segal M. Calcium Sensors STIM1 and STIM2 Regulate Different Calcium Functions in Cultured Hippocampal Neurons. Front Synaptic Neurosci 2021; 12:573714. [PMID: 33469426 PMCID: PMC7813759 DOI: 10.3389/fnsyn.2020.573714] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022] Open
Abstract
There are growing indications for the involvement of calcium stores in the plastic properties of neurons and particularly in dendritic spines of central neurons. The store-operated calcium entry (SOCE) channels are assumed to be activated by the calcium sensor stromal interaction molecule (STIM)which leads to activation of its associated Orai channel. There are two STIM species, and the differential role of the two in SOCE is not entirely clear. In the present study, we were able to distinguish between transfected STIM1, which is more mobile primarily in young neurons, and STIM2 which is less mobile and more prominent in older neurons in culture. STIM1 mobility is associated with spontaneous calcium sparks, local transient rise in cytosolic [Ca2+]i, and in the formation and elongation of dendritic filopodia/spines. In contrast, STIM2 is associated with older neurons, where it is mobile and moves into dendritic spines primarily when cytosolic [Ca2+]i levels are reduced, apparently to activate resident Orai channels. These results highlight a role for STIM1 in the regulation of [Ca2+]i fluctuations associated with the formation of dendritic spines or filopodia in the developing neuron, whereas STIM2 is associated with the maintenance of calcium entry into stores in the adult neuron.
Collapse
Affiliation(s)
- Liliya Kushnireva
- Department of Neurobiology, The Weizmann Institute, Rehovot, Israel.,Faculty of Biology, Perm State University, Perm, Russia
| | - Eduard Korkotian
- Department of Neurobiology, The Weizmann Institute, Rehovot, Israel.,Faculty of Biology, Perm State University, Perm, Russia
| | - Menahem Segal
- Department of Neurobiology, The Weizmann Institute, Rehovot, Israel
| |
Collapse
|
42
|
Shen Q, Liu L, Gu X, Xing D. Photobiomodulation suppresses JNK3 by activation of ERK/MKP7 to attenuate AMPA receptor endocytosis in Alzheimer's disease. Aging Cell 2021; 20:e13289. [PMID: 33336891 PMCID: PMC7811840 DOI: 10.1111/acel.13289] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/07/2020] [Accepted: 11/27/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD), a severe age‐related neurodegenerative disorder, lacks effective therapeutic methods at present. Physical approaches such as gamma frequency light flicker that can effectively reduce amyloid load have been reported recently. Our previous research showed that a physical method named photobiomodulation (PBM) therapy rescues Aβ‐induced dendritic atrophy in vitro. However, it remains to be further investigated the mechanism by which PBM affects AD‐related multiple pathological features to improve learning and memory deficits. Here, we found that PBM attenuated Aβ‐induced synaptic dysfunction and neuronal death through MKP7‐dependent suppression of JNK3, a brain‐specific JNK isoform related to neurodegeneration. The results showed PBM‐attenuated amyloid load, AMPA receptor endocytosis, dendrite injury, and inflammatory responses, thereby rescuing memory deficits in APP/PS1 mice. We noted JNK3 phosphorylation was dramatically decreased after PBM treatment in vivo and in vitro. Mechanistically, PBM activated ERK, which subsequently phosphorylated and stabilized MKP7, resulting in JNK3 inactivation. Furthermore, activation of ERK/MKP7 signaling by PBM increased the level of AMPA receptor subunit GluR 1 phosphorylation and attenuated AMPA receptor endocytosis in an AD pathological model. Collectively, these data demonstrated that PBM has potential therapeutic value in reducing multiple pathological features associated with AD, which is achieved by regulating JNK3, thus providing a noninvasive, and drug‐free therapeutic strategy to impede AD progression.
Collapse
Affiliation(s)
- Qi Shen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science South China Normal University Guangzhou China
- College of Biophotonics South China Normal University Guangzhou China
| | - Lei Liu
- College of Biophotonics South China Normal University Guangzhou China
| | - Xiaotong Gu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science South China Normal University Guangzhou China
- College of Biophotonics South China Normal University Guangzhou China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science South China Normal University Guangzhou China
- College of Biophotonics South China Normal University Guangzhou China
| |
Collapse
|
43
|
Tamada H, Blanc J, Korogod N, Petersen CC, Knott GW. Ultrastructural comparison of dendritic spine morphology preserved with cryo and chemical fixation. eLife 2020; 9:56384. [PMID: 33274717 PMCID: PMC7748412 DOI: 10.7554/elife.56384] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 12/04/2020] [Indexed: 12/25/2022] Open
Abstract
Previously, we showed that cryo fixation of adult mouse brain tissue gave a truer representation of brain ultrastructure in comparison with a standard chemical fixation method (Korogod et al., 2015). Extracellular space matched physiological measurements, there were larger numbers of docked vesicles and less glial coverage of synapses and blood capillaries. Here, using the same preservation approaches, we compared the morphology of dendritic spines. We show that the length of the spine and the volume of its head is unchanged; however, the spine neck width is thinner by more than 30% after cryo fixation. In addition, the weak correlation between spine neck width and head volume seen after chemical fixation was not present in cryo-fixed spines. Our data suggest that spine neck geometry is independent of the spine head volume, with cryo fixation showing enhanced spine head compartmentalization and a higher predicted electrical resistance between spine head and parent dendrite.
Collapse
Affiliation(s)
- Hiromi Tamada
- Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Biological Electron Microscopy Facility, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Functional Anatomy and Neuroscience, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Japan Society of the Promotion of Sciences (JSPS), Tokyo, Japan
| | - Jerome Blanc
- Biological Electron Microscopy Facility, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Natalya Korogod
- Biological Electron Microscopy Facility, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,School of Health Sciences (HESAV), University of Applied Sciences and Arts Western Switzerland (HES-SO), Lausanne, Switzerland
| | - Carl Ch Petersen
- Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Graham W Knott
- Biological Electron Microscopy Facility, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
44
|
Györffy BA, Tóth V, Török G, Gulyássy P, Kovács RÁ, Vadászi H, Micsonai A, Tóth ME, Sántha M, Homolya L, Drahos L, Juhász G, Kékesi KA, Kardos J. Synaptic mitochondrial dysfunction and septin accumulation are linked to complement-mediated synapse loss in an Alzheimer's disease animal model. Cell Mol Life Sci 2020; 77:5243-5258. [PMID: 32034429 PMCID: PMC7671981 DOI: 10.1007/s00018-020-03468-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/25/2019] [Accepted: 01/21/2020] [Indexed: 12/21/2022]
Abstract
Synaptic functional disturbances with concomitant synapse loss represent central pathological hallmarks of Alzheimer's disease. Excessive accumulation of cytotoxic amyloid oligomers is widely recognized as a key event that underlies neurodegeneration. Certain complement components are crucial instruments of widespread synapse loss because they can tag synapses with functional impairments leading to their engulfment by microglia. However, an exact understanding of the affected synaptic functions that predispose to complement-mediated synapse elimination is lacking. Therefore, we conducted systematic proteomic examinations on synaptosomes prepared from an amyloidogenic mouse model of Alzheimer's disease (APP/PS1). Synaptic fractions were separated according to the presence of the C1q-tag using fluorescence-activated synaptosome sorting and subjected to proteomic comparisons. The results raised the decline of mitochondrial functions in the C1q-tagged synapses of APP/PS1 mice based on enrichment analyses, which was verified using flow cytometry. Additionally, proteomics results revealed extensive alterations in the level of septin protein family members, which are known to dynamically form highly organized pre- and postsynaptic supramolecular structures, thereby affecting synaptic transmission. High-resolution microscopy investigations demonstrated that synapses with considerable amounts of septin-3 and septin-5 show increased accumulation of C1q in APP/PS1 mice compared to the wild-type ones. Moreover, a strong positive correlation was apparent between synaptic septin-3 levels and C1q deposition as revealed via flow cytometry and confocal microscopy examinations. In sum, our results imply that deterioration of synaptic mitochondrial functions and alterations in the organization of synaptic septins are associated with complement-dependent synapse loss in Alzheimer's disease.
Collapse
Affiliation(s)
- Balázs A Györffy
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Vilmos Tóth
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - György Török
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Centre of Excellence, Budapest, Hungary
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Péter Gulyássy
- MS Proteomics Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Réka Á Kovács
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Henrietta Vadászi
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - András Micsonai
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Melinda E Tóth
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Miklós Sántha
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - László Homolya
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Centre of Excellence, Budapest, Hungary
| | - László Drahos
- MS Proteomics Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Gábor Juhász
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- CRU Hungary Ltd., Göd, Hungary
| | - Katalin A Kékesi
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Physiology and Neurobiology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - József Kardos
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary.
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
45
|
Ageta-Ishihara N, Kinoshita M. Developmental and postdevelopmental roles of septins in the brain. Neurosci Res 2020; 170:6-12. [PMID: 33159992 DOI: 10.1016/j.neures.2020.08.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/21/2020] [Accepted: 08/23/2020] [Indexed: 11/25/2022]
Abstract
Morphogenetic processes during brain development and postdevelopmental remodeling of neural architecture depend on the exquisite interplay between the microtubule- and actin-based cytoskeletal systems. Accumulation of evidence indicates cooperative roles of another cytoskeletal system composed of the septin family. Here we overview experimental findings on mammalian septins and their hypothetical roles in the proliferation of neural progenitor cells, neurite development, synapse formation and regulations. The diverse, mostly unexpected phenotypes obtained from gain- and loss-of-function mutants point to unknown molecular network to be elucidated, which may underlie pathogenetic processes of infectious diseases and neuropsychiatric disorders in humans.
Collapse
Affiliation(s)
- Natsumi Ageta-Ishihara
- Division of Biological Science, Nagoya University Graduate School of Science, Furo, Chikusa, Nagoya 464-8602, Japan.
| | - Makoto Kinoshita
- Division of Biological Science, Nagoya University Graduate School of Science, Furo, Chikusa, Nagoya 464-8602, Japan.
| |
Collapse
|
46
|
SEPT7 regulates Ca 2+ entry through Orai channels in human neural progenitor cells and neurons. Cell Calcium 2020; 90:102252. [PMID: 32682163 DOI: 10.1016/j.ceca.2020.102252] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/25/2020] [Accepted: 07/06/2020] [Indexed: 12/16/2022]
Abstract
Human neural progenitor cells (hNPCs) are self-renewing cells of neural lineage that can be differentiated into neurons of different subtypes. Here we show that SEPT7, a member of the family of filament-forming GTPases called septins, prevents constitutive Ca2+ entry through the store-operated Ca2+ entry channel, Orai in hNPCs and in differentiated neurons and is thus required for neuronal calcium homeostasis. Previous work in Drosophila neurons has shown that loss of one copy of the evolutionarily-conserved dSEPT7 gene leads to elevated Ca2+ entry via Orai, in the absence of ER-Ca2+ store depletion. We have identified an N-terminal polybasic region of SEPT7, known to interact with membrane-localized phospholipids, as essential for spontaneous calcium entry through Orai in hNPCs, whereas the GTPase domain of dSEPT7 is dispensable for this purpose. Re-organisation of Orai1 and the ER-Ca2+ sensor STIM1 observed near the plasma membrane in SEPT7 KD hNPCs, supports the idea that Septin7 containing heteromers prevent Ca2+ entry through a fraction of STIM-Orai complexes. Possible mechanisms by which SEPT7 reduction leads to opening of Orai channels in the absence of store-depletion are discussed.
Collapse
|
47
|
Radler MR, Suber A, Spiliotis ET. Spatial control of membrane traffic in neuronal dendrites. Mol Cell Neurosci 2020; 105:103492. [PMID: 32294508 PMCID: PMC7317674 DOI: 10.1016/j.mcn.2020.103492] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Neuronal dendrites are highly branched and specialized compartments with distinct structures and secretory organelles (e.g., spines, Golgi outposts), and a unique cytoskeletal organization that includes microtubules of mixed polarity. Dendritic membranes are enriched with proteins, which specialize in the formation and function of the post-synaptic membrane of the neuronal synapse. How these proteins partition preferentially in dendrites, and how they traffic in a manner that is spatiotemporally accurate and regulated by synaptic activity are long-standing questions of neuronal cell biology. Recent studies have shed new insights into the spatial control of dendritic membrane traffic, revealing new classes of proteins (e.g., septins) and cytoskeleton-based mechanisms with dendrite-specific functions. Here, we review these advances by revisiting the fundamental mechanisms that control membrane traffic at the levels of protein sorting and motor-driven transport on microtubules and actin filaments. Overall, dendrites possess unique mechanisms for the spatial control of membrane traffic, which might have specialized and co-evolved with their highly arborized morphology.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Ayana Suber
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut St, Philadelphia, PA 19104, USA.
| |
Collapse
|
48
|
Ito H, Morishita R, Noda M, Iwamoto I, Nagata KI. Biochemical and morphological characterization of SEPT1 in mouse brain. Med Mol Morphol 2020; 53:221-228. [PMID: 32146512 DOI: 10.1007/s00795-020-00248-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/26/2020] [Indexed: 01/21/2023]
Abstract
Septins are a highly conserved family of GTPases which are identified in diverse organisms ranging from yeast to humans. In mammals, nervous tissues abundantly contain septins and associations of septins with neurological disorders such as Alzheimer's disease and Parkinson's disease have been reported. However, roles of septins in the brain development have not been fully understood. In this study, we produced a specific antibody against mouse SEPT1 and carried out biochemical and morphological characterization of SEPT1. When the expression profile of SEPT1 during mouse brain development was analyzed by western blotting, we found that SEPT1 expression began to increase after birth and the increase continued until postnatal day 22. Subcellular fractionation of mouse brain and subsequent western blot analysis revealed the distribution of SEPT1 in synaptic fractions. Immunofluorescent analyses showed the localization of SEPT1 at synapses in primary cultured mouse hippocampal neurons. We also found the distribution of SEPT1 at synapses in mouse brain by immunohistochemistry. These results suggest that SEPT1 participates in various synaptic events such as the signaling, the neurotransmitter release, and the synapse formation/maintenance.
Collapse
Affiliation(s)
- Hidenori Ito
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai, Aichi, 480-0392, Japan
| | - Rika Morishita
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai, Aichi, 480-0392, Japan
| | - Mariko Noda
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai, Aichi, 480-0392, Japan
| | - Ikuko Iwamoto
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai, Aichi, 480-0392, Japan
| | - Koh-Ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai, Aichi, 480-0392, Japan. .,Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
49
|
Gönczi M, Dienes B, Dobrosi N, Fodor J, Balogh N, Oláh T, Csernoch L. Septins, a cytoskeletal protein family, with emerging role in striated muscle. J Muscle Res Cell Motil 2020; 42:251-265. [PMID: 31955380 PMCID: PMC8332580 DOI: 10.1007/s10974-020-09573-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 01/06/2020] [Indexed: 12/15/2022]
Abstract
Appropriate organization of cytoskeletal components are required for normal distribution and intracellular localization of different ion channels and proteins involved in calcium homeostasis, signal transduction, and contractile function of striated muscle. Proteins of the contractile system are in direct or indirect connection with the extrasarcomeric cytoskeleton. A number of other molecules which have essential role in regulating stretch-, voltage-, and chemical signal transduction from the surface into the cytoplasm or other intracellular compartments are already well characterized. Sarcomere, the basic contractile unit, is comprised of a precisely organized system of thin (actin), and thick (myosin) filaments. Intermediate filaments connect the sarcomeres and other organelles (mitochondria and nucleus), and are responsible for the cellular integrity. Interacting proteins have a very diverse function in coupling of the intracellular assembly components and regulating the normal physiological function. Despite the more and more intense investigations of a new cytoskeletal protein family, the septins, only limited information is available regarding their expression and role in striated, especially in skeletal muscles. In this review we collected basic and specified knowledge regarding this protein group and emphasize the importance of this emerging field in skeletal muscle biology.
Collapse
Affiliation(s)
- Mónika Gönczi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, 4012, Hungary
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, 4012, Hungary
| | - Nóra Dobrosi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, 4012, Hungary
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, 4012, Hungary
| | - Norbert Balogh
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, 4012, Hungary.,Doctoral School of Molecular Medicine, University of Debrecen, Debrecen, 4012, Hungary
| | - Tamás Oláh
- Center of Experimental Orthopaedics, Saarland University, 66421, Homburg, Saar, Germany
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, 4012, Hungary.
| |
Collapse
|
50
|
Qiu R, Runxiang Q, Geng A, Liu J, Xu CW, Menon MB, Gaestel M, Lu Q. SEPT7 Interacts with KIF20A and Regulates the Proliferative State of Neural Progenitor Cells During Cortical Development. Cereb Cortex 2019; 30:3030-3043. [PMID: 31813992 DOI: 10.1093/cercor/bhz292] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/01/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023] Open
Abstract
Balanced proliferation and differentiation of neural progenitor cells (NPCs) are critical for brain development, but how the process is regulated and what components of the cell division machinery is involved are not well understood. Here we report that SEPT7, a cell division regulator originally identified in Saccharomyces cerevisiae, interacts with KIF20A in the intercellular bridge of dividing NPCs and plays an essential role in maintaining the proliferative state of NPCs during cortical development. Knockdown of SEPT7 in NPCs results in displacement of KIF20A from the midbody and early neuronal differentiation. NPC-specific inducible knockout of Sept7 causes early cell cycle exit, precocious neuronal differentiation, and ventriculomegaly in the cortex, but surprisingly does not lead to noticeable cytokinesis defect. Our data uncover an interaction of SEPT7 and KIF20A during NPC divisions and demonstrate a crucial role of SEPT7 in cell fate determination. In addition, this study presents a functional approach for identifying additional cell fate regulators of the mammalian brain.
Collapse
Affiliation(s)
- Runxiang Qiu
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Qiu Runxiang
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Anqi Geng
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.,Institute of Medical Research, Northwestern Polytechnical University, Xian, Shaanxi Province, China
| | - Jiancheng Liu
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - C Wilson Xu
- Balto Pharmaceuticals, Inc., South Pasadena, CA 91030, USA
| | - Manoj B Menon
- Institute of Cell Biochemistry, Hannover Medical School, Hannover 30625, Germany.,Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New-Delhi 110016, India
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover 30625, Germany
| | - Qiang Lu
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|