1
|
Xie L, Kang F, Qin T, Kang Y, Liang T, Xie H, Froese CD, Xie H, Au A, Yip CM, Trimble WS, Gaisano HY. Septin5 deletion enhances β-cell exocytosis by releasing microtubule-tethered insulin granules onto plasma membrane. Nat Commun 2025; 16:2725. [PMID: 40108136 PMCID: PMC11923188 DOI: 10.1038/s41467-025-57421-5] [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: 02/22/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Septin5 interacts with SNARE proteins to regulate exocytosis in neurons, but its role in pancreatic β-cells is unknown. Here, we report that Septin5 is abundant in rodent and human β-cells, deletion of which dramatically enhances biphasic glucose-stimulated insulin secretion, including in type 2 diabetes (T2D). Super-resolution imaging shows that Septin5 is preferentially assembled in microtubule-plasma membrane contact sites in a microtubule-dependent manner, which provides discrete harbor for secretory granule anchoring. By decreasing the stability of the cortical microtubule meshwork, Septin5 depletion increases insulin granule dynamics and access to the plasma membrane. Analysis of spatiotemporal coupling of fusion events and localized Ca2+ influx through L-type Ca2+ channels show that Septin5 depletion increases releasable granule pool clustering on Ca2+ channels, previously shown to be impaired in T2D, thus rectifying this T2D defect. Hence, inhibition of Septin5 can improve insulin secretion.
Collapse
Affiliation(s)
- Li Xie
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
| | - Fei Kang
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
| | - Tairan Qin
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Youhou Kang
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Tao Liang
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Huanli Xie
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Carol D Froese
- The program in Cell Biology, Department of Biochemistry, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Hong Xie
- The program in Cell Biology, Department of Biochemistry, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Aaron Au
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Christopher M Yip
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - William S Trimble
- The program in Cell Biology, Department of Biochemistry, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Herbert Y Gaisano
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
2
|
Das A, Kunwar A. Septins: Structural Insights, Functional Dynamics, and Implications in Health and Disease. J Cell Biochem 2025; 126:e30660. [PMID: 39324363 DOI: 10.1002/jcb.30660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/03/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
Septins are a class of proteins with diverse and vital roles in cell biology. Structurally, they form hetero-oligomeric complexes and assemble into filaments, contributing to the organization of cells. These filaments act as scaffolds, aiding in processes like membrane remodeling, cytokinesis, and cell motility. Functionally, septins are essential to cell division, playing essential roles in cytokinetic furrow formation and maintaining the structural integrity of the contractile ring. They also regulate membrane trafficking and help organize intracellular organelles. In terms of physiology, septins facilitate cell migration, phagocytosis, and immune responses by maintaining membrane integrity and influencing cytoskeletal dynamics. Septin dysfunction is associated with pathophysiological conditions. Mutations in septin genes have been linked to neurodegenerative diseases, such as hereditary spastic paraplegias, underscoring their significance in neuronal function. Septins also play a role in cancer and infectious diseases, making them potential targets for therapeutic interventions. Septins serve as pivotal components of intracellular signaling networks, engaging with diverse proteins like kinases and phosphatases. By modulating the activity of these molecules, septins regulate vital cellular pathways. This integral role in signaling makes septins central to orchestrating cellular responses to environmental stimuli. This review mainly focuses on the human septins, their structural composition, regulatory functions, and implication in pathophysiological conditions underscores their importance in fundamental cellular biology. Moreover, their potential as therapeutic targets across various diseases further emphasizes their significance.
Collapse
Affiliation(s)
- Aurosikha Das
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Ambarish Kunwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| |
Collapse
|
3
|
Lai HY, Yu KH, Tsai KC, Lee CC, Wang HY, Hsieh YP, Chiang KY, Kuo PL, Huang TT, Hung HY. The first attempt in synthesis, identification, and evaluation of SEPT9 inhibitors on human oral squamous carcinomas. Bioorg Chem 2025; 154:108068. [PMID: 39705938 DOI: 10.1016/j.bioorg.2024.108068] [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: 09/23/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Septin 9 (SEPT9), a GTPase, known as the fourth cytoskeleton, is widely expressed in various cells and tissues. The functions of SEPT9 are partly similar to other cytoskeletons as a structure protein. Further, SEPT9 can interact with other cytoskeletons, participating in actin dynamics and microtubule regulation. SEPT9 is associated with various diseases, such as cancers. Thus, it could be a potential drug target. However, there are no small molecule SEPT9 inhibitors and the only reported septin inhibitor, forchlorfenuron, has no effects on SEPT9 inhibition from our study results. Therefore, the derivatives of forchlorfenuron were synthesized, and their activities were evaluated by a direct SEPT9 inhibition screening platform, followed by localized surface plasmon resonance (LSPR) and cell-based assays. The screening results conveyed that 6b, 8a, and 8b are SEPT9 inhibitors with IC50 values of 91, 99, and 95 μM, respectively. Also, their binding affinities were 4, 18, and 22 μM, respectively, validated through LSPR. Eventually, the SAR concludes that at the para position, small substituents are tolerated, while at the ortho position, a bulky benzene ring substituent can be the best candidate. In cell-based assays, the IC50 of 6a, 8a, and 8b of human oral squamous carcinomas cytotoxicity were 122, 20, and 21 µM, respectively. Additionally, significant suppression of the cell migration and invasion ability was observed with the 8b treatment. The co-localization study revealed that 8b effectively disrupted the structural organization of SEPT9, microtubules, and actins. This is the first article to systematically study SEPT9 inhibitors and their biological properties, hoping to shed some light on septin research.
Collapse
Affiliation(s)
- Hsuan-Yu Lai
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Ko-Hua Yu
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Keng-Chang Tsai
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Ministry of Health and Welfare, National Research Institute of Chinese Medicine, Taipei 112, Taiwan
| | - Chao-Chang Lee
- Ministry of Health and Welfare, National Research Institute of Chinese Medicine, Taipei 112, Taiwan
| | - Han-Yu Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ping Hsieh
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuan-Yi Chiang
- Institute of Oral Medicine, School of Dentistry, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan
| | - Pao-Lin Kuo
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Tze-Ta Huang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Oral Medicine, School of Dentistry, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan; Department of Stomatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan.
| | - Hsin-Yi Hung
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
| |
Collapse
|
4
|
Whitfield M. The annulus: composition, role and importance in sperm flagellum biogenesis and male fertility. Basic Clin Androl 2024; 34:25. [PMID: 39676174 DOI: 10.1186/s12610-024-00241-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/23/2024] [Indexed: 12/17/2024] Open
Abstract
The annulus is an electron-dense ring structure that surrounds the axoneme and compartmentalizes the sperm flagellum into two parts: the midpiece and the principal piece. The function of the annulus as a diffusion barrier in the mature spermatozoon is now well described but its function during spermiogenesis remains unclear. The intriguing spatio-temporal dynamics of the annulus during spermiogenesis and its position at the interface of the two main flagellar compartments have been highlighted for more than 50 years, and suggest a major role in this process. During the last decade, numerous studies contributed in establishing a repertoire of proteins known to be located at the annulus. Mutant mouse models of invalidation of these proteins have provided essential information and clues for novel hypotheses regarding the functions and regulation of this structure. Importantly, the recent identification in humans of homozygous mutations of genes coding for annulus proteins and leading to sterility have reinforced the importance of this ring structure for sperm physiology and male fertility. This review provides a comprehensive description of all the knowledge obtained in the last several years regarding the annulus composition and functions, both in mice and in humans.
Collapse
Affiliation(s)
- Marjorie Whitfield
- Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, Université Grenoble Alpes, Team 'Physiopathology and Pathophysiology of Sperm cells', 38000, Grenoble, France.
| |
Collapse
|
5
|
Cohen D, Fernandez D, Lázaro-Diéguez F, Überheide B, Müsch A. Borg5 restricts contractility and motility in epithelial MDCK cells. J Cell Sci 2024; 137:jcs261705. [PMID: 39503295 PMCID: PMC11698036 DOI: 10.1242/jcs.261705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 10/30/2024] [Indexed: 12/12/2024] Open
Abstract
The Borg (or Cdc42EP) family consists of septin-binding proteins that are known to promote septin-dependent stress fibers and acto-myosin contractility. We show here that epithelial Borg5 (also known as Cdc42EP1) instead limits contractility, cell-cell adhesion tension and motility, as is required for the acquisition of columnar, isotropic cell morphology in mature MDCK monolayers. Borg5 depletion inhibited the development of the lateral F-actin cortex and stimulated microtubule-dependent leading-edge lamellae as well as radial stress fibers and, independently of the basal F-actin phenotype, caused anisotropy of apical surfaces within compacted monolayers. We determined that Borg5 limits colocalization of septin proteins with microtubules, and that like septin 2, Borg5 interacts with the rod-domain of myosin IIA (herein referring to the MYH9 heavy chain). The interaction of myosin IIA with Borg5 was reduced in the presence of septins. Because septins also mediate myosin activation, we propose that Borg5 limits contractility in MDCK cells in part by counteracting septin-associated myosin activity.
Collapse
Affiliation(s)
- David Cohen
- Albert-Einstein College of Medicine, Bronx, NY 10461, USA
| | - Dawn Fernandez
- Albert-Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Beatrix Überheide
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anne Müsch
- Albert-Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
6
|
Robinson BP, Bass NR, Bhakt P, Spiliotis ET. Septin-coated microtubules promote maturation of multivesicular bodies by inhibiting their motility. J Cell Biol 2024; 223:e202308049. [PMID: 38668767 PMCID: PMC11046855 DOI: 10.1083/jcb.202308049] [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: 08/09/2023] [Revised: 02/06/2024] [Accepted: 04/10/2024] [Indexed: 04/29/2024] Open
Abstract
The microtubule cytoskeleton consists of microtubule subsets with distinct compositions of microtubule-associated proteins, which instruct the position and traffic of subcellular organelles. In the endocytic pathway, these microtubule-associated cues are poorly understood. Here, we report that in MDCK cells, endosomes with multivesicular body (MVB) and late endosome (LE) markers localize preferentially to microtubules coated with septin GTPases. Compared with early endosomes, CD63-containing MVBs/LEs are largely immotile on septin-coated microtubules. In vitro reconstitution assays revealed that the motility of isolated GFP-CD63 endosomes is directly inhibited by microtubule-associated septins. Quantification of CD63-positive endosomes containing the early endosome antigen (EEA1), the Rab7 effector and dynein adaptor RILP or Rab27a, showed that intermediary EEA1- and RILP-positive GFP-CD63 preferentially associate with septin-coated microtubules. Septin knockdown enhanced GFP-CD63 motility and decreased the percentage of CD63-positive MVBs/LEs with lysobiphosphatidic acid without impacting the fraction of EEA1-positive CD63. These results suggest that MVB maturation involves immobilization on septin-coated microtubules, which may facilitate multivesiculation and/or organelle-organelle contacts.
Collapse
Affiliation(s)
| | - Naomi R. Bass
- Department of Biology, Drexel University, Philadelphia, PA, USA
| | - Priyanka Bhakt
- Department of Biology, Drexel University, Philadelphia, PA, USA
| | - Elias T. Spiliotis
- Department of Biology, Drexel University, Philadelphia, PA, USA
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
7
|
Hecht M, Alber N, Marhoffer P, Johnsson N, Gronemeyer T. The concerted action of SEPT9 and EPLIN modulates the adhesion and migration of human fibroblasts. Life Sci Alliance 2024; 7:e202201686. [PMID: 38719752 PMCID: PMC11077590 DOI: 10.26508/lsa.202201686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Septins are cytoskeletal proteins that participate in cell adhesion, migration, and polarity establishment. The septin subunit SEPT9 directly interacts with the single LIM domain of epithelial protein lost in neoplasm (EPLIN), an actin-bundling protein. Using a human SEPT9 KO fibroblast cell line, we show that cell adhesion and migration are regulated by the interplay between both proteins. The low motility of SEPT9-depleted cells could be partly rescued by increased levels of EPLIN. The normal organization of actin-related filopodia and stress fibers was directly dependent on the expression level of SEPT9 and EPLIN. Increased levels of SEPT9 and EPLIN enhanced the size of focal adhesions in cell protrusions, correlating with stabilization of actin bundles. Conversely, decreased levels had the opposite effect. Our work thus establishes the interaction between SEPT9 and EPLIN as an important link between the septin and the actin cytoskeleton, influencing cell adhesion, motility, and migration.
Collapse
Affiliation(s)
- Matthias Hecht
- Institute of Molecular Genetics and Cell Biology, James Franck Ring N27, Ulm University, Ulm, Germany
| | - Nane Alber
- Institute of Molecular Genetics and Cell Biology, James Franck Ring N27, Ulm University, Ulm, Germany
| | - Pia Marhoffer
- Institute of Molecular Genetics and Cell Biology, James Franck Ring N27, Ulm University, Ulm, Germany
| | - Nils Johnsson
- Institute of Molecular Genetics and Cell Biology, James Franck Ring N27, Ulm University, Ulm, Germany
| | - Thomas Gronemeyer
- Institute of Molecular Genetics and Cell Biology, James Franck Ring N27, Ulm University, Ulm, Germany
| |
Collapse
|
8
|
Princen K, Van Dooren T, van Gorsel M, Louros N, Yang X, Dumbacher M, Bastiaens I, Coupet K, Dupont S, Cuveliers E, Lauwers A, Laghmouchi M, Vanwelden T, Carmans S, Van Damme N, Duhamel H, Vansteenkiste S, Prerad J, Pipeleers K, Rodiers O, De Ridder L, Claes S, Busschots Y, Pringels L, Verhelst V, Debroux E, Brouwer M, Lievens S, Tavernier J, Farinelli M, Hughes-Asceri S, Voets M, Winderickx J, Wera S, de Wit J, Schymkowitz J, Rousseau F, Zetterberg H, Cummings JL, Annaert W, Cornelissen T, De Winter H, De Witte K, Fivaz M, Griffioen G. Pharmacological modulation of septins restores calcium homeostasis and is neuroprotective in models of Alzheimer's disease. Science 2024; 384:eadd6260. [PMID: 38815015 PMCID: PMC11827694 DOI: 10.1126/science.add6260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 04/04/2024] [Indexed: 06/01/2024]
Abstract
Abnormal calcium signaling is a central pathological component of Alzheimer's disease (AD). Here, we describe the identification of a class of compounds called ReS19-T, which are able to restore calcium homeostasis in cell-based models of tau pathology. Aberrant tau accumulation leads to uncontrolled activation of store-operated calcium channels (SOCCs) by remodeling septin filaments at the cell cortex. Binding of ReS19-T to septins restores filament assembly in the disease state and restrains calcium entry through SOCCs. In amyloid-β and tau-driven mouse models of disease, ReS19-T agents restored synaptic plasticity, normalized brain network activity, and attenuated the development of both amyloid-β and tau pathology. Our findings identify the septin cytoskeleton as a potential therapeutic target for the development of disease-modifying AD treatments.
Collapse
Affiliation(s)
| | | | | | - Nikolaos Louros
- Switch Laboratory, VIB Center for Brain and Disease Research, 3000 Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Xiaojuan Yang
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research and Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | | | | | | | - Shana Dupont
- reMYND NV, Bio-Incubator, 3001 Leuven-Heverlee, Belgium
| | - Eva Cuveliers
- reMYND NV, Bio-Incubator, 3001 Leuven-Heverlee, Belgium
| | | | | | | | - Sofie Carmans
- reMYND NV, Bio-Incubator, 3001 Leuven-Heverlee, Belgium
| | | | - Hein Duhamel
- reMYND NV, Bio-Incubator, 3001 Leuven-Heverlee, Belgium
| | | | - Jovan Prerad
- reMYND NV, Bio-Incubator, 3001 Leuven-Heverlee, Belgium
| | | | | | | | - Sofie Claes
- reMYND NV, Bio-Incubator, 3001 Leuven-Heverlee, Belgium
| | | | | | | | | | - Marinka Brouwer
- Laboratory of Synapse Biology, VIB Center for Brain & Disease Research and KU Leuven Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Sam Lievens
- Cytokine Receptor Lab, VIB Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Jan Tavernier
- Cytokine Receptor Lab, VIB Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | | | | | - Marieke Voets
- reMYND NV, Bio-Incubator, 3001 Leuven-Heverlee, Belgium
| | - Joris Winderickx
- reMYND NV, Bio-Incubator, 3001 Leuven-Heverlee, Belgium
- Functional Biology, Department of Biology, KU Leuven, 3001 Leuven-Heverlee, Belgium
| | - Stefaan Wera
- reMYND NV, Bio-Incubator, 3001 Leuven-Heverlee, Belgium
- ViroVet NV, 3001 Leuven-Heverlee, Belgium
| | - Joris de Wit
- Laboratory of Synapse Biology, VIB Center for Brain & Disease Research and KU Leuven Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB Center for Brain and Disease Research, 3000 Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Frederic Rousseau
- Switch Laboratory, VIB Center for Brain and Disease Research, 3000 Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Jeffrey L. Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas, Las Vegas, NV 89154, USA
| | - Wim Annaert
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research and Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | | | - Hans De Winter
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Koen De Witte
- reMYND NV, Bio-Incubator, 3001 Leuven-Heverlee, Belgium
| | - Marc Fivaz
- reMYND NV, Bio-Incubator, 3001 Leuven-Heverlee, Belgium
| | | |
Collapse
|
9
|
Groh AC, Möller-Kerutt A, Gilhaus K, Höffken V, Nedvetsky P, Kleimann S, Behrens M, Ghosh S, Hansen U, Krahn MP, Ebnet K, Pavenstädt H, Ludwig A, Weide T. PALS1 is a key regulator of the lateral distribution of tight junction proteins in renal epithelial cells. J Cell Sci 2024; 137:jcs261303. [PMID: 38265145 DOI: 10.1242/jcs.261303] [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: 05/03/2023] [Accepted: 12/04/2023] [Indexed: 01/25/2024] Open
Abstract
The evolutionarily conserved apical Crumbs (CRB) complex, consisting of the core components CRB3a (an isoform of CRB3), PALS1 and PATJ, plays a key role in epithelial cell-cell contact formation and cell polarization. Recently, we observed that deletion of one Pals1 allele in mice results in functional haploinsufficiency characterized by renal cysts. Here, to address the role of PALS1 at the cellular level, we generated CRISPR/Cas9-mediated PALS1-knockout MDCKII cell lines. The loss of PALS1 resulted in increased paracellular permeability, indicating an epithelial barrier defect. This defect was associated with a redistribution of several tight junction-associated proteins from bicellular to tricellular contacts. PALS1-dependent localization of tight junction proteins at bicellular junctions required its interaction with PATJ. Importantly, reestablishment of the tight junction belt upon transient F-actin depolymerization or upon Ca2+ removal was strongly delayed in PALS1-deficient cells. Additionally, the cytoskeleton regulator RhoA was redistributed from junctions into the cytosol under PALS1 knockout. Together, our data uncover a critical role of PALS1 in the coupling of tight junction proteins to the F-actin cytoskeleton, which ensures their correct distribution along bicellular junctions and the formation of tight epithelial barrier.
Collapse
Affiliation(s)
- Ann-Christin Groh
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Annika Möller-Kerutt
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Kevin Gilhaus
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Verena Höffken
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Pavel Nedvetsky
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Medical Cell Biology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Simon Kleimann
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Malina Behrens
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Sujasha Ghosh
- School of Biological Sciences and NTU Institute of Structural Biology (NISB), Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore City, Singapore
| | - Uwe Hansen
- University Hospital of Münster, Institute of Musculoskeletal Medicine (IMM), Head Core Facility Electron Microscopy, Domagkstraße 3, 48149 Münster, Germany
| | - Michael P Krahn
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Medical Cell Biology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Klaus Ebnet
- Institute-associated Research Group "Cell adhesion and cell polarity", Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Von-Esmarch-Straße 56, 48149 Münster, Germany
| | - Hermann Pavenstädt
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Alexander Ludwig
- School of Biological Sciences and NTU Institute of Structural Biology (NISB), Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore City, Singapore
| | - Thomas Weide
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| |
Collapse
|
10
|
Mageswaran SK, Grotjahn DA, Zeng X, Barad BA, Medina M, Hoang MH, Dobro MJ, Chang YW, Xu M, Yang WY, Jensen GJ. Nanoscale details of mitochondrial constriction revealed by cryoelectron tomography. Biophys J 2023; 122:3768-3782. [PMID: 37533259 PMCID: PMC10541493 DOI: 10.1016/j.bpj.2023.07.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 08/04/2023] Open
Abstract
Mitochondria adapt to changing cellular environments, stress stimuli, and metabolic demands through dramatic morphological remodeling of their shape, and thus function. Such mitochondrial dynamics is often dependent on cytoskeletal filament interactions. However, the precise organization of these filamentous assemblies remains speculative. Here, we apply cryogenic electron tomography to directly image the nanoscale architecture of the cytoskeletal-membrane interactions involved in mitochondrial dynamics in response to damage. We induced mitochondrial damage via membrane depolarization, a cellular stress associated with mitochondrial fragmentation and mitophagy. We find that, in response to acute membrane depolarization, mammalian mitochondria predominantly organize into tubular morphology that abundantly displays constrictions. We observe long bundles of both unbranched actin and septin filaments enriched at these constrictions. We also observed septin-microtubule interactions at these sites and elsewhere, suggesting that these two filaments guide each other in the cytosolic space. Together, our results provide empirical parameters for the architecture of mitochondrial constriction factors to validate/refine existing models and inform the development of new ones.
Collapse
Affiliation(s)
- Shrawan Kumar Mageswaran
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California; Department of Biophysics and Biochemistry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Danielle Ann Grotjahn
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California; Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California.
| | - Xiangrui Zeng
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Benjamin Asher Barad
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California
| | - Michaela Medina
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California
| | - My Hanh Hoang
- Department of Biophysics and Biochemistry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Yi-Wei Chang
- Department of Biophysics and Biochemistry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Min Xu
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Wei Yuan Yang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Grant J Jensen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California; Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah.
| |
Collapse
|
11
|
Yanda MK, Ciobanu C, Guggino WB, Cebotaru L. CFTR and PC2, partners in the primary cilia in autosomal dominant polycystic kidney disease. Am J Physiol Cell Physiol 2023; 325:C682-C693. [PMID: 37519231 PMCID: PMC10635646 DOI: 10.1152/ajpcell.00197.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
Defects in the primary cilium are associated with autosomal dominant polycystic kidney disease (ADPKD). We used a combination of animal models, Western blotting, and confocal microscopy and discovered that CFTR and polycystin 2 (PC2) are both colocalized to the cilium in normal kidneys, with the levels of both being decreased in cystic epithelia. Cilia were longer in CFTR-null mice and in cystic cells in our ADPKD animal models. We examined septin 2, known to play a role in cilia length, to act as a diffusion barrier and to serve as an enhancer of proliferation. We found that septin 2 protein levels were upregulated and colocalized strongly with CFTR in cystic cells. Application of VX-809, the CFTR corrector, restored CFTR and PC2 toward normal in the cilia, decreased the protein levels of septin 2, and drastically reduced septin 2 colocalization with CFTR. Our data suggest that CFTR is present in the cilia and plays a role there, perhaps through its conductance of Cl-. We also postulate that septin 2 is important for localizing CFTR to the apical membrane in cystic epithelia.NEW & NOTEWORTHY CFTR is present in the primary cilia together with polycystin 2 (PC2). Ablation of CFTR makes cilia longer suggesting that CFTR plays a role there, perhaps through its conductance of Cl.
Collapse
Affiliation(s)
- Murali K Yanda
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Cristian Ciobanu
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - William B Guggino
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Liudmila Cebotaru
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
12
|
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
|
13
|
Fu L, Wang X, Yang Y, Chen M, Kuerban A, Liu H, Dong Y, Cai Q, Ma M, Wu X. Septin11 promotes hepatocellular carcinoma cell motility by activating RhoA to regulate cytoskeleton and cell adhesion. Cell Death Dis 2023; 14:280. [PMID: 37080972 PMCID: PMC10119145 DOI: 10.1038/s41419-023-05726-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 02/25/2023] [Accepted: 03/09/2023] [Indexed: 04/22/2023]
Abstract
Septins as GTPases in the cytoskeleton, are linked to a broad spectrum of cellular functions, including cell migration and the progression of hepatocellular carcinoma (HCC). However, roles of SEPT11, the new member of septin, have been hardly understood in HCC. In the study, the clinical significance and biological function of SEPT11 in HCC was explored. SEPT11 was screened out by combining ATAC-seq with mRNA-seq. Role of SEPT11 in HCC was further investigated by using overexpression, shRNA and CRISPR/Cas9-mediated SEPT11-knockout cells or in vivo models. We found RNA-seq and ATAC-seq highlights LncRNA AY927503 (AY) induced SEPT11 transcription, resulting in Rho GTPase activation and cytoskeleton actin aggregation. The GTP-binding protein SEPT11 is thus considered, as a downstream factor of AY, highly expressed in various tumors, including HCC, and associated with poor prognosis of the patients. In vitro, SEPT11 overexpression promotes the migration and invasion of HCC cells, while SEPT11-knockout inhibits migration and invasion. In vivo, SEPT11-overexpressed HCC cells show high metastasis incidents but don't significantly affect proliferation. Meanwhile, we found SEPT11 targets RhoA, thereby regulating cytoskeleton rearrangement and abnormal cell adhesion through ROCK1/cofilin and FAK/paxillin signaling pathways, promoting invasion and migration of HCC. Further, we found SEPT11 facilitates the binding of GEF-H1 to RhoA, which enhances the activity of RhoA. Overall, our study confirmed function of SEPT11 in promoting metastasis in HCC, and preliminarily explored its related molecular mechanism. SEPT11 acts as an oncogene in HCC, also draws further interest regarding its clinical application as a potential therapeutic target.
Collapse
Affiliation(s)
- Lisheng Fu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Department of Cardiology of Huadong Hospital Affiliated to Fudan University, Fudan University, 200032, Shanghai, People's Republic of China
| | - Xiaoyan Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 200032, Shanghai, People's Republic of China
| | - Ying Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Department of Cardiology of Huadong Hospital Affiliated to Fudan University, Fudan University, 200032, Shanghai, People's Republic of China
| | - MeiHua Chen
- NHC Key Laboratory of Glycoconjugates, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 200032, Shanghai, People's Republic of China
| | - Adilijiang Kuerban
- Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Fudan University, 200040, Shanghai, People's Republic of China
| | - Haojie Liu
- NHC Key Laboratory of Glycoconjugates, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 200032, Shanghai, People's Republic of China
| | - Yiwei Dong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Department of Cardiology of Huadong Hospital Affiliated to Fudan University, Fudan University, 200032, Shanghai, People's Republic of China
| | - QianQian Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Department of Cardiology of Huadong Hospital Affiliated to Fudan University, Fudan University, 200032, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 201318, Shanghai, China.
| | - Mingzhe Ma
- Department of Gastric Surgery, Shanghai Cancer Center of Fudan University, 200032, Shanghai, People's Republic of China.
| | - XingZhong Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Department of Cardiology of Huadong Hospital Affiliated to Fudan University, Fudan University, 200032, Shanghai, People's Republic of China.
| |
Collapse
|
14
|
Kho M, Hladyshau S, Tsygankov D, Nie S. Coordinated regulation of Cdc42ep1, actin, and septin filaments during neural crest cell migration. Front Cell Dev Biol 2023; 11:1106595. [PMID: 36923257 PMCID: PMC10009165 DOI: 10.3389/fcell.2023.1106595] [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: 11/23/2022] [Accepted: 02/15/2023] [Indexed: 03/02/2023] Open
Abstract
The septin cytoskeleton has been demonstrated to interact with other cytoskeletal components to regulate various cellular processes, including cell migration. However, the mechanisms of how septin regulates cell migration are not fully understood. In this study, we use the highly migratory neural crest cells of frog embryos to examine the role of septin filaments in cell migration. We found that septin filaments are required for the proper migration of neural crest cells by controlling both the speed and the direction of cell migration. We further determined that septin filaments regulate these features of cell migration by interacting with actin stress fibers. In neural crest cells, septin filaments co-align with actin stress fibers, and the loss of septin filaments leads to impaired stability and contractility of actin stress fibers. In addition, we showed that a partial loss of septin filaments leads to drastic changes in the orientations of newly formed actin stress fibers, suggesting that septin filaments help maintain the persistent orientation of actin stress fibers during directed cell migration. Lastly, our study revealed that these activities of septin filaments depend on Cdc42ep1, which colocalizes with septin filaments in the center of neural crest cells. Cdc42ep1 interacts with septin filaments in a reciprocal manner, with septin filaments recruiting Cdc42ep1 to the cell center and Cdc42ep1 supporting the formation of septin filaments.
Collapse
Affiliation(s)
- Mary Kho
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Siarhei Hladyshau
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Denis Tsygankov
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Shuyi Nie
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
15
|
Merenich D, Nakos K, Pompan T, Donovan SJ, Gill A, Patel P, Spiliotis ET, Myers KA. Septins guide noncentrosomal microtubules to promote focal adhesion disassembly in migrating cells. Mol Biol Cell 2022; 33:ar40. [PMID: 35274967 PMCID: PMC9282018 DOI: 10.1091/mbc.e21-06-0334] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/07/2022] [Accepted: 03/04/2022] [Indexed: 11/30/2022] Open
Abstract
Endothelial cell migration is critical for vascular angiogenesis and is compromised to facilitate tumor metastasis. The migratory process requires the coordinated assembly and disassembly of focal adhesions (FA), actin, and microtubules (MT). MT dynamics at FAs deliver vesicular cargoes and enhance actomyosin contractility to promote FA turnover and facilitate cell advance. Noncentrosomal (NC) MTs regulate FA dynamics and are sufficient to drive cell polarity, but how NC MTs target FAs to control FA turnover is not understood. Here, we show that Rac1 induces the assembly of FA-proximal septin filaments that promote NC MT growth into FAs and inhibit mitotic centromere-associated kinesin (MCAK)-associated MT disassembly, thereby maintaining intact MT plus ends proximal to FAs. Septin-associated MT rescue is coupled with accumulation of Aurora-A kinase and cytoplasmic linker-associated protein (CLASP) localization to the MT between septin and FAs. In this way, NC MTs are strategically positioned to undergo MCAK- and CLASP-regulated bouts of assembly and disassembly into FAs, thereby regulating FA turnover and cell migration.
Collapse
Affiliation(s)
- Daniel Merenich
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | | | - Taylor Pompan
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | - Samantha J. Donovan
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | - Amrik Gill
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | - Pranav Patel
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| | | | - Kenneth A. Myers
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, PA 19104
| |
Collapse
|
16
|
Fischer M, Frank D, Rösler R, Johnsson N, Gronemeyer T. Biochemical Characterization of a Human Septin Octamer. Front Cell Dev Biol 2022; 10:771388. [PMID: 35309913 PMCID: PMC8928218 DOI: 10.3389/fcell.2022.771388] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 02/14/2022] [Indexed: 11/25/2022] Open
Abstract
Septins are part of the cytoskeleton and polymerize into non-polar filaments of heteromeric hexamers or octamers. They belong to the class of P-loop GTPases but the roles of GTP binding and hydrolysis on filament formation and dynamics are not well understood. The basic human septin building block is the septin rod, a hetero-octamer composed of SEPT2, SEPT6, SEPT7, and SEPT9 with a stoichiometry of 2:2:2:2 (2-6-7-9-9-7-6-2). Septin rods polymerize by end-to-end and lateral joining into linear filaments and higher ordered structures such as rings, sheets, and gauzes. We purified a recombinant human septin octamer from E. coli for in vitro experimentation that is able to polymerize into filaments. We could show that the C-terminal region of the central SEPT9 subunit contributes to filament formation and that the human septin rod decreases the rate of in vitro actin polymerization. We provide further first kinetic data on the nucleotide uptake- and exchange properties of human hexameric and octameric septin rods. We could show that nucleotide uptake prior to hydrolysis is a dynamic process and that a bound nucleotide is exchangeable. However, the hydrolyzed γ-phosphate is not released from the native protein complex. We consequently propose that GTP hydrolysis in human septins does not follow the typical mechanism known from other small GTPases.
Collapse
Affiliation(s)
- Martin Fischer
- Institute of Molecular Genetics and Cell Biology, Ulm University, Ulm, Germany
| | - Dominik Frank
- Institute of Molecular Genetics and Cell Biology, Ulm University, Ulm, Germany
| | - Reinhild Rösler
- Core Unit Mass Spectrometry and Proteomics, Ulm University, Ulm, Germany
| | - Nils Johnsson
- Institute of Molecular Genetics and Cell Biology, Ulm University, Ulm, Germany
| | - Thomas Gronemeyer
- Institute of Molecular Genetics and Cell Biology, Ulm University, Ulm, Germany
| |
Collapse
|
17
|
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: 14] [Impact Index Per Article: 3.5] [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
|
18
|
Iv F, Martins CS, Castro-Linares G, Taveneau C, Barbier P, Verdier-Pinard P, Camoin L, Audebert S, Tsai FC, Ramond L, Llewellyn A, Belhabib M, Nakazawa K, Di Cicco A, Vincentelli R, Wenger J, Cabantous S, Koenderink GH, Bertin A, Mavrakis M. Insights into animal septins using recombinant human septin octamers with distinct SEPT9 isoforms. J Cell Sci 2021; 134:jcs258484. [PMID: 34350965 DOI: 10.1242/jcs.258484] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/02/2021] [Indexed: 01/22/2023] Open
Abstract
Septin GTP-binding proteins contribute essential biological functions that range from the establishment of cell polarity to animal tissue morphogenesis. Human septins in cells form hetero-octameric septin complexes containing the ubiquitously expressed SEPT9 subunit (also known as SEPTIN9). Despite the established role of SEPT9 in mammalian development and human pathophysiology, biochemical and biophysical studies have relied on monomeric SEPT9, thus not recapitulating its native assembly into hetero-octameric complexes. We established a protocol that enabled, for the first time, the isolation of recombinant human septin octamers containing distinct SEPT9 isoforms. A combination of biochemical and biophysical assays confirmed the octameric nature of the isolated complexes in solution. Reconstitution studies showed that octamers with either a long or a short SEPT9 isoform form filament assemblies, and can directly bind and cross-link actin filaments, raising the possibility that septin-decorated actin structures in cells reflect direct actin-septin interactions. Recombinant SEPT9-containing octamers will make it possible to design cell-free assays to dissect the complex interactions of septins with cell membranes and the actin and microtubule cytoskeleton.
Collapse
Affiliation(s)
- Francois Iv
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Carla Silva Martins
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Gerard Castro-Linares
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Cyntia Taveneau
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire Physico Chimie Curie, 75005 Paris, France
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Australia; Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, 3800 Clayton, Australia
| | - Pascale Barbier
- Aix-Marseille Univ, CNRS, UMR 7051, Institut de Neurophysiopathologie (INP), 13005 Marseille, France
| | - Pascal Verdier-Pinard
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, 13009 Marseille, France
| | - Luc Camoin
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France
| | - Stéphane Audebert
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France
| | - Feng-Ching Tsai
- Department of Living Matter, AMOLF, 1098 XG Amsterdam, The Netherlands
| | - Laurie Ramond
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Alex Llewellyn
- 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
| | - Koyomi Nakazawa
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire Physico Chimie Curie, 75005 Paris, France
| | - Aurélie Di Cicco
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire Physico Chimie Curie, 75005 Paris, France
| | - Renaud Vincentelli
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS UMR7257, Aix Marseille Univ, 13009 Marseille, France
| | - Jerome Wenger
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| | - Stéphanie Cabantous
- Centre de Recherche en Cancérologie de Toulouse (CRCT), Inserm, Université Paul Sabatier-Toulouse III, CNRS, 31037 Toulouse, France
| | - Gijsje H Koenderink
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
- Department of Living Matter, AMOLF, 1098 XG Amsterdam, The Netherlands
| | - Aurélie Bertin
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire Physico Chimie Curie, 75005 Paris, France
| | - Manos Mavrakis
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Marseille, 13013 Marseille, France
| |
Collapse
|
19
|
Salameh J, Cantaloube I, Benoit B, Poüs C, Baillet A. Cdc42 and its BORG2 and BORG3 effectors control the subcellular localization of septins between actin stress fibers and microtubules. Curr Biol 2021; 31:4088-4103.e5. [PMID: 34329591 DOI: 10.1016/j.cub.2021.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 05/28/2021] [Accepted: 07/02/2021] [Indexed: 01/22/2023]
Abstract
Cell resistance to taxanes involves several complementary mechanisms, among which septin relocalization from actin stress fibers to microtubules plays an early role. By investigating the molecular mechanism underlying this relocalization, we found that acute paclitaxel treatment triggers the release from stress fibers and subsequent proteasome-mediated degradation of binder of Rho GTPases 2 (BORG2)/Cdc42 effector protein 3 (Cdc42EP3) and to a lesser extent of BORG3/Cdc42EP5, two Cdc42 effectors that link septins to actin in interphase cells. BORG2 or BORG3 silencing not only caused septin detachment from stress fibers but also mimicked the effects of paclitaxel by triggering both septin relocalization to microtubules and significant drug resistance. Conversely, BORG2 or BORG3 overexpression retained septins on actin fibers even after paclitaxel treatment, without affecting paclitaxel sensitivity. We found that drug-induced inhibition of Cdc42 resulted in a drop in BORG2 level and in the relocalization of septins to microtubules. Accordingly, although septins relocalized when overexpressing an inactive mutant of Cdc42, the expression of a constitutively active mutant acted locally at actin stress fibers to prevent septin release, even after paclitaxel treatment. These findings reveal the role of Cdc42 upstream of BORG2 and BORG3 in controlling the interplay between septins, actin fibers, and microtubules in basal condition and in response to taxanes.
Collapse
Affiliation(s)
- Joëlle Salameh
- INSERM UMR-S 1193, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| | - Isabelle Cantaloube
- INSERM UMR-S 1193, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| | - Béatrice Benoit
- INSERM UMR-S 1193, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| | - Christian Poüs
- INSERM UMR-S 1193, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France; Laboratoire de Biochimie-Hormonologie, Hôpital Antoine Béclère, AP-HP, Clamart, France.
| | - Anita Baillet
- INSERM UMR-S 1193, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France.
| |
Collapse
|
20
|
Spiliotis ET, Kesisova IA. Spatial regulation of microtubule-dependent transport by septin GTPases. Trends Cell Biol 2021; 31:979-993. [PMID: 34253430 DOI: 10.1016/j.tcb.2021.06.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 01/21/2023]
Abstract
The intracellular long-range transport of membrane vesicles and organelles is mediated by microtubule motors (kinesins, dynein) which move cargo with spatiotemporal accuracy and efficiency. How motors navigate the microtubule network and coordinate their activity on membrane cargo are fundamental but poorly understood questions. New studies show that microtubule-dependent membrane traffic is spatially controlled by septins - a unique family of multimerizing GTPases that associate with microtubules and membrane organelles. We review how septins selectively regulate motor interactions with microtubules and membrane cargo. We posit that septins provide a novel traffic code that specifies the movement and directionality of select motor-cargo complexes on distinct microtubule tracks.
Collapse
Affiliation(s)
- Elias T Spiliotis
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA.
| | - Ilona A Kesisova
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| |
Collapse
|
21
|
Wang X, Wang W, Wang X, Wang M, Zhu L, Garba F, Fu C, Zieger B, Liu X, Liu X, Yao X. The septin complex links the catenin complex to the actin cytoskeleton for establishing epithelial cell polarity. J Mol Cell Biol 2021; 13:395-408. [PMID: 34143183 PMCID: PMC8436676 DOI: 10.1093/jmcb/mjab036] [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: 11/25/2020] [Revised: 03/16/2021] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
Cell polarity is essential for spatially regulating of physiological processes in metazoans by which hormonal stimulation‒secretion coupling is precisely coupled for tissue homeostasis and organ communications. However, the molecular mechanisms underlying epithelial cell polarity establishment remain elusive. Here, we show that septin cytoskeleton interacts with catenin complex to organize a functional domain to separate apical from basal membranes in polarized epithelial cells. Using polarized epithelial cell monolayer as a model system with transepithelial electrical resistance as functional readout, our studies show that septins are essential for epithelial cell polarization. Our proteomic analyses discovered a novel septin‒catenin complex during epithelial cell polarization. The functional relevance of septin‒catenin complex was then examined in three-dimensional (3D) culture in which suppression of septins resulted in deformation of apical lumen in cysts, a hallmark seen in polarity-deficient 3D cultures and animals. Mechanistically, septin cytoskeleton stabilizes the association of adherens catenin complex with actin cytoskeleton, and depletion or disruption of septin cytoskeleton liberates adherens junction and polarity complexes into the cytoplasm. Together, these findings reveal a previously unrecognized role for septin cytoskeleton in the polarization of the apical‒basal axis and lumen formation in polarized epithelial cells.
Collapse
Affiliation(s)
- Xueying Wang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China.,Keck Center for Organoids Plasticity Control, Atlanta, GA 30310, USA
| | - Wenwen Wang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China.,Anhui Key Laboratory for Cellular Dynamics & Chemical Biology and CAS Center for Excellence in Molecular Cell Science, Hefei 230027, China.,Keck Center for Organoids Plasticity Control, Atlanta, GA 30310, USA
| | - Xiwei Wang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China.,Keck Center for Organoids Plasticity Control, Atlanta, GA 30310, USA
| | - Ming Wang
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China
| | - Lijuan Zhu
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China.,Anhui Key Laboratory for Cellular Dynamics & Chemical Biology and CAS Center for Excellence in Molecular Cell Science, Hefei 230027, China
| | - Fatima Garba
- Keck Center for Organoids Plasticity Control, Atlanta, GA 30310, USA
| | - Chuanhai Fu
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China.,Anhui Key Laboratory for Cellular Dynamics & Chemical Biology and CAS Center for Excellence in Molecular Cell Science, Hefei 230027, China
| | - Barbara Zieger
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Xu Liu
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China.,Keck Center for Organoids Plasticity Control, Atlanta, GA 30310, USA
| | - Xing Liu
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China.,Anhui Key Laboratory for Cellular Dynamics & Chemical Biology and CAS Center for Excellence in Molecular Cell Science, Hefei 230027, China
| | - Xuebiao Yao
- MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China.,Anhui Key Laboratory for Cellular Dynamics & Chemical Biology and CAS Center for Excellence in Molecular Cell Science, Hefei 230027, China
| |
Collapse
|
22
|
Spiliotis ET, McMurray MA. Masters of asymmetry - lessons and perspectives from 50 years of septins. Mol Biol Cell 2021; 31:2289-2297. [PMID: 32991244 PMCID: PMC7851956 DOI: 10.1091/mbc.e19-11-0648] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Septins are a unique family of GTPases, which were discovered 50 years ago as essential genes for the asymmetric cell shape and division of budding yeast. Septins assemble into filamentous nonpolar polymers, which associate with distinct membrane macrodomains and subpopulations of actin filaments and microtubules. While structurally a cytoskeleton-like element, septins function predominantly as spatial regulators of protein localization and interactions. Septin scaffolds and barriers have provided a long-standing paradigm for the generation and maintenance of asymmetry in cell membranes. Septins also promote asymmetry by regulating the spatial organization of the actin and microtubule cytoskeleton, and biasing the directionality of membrane traffic. In this 50th anniversary perspective, we highlight how septins have conserved and adapted their roles as effectors of membrane and cytoplasmic asymmetry across fungi and animals. We conclude by outlining principles of septin function as a module of symmetry breaking, which alongside the monomeric small GTPases provides a core mechanism for the biogenesis of molecular asymmetry and cell polarity.
Collapse
Affiliation(s)
| | - Michael A McMurray
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
23
|
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
|
24
|
Hu C, Feng P, Yang Q, Xiao L. Clinical and Neurobiological Aspects of TAO Kinase Family in Neurodevelopmental Disorders. Front Mol Neurosci 2021; 14:655037. [PMID: 33867937 PMCID: PMC8044823 DOI: 10.3389/fnmol.2021.655037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/04/2021] [Indexed: 12/20/2022] Open
Abstract
Despite the complexity of neurodevelopmental disorders (NDDs), from their genotype to phenotype, in the last few decades substantial progress has been made in understanding their pathophysiology. Recent accumulating evidence shows the relevance of genetic variants in thousand and one (TAO) kinases as major contributors to several NDDs. Although it is well-known that TAO kinases are a highly conserved family of STE20 kinase and play important roles in multiple biological processes, the emerging roles of TAO kinases in neurodevelopment and NDDs have yet to be intensively discussed. In this review article, we summarize the potential roles of the TAO kinases based on structural and biochemical analyses, present the genetic data from clinical investigations, and assess the mechanistic link between the mutations of TAO kinases, neuropathology, and behavioral impairment in NDDs. We then offer potential perspectives from basic research to clinical therapies, which may contribute to fully understanding how TAO kinases are involved in NDDs.
Collapse
Affiliation(s)
- Chun Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, South China Normal University, Guangzhou, China.,Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Pan Feng
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, South China Normal University, Guangzhou, China.,Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Qian Yang
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, South China Normal University, Guangzhou, China.,Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Lin Xiao
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, South China Normal University, Guangzhou, China.,Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| |
Collapse
|
25
|
Wang M, Du Y, Gao S, Wang Z, Qu P, Gao Y, Wang J, Liu Z, Zhang J, Zhang Y, Qing S, Wang Y. Sperm-borne miR-202 targets SEPT7 and regulates first cleavage of bovine embryos via cytoskeletal remodeling. Development 2021; 148:dev.189670. [PMID: 33472846 DOI: 10.1242/dev.189670] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 01/08/2021] [Indexed: 01/28/2023]
Abstract
In mammals, sperm-borne regulators can be transferred to oocytes during fertilization and have different effects on the formation of pronuclei, the first cleavage of zygotes, the development of preimplantation embryos and even the metabolism of individuals after birth. The regulatory role of sperm microRNAs (miRNAs) in the development of bovine preimplantation embryos has not been reported in detail. By constructing and screening miRNA expression libraries, we found that miR-202 was highly enriched in bovine sperm. As a target gene of miR-202, co-injection of SEPT7 siRNA can partially reverse the accelerated first cleavage of bovine embryos caused by miR-202 inhibitor. In addition, both a miR-202 mimic and SEPT7 siRNA delayed the first cleavage of somatic cell nuclear transfer (SCNT) embryos, suggesting that miR-202-SEPT7 mediates the delay of first cleavage of bovine embryos. By further exploring the relationship between miR-202/SEPT7, HDAC6 and acetylated α-tubulin during embryonic development, we investigated how sperm-borne miR-202 regulates the first cleavage process of bovine embryos by SEPT7 and demonstrate the potential of sperm-borne miRNAs to improve the efficiency of SCNT.
Collapse
Affiliation(s)
- Mengyun Wang
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling 712100, Shaanxi Province, China.,School of Life Science and Technology, Harbin Institute of Technology, Science Park of Harbin Institute of Technology, Harbin 150000, China
| | - Yue Du
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling 712100, Shaanxi Province, China.,Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Song Gao
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling 712100, Shaanxi Province, China
| | - Zheng Wang
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling 712100, Shaanxi Province, China
| | - Pengxiang Qu
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling 712100, Shaanxi Province, China
| | - Yang Gao
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling 712100, Shaanxi Province, China
| | - Jingyi Wang
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling 712100, Shaanxi Province, China
| | - Zhengqi Liu
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling 712100, Shaanxi Province, China
| | - Jingcheng Zhang
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling 712100, Shaanxi Province, China
| | - Yong Zhang
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling 712100, Shaanxi Province, China
| | - Suzhu Qing
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling 712100, Shaanxi Province, China
| | - Yongsheng Wang
- College of Veterinary Medicine, Northwest A&F University, Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling 712100, Shaanxi Province, China
| |
Collapse
|
26
|
Dulal N, Rogers AM, Proko R, Bieger BD, Liyanage R, Krishnamurthi VR, Wang Y, Egan MJ. Turgor-dependent and coronin-mediated F-actin dynamics drive septin disc-to-ring remodeling in the blast fungus Magnaporthe oryzae. J Cell Sci 2021; 134:jcs.251298. [PMID: 33414165 DOI: 10.1242/jcs.251298] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 12/16/2020] [Indexed: 12/16/2022] Open
Abstract
The fungus Magnaporthe oryzae uses a specialized pressure-generating infection cell called an appressorium to break into rice leaves and initiate disease. Appressorium functionality is dependent on the formation of a cortical septin ring during its morphogenesis, but precisely how this structure assembles is unclear. Here, we show that F-actin rings are recruited to the circumference of incipient septin disc-like structures in a pressure-dependent manner, and that this is necessary for their contraction and remodeling into rings. We demonstrate that the structural integrity of these incipient septin discs requires both an intact F-actin and microtubule cytoskeleton and provide fundamental new insight into their functional organization within the appressorium. Lastly, using proximity-dependent labeling, we identify the actin modulator coronin as a septin-proximal protein and show that F-actin-mediated septin disc-to-ring remodeling is perturbed in the genetic absence of coronin. Taken together, our findings provide new insight into the dynamic remodeling of infection-specific higher-order septin structures in a globally significant fungal plant pathogen.
Collapse
Affiliation(s)
- Nawaraj Dulal
- Department of Entomology and Plant Pathology, University of Arkansas Systems Division of Agriculture, Fayetteville, AR 72701, USA
| | - Audra Mae Rogers
- Department of Entomology and Plant Pathology, University of Arkansas Systems Division of Agriculture, Fayetteville, AR 72701, USA
| | - Rinalda Proko
- Department of Entomology and Plant Pathology, University of Arkansas Systems Division of Agriculture, Fayetteville, AR 72701, USA.,Cell and Molecular Biology graduate program, University of Arkansas, Fayetteville, AR 72701, USA
| | - Baronger Dowell Bieger
- Department of Entomology and Plant Pathology, University of Arkansas Systems Division of Agriculture, Fayetteville, AR 72701, USA.,Cell and Molecular Biology graduate program, University of Arkansas, Fayetteville, AR 72701, USA
| | - Rohana Liyanage
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA
| | | | - Yong Wang
- Cell and Molecular Biology graduate program, University of Arkansas, Fayetteville, AR 72701, USA.,Department of Physics, University of Arkansas, Fayetteville, AR 72701, USA.,Microelectronics-Photonics graduate program, University of Arkansas, Fayetteville, AR 72701, USA
| | - Martin John Egan
- Department of Entomology and Plant Pathology, University of Arkansas Systems Division of Agriculture, Fayetteville, AR 72701, USA .,Cell and Molecular Biology graduate program, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
27
|
Protein Kinase A-Mediated Septin7 Phosphorylation Disrupts Septin Filaments and Ciliogenesis. Cells 2021; 10:cells10020361. [PMID: 33572403 PMCID: PMC7916176 DOI: 10.3390/cells10020361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 01/22/2023] Open
Abstract
Septins are GTP-binding proteins that form heteromeric filaments for proper cell growth and migration. Among the septins, septin7 (SEPT7) is an important component of all septin filaments. Here we show that protein kinase A (PKA) phosphorylates SEPT7 at Thr197, thus disrupting septin filament dynamics and ciliogenesis. The Thr197 residue of SEPT7, a PKA phosphorylating site, was conserved among different species. Treatment with cAMP or overexpression of PKA catalytic subunit (PKACA2) induced SEPT7 phosphorylation, followed by disruption of septin filament formation. Constitutive phosphorylation of SEPT7 at Thr197 reduced SEPT7‒SEPT7 interaction, but did not affect SEPT7‒SEPT6‒SEPT2 or SEPT4 interaction. Moreover, we noted that SEPT7 interacted with PKACA2 via its GTP-binding domain. Furthermore, PKA-mediated SEPT7 phosphorylation disrupted primary cilia formation. Thus, our data uncover the novel biological function of SEPT7 phosphorylation in septin filament polymerization and primary cilia formation.
Collapse
|
28
|
Kesisova IA, Robinson BP, Spiliotis ET. A septin GTPase scaffold of dynein-dynactin motors triggers retrograde lysosome transport. J Cell Biol 2021; 220:211663. [PMID: 33416861 PMCID: PMC7802366 DOI: 10.1083/jcb.202005219] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/22/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
The metabolic and signaling functions of lysosomes depend on their intracellular positioning and trafficking, but the underlying mechanisms are little understood. Here, we have discovered a novel septin GTPase-based mechanism for retrograde lysosome transport. We found that septin 9 (SEPT9) associates with lysosomes, promoting the perinuclear localization of lysosomes in a Rab7-independent manner. SEPT9 targeting to mitochondria and peroxisomes is sufficient to recruit dynein and cause perinuclear clustering. We show that SEPT9 interacts with both dynein and dynactin through its GTPase domain and N-terminal extension, respectively. Strikingly, SEPT9 associates preferentially with the dynein intermediate chain (DIC) in its GDP-bound state, which favors dimerization and assembly into septin multimers. In response to oxidative cell stress induced by arsenite, SEPT9 localization to lysosomes is enhanced, promoting the perinuclear clustering of lysosomes. We posit that septins function as GDP-activated scaffolds for the cooperative assembly of dynein-dynactin, providing an alternative mechanism of retrograde lysosome transport at steady state and during cellular adaptation to stress.
Collapse
|
29
|
Marttinen M, Ferreira CB, Paldanius KMA, Takalo M, Natunen T, Mäkinen P, Leppänen L, Leinonen V, Tanigaki K, Kang G, Hiroi N, Soininen H, Rilla K, Haapasalo A, Hiltunen M. Presynaptic Vesicle Protein SEPTIN5 Regulates the Degradation of APP C-Terminal Fragments and the Levels of Aβ. Cells 2020; 9:cells9112482. [PMID: 33203136 PMCID: PMC7696542 DOI: 10.3390/cells9112482] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/05/2020] [Accepted: 11/10/2020] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by aberrant amyloid-β (Aβ) and hyperphosphorylated tau aggregation. We have previously investigated the involvement of SEPTIN family members in AD-related cellular processes and discovered a role for SEPTIN8 in the sorting and accumulation of β-secretase. Here, we elucidated the potential role of SEPTIN5, an interaction partner of SEPTIN8, in the cellular processes relevant for AD, including amyloid precursor protein (APP) processing and the generation of Aβ. The in vitro and in vivo studies both revealed that the downregulation of SEPTIN5 reduced the levels of APP C-terminal fragments (APP CTFs) and Aβ in neuronal cells and in the cortex of Septin5 knockout mice. Mechanistic elucidation revealed that the downregulation of SEPTIN5 increased the degradation of APP CTFs, without affecting the secretory pathway-related trafficking or the endocytosis of APP. Furthermore, we found that the APP CTFs were degraded, to a large extent, via the autophagosomal pathway and that the downregulation of SEPTIN5 enhanced autophagosomal activity in neuronal cells as indicated by altered levels of key autophagosomal markers. Collectively, our data suggest that the downregulation of SEPTIN5 increases the autophagy-mediated degradation of APP CTFs, leading to reduced levels of Aβ in neuronal cells.
Collapse
Affiliation(s)
- Mikael Marttinen
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland; (M.M.); (K.M.A.P.); (M.T.); (T.N.); (P.M.); (L.L.); (K.R.)
| | - Catarina B. Ferreira
- Instituto de Medicina Molecular—João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal;
| | - Kaisa M. A. Paldanius
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland; (M.M.); (K.M.A.P.); (M.T.); (T.N.); (P.M.); (L.L.); (K.R.)
| | - Mari Takalo
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland; (M.M.); (K.M.A.P.); (M.T.); (T.N.); (P.M.); (L.L.); (K.R.)
| | - Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland; (M.M.); (K.M.A.P.); (M.T.); (T.N.); (P.M.); (L.L.); (K.R.)
| | - Petra Mäkinen
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland; (M.M.); (K.M.A.P.); (M.T.); (T.N.); (P.M.); (L.L.); (K.R.)
| | - Luukas Leppänen
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland; (M.M.); (K.M.A.P.); (M.T.); (T.N.); (P.M.); (L.L.); (K.R.)
| | - Ville Leinonen
- Institute of Clinical Medicine–Neurosurgery, University of Eastern Finland, 70210 Kuopio, Finland;
- Neurology of Neuro Center Kuopio University Hospital, 70210 Kuopio, Finland
| | - Kenji Tanigaki
- Research Institute, Shiga Medical Center, Shiga 524-8524, Japan;
| | - Gina Kang
- Department of Pharmacology, Department of Integrative and Systems Physiology, Department of Cell Systems and Anatomy, Department of Psychiatry, University of Texas Health Science Center, San Antonio, TX 77030, USA; (G.K.); (N.H.)
| | - Noboru Hiroi
- Department of Pharmacology, Department of Integrative and Systems Physiology, Department of Cell Systems and Anatomy, Department of Psychiatry, University of Texas Health Science Center, San Antonio, TX 77030, USA; (G.K.); (N.H.)
| | - Hilkka Soininen
- Institute of Clinical Medicine–Neurology, University of Eastern Finland, 70210 Kuopio, Finland;
| | - Kirsi Rilla
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland; (M.M.); (K.M.A.P.); (M.T.); (T.N.); (P.M.); (L.L.); (K.R.)
| | - Annakaisa Haapasalo
- A.I Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
- Correspondence: (A.H.); (M.H.); Tel.: +358-40-355-2768 (A.H.); +358-40-355-2014 (M.H.)
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, 70210 Kuopio, Finland; (M.M.); (K.M.A.P.); (M.T.); (T.N.); (P.M.); (L.L.); (K.R.)
- Correspondence: (A.H.); (M.H.); Tel.: +358-40-355-2768 (A.H.); +358-40-355-2014 (M.H.)
| |
Collapse
|
30
|
Maloney SM, Hoover CA, Morejon-Lasso LV, Prosperi JR. Mechanisms of Taxane Resistance. Cancers (Basel) 2020; 12:E3323. [PMID: 33182737 PMCID: PMC7697134 DOI: 10.3390/cancers12113323] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
The taxane family of chemotherapy drugs has been used to treat a variety of mostly epithelial-derived tumors and remain the first-line treatment for some cancers. Despite the improved survival time and reduction of tumor size observed in some patients, many have no response to the drugs or develop resistance over time. Taxane resistance is multi-faceted and involves multiple pathways in proliferation, apoptosis, metabolism, and the transport of foreign substances. In this review, we dive deeper into hypothesized resistance mechanisms from research during the last decade, with a focus on the cancer types that use taxanes as first-line treatment but frequently develop resistance to them. Furthermore, we will discuss current clinical inhibitors and those yet to be approved that target key pathways or proteins and aim to reverse resistance in combination with taxanes or individually. Lastly, we will highlight taxane response biomarkers, specific genes with monitored expression and correlated with response to taxanes, mentioning those currently being used and those that should be adopted. The future directions of taxanes involve more personalized approaches to treatment by tailoring drug-inhibitor combinations or alternatives depending on levels of resistance biomarkers. We hope that this review will identify gaps in knowledge surrounding taxane resistance that future research or clinical trials can overcome.
Collapse
Affiliation(s)
- Sara M. Maloney
- Harper Cancer Research Institute, South Bend, IN 46617, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA
| | - Camden A. Hoover
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| | - Lorena V. Morejon-Lasso
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| | - Jenifer R. Prosperi
- Harper Cancer Research Institute, South Bend, IN 46617, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| |
Collapse
|
31
|
Novel Functions of the Septin Cytoskeleton: Shaping Up Tissue Inflammation and Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:40-51. [PMID: 33039354 DOI: 10.1016/j.ajpath.2020.09.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/24/2020] [Accepted: 09/16/2020] [Indexed: 12/19/2022]
Abstract
Chronic inflammatory diseases cause profound alterations in tissue homeostasis, including unchecked activation of immune and nonimmune cells leading to disease complications such as aberrant tissue repair and fibrosis. Current anti-inflammatory therapies are often insufficient in preventing or reversing these complications. Remodeling of the intracellular cytoskeleton is critical for cell activation in inflamed and fibrotic tissues; however, the cytoskeleton has not been adequately explored as a therapeutic target in inflammation. Septins are GTP-binding proteins that self-assemble into higher order cytoskeletal structures. The septin cytoskeleton exhibits a number of critical cellular functions, including regulation of cell shape and polarity, cytokinesis, cell migration, vesicle trafficking, and receptor signaling. Surprisingly, little is known about the role of the septin cytoskeleton in inflammation. This article reviews emerging evidence implicating different septins in the regulation of host-pathogen interactions, immune cell functions, and tissue fibrosis. Targeting of the septin cytoskeleton as a potential future therapeutic intervention in human inflammatory and fibrotic diseases is also discussed.
Collapse
|
32
|
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: 25] [Impact Index Per Article: 5.0] [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
|
33
|
Sun J, Zheng MY, Li YW, Zhang SW. Structure and function of Septin 9 and its role in human malignant tumors. World J Gastrointest Oncol 2020; 12:619-631. [PMID: 32699577 PMCID: PMC7340996 DOI: 10.4251/wjgo.v12.i6.619] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/03/2020] [Accepted: 04/25/2020] [Indexed: 02/05/2023] Open
Abstract
The treatment and prognosis of malignant tumors are closely related to the time when the tumors are diagnosed; the earlier the diagnosis of the tumor, the better the prognosis. However, most tumors are not detected in the early stages of screening and diagnosis. It is of great clinical significance to study the correlation between multiple pathogeneses of tumors and explore simple, safe, specific, and sensitive molecular indicators for early screening, diagnosis, and prognosis. The Septin 9 (SEPT9) gene has been found to be associated with a variety of human diseases, and it plays a role in the development of tumors. SEPT9 is a member of the conserved family of cytoskeletal GTPase, which consists of a P-loop-based GTP-binding domain flanked by a variable N-terminal region and a C-terminal region. SEPT9 is involved in many biological processes such as cytokinesis, polarization, vesicle trafficking, membrane reconstruction, deoxyribonucleic acid repair, cell migration, and apoptosis. Several studies have shown that SEPT9 may serve as a marker for early screening, diagnosis, and prognosis of some malignant tumors, and have the potential to become a new target for anti-cancer therapy. This article reviews the progress in research on the SEPT9 gene in early screening, diagnosis, and prognosis of tumors.
Collapse
Affiliation(s)
- Jie Sun
- Department of Pathology, Tianjin Union Medical Center, Tianjin 300121, China
| | - Min-Ying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin 300121, China
| | - Yu-Wei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin 300121, China
| | - Shi-Wu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin 300121, China
| |
Collapse
|
34
|
Collins KB, Kang H, Matsche J, Klomp JE, Rehman J, Malik AB, Karginov AV. Septin2 mediates podosome maturation and endothelial cell invasion associated with angiogenesis. J Cell Biol 2020; 219:e201903023. [PMID: 31865373 PMCID: PMC7041690 DOI: 10.1083/jcb.201903023] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 09/14/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Podosomes are compartmentalized actin-rich adhesions, defined by their ability to locally secrete proteases and remodel extracellular matrix. Matrix remodeling by endothelial podosomes facilitates invasion and thereby vessel formation. However, the mechanisms underlying endothelial podosome formation and function remain unclear. Here, we demonstrate that Septin2, Septin6, and Septin7 are required for maturation of nascent endothelial podosomes into matrix-degrading organelles. We show that podosome development occurs through initial mobilization of the scaffolding protein Tks5 and F-actin accumulation, followed by later recruitment of Septin2. Septin2 localizes around the perimeter of podosomes in close proximity to the basolateral plasma membrane, and phosphoinositide-binding residues of Septin2 are required for podosome function. Combined, our results suggest that the septin cytoskeleton forms a diffusive barrier around nascent podosomes to promote their maturation. Finally, we show that Septin2-mediated regulation of podosomes is critical for endothelial cell invasion associated with angiogenesis. Therefore, targeting of Septin2-mediated podosome formation is a potentially attractive anti-angiogenesis strategy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andrei V. Karginov
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL
| |
Collapse
|
35
|
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
|
36
|
O'Loughlin T, Kendrick-Jones J, Buss F. Approaches to Identify and Characterise MYO6-Cargo Interactions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1239:355-380. [PMID: 32451866 DOI: 10.1007/978-3-030-38062-5_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Given the prevalence and importance of the actin cytoskeleton and the host of associated myosin motors, it comes as no surprise to find that they are linked to a plethora of cellular functions and pathologies. Although our understanding of the biophysical properties of myosin motors has been aided by the high levels of conservation in their motor domains and the extensive work on myosin in skeletal muscle contraction, our understanding of how the nonmuscle myosins participate in such a wide variety of cellular processes is less clear. It is now well established that the highly variable myosin tails are responsible for targeting these myosins to distinct cellular sites for specific functions, and although a number of adaptor proteins have been identified, our current understanding of the cellular processes involved is rather limited. Furthermore, as more adaptor proteins, cargoes and complexes are identified, the importance of elucidating the regulatory mechanisms involved is essential. Ca2+, and now phosphorylation and ubiquitination, are emerging as important regulators of cargo binding, and it is likely that other post-translational modifications are also involved. In the case of myosin VI (MYO6), a number of immediate binding partners have been identified using traditional approaches such as yeast two-hybrid screens and affinity-based pull-downs. However, these methods have only been successful in identifying the cargo adaptors, but not the cargoes themselves, which may often comprise multi-protein complexes. Furthermore, motor-adaptor-cargo interactions are dynamic by nature and often weak, transient and highly regulated and therefore difficult to capture using traditional affinity-based methods. In this chapter we will discuss the various approaches including functional proteomics that have been used to uncover and characterise novel MYO6-associated proteins and complexes and how this work contributes to a fuller understanding of the targeting and function(s) of this unique myosin motor.
Collapse
Affiliation(s)
- Thomas O'Loughlin
- Cambridge Institute for Medical Research, University of Cambridge, The Keith Peters Building, Cambridge, UK
| | | | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, The Keith Peters Building, Cambridge, UK.
| |
Collapse
|
37
|
Hahn I, Voelzmann A, Liew YT, Costa-Gomes B, Prokop A. The model of local axon homeostasis - explaining the role and regulation of microtubule bundles in axon maintenance and pathology. Neural Dev 2019; 14:11. [PMID: 31706327 PMCID: PMC6842214 DOI: 10.1186/s13064-019-0134-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Axons are the slender, cable-like, up to meter-long projections of neurons that electrically wire our brains and bodies. In spite of their challenging morphology, they usually need to be maintained for an organism's lifetime. This makes them key lesion sites in pathological processes of ageing, injury and neurodegeneration. The morphology and physiology of axons crucially depends on the parallel bundles of microtubules (MTs), running all along to serve as their structural backbones and highways for life-sustaining cargo transport and organelle dynamics. Understanding how these bundles are formed and then maintained will provide important explanations for axon biology and pathology. Currently, much is known about MTs and the proteins that bind and regulate them, but very little about how these factors functionally integrate to regulate axon biology. As an attempt to bridge between molecular mechanisms and their cellular relevance, we explain here the model of local axon homeostasis, based on our own experiments in Drosophila and published data primarily from vertebrates/mammals as well as C. elegans. The model proposes that (1) the physical forces imposed by motor protein-driven transport and dynamics in the confined axonal space, are a life-sustaining necessity, but pose a strong bias for MT bundles to become disorganised. (2) To counterbalance this risk, MT-binding and -regulating proteins of different classes work together to maintain and protect MT bundles as necessary transport highways. Loss of balance between these two fundamental processes can explain the development of axonopathies, in particular those linking to MT-regulating proteins, motors and transport defects. With this perspective in mind, we hope that more researchers incorporate MTs into their work, thus enhancing our chances of deciphering the complex regulatory networks that underpin axon biology and pathology.
Collapse
Affiliation(s)
- Ines Hahn
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - André Voelzmann
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Yu-Ting Liew
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Beatriz Costa-Gomes
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Andreas Prokop
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK.
| |
Collapse
|
38
|
Vakhrusheva A, Endzhievskaya S, Zhuikov V, Nekrasova T, Parshina E, Ovsiannikova N, Popov V, Bagrov D, Minin AА, Sokolova OS. The role of vimentin in directional migration of rat fibroblasts. Cytoskeleton (Hoboken) 2019; 76:467-476. [PMID: 31626376 DOI: 10.1002/cm.21572] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 10/05/2019] [Accepted: 10/14/2019] [Indexed: 12/16/2022]
Abstract
Cell migration is one of the most important processes in which the cytoskeleton plays a main role. The cytoskeleton network is formed by tubulin microtubules, actin filaments, and intermediate filaments (IFs). While the structure and functions of the two aforementioned proteins have been extensively investigated during the last decades, vimentin IFs structure and their role in cell migration and adhesion remain unclear. Here, we investigated polarity determination in rat fibroblasts with either a knocked out vim gene or with a mutation that blocks filament formation on the stage of unit-length filaments (ULFs). Structured illumination microscopy has demonstrated the difference in the morphology of IFs in wild-type fibroblasts and of ULFs in mutant fibroblasts. We have developed an approach to measure cell stiffness separately on the trailing and leading edges using atomic force microscopy. Young's modulus values on the leading and trailing edge of migrating rat fibroblasts differ approximately by two times, being larger on the leading edge. The knockout of the vim gene leads to having comparable values of Young's moduli on both edges. Vimentin-null cells change the direction of migration more frequently than those expressing wild-type or mutated vimentin. Our results have shown the principle role of vimentin, not only in the form of IFs, but also as ULFs, in the determination of the polarity and the directionality of fibroblast migration.
Collapse
Affiliation(s)
- Anna Vakhrusheva
- Lomonosov Moscow State University, Department of Biology, Moscow, Russia
| | - Sofia Endzhievskaya
- Institute of Protein Research of Russian Academy of Sciences, Department of Cell Biology, Moscow, Russia
| | - Vsevolod Zhuikov
- Research Centre of Biotechnology of Russian Academy of Sciences, Moscow, Russia
| | - Tatyana Nekrasova
- Institute of Protein Research of Russian Academy of Sciences, Department of Cell Biology, Moscow, Russia
| | - Evgenia Parshina
- Lomonosov Moscow State University, Department of Biology, Moscow, Russia
| | - Natalia Ovsiannikova
- Lomonosov Moscow State University, Belozersky Institute of Physico-chemical biology, Moscow, Russia
| | - Vladimir Popov
- Lomonosov Moscow State University, Department of Physics, Moscow, Russia
| | - Dmitry Bagrov
- Lomonosov Moscow State University, Department of Biology, Moscow, Russia
| | - Alexander А Minin
- Institute of Protein Research of Russian Academy of Sciences, Department of Cell Biology, Moscow, Russia
| | - Olga S Sokolova
- Lomonosov Moscow State University, Department of Biology, Moscow, Russia
| |
Collapse
|
39
|
Bashirzadeh Y, Liu AP. Encapsulation of the cytoskeleton: towards mimicking the mechanics of a cell. SOFT MATTER 2019; 15:8425-8436. [PMID: 31621750 DOI: 10.1039/c9sm01669d] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The cytoskeleton of a cell controls all the aspects of cell shape changes and motility from its physiological functions for survival to reproduction to death. The structure and dynamics of the cytoskeletal components: actin, microtubules, intermediate filaments, and septins - recently regarded as the fourth member of the cytoskeleton family - are conserved during evolution. Such conserved and effective control over the mechanics of the cell makes the cytoskeletal components great candidates for in vitro reconstitution and bottom-up synthetic biology studies. Here, we review the recent efforts in reconstitution of the cytoskeleton in and on membrane-enclosed biomimetic systems and argue that co-reconstitution and synergistic interplay between cytoskeletal filaments might be indispensable for efficient mechanical functionality of active minimal cells. Further, mechanical equilibrium in adherent eukaryotic cells is achieved by the formation of integrin-based focal contacts with extracellular matrix (ECM) and the transmission of stresses generated by actomyosin contraction to ECM. Therefore, a minimal mimic of such balance of forces and quasi-static kinetics of the cell by bottom-up reconstitution requires a careful construction of contractile machineries and their link with adhesive contacts. In this review, in addition to cytoskeletal crosstalk, we provide a perspective on reconstruction of cell mechanical equilibrium by reconstitution of cortical actomyosin networks in lipid membrane vesicles adhered on compliant substrates and also discuss future perspectives of this active research area.
Collapse
Affiliation(s)
- Yashar Bashirzadeh
- Department of Mechanical Engineering, University of Michigan, 2350 Hayward Street, Ann Arbor, Michigan, USA.
| | | |
Collapse
|
40
|
Nakos K, Radler MR, Spiliotis ET. Septin 2/6/7 complexes tune microtubule plus-end growth and EB1 binding in a concentration- and filament-dependent manner. Mol Biol Cell 2019; 30:2913-2928. [PMID: 31577529 PMCID: PMC6822581 DOI: 10.1091/mbc.e19-07-0362] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Septins (SEPTs) are filamentous guanosine-5′-triphosphate (GTP)-binding proteins, which affect microtubule (MT)-dependent functions including membrane trafficking and cell division, but their precise role in MT dynamics is poorly understood. Here, in vitro reconstitution of MT dynamics with SEPT2/6/7, the minimal subunits of septin heteromers, shows that SEPT2/6/7 has a biphasic concentration-dependent effect on MT growth. Lower concentrations of SEPT2/6/7 enhance MT plus-end growth and elongation, while higher and intermediate concentrations inhibit and pause plus-end growth, respectively. We show that SEPT2/6/7 has a modest preference for GTP- over guanosine diphosphate (GDP)-bound MT lattice and competes with end-binding protein 1 (EB1) for binding to guanosine 5′-O-[γ-thio]triphosphate (GTPγS)-stabilized MTs, which mimic the EB1-preferred GDP-Pi state of polymerized tubulin. Strikingly, SEPT2/6/7 triggers EB1 dissociation from plus-end tips in cis by binding to the MT lattice and in trans when MT plus ends collide with SEPT2/6/7 filaments. At these intersections, SEPT2/6/7 filaments were more potent barriers than actin filaments in pausing MT growth and dissociating EB1 in vitro and in live cells. These data demonstrate that SEPT2/6/7 complexes and filaments can directly impact MT plus-end growth and the tracking of plus end–binding proteins and thereby may facilitate the capture of MT plus ends at intracellular sites of septin enrichment.
Collapse
Affiliation(s)
| | - Megan R Radler
- Department of Biology, Drexel University, Philadelphia, PA 19104
| | | |
Collapse
|
41
|
Zeng Y, Cao Y, Liu L, Zhao J, Zhang T, Xiao L, Jia M, Tian Q, Yu H, Chen S, Cai Y. SEPT9_i1 regulates human breast cancer cell motility through cytoskeletal and RhoA/FAK signaling pathway regulation. Cell Death Dis 2019; 10:720. [PMID: 31558699 PMCID: PMC6763430 DOI: 10.1038/s41419-019-1947-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 08/09/2019] [Accepted: 08/26/2019] [Indexed: 02/08/2023]
Abstract
Increasing cell mobility is the basis of tumor invasion and metastasis, and is therefore a therapeutic target for preventing the spread of many types of cancer. Septins are a family of cytoskeletal proteins with GTPase activity, and play a role in many important cellular functions, including cell migration. SEPT9 isoform 1 protein (SEPT9_i1) has been associated with breast tumor development and the enhancement of cell migration; however, the exact mechanism of how SEPT9_i1 might affect breast cancer progression remains to be elucidated. Here, we report that the expression of SEPT9_i1 positively correlated with paxillin, and both were significantly upregulated in invasive breast cancer tissues of patients with lymph node metastases. Lentivirus-mediated shRNA knockdown of SEPT9 in MCF-7 cells diminished tumor cell migration, focal adhesion (FA) maturation and the expression of β-actin, β-tubulin, Cdc42, RhoA, and Rac, whereas overexpression of SEPT9_i1 in SEPT9-knockdown MCF-7 cells promoted cell migration, FA maturation and relevant protein expression. Furthermore, overexpression of SEPT9_i1 in MCF-7 cells markedly increased FAK/Src/paxillin signaling, at least in part through RhoA/ROCK1 upstream activation. Transcriptome profiling suggested that SEPT9_i1 may directly affect “Focal adhesion” and “Regulation of actin cytoskeleton” signaling mechanisms. Finally, overexpression of SEPT9_i1 markedly enhanced lung metastases in vivo 6 weeks after tumor inoculation. These findings suggest that a mechanism of Septin-9-induced aberrant cancer cell migration is through cytoskeletal regulation and FA modulation, and encourages the use of SEPT9 as novel therapeutic target in the prevention of tumor metastasis.
Collapse
Affiliation(s)
- Yongqiu Zeng
- Key Laboratory of Obstetric, Gynecologic, and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, Sichuan, China. .,Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China.
| | - Yang Cao
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Lan Liu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Jiao Zhao
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Ting Zhang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Lifan Xiao
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Man Jia
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Qiang Tian
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Hong Yu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Shaokun Chen
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Yansen Cai
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
42
|
de Jonge JJ, Batters C, O'Loughlin T, Arden SD, Buss F. The MYO6 interactome: selective motor-cargo complexes for diverse cellular processes. FEBS Lett 2019; 593:1494-1507. [PMID: 31206648 DOI: 10.1002/1873-3468.13486] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 12/16/2022]
Abstract
Myosins of class VI (MYO6) are unique actin-based motor proteins that move cargo towards the minus ends of actin filaments. As the sole myosin with this directionality, it is critically important in a number of biological processes. Indeed, loss or overexpression of MYO6 in humans is linked to a variety of pathologies including deafness, cardiomyopathy, neurodegenerative diseases as well as cancer. This myosin interacts with a wide variety of direct binding partners such as for example the selective autophagy receptors optineurin, TAX1BP1 and NDP52 and also Dab2, GIPC, TOM1 and LMTK2, which mediate distinct functions of different MYO6 isoforms along the endocytic pathway. Functional proteomics has recently been used to identify the wider MYO6 interactome including several large functionally distinct multi-protein complexes, which highlight the importance of this myosin in regulating the actin and septin cytoskeleton. Interestingly, adaptor-binding not only triggers cargo attachment, but also controls the inactive folded conformation and dimerisation of MYO6. Thus, the C-terminal tail domain mediates cargo recognition and binding, but is also crucial for modulating motor activity and regulating cytoskeletal track dynamics.
Collapse
Affiliation(s)
| | | | - Thomas O'Loughlin
- Cambridge Institute for Medical Research, University of Cambridge, UK
| | - Susan D Arden
- Cambridge Institute for Medical Research, University of Cambridge, UK
| | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, UK
| |
Collapse
|
43
|
Lin XL, Sun QC, Lu Y, Han XQ, Zhao T, Zhou XH. [Proteomic analysis and verification of protein expression after upregulation of human CD99 in Hodgkin lymphoma cell line L428]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:490-496. [PMID: 31340622 PMCID: PMC7342398 DOI: 10.3760/cma.j.issn.0253-2727.2019.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Indexed: 11/22/2022]
Abstract
Objective: To investigate the proteins expression difference after upregulation of human CD99 in Hodgkin Lymphoma cell line, L428 cell, and verify the function of differential proteins. Methods: The differential proteins were detected by two-dimensional fluorescence difference gel electrophoresis and mass spectrometry analysis, cluster analysis was done by GOfact. Results: There were 38 proteins screened out, of which 21 proteins were positively associated with CD99, while 17 proteins were negative. Among the 38 proteins, 32 proteins participated in biological process, and 35 proteins were involved in the composition and construction. And 28 proteins participated in multifaceted biological activities including antioxidation, protein binding, catalytic activity, regulation of enzyme, signal transduction, molecular structure, regulation of translation and ion transport. Conclusions: The changes of the differential proteins, correlated with cytoskeleton, cell differentiation, signal pathway and regulating gene expression, are closely relevant to the translation between Hodgkin/Reed-Sternberg and B lymphocyte cell.
Collapse
Affiliation(s)
- X L Lin
- Department of Pathology, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | | | | | | | | | | |
Collapse
|
44
|
Karasmanis EP, Hwang D, Nakos K, Bowen JR, Angelis D, Spiliotis ET. A Septin Double Ring Controls the Spatiotemporal Organization of the ESCRT Machinery in Cytokinetic Abscission. Curr Biol 2019; 29:2174-2182.e7. [PMID: 31204162 DOI: 10.1016/j.cub.2019.05.050] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/29/2019] [Accepted: 05/20/2019] [Indexed: 01/01/2023]
Abstract
Abscission is the terminal step of mitosis that physically separates two daughter cells [1, 2]. Abscission requires the endocytic sorting complex required for transport (ESCRT), a molecular machinery of multiple subcomplexes (ESCRT-I/II/III) that promotes membrane remodeling and scission [3-5]. Recruitment of ESCRT-I/II complexes to the midbody of telophase cells initiates ESCRT-III assembly into two rings, which subsequently expand into helices and spirals that narrow down to the incipient site of abscission [6-8]. ESCRT-III assembly is highly dynamic and spatiotemporally ordered, but the underlying mechanisms are poorly understood. Here, we report that, after cleavage furrow closure, septins form a membrane-bound double ring that controls the organization and function of ESCRT-III. The septin double ring demarcates the sites of ESCRT-III assembly into rings and disassembles before ESCRT-III rings expand into helices and spirals. We show that septin 9 (SEPT9) depletion, which abrogates abscission, impairs recruitment of VPS25 (ESCRT-II) and CHMP6 (ESCRT-III). Strikingly, ESCRT-III subunits (CHMP4B and CHMP2A/B) accumulate to the midbody, but they are highly disorganized, failing to form symmetric rings and to expand laterally into the cone-shaped helices and spirals of abscission. We found that SEPT9 interacts directly with the ubiquitin E2 variant (UEV) domain of ESCRT-I protein TSG101 through two N-terminal PTAP motifs, which are required for the recruitment of VPS25 and CHMP6, and the spatial organization of ESCRT-III (CHMP4B and CHMP2B) into functional rings. These results reveal that septins function in the ESCRT-I-ESCRT-II-CHMP6 pathway of ESCRT-III assembly and provide a framework for the spatiotemporal control of the ESCRT machinery of cytokinetic abscission.
Collapse
Affiliation(s)
- Eva P Karasmanis
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Daniel Hwang
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | | | - Jonathan R Bowen
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Dimitrios Angelis
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
45
|
An Interaction Network of the Human SEPT9 Established by Quantitative Mass Spectrometry. G3-GENES GENOMES GENETICS 2019; 9:1869-1880. [PMID: 30975701 PMCID: PMC6553528 DOI: 10.1534/g3.119.400197] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Septins regulate the organization of the actin cytoskeleton, vesicle transport and fusion, chromosome alignment and segregation, and cytokinesis in mammalian cells. SEPT9 is part of the core septin hetero-octamer in human cells which is composed of SEPT2, SEPT6, SEPT7, and SEPT9. SEPT9 has been linked to a variety of intracellular functions as well as to diseases and diverse types of cancer. A targeted high-throughput approach to systematically identify the interaction partners of SEPT9 has not yet been performed. We applied a quantitative proteomics approach to establish an interactome of SEPT9 in human fibroblast cells. Among the newly identified interaction partners were members of the myosin family and LIM domain containing proteins. Fluorescence microscopy of SEPT9 and its interaction partners provides additional evidence that SEPT9 might participate in vesicle transport from and to the plasma membrane as well as in the attachment of actin stress fibers to cellular adhesions.
Collapse
|
46
|
Booth DS, Szmidt-Middleton H, King N. Transfection of choanoflagellates illuminates their cell biology and the ancestry of animal septins. Mol Biol Cell 2018; 29:3026-3038. [PMID: 30281390 PMCID: PMC6333174 DOI: 10.1091/mbc.e18-08-0514] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 12/17/2022] Open
Abstract
As the closest living relatives of animals, choanoflagellates offer unique insights into animal origins and core mechanisms underlying animal cell biology. However, unlike traditional model organisms, such as yeast, flies, and worms, choanoflagellates have been refractory to DNA delivery methods for expressing foreign genes. Here we report a robust method for expressing transgenes in the choanoflagellate Salpingoeca rosetta, overcoming barriers that have previously hampered DNA delivery and expression. To demonstrate how this method accelerates the study of S. rosetta cell biology, we engineered a panel of fluorescent protein markers that illuminate key features of choanoflagellate cells. We then investigated the localization of choanoflagellate septins, a family of GTP-binding cytoskeletal proteins that are hypothesized to regulate multicellular rosette development in S. rosetta. Fluorescently tagged septins localized to the basal poles of S. rosetta single cells and rosettes in a pattern resembling septin localization in animal epithelia. The establishment of transfection in S. rosetta and its application to the study of septins represent critical advances in the use of S. rosetta as an experimental model for investigating choanoflagellate cell biology, core mechanisms underlying animal cell biology, and the origin of animals.
Collapse
Affiliation(s)
- David S. Booth
- Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Heather Szmidt-Middleton
- Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Nicole King
- Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
47
|
Nakos K, Rosenberg M, Spiliotis ET. Regulation of microtubule plus end dynamics by septin 9. Cytoskeleton (Hoboken) 2018; 76:83-91. [PMID: 30144301 DOI: 10.1002/cm.21488] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/05/2018] [Accepted: 08/10/2018] [Indexed: 12/18/2022]
Abstract
Septins are GTP-binding proteins that associate with the microtubule (MT) and actin cytoskeleton. Septins affect MT organization and posttranslational modifications, but their role in MT dynamics is less understood. Here, we reconstituted MT dynamics in the presence of the MT-binding septin (SEPT9) using an in vitro cell-free assay, which images the polymerization of tubulin from guanosine-5'-[(α,β)-methyleno]triphosphate (GMPCPP)-stabilized MT seeds. We found that submicromolar concentrations of SEPT9 suppress MT catastrophe and enhance the growth of MT plus ends to great lengths, while low micromolar concentrations of SEPT9 stabilize MTs by inhibiting dynamic instability. We show that SEPT9 associates preferentially with the lattice of GMPCPP-stabilized MT seeds and surprisingly recruits soluble tubulin to the MT lattice. Notably, the effects of SEPT9 on MT dynamics are dependent on its G-G dimerization interface, which is formed by the pockets of the GTP-binding domains. A mutation (H530D) that disrupts G-G dimerization abrogates the effects of SEPT9 on MT dynamics and diminishes its ability to recruit tubulin to the MT lattice. Taken together, these results suggest that SEPT9 promotes the formation and maintenance of long stable MTs through a mechanism that may involve recruitment of unpolymerized tubulin to the MT lattice.
Collapse
Affiliation(s)
| | | | - Elias T Spiliotis
- Department of Biology, Drexel University, Philadelphia, Pennsylvania
| |
Collapse
|
48
|
Wasik AA, Dash SN, Lehtonen S. Septins in kidney: A territory little explored. Cytoskeleton (Hoboken) 2018; 76:154-162. [PMID: 30004646 PMCID: PMC6585700 DOI: 10.1002/cm.21477] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/15/2018] [Accepted: 07/03/2018] [Indexed: 12/15/2022]
Abstract
Septins are a conserved family of GTP‐binding proteins that assemble into cytoskeletal filaments to function in a highly sophisticated and physiologically regulated manner. Originally septins were discovered in the budding yeast as membrane‐associated filaments that affect cell polarity and cytokinesis. In the last decades, much progress has been made in understanding the biochemical properties and cell biological functions of septins. In line with this, mammalian septins have been shown to be involved in various cellular processes, including regulation of cell polarity, cytoskeletal organization, vesicle trafficking, ciliogenesis, and cell–pathogen interactions. A growing number of studies have shown that septins play important roles in tissue and organ development and physiology; yet, little is known about their role in the kidney. In the following review, we discuss the structure and functions of septins in general and summarize the evidence for their presence and roles in the kidney.
Collapse
Affiliation(s)
- Anita A Wasik
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Surjya N Dash
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Sanna Lehtonen
- Department of Pathology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
49
|
Karasmanis EP, Phan CT, Angelis D, Kesisova IA, Hoogenraad CC, McKenney RJ, Spiliotis ET. Polarity of Neuronal Membrane Traffic Requires Sorting of Kinesin Motor Cargo during Entry into Dendrites by a Microtubule-Associated Septin. Dev Cell 2018; 46:204-218.e7. [PMID: 30016622 DOI: 10.1016/j.devcel.2018.06.013] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/04/2018] [Accepted: 06/15/2018] [Indexed: 01/22/2023]
Abstract
Neuronal function requires axon-dendrite membrane polarity, which depends on sorting of membrane traffic during entry into axons. Due to a microtubule network of mixed polarity, dendrites receive vesicles from the cell body without apparent capacity for directional sorting. We found that, during entry into dendrites, axonally destined cargos move with a retrograde bias toward the cell body, while dendritically destined cargos are biased in the anterograde direction. A microtubule-associated septin (SEPT9), which localizes specifically in dendrites, impedes axonal cargo of kinesin-1/KIF5 and boosts kinesin-3/KIF1 motor cargo further into dendrites. In neurons and in vitro single-molecule motility assays, SEPT9 suppresses kinesin-1/KIF5 and enhances kinesin-3/KIF1 in a manner that depends on a lysine-rich loop of the kinesin motor domain. This differential regulation impacts partitioning of neuronal membrane proteins into axons-dendrites. Thus, polarized membrane traffic requires sorting during entry into dendrites by a septin-mediated mechanism that bestows directional bias on microtubules of mixed orientation.
Collapse
Affiliation(s)
- Eva P Karasmanis
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Cat-Thi Phan
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Dimitrios Angelis
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Ilona A Kesisova
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Casper C Hoogenraad
- Cell Biology, Department of Biology, Utrecht University, Utrecht 3584 CH, the Netherlands
| | - Richard J McKenney
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA 95616, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
50
|
Shindo A, Audrey A, Takagishi M, Takahashi M, Wallingford JB, Kinoshita M. Septin-dependent remodeling of cortical microtubule drives cell reshaping during epithelial wound healing. J Cell Sci 2018; 131:jcs212647. [PMID: 29777035 PMCID: PMC6031381 DOI: 10.1242/jcs.212647] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 05/10/2018] [Indexed: 12/31/2022] Open
Abstract
Wounds in embryos heal rapidly through contraction of the wound edges. Despite well-recognized significance of the actomyosin purse string for wound closure, roles for other cytoskeletal components are largely unknown. Here, we report that the septin cytoskeleton cooperates with actomyosin and microtubules to coordinate circumferential contraction of the wound margin and concentric elongation of wound-proximal cells in Xenopus laevis embryos. Microtubules reoriented radially, forming bundles along lateral cell cortices in elongating wound-proximal cells. Depletion of septin 7 (Sept7) slowed wound closure by attenuating the wound edge contraction and cell elongation. ROCK/Rho-kinase inhibitor-mediated suppression of actomyosin contractility enhanced the Sept7 phenotype, whereas the Sept7 depletion did not affect the accumulation of actomyosin at the wound edge. The cortical microtubule bundles were reduced in wound-proximal cells in Sept7 knockdown (Sept7-KD) embryos, but forced bundling of microtubules mediated by the microtubule-stabilizing protein Map7 did not rescue the Sept7-KD phenotype. Nocodazole-mediated microtubule depolymerization enhanced the Sept7-KD phenotype, suggesting that Sept7 is required for microtubule reorganization during cell elongation. Our findings indicate that septins are required for the rapid wound closure by facilitating cortical microtubule reorganization and the concentric elongation of surrounding cells.
Collapse
Affiliation(s)
- Asako Shindo
- Division of Biological Sciences, Department of Molecular Biology, Nagoya University Graduate School of Science, Nagoya 464-8602, Japan
- Department of Molecular Biosciences, University of Texas at Austin, Austin 78712, USA
| | - Anastasia Audrey
- Division of Biological Sciences, Department of Molecular Biology, Nagoya University Graduate School of Science, Nagoya 464-8602, Japan
| | - Maki Takagishi
- Department of Tumor Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Masahide Takahashi
- Department of Tumor Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin 78712, USA
| | - Makoto Kinoshita
- Division of Biological Sciences, Department of Molecular Biology, Nagoya University Graduate School of Science, Nagoya 464-8602, Japan
| |
Collapse
|