1
|
Terada Y, Obara K, Yoshioka Y, Ochiya T, Bito H, Tsuchida K, Ageta H, Ageta-Ishihara N. Intracellular dynamics of ubiquitin-like 3 visualized using an inducible fluorescent timer expression system. Biol Open 2024; 13:bio060345. [PMID: 39498724 DOI: 10.1242/bio.060345] [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: 08/16/2024] [Accepted: 10/03/2024] [Indexed: 11/07/2024] Open
Abstract
Exosomes are small extracellular vesicles (sEVs) secreted via multivesicular bodies (MVBs)/late endosomes and mediators of cell-cell communication. We previously reported a novel post-translational modification by ubiquitin-like 3 (UBL3). UBL3 is localized in MVBs and the plasma membrane and released outside as sEVs, including exosomes. Approximately 60% of proteins sorted in sEVs are affected by UBL3 and localized in various organelles, the plasma membrane, and the cytosol, suggesting that its dynamic movement in the cell before entering the MVBs. To examine the intracellular dynamics of UBL3, we constructed a sophisticated visualization system via fusing fluorescent timers that changed from blue to red form over time with UBL3 and by its expression under Tet-on regulation. Intriguingly, we found that after synthesis, UBL3 was initially distributed within the cytosol. Subsequently, UBL3 was localized to MVBs and the plasma membrane and finally showed predominant accumulation in MVBs. Furthermore, by super-resolution microscopy analysis, UBL3 was found to be associated with one of its substrates, α-tubulin, in the cytosol, and the complex was subsequently transported to MVBs. This spatiotemporal visualization system for UBL3 will form a basis for further studies to elucidate when and where UBL3 associates with its substrates/binding proteins before localization in MVBs.
Collapse
Affiliation(s)
- Yuka Terada
- Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Chiba 274-8510, Japan
| | - Kumi Obara
- Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Chiba 274-8510, Japan
| | - Yusuke Yoshioka
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Shinjyuku-ku, Tokyo 160-0023, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Shinjyuku-ku, Tokyo 160-0023, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kunihiro Tsuchida
- Division for Therapies Against Intractable Diseases, Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Hiroshi Ageta
- Division for Therapies Against Intractable Diseases, Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Natsumi Ageta-Ishihara
- Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Chiba 274-8510, Japan
| |
Collapse
|
2
|
Verma H, Kaur S, Kaur S, Gangwar P, Dhiman M, Mantha AK. Role of Cytoskeletal Elements in Regulation of Synaptic Functions: Implications Toward Alzheimer's Disease and Phytochemicals-Based Interventions. Mol Neurobiol 2024; 61:8320-8343. [PMID: 38491338 DOI: 10.1007/s12035-024-04053-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 02/13/2024] [Indexed: 03/18/2024]
Abstract
Alzheimer's disease (AD), a multifactorial disease, is characterized by the accumulation of neurofibrillary tangles (NFTs) and amyloid beta (Aβ) plaques. AD is triggered via several factors like alteration in cytoskeletal proteins, a mutation in presenilin 1 (PSEN1), presenilin 2 (PSEN2), amyloid precursor protein (APP), and post-translational modifications (PTMs) in the cytoskeletal elements. Owing to the major structural and functional role of cytoskeletal elements, like the organization of axon initial segmentation, dendritic spines, synaptic regulation, and delivery of cargo at the synapse; modulation of these elements plays an important role in AD pathogenesis; like Tau is a microtubule-associated protein that stabilizes the microtubules, and it also causes inhibition of nucleo-cytoplasmic transportation by disrupting the integrity of nuclear pore complex. One of the major cytoskeletal elements, actin and its dynamics, regulate the dendritic spine structure and functions; impairments have been documented towards learning and memory defects. The second major constituent of these cytoskeletal elements, microtubules, are necessary for the delivery of the cargo, like ion channels and receptors at the synaptic membranes, whereas actin-binding protein, i.e., Cofilin's activation form rod-like structures, is involved in the formation of paired helical filaments (PHFs) observed in AD. Also, the glial cells rely on their cytoskeleton to maintain synaptic functionality. Thus, making cytoskeletal elements and their regulation in synaptic structure and function as an important aspect to be focused for better management and targeting AD pathology. This review advocates exploring phytochemicals and Ayurvedic plant extracts against AD by elucidating their neuroprotective mechanisms involving cytoskeletal modulation and enhancing synaptic plasticity. However, challenges include their limited bioavailability due to the poor solubility and the limited potential to cross the blood-brain barrier (BBB), emphasizing the need for targeted strategies to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Harkomal Verma
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, VPO - Ghudda, Bathinda, 151 401, Punjab, India
| | - Sharanjot Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, India
| | - Sukhchain Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, India
| | - Prabhakar Gangwar
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, VPO - Ghudda, Bathinda, 151 401, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Village Ghudda, Bathinda, Punjab, India
| | - Anil Kumar Mantha
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Village Ghudda, VPO - Ghudda, Bathinda, 151 401, Punjab, India.
| |
Collapse
|
3
|
Das A, Kunwar A. Septins: Structural Insights, Functional Dynamics, and Implications in Health and Disease. J Cell Biochem 2024:e30660. [PMID: 39324363 DOI: 10.1002/jcb.30660] [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: 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
|
4
|
Jędrzejczak P, Saramowicz K, Kuś J, Barczuk J, Rozpędek-Kamińska W, Siwecka N, Galita G, Wiese W, Majsterek I. SEPT9_i1 and Septin Dynamics in Oncogenesis and Cancer Treatment. Biomolecules 2024; 14:1194. [PMID: 39334960 PMCID: PMC11430720 DOI: 10.3390/biom14091194] [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: 08/15/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Despite significant advancements in the field of oncology, cancers still pose one of the greatest challenges of modern healthcare. Given the cytoskeleton's pivotal role in regulating mechanisms critical to cancer development, further studies of the cytoskeletal elements could yield new practical applications. Septins represent a group of relatively well-conserved GTP-binding proteins that constitute the fourth component of the cytoskeleton. Septin 9 (SEPT9) has been linked to a diverse spectrum of malignancies and appears to be the most notable septin member in that category. SEPT9 constitutes a biomarker of colorectal cancer (CRC) and has been positively correlated with a high clinical stage in breast cancer, cervical cancer, and head and neck squamous cell carcinoma. SEPT9_i1 represents the most extensively studied isoform of SEPT9, which substantially contributes to carcinogenesis, metastasis, and treatment resistance. Nevertheless, the mechanistic basis of SEPT9_i1 oncogenicity remains to be fully elucidated. In this review, we highlight SEPT9's and SEPT9_i1's structures and interactions with Hypoxia Inducible Factor α (HIF-1 α) and C-Jun N-Terminal Kinase (JNK), as well as discuss SEPT9_i1's contribution to aneuploidy, cell invasiveness, and taxane resistance-key phenomena in the progression of malignancies. Finally, we emphasize forchlorfenuron and other septin inhibitors as potential chemotherapeutics and migrastatics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (P.J.); (K.S.); (J.K.); (J.B.); (W.R.-K.); (N.S.); (G.G.); (W.W.)
| |
Collapse
|
5
|
Ageta H, Nishioka T, Yamaguchi H, Tsuchida K, Ageta-Ishihara N. Comprehensive identification of ubiquitin-like 3 (UBL3)-interacting proteins in the mouse brain. Mol Brain 2024; 17:57. [PMID: 39148092 PMCID: PMC11325695 DOI: 10.1186/s13041-024-01131-4] [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: 11/30/2023] [Accepted: 08/07/2024] [Indexed: 08/17/2024] Open
Abstract
Discovery of novel post-translational modifications provides new insights into changes in protein function, localization, and stability. They are also key elements in understanding disease mechanisms and developing therapeutic strategies. We have previously reported that ubiquitin-like 3 (UBL3) serves as a novel post-translational modifier that is highly expressed in the cerebral cortex and hippocampus, in addition to various other organs, and that 60% of proteins contained in small extracellular vesicles (sEVs), including exosomes, are influenced by UBL3. In this study, we generated transgenic mice expressing biotinylated UBL3 in the forebrain under control of the alpha-CaMKII promoter (Ubl3Tg/+). Western blot analysis revealed that the expression of UBL3 in the cerebral cortex and hippocampus was 6- to 7-fold higher than that in the cerebellum. Therefore, we performed immunoprecipitation of protein extracts from the cerebral cortex of Ubl3+/+ and Ubl3Tg/+ mice using avidin beads to comprehensively discover UBL3 interacting proteins, identifying 35 new UBL3 interacting proteins. Nine proteins were annotated as extracellular exosomes. Gene Ontology (GO) analysis suggested a new relationship between sEVs and RNA metabolism in neurodegenerative diseases. We confirmed the association of endogenous UBL3 with the RNA-binding proteins FUS and HPRT1-both listed in the Neurodegenerative Diseases Variation Database (NDDVD)-and with LYPLA1, which is involved in Huntington's disease, using immunoprecipitation (IP)-western blotting analysis. These UBL3 interacting proteins will accelerate the continued elucidation of sEV research about proteins regulated by novel post-translational modifications by UBL3 in the brain.
Collapse
Affiliation(s)
- Hiroshi Ageta
- Division for Therapies Against Intractable Diseases, Center for Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| | - Tomoki Nishioka
- Open Facility Center, Research Promotion Headquarters, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Hisateru Yamaguchi
- Department of Medical Technology, Yokkaichi Nursing and Medical Care University, Yokkaichi, 512-8045, Japan
| | - Kunihiro Tsuchida
- Division for Therapies Against Intractable Diseases, Center for Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| | - Natsumi Ageta-Ishihara
- Department of Biomolecular Science, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan.
| |
Collapse
|
6
|
Scott-Hewitt N, Mahoney M, Huang Y, Korte N, Yvanka de Soysa T, Wilton DK, Knorr E, Mastro K, Chang A, Zhang A, Melville D, Schenone M, Hartigan C, Stevens B. Microglial-derived C1q integrates into neuronal ribonucleoprotein complexes and impacts protein homeostasis in the aging brain. Cell 2024; 187:4193-4212.e24. [PMID: 38942014 DOI: 10.1016/j.cell.2024.05.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 01/08/2024] [Accepted: 05/31/2024] [Indexed: 06/30/2024]
Abstract
Neuroimmune interactions mediate intercellular communication and underlie critical brain functions. Microglia, CNS-resident macrophages, modulate the brain through direct physical interactions and the secretion of molecules. One such secreted factor, the complement protein C1q, contributes to complement-mediated synapse elimination in both developmental and disease models, yet brain C1q protein levels increase significantly throughout aging. Here, we report that C1q interacts with neuronal ribonucleoprotein (RNP) complexes in an age-dependent manner. Purified C1q protein undergoes RNA-dependent liquid-liquid phase separation (LLPS) in vitro, and the interaction of C1q with neuronal RNP complexes in vivo is dependent on RNA and endocytosis. Mice lacking C1q have age-specific alterations in neuronal protein synthesis in vivo and impaired fear memory extinction. Together, our findings reveal a biophysical property of C1q that underlies RNA- and age-dependent neuronal interactions and demonstrate a role of C1q in critical intracellular neuronal processes.
Collapse
Affiliation(s)
- Nicole Scott-Hewitt
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Matthew Mahoney
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Youtong Huang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nils Korte
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - T Yvanka de Soysa
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniel K Wilton
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Emily Knorr
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kevin Mastro
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Allison Chang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Allison Zhang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - David Melville
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Monica Schenone
- The Broad Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Christina Hartigan
- The Broad Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Investigator, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Pérez Baca MDR, Jacobs EZ, Vantomme L, Leblanc P, Bogaert E, Dheedene A, De Cock L, Haghshenas S, Foroutan A, Levy MA, Kerkhof J, McConkey H, Chen CA, Batzir NA, Wang X, Palomares M, Carels M, Dermaut B, Sadikovic B, Menten B, Yuan B, Vergult S, Callewaert B. Haploinsufficiency of ZFHX3, encoding a key player in neuronal development, causes syndromic intellectual disability. Am J Hum Genet 2024; 111:509-528. [PMID: 38412861 PMCID: PMC10940049 DOI: 10.1016/j.ajhg.2024.01.013] [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: 06/07/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/29/2024] Open
Abstract
Neurodevelopmental disorders (NDDs) result from impaired development and functioning of the brain. Here, we identify loss-of-function (LoF) variation in ZFHX3 as a cause for syndromic intellectual disability (ID). ZFHX3 is a zinc-finger homeodomain transcription factor involved in various biological processes, including cell differentiation and tumorigenesis. We describe 42 individuals with protein-truncating variants (PTVs) or (partial) deletions of ZFHX3, exhibiting variable intellectual disability and autism spectrum disorder, recurrent facial features, relative short stature, brachydactyly, and, rarely, cleft palate. ZFHX3 LoF associates with a specific methylation profile in whole blood extracted DNA. Nuclear abundance of ZFHX3 increases during human brain development and neuronal differentiation. ZFHX3 was found to interact with the chromatin remodeling BRG1/Brm-associated factor complex and the cleavage and polyadenylation complex, suggesting a function in chromatin remodeling and mRNA processing. Furthermore, ChIP-seq for ZFHX3 revealed that it predominantly binds promoters of genes involved in nervous system development. We conclude that loss-of-function variants in ZFHX3 are a cause of syndromic ID associating with a specific DNA methylation profile.
Collapse
Affiliation(s)
- María Del Rocío Pérez Baca
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Eva Z Jacobs
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Lies Vantomme
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Pontus Leblanc
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Elke Bogaert
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Annelies Dheedene
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Laurenz De Cock
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Sadegheh Haghshenas
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Aidin Foroutan
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada; Children's Health Research Institute, Lawson Research Institute, London, ON N6C 2R5, Canada
| | - Michael A Levy
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Jennifer Kerkhof
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Haley McConkey
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada; Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada
| | - Chun-An Chen
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Nurit Assia Batzir
- Schneider Children's Medical Center of Israel, Petach Tikvah 4920235, Israel
| | - Xia Wang
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - María Palomares
- INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Marieke Carels
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; VIB UGent Center for Inflammation Research, Department for Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Bart Dermaut
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Bekim Sadikovic
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada; Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada
| | - Björn Menten
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Bo Yuan
- Seattle Children's Hospital, Seattle and Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98105, USA
| | - Sarah Vergult
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium.
| | - Bert Callewaert
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
8
|
Villar-Pazos S, Thomas L, Yang Y, Chen K, Lyles JB, Deitch BJ, Ochaba J, Ling K, Powers B, Gingras S, Kordasiewicz HB, Grubisha MJ, Huang YH, Thomas G. Neural deficits in a mouse model of PACS1 syndrome are corrected with PACS1- or HDAC6-targeting therapy. Nat Commun 2023; 14:6547. [PMID: 37848409 PMCID: PMC10582149 DOI: 10.1038/s41467-023-42176-8] [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: 01/26/2023] [Accepted: 09/29/2023] [Indexed: 10/19/2023] Open
Abstract
PACS1 syndrome is a neurodevelopmental disorder (NDD) caused by a recurrent de novo missense mutation in PACS1 (p.Arg203Trp (PACS1R203W)). The mechanism by which PACS1R203W causes PACS1 syndrome is unknown, and no curative treatment is available. Here, we use patient cells and PACS1 syndrome mice to show that PACS1 (or PACS-1) is an HDAC6 effector and that the R203W substitution increases the PACS1/HDAC6 interaction, aberrantly potentiating deacetylase activity. Consequently, PACS1R203W reduces acetylation of α-tubulin and cortactin, causing the Golgi ribbon in hippocampal neurons and patient-derived neural progenitor cells (NPCs) to fragment and overpopulate dendrites, increasing their arborization. The dendrites, however, are beset with varicosities, diminished spine density, and fewer functional synapses, characteristic of NDDs. Treatment of PACS1 syndrome mice or patient NPCs with PACS1- or HDAC6-targeting antisense oligonucleotides, or HDAC6 inhibitors, restores neuronal structure and synaptic transmission in prefrontal cortex, suggesting that targeting PACS1R203W/HDAC6 may be an effective therapy for PACS1 syndrome.
Collapse
Affiliation(s)
- Sabrina Villar-Pazos
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter Campus (VBC), Vienna, Austria
| | - Laurel Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Yunhan Yang
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Kun Chen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jenea B Lyles
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Bradley J Deitch
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | | | - Karen Ling
- Ionis Pharmaceuticals, Carlsbad, CA, USA
| | | | - Sebastien Gingras
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Melanie J Grubisha
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yanhua H Huang
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Gary Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
9
|
Smith G, Sweeney ST, O’Kane CJ, Prokop A. How neurons maintain their axons long-term: an integrated view of axon biology and pathology. Front Neurosci 2023; 17:1236815. [PMID: 37564364 PMCID: PMC10410161 DOI: 10.3389/fnins.2023.1236815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/06/2023] [Indexed: 08/12/2023] Open
Abstract
Axons are processes of neurons, up to a metre long, that form the essential biological cables wiring nervous systems. They must survive, often far away from their cell bodies and up to a century in humans. This requires self-sufficient cell biology including structural proteins, organelles, and membrane trafficking, metabolic, signalling, translational, chaperone, and degradation machinery-all maintaining the homeostasis of energy, lipids, proteins, and signalling networks including reactive oxygen species and calcium. Axon maintenance also involves specialised cytoskeleton including the cortical actin-spectrin corset, and bundles of microtubules that provide the highways for motor-driven transport of components and organelles for virtually all the above-mentioned processes. Here, we aim to provide a conceptual overview of key aspects of axon biology and physiology, and the homeostatic networks they form. This homeostasis can be derailed, causing axonopathies through processes of ageing, trauma, poisoning, inflammation or genetic mutations. To illustrate which malfunctions of organelles or cell biological processes can lead to axonopathies, we focus on axonopathy-linked subcellular defects caused by genetic mutations. Based on these descriptions and backed up by our comprehensive data mining of genes linked to neural disorders, we describe the 'dependency cycle of local axon homeostasis' as an integrative model to explain why very different causes can trigger very similar axonopathies, providing new ideas that can drive the quest for strategies able to battle these devastating diseases.
Collapse
Affiliation(s)
- Gaynor Smith
- Cardiff University, School of Medicine, College of Biomedical and Life Sciences, Cardiff, United Kingdom
| | - Sean T. Sweeney
- Department of Biology, University of York and York Biomedical Research Institute, York, United Kingdom
| | - Cahir J. O’Kane
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Andreas Prokop
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biology, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
10
|
Werner B, Yadav S. Phosphoregulation of the septin cytoskeleton in neuronal development and disease. Cytoskeleton (Hoboken) 2023; 80:275-289. [PMID: 36127729 PMCID: PMC10025170 DOI: 10.1002/cm.21728] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/13/2022] [Accepted: 09/12/2022] [Indexed: 11/06/2022]
Abstract
Septins are highly conserved GTP-binding proteins that oligomerize and form higher order structures. The septin cytoskeleton plays an important role in cellular organization, intracellular transport, and cytokinesis. Kinase-mediated phosphorylation of septins regulates various aspects of their function, localization, and dynamics. Septins are enriched in the mammalian nervous system where they contribute to neurodevelopment and neuronal function. Emerging research has implicated aberrant changes in septin cytoskeleton in several human diseases. The mechanisms through which aberrant phosphorylation by kinases contributes to septin dysfunction in neurological disorders are poorly understood and represent an important question for future research with therapeutic implications. This review summarizes the current state of knowledge of the diversity of kinases that interact with and phosphorylate mammalian septins, delineates how phosphoregulation impacts septin dynamics, and describes how aberrant septin phosphorylation contributes to neurological disorders.
Collapse
Affiliation(s)
- Bailey Werner
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
11
|
Torraca V, Bielecka MK, Gomes MC, Brokatzky D, Busch‐Nentwich EM, Mostowy S. Zebrafish null mutants of Sept6 and Sept15 are viable but more susceptible to Shigella infection. Cytoskeleton (Hoboken) 2023; 80:266-274. [PMID: 36855298 PMCID: PMC10952258 DOI: 10.1002/cm.21750] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/21/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023]
Abstract
Septins are evolutionarily conserved GTP-binding proteins known for their roles in cell division and host defence against Shigella infection. Although septin group members are viewed to function as hetero-oligomeric complexes, the role of individual septins within these complexes or in isolation is poorly understood. Decades of work using mouse models has shown that some septins (including SEPT7) are essential for animal development, while others (including SEPT6) are dispensable, suggesting that some septins may compensate for the absence of others. The zebrafish genome encodes 19 septin genes, representing the full complement of septin groups described in mice and humans. In this report, we characterise null mutants for zebrafish Sept6 (a member of the SEPT6 group) and Sept15 (a member of the SEPT7 group) and test their role in zebrafish development and host defence against Shigella infection. We show that null mutants for Sept6 and Sept15 are both viable, and that expression of other zebrafish septins are not significantly affected by their mutation. Consistent with previous reports using knockdown of Sept2, Sept7b, and Sept15, we show that Sept6 and Sept15 are required for host defence against Shigella infection. These results highlight Shigella infection of zebrafish as a powerful system to study the role of individual septins in vivo.
Collapse
Affiliation(s)
- Vincenzo Torraca
- Department of Infection BiologyLondon School of Hygiene & Tropical MedicineLondonUK
- School of Life SciencesUniversity of WestminsterLondonUK
| | | | - Margarida C. Gomes
- Department of Infection BiologyLondon School of Hygiene & Tropical MedicineLondonUK
| | - Dominik Brokatzky
- Department of Infection BiologyLondon School of Hygiene & Tropical MedicineLondonUK
| | - Elisabeth M. Busch‐Nentwich
- Department of Medicine, Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID)University of CambridgeCambridgeUK
- School of Biological and Behavioural Sciences, Faculty of Science and EngineeringQueen Mary University of LondonLondonUK
| | - Serge Mostowy
- Department of Infection BiologyLondon School of Hygiene & Tropical MedicineLondonUK
| |
Collapse
|
12
|
Naren P, Samim KS, Tryphena KP, Vora LK, Srivastava S, Singh SB, Khatri DK. Microtubule acetylation dyshomeostasis in Parkinson's disease. Transl Neurodegener 2023; 12:20. [PMID: 37150812 PMCID: PMC10165769 DOI: 10.1186/s40035-023-00354-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/06/2023] [Indexed: 05/09/2023] Open
Abstract
The inter-neuronal communication occurring in extensively branched neuronal cells is achieved primarily through the microtubule (MT)-mediated axonal transport system. This mechanistically regulated system delivers cargos (proteins, mRNAs and organelles such as mitochondria) back and forth from the soma to the synapse. Motor proteins like kinesins and dynein mechanistically regulate polarized anterograde (from the soma to the synapse) and retrograde (from the synapse to the soma) commute of the cargos, respectively. Proficient axonal transport of such cargos is achieved by altering the microtubule stability via post-translational modifications (PTMs) of α- and β-tubulin heterodimers, core components constructing the MTs. Occurring within the lumen of MTs, K40 acetylation of α-tubulin via α-tubulin acetyl transferase and its subsequent deacetylation by HDAC6 and SIRT2 are widely scrutinized PTMs that make the MTs highly flexible, which in turn promotes their lifespan. The movement of various motor proteins, including kinesin-1 (responsible for axonal mitochondrial commute), is enhanced by this PTM, and dyshomeostasis of neuronal MT acetylation has been observed in a variety of neurodegenerative conditions, including Alzheimer's disease and Parkinson's disease (PD). PD is the second most common neurodegenerative condition and is closely associated with impaired MT dynamics and deregulated tubulin acetylation levels. Although the relationship between status of MT acetylation and progression of PD pathogenesis has become a chicken-and-egg question, our review aims to provide insights into the MT-mediated axonal commute of mitochondria and dyshomeostasis of MT acetylation in PD. The enzymatic regulators of MT acetylation along with their synthetic modulators have also been briefly explored. Moving towards a tubulin-based therapy that enhances MT acetylation could serve as a disease-modifying treatment in neurological conditions that lack it.
Collapse
Affiliation(s)
- Padmashri Naren
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Khan Sabiya Samim
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Kamatham Pushpa Tryphena
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| | - Shashi Bala Singh
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Dharmendra Kumar Khatri
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| |
Collapse
|
13
|
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
|
14
|
Niu X, Mao CX, Wang S, Wang X, Zhang Y, Hu J, Bi R, Liu Z, Shan J. α-Tubulin acetylation at lysine 40 regulates dendritic arborization and larval locomotion by promoting microtubule stability in Drosophila. PLoS One 2023; 18:e0280573. [PMID: 36827311 PMCID: PMC9955671 DOI: 10.1371/journal.pone.0280573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 01/03/2023] [Indexed: 02/25/2023] Open
Abstract
Posttranslational modification of tubulin increases the dynamic complexity and functional diversity of microtubules. Acetylation of α-tubulin at Lys-40 is a highly conserved posttranslational modification that has been shown to improve the flexibility and resilience of microtubules. Here we studied the in vivo functions of α-tubulin acetylation by knocking-out Atat, the Drosophila α-tubulin acetyltransferase, and by mutating Lys-40 to Arg in α1-tubulin. We found a reduction in the dendritic arborization of larval class I dendritic arborization (da) neurons in both mutants. The dendritic developmental defects in atat mutants could be reversed by enhancing the stability of microtubules either through knocking down the microtubule severing protein Katanin 60 or through overexpressing tubulin-specific chaperone E, suggesting that α-tubulin deacetylation impairsed dendritic morphology by decreasing the stability of microtubules. Using time-lapse recordings, we found that atat and α1-tubulinK40R mutations dramatically increased the number of dendritic protrusions that were likely to be immature dendritic precursors. Finally, we showed that both Atat and α-tubulin acetylation were required in class I da neurons to control larval locomotion. These findings add novel insight into the current knowledge of the role of α-tubulin acetylation in regulating neuronal development and functions.
Collapse
Affiliation(s)
- Xiaoxiao Niu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life science, Hubei University, Wuhan, China
| | - Chuan-Xi Mao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life science, Hubei University, Wuhan, China
| | - Shan Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life science, Hubei University, Wuhan, China
| | - Xiongxiong Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life science, Hubei University, Wuhan, China
| | - Youyu Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life science, Hubei University, Wuhan, China
| | - Juncheng Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life science, Hubei University, Wuhan, China
| | - Ran Bi
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life science, Hubei University, Wuhan, China
| | - Zhihua Liu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life science, Hubei University, Wuhan, China
- * E-mail: (SJ); (ZL)
| | - Jin Shan
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life science, Hubei University, Wuhan, China
- * E-mail: (SJ); (ZL)
| |
Collapse
|
15
|
Ho CT, Gupton SL. Cytoskeleton: Septin wreaths regulate actin in neuritogenesis. Curr Biol 2023; 33:R98-R100. [PMID: 36750031 DOI: 10.1016/j.cub.2022.12.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The shape of a neuron changes dramatically during development. New work reports a novel septin cytoskeleton network that is important in establishing proper neuronal morphology.
Collapse
Affiliation(s)
- Chris T Ho
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Medical Biomolecular Research Building 4332, Campus Box 7090, Chapel Hill, NC 27599-7545, USA
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Medical Biomolecular Research Building 4332, Campus Box 7090, Chapel Hill, NC 27599-7545, USA; UNC Neuroscience Center, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA.
| |
Collapse
|
16
|
Radler MR, Liu X, Peng M, Doyle B, Toyo-Oka K, Spiliotis ET. Pyramidal neuron morphogenesis requires a septin network that stabilizes filopodia and suppresses lamellipodia during neurite initiation. Curr Biol 2023; 33:434-448.e8. [PMID: 36538929 PMCID: PMC9905282 DOI: 10.1016/j.cub.2022.11.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/31/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022]
Abstract
Pyramidal neurons are a major cell type of the forebrain, consisting of a pyramidally shaped soma with axonal and apicobasal dendritic processes. It is poorly understood how the neuronal soma develops its pyramidal morphology, while generating neurites of the proper shape and orientation. Here, we discovered that the spherical somata of immature neurite-less neurons possess a circumferential wreath-like network of septin filaments, which promotes neuritogenesis by balancing the protrusive activity of lamellipodia and filopodia. In embryonic rat hippocampal and mouse cortical neurons, the septin wreath network consists of curvilinear filaments that contain septins 5, 7, and 11 (Sept5/7/11). The Sept5/7/11 wreath network demarcates a zone of myosin II enrichment and Arp2/3 diminution at the base of filopodial actin bundles. In Sept7-depleted neurons, cell bodies are enlarged with hyperextended lamellae and abnormally shaped neurites that originate from lamellipodia. This phenotype is accompanied by diminished myosin II and filopodia lifetimes and increased Arp2/3 and lamellipodial activity. Inhibition of Arp2/3 rescues soma and neurite phenotypes, indicating that the septin wreath network suppresses the extension of lamellipodia, facilitating the formation of neurites from the filopodia of a consolidated soma. We show that this septin function is critical for developing a pyramidally shaped soma with properly distributed and oriented dendrites in cultured rat hippocampal neurons and in vivo in mouse perinatal cortical neurons. Therefore, the somatic septin cytoskeleton provides a key morphogenetic mechanism for neuritogenesis and the development of pyramidal neurons.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Xiaonan Liu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Megan Peng
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Brenna Doyle
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
17
|
Stewart VD, Cadieux J, Thulasiram MR, Douglas TC, Drewnik DA, Selamat S, Lao Y, Spicer V, Hannila SS. Myelin‐associated glycoprotein alters the neuronal secretome and stimulates the release of
TGFβ
and proteins that affect neural plasticity. FEBS Lett 2022; 596:2952-2973. [DOI: 10.1002/1873-3468.14496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/12/2022]
Affiliation(s)
- Vanessa D. Stewart
- Department of Human Anatomy and Cell Science University of Manitoba Room 130, Basic Medical Sciences Building, 745 Bannatyne Avenue R3E 0J9 Winnipeg Manitoba Canada
| | - Justine Cadieux
- Department of Human Anatomy and Cell Science University of Manitoba Room 130, Basic Medical Sciences Building, 745 Bannatyne Avenue R3E 0J9 Winnipeg Manitoba Canada
| | - Matsya R. Thulasiram
- Department of Human Anatomy and Cell Science University of Manitoba Room 130, Basic Medical Sciences Building, 745 Bannatyne Avenue R3E 0J9 Winnipeg Manitoba Canada
| | - Tinsley Claire Douglas
- Department of Human Anatomy and Cell Science University of Manitoba Room 130, Basic Medical Sciences Building, 745 Bannatyne Avenue R3E 0J9 Winnipeg Manitoba Canada
| | - Dennis A. Drewnik
- Department of Human Anatomy and Cell Science University of Manitoba Room 130, Basic Medical Sciences Building, 745 Bannatyne Avenue R3E 0J9 Winnipeg Manitoba Canada
| | - Suhaila Selamat
- Department of Human Anatomy and Cell Science University of Manitoba Room 130, Basic Medical Sciences Building, 745 Bannatyne Avenue R3E 0J9 Winnipeg Manitoba Canada
| | - Ying Lao
- Centre for Proteomics and Systems Biology University of Manitoba Room 799, John Buhler Research Centre, 715 McDermot Avenue R3E 3P4 Winnipeg Manitoba Canada
| | - Victor Spicer
- Centre for Proteomics and Systems Biology University of Manitoba Room 799, John Buhler Research Centre, 715 McDermot Avenue R3E 3P4 Winnipeg Manitoba Canada
| | - Sari S. Hannila
- Department of Human Anatomy and Cell Science University of Manitoba Room 130, Basic Medical Sciences Building, 745 Bannatyne Avenue R3E 0J9 Winnipeg Manitoba Canada
| |
Collapse
|
18
|
Neuronal-specific septin-3 binds Atg8/LC3B, accumulates and localizes to autophagosomes during induced autophagy. Cell Mol Life Sci 2022; 79:471. [PMID: 35932293 PMCID: PMC9356936 DOI: 10.1007/s00018-022-04488-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/27/2022] [Accepted: 07/13/2022] [Indexed: 11/03/2022]
Abstract
In synapses that show signs of local apoptosis and mitochondrial stress and undergo neuro-immunological synapse pruning, an increase in the levels of the presynaptic protein, neuronal-specific septin-3 can be observed. Septin-3 is a member of the septin GTPase family with the ability to form multimers and contribute to the cytoskeleton. However, the function of septin-3 remains elusive. Here, we provide evidence that septin-3 is capable of binding the most-studied autophagy protein Atg8 homolog microtubule-associated protein 1 light chain 3B (LC3B), besides another homolog, GABA receptor-associated protein-like 2 (GABARAPL2). Moreover, we demonstrate that colocalization of septin-3 and LC3B increases upon chemical autophagy induction in primary neuronal cells. Septin-3 is accumulated in primary neurons upon autophagy enhancement or blockade, similar to autophagy proteins. Using electron microscopy, we also show that septin-3 localizes to LC3B positive membranes and can be found at mitochondria. However, colocalization results of septin-3 and the early mitophagy marker PTEN-induced kinase 1 (PINK1) do not support that binding of septin-3 to mitochondria is mitophagy related. We conclude that septin-3 correlates with synaptic/neuronal autophagy, binds Atg8 and localizes to autophagic membranes that can be enhanced with chemical autophagy induction. Based on our results, elevated septin-3 levels might indicate enhanced or impeded autophagy in neurons.
Collapse
|
19
|
Radler MR, Spiliotis ET. Right place, right time - Spatial guidance of neuronal morphogenesis by septin GTPases. Curr Opin Neurobiol 2022; 75:102557. [PMID: 35609489 PMCID: PMC9968515 DOI: 10.1016/j.conb.2022.102557] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/07/2022] [Accepted: 04/20/2022] [Indexed: 12/21/2022]
Abstract
Neuronal morphogenesis is guided by outside-in signals and inside-out mechanisms, which require spatiotemporal precision. How the intracellular mechanisms of neuronal morphogenesis are spatiotemporally controlled is not well understood. Septins comprise a unique GTPase module, which consists of complexes with differential localizations and functions. Septins demarcate distinct membrane domains in neural precursor cells, orienting the axis of cell division and the sites of neurite formation. By controlling the localization of membrane and cytoskeletal proteins, septins promote axon-dendrite formation and polarity. Furthermore, septins modulate vesicle exocytosis at pre-synaptic terminals, and stabilize dendritic spines and post-synaptic densities in a phospho-regulatable manner. We posit that neuronal septins are topologically and functionally specialized for the spatiotemporal regulation of neuronal morphogenesis and plasticity.
Collapse
Affiliation(s)
- Megan R. Radler
- Department of Biology, Drexel University, Papadakis Integrated Sciences Building 423, 3245 Chestnut St, Philadelphia, PA 19104, USA
| | - Elias T. Spiliotis
- Department of Biology, Drexel University, Papadakis Integrated Sciences Building 423, 3245 Chestnut St, Philadelphia, PA 19104, USA
| |
Collapse
|
20
|
Kim OV, Litvinov RI, Mordakhanova ER, Bi E, Vagin O, Weisel JW. Contribution of septins to human platelet structure and function. iScience 2022; 25:104654. [PMID: 35832887 PMCID: PMC9272382 DOI: 10.1016/j.isci.2022.104654] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 04/23/2022] [Accepted: 06/17/2022] [Indexed: 11/29/2022] Open
Abstract
Although septins have been well-studied in nucleated cells, their role in anucleate blood platelets remains obscure. Here, we elucidate the contribution of septins to human platelet structure and functionality. We show that Septin-2 and Septin-9 are predominantly distributed at the periphery of resting platelets and co-localize strongly with microtubules. Activation of platelets by thrombin causes clustering of septins and impairs their association with microtubules. Inhibition of septin dynamics with forchlorfenuron (FCF) reduces thrombin-induced densification of septins and lessens their colocalization with microtubules in resting and activated platelets. Exposure to FCF alters platelet shape, suggesting that septins stabilize platelet cytoskeleton. FCF suppresses platelet integrin αIIbβ3 activation, promotes phosphatidylserine exposure on activated platelets, and induces P-selectin expression on resting platelets, suggesting septin involvement in these processes. Inhibition of septin dynamics substantially reduces platelet contractility and abrogates their spreading on fibrinogen-coated surfaces. Overall, septins strongly contribute to platelet structure, activation and biomechanics.
Collapse
Affiliation(s)
- Oleg V. Kim
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rustem I. Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elmira R. Mordakhanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Erfei Bi
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olga Vagin
- Department of Pediatrics, Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - John W. Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
21
|
Menon MB, Gaestel M. Editorial: Emerging Functions of Septins—Volume II. Front Cell Dev Biol 2022; 10:949824. [PMID: 35784463 PMCID: PMC9246257 DOI: 10.3389/fcell.2022.949824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 05/31/2022] [Indexed: 11/26/2022] Open
Affiliation(s)
- Manoj B. Menon
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
- *Correspondence: Manoj B. Menon, ; Matthias Gaestel,
| | - Matthias Gaestel
- Institute for Cell Biochemistry, Hannover Medical School, Hannover, Germany
- *Correspondence: Manoj B. Menon, ; Matthias Gaestel,
| |
Collapse
|
22
|
Byeon S, Werner B, Falter R, Davidsen K, Snyder C, Ong SE, Yadav S. Proteomic Identification of Phosphorylation-Dependent Septin 7 Interactors that Drive Dendritic Spine Formation. Front Cell Dev Biol 2022; 10:836746. [PMID: 35602601 PMCID: PMC9114808 DOI: 10.3389/fcell.2022.836746] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
Septins are a family of cytoskeletal proteins that regulate several important aspects of neuronal development. Septin 7 (Sept7) is enriched at the base of dendritic spines in excitatory neurons and mediates both spine formation and spine and synapse maturation. Phosphorylation at a conserved C-terminal tail residue of Sept7 mediates its translocation into the dendritic spine head to allow spine and synapse maturation. The mechanistic basis for postsynaptic stability and compartmentalization conferred by phosphorylated Sept7, however, is unclear. We report herein the proteomic identification of Sept7 phosphorylation-dependent neuronal interactors. Using Sept7 C-terminal phosphopeptide pulldown and biochemical assays, we show that the 14-3-3 family of proteins specifically interacts with Sept7 when phosphorylated at the T426 residue. Biochemically, we validate the interaction between Sept7 and 14-3-3 isoform gamma and show that 14-3-3 gamma is also enriched in the mature dendritic spine head. Furthermore, we demonstrate that interaction of phosphorylated Sept7 with 14-3-3 protects it from dephosphorylation, as expression of a 14-3-3 antagonist significantly decreases phosphorylated Sept7 in neurons. This study identifies 14-3-3 proteins as an important physiological regulator of Sept7 function in neuronal development.
Collapse
Affiliation(s)
- Sujin Byeon
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
| | - Bailey Werner
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Reilly Falter
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Kristian Davidsen
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Calvin Snyder
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Shao-En Ong
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Smita Yadav
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
23
|
Li M, Hu J, Mao H, Li D, Jiang Z, Sun Z, Yu T, Hu C, Xu X. Grass Carp ( Ctenopharyngodon idella) KAT8 Inhibits IFN 1 Response Through Acetylating IRF3/IRF7. Front Immunol 2022; 12:808159. [PMID: 35046960 PMCID: PMC8761793 DOI: 10.3389/fimmu.2021.808159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/09/2021] [Indexed: 12/30/2022] Open
Abstract
Post-translational modifications (PTMs), such as phosphorylation and ubiquitination, etc., have been reported to modulate the activities of IRF3 and IRF7. In this study, we found an acetyltransferase KAT8 in grass carp (CiKAT8, MW286472) that acetylated IRF3/IRF7 and then resulted in inhibition of IFN 1 response. CiKAT8 expression was up-regulated in the cells under poly I:C, B-DNA or Z-DNA stimulation as well as GCRV(strain 873) or SVCV infection. The acetyltransferase domain (MYST domain) of KAT8 promoted the acetylation of IRF3 and IRF7 through the direct interaction with them. So, the domain is essential for KAT8 function. Expectedly, KAT8 without MYST domain (KAT8-△264-487) was granularly aggregated in the nucleus and failed to down-regulate IFN 1 expression. Subcellular localization analysis showed that KAT8 protein was evenly distributed in the nucleus. In addition, we found that KAT8 inhibited the recruitment of IRF3 and IRF7 to ISRE response element. Taken together, our findings revealed that grass carp KAT8 blocked the activities of IRF3 and IRF7 by acetylating them, resulting in a low affinity interaction of ISRE response element with IRF3 and IRF7, and then inhibiting nucleic acids-induced innate immune response.
Collapse
Affiliation(s)
- Meifeng Li
- School of Life Science, Nanchang University, Nanchang, China
| | - Jihuan Hu
- School of Life Science, Nanchang University, Nanchang, China
| | - Huiling Mao
- School of Life Science, Nanchang University, Nanchang, China
| | - Dongming Li
- Fuzhou Medical College, Nanchang University, Fuzhou, China
| | - Zeyin Jiang
- School of Life Science, Nanchang University, Nanchang, China
| | - Zhichao Sun
- School of Life Science, Nanchang University, Nanchang, China
| | - Tingting Yu
- School of Life Science, Nanchang University, Nanchang, China
| | - Chengyu Hu
- School of Life Science, Nanchang University, Nanchang, China
| | - Xiaowen Xu
- School of Life Science, Nanchang University, Nanchang, China.,State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
24
|
Balmik AA, Chinnathambi S. Inter-relationship of Histone Deacetylase-6 with cytoskeletal organization and remodeling. Eur J Cell Biol 2022; 101:151202. [DOI: 10.1016/j.ejcb.2022.151202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 11/30/2022] Open
|
25
|
Cavini IA, Leonardo DA, Rosa HVD, Castro DKSV, D'Muniz Pereira H, Valadares NF, Araujo APU, Garratt RC. The Structural Biology of Septins and Their Filaments: An Update. Front Cell Dev Biol 2021; 9:765085. [PMID: 34869357 PMCID: PMC8640212 DOI: 10.3389/fcell.2021.765085] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/27/2021] [Indexed: 01/22/2023] Open
Abstract
In order to fully understand any complex biochemical system from a mechanistic point of view, it is necessary to have access to the three-dimensional structures of the molecular components involved. Septins and their oligomers, filaments and higher-order complexes are no exception. Indeed, the spontaneous recruitment of different septin monomers to specific positions along a filament represents a fascinating example of subtle molecular recognition. Over the last few years, the amount of structural information available about these important cytoskeletal proteins has increased dramatically. This has allowed for a more detailed description of their individual domains and the different interfaces formed between them, which are the basis for stabilizing higher-order structures such as hexamers, octamers and fully formed filaments. The flexibility of these structures and the plasticity of the individual interfaces have also begun to be understood. Furthermore, recently, light has been shed on how filaments may bundle into higher-order structures by the formation of antiparallel coiled coils involving the C-terminal domains. Nevertheless, even with these advances, there is still some way to go before we fully understand how the structure and dynamics of septin assemblies are related to their physiological roles, including their interactions with biological membranes and other cytoskeletal components. In this review, we aim to bring together the various strands of structural evidence currently available into a more coherent picture. Although it would be an exaggeration to say that this is complete, recent progress seems to suggest that headway is being made in that direction.
Collapse
Affiliation(s)
- Italo A Cavini
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| | - Diego A Leonardo
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| | - Higor V D Rosa
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| | - Danielle K S V Castro
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil.,São Carlos Institute of Chemistry, University of São Paulo, São Carlos, Brazil
| | | | | | - Ana P U Araujo
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| | - Richard C Garratt
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| |
Collapse
|
26
|
Zhovmer AS, Manning A, Smith C, Hayes JB, Burnette DT, Wang J, Cartagena-Rivera AX, Dokholyan NV, Singh RK, Tabdanov ED. Mechanical Counterbalance of Kinesin and Dynein Motors in a Microtubular Network Regulates Cell Mechanics, 3D Architecture, and Mechanosensing. ACS NANO 2021; 15:17528-17548. [PMID: 34677937 PMCID: PMC9291236 DOI: 10.1021/acsnano.1c04435] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Microtubules (MTs) and MT motor proteins form active 3D networks made of unstretchable cables with rod-like bending mechanics that provide cells with a dynamically changing structural scaffold. In this study, we report an antagonistic mechanical balance within the dynein-kinesin microtubular motor system. Dynein activity drives the microtubular network inward compaction, while isolated activity of kinesins bundles and expands MTs into giant circular bands that deform the cell cortex into discoids. Furthermore, we show that dyneins recruit MTs to sites of cell adhesion, increasing the topographic contact guidance of cells, while kinesins antagonize it via retraction of MTs from sites of cell adhesion. Actin-to-microtubule translocation of septin-9 enhances kinesin-MT interactions, outbalances the activity of kinesins over that of dyneins, and induces the discoid architecture of cells. These orthogonal mechanisms of MT network reorganization highlight the existence of an intricate mechanical balance between motor activities of kinesins and dyneins that controls cell 3D architecture, mechanics, and cell-microenvironment interactions.
Collapse
Affiliation(s)
- Alexander S. Zhovmer
- Center
for Biologics Evaluation and Research, U.S.
Food and Drug Administration, Silver Spring, Maryland 20903, United States
| | - Alexis Manning
- Center
for Biologics Evaluation and Research, U.S.
Food and Drug Administration, Silver Spring, Maryland 20903, United States
| | - Chynna Smith
- Section
on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - James B. Hayes
- Department
of Cell and Developmental Biology, Vanderbilt Medical Center, University of Vanderbilt, Nashville, Tennessee 37232, United States
| | - Dylan T. Burnette
- Department
of Cell and Developmental Biology, Vanderbilt Medical Center, University of Vanderbilt, Nashville, Tennessee 37232, United States
| | - Jian Wang
- Department
of Pharmacology, Penn State College of Medicine, Pennsylvania State University, Hummelstown, Pennsylvania 17036, United States
| | - Alexander X. Cartagena-Rivera
- Section
on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Nikolay V. Dokholyan
- Department
of Pharmacology, Penn State College of Medicine, Pennsylvania State University, Hummelstown, Pennsylvania 17036, United States
- Department
of Biochemistry & Molecular Biology, Penn State College of Medicine, Pennsylvania State University, Hershey, Pennsylvania 17033, United States
| | - Rakesh K. Singh
- Department
of Obstetrics and Gynecology, University
of Rochester Medical Center, Rochester, New York 14620, United States
| | - Erdem D. Tabdanov
- Department
of Pharmacology, Penn State College of Medicine, Pennsylvania State University, Hummelstown, Pennsylvania 17036, United States
| |
Collapse
|
27
|
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: 4.7] [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
|
28
|
Bedogni F, Hevner RF. Cell-Type-Specific Gene Expression in Developing Mouse Neocortex: Intermediate Progenitors Implicated in Axon Development. Front Mol Neurosci 2021; 14:686034. [PMID: 34321999 PMCID: PMC8313239 DOI: 10.3389/fnmol.2021.686034] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/03/2021] [Indexed: 01/06/2023] Open
Abstract
Cerebral cortex projection neurons (PNs) are generated from intermediate progenitors (IPs), which are in turn derived from radial glial progenitors (RGPs). To investigate developmental processes in IPs, we profiled IP transcriptomes in embryonic mouse neocortex, using transgenic Tbr2-GFP mice, cell sorting, and microarrays. These data were used in combination with in situ hybridization to ascertain gene sets specific for IPs, RGPs, PNs, interneurons, and other neural and non-neural cell types. RGP-selective transcripts (n = 419) included molecules for Notch receptor signaling, proliferation, neural stem cell identity, apical junctions, necroptosis, hippo pathway, and NF-κB pathway. RGPs also expressed specific genes for critical interactions with meningeal and vascular cells. In contrast, IP-selective genes (n = 136) encoded molecules for activated Delta ligand presentation, epithelial-mesenchymal transition, core planar cell polarity (PCP), axon genesis, and intrinsic excitability. Interestingly, IPs expressed several “dependence receptors” (Unc5d, Dcc, Ntrk3, and Epha4) that induce apoptosis in the absence of ligand, suggesting a competitive mechanism for IPs and new PNs to detect key environmental cues or die. Overall, our results imply a novel role for IPs in the patterning of neuronal polarization, axon differentiation, and intrinsic excitability prior to mitosis. Significantly, IPs highly express Wnt-PCP, netrin, and semaphorin pathway molecules known to regulate axon polarization in other systems. In sum, IPs not only amplify neurogenesis quantitatively, but also molecularly “prime” new PNs for axogenesis, guidance, and excitability.
Collapse
Affiliation(s)
| | - Robert F Hevner
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
29
|
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: 8] [Impact Index Per Article: 2.7] [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
|
30
|
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
|
31
|
Di Paolo A, Farias J, Garat J, Macklin A, Ignatchenko V, Kislinger T, Sotelo Silveira J. Rat Sciatic Nerve Axoplasm Proteome Is Enriched with Ribosomal Proteins during Regeneration Processes. J Proteome Res 2021; 20:2506-2520. [PMID: 33793244 DOI: 10.1021/acs.jproteome.0c00980] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Axons are complex subcellular compartments that are extremely long in relation to cell bodies, especially in peripheral nerves. Many processes are required and regulated during axon injury, including anterograde and retrograde transport, glia-to-axon macromolecular transfer, and local axonal protein synthesis. Many in vitro omics approaches have been used to gain insight into these processes, but few have been applied in vivo. Here we adapted the osmotic ex vivo axoplasm isolation method and analyzed the adult rat sciatic-nerve-extruded axoplasm by label-free quantitative proteomics before and after injury. 2087 proteins groups were detected in the axoplasm, revealing translation machinery and microtubule-associated proteins as the most overrepresented biological processes. Ribosomal proteins (73) were detected in the uninjured axoplasm and increased their levels after injury but not within whole sciatic nerves. Meta-analysis showed that detected ribosomal proteins were present in in vitro axonal proteomes. Because local protein synthesis is important for protein localization, we were interested in detecting the most abundant newly synthesized axonal proteins in vivo. With an MS/MS-BONCAT approach, we detected 42 newly synthesized protein groups. Overall, our work indicates that proteomics profiling is useful for local axonal interrogation and suggests that ribosomal proteins may play an important role, especially during injury.
Collapse
Affiliation(s)
- Andres Di Paolo
- Departamento de Proteínas y Ácidos Nucleicos, IIBCE, 11600 Montevideo, Uruguay.,Departamento de Genómica, IIBCE, 11600 Montevideo, Uruguay
| | | | - Joaquin Garat
- Departamento de Genómica, IIBCE, 11600 Montevideo, Uruguay
| | - Andrew Macklin
- Princess Margaret Cancer Centre, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Vladimir Ignatchenko
- Princess Margaret Cancer Centre, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, 101 College Street, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - José Sotelo Silveira
- Departamento de Genómica, IIBCE, 11600 Montevideo, Uruguay.,Departamento de Biología Celular y Molecular, Facultad de Ciencias, 11400 Montevideo, Uruguay
| |
Collapse
|
32
|
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: 3.7] [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
|
33
|
Huang W, Zhu L, Cao G, Xie P, Song Y, Huang J, Chen X, Cai Z. Integrated Proteomics and Metabolomics Assessment Indicated Metabolic Alterations in Hypothalamus of Mice Exposed to Triclosan. Chem Res Toxicol 2021; 34:1319-1328. [PMID: 33611912 DOI: 10.1021/acs.chemrestox.0c00514] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Triclosan (TCS) is a ubiquitous antimicrobial used in many daily consumer products. It has been reported to induce endocrine disrupting effects at low doses in mammals, disturbing sex hormone function and thyroid function. The hypothalamus plays a crucial role in the maintenance of neuroendocrine function and energy homeostasis. We speculated that the adverse effects of TCS might be related to the disturbance of metabolic processes in hypothalamus. The present study aimed at investigating the effects of TCS exposure on the protein and metabolite profiles in hypothalamus of mice. Male C57BL/6 mice were orally exposed to TCS at the dosage of 10 mg/kg/d for 13 weeks. The hypothalamus was isolated and processed for mass spectrometry (MS)-based proteomics and metabolomics analyses. The results showed that a 10.6% decrease (P = 0.066) in body weight gain was observed in the TCS exposure group compared with vehicle control group. Differential analysis defined 52 proteins and 57 metabolites that delineated TCS exposed mice from vehicle controls. Among the differential features, multiple proteins and metabolites were found to play vital roles in neuronal signaling and function. Bioinformatics analysis revealed that these differentially expressed proteins and metabolites were involved in four major biological processes, including glucose metabolism, purine metabolism, neurotransmitter release, and neural plasticity, suggesting the disturbance of homeostasis in energy metabolism, mitochondria function, neurotransmitter system, and neuronal function. Our results may provide insights into the neurotoxicity of TCS and extend our understanding of the biological effects induced by TCS exposure.
Collapse
Affiliation(s)
- Wei Huang
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China.,School of Environment, Jinan University, Guangzhou 510632, China
| | - Lin Zhu
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China
| | - Guodong Cao
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China
| | - Peisi Xie
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yuanyuan Song
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jialing Huang
- School of Environment, Jinan University, Guangzhou 510632, China
| | - Xiangfeng Chen
- Shandong Analysis and Test Center, Qilu University of Technology, Jinan, Shandong, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
34
|
Yang Q, Li Q, Liu Z, Wang D, Guo Y, Li X, Tang Y, Li H, Dong B, Zhi C. Dendrites in Zn-Based Batteries. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2001854. [PMID: 33103828 DOI: 10.1002/adma.202001854] [Citation(s) in RCA: 277] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/01/2020] [Indexed: 05/18/2023]
Abstract
Aqueous Zn batteries that provide a synergistic integration of absolute safety and high energy density have been considered as highly promising energy-storage systems for powering electronics. Despite the rapid progress made in developing high-performance cathodes and electrolytes, the underestimated but non-negligible dendrites of Zn anode have been observed to shorten battery lifespan. Herein, this dendrite issue in Zn anodes, with regard to fundamentals, protection strategies, characterization techniques, and theoretical simulations, is systematically discussed. An overall comparison between the Zn dendrite and its Li and Al counterparts, to highlight their differences in both origin and topology, is given. Subsequently, in-depth clarifications of the specific influence factors of Zn dendrites, including the accumulation effect and the cathode loading mass (a distinct factor for laboratory studies and practical applications) are presented. Recent advances in Zn dendrite protection are then comprehensively summarized and categorized to generate an overview of respective superiorities and limitations of various strategies. Accordingly, theoretical computations and advanced characterization approaches are introduced as mechanism guidelines and measurement criteria for dendrite suppression, respectively. The concluding section emphasizes future challenges in addressing the Zn dendrite issue and potential approaches to further promoting the lifespan of Zn batteries.
Collapse
Affiliation(s)
- Qi Yang
- Department of Materials Science and Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR, 999077, China
| | - Qing Li
- Department of Materials Science and Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR, 999077, China
| | - Zhuoxin Liu
- Department of Materials Science and Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR, 999077, China
| | - Donghong Wang
- Department of Materials Science and Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR, 999077, China
| | - Ying Guo
- Department of Materials Science and Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR, 999077, China
| | - Xinliang Li
- Department of Materials Science and Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR, 999077, China
| | - Yongchao Tang
- Songshan Lake Materials Laboratory, Dongguan, 523808, China
| | - Hongfei Li
- Songshan Lake Materials Laboratory, Dongguan, 523808, China
| | - Binbin Dong
- National Engineering Research Center for Advanced Polymer Processing Technology, Zhengzhou University, Zhengzhou, Henan, 450002, China
| | - Chunyi Zhi
- Department of Materials Science and Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR, 999077, China
- Center for Advanced Nuclear Safety and Sustainable Development, City University of Hong Kong, Kowloon, 999077, Hong Kong
| |
Collapse
|
35
|
Ageta-Ishihara N, Kinoshita M. Developmental and postdevelopmental roles of septins in the brain. Neurosci Res 2020; 170:6-12. [PMID: 33159992 DOI: 10.1016/j.neures.2020.08.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/21/2020] [Accepted: 08/23/2020] [Indexed: 11/25/2022]
Abstract
Morphogenetic processes during brain development and postdevelopmental remodeling of neural architecture depend on the exquisite interplay between the microtubule- and actin-based cytoskeletal systems. Accumulation of evidence indicates cooperative roles of another cytoskeletal system composed of the septin family. Here we overview experimental findings on mammalian septins and their hypothetical roles in the proliferation of neural progenitor cells, neurite development, synapse formation and regulations. The diverse, mostly unexpected phenotypes obtained from gain- and loss-of-function mutants point to unknown molecular network to be elucidated, which may underlie pathogenetic processes of infectious diseases and neuropsychiatric disorders in humans.
Collapse
Affiliation(s)
- Natsumi Ageta-Ishihara
- Division of Biological Science, Nagoya University Graduate School of Science, Furo, Chikusa, Nagoya 464-8602, Japan.
| | - Makoto Kinoshita
- Division of Biological Science, Nagoya University Graduate School of Science, Furo, Chikusa, Nagoya 464-8602, Japan.
| |
Collapse
|
36
|
Chen TY, Lin TC, Kuo PL, Chen ZR, Cheng HL, Chao YY, Syu JS, Lu FI, Wang CY. Septin 7 is a centrosomal protein that ensures S phase entry and microtubule nucleation by maintaining the abundance of p150 glued. J Cell Physiol 2020; 236:2706-2724. [PMID: 32869310 DOI: 10.1002/jcp.30037] [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: 01/02/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022]
Abstract
Septins play important roles in regulating development and differentiation. Septin 7 (SEPT7) is a crucial component in orchestrating the septin core complex into highly ordered filamentous structures. Here, we showed that genetic depletion of SEPT7 or treatment with forchlorfenuron (FCF; a compound known to affect septin filament assembly) led to reduced the S phase entry in cell models and zebrafish embryos. In addition to colocalizing with actin filaments, SEPT7 resided in the centrosome, and SEPT7 depletion led to aberrant mitotic spindle pole formation. This mitotic defect was rescued in SEPT7-deficient cells by wild-type SEPT7, suggesting that SEPT7 maintained mitotic spindle poles. In addition, we observed disorganized microtubule nucleation and reduced cell migration with SEPT7 depletion. Furthermore, SEPT7 formed a complex with and maintained the abundance of p150glued , the component of centriole subdistal appendages. Depletion of p150glued resulted in a phenotype reminiscent of SEPT7-deficient cells, and overexpression of p150glued reversed the defective phenotypes. Thus, SEPT7 is a centrosomal protein that maintains proper cell proliferation and microtubule array formation via maintaining the abundance of p150glued .
Collapse
Affiliation(s)
- Ting-Yu Chen
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Chien Lin
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pao-Lin Kuo
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Zi-Rong Chen
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan.,The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Hui-Ling Cheng
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ying Chao
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jhih-Siang Syu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Fu-I Lu
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan.,The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Chia-Yih Wang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
37
|
Yin Q, Shen L, Qi Y, Song D, Ye L, Peng Y, Wang Y, Jin Z, Ning G, Wang W, Lin D, Wang S. Decreased SIRT1 expression in the peripheral blood of patients with Graves' disease. J Endocrinol 2020; 246:161-173. [PMID: 32485674 PMCID: PMC7354706 DOI: 10.1530/joe-19-0501] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/02/2020] [Indexed: 11/08/2022]
Abstract
SIRT1, a class III histone/protein deacetylase (HDAC), has been associated with autoimmune diseases. There is a paucity of data about the role of SIRT1 in Graves' disease. The aim of this study was to investigate the role of SIRT1 in the pathogenesis of GD. Here, we showed that SIRT1 expression and activity were significantly decreased in GD patients compared with healthy controls. The NF-κB pathway was activated in the peripheral blood of GD patients. The reduced SIRT1 levels correlated strongly with clinical parameters. In euthyroid patients, SIRT1 expression was markedly upregulated and NF-κB downstream target gene expression was significantly reduced. SIRT1 inhibited the NF-κB pathway activity by deacetylating P65. These results demonstrate that reduced SIRT1 expression and activity contribute to the activation of the NF-κB pathway and may be involved in the pathogenesis of GD.
Collapse
Affiliation(s)
- Qinglei Yin
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Liyun Shen
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Yicheng Qi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, RenJi Hospital, Shanghai Jiao-Tong University School of Medicine, Pudong, Shanghai, China
| | - Dalong Song
- Reproductive Medicine Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Lei Ye
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Ying Peng
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Yanqiu Wang
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Zhou Jin
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Guang Ning
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Weiqing Wang
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
| | - Dongping Lin
- Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Affiliated Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- Correspondence should be addressed to D Lin and S Wang: or
| | - Shu Wang
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Institute of EndocrineRuijin Hospital, Shanghai Jiao-Tong University School of Medicine, China
- Correspondence should be addressed to D Lin and S Wang: or
| |
Collapse
|
38
|
SEPT7 regulates Ca 2+ entry through Orai channels in human neural progenitor cells and neurons. Cell Calcium 2020; 90:102252. [PMID: 32682163 DOI: 10.1016/j.ceca.2020.102252] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [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
|
39
|
Niklison-Chirou MV, Agostini M, Amelio I, Melino G. Regulation of Adult Neurogenesis in Mammalian Brain. Int J Mol Sci 2020; 21:ijms21144869. [PMID: 32660154 PMCID: PMC7402357 DOI: 10.3390/ijms21144869] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022] Open
Abstract
Adult neurogenesis is a multistage process by which neurons are generated and integrated into existing neuronal circuits. In the adult brain, neurogenesis is mainly localized in two specialized niches, the subgranular zone (SGZ) of the dentate gyrus and the subventricular zone (SVZ) adjacent to the lateral ventricles. Neurogenesis plays a fundamental role in postnatal brain, where it is required for neuronal plasticity. Moreover, perturbation of adult neurogenesis contributes to several human diseases, including cognitive impairment and neurodegenerative diseases. The interplay between extrinsic and intrinsic factors is fundamental in regulating neurogenesis. Over the past decades, several studies on intrinsic pathways, including transcription factors, have highlighted their fundamental role in regulating every stage of neurogenesis. However, it is likely that transcriptional regulation is part of a more sophisticated regulatory network, which includes epigenetic modifications, non-coding RNAs and metabolic pathways. Here, we review recent findings that advance our knowledge in epigenetic, transcriptional and metabolic regulation of adult neurogenesis in the SGZ of the hippocampus, with a special attention to the p53-family of transcription factors.
Collapse
Affiliation(s)
- Maria Victoria Niklison-Chirou
- Centre for Therapeutic Innovation (CTI-Bath), Department of Pharmacy & Pharmacology, University of Bath, Bath BA2 7AY, UK;
- Blizard Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Massimiliano Agostini
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.A.); (I.A.)
| | - Ivano Amelio
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.A.); (I.A.)
- School of Life Sciences, University of Nottingham, Nottingham NG7 2HU, UK
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.A.); (I.A.)
- Correspondence:
| |
Collapse
|
40
|
Jiang Y, Liu L, Xiang Q, He X, Wang Y, Zhou D, Zou C, Chen Q, Peng M, He J, Jiang X, Xiang T, Yang Y. SEPT9_v2, frequently silenced by promoter hypermethylation, exerts anti-tumor functions through inactivation of Wnt/β-catenin signaling pathway via miR92b-3p/FZD10 in nasopharyngeal carcinoma cells. Clin Epigenetics 2020; 12:41. [PMID: 32138771 PMCID: PMC7059696 DOI: 10.1186/s13148-020-00833-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/26/2020] [Indexed: 12/24/2022] Open
Abstract
Background Nasopharyngeal carcinoma tends to present at an advanced stage because the primary anatomic site is located in a less visible area and its clinical symptoms are nonspecific. Prognosis of advanced nasopharyngeal carcinoma cases remains disappointing. SEPT9 is a methylation-based biomarker approved by the US Food and Drug Administration for colorectal cancer screening and diagnosis. Interestingly, downregulation of SEPT9, especially SEPT9_v2, mediated by promoter hypermethylation has been also detected in head and neck squamous cell carcinoma than in head and neck squamous epithelium, while other SEPT9 variants did not. These reasons above indicate a crucial role of SEPT9_v2 in cancer progression. Therefore, we address the methylation status of SEPT9_v2 in nasopharyngeal carcinoma and explore the role of SEPT9_v2 in nasopharyngeal carcinoma proliferation and cancer progression. Results SEPT9_v2 expression was found to be downregulated via promoter methylation in nasopharyngeal carcinoma cell lines and tissues. Ectopic expression of SEPT9_v2 induced G0/G1 cell cycle arrest and apoptosis, which exerted an inhibitory effect in cell proliferation and colony formation. Additionally, nasopharyngeal carcinoma cell migration and invasion were shown to be inhibited by SEPT9_v2. Furthermore, our data suggested that SEPT9_v2 inhibits proliferation and migration of nasopharyngeal carcinoma cells through inactivation of the Wnt/β-catenin signaling pathway via miR92b-3p/FZD10. Conclusions This study delineates SEPT9_v2, frequently silenced by promoter hypermethylation, exerts anti-tumor functions through inactivation of the Wnt/β-catenin signaling pathway via miR92b-3p/FZD10 in nasopharyngeal carcinoma cells and, hence, SEPT9_v2 may be a promising therapeutic target and biomarker for nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Lei Liu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Qin Xiang
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Xiaoqian He
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Yan Wang
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Dishu Zhou
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Can Zou
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Qian Chen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Mingyu Peng
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Jin He
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Xianyao Jiang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Tingxiu Xiang
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Yucheng Yang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
41
|
Qiu R, Runxiang Q, Geng A, Liu J, Xu CW, Menon MB, Gaestel M, Lu Q. SEPT7 Interacts with KIF20A and Regulates the Proliferative State of Neural Progenitor Cells During Cortical Development. Cereb Cortex 2019; 30:3030-3043. [PMID: 31813992 DOI: 10.1093/cercor/bhz292] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/01/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023] Open
Abstract
Balanced proliferation and differentiation of neural progenitor cells (NPCs) are critical for brain development, but how the process is regulated and what components of the cell division machinery is involved are not well understood. Here we report that SEPT7, a cell division regulator originally identified in Saccharomyces cerevisiae, interacts with KIF20A in the intercellular bridge of dividing NPCs and plays an essential role in maintaining the proliferative state of NPCs during cortical development. Knockdown of SEPT7 in NPCs results in displacement of KIF20A from the midbody and early neuronal differentiation. NPC-specific inducible knockout of Sept7 causes early cell cycle exit, precocious neuronal differentiation, and ventriculomegaly in the cortex, but surprisingly does not lead to noticeable cytokinesis defect. Our data uncover an interaction of SEPT7 and KIF20A during NPC divisions and demonstrate a crucial role of SEPT7 in cell fate determination. In addition, this study presents a functional approach for identifying additional cell fate regulators of the mammalian brain.
Collapse
Affiliation(s)
- Runxiang Qiu
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Qiu Runxiang
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Anqi Geng
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.,Institute of Medical Research, Northwestern Polytechnical University, Xian, Shaanxi Province, China
| | - Jiancheng Liu
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - C Wilson Xu
- Balto Pharmaceuticals, Inc., South Pasadena, CA 91030, USA
| | - Manoj B Menon
- Institute of Cell Biochemistry, Hannover Medical School, Hannover 30625, Germany.,Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New-Delhi 110016, India
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover 30625, Germany
| | - Qiang Lu
- Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
42
|
Bucher M, Fanutza T, Mikhaylova M. Cytoskeletal makeup of the synapse: Shaft versus spine. Cytoskeleton (Hoboken) 2019; 77:55-64. [PMID: 31762205 DOI: 10.1002/cm.21583] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/13/2022]
Abstract
The ability of neurons to communicate and store information depends on the activity of synapses which can be located on small protrusions (dendritic spines) or directly on the dendritic shaft. The formation, plasticity, and stability of synapses are regulated by the neuronal cytoskeleton. Actin filaments together with microtubules, neurofilaments, septins, and scaffolding proteins orchestrate the structural organization of both shaft and spine synapses, enabling their efficacy in response to synaptic activation. Synapses critically depend on several factors, which are also mediated by the cytoskeleton, including transport and delivery of proteins from the soma, protein synthesis, as well as surface diffusion of membrane proteins. In this minireview, we focus on recent progress made in the field of cytoskeletal elements of the postsynapse and discuss the differences and similarities between synapses located in the spines versus dendritic shaft.
Collapse
Affiliation(s)
- Michael Bucher
- DFG Emmy Noether Group 'Neuronal Protein Transport', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tomas Fanutza
- DFG Emmy Noether Group 'Neuronal Protein Transport', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marina Mikhaylova
- DFG Emmy Noether Group 'Neuronal Protein Transport', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
43
|
Yan C, Wang F, Peng Y, Williams CR, Jenkins B, Wildonger J, Kim HJ, Perr JB, Vaughan JC, Kern ME, Falvo MR, O'Brien ET, Superfine R, Tuthill JC, Xiang Y, Rogers SL, Parrish JZ. Microtubule Acetylation Is Required for Mechanosensation in Drosophila. Cell Rep 2019; 25:1051-1065.e6. [PMID: 30355484 DOI: 10.1016/j.celrep.2018.09.075] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 08/04/2018] [Accepted: 09/24/2018] [Indexed: 01/13/2023] Open
Abstract
At the cellular level, α-tubulin acetylation alters the structure of microtubules to render them mechanically resistant to compressive forces. How this biochemical property of microtubule acetylation relates to mechanosensation remains unknown, although prior studies have shown that microtubule acetylation influences touch perception. Here, we identify the major Drosophila α-tubulin acetylase (dTAT) and show that it plays key roles in several forms of mechanosensation. dTAT is highly expressed in the larval peripheral nervous system (PNS), but it is largely dispensable for neuronal morphogenesis. Mutation of the acetylase gene or the K40 acetylation site in α-tubulin impairs mechanical sensitivity in sensory neurons and behavioral responses to gentle touch, harsh touch, gravity, and vibration stimuli, but not noxious thermal stimulus. Finally, we show that dTAT is required for mechanically induced activation of NOMPC, a microtubule-associated transient receptor potential channel, and functions to maintain integrity of the microtubule cytoskeleton in response to mechanical stimulation.
Collapse
Affiliation(s)
- Connie Yan
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Fei Wang
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Yun Peng
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Claire R Williams
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Brian Jenkins
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jill Wildonger
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Hyeon-Jin Kim
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Jonathan B Perr
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA; Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Megan E Kern
- Department of Physics & Astronomy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | - Michael R Falvo
- Department of Physics & Astronomy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | - E Timothy O'Brien
- Department of Physics & Astronomy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | - Richard Superfine
- Department of Applied and Physical Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | - John C Tuthill
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Yang Xiang
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Stephen L Rogers
- Department of Biology, Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280, USA; Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280, USA.
| | - Jay Z Parrish
- Department of Biology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
44
|
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: 30] [Impact Index Per Article: 6.0] [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
|
45
|
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: 19] [Impact Index Per Article: 3.8] [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
|
46
|
Liang T, Hou X, Zhou Y, Yang X, Fang H. Design, Synthesis, and Biological Evaluation of 2,4-Imidazolinedione Derivatives as HDAC6 Isoform-Selective Inhibitors. ACS Med Chem Lett 2019; 10:1122-1127. [PMID: 31413795 DOI: 10.1021/acsmedchemlett.9b00084] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 07/05/2019] [Indexed: 01/23/2023] Open
Abstract
Histone deacetylase 6 (HDAC6) has emerged as a promising drug target for various human diseases, including diverse neurodegenerative diseases and cancer. Herein, we reported a series of 2,4-imidazolinedione derivatives as novel HDAC6 isoform-selective inhibitors based on structure-based drug design. Most target compounds exhibit good profiles in a preliminary screening concerning HDAC6 inhibitory activities. Moreover, the most active compound 10c increases the acetylation level of α-tubulin with little effect on the acetylation of histone H3. Further biological evaluation suggested that potent compound 10c, which possesses good antiproliferative activity, could induce apoptosis in HL-60 cell by activating caspase 3.
Collapse
Affiliation(s)
- Tao Liang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, P. R. China
| | - Xuben Hou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, P. R. China
| | - Yi Zhou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, P. R. China
| | - Xinying Yang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, P. R. China
| | - Hao Fang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong 250012, P. R. China
| |
Collapse
|
47
|
Falk J, Boubakar L, Castellani V. Septin functions during neuro-development, a yeast perspective. Curr Opin Neurobiol 2019; 57:102-109. [DOI: 10.1016/j.conb.2019.01.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/09/2019] [Accepted: 01/13/2019] [Indexed: 12/24/2022]
|
48
|
Abstract
Septins are widely recognized as a component of the cytoskeleton that is essential for cell division, and new work has shown that septins can recognise cell shape by assembling into filaments on membrane regions that display micrometer-scale curvature (e.g. at the cytokinetic furrow). Moreover, infection biology studies have illuminated important roles for septins in mediating the outcome of host-microbe interactions. In this Review, we discuss a selection of mechanistic insights recently gained from studying three infection paradigms: the rice blast fungus Magnaporthe oryzae, the poxvirus family member vaccinia virus and the Gram-negative bacterium Shigella flexneri These studies have respectively discovered that higher-order septin assemblies enable fungal invasion into plant cells, entrap viral particles at the plasma membrane and recognize dividing bacterial cells for delivery to lysosomes. Collectively, these insights illustrate how studying septin biology during microbial infection can provide fundamental advances in both cell and infection biology, and suggest new concepts underlying infection control.
Collapse
Affiliation(s)
- Hoan Van Ngo
- Department of Immunology & Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Serge Mostowy
- Department of Immunology & Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| |
Collapse
|
49
|
Banko M, Mucha-Kruczynska I, Weise C, Heyd F, Ewers H. A homozygous genome-edited Sept2-EGFP fibroblast cell line. Cytoskeleton (Hoboken) 2019; 76:73-82. [PMID: 30924304 PMCID: PMC6593442 DOI: 10.1002/cm.21518] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/22/2019] [Accepted: 03/23/2019] [Indexed: 01/09/2023]
Abstract
Septins are a conserved, essential family of GTPases that interact with actin, microtubules, and membranes and form scaffolds and diffusion barriers in cells. Several of the 13 known mammalian septins assemble into nonpolar, multimeric complexes that can further polymerize into filamentous structures. While some GFP‐coupled septins have been described, overexpression of GFP‐tagged septins often leads to artifacts in localization and function. To overcome this ubiquitous problem, we have here generated a genome‐edited rat fibroblast cell line expressing Septin 2 (Sept2) coupled to enhanced green fluorescent protein (EGFP) from both chromosomal loci. We characterize these cells by genomic polymerase chain reaction (PCR) for genomic integration, by western blot and reverse transcriptase‐PCR for expression, by immunofluorescence and immunoprecipitation for the colocalization of septins with one another and cellular structures and for complex formation of different septins. By live cell imaging, proliferation and migration assays we investigate proper function of septins in these cells. We find that EGFP is incorporated into both chromosomal loci and only EGFP‐coupled Sept2 is expressed in homozygous cells. We find that endogenous Sept2‐EGFP exhibits expression levels, localization and incorporation into cellular septin complexes similar to the wt in these cells. The expression level of other septins is not perturbed and cell division and cell migration proceed normally. We expect our cell line to be a useful tool for the cell biology of septins, especially for quantitative biology.
Collapse
Affiliation(s)
- Monika Banko
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK.,Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Iwona Mucha-Kruczynska
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK.,Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Christoph Weise
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Florian Heyd
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Helge Ewers
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK.,Department of Biology, ETH Zürich, Zürich, Switzerland.,Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
50
|
Aprile D, Fruscione F, Baldassari S, Fadda M, Ferrante D, Falace A, Buhler E, Sartorelli J, Represa A, Baldelli P, Benfenati F, Zara F, Fassio A. TBC1D24 regulates axonal outgrowth and membrane trafficking at the growth cone in rodent and human neurons. Cell Death Differ 2019; 26:2464-2478. [PMID: 30858606 DOI: 10.1038/s41418-019-0313-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 01/25/2019] [Accepted: 02/20/2019] [Indexed: 01/04/2023] Open
Abstract
Mutations in TBC1D24 are described in patients with a spectrum of neurological diseases, including mild and severe epilepsies and complex syndromic phenotypes such as Deafness, Onycodystrophy, Osteodystrophy, Mental Retardation and Seizure (DOORS) syndrome. The product of TBC1D24 is a multifunctional protein involved in neuronal development, regulation of synaptic vesicle trafficking, and protection from oxidative stress. Although pathogenic mutations in TBC1D24 span the entire coding sequence, no clear genotype/phenotype correlations have emerged. However most patients bearing predicted loss of function mutations exhibit a severe neurodevelopmental disorder. Aim of the study is to investigate the impact of TBC1D24 knockdown during the first stages of neuronal differentiation when axonal specification and outgrowth take place. In rat cortical primary neurons silenced for TBC1D24, we found defects in axonal specification, the maturation of axonal initial segment and action potential firing. The axonal phenotype was accompanied by an impairment of endocytosis at the growth cone and an altered activation of the TBC1D24 molecular partner ADP ribosylation factor 6. Accordingly, acute knockdown of TBC1D24 in cerebrocortical neurons in vivo analogously impairs callosal projections. The axonal defect was also investigated in human induced pluripotent stem cell-derived neurons from patients carrying TBC1D24 mutations. Reprogrammed neurons from a patient with severe developmental encephalopathy show significant axon formation defect that were absent from reprogrammed neurons of a patient with mild early onset epilepsy. Our data reveal that alterations of membrane trafficking at the growth cone induced by TBC1D24 loss of function cause axonal and excitability defects. The axonal phenotype correlates with the disease severity and highlight an important role for TBC1D24 in connectivity during brain development.
Collapse
Affiliation(s)
- Davide Aprile
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Floriana Fruscione
- Laboratory of Neurogenetics and Neuroscience, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Simona Baldassari
- Laboratory of Neurogenetics and Neuroscience, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Manuela Fadda
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Daniele Ferrante
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Antonio Falace
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Children's Hospital A. Meyer-University of Florence, Florence, Italy
| | | | - Jacopo Sartorelli
- Laboratory of Neurogenetics and Neuroscience, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Alfonso Represa
- INMED, Aix-Marseille University, INSERM U1249, Marseille, France
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fabio Benfenati
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Center of Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Federico Zara
- Laboratory of Neurogenetics and Neuroscience, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Anna Fassio
- Department of Experimental Medicine, University of Genoa, Genoa, Italy. .,IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|