1
|
Kodba S, Öztop A, van Berkum E, Katrukha EA, Iwanski MK, Nijenhuis W, Kapitein LC, Chaigne A. Aurora B controls microtubule stability to regulate abscission dynamics in stem cells. Cell Rep 2025; 44:115238. [PMID: 39854207 DOI: 10.1016/j.celrep.2025.115238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/11/2024] [Accepted: 01/06/2025] [Indexed: 01/26/2025] Open
Abstract
Abscission is the last step of cell division. It separates the two sister cells and consists of cutting the cytoplasmic bridge. Abscission is mediated by the ESCRT membrane remodeling machinery, which also triggers the severing of a thick bundle of microtubules. Here, we show that rather than being passive actors in abscission, microtubules control abscission speed. Using mouse embryonic stem cells, which transition from slow to fast abscission during exit from naive pluripotency, we investigate the molecular mechanism for the regulation of abscission dynamics and identify crosstalk between Aurora B activity and microtubule stability. We demonstrate that naive stem cells maintain high Aurora B activity on the bridge after cytokinesis. This high Aurora B activity leads to transient microtubule stabilization that delays abscission by decreasing MCAK recruitment to the midbody. In turn, stable microtubules promote the activity of Aurora B. Overall, our data demonstrate that Aurora B-dependent microtubule stability controls abscission dynamics.
Collapse
Affiliation(s)
- Snježana Kodba
- Cell Biology, Neurobiology, and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, 3584 CS Utrecht, the Netherlands
| | - Amber Öztop
- Cell Biology, Neurobiology, and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, 3584 CS Utrecht, the Netherlands
| | - Eri van Berkum
- Cell Biology, Neurobiology, and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, 3584 CS Utrecht, the Netherlands
| | - Eugene A Katrukha
- Cell Biology, Neurobiology, and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, 3584 CS Utrecht, the Netherlands; Centre for Living Technologies, Alliance TU/e, WUR, UU, UMC Utrecht, Princetonlaan 6, 3584 CB Utrecht, the Netherlands
| | - Malina K Iwanski
- Cell Biology, Neurobiology, and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, 3584 CS Utrecht, the Netherlands
| | - Wilco Nijenhuis
- Cell Biology, Neurobiology, and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, 3584 CS Utrecht, the Netherlands; Centre for Living Technologies, Alliance TU/e, WUR, UU, UMC Utrecht, Princetonlaan 6, 3584 CB Utrecht, the Netherlands
| | - Lukas C Kapitein
- Cell Biology, Neurobiology, and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, 3584 CS Utrecht, the Netherlands; Centre for Living Technologies, Alliance TU/e, WUR, UU, UMC Utrecht, Princetonlaan 6, 3584 CB Utrecht, the Netherlands
| | - Agathe Chaigne
- Cell Biology, Neurobiology, and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, 3584 CS Utrecht, the Netherlands.
| |
Collapse
|
2
|
Serrano T, Casartelli N, Ghasemi F, Wioland H, Cuvelier F, Salles A, Moya-Nilges M, Welker L, Bernacchi S, Ruff M, Jégou A, Romet-Lemonne G, Schwartz O, Frémont S, Echard A. HIV-1 budding requires cortical actin disassembly by the oxidoreductase MICAL1. Proc Natl Acad Sci U S A 2024; 121:e2407835121. [PMID: 39556735 PMCID: PMC11621841 DOI: 10.1073/pnas.2407835121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/30/2024] [Indexed: 11/20/2024] Open
Abstract
Many enveloped viruses bud from the plasma membrane that is tightly associated with a dense and thick actin cortex. This actin network represents a significant challenge for membrane deformation and scission, and how it is remodeled during the late steps of the viral cycle is largely unknown. Using superresolution microscopy, we show that HIV-1 buds in areas of the plasma membrane with low cortical F-actin levels. We find that the cellular oxidoreductase MICAL1 locally depolymerizes actin at budding sites to promote HIV-1 budding and release. Upon MICAL1 depletion, F-actin abnormally remains at viral budding sites, incompletely budded viruses accumulate at the plasma membrane and viral release is impaired. Remarkably, normal viral release can be restored in MICAL1-depleted cells by inhibiting Arp2/3-dependent branched actin networks. Mechanistically, we find that MICAL1 directly disassembles branched-actin networks and controls the timely recruitment of the Endosomal Sorting Complexes Required for Transport scission machinery during viral budding. In addition, the MICAL1 activator Rab35 is recruited at budding sites, functions in the same pathway as MICAL1, and is also required for viral release. This work reveals a role for oxidoreduction in triggering local actin depolymerization to control HIV-1 budding, a mechanism that may be widely used by other viruses. The debranching activity of MICAL1 could be involved beyond viral budding in various other cellular functions requiring local plasma membrane deformation.
Collapse
Affiliation(s)
- Thomas Serrano
- Membrane Traffic and Cell Division Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3691, ParisF-75015, France
| | - Nicoletta Casartelli
- Virology department, Virus and Immunity Lab, Institut Pasteur, Université Paris Cité, ParisF-75015, France
| | - Foad Ghasemi
- Université Paris Cité, CNRS, Institut Jacques Monod, ParisF-75013, France
| | - Hugo Wioland
- Université Paris Cité, CNRS, Institut Jacques Monod, ParisF-75013, France
| | - Frédérique Cuvelier
- Membrane Traffic and Cell Division Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3691, ParisF-75015, France
| | - Audrey Salles
- Institut Pasteur, Université Paris Cité, Photonic Bio-Imaging Unit, Centre de Ressources et Recherches Technologiques (UTechS-PBI, C2RT), ParisF-75015, France
| | - Maryse Moya-Nilges
- Institut Pasteur, Université Paris Cité, Ultrastructural BioImaging, ParisF-75015, France
| | - Lisa Welker
- Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire, Architecture et Réactivité de l’ARN, CNRS UPR9002, StrasbourgF-67084, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Integrated Structural Biology, CNRS UMR 7104, Inserm U 1258, University of Strasbourg, IllkirchF-67404, France
| | - Serena Bernacchi
- Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire, Architecture et Réactivité de l’ARN, CNRS UPR9002, StrasbourgF-67084, France
| | - Marc Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Integrated Structural Biology, CNRS UMR 7104, Inserm U 1258, University of Strasbourg, IllkirchF-67404, France
| | - Antoine Jégou
- Université Paris Cité, CNRS, Institut Jacques Monod, ParisF-75013, France
| | | | - Olivier Schwartz
- Virology department, Virus and Immunity Lab, Institut Pasteur, Université Paris Cité, ParisF-75015, France
| | - Stéphane Frémont
- Membrane Traffic and Cell Division Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3691, ParisF-75015, France
| | - Arnaud Echard
- Membrane Traffic and Cell Division Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3691, ParisF-75015, France
| |
Collapse
|
3
|
Horvath M, Schrofel A, Kowalska K, Sabo J, Vlasak J, Nourisanami F, Sobol M, Pinkas D, Knapp K, Koupilova N, Novacek J, Veverka V, Lansky Z, Rozbesky D. Structural basis of MICAL autoinhibition. Nat Commun 2024; 15:9810. [PMID: 39532862 PMCID: PMC11557892 DOI: 10.1038/s41467-024-54131-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
MICAL proteins play a crucial role in cellular dynamics by binding and disassembling actin filaments, impacting processes like axon guidance, cytokinesis, and cell morphology. Their cellular activity is tightly controlled, as dysregulation can lead to detrimental effects on cellular morphology. Although previous studies have suggested that MICALs are autoinhibited, and require Rab proteins to become active, the detailed molecular mechanisms remained unclear. Here, we report the cryo-EM structure of human MICAL1 at a nominal resolution of 3.1 Å. Structural analyses, alongside biochemical and functional studies, show that MICAL1 autoinhibition is mediated by an intramolecular interaction between its N-terminal catalytic and C-terminal coiled-coil domains, blocking F-actin interaction. Moreover, we demonstrate that allosteric changes in the coiled-coil domain and the binding of the tripartite assembly of CH-L2α1-LIM domains to the coiled-coil domain are crucial for MICAL activation and autoinhibition. These mechanisms appear to be evolutionarily conserved, suggesting a potential universality across the MICAL family.
Collapse
Affiliation(s)
- Matej Horvath
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Adam Schrofel
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Karolina Kowalska
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jan Sabo
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| | - Jonas Vlasak
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Farahdokht Nourisanami
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Margarita Sobol
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Daniel Pinkas
- Central European Institute of Technology, Masaryk University, Brno, Czechia
| | - Krystof Knapp
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Nicola Koupilova
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jiri Novacek
- Central European Institute of Technology, Masaryk University, Brno, Czechia
| | - Vaclav Veverka
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czechia
| | - Zdenek Lansky
- Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
| | - Daniel Rozbesky
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia.
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia.
| |
Collapse
|
4
|
Tan R, Hoare M, Bellomio P, Broas S, Camacho K, Swovick K, Welle KA, Hryhorenko JR, Ghaemmaghami S. Formylation facilitates the reduction of oxidized initiator methionines. Proc Natl Acad Sci U S A 2024; 121:e2403880121. [PMID: 39499632 PMCID: PMC11572973 DOI: 10.1073/pnas.2403880121] [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: 02/23/2024] [Accepted: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
Within a cell, protein-bound methionines can be chemically or enzymatically oxidized, and subsequently reduced by methionine sulfoxide reductases (Msrs). Methionine oxidation can result in structural damage or be the basis of functional regulation of enzymes. In addition to participating in redox reactions, methionines play an important role as the initiator residue of translated proteins where they are commonly modified at their α-amine group by formylation or acetylation. Here, we investigated how formylation and acetylation of initiator methionines impact their propensity for oxidation and reduction. We show that in vitro, N-terminal methionine residues are particularly prone to chemical oxidation and that their modification by formylation or acetylation greatly enhances their subsequent enzymatic reduction by MsrA and MsrB. Concordantly, in vivo ablation of methionyl-tRNA formyltransferase (MTF) in Escherichia coli increases the prevalence of oxidized methionines within synthesized proteins. We show that oxidation of formylated initiator methionines is detrimental in part because it obstructs their ensuing deformylation by peptide deformylase (PDF) and hydrolysis by methionyl aminopeptidase (MAP). Thus, by facilitating their reduction, formylation mitigates the misprocessing of oxidized initiator methionines.
Collapse
Affiliation(s)
- Ruiyue Tan
- Department of Biology, University of Rochester, Rochester, NY14627
| | - Margaret Hoare
- Department of Biology, University of Rochester, Rochester, NY14627
| | - Philip Bellomio
- Department of Biology, University of Rochester, Rochester, NY14627
| | - Sarah Broas
- Department of Biology, University of Rochester, Rochester, NY14627
| | | | - Kyle Swovick
- Mass Spectrometry Resource Laboratory, University of Rochester Medical Center, Rochester, NY14627
| | - Kevin A. Welle
- Mass Spectrometry Resource Laboratory, University of Rochester Medical Center, Rochester, NY14627
| | - Jennifer R. Hryhorenko
- Mass Spectrometry Resource Laboratory, University of Rochester Medical Center, Rochester, NY14627
| | - Sina Ghaemmaghami
- Department of Biology, University of Rochester, Rochester, NY14627
- Mass Spectrometry Resource Laboratory, University of Rochester Medical Center, Rochester, NY14627
| |
Collapse
|
5
|
Huang CD, Shi Y, Wang F, Wu PF, Chen JG. Methionine oxidation of actin cytoskeleton attenuates traumatic memory retention via reactivating dendritic spine morphogenesis. Redox Biol 2024; 77:103391. [PMID: 39405981 PMCID: PMC11525628 DOI: 10.1016/j.redox.2024.103391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 11/03/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is characterized by hypermnesia of the trauma and a persistent fear response. The molecular mechanisms underlying the retention of traumatic memories remain largely unknown, which hinders the development of more effective treatments. Utilizing auditory fear conditioning, we demonstrate that a redox-dependent dynamic pathway for dendritic spine morphogenesis in the basolateral amygdala (BLA) is crucial for traumatic memory retention. Exposure to a fear-induced event markedly increased the reduction of oxidized filamentous actin (F-actin) and decreased the expression of the molecule interacting with CasL 1 (MICAL1), a methionine-oxidizing enzyme that directly oxidizes and depolymerizes F-actin, leading to cytoskeletal dynamic abnormalities in the BLA, which impairs dendritic spine morphogenesis and contributes to the persistence of fearful memories. Following fear conditioning, overexpression of MICAL1 in the BLA inhibited freezing behavior during fear memory retrieval via reactivating cytokinesis, whereas overexpression of methionine sulfoxide reductase B 1, a key enzyme that reduces oxidized F-actin monomer, increased freezing behavior during retrieval. Notably, intra-BLA injection of semaphorin 3A, an endogenous activator of MICAL1, rapidly disrupted fear memory within a short time window after conditioning. Collectively, our results indicate that redox modulation of actin cytoskeleton in the BLA is functionally linked to fear memory retention and PTSD-like memory.
Collapse
Affiliation(s)
- Cun-Dong Huang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yu Shi
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Hubei Shizhen Laboratory, Wuhan, Hubei, 430030, China.
| | - Peng-Fei Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Hubei Shizhen Laboratory, Wuhan, Hubei, 430030, China.
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Hubei Shizhen Laboratory, Wuhan, Hubei, 430030, China.
| |
Collapse
|
6
|
Öztop A, Chaigne A. Molecular and mechanical mechanisms of animal cell abscission. FEBS Lett 2024. [PMID: 39324548 DOI: 10.1002/1873-3468.15015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 09/27/2024]
Affiliation(s)
- Amber Öztop
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, Utrecht, 3584 CS, the Netherlands
| | - Agathe Chaigne
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, Utrecht, 3584 CS, the Netherlands
| |
Collapse
|
7
|
Dong J, Zhu XN, Zeng PM, Cao DD, Yang Y, Hu J, Luo ZG. A hominoid-specific signaling axis regulating the tempo of synaptic maturation. Cell Rep 2024; 43:114548. [PMID: 39052482 DOI: 10.1016/j.celrep.2024.114548] [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: 11/13/2023] [Revised: 04/15/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
Human cortical neurons (hCNs) exhibit high dendritic complexity and synaptic density, and the maturation process is greatly protracted. However, the molecular mechanism governing these specific features remains unclear. Here, we report that the hominoid-specific gene TBC1D3 promotes dendritic arborization and protracts the pace of synaptogenesis. Ablation of TBC1D3 in induced hCNs causes reduction of dendritic growth and precocious synaptic maturation. Forced expression of TBC1D3 in the mouse cortex protracts synaptic maturation while increasing dendritic growth. Mechanistically, TBC1D3 functions via interaction with MICAL1, a monooxygenase that mediates oxidation of actin filament. At the early stage of differentiation, the TBC1D3/MICAL1 interaction in the cytosol promotes dendritic growth via F-actin oxidation and enhanced actin dynamics. At late stages, TBC1D3 escorts MICAL1 into the nucleus and downregulates the expression of genes related with synaptic maturation through interaction with the chromatin remodeling factor ATRX. Thus, this study delineates the molecular mechanisms underlying human neuron development.
Collapse
Affiliation(s)
- Jian Dong
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Xiao-Na Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Peng-Ming Zeng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Dong-Dong Cao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Yang Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhen-Ge Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
8
|
Lin L, Dong J, Xu S, Xiao J, Yu C, Niu F, Wei Z. Autoinhibition and relief mechanisms for MICAL monooxygenases in F-actin disassembly. Nat Commun 2024; 15:6824. [PMID: 39122694 PMCID: PMC11315924 DOI: 10.1038/s41467-024-50940-7] [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: 02/16/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
MICAL proteins represent a unique family of actin regulators crucial for synapse development, membrane trafficking, and cytokinesis. Unlike classical actin regulators, MICALs catalyze the oxidation of specific residues within actin filaments to induce robust filament disassembly. The potent activity of MICALs requires tight control to prevent extensive damage to actin cytoskeleton. However, the molecular mechanism governing MICALs' activity regulation remains elusive. Here, we report the cryo-EM structure of MICAL1 in the autoinhibited state, unveiling a head-to-tail interaction that allosterically blocks enzymatic activity. The structure also reveals the assembly of C-terminal domains via a tripartite interdomain interaction, stabilizing the inhibitory conformation of the RBD. Our structural, biochemical, and cellular analyses elucidate a multi-step mechanism to relieve MICAL1 autoinhibition in response to the dual-binding of two Rab effectors, revealing its intricate activity regulation mechanisms. Furthermore, our mutagenesis study of MICAL3 suggests the conserved autoinhibition and relief mechanisms among MICALs.
Collapse
Affiliation(s)
- Leishu Lin
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jiayuan Dong
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shun Xu
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jinman Xiao
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China
| | - Cong Yu
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Fengfeng Niu
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China.
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Zhiyi Wei
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China.
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
9
|
Prever L, Squillero G, Hirsch E, Gulluni F. Linking phosphoinositide function to mitosis. Cell Rep 2024; 43:114273. [PMID: 38843397 DOI: 10.1016/j.celrep.2024.114273] [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: 03/18/2024] [Revised: 04/12/2024] [Accepted: 05/09/2024] [Indexed: 07/02/2024] Open
Abstract
Phosphoinositides (PtdIns) are a family of differentially phosphorylated lipid second messengers localized to the cytoplasmic leaflet of both plasma and intracellular membranes. Kinases and phosphatases can selectively modify the PtdIns composition of different cellular compartments, leading to the recruitment of specific binding proteins, which control cellular homeostasis and proliferation. Thus, while PtdIns affect cell growth and survival during interphase, they are also emerging as key drivers in multiple temporally defined membrane remodeling events of mitosis, like cell rounding, spindle orientation, cytokinesis, and abscission. In this review, we summarize and discuss what is known about PtdIns function during mitosis and how alterations in the production and removal of PtdIns can interfere with proper cell division.
Collapse
Affiliation(s)
- Lorenzo Prever
- University of Turin, Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", Via Nizza 52, 10126 Turin, Italy
| | - Gabriele Squillero
- University of Turin, Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", Via Nizza 52, 10126 Turin, Italy
| | - Emilio Hirsch
- University of Turin, Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", Via Nizza 52, 10126 Turin, Italy.
| | - Federico Gulluni
- University of Turin, Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", Via Nizza 52, 10126 Turin, Italy.
| |
Collapse
|
10
|
Richard A, Berthelet J, Judith D, Advedissian T, Espadas J, Jannot G, Amo A, Loew D, Lombard B, Casanova AG, Reynoird N, Roux A, Berlioz-Torrent C, Echard A, Weitzman JB, Medjkane S. Methylation of ESCRT-III components regulates the timing of cytokinetic abscission. Nat Commun 2024; 15:4023. [PMID: 38740816 PMCID: PMC11091153 DOI: 10.1038/s41467-024-47717-3] [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: 09/20/2023] [Accepted: 04/10/2024] [Indexed: 05/16/2024] Open
Abstract
Abscission is the final stage of cytokinesis, which cleaves the intercellular bridge (ICB) connecting two daughter cells. Abscission requires tight control of the recruitment and polymerization of the Endosomal Protein Complex Required for Transport-III (ESCRT-III) components. We explore the role of post-translational modifications in regulating ESCRT dynamics. We discover that SMYD2 methylates the lysine 6 residue of human CHMP2B, a key ESCRT-III component, at the ICB, impacting the dynamic relocation of CHMP2B to sites of abscission. SMYD2 loss-of-function (genetically or pharmacologically) causes CHMP2B hypomethylation, delayed CHMP2B polymerization and delayed abscission. This is phenocopied by CHMP2B lysine 6 mutants that cannot be methylated. Conversely, SMYD2 gain-of-function causes CHMP2B hypermethylation and accelerated abscission, specifically in cells undergoing cytokinetic challenges, thereby bypassing the abscission checkpoint. Additional experiments highlight the importance of CHMP2B methylation beyond cytokinesis, namely during ESCRT-III-mediated HIV-1 budding. We propose that lysine methylation signaling fine-tunes the ESCRT-III machinery to regulate the timing of cytokinetic abscission and other ESCRT-III dependent functions.
Collapse
Affiliation(s)
- Aurélie Richard
- Université Paris Cité, CNRS, UMR7126 Epigenetics and Cell Fate, F-75013, Paris, France
| | - Jérémy Berthelet
- Université Paris Cité, CNRS, UMR7126 Epigenetics and Cell Fate, F-75013, Paris, France
| | - Delphine Judith
- Université Paris Cité, Inserm, CNRS, Institut Cochin, F-75014, Paris, France
| | - Tamara Advedissian
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 Rue du Dr Roux, F-75015, Paris, France
| | - Javier Espadas
- Department of Biochemistry, University of Geneva, CH-1211, Geneva, Switzerland
| | - Guillaume Jannot
- Université Paris Cité, CNRS, UMR7126 Epigenetics and Cell Fate, F-75013, Paris, France
| | - Angélique Amo
- Université Paris Cité, CNRS, UMR7126 Epigenetics and Cell Fate, F-75013, Paris, France
| | - Damarys Loew
- Institut Curie, PSL Research University, Centre de Recherche, CurieCoreTech Mass Spectrometry Proteomics, F-75005, Paris, France
| | - Berangere Lombard
- Institut Curie, PSL Research University, Centre de Recherche, CurieCoreTech Mass Spectrometry Proteomics, F-75005, Paris, France
| | - Alexandre G Casanova
- Université Grenoble Alpes, CNRS UMR5309, INSERM U1209, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Nicolas Reynoird
- Université Grenoble Alpes, CNRS UMR5309, INSERM U1209, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Aurélien Roux
- Department of Biochemistry, University of Geneva, CH-1211, Geneva, Switzerland
| | | | - Arnaud Echard
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 Rue du Dr Roux, F-75015, Paris, France
| | - Jonathan B Weitzman
- Université Paris Cité, CNRS, UMR7126 Epigenetics and Cell Fate, F-75013, Paris, France
| | - Souhila Medjkane
- Université Paris Cité, CNRS, UMR7126 Epigenetics and Cell Fate, F-75013, Paris, France.
| |
Collapse
|
11
|
Aguila A, Salah S, Kulasekaran G, Shweiki M, Shaul-Lotan N, Mor-Shaked H, Daana M, Harel T, McPherson PS. A neurodevelopmental disorder associated with a loss-of-function missense mutation in RAB35. J Biol Chem 2024; 300:107124. [PMID: 38432637 PMCID: PMC10966776 DOI: 10.1016/j.jbc.2024.107124] [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: 08/01/2023] [Revised: 02/01/2024] [Accepted: 02/18/2024] [Indexed: 03/05/2024] Open
Abstract
Rab35 (Ras-associated binding protein) is a small GTPase that regulates endosomal membrane trafficking and functions in cell polarity, cytokinesis, and growth factor signaling. Altered Rab35 function contributes to progression of glioblastoma, defects in primary cilia formation, and altered cytokinesis. Here, we report a pediatric patient with global developmental delay, hydrocephalus, a Dandy-Walker malformation, axial hypotonia with peripheral hypertonia, visual problems, and conductive hearing impairment. Exome sequencing identified a homozygous missense variant in the GTPase fold of RAB35 (c.80G>A; p.R27H) as the most likely candidate. Functional analysis of the R27H-Rab35 variant protein revealed enhanced interaction with its guanine-nucleotide exchange factor, DENND1A and decreased interaction with a known effector, MICAL1, indicating that the protein is in an inactive conformation. Cellular expression of the variant drives the activation of Arf6, a small GTPase under negative regulatory control of Rab35. Importantly, variant expression leads to delayed cytokinesis and altered length, number, and Arl13b composition of primary cilia, known factors in neurodevelopmental disease. Our findings provide evidence of altered Rab35 function as a causative factor of a neurodevelopmental disorder.
Collapse
Affiliation(s)
- Adriana Aguila
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Somaya Salah
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel
| | - Gopinath Kulasekaran
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Moatasem Shweiki
- Neurosurgery Department, Hadassah Medical Center, Jerusalem, Israel
| | - Nava Shaul-Lotan
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel
| | - Hagar Mor-Shaked
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Muhannad Daana
- Child Development Centers, Clalit Health Care Services, Yokne'am Illit, Israel
| | - Tamar Harel
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Peter S McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
12
|
van Soest DMK, Polderman PE, den Toom WTF, Keijer JP, van Roosmalen MJ, Leyten TMF, Lehmann J, Zwakenberg S, De Henau S, van Boxtel R, Burgering BMT, Dansen TB. Mitochondrial H 2O 2 release does not directly cause damage to chromosomal DNA. Nat Commun 2024; 15:2725. [PMID: 38548751 PMCID: PMC10978998 DOI: 10.1038/s41467-024-47008-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 03/18/2024] [Indexed: 04/01/2024] Open
Abstract
Reactive Oxygen Species (ROS) derived from mitochondrial respiration are frequently cited as a major source of chromosomal DNA mutations that contribute to cancer development and aging. However, experimental evidence showing that ROS released by mitochondria can directly damage nuclear DNA is largely lacking. In this study, we investigated the effects of H2O2 released by mitochondria or produced at the nucleosomes using a titratable chemogenetic approach. This enabled us to precisely investigate to what extent DNA damage occurs downstream of near- and supraphysiological amounts of localized H2O2. Nuclear H2O2 gives rise to DNA damage and mutations and a subsequent p53 dependent cell cycle arrest. Mitochondrial H2O2 release shows none of these effects, even at levels that are orders of magnitude higher than what mitochondria normally produce. We conclude that H2O2 released from mitochondria is unlikely to directly damage nuclear genomic DNA, limiting its contribution to oncogenic transformation and aging.
Collapse
Affiliation(s)
- Daan M K van Soest
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, Utrecht, 3584 CG, The Netherlands
| | - Paulien E Polderman
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, Utrecht, 3584 CG, The Netherlands
| | - Wytze T F den Toom
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, Utrecht, 3584 CG, The Netherlands
| | - Janneke P Keijer
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, Utrecht, 3584 CG, The Netherlands
| | - Markus J van Roosmalen
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht, 3584 CS, The Netherlands
| | - Tim M F Leyten
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, Utrecht, 3584 CG, The Netherlands
| | - Johannes Lehmann
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, Utrecht, 3584 CG, The Netherlands
| | - Susan Zwakenberg
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, Utrecht, 3584 CG, The Netherlands
| | - Sasha De Henau
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, Utrecht, 3584 CG, The Netherlands
| | - Ruben van Boxtel
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht, 3584 CS, The Netherlands
- Oncode Institute, Jaarbeursplein 6, Utrecht, 3521 AL, The Netherlands
| | - Boudewijn M T Burgering
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, Utrecht, 3584 CG, The Netherlands
- Oncode Institute, Jaarbeursplein 6, Utrecht, 3521 AL, The Netherlands
| | - Tobias B Dansen
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, Utrecht, 3584 CG, The Netherlands.
| |
Collapse
|
13
|
Goupil E, Lacroix L, Brière J, Guga S, Saba-El-Leil MK, Meloche S, Labbé JC. OSGN-1 is a conserved flavin-containing monooxygenase required to stabilize the intercellular bridge in late cytokinesis. Proc Natl Acad Sci U S A 2024; 121:e2308570121. [PMID: 38442170 PMCID: PMC10945809 DOI: 10.1073/pnas.2308570121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 01/24/2024] [Indexed: 03/07/2024] Open
Abstract
Cytokinesis is the last step of cell division and is regulated by the small GTPase RhoA. RhoA activity is required for all steps of cytokinesis, including prior to abscission when daughter cells are ultimately physically separated. Like germ cells in all animals, the Caenorhabditis elegans embryonic germline founder cell initiates cytokinesis but does not complete abscission, leaving a stable intercellular bridge between the two daughter cells. Here, we identify and characterize C. elegans OSGN-1 as a cytokinetic regulator that promotes RhoA activity during late cytokinesis. Sequence analyses and biochemical reconstitutions reveal that OSGN-1 is a flavin-containing monooxygenase (MO). Genetic analyses indicate that the MO activity of OSGN-1 is required to maintain active RhoA at the end of cytokinesis in the germline founder cell and to stabilize the intercellular bridge. Deletion of OSGIN1 in human cells results in an increase in binucleation as a result of cytokinetic furrow regression, and this phenotype can be rescued by expressing a catalytically active form of C. elegans OSGN-1, indicating that OSGN-1 and OSGIN1 are functional orthologs. We propose that OSGN-1 and OSGIN1 are conserved MO enzymes required to maintain RhoA activity at the intercellular bridge during late cytokinesis and thus favor its stability, enabling proper abscission in human cells and bridge stabilization in C. elegans germ cells.
Collapse
Affiliation(s)
- Eugénie Goupil
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QCH3C 3J7, Canada
| | - Léa Lacroix
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QCH3C 3J7, Canada
| | - Jonathan Brière
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QCH3C 3J7, Canada
| | - Sandra Guga
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QCH3C 3J7, Canada
| | - Marc K. Saba-El-Leil
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QCH3C 3J7, Canada
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QCH3C 3J7, Canada
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, QCH3C 3J7, Canada
| | - Jean-Claude Labbé
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QCH3C 3J7, Canada
- Department of Pathology and Cell Biology, Université de Montréal, Montréal, QCH3C 3J7, Canada
| |
Collapse
|
14
|
Advedissian T, Frémont S, Echard A. Cytokinetic abscission requires actin-dependent microtubule severing. Nat Commun 2024; 15:1949. [PMID: 38431632 PMCID: PMC10908825 DOI: 10.1038/s41467-024-46062-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 02/09/2024] [Indexed: 03/05/2024] Open
Abstract
Cell division is completed by the abscission of the intercellular bridge connecting the daughter cells. Abscission requires the polymerization of an ESCRT-III cone close to the midbody to both recruit the microtubule severing enzyme spastin and scission the plasma membrane. Here, we found that the microtubule and the membrane cuts are two separate events that are regulated differently. Using HeLa cells, we uncovered that the F-actin disassembling protein Cofilin-1 controls the disappearance of a transient pool of branched F-actin which is precisely assembled at the tip of the ESCRT-III cone shortly before the microtubule cut. Functionally, Cofilin-1 and Arp2/3-mediated branched F-actin favor abscission by promoting local severing of the microtubules but do not participate later in the membrane scission event. Mechanistically, we propose that branched F-actin functions as a physical barrier that limits ESCRT-III cone elongation and thereby favors stable spastin recruitment. Our work thus reveals that F-actin controls the timely and local disassembly of microtubules required for cytokinetic abscission.
Collapse
Affiliation(s)
- Tamara Advedissian
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015, Paris, France
| | - Stéphane Frémont
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015, Paris, France
| | - Arnaud Echard
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015, Paris, France.
| |
Collapse
|
15
|
Garrido-Bazán V, Guzmán-Ocampo DC, Domínguez L, Aguirre J. Filamentous actin destabilization by H 2O 2 favors DnmA aggregation, with crucial roles of cysteines 450 and 776 in mitochondrial and peroxisomal division in Aspergillus nidulans. mBio 2023; 14:e0282223. [PMID: 38014993 PMCID: PMC10746283 DOI: 10.1128/mbio.02822-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023] Open
Abstract
IMPORTANCE Mitochondria constitute major sources of H2O2 and other reactive oxygen species in eukaryotic cells. The division of these organelles is crucial for multiple processes in cell biology and relies on highly regulated mechano-GTPases that are oligomerization dependent and belong to the dynamin-related protein family, like A. nidulans DnmA. Our previous work demonstrated that H2O2 induces mitochondrial constriction, division, and remodeling of the outer membrane. Here, we show that H2O2 also induces a DnmA aggregation consistent with higher-order oligomerization and its recruitment to mitochondria. The study of this response uncovered that H2O2 induces the depolymerization and reorganization of actin as well as the critical role that cysteines 450 and 776 play in DnmA function. Our results provide new insights into the mechanisms of reactive oxygen species cell signaling and how they can regulate the dynamics of the actin cytoskeleton and the division of mitochondria and peroxisomes.
Collapse
Affiliation(s)
- Verónica Garrido-Bazán
- Instituto de Fisiología Celular, Departamento de Biología Celular y Desarrollo, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Dulce C. Guzmán-Ocampo
- Facultad de Química, Departamento de Fisicoquímica, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Laura Domínguez
- Facultad de Química, Departamento de Fisicoquímica, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Jesús Aguirre
- Instituto de Fisiología Celular, Departamento de Biología Celular y Desarrollo, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
16
|
Goode BL, Eskin J, Shekhar S. Mechanisms of actin disassembly and turnover. J Cell Biol 2023; 222:e202309021. [PMID: 37948068 PMCID: PMC10638096 DOI: 10.1083/jcb.202309021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Cellular actin networks exhibit a wide range of sizes, shapes, and architectures tailored to their biological roles. Once assembled, these filamentous networks are either maintained in a state of polarized turnover or induced to undergo net disassembly. Further, the rates at which the networks are turned over and/or dismantled can vary greatly, from seconds to minutes to hours or even days. Here, we review the molecular machinery and mechanisms employed in cells to drive the disassembly and turnover of actin networks. In particular, we highlight recent discoveries showing that specific combinations of conserved actin disassembly-promoting proteins (cofilin, GMF, twinfilin, Srv2/CAP, coronin, AIP1, capping protein, and profilin) work in concert to debranch, sever, cap, and depolymerize actin filaments, and to recharge actin monomers for new rounds of assembly.
Collapse
Affiliation(s)
- Bruce L. Goode
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA
| | - Julian Eskin
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA
| | - Shashank Shekhar
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, GA, USA
| |
Collapse
|
17
|
Carlton JG, Baum B. Roles of ESCRT-III polymers in cell division across the tree of life. Curr Opin Cell Biol 2023; 85:102274. [PMID: 37944425 PMCID: PMC7615534 DOI: 10.1016/j.ceb.2023.102274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/12/2023] [Accepted: 10/12/2023] [Indexed: 11/12/2023]
Abstract
Every cell becomes two through a carefully orchestrated process of division. Prior to division, contractile machinery must first be assembled at the cell midzone to ensure that the cut, when it is made, bisects the two separated copies of the genetic material. Second, this contractile machinery must be dynamically tethered to the limiting plasma membrane so as to bring the membrane with it as it constricts. Finally, the connecting membrane must be severed to generate two physically separate daughter cells. In several organisms across the tree of life, Endosomal Sorting Complex Required for Transport (ESCRT)-III family proteins aid cell division by forming composite polymers that function together with the Vps4 AAA-ATPase to constrict and cut the membrane tube connecting nascent daughter cells from the inside. In this review, we discuss unique features of ESCRT-III that enable it to play this role in division in many archaea and eukaryotes.
Collapse
Affiliation(s)
- Jeremy Graham Carlton
- Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Hospital, London, SE1 1UL, UK; Organelle Dynamics Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| | - Buzz Baum
- Medical Research Council Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK.
| |
Collapse
|
18
|
Serres MP, Shaughnessy R, Escot S, Hammich H, Cuvelier F, Salles A, Rocancourt M, Verdon Q, Gaffuri AL, Sourigues Y, Malherbe G, Velikovsky L, Chardon F, Sassoon N, Tinevez JY, Callebaut I, Formstecher E, Houdusse A, David NB, Pylypenko O, Echard A. MiniBAR/GARRE1 is a dual Rac and Rab effector required for ciliogenesis. Dev Cell 2023; 58:2477-2494.e8. [PMID: 37875118 DOI: 10.1016/j.devcel.2023.09.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/07/2023] [Accepted: 09/29/2023] [Indexed: 10/26/2023]
Abstract
Cilia protrude from the cell surface and play critical roles in intracellular signaling, environmental sensing, and development. Reduced actin-dependent contractility and intracellular trafficking are both required for ciliogenesis, but little is known about how these processes are coordinated. Here, we identified a Rac1- and Rab35-binding protein with a truncated BAR (Bin/amphiphysin/Rvs) domain that we named MiniBAR (also known as KIAA0355/GARRE1), which plays a key role in ciliogenesis. MiniBAR colocalizes with Rac1 and Rab35 at the plasma membrane and on intracellular vesicles trafficking to the ciliary base and exhibits fast pulses at the ciliary membrane. MiniBAR depletion leads to short cilia, resulting from abnormal Rac-GTP/Rho-GTP levels and increased acto-myosin-II-dependent contractility together with defective trafficking of IFT88 and ARL13B into cilia. MiniBAR-depleted zebrafish embryos display dysfunctional short cilia and hallmarks of ciliopathies, including left-right asymmetry defects. Thus, MiniBAR is a dual Rac and Rab effector that controls both actin cytoskeleton and membrane trafficking for ciliogenesis.
Collapse
Affiliation(s)
- Murielle P Serres
- Institut Pasteur, Université de Paris, CNRS UMR3691, Membrane Traffic and Cell Division Laboratory, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Ronan Shaughnessy
- Institut Pasteur, Université de Paris, CNRS UMR3691, Membrane Traffic and Cell Division Laboratory, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Sophie Escot
- Laboratoire d'Optique et Biosciences (LOB), CNRS, INSERM, Ecole Polytechnique, Institut Polytechnique de Paris, 91120 Palaiseau, France
| | - Hussein Hammich
- Institut Curie, PSL Research University, CNRS UMR144, Structural Motility, 26 rue d'Ulm, 75005 Paris, France
| | - Frédérique Cuvelier
- Institut Pasteur, Université de Paris, CNRS UMR3691, Membrane Traffic and Cell Division Laboratory, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Audrey Salles
- Institut Pasteur, Université de Paris, UTechS Photonic BioImaging (UTechS PBI), Centre de Recherche et de Ressources Technologiques C2RT, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Murielle Rocancourt
- Institut Pasteur, Université de Paris, CNRS UMR3691, Membrane Traffic and Cell Division Laboratory, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Quentin Verdon
- Institut Pasteur, Université de Paris, CNRS UMR3691, Membrane Traffic and Cell Division Laboratory, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Anne-Lise Gaffuri
- Institut Pasteur, Université de Paris, CNRS UMR3691, Membrane Traffic and Cell Division Laboratory, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Yannick Sourigues
- Institut Curie, PSL Research University, CNRS UMR144, Structural Motility, 26 rue d'Ulm, 75005 Paris, France
| | - Gilles Malherbe
- Institut Curie, PSL Research University, CNRS UMR144, Structural Motility, 26 rue d'Ulm, 75005 Paris, France
| | - Leonid Velikovsky
- Institut Curie, PSL Research University, CNRS UMR144, Structural Motility, 26 rue d'Ulm, 75005 Paris, France
| | - Florian Chardon
- Institut Curie, PSL Research University, CNRS UMR144, Structural Motility, 26 rue d'Ulm, 75005 Paris, France
| | - Nathalie Sassoon
- Institut Pasteur, Université de Paris, CNRS UMR3691, Membrane Traffic and Cell Division Laboratory, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Jean-Yves Tinevez
- Institut Pasteur, Université de Paris, Image Analysis Hub, 25-28 rue du Dr Roux, 75015 Paris, France
| | - Isabelle Callebaut
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, Paris, France
| | - Etienne Formstecher
- Hybrigenics Services SAS, 1 rue Pierre Fontaine 91000 Evry, Courcouronnes, France
| | - Anne Houdusse
- Institut Curie, PSL Research University, CNRS UMR144, Structural Motility, 26 rue d'Ulm, 75005 Paris, France
| | - Nicolas B David
- Laboratoire d'Optique et Biosciences (LOB), CNRS, INSERM, Ecole Polytechnique, Institut Polytechnique de Paris, 91120 Palaiseau, France
| | - Olena Pylypenko
- Institut Curie, PSL Research University, CNRS UMR144, Structural Motility, 26 rue d'Ulm, 75005 Paris, France
| | - Arnaud Echard
- Institut Pasteur, Université de Paris, CNRS UMR3691, Membrane Traffic and Cell Division Laboratory, 25-28 rue du Dr Roux, 75015 Paris, France.
| |
Collapse
|
19
|
Miao H, Millage M, Rollins KR, Blankenship JT. A Rab39-Klp98A-Rab35 endocytic recycling pathway is essential for rapid Golgi-dependent furrow ingression. Development 2023; 150:dev201547. [PMID: 37590130 PMCID: PMC10445802 DOI: 10.1242/dev.201547] [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: 12/15/2022] [Accepted: 07/18/2023] [Indexed: 08/19/2023]
Abstract
Ingression of the plasma membrane is an essential part of the cell topology-distorting repertoire and a key element in animal cell cytokinesis. Many embryos have rapid cleavage stages in which they are furrowing powerhouses, quickly forming and disassembling cleavage furrows on timescales of just minutes. Previous work has shown that cytoskeletal proteins and membrane trafficking coordinate to drive furrow ingression, but where these membrane stores are derived from and how they are directed to furrowing processes has been less clear. Here, we identify an extensive Rab35/Rab4>Rab39/Klp98A>trans-Golgi network (TGN) endocytic recycling pathway necessary for fast furrow ingression in the Drosophila embryo. Rab39 is present in vesiculotubular compartments at the TGN where it receives endocytically derived cargo through a Rab35/Rab4-dependent pathway. A Kinesin-3 family member, Klp98A, drives the movements and tubulation activities of Rab39, and disruption of this Rab39-Klp98A-Rab35 pathway causes deep furrow ingression defects and genomic instability. These data suggest that an endocytic recycling pathway rapidly remobilizes membrane cargo from the cell surface and directs it to the trans-Golgi network to permit the initiation of new cycles of cleavage furrow formation.
Collapse
Affiliation(s)
- Hui Miao
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| | - Megan Millage
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| | | | - J. Todd Blankenship
- Department of Biological Sciences, University of Denver, Denver, CO 80208, USA
| |
Collapse
|
20
|
Pust S, Brech A, Wegner CS, Stenmark H, Haglund K. Vesicle-mediated transport of ALIX and ESCRT-III to the intercellular bridge during cytokinesis. Cell Mol Life Sci 2023; 80:235. [PMID: 37523003 PMCID: PMC10390626 DOI: 10.1007/s00018-023-04864-y] [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: 12/19/2022] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 08/01/2023]
Abstract
Cellular abscission is the final step of cytokinesis that leads to the physical separation of the two daughter cells. The scaffold protein ALIX and the ESCRT-I protein TSG101 contribute to recruiting ESCRT-III to the midbody, which orchestrates the final membrane scission of the intercellular bridge. Here, we addressed the transport mechanisms of ALIX and ESCRT-III subunit CHMP4B to the midbody. Structured illumination microscopy revealed gradual accumulation of ALIX at the midbody, resulting in the formation of spiral-like structures extending from the midbody to the abscission site, which strongly co-localized with CHMP4B. Live-cell microscopy uncovered that ALIX appeared together with CHMP4B in vesicular structures, whose motility was microtubule-dependent. Depletion of ALIX led to structural alterations of the midbody and delayed recruitment of CHMP4B, resulting in delayed abscission. Likewise, depletion of the kinesin-1 motor KIF5B reduced the motility of ALIX-positive vesicles and delayed midbody recruitment of ALIX, TSG101 and CHMP4B, accompanied by impeded abscission. We propose that ALIX, TSG101 and CHMP4B are associated with endosomal vesicles transported on microtubules by kinesin-1 to the cytokinetic bridge and midbody, thereby contributing to their function in abscission.
Collapse
Affiliation(s)
- Sascha Pust
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379, Oslo, Norway.
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379, Oslo, Norway.
| | - Andreas Brech
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379, Oslo, Norway
| | - Catherine Sem Wegner
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379, Oslo, Norway
| | - Harald Stenmark
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379, Oslo, Norway
| | - Kaisa Haglund
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379, Oslo, Norway.
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, 0379, Oslo, Norway.
| |
Collapse
|
21
|
Kumar R, Francis V, Ioannou MS, Aguila A, Khan M, Banks E, Kulasekaran G, McPherson PS. DENND2B activates Rab35 at the intercellular bridge, regulating cytokinetic abscission and tetraploidy. Cell Rep 2023; 42:112795. [PMID: 37454296 DOI: 10.1016/j.celrep.2023.112795] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/05/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023] Open
Abstract
Cytokinesis relies on membrane trafficking pathways regulated by Rabs and guanine nucleotide exchange factors (GEFs). During cytokinesis, the intercellular cytokinetic bridge (ICB) connecting daughter cells undergoes abscission, which requires actin depolymerization. Rab35 recruits MICAL1 to oxidize and depolymerize actin filaments. We show that DENND2B, a protein linked to cancer and congenital disorders, functions as a Rab35 GEF, recruiting and activating Rab35 at the ICB. DENND2B's N-terminal region also interacts with an active form of Rab35, suggesting that DENND2B is both a Rab35 GEF and effector. Knockdown of DENND2B delays abscission, leading to multinucleated cells and filamentous actin (F-actin) accumulation at the ICB, impairing recruitment of ESCRT-III at the abscission site. Additionally, F-actin accumulation triggers the formation of a chromatin bridge, activating the NoCut/abscission checkpoint, and DENND2B knockdown activates Aurora B kinase, a hallmark of checkpoint activation. Thus, our study identifies DENND2B as a crucial player in cytokinetic abscission.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada.
| | - Vincent Francis
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Maria S Ioannou
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Adriana Aguila
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Maleeha Khan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Emily Banks
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Gopinath Kulasekaran
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Peter S McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada.
| |
Collapse
|
22
|
Rajan S, Kudryashov DS, Reisler E. Actin Bundles Dynamics and Architecture. Biomolecules 2023; 13:450. [PMID: 36979385 PMCID: PMC10046292 DOI: 10.3390/biom13030450] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/04/2023] Open
Abstract
Cells use the actin cytoskeleton for many of their functions, including their division, adhesion, mechanosensing, endo- and phagocytosis, migration, and invasion. Actin bundles are the main constituent of actin-rich structures involved in these processes. An ever-increasing number of proteins that crosslink actin into bundles or regulate their morphology is being identified in cells. With recent advances in high-resolution microscopy and imaging techniques, the complex process of bundles formation and the multiple forms of physiological bundles are beginning to be better understood. Here, we review the physiochemical and biological properties of four families of highly conserved and abundant actin-bundling proteins, namely, α-actinin, fimbrin/plastin, fascin, and espin. We describe the similarities and differences between these proteins, their role in the formation of physiological actin bundles, and their properties-both related and unrelated to their bundling abilities. We also review some aspects of the general mechanism of actin bundles formation, which are known from the available information on the activity of the key actin partners involved in this process.
Collapse
Affiliation(s)
- Sudeepa Rajan
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Dmitri S. Kudryashov
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH 43210, USA
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
23
|
Rajan S, Terman JR, Reisler E. MICAL-mediated oxidation of actin and its effects on cytoskeletal and cellular dynamics. Front Cell Dev Biol 2023; 11:1124202. [PMID: 36875759 PMCID: PMC9982024 DOI: 10.3389/fcell.2023.1124202] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/02/2023] [Indexed: 02/19/2023] Open
Abstract
Actin and its dynamic structural remodelings are involved in multiple cellular functions, including maintaining cell shape and integrity, cytokinesis, motility, navigation, and muscle contraction. Many actin-binding proteins regulate the cytoskeleton to facilitate these functions. Recently, actin's post-translational modifications (PTMs) and their importance to actin functions have gained increasing recognition. The MICAL family of proteins has emerged as important actin regulatory oxidation-reduction (Redox) enzymes, influencing actin's properties both in vitro and in vivo. MICALs specifically bind to actin filaments and selectively oxidize actin's methionine residues 44 and 47, which perturbs filaments' structure and leads to their disassembly. This review provides an overview of the MICALs and the impact of MICAL-mediated oxidation on actin's properties, including its assembly and disassembly, effects on other actin-binding proteins, and on cells and tissue systems.
Collapse
Affiliation(s)
- Sudeepa Rajan
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jonathan R. Terman
- Departments of Neuroscience and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
24
|
Richter SM, Massman LC, Stuehr DJ, Sweeny EA. Functional interactions between NADPH oxidase 5 and actin. Front Cell Dev Biol 2023; 11:1116833. [PMID: 36776559 PMCID: PMC9909703 DOI: 10.3389/fcell.2023.1116833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
NADPH oxidase 5 (NOX5) is a transmembrane oxidative signaling enzyme which produces superoxide in response to intracellular calcium flux. Increasing evidence indicates that NOX5 is involved in a variety of physiological processes as well as human disease, however, details of NOX5 signaling pathways and targets of NOX5 mediated oxidative modifications remain poorly resolved. Actin dynamics have previously been shown to be modulated by oxidative modification, however, a direct connection to NOX5 expression and activity has not been fully explored. Here we show that NOX5 and actin interact in the cell, and each modulate the activity of the other. Using actin effector molecules jasplakinolide, cytochalasin D and latrunculin A, we show that changes in actin dynamics affect NOX5 superoxide production. In tandem, NOX5 oxidatively modifies actin, and shifts the ratio of filamentous to monomeric actin. Finally, we show that knockdown of NOX5 in the pancreatic cancer cell line PSN-1 impairs cell migration. Together our findings indicate an important link between actin dynamics and oxidative signaling through NOX5.
Collapse
Affiliation(s)
- Samantha M Richter
- Department of Biochemistry, The Medical College of Wisconsin, Milwaukee, WI, United States
| | - Lilyanna C Massman
- Department of Biochemistry, The Medical College of Wisconsin, Milwaukee, WI, United States
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, United States
| | - Elizabeth A Sweeny
- Department of Biochemistry, The Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
25
|
Andrade V, Echard A. Mechanics and regulation of cytokinetic abscission. Front Cell Dev Biol 2022; 10:1046617. [PMID: 36506096 PMCID: PMC9730121 DOI: 10.3389/fcell.2022.1046617] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Cytokinetic abscission leads to the physical cut of the intercellular bridge (ICB) connecting the daughter cells and concludes cell division. In different animal cells, it is well established that the ESCRT-III machinery is responsible for the constriction and scission of the ICB. Here, we review the mechanical context of abscission. We first summarize the evidence that the ICB is initially under high tension and explain why, paradoxically, this can inhibit abscission in epithelial cells by impacting on ESCRT-III assembly. We next detail the different mechanisms that have been recently identified to release ICB tension and trigger abscission. Finally, we discuss whether traction-induced mechanical cell rupture could represent an ancient alternative mechanism of abscission and suggest future research avenues to further understand the role of mechanics in regulating abscission.
Collapse
Affiliation(s)
- Virginia Andrade
- CNRS UMR3691, Membrane Traffic and Cell Division Unit, Institut Pasteur, Université Paris Cité, Paris, France,Collège Doctoral, Sorbonne Université, Paris, France
| | - Arnaud Echard
- CNRS UMR3691, Membrane Traffic and Cell Division Unit, Institut Pasteur, Université Paris Cité, Paris, France,*Correspondence: Arnaud Echard,
| |
Collapse
|
26
|
Li Y, Li F. Mechanism and Prospect of Gastrodin in Osteoporosis, Bone Regeneration, and Osseointegration. Pharmaceuticals (Basel) 2022; 15:1432. [PMID: 36422561 PMCID: PMC9698149 DOI: 10.3390/ph15111432] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 11/14/2023] Open
Abstract
Gastrodin, a traditional Chinese medicine ingredient, is widely used to treat vascular and neurological diseases. However, recently, an increasing number of studies have shown that gastrodin has anti-osteoporosis effects, and its mechanisms of action include its antioxidant effect, anti-inflammatory effect, and anti-apoptotic effect. In addition, gastrodin has many unique advantages in promoting bone healing in tissue engineering, such as inducing high hydrophilicity in the material surface, its anti-inflammatory effect, and pro-vascular regeneration. Therefore, this paper summarized the effects and mechanisms of gastrodin on osteoporosis and bone regeneration in the current research. Here we propose an assumption that the use of gastrodin in the surface loading of oral implants may greatly promote the osseointegration of implants and increase the success rate of implants. In addition, we speculated on the potential mechanisms of gastrodin against osteoporosis, by affecting actin filament polymerization, renin-angiotensin system (RAS) and ferroptosis, and proposed that the potential combination of gastrodin with Mg2+, angiotensin type 2 receptor blockers or artemisinin may greatly inhibit osteoporosis. The purpose of this review is to provide a reference for more in-depth research and application of gastrodin in the treatment of osteoporosis and implant osseointegration in the future.
Collapse
Affiliation(s)
| | - Fenglan Li
- Department of Prosthodontics, Shanxi Provincial People’s Hospital, Shanxi Medical University, Taiyuan 030000, China
| |
Collapse
|
27
|
McGarry DJ, Castino G, Lilla S, Carnet A, Kelly L, Micovic K, Zanivan S, Olson MF. MICAL1 activation by PAK1 mediates actin filament disassembly. Cell Rep 2022; 41:111442. [PMID: 36198272 DOI: 10.1016/j.celrep.2022.111442] [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: 09/28/2021] [Revised: 06/14/2022] [Accepted: 09/09/2022] [Indexed: 11/03/2022] Open
Abstract
The MICAL1 monooxygenase is an important regulator of filamentous actin (F-actin) structures. Although MICAL1 has been shown to be regulated via protein-protein interactions at the autoinhibitory carboxyl terminus, a link between actin-regulatory RHO GTPase signaling pathways and MICAL1 has not been established. We show that the CDC42 GTPase effector PAK1 associates with and phosphorylates MICAL1 on two serine residues, leading to accelerated F-actin disassembly. PAK1 binds to the amino-terminal catalytic monooxygenase and calponin homology domains, distinct from the autoinhibitory carboxyl terminus. Extracellular ligand stimulation leads to PAK-dependent phosphorylation, linking external signals to MICAL1 phosphorylation. Mass spectrometry indicates that MICAL1 co-expression with CDC42 and PAK1 increases MICAL1 association with hundreds of proteins, including the previously described MICAL1-interacting proteins RAB10 and RAB7A. These results provide insights into a redox-mediated pathway linking extracellular signals to cytoskeleton regulation via a RHO GTPase and indicate a means of communication between RHO and RAB GTPases.
Collapse
Affiliation(s)
- David J McGarry
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
| | - Giovanni Castino
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
| | - Sergio Lilla
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Alexandre Carnet
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
| | - Loughlin Kelly
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
| | - Katarina Micovic
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
| | - Sara Zanivan
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Michael F Olson
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada.
| |
Collapse
|
28
|
Francis CR, Kincross H, Kushner EJ. Rab35 governs apicobasal polarity through regulation of actin dynamics during sprouting angiogenesis. Nat Commun 2022; 13:5276. [PMID: 36075898 PMCID: PMC9458672 DOI: 10.1038/s41467-022-32853-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/17/2022] [Indexed: 12/01/2022] Open
Abstract
In early blood vessel development, trafficking programs, such as those using Rab GTPases, are tasked with delivering vesicular cargo with high spatiotemporal accuracy. However, the function of many Rab trafficking proteins remain ill-defined in endothelial tissue; therefore, their relevance to blood vessel development is unknown. Rab35 has been shown to play an enigmatic role in cellular behaviors which differs greatly between tissue-type and organism. Importantly, Rab35 has never been characterized for its potential contribution in sprouting angiogenesis; thus, our goal was to map Rab35’s primary function in angiogenesis. Our results demonstrate that Rab35 is critical for sprout formation; in its absence, apicobasal polarity is entirely lost in vitro and in vivo. To determine mechanism, we systematically explored established Rab35 effectors and show that none are operative in endothelial cells. However, we find that Rab35 partners with DENNd1c, an evolutionarily divergent guanine exchange factor, to localize to actin. Here, Rab35 regulates actin polymerization through limiting Rac1 and RhoA activity, which is required to set up proper apicobasal polarity during sprout formation. Our findings establish that Rab35 is a potent brake of actin remodeling during blood vessel development. The promiscuous GTPase Rab35 has been shown to be involved in many important cellular functions. In this article, Francis et al. illustrate how Rab35 acts as a critical brake to actin remodeling during sprouting angiogenesis and how it is necessary for proper blood vessel development.
Collapse
Affiliation(s)
- Caitlin R Francis
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Hayle Kincross
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Erich J Kushner
- Department of Biological Sciences, University of Denver, Denver, CO, USA.
| |
Collapse
|
29
|
Xue B, Li H, Liu S, Feng Q, Xu Y, Deng R, Chen S, Wang J, Li X, Wan M, Tang S, Zhu H. The redox cycling of STAT2 maintains innate immune homeostasis. Cell Rep 2022; 40:111215. [PMID: 35977519 DOI: 10.1016/j.celrep.2022.111215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/30/2022] [Accepted: 07/22/2022] [Indexed: 11/30/2022] Open
Abstract
Interferons (IFNs) are essential in antiviral defense, antitumor effects, and immunoregulatory activities. Although methionine oxidation is associated with various physiological and pathophysiological processes in plants, animals, and humans, its role in immunity remains unclear. We find that the redox cycling of signal transducer and activator of transcription 2 (STAT2) is an intrinsic cellular biological process, and that impairment of the redox status contributes to STAT2 methionine oxidation, inhibiting its activation. IFN protects STAT2 from methionine oxidation through the recruitment of methionine sulfoxide reductase MSRB2, whose enzymatic activity is enhanced by N-acetyltransferase 9 (NAT9), a chaperone of STAT2 defined in this study, upon IFN treatment. Consequently, loss of Nat9 renders mice more susceptible to viral infection. Our study highlights the key function of methionine oxidation in immunity, which provides evidence for the decline of immune function by aging and may provide insights into the clinical applications of IFN in immune-related diseases.
Collapse
Affiliation(s)
- Binbin Xue
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan, China
| | - Huiyi Li
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan, China
| | - Shun Liu
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan, China
| | - Qing Feng
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan, China
| | - Yan Xu
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan, China
| | - Rilin Deng
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan, China
| | - Shengwen Chen
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan, China
| | - Jingjing Wang
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan, China
| | - Xinran Li
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan, China
| | - Mengyu Wan
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan, China
| | - Songqing Tang
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan, China
| | - Haizhen Zhu
- Institute of Pathogen Biology and Immunology of College of Biology, Hunan Provincial Key Laboratory of Medical Virology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan, China; Research Center of Cancer Prevention and Treatment, Translational Medicine Research Center of Liver Cancer, Hunan Cancer Hospital, Changsha, Hunan, China.
| |
Collapse
|
30
|
Fine-tuning cell organelle dynamics during mitosis by small GTPases. Front Med 2022; 16:339-357. [PMID: 35759087 DOI: 10.1007/s11684-022-0926-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/24/2022] [Indexed: 11/04/2022]
Abstract
During mitosis, the allocation of genetic material concurs with organelle transformation and distribution. The coordination of genetic material inheritance with organelle dynamics directs accurate mitotic progression, cell fate determination, and organismal homeostasis. Small GTPases belonging to the Ras superfamily regulate various cell organelles during division. Being the key regulators of membrane dynamics, the dysregulation of small GTPases is widely associated with cell organelle disruption in neoplastic and non-neoplastic diseases, such as cancer and Alzheimer's disease. Recent discoveries shed light on the molecular properties of small GTPases as sophisticated modulators of a remarkably complex and perfect adaptors for rapid structure reformation. This review collects current knowledge on small GTPases in the regulation of cell organelles during mitosis and highlights the mediator role of small GTPase in transducing cell cycle signaling to organelle dynamics during mitosis.
Collapse
|
31
|
Rouyère C, Serrano T, Frémont S, Echard A. Oxidation and reduction of actin: Origin, impact in vitro and functional consequences in vivo. Eur J Cell Biol 2022; 101:151249. [PMID: 35716426 DOI: 10.1016/j.ejcb.2022.151249] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/13/2022] [Accepted: 06/06/2022] [Indexed: 11/15/2022] Open
Abstract
Actin is among the most abundant proteins in eukaryotic cells and assembles into dynamic filamentous networks regulated by many actin binding proteins. The actin cytoskeleton must be finely tuned, both in space and time, to fulfill key cellular functions such as cell division, cell shape changes, phagocytosis and cell migration. While actin oxidation by reactive oxygen species (ROS) at non-physiological levels are known for long to impact on actin polymerization and on the cellular actin cytoskeleton, growing evidence shows that direct and reversible oxidation/reduction of specific actin amino acids plays an important and physiological role in regulating the actin cytoskeleton. In this review, we describe which actin amino acid residues can be selectively oxidized and reduced in many different ways (e.g. disulfide bond formation, glutathionylation, carbonylation, nitration, nitrosylation and other oxidations), the cellular enzymes at the origin of these post-translational modifications, and the impact of actin redox modifications both in vitro and in vivo. We show that the regulated balance of oxidation and reduction of key actin amino acid residues contributes to the control of actin filament polymerization and disassembly at the subcellular scale and highlight how improper redox modifications of actin can lead to pathological conditions.
Collapse
Affiliation(s)
- Clémentine Rouyère
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015 Paris, France; Sorbonne Université, Collège Doctoral, F-75005 Paris, France
| | - Thomas Serrano
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015 Paris, France
| | - Stéphane Frémont
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015 Paris, France
| | - Arnaud Echard
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015 Paris, France.
| |
Collapse
|
32
|
Bettinger JQ, Simon M, Korotkov A, Welle KA, Hryhorenko JR, Seluanov A, Gorbunova V, Ghaemmaghami S. Accurate Proteomewide Measurement of Methionine Oxidation in Aging Mouse Brains. J Proteome Res 2022; 21:1495-1509. [PMID: 35584362 PMCID: PMC9171897 DOI: 10.1021/acs.jproteome.2c00127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Indexed: 11/28/2022]
Abstract
The oxidation of methionine has emerged as an important post-translational modification of proteins. A number of studies have suggested that the oxidation of methionines in select proteins can have diverse impacts on cell physiology, ranging from detrimental effects on protein stability to functional roles in cell signaling. Despite its importance, the large-scale investigation of methionine oxidation in a complex matrix, such as the cellular proteome, has been hampered by technical limitations. We report a methodology, methionine oxidation by blocking (MobB), that allows for accurate and precise quantification of low levels of methionine oxidation typically observed in vivo. To demonstrate the utility of this methodology, we analyzed the brain tissues of young (6 m.o.) and old (20 m.o.) mice and identified over 280 novel sites for in vivo methionine oxidation. We further demonstrated that oxidation stoichiometries for specific methionine residues are highly consistent between individual animals and methionine sulfoxides are enriched in clusters of functionally related gene products including membrane and extracellular proteins. However, we did not detect significant changes in methionine oxidation in brains of old mice. Our results suggest that under normal conditions, methionine oxidation may be a biologically regulated process rather than a result of stochastic chemical damage.
Collapse
Affiliation(s)
- John Q. Bettinger
- Department
of Biology, University of Rochester, Rochester, New York 14627, United States
| | - Matthew Simon
- Department
of Biology, University of Rochester, Rochester, New York 14627, United States
| | - Anatoly Korotkov
- Department
of Biology, University of Rochester, Rochester, New York 14627, United States
| | - Kevin A. Welle
- Department
of Medicine, University of Rochester Medical
Center, Rochester, New York 14627, United States
| | - Jennifer R. Hryhorenko
- Department
of Medicine, University of Rochester Medical
Center, Rochester, New York 14627, United States
| | - Andrei Seluanov
- Department
of Biology, University of Rochester, Rochester, New York 14627, United States
- Department
of Medicine, University of Rochester Medical
Center, Rochester, New York 14627, United States
| | - Vera Gorbunova
- Department
of Biology, University of Rochester, Rochester, New York 14627, United States
- Department
of Medicine, University of Rochester Medical
Center, Rochester, New York 14627, United States
| | - Sina Ghaemmaghami
- Department
of Biology, University of Rochester, Rochester, New York 14627, United States
- University
of Rochester Mass Spectrometry Resource Laboratory, Rochester, New York 14627, United States
| |
Collapse
|
33
|
Andrade V, Bai J, Gupta-Rossi N, Jimenez AJ, Delevoye C, Lamaze C, Echard A. Caveolae promote successful abscission by controlling intercellular bridge tension during cytokinesis. SCIENCE ADVANCES 2022; 8:eabm5095. [PMID: 35417244 PMCID: PMC9007517 DOI: 10.1126/sciadv.abm5095] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
During cytokinesis, the intercellular bridge (ICB) connecting the daughter cells experiences pulling forces, which delay abscission by preventing the assembly of the ESCRT scission machinery. Abscission is thus triggered by tension release, but how ICB tension is controlled is unknown. Here, we report that caveolae, which are known to regulate membrane tension upon mechanical stress in interphase cells, are located at the midbody, at the abscission site, and at the ICB/cell interface in dividing cells. Functionally, the loss of caveolae delays ESCRT-III recruitment during cytokinesis and impairs abscission. This is the consequence of a twofold increase of ICB tension measured by laser ablation, associated with a local increase in myosin II activity at the ICB/cell interface. We thus propose that caveolae buffer membrane tension and limit contractibility at the ICB to promote ESCRT-III assembly and cytokinetic abscission. Together, this work reveals an unexpected connection between caveolae and the ESCRT machinery and the first role of caveolae in cell division.
Collapse
Affiliation(s)
- Virginia Andrade
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015 Paris, France
- Sorbonne Université, Collège doctoral, F-75005 Paris, France
| | - Jian Bai
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015 Paris, France
- Sorbonne Université, Collège doctoral, F-75005 Paris, France
| | - Neetu Gupta-Rossi
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015 Paris, France
| | - Ana Joaquina Jimenez
- Dynamics of Intracellular Organization Laboratory, Institut Curie, PSL Research University, CNRS UMR 144, Sorbonne Université, 75005 Paris, France
| | - Cédric Delevoye
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005 Paris, France
- Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 75005 Paris, France
| | - Christophe Lamaze
- Institut Curie, PSL Research University, INSERM U1143, CNRS UMR 3666, Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, 26 rue d’Ulm, 75005 Paris, France
| | - Arnaud Echard
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015 Paris, France
- Corresponding author.
| |
Collapse
|
34
|
Ono Y, Matsuzawa K, Ikenouchi J. mTORC2 suppresses cell death induced by hypo-osmotic stress by promoting sphingomyelin transport. J Cell Biol 2022; 221:213090. [PMID: 35319770 PMCID: PMC8952684 DOI: 10.1083/jcb.202106160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/28/2021] [Accepted: 01/24/2022] [Indexed: 11/22/2022] Open
Abstract
Epithelial cells are constantly exposed to osmotic stress. The influx of water molecules into the cell in a hypo-osmotic environment increases plasma membrane tension as it rapidly expands. Therefore, the plasma membrane must be supplied with membrane lipids since expansion beyond its elastic limit will cause the cell to rupture. However, the molecular mechanism to maintain a constant plasma membrane tension is not known. In this study, we found that the apical membrane selectively expands when epithelial cells are exposed to hypo-osmotic stress. This requires the activation of mTORC2, which enhances the transport of secretory vesicles containing sphingomyelin, the major lipid of the apical membrane. We further show that the mTORC2–Rab35 axis plays an essential role in the defense against hypotonic stress by promoting the degradation of the actin cortex through the up-regulation of PI(4,5)P2 metabolism, which facilitates the apical tethering of sphingomyelin-loaded vesicles to relieve plasma membrane tension.
Collapse
Affiliation(s)
- Yumiko Ono
- Department of Biology, Faculty of Sciences, Kyushu University, Nishi-ku, Fukuoka, Japan
| | - Kenji Matsuzawa
- Department of Biology, Faculty of Sciences, Kyushu University, Nishi-ku, Fukuoka, Japan
| | - Junichi Ikenouchi
- Department of Biology, Faculty of Sciences, Kyushu University, Nishi-ku, Fukuoka, Japan
| |
Collapse
|
35
|
Zhang Z, Liu R, Wang Y, Wang Y, Shuai Y, Ke C, Jin R, Wang X, Luo J. Phosphorylation of MICAL2 by ARG promotes head and neck cancer tumorigenesis by regulating skeletal rearrangement. Oncogene 2022; 41:334-346. [PMID: 34750518 DOI: 10.1038/s41388-021-02101-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/22/2022]
Abstract
The actin cytoskeletal architecture provides the structural underpinnings for crucial cellular behaviors. In cancer cells, changes in the actin cytoskeleton may serve as prerequisites for proliferation, invasion, and metastatic dissemination. However, the underlying mechanisms remain largely unknown. Here, we show that MICAL2, which is increased in head and neck squamous cell carcinoma (HNSCC) and inversely associated with patient survival, promotes HNSCC growth, invasion, and migration. MICAL2 serves as a flavoprotein monooxygenase and directly induces actin filament depolymerization by specifically oxidizing the methionine 44 and 47 residues of F-actin. The kinase ARG interacts with MICAL2 and augments MICAL2-mediated actin disassembly. Direct phosphorylation assay and mass spectrometry confirmed that ARG phosphorylates MICAL2 at Tyr445, Tyr463, and Tyr488. Substitution of the Tyr445 or Tyr463 residue of purified recombinant MICAL2-redox with phenylalanine (generating a non-phosphorylatable mutant) abolishes the enhanced MICAL2-mediated F-actin disassembly induced by ARG. Consistently, ectopic expression of non-phosphorylatable MICAL2 mutants (MICAL2Y445F and MICAL2Y463F, not MICAL2Y488F) failed to ameliorate HNSCC cell growth, whereas expression of wild-type MICAL2 or MICAL2Y488F rescued the impaired proliferation induced by MICAL2 knockdown. Moreover, CCG-1423, an inhibitor of MICAL2, was shown to inhibit HNSCC cell proliferation, invasion, and migration. Taken together, our findings indicate that phosphorylation of MICAL2 at Tyr445 and Tyr463 by ARG mediates F-actin disassembly and promotes HNSCC progression.
Collapse
Affiliation(s)
- Ze Zhang
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ruoyan Liu
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Yafei Wang
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yun Wang
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yanjie Shuai
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chuangwu Ke
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Rui Jin
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xudong Wang
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jingtao Luo
- Department of Maxillofacial and Otorhinolaryngology Oncology and Department of Head and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China. .,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China. .,Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
36
|
Gu H, Li Y, Cui X, Cao H, Hou Z, Ti Y, Liu D, Gao J, Wang Y, Wen P. MICAL1 inhibits colorectal cancer cell migration and proliferation by regulating the EGR1/β-catenin signaling pathway. Biochem Pharmacol 2022; 195:114870. [PMID: 34902339 DOI: 10.1016/j.bcp.2021.114870] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/18/2021] [Accepted: 12/03/2021] [Indexed: 12/26/2022]
Abstract
MICAL1 has been reported to be involved in the malignant processes of several types of cancer cells, however, the roles of MICAL1 in colorectal cancer (CRC) have not been well-characterized. This study aims to investigate the cellular functions and molecular mechanisms of MICAL1 in CRC cells. Here, we found that both mRNA and protein levels of MICAL1 were down-regulated in colorectal cancer tissues compared with matched adjacent non-tumor tissues, and the expression level of MICAL1 was correlated with the metastatic status of colorectal cancer. Importantly, overexpression of MICAL1 significantly inhibited colorectal cancer cell migration and growth, and increased the level of E-cadherin and Occludin, and suppressed the expression level of Vimentin and N-cadherin; while silencing of MICAL1 promoted CRC cell migration and enhanced EMT. In addition, MICAL1 overexpression significantly inhibited the proliferation and growth of CRC in vitro and in vivo. Moreover, RNA sequencing and bioinformatics analysis identified that MICAL1 was closely correlated with "cell migration", "cell cycle" and "β-catenin signaling" genesets. Mechanistically, overexpression of MICAL1 downregulated the mRNA level of EGR1 and β-catenin, decreased the protein level and nuclear translocation of β-catenin, and inhibited the transcriptions of β-catenin downstream targets, c-myc and cyclin D1. The ectopic expression of EGR1 or β-catenin can significantly block the MICAL1-mediated inhibitory effects. Collectively, MICAL1 is down-regulated in CRC, and plays an inhibitory role in the migration and growth of CRC cells by suppressing the ERG1/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Huanyu Gu
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Yi Li
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Xiuping Cui
- Life Science Institute, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Huiru Cao
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Zhijuan Hou
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Yunhe Ti
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Dahua Liu
- Biological Anthropology Institute, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Jing Gao
- Department of Ultrasonography, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Yu Wang
- Life Science Institute, Jinzhou Medical University, Jinzhou 121000, Liaoning, China.
| | - Pushuai Wen
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121000, Liaoning, China; Biological Anthropology Institute, Jinzhou Medical University, Jinzhou 121000, Liaoning, China.
| |
Collapse
|
37
|
Early Pregnancy Exposure to Ambient Air Pollution among Late-Onset Preeclamptic Cases Is Associated with Placental DNA Hypomethylation of Specific Genes and Slower Placental Maturation. TOXICS 2021; 9:toxics9120338. [PMID: 34941772 PMCID: PMC8708250 DOI: 10.3390/toxics9120338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 01/19/2023]
Abstract
Exposure to ambient air pollution during pregnancy has been associated with an increased risk of preeclampsia (PE). Some suggested mechanisms behind this association are changes in placental DNA methylation and gene expression. The objective of this study was to identify how early pregnancy exposure to ambient nitrogen oxides (NOx) among PE cases and normotensive controls influence DNA methylation (EPIC array) and gene expression (RNA-seq). The study included placentas from 111 women (29 PE cases/82 controls) in Scania, Sweden. First-trimester NOx exposure was assessed at the participants’ residence using a dispersion model and categorized via median split into high or low NOx. Placental gestational epigenetic age was derived from the DNA methylation data. We identified six differentially methylated positions (DMPs, q < 0.05) comparing controls with low NOx vs. cases with high NOx and 14 DMPs comparing cases and controls with high NOx. Placentas with female fetuses showed more DMPs (N = 309) than male-derived placentas (N = 1). Placentas from PE cases with high NOx demonstrated gestational age deceleration compared to controls with low NOx (p = 0.034). No differentially expressed genes (DEGs, q < 0.05) were found. In conclusion, early pregnancy exposure to NOx affected placental DNA methylation in PE, resulting in placental immaturity and showing sexual dimorphism.
Collapse
|
38
|
The Abscission Checkpoint: A Guardian of Chromosomal Stability. Cells 2021; 10:cells10123350. [PMID: 34943860 PMCID: PMC8699595 DOI: 10.3390/cells10123350] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/11/2022] Open
Abstract
The abscission checkpoint contributes to the fidelity of chromosome segregation by delaying completion of cytokinesis (abscission) when there is chromatin lagging in the intercellular bridge between dividing cells. Although additional triggers of an abscission checkpoint-delay have been described, including nuclear pore defects, replication stress or high intercellular bridge tension, this review will focus only on chromatin bridges. In the presence of such abnormal chromosomal tethers in mammalian cells, the abscission checkpoint requires proper localization and optimal kinase activity of the Chromosomal Passenger Complex (CPC)-catalytic subunit Aurora B at the midbody and culminates in the inhibition of Endosomal Sorting Complex Required for Transport-III (ESCRT-III) components at the abscission site to delay the final cut. Furthermore, cells with an active checkpoint stabilize the narrow cytoplasmic canal that connects the two daughter cells until the chromatin bridges are resolved. Unsuccessful resolution of chromatin bridges in checkpoint-deficient cells or in cells with unstable intercellular canals can lead to chromatin bridge breakage or tetraploidization by regression of the cleavage furrow. In turn, these outcomes can lead to accumulation of DNA damage, chromothripsis, generation of hypermutation clusters and chromosomal instability, which are associated with cancer formation or progression. Recently, many important questions regarding the mechanisms of the abscission checkpoint have been investigated, such as how the presence of chromatin bridges is signaled to the CPC, how Aurora B localization and kinase activity is regulated in late midbodies, the signaling pathways by which Aurora B implements the abscission delay, and how the actin cytoskeleton is remodeled to stabilize intercellular canals with DNA bridges. Here, we review recent progress toward understanding the mechanisms of the abscission checkpoint and its role in guarding genome integrity at the chromosome level, and consider its potential implications for cancer therapy.
Collapse
|
39
|
McGarry DJ, Armstrong G, Castino G, Mason S, Clark W, Shaw R, McGarry L, Blyth K, Olson MF. MICAL1 regulates actin cytoskeleton organization, directional cell migration and the growth of human breast cancer cells as orthotopic xenograft tumours. Cancer Lett 2021; 519:226-236. [PMID: 34314753 DOI: 10.1016/j.canlet.2021.07.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/19/2021] [Accepted: 07/22/2021] [Indexed: 02/09/2023]
Abstract
The Molecule Interacting with CasL 1 (MICAL1) monooxygenase has emerged as an important regulator of cytoskeleton organization via actin oxidation. Although filamentous actin (F-actin) increases MICAL1 monooxygenase activity, hydrogen peroxide (H2O2) is also generated in the absence of F-actin, suggesting that diffusible H2O2 might have additional functions. MICAL1 gene disruption by CRISPR/Cas9 in MDA MB 231 human breast cancer cells knocked out (KO) protein expression, which affected F-actin organization, cell size and motility. Transcriptomic profiling revealed that MICAL1 deletion significantly affected the expression of over 700 genes, with the majority being reduced in their expression levels. In addition, the absolute magnitudes of reduced gene expression were significantly greater than the magnitudes of increased gene expression. Gene set enrichment analysis (GSEA) identified receptor regulator activity as the most significant negatively enriched molecular function gene set. The prominent influence exerted by MICAL1 on F-actin structures was also associated with changes in the expression of several serum-response factor (SRF) regulated genes in KO cells. Moreover, MICAL1 disruption attenuated breast cancer tumour growth in vivo. Elevated MICAL1 gene expression was observed in invasive breast cancer samples from human patients relative to normal tissue, while MICAL1 amplification or point mutations were associated with reduced progression free survival. Collectively, these results demonstrate that MICAL1 gene disruption altered cytoskeleton organization, cell morphology and migration, gene expression, and impaired tumour growth in an orthotopic in vivo breast cancer model, suggesting that pharmacological MICAL1 inhibition could have therapeutic benefits for cancer patients.
Collapse
Affiliation(s)
- David J McGarry
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Garett Armstrong
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Giovanni Castino
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Susan Mason
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | - Robin Shaw
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Lynn McGarry
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Karen Blyth
- Cancer Research UK Beatson Institute, Glasgow, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Michael F Olson
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
40
|
Grintsevich EE, Ahmed G, Ginosyan AA, Wu H, Rich SK, Reisler E, Terman JR. Profilin and Mical combine to impair F-actin assembly and promote disassembly and remodeling. Nat Commun 2021; 12:5542. [PMID: 34545088 PMCID: PMC8452626 DOI: 10.1038/s41467-021-25781-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/24/2021] [Indexed: 11/30/2022] Open
Abstract
Cellular events require the spatiotemporal interplay between actin assembly and actin disassembly. Yet, how different factors promote the integration of these two opposing processes is unclear. In particular, cellular monomeric (G)-actin is complexed with profilin, which inhibits spontaneous actin nucleation but fuels actin filament (F-actin) assembly by elongation-promoting factors (formins, Ena/VASP). In contrast, site-specific F-actin oxidation by Mical promotes F-actin disassembly and release of polymerization-impaired Mical-oxidized (Mox)-G-actin. Here we find that these two opposing processes connect with one another to orchestrate actin/cellular remodeling. Specifically, we find that profilin binds Mox-G-actin, yet these complexes do not fuel elongation factors’-mediated F-actin assembly, but instead inhibit polymerization and promote further Mox-F-actin disassembly. Using Drosophila as a model system, we show that similar profilin–Mical connections occur in vivo – where they underlie F-actin/cellular remodeling that accompanies Semaphorin–Plexin cellular/axon repulsion. Thus, profilin and Mical combine to impair F-actin assembly and promote F-actin disassembly, while concomitantly facilitating cellular remodeling and plasticity. Actin-based structures in cells and tissues are built and maintained through a poorly understood balance between assembly and disassembly. Here, our findings provide insights into how factors known to promote these opposing effects dynamically integrate to shape cells and tissue systems.
Collapse
Affiliation(s)
- Elena E Grintsevich
- Department of Chemistry and Biochemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA. .,Department of Chemistry and Biochemistry, California State University, Long Beach (CSULB), Long Beach, CA, 90840, USA.
| | - Giasuddin Ahmed
- Departments of Neuroscience and Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Anush A Ginosyan
- Department of Chemistry and Biochemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Heng Wu
- Departments of Neuroscience and Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Shannon K Rich
- Departments of Neuroscience and Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA. .,Molecular Biology Institute, UCLA, Los Angeles, CA, 90095, USA.
| | - Jonathan R Terman
- Departments of Neuroscience and Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
41
|
Gibieža P, Petrikaitė V. The regulation of actin dynamics during cell division and malignancy. Am J Cancer Res 2021; 11:4050-4069. [PMID: 34659876 PMCID: PMC8493394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/29/2021] [Indexed: 06/13/2023] Open
Abstract
Actin is the most abundant protein in almost all the eukaryotic cells. Actin amino acid sequences are highly conserved and have not changed a lot during the progress of evolution, varying by no more than 20% in the completely different species, such as humans and algae. The network of actin filaments plays a crucial role in regulating cells' cytoskeleton that needs to undergo dynamic tuning and structural changes in order for various functional processes, such as cell motility, migration, adhesion, polarity establishment, cell growth and cell division, to take place in live cells. Owing to its fundamental role in the cell, actin is a prominent regulator of cell division, a process, whose success directly depends on morphological changes of actin cytoskeleton and correct segregation of duplicated chromosomes. Disorganization of actin framework during the last stage of cell division, known as cytokinesis, can lead to multinucleation and formation of polyploidy in post-mitotic cells, eventually developing into cancer. In this review, we will cover the principles of actin regulation during cell division and discuss how the control of actin dynamics is altered during the state of malignancy.
Collapse
Affiliation(s)
- Paulius Gibieža
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Kaunas, LT-50162, Lithuania
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Kaunas, LT-50162, Lithuania
| |
Collapse
|
42
|
Jewett CE, Soh AWJ, Lin CH, Lu Q, Lencer E, Westlake CJ, Pearson CG, Prekeris R. RAB19 Directs Cortical Remodeling and Membrane Growth for Primary Ciliogenesis. Dev Cell 2021; 56:325-340.e8. [PMID: 33561422 DOI: 10.1016/j.devcel.2020.12.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 10/09/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022]
Abstract
Primary cilia are sensory organelles that utilize the compartmentalization of membrane and cytoplasm to communicate signaling events, and yet, how the formation of a cilium is coordinated with reorganization of the cortical membrane and cytoskeleton is unclear. Using polarized epithelia, we find that cortical actin clearing and apical membrane partitioning occur where the centrosome resides at the cell surface prior to ciliation. RAB19, a previously uncharacterized RAB, associates with the RAB-GAP TBC1D4 and the HOPS-tethering complex to coordinate cortical clearing and ciliary membrane growth, which is essential for ciliogenesis. This RAB19-directed pathway is not exclusive to polarized epithelia, as RAB19 loss in nonpolarized cell types blocks ciliogenesis with a docked ciliary vesicle. Remarkably, inhibiting actomyosin contractility can substitute for the function of the RAB19 complex and restore ciliogenesis in knockout cells. Together, this work provides a mechanistic understanding behind a cytoskeletal clearing and membrane partitioning step required for ciliogenesis.
Collapse
Affiliation(s)
- Cayla E Jewett
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Adam W J Soh
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Carrie H Lin
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Quanlong Lu
- Laboratory of Cell and Developmental Signaling, National Cancer Institute-Frederick, Frederick, MD 21702, USA
| | - Ezra Lencer
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christopher J Westlake
- Laboratory of Cell and Developmental Signaling, National Cancer Institute-Frederick, Frederick, MD 21702, USA
| | - Chad G Pearson
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
43
|
Record J, Saeed MB, Venit T, Percipalle P, Westerberg LS. Journey to the Center of the Cell: Cytoplasmic and Nuclear Actin in Immune Cell Functions. Front Cell Dev Biol 2021; 9:682294. [PMID: 34422807 PMCID: PMC8375500 DOI: 10.3389/fcell.2021.682294] [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: 03/18/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Actin cytoskeletal dynamics drive cellular shape changes, linking numerous cell functions to physiological and pathological cues. Mutations in actin regulators that are differentially expressed or enriched in immune cells cause severe human diseases known as primary immunodeficiencies underscoring the importance of efficienct actin remodeling in immune cell homeostasis. Here we discuss recent findings on how immune cells sense the mechanical properties of their environement. Moreover, while the organization and biochemical regulation of cytoplasmic actin have been extensively studied, nuclear actin reorganization is a rapidly emerging field that has only begun to be explored in immune cells. Based on the critical and multifaceted contributions of cytoplasmic actin in immune cell functionality, nuclear actin regulation is anticipated to have a large impact on our understanding of immune cell development and functionality.
Collapse
Affiliation(s)
- Julien Record
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Mezida B. Saeed
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Tomas Venit
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
| | - Piergiorgio Percipalle
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Lisa S. Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
44
|
Galloni C, Carra D, Abella JV, Kjær S, Singaravelu P, Barry DJ, Kogata N, Guérin C, Blanchoin L, Way M. MICAL2 enhances branched actin network disassembly by oxidizing Arp3B-containing Arp2/3 complexes. J Cell Biol 2021; 220:e202102043. [PMID: 34106209 PMCID: PMC8193582 DOI: 10.1083/jcb.202102043] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 04/27/2021] [Accepted: 05/20/2021] [Indexed: 01/24/2023] Open
Abstract
The mechanisms regulating the disassembly of branched actin networks formed by the Arp2/3 complex still remain to be fully elucidated. In addition, the impact of Arp3 isoforms on the properties of Arp2/3 are also unexplored. We now demonstrate that Arp3 and Arp3B isocomplexes promote actin assembly equally efficiently but generate branched actin networks with different disassembly rates. Arp3B dissociates significantly faster than Arp3 from the network, and its depletion increases actin stability. This difference is due to the oxidation of Arp3B, but not Arp3, by the methionine monooxygenase MICAL2, which is recruited to the actin network by coronin 1C. Substitution of Arp3B Met293 by threonine, the corresponding residue in Arp3, increases actin network stability. Conversely, replacing Arp3 Thr293 with glutamine to mimic Met oxidation promotes disassembly. The ability of MICAL2 to enhance network disassembly also depends on cortactin. Our observations demonstrate that coronin 1C, cortactin, and MICAL2 act together to promote disassembly of branched actin networks by oxidizing Arp3B-containing Arp2/3 complexes.
Collapse
Affiliation(s)
- Chiara Galloni
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, UK
| | - Davide Carra
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, UK
| | - Jasmine V.G. Abella
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, UK
| | - Svend Kjær
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Pavithra Singaravelu
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, University of Grenoble-Alpes, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Grenoble, France
- CytoMorpho Lab, Institut de Recherche Saint Louis, University of Paris, Institut National de la Santé et de la Recherche Médicale, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Paris, France
| | - David J. Barry
- Advanced Light Microscopy Facility, The Francis Crick Institute, London, UK
| | - Naoko Kogata
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, UK
| | - Christophe Guérin
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, University of Grenoble-Alpes, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Grenoble, France
- CytoMorpho Lab, Institut de Recherche Saint Louis, University of Paris, Institut National de la Santé et de la Recherche Médicale, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Paris, France
| | - Laurent Blanchoin
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, University of Grenoble-Alpes, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Grenoble, France
- CytoMorpho Lab, Institut de Recherche Saint Louis, University of Paris, Institut National de la Santé et de la Recherche Médicale, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Paris, France
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, UK
- Department of Infectious Disease, Imperial College, London, UK
| |
Collapse
|
45
|
Belicova L, Repnik U, Delpierre J, Gralinska E, Seifert S, Valenzuela JI, Morales-Navarrete HA, Franke C, Räägel H, Shcherbinina E, Prikazchikova T, Koteliansky V, Vingron M, Kalaidzidis YL, Zatsepin T, Zerial M. Anisotropic expansion of hepatocyte lumina enforced by apical bulkheads. J Cell Biol 2021; 220:212522. [PMID: 34328499 PMCID: PMC8329733 DOI: 10.1083/jcb.202103003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/11/2021] [Accepted: 07/08/2021] [Indexed: 12/20/2022] Open
Abstract
Lumen morphogenesis results from the interplay between molecular pathways and mechanical forces. In several organs, epithelial cells share their apical surfaces to form a tubular lumen. In the liver, however, hepatocytes share the apical surface only between adjacent cells and form narrow lumina that grow anisotropically, generating a 3D network of bile canaliculi (BC). Here, by studying lumenogenesis in differentiating mouse hepatoblasts in vitro, we discovered that adjacent hepatocytes assemble a pattern of specific extensions of the apical membrane traversing the lumen and ensuring its anisotropic expansion. These previously unrecognized structures form a pattern, reminiscent of the bulkheads of boats, also present in the developing and adult liver. Silencing of Rab35 resulted in loss of apical bulkheads and lumen anisotropy, leading to cyst formation. Strikingly, we could reengineer hepatocyte polarity in embryonic liver tissue, converting BC into epithelial tubes. Our results suggest that apical bulkheads are cell-intrinsic anisotropic mechanical elements that determine the elongation of BC during liver tissue morphogenesis.
Collapse
Affiliation(s)
- Lenka Belicova
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Urska Repnik
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Julien Delpierre
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Elzbieta Gralinska
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Sarah Seifert
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | | | - Christian Franke
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Helin Räägel
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Nelson Laboratories LLC, Salt Lake City, UT
| | | | | | | | - Martin Vingron
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | | | - Timofei Zatsepin
- Skolkovo Institute of Science and Technology, Skolkovo, Russia.,Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Marino Zerial
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
46
|
Erdmann C, Hassoun R, Schmitt S, Kikuti C, Houdusse A, Mazur AJ, Mügge A, Hamdani N, Geyer M, Jaquet K, Mannherz HG. Integration of Cardiac Actin Mutants Causing Hypertrophic (p.A295S) and Dilated Cardiomyopathy (p.R312H and p.E361G) into Cellular Structures. Antioxidants (Basel) 2021; 10:antiox10071082. [PMID: 34356314 PMCID: PMC8301065 DOI: 10.3390/antiox10071082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/26/2021] [Accepted: 06/30/2021] [Indexed: 02/03/2023] Open
Abstract
The human mutant cardiac α-actins p.A295S or p.R312H and p.E361G, correlated with hypertrophic or dilated cardiomyopathy, respectively, were expressed by the baculovirus/Sf21 insect cell system and purified to homogeneity. The purified cardiac actins maintained their native state but showed differences in Ca2+-sensitivity to stimulate the myosin-subfragment1 ATPase. Here we analyzed the interactions of these c-actins with actin-binding and -modifying proteins implicated in cardiomyocyte differentiation. We demonstrate that Arp2/3 complex and the formin mDia3 stimulated the polymerization rate and extent of the c-actins, albeit to different degrees. In addition, we tested the effect of the MICAL-1 monooxygenase, which modifies the supramolecular actin organization during development and adaptive processes. MICAL-1 oxidized these c-actin variants and induced their de-polymerization, albeit at different rates. Transfection experiments using MDCK cells demonstrated the preferable incorporation of wild type and p.A295S c-actins into their microfilament system but of p.R312H and p.E361G actins into the submembranous actin network. Transduction of neonatal rat cardiomyocytes with adenoviral constructs coding HA-tagged c-actin variants showed their incorporation into microfilaments after one day in culture and thereafter into thin filaments of nascent sarcomeric structures at their plus ends (Z-lines) except the p.E361G mutant, which preferentially incorporated at the minus ends.
Collapse
Affiliation(s)
- Constanze Erdmann
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, D-44780 Bochum, Germany;
| | - Roua Hassoun
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, D-44780 Bochum, Germany; (R.H.); (A.M.); (N.H.); (K.J.)
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, D-44780 Bochum, Germany
| | - Sebastian Schmitt
- Institute of Structural Biology, University of Bonn, D-53127 Bonn, Germany; (S.S.); (M.G.)
| | - Carlos Kikuti
- Institut Curie, Structural Motility Team, F-75005 Paris, France; (C.K.); (A.H.)
| | - Anne Houdusse
- Institut Curie, Structural Motility Team, F-75005 Paris, France; (C.K.); (A.H.)
| | - Antonina J. Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Pl-50-383 Wroclaw, Poland;
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, D-44780 Bochum, Germany; (R.H.); (A.M.); (N.H.); (K.J.)
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, D-44780 Bochum, Germany
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, D-44780 Bochum, Germany; (R.H.); (A.M.); (N.H.); (K.J.)
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, D-44780 Bochum, Germany
| | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, D-53127 Bonn, Germany; (S.S.); (M.G.)
| | - Kornelia Jaquet
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, D-44780 Bochum, Germany; (R.H.); (A.M.); (N.H.); (K.J.)
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, D-44780 Bochum, Germany
| | - Hans Georg Mannherz
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, D-44780 Bochum, Germany;
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, D-44780 Bochum, Germany; (R.H.); (A.M.); (N.H.); (K.J.)
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, D-44780 Bochum, Germany
- Correspondence: ; Fax: +49-234-3214474
| |
Collapse
|
47
|
Iannantuono NVG, Emery G. Rab11FIP1 maintains Rab35 at the intercellular bridge to promote actin removal and abscission. J Cell Sci 2021; 134:jcs244384. [PMID: 34152390 DOI: 10.1242/jcs.244384] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/20/2021] [Indexed: 11/20/2022] Open
Abstract
Cytokinesis occurs at the end of mitosis/meiosis wherein the cytoplasms of daughter cells are separated. Before abscission, an intercellular bridge containing the remaining furrowing machinery, mitotic spindle and actin cytoskeleton connects the two daughter cells. To remove this actin and allow for the separation of daughter cells, Rab35 vesicles, loaded with the actin oxidizer MICAL1 and the inositol polyphosphate 5-phosphatase OCRL, are recruited to the midbody in a fine-tuned spatiotemporal manner. However, importantly, the means by which these vesicles are recruited is currently unclear. Here, we demonstrate that Rab11FIP1 is recruited to the midbody after Rab35 to scaffold it at the bridge and maintain Rab35 in this region. In the absence of Rab11FIP1, Rab35 dramatically drops from the midbody, inducing defects, such as cytokinetic delays and binucleation due to actin overaccumulation at the intercellular bridge, which can be rescued with Latrunculin A treatment. Importantly, we show that Rab11FIP1 is critical for Rab35 function in actin removal prior to cytokinesis. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Nicholas V G Iannantuono
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Downtown station, Montréal, Québec H3C 3J7, Canada
| | - Gregory Emery
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, P.O. Box 6128, Downtown station, Montréal, Québec H3C 3J7, Canada
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| |
Collapse
|
48
|
Gibieža P, Petrikaitė V. The dual functions of Rab11 and Rab35 GTPases-regulation of cell division and promotion of tumorigenicity. Am J Cancer Res 2021; 11:1861-1872. [PMID: 34094658 PMCID: PMC8167671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/01/2021] [Indexed: 06/12/2023] Open
Abstract
The broad studies of cancer have led researchers to the creditable understanding of biological and environmental factors that make benign cells to become malignant, as well as the developmental aspects of the tumour cells, known as the "hallmarks of cancer". However, additional research is needed to uncover the features of cancer biology, which would allow to design new and more effective treatment strategies for cancer patients. Since RabGTPases and their effectors are frequently altered in cancer, their role in a regulation of cell division leading to the acquisition of cancer cell-like phenotype has drawn a lot of attention from different research groups in recent years. Both, Rab11 and Rab35 belong to a superfamily of small monomeric GTPases that regulate a diverse array of cellular functions. Lately, Rab11 and Rab35 were declared as oncogenic, and because of their association with abundant cellular functions, a linkage to the induction of cancer, has been proposed. Although the clear connection between the improper regulation of Rab11 or Rab35 and the initiation of tumorigenicity has only beginning to emerge, in this review we will discuss the newest findings regarding the participation of RabGTPases in a control of cell division and promotion of tumorigenesis, trying to link the actual function to the cancer causality.
Collapse
Affiliation(s)
- Paulius Gibieža
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Kaunas, LT-50162, Lithuania
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences Kaunas, LT-50162, Lithuania
| |
Collapse
|
49
|
Willoughby PM, Allen M, Yu J, Korytnikov R, Chen T, Liu Y, So I, Macpherson N, Mitchell JA, Fernandez-Gonzalez R, Bruce AE. The recycling endosome protein Rab25 coordinates collective cell movements in the zebrafish surface epithelium. eLife 2021; 10:66060. [PMID: 33755014 PMCID: PMC8034978 DOI: 10.7554/elife.66060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/22/2021] [Indexed: 12/16/2022] Open
Abstract
In emerging epithelial tissues, cells undergo dramatic rearrangements to promote tissue shape changes. Dividing cells remain interconnected via transient cytokinetic bridges. Bridges are cleaved during abscission and currently, the consequences of disrupting abscission in developing epithelia are not well understood. We show that the Rab GTPase Rab25 localizes near cytokinetic midbodies and likely coordinates abscission through endomembrane trafficking in the epithelium of the zebrafish gastrula during epiboly. In maternal-zygotic Rab25a and Rab25b mutant embryos, morphogenic activity tears open persistent apical cytokinetic bridges that failed to undergo timely abscission. Cytokinesis defects result in anisotropic cell morphologies that are associated with a reduction of contractile actomyosin networks. This slows cell rearrangements and alters the viscoelastic responses of the tissue, all of which likely contribute to delayed epiboly. We present a model in which Rab25 trafficking coordinates cytokinetic bridge abscission and cortical actin density, impacting local cell shape changes and tissue-scale forces.
Collapse
Affiliation(s)
| | - Molly Allen
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Jessica Yu
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, University of Toronto, Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Roman Korytnikov
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Tianhui Chen
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Yupeng Liu
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Isis So
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Neil Macpherson
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Rodrigo Fernandez-Gonzalez
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, University of Toronto, Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Ashley Ee Bruce
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| |
Collapse
|
50
|
Signal-regulated oxidation of proteins via MICAL. Biochem Soc Trans 2021; 48:613-620. [PMID: 32219383 DOI: 10.1042/bst20190866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/09/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022]
Abstract
Processing of and responding to various signals is an essential cellular function that influences survival, homeostasis, development, and cell death. Extra- or intracellular signals are perceived via specific receptors and transduced in a particular signalling pathway that results in a precise response. Reversible post-translational redox modifications of cysteinyl and methionyl residues have been characterised in countless signal transduction pathways. Due to the low reactivity of most sulfur-containing amino acid side chains with hydrogen peroxide, for instance, and also to ensure specificity, redox signalling requires catalysis, just like phosphorylation signalling requires kinases and phosphatases. While reducing enzymes of both cysteinyl- and methionyl-derivates have been characterised in great detail before, the discovery and characterisation of MICAL proteins evinced the first examples of specific oxidases in signal transduction. This article provides an overview of the functions of MICAL proteins in the redox regulation of cellular functions.
Collapse
|