1
|
Woodward BL, Lahiri S, Chauhan AS, Garcia MR, Goodley LE, Clarke TL, Pal M, Agathanggelou A, Jhujh SS, Ganesh AN, Hollins FM, Deforie VG, Maroofian R, Efthymiou S, Meinhardt A, Mathew CG, Simpson MA, Mefford HC, Faqeih EA, Rosenzweig SD, Volpi S, Di Matteo G, Cancrini C, Scardamaglia A, Shackley F, Davies EG, Ibrahim S, Arkwright PD, Zaki MS, Stankovic T, Taylor AMR, Mazur AJ, Di Donato N, Houlden H, Rothenberg E, Stewart GS. Inherited deficiency of DIAPH1 identifies a DNA double strand break repair pathway regulated by γ-actin. Nat Commun 2025; 16:4491. [PMID: 40368919 PMCID: PMC12078678 DOI: 10.1038/s41467-025-59553-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
DNA double strand break repair (DSBR) represents a fundamental process required to maintain genome stability and prevent the onset of disease. Whilst cell cycle phase and the chromatin context largely dictate which repair pathway is utilised to restore damaged DNA, it has been recently shown that nuclear actin filaments play a major role in clustering DNA breaks to facilitate DSBR by homologous recombination (HR). However, the mechanism with which nuclear actin and the different actin nucleating factors regulate HR is unclear. Interestingly, patients with biallelic mutations in the actin nucleating factor DIAPH1 exhibit a striking overlap of clinical features with the HR deficiency disorders, Nijmegen Breakage Syndrome (NBS) and Warsaw Breakage Syndrome (WABS). This suggests that DIAPH1 may play a role in regulating HR and that some of the clinical deficits associated with DIAPH1 mutations may be caused by an underlying DSBR defect. In keeping with this clinical similarity, we demonstrate that cells from DIAL (DIAPH1 Loss-of-function) Syndrome patients display an HR repair defect comparable to loss of NBS1. Moreover, we show that this DSBR defect is also observed in a subset of patients with Baraitser-Winter Cerebrofrontofacial (BWCFF) syndrome associated with mutations in ACTG1 (γ-actin) but not ACTB (β-actin). Lastly, we demonstrate that DIAPH1 and γ-actin promote HR-dependent repair by facilitating the relocalisation of the MRE11/RAD50/NBS1 complex to sites of DNA breaks to initiate end-resection. Taken together, these data provide a mechanistic explanation for the overlapping clinical symptoms exhibited by patients with DIAL syndrome, BWCFF syndrome and NBS.
Collapse
Affiliation(s)
- Beth L Woodward
- Department of Cancer and Genomic Sciences, College of Medical and Health, University of Birmingham, Birmingham, UK
| | - Sudipta Lahiri
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Anoop S Chauhan
- Department of Cancer and Genomic Sciences, College of Medical and Health, University of Birmingham, Birmingham, UK
| | - Marcos Rios Garcia
- Department of Cancer and Genomic Sciences, College of Medical and Health, University of Birmingham, Birmingham, UK
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Lucy E Goodley
- Department of Cancer and Genomic Sciences, College of Medical and Health, University of Birmingham, Birmingham, UK
| | - Thomas L Clarke
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Mohinder Pal
- School of Natural Sciences, University of Kent, Canterbury, UK
| | - Angelo Agathanggelou
- Department of Cancer and Genomic Sciences, College of Medical and Health, University of Birmingham, Birmingham, UK
| | - Satpal S Jhujh
- Department of Cancer and Genomic Sciences, College of Medical and Health, University of Birmingham, Birmingham, UK
| | - Anil N Ganesh
- Department of Cancer and Genomic Sciences, College of Medical and Health, University of Birmingham, Birmingham, UK
| | - Fay M Hollins
- Department of Cancer and Genomic Sciences, College of Medical and Health, University of Birmingham, Birmingham, UK
| | - Valentina Galassi Deforie
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, UK
| | - Reza Maroofian
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, UK
| | - Stephanie Efthymiou
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, UK
| | - Andrea Meinhardt
- Institute for Clinical Genetics, University Hospital Carl Gustav Carus at TUD Dresden University of Technology and Faculty of Medicine of TUD Dresden University of Technology, Dresden, Germany
| | - Christopher G Mathew
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
- Department of Medical and Molecular Genetics, Faculty of Life Science and Medicine, King's College London, Guy's Hospital, London, UK
| | - Michael A Simpson
- Department of Medical and Molecular Genetics, Faculty of Life Science and Medicine, King's College London, Guy's Hospital, London, UK
| | - Heather C Mefford
- Center for Pediatric Neurological Disease Research, St. Jude Children's Hospital, Memphis, TN, USA
| | - Eissa A Faqeih
- King Fahad Medical City, Children's Hospital,, Riyadh, Kingdom of Saudi Arabia
| | - Sergio D Rosenzweig
- Immunology Service, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Stefano Volpi
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- DINOGMI, Università degli Studi di Genova, Genoa, Italy
| | - Gigliola Di Matteo
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
- Research Unit of Primary Immunodeficiencies, Unit of Clinical Immunology and Vaccinology, Scientific Institute for Research and Healthcare (IRCCS) Bambino Gesù Children Hospital, Rome, Italy
| | - Caterina Cancrini
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
- Research Unit of Primary Immunodeficiencies, Unit of Clinical Immunology and Vaccinology, Scientific Institute for Research and Healthcare (IRCCS) Bambino Gesù Children Hospital, Rome, Italy
| | - Annarita Scardamaglia
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, UK
| | - Fiona Shackley
- Paediatric Immunology, Allergy and Infectious Diseases, Sheffield Children's Hospital NHS Foundation Trust, Sheffield, UK
| | - E Graham Davies
- Department of Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Shahnaz Ibrahim
- Department of pediatrics and child health, Aga Khan University, Karachi, Pakistan
| | - Peter D Arkwright
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - Tatjana Stankovic
- Department of Cancer and Genomic Sciences, College of Medical and Health, University of Birmingham, Birmingham, UK
| | - A Malcolm R Taylor
- Department of Cancer and Genomic Sciences, College of Medical and Health, University of Birmingham, Birmingham, UK
| | - Antonina J Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Nataliya Di Donato
- Institute for Human Genetics, Hannover Medical School, Hannover, Germany
| | - Henry Houlden
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, UK
| | - Eli Rothenberg
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Grant S Stewart
- Department of Cancer and Genomic Sciences, College of Medical and Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
2
|
Zhao B, Ye DM, Li S, Zhang Y, Zheng Y, Kang J, Wang L, Zhao N, Ahmad B, Sun J, Yu T, Wu H. FMNL3 Promotes Migration and Invasion of Breast Cancer Cells via Inhibiting Rad23B-Induced Ubiquitination of Twist1. J Cell Physiol 2025; 240:e31481. [PMID: 39582466 DOI: 10.1002/jcp.31481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024]
Abstract
Breast cancer is a heterogeneous malignant tumor, and its high metastasis rate depends on the abnormal activation of cell dynamics. Formin-like protein 3 (FMNL3) plays an important role in the formation of various cytoskeletons that participate in cell movement. The objective of this study was to explore the function of FMNL3 in breast cancer progression and endeavor to reveal the molecular mechanism of this phenomenon. We found that FMNL3 was abnormally highly expressed in aggressive breast cancer cells and tissues, and it significantly inhibited E-cadherin expression. FMNL3 could specifically interact with Twist1 rather than other epithelial-mesenchymal transition transcription factors (EMT-TFs). We also found that FMNL3 enhanced the repressive effect of Twist1 on CDH1 transcription in breast cancer cells. Further mechanism studies showed that FMNL3 suppressed the ubiquitin degradation of Twist1 by inhibiting the interaction between Twist1 and Rad23B, the ubiquitin transfer protein of Twist1. In vitro functional experiments, it was confirmed that FMNL3 promoted the migration and invasion of breast cancer cells by regulating Twist1. Furthermore, Twist1 could directly bind to the fmnl3 promoter to facilitate FMNL3 transcription. To conclude, this study indicated that FMNL3 acted as a pro-metastasis factor in breast cancer by promoting Twist1 stability to suppress CDH1 transcription.
Collapse
Affiliation(s)
- Binggong Zhao
- School of Bioengineering and Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning, China
| | - Dong-Man Ye
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Shujing Li
- School of Bioengineering and Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning, China
| | - Yong Zhang
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Yang Zheng
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Jie Kang
- School of Bioengineering and Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning, China
| | - Luhong Wang
- School of Bioengineering and Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning, China
| | - Nannan Zhao
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Bashir Ahmad
- School of Bioengineering and Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning, China
| | - Jing Sun
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning, China
| | - Huijian Wu
- School of Bioengineering and Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning, China
| |
Collapse
|
3
|
Courtemanche N, Henty-Ridilla JL. Actin filament dynamics at barbed ends: New structures, new insights. Curr Opin Cell Biol 2024; 90:102419. [PMID: 39178734 PMCID: PMC11492572 DOI: 10.1016/j.ceb.2024.102419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/30/2024] [Accepted: 07/31/2024] [Indexed: 08/26/2024]
Abstract
The dynamic actin cytoskeleton contributes to many critical biological processes by providing the structural support underlying the morphology of most cells, facilitating intracellular transport, and generating forces required for cell motility and division. To execute many of these functions, actin monomers polymerize into polarized filaments that display different structural and biochemical properties at each end. Filament dynamics are regulated by diverse regulatory proteins which collaborate to dictate rates of elongation and disassembly, particularly at the fast-growing barbed (plus) end. This review highlights the biochemical mechanisms of six barbed end regulatory proteins: formin, profilin, capping protein, IQGAP1, cyclase-associated protein, and twinfilin. We discuss how individual proteins influence actin dynamics and how several intriguing complex assemblies influence the polymerization fate of actin filaments. Understanding these mechanisms offers insights into how actin is regulated in essential cell processes and dysregulated in disease.
Collapse
Affiliation(s)
- Naomi Courtemanche
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Jessica L Henty-Ridilla
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
4
|
Palmer NJ, Barrie KR, Dominguez R. Mechanisms of actin filament severing and elongation by formins. Nature 2024; 632:437-442. [PMID: 38843827 PMCID: PMC11375965 DOI: 10.1038/s41586-024-07637-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/31/2024] [Indexed: 08/02/2024]
Abstract
Humans express 15 formins that play crucial roles in actin-based processes, including cytokinesis, cell motility and mechanotransduction1,2. However, the lack of structures bound to the actin filament (F-actin) has been a major impediment to understanding formin function. Whereas formins are known for their ability to nucleate and elongate F-actin3-7, some formins can additionally depolymerize, sever or bundle F-actin. Two mammalian formins, inverted formin 2 (INF2) and diaphanous 1 (DIA1, encoded by DIAPH1), exemplify this diversity. INF2 shows potent severing activity but elongates weakly8-11 whereas DIA1 has potent elongation activity but does not sever4,8. Using cryo-electron microscopy (cryo-EM) we show five structural states of INF2 and two of DIA1 bound to the middle and barbed end of F-actin. INF2 and DIA1 bind differently to these sites, consistent with their distinct activities. The formin-homology 2 and Wiskott-Aldrich syndrome protein-homology 2 (FH2 and WH2, respectively) domains of INF2 are positioned to sever F-actin, whereas DIA1 appears unsuited for severing. These structures also show how profilin-actin is delivered to the fast-growing barbed end, and how this is followed by a transition of the incoming monomer into the F-actin conformation and the release of profilin. Combined, the seven structures presented here provide step-by-step visualization of the mechanisms of F-actin severing and elongation by formins.
Collapse
Affiliation(s)
- Nicholas J Palmer
- Department of Physiology and Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyle R Barrie
- Department of Physiology and Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Roberto Dominguez
- Department of Physiology and Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Shah R, Panagiotou TC, Cole GB, Moraes TF, Lavoie BD, McCulloch CA, Wilde A. The DIAPH3 linker specifies a β-actin network that maintains RhoA and Myosin-II at the cytokinetic furrow. Nat Commun 2024; 15:5250. [PMID: 38897998 PMCID: PMC11187180 DOI: 10.1038/s41467-024-49427-2] [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: 10/10/2023] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Cytokinesis is the final step of the cell division cycle that leads to the formation of two new cells. Successful cytokinesis requires significant remodelling of the plasma membrane by spatially distinct β- and γ-actin networks. These networks are generated by the formin family of actin nucleators, DIAPH3 and DIAPH1 respectively. Here we show that β- and γ-actin perform specialized and non-redundant roles in cytokinesis and cannot substitute for one another. Expression of hybrid DIAPH1 and DIAPH3 proteins with altered actin isoform specificity relocalized cytokinetic actin isoform networks within the cell, causing cytokinetic failure. Consistent with this we show that β-actin networks, but not γ-actin networks, are required for the maintenance of non-muscle myosin II and RhoA at the cytokinetic furrow. These data suggest that independent and spatially distinct actin isoform networks form scaffolds of unique interactors that facilitate localized biochemical activities to ensure successful cell division.
Collapse
Affiliation(s)
- Riya Shah
- Department of Biochemistry, University of Toronto, 661 University Ave, Toronto, ON, M5G 1M1, Canada
| | - Thomas C Panagiotou
- Department of Molecular Genetics, University of Toronto, 661 University Ave, Toronto, ON, M5G 1M1, Canada
| | - Gregory B Cole
- Department of Biochemistry, University of Toronto, 661 University Ave, Toronto, ON, M5G 1M1, Canada
| | - Trevor F Moraes
- Department of Biochemistry, University of Toronto, 661 University Ave, Toronto, ON, M5G 1M1, Canada
| | - Brigitte D Lavoie
- Department of Molecular Genetics, University of Toronto, 661 University Ave, Toronto, ON, M5G 1M1, Canada
| | | | - Andrew Wilde
- Department of Biochemistry, University of Toronto, 661 University Ave, Toronto, ON, M5G 1M1, Canada.
- Department of Molecular Genetics, University of Toronto, 661 University Ave, Toronto, ON, M5G 1M1, Canada.
| |
Collapse
|
6
|
Oosterheert W, Boiero Sanders M, Funk J, Prumbaum D, Raunser S, Bieling P. Molecular mechanism of actin filament elongation by formins. Science 2024; 384:eadn9560. [PMID: 38603491 DOI: 10.1126/science.adn9560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/05/2024] [Indexed: 04/13/2024]
Abstract
Formins control the assembly of actin filaments (F-actin) that drive cell morphogenesis and motility in eukaryotes. However, their molecular interaction with F-actin and their mechanism of action remain unclear. In this work, we present high-resolution cryo-electron microscopy structures of F-actin barbed ends bound by three distinct formins, revealing a common asymmetric formin conformation imposed by the filament. Formation of new intersubunit contacts during actin polymerization sterically displaces formin and triggers its translocation. This "undock-and-lock" mechanism explains how actin-filament growth is coordinated with formin movement. Filament elongation speeds are controlled by the positioning and stability of actin-formin interfaces, which distinguish fast and slow formins. Furthermore, we provide a structure of the actin-formin-profilin ring complex, which resolves how profilin is rapidly released from the barbed end during filament elongation.
Collapse
Affiliation(s)
- Wout Oosterheert
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Micaela Boiero Sanders
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Johanna Funk
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Daniel Prumbaum
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Peter Bieling
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| |
Collapse
|
7
|
Ali H, Malik MZ, Abu-Farha M, Abubaker J, Cherian P, Nizam R, Jacob S, Bahbahani Y, Naim M, Ahmad S, Al-Sayegh M, Thanaraj TA, Ong ACM, Harris PC, Al-Mulla F. Global analysis of urinary extracellular vesicle small RNAs in autosomal dominant polycystic kidney disease. J Gene Med 2024; 26:e3674. [PMID: 38404150 DOI: 10.1002/jgm.3674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/15/2024] [Accepted: 01/26/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is the most prevalent monogenic renal disease progressing to end-stage renal disease. There is a pressing need for the identification of early ADPKD biomarkers to enable timely intervention and the development of effective therapeutic approaches. Here, we profiled human urinary extracellular vesicles small RNAs by small RNA sequencing in patients with ADPKD and compared their differential expression considering healthy control individuals to identify dysregulated small RNAs and analyze downstream interaction to gain insight about molecular pathophysiology. METHODS This is a cross-sectional study where urine samples were collected from a total of 23 PKD1-ADPKD patients and 28 healthy individuals. Urinary extracellular vesicles were purified, and small RNA was isolated and sequenced. Differentially expressed Small RNA were identified and functional enrichment analysis of the critical miRNAs was performed to identify driver genes and affected pathways. RESULTS miR-320b, miR-320c, miR-146a-5p, miR-199b-3p, miR-671-5p, miR-1246, miR-8485, miR-3656, has_piR_020497, has_piR_020496 and has_piR_016271 were significantly upregulated in ADPKD patient urine extracellular vesicles and miRNA-29c was significantly downregulated. Five 'driver' target genes (FBRS, EDC3, FMNL3, CTNNBIP1 and KMT2A) were identified. CONCLUSIONS The findings of the present study make significant contributions to the understanding of ADPKD pathogenesis and to the identification of novel biomarkers and potential drug targets aimed at slowing disease progression in ADPKD.
Collapse
Affiliation(s)
- Hamad Ali
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center (HSC), Kuwait University, Jabriya, Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait
- Division of Nephrology, Mubarak Al-Kabeer Hospital, Ministry of Health, Jabriya, Kuwait
| | - Md Zubbair Malik
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Mohamed Abu-Farha
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Jehad Abubaker
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Preethi Cherian
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Rasheeba Nizam
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Sindhu Jacob
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Yousif Bahbahani
- Division of Nephrology, Mubarak Al-Kabeer Hospital, Ministry of Health, Jabriya, Kuwait
- Medical Division, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Medhat Naim
- Division of Nephrology, Mubarak Al-Kabeer Hospital, Ministry of Health, Jabriya, Kuwait
| | - Sajjad Ahmad
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Mohammad Al-Sayegh
- Biology Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | | | - Albert C M Ong
- Academic Nephrology Unit, Division of Clinical Medicine, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield, UK
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Fahd Al-Mulla
- Department of Translational Medicine, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| |
Collapse
|
8
|
Carman PJ, Rebowski G, Dominguez R, Alqassim SS. Single particle cryo-EM analysis of Rickettsia conorii Sca2 reveals a formin-like core. J Struct Biol 2023; 215:107960. [PMID: 37028467 PMCID: PMC10200769 DOI: 10.1016/j.jsb.2023.107960] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/16/2023] [Accepted: 04/01/2023] [Indexed: 04/08/2023]
Abstract
Spotted fever group Rickettsia undergo actin-based motility inside infected eukaryotic cells using Sca2 (surface cell antigen 2): an ∼ 1800 amino-acid monomeric autotransporter protein that is surface-attached to the bacterium and responsible for the assembly of long unbranched actin tails. Sca2 is the only known functional mimic of eukaryotic formins, yet it shares no sequence similarities to the latter. Using structural and biochemical approaches we have previously shown that Sca2 uses a novel actin assembly mechanism. The first ∼ 400 amino acids fold into helix-loop-helix repeats that form a crescent shape reminiscent of a formin FH2 monomer. Additionally, the N- and C- terminal halves of Sca2 display intramolecular interaction in an end-to-end manner and cooperate for actin assembly, mimicking a formin FH2 dimer. Towards a better structural understanding of this mechanism, we performed single-particle cryo-electron microscopy analysis of Sca2. While high-resolution structural details remain elusive, our model confirms the presence of a formin-like core: Sca2 indeed forms a doughnut shape, similar in diameter to a formin FH2 dimer and can accommodate two actin subunits. Extra electron density, thought to be contributed by the C-terminal repeat domain (CRD), covering one side is also observed. This structural analysis allows us to propose an updated model where nucleation proceeds by encircling two actin subunits, and elongation proceeds either by a formin-like mechanism that necessitates conformational changes in the observed Sca2 model, or via an insertional mechanism akin to that observed in the ParMRC system.
Collapse
Affiliation(s)
- Peter J Carman
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Grzegorz Rebowski
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Roberto Dominguez
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Saif S Alqassim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.
| |
Collapse
|
9
|
Ponlachantra K, Suginta W, Robinson RC, Kitaoku Y. AlphaFold2: A versatile tool to predict the appearance of functional adaptations in evolution: Profilin interactions in uncultured Asgard archaea: Profilin interactions in uncultured Asgard archaea. Bioessays 2023; 45:e2200119. [PMID: 36461738 DOI: 10.1002/bies.202200119] [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: 06/17/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 12/05/2022]
Abstract
The release of AlphaFold2 (AF2), a deep-learning-aided, open-source protein structure prediction program, from DeepMind, opened a new era of molecular biology. The astonishing improvement in the accuracy of the structure predictions provides the opportunity to characterize protein systems from uncultured Asgard archaea, key organisms in evolutionary biology. Despite the accumulation in metagenomics-derived Asgard archaea eukaryotic-like protein sequences, limited structural and biochemical information have restricted the insight in their potential functions. In this review, we focus on profilin, an actin-dynamics regulating protein, which in eukaryotes, modulates actin polymerization through (1) direct actin interaction, (2) polyproline binding, and (3) phospholipid binding. We assess AF2-predicted profilin structures in their potential abilities to participate in these activities. We demonstrate that AF2 is a powerful new tool for understanding the emergence of biological functional traits in evolution.
Collapse
Affiliation(s)
- Khongpon Ponlachantra
- School of Biomolecular Science and Engineering (BSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, Thailand
| | - Wipa Suginta
- School of Biomolecular Science and Engineering (BSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, Thailand
| | - Robert C Robinson
- School of Biomolecular Science and Engineering (BSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, Thailand.,Research Institute for Interdisciplinary Science (RIIS), Okayama University, Okayama, Japan
| | - Yoshihito Kitaoku
- Research Institute for Interdisciplinary Science (RIIS), Okayama University, Okayama, Japan
| |
Collapse
|
10
|
Maufront J, Guichard B, Cao LY, Cicco AD, Jégou A, Romet-Lemonne G, Bertin A. Direct observation of the conformational states of formin mDia1 at actin filament barbed ends and along the filament. Mol Biol Cell 2022; 34:ar2. [PMID: 36383775 PMCID: PMC9816646 DOI: 10.1091/mbc.e22-10-0472] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The fine regulation of actin polymerization is essential to control cell motility and architecture and to perform essential cellular functions. Formins are key regulators of actin filament assembly, known to processively elongate filament barbed ends and increase their polymerization rate. Different models have been extrapolated to describe the molecular mechanism governing the processive motion of formin FH2 domains at polymerizing barbed ends. Using negative stain electron microscopy, we directly identified for the first time two conformations of the mDia1 formin FH2 domains in interaction with the barbed ends of actin filaments. These conformations agree with the speculated open and closed conformations of the "stair-stepping" model. We observed the FH2 dimers to be in the open conformation for 79% of the data, interacting with the two terminal actin subunits of the barbed end while they interact with three actin subunits in the closed conformation. In addition, we identified and characterized the structure of single FH2 dimers encircling the core of actin filaments, and reveal their ability to spontaneously depart from barbed ends.
Collapse
Affiliation(s)
- Julien Maufront
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire Physico Chimie Curie,75005 Paris, France
| | - Bérengère Guichard
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Lu-Yan Cao
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Aurélie Di Cicco
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire Physico Chimie Curie,75005 Paris, France
| | - Antoine Jégou
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France,*Address correspondence to: Aurélie Bertin (); Guillaume Romet-Lemonne (); Antoine Jégou ()
| | - Guillaume Romet-Lemonne
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France,*Address correspondence to: Aurélie Bertin (); Guillaume Romet-Lemonne (); Antoine Jégou ()
| | - Aurélie Bertin
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR 168, Laboratoire Physico Chimie Curie,75005 Paris, France,*Address correspondence to: Aurélie Bertin (); Guillaume Romet-Lemonne (); Antoine Jégou ()
| |
Collapse
|
11
|
Lappalainen P, Kotila T, Jégou A, Romet-Lemonne G. Biochemical and mechanical regulation of actin dynamics. Nat Rev Mol Cell Biol 2022; 23:836-852. [PMID: 35918536 DOI: 10.1038/s41580-022-00508-4] [Citation(s) in RCA: 141] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 12/30/2022]
Abstract
Polymerization of actin filaments against membranes produces force for numerous cellular processes, such as migration, morphogenesis, endocytosis, phagocytosis and organelle dynamics. Consequently, aberrant actin cytoskeleton dynamics are linked to various diseases, including cancer, as well as immunological and neurological disorders. Understanding how actin filaments generate forces in cells, how force production is regulated by the interplay between actin-binding proteins and how the actin-regulatory machinery responds to mechanical load are at the heart of many cellular, developmental and pathological processes. During the past few years, our understanding of the mechanisms controlling actin filament assembly and disassembly has evolved substantially. It has also become evident that the activities of key actin-binding proteins are not regulated solely by biochemical signalling pathways, as mechanical regulation is critical for these proteins. Indeed, the architecture and dynamics of the actin cytoskeleton are directly tuned by mechanical load. Here we discuss the general mechanisms by which key actin regulators, often in synergy with each other, control actin filament assembly, disassembly, and monomer recycling. By using an updated view of actin dynamics as a framework, we discuss how the mechanics and geometry of actin networks control actin-binding proteins, and how this translates into force production in endocytosis and mesenchymal cell migration.
Collapse
Affiliation(s)
- Pekka Lappalainen
- Institute of Biotechnology and Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland.
| | - Tommi Kotila
- Institute of Biotechnology and Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Antoine Jégou
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | | |
Collapse
|
12
|
Schutt CE, Karlén M, Karlsson R. A structural model of the profilin-formin pacemaker system for actin filament elongation. Sci Rep 2022; 12:20515. [PMID: 36443454 PMCID: PMC9705415 DOI: 10.1038/s41598-022-25011-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
The formins constitute a large class of multi-domain polymerases that catalyze the localization and growth of unbranched actin filaments in cells from yeast to mammals. The conserved FH2 domains form dimers that bind actin at the barbed end of growing filaments and remain attached as new subunits are added. Profilin-actin is recruited and delivered to the barbed end by formin FH1 domains via the binding of profilin to interspersed tracts of poly-L-proline. We present a structural model showing that profilin-actin can bind the FH2 dimer at the barbed end stabilizing a state where profilin prevents its associated actin subunit from directly joining the barbed end. It is only with the dissociation of profilin from the polymerase that an actin subunit rotates and docks into its helical position, consistent with observations that under physiological conditions optimal elongation rates depend on the dissociation rate of profilin, independently of cellular concentrations of actin subunits.
Collapse
Affiliation(s)
- Clarence E. Schutt
- grid.16750.350000 0001 2097 5006Department of Chemistry, Princeton University, Princeton, NJ USA
| | | | - Roger Karlsson
- Department of Molecular Biosciences, WGI, Stockholm University, Stockholm, Sweden.
| |
Collapse
|
13
|
Orman M, Landis M, Oza A, Nambiar D, Gjeci J, Song K, Huang V, Klestzick A, Hachicho C, Liu SQ, Kamm JM, Bartolini F, Vadakkan JJ, Rojas CM, Vizcarra CL. Alterations to the broad-spectrum formin inhibitor SMIFH2 modulate potency but not specificity. Sci Rep 2022; 12:13520. [PMID: 35941181 PMCID: PMC9360399 DOI: 10.1038/s41598-022-17685-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 07/29/2022] [Indexed: 12/23/2022] Open
Abstract
SMIFH2 is a small molecule inhibitor of the formin family of cytoskeletal regulators that was originally identified in a screen for suppression of actin polymerization induced by the mouse formin Diaphanous 1 (mDia1). Despite widespread use of this compound, it is unknown whether SMIFH2 inhibits all human formins. Additionally, the nature of protein/inhibitor interactions remains elusive. We assayed SMIFH2 against human formins representing six of the seven mammalian classes and found inhibitory activity against all formins tested. We synthesized a panel of SMIFH2 derivatives and found that, while many alterations disrupt SMIFH2 activity, substitution of an electron-donating methoxy group in place of the bromine along with halogenation of the furan ring increases potency by approximately five-fold. Similar to SMIFH2, the active derivatives are also pan-inhibitors for the formins tested. This result suggests that while potency can be improved, the goal of distinguishing between highly conserved FH2 domains may not be achievable using the SMIFH2 scaffold.
Collapse
Affiliation(s)
- Marina Orman
- Department of Chemistry, Barnard College, New York, NY, USA
| | - Maya Landis
- Department of Chemistry, Barnard College, New York, NY, USA
| | - Aisha Oza
- Department of Chemistry, Barnard College, New York, NY, USA
| | | | - Joana Gjeci
- Department of Chemistry, Barnard College, New York, NY, USA
| | - Kristen Song
- Department of Chemistry, Barnard College, New York, NY, USA
| | - Vivian Huang
- Department of Chemistry, Barnard College, New York, NY, USA
| | | | - Carla Hachicho
- Department of Chemistry, Barnard College, New York, NY, USA
| | - Su Qing Liu
- Department of Chemistry, Barnard College, New York, NY, USA
| | - Judith M Kamm
- Department of Chemistry, Barnard College, New York, NY, USA
| | - Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | | | | | | |
Collapse
|
14
|
Billault-Chaumartin I, Michon L, Anderson CA, Yde SE, Suarez C, Iwaszkiewicz J, Zoete V, Kovar DR, Martin SG. Actin assembly requirements of the formin Fus1 to build the fusion focus. J Cell Sci 2022; 135:jcs260289. [PMID: 35673994 PMCID: PMC9377709 DOI: 10.1242/jcs.260289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 12/28/2022] Open
Abstract
In formin-family proteins, actin filament nucleation and elongation activities reside in the formin homology 1 (FH1) and FH2 domains, with reaction rates that vary by at least 20-fold between formins. Each cell expresses distinct formins that assemble one or several actin structures, raising the question of what confers each formin its specificity. Here, using the formin Fus1 in Schizosaccharomyces pombe, we systematically probed the importance of formin nucleation and elongation rates in vivo. Fus1 assembles the actin fusion focus, necessary for gamete fusion to form the zygote during sexual reproduction. By constructing chimeric formins with combinations of FH1 and FH2 domains previously characterized in vitro, we establish that changes in formin nucleation and elongation rates have direct consequences on fusion focus architecture, and that Fus1 native high nucleation and low elongation rates are optimal for fusion focus assembly. We further describe a point mutant in Fus1 FH2 that preserves native nucleation and elongation rates in vitro but alters function in vivo, indicating an additional FH2 domain property. Thus, rates of actin assembly are tailored for assembly of specific actin structures.
Collapse
Affiliation(s)
- Ingrid Billault-Chaumartin
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore building, CH-1015 Lausanne, Switzerland
| | - Laetitia Michon
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore building, CH-1015 Lausanne, Switzerland
| | - Caitlin A. Anderson
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah E. Yde
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Cristian Suarez
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Justyna Iwaszkiewicz
- Molecular Modeling Group, Swiss Institute of Bioinformatics, Amphipôle Building, CH-1015 Lausanne, Switzerland
| | - Vincent Zoete
- Molecular Modeling Group, Swiss Institute of Bioinformatics, Amphipôle Building, CH-1015 Lausanne, Switzerland
- Department of Oncology UNIL-CHUV, University of Lausanne, Ludwig Institute for Cancer Research, Route de la Corniche 9A, CH-1066 Epalinges, Switzerland
| | - David R. Kovar
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Sophie G. Martin
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore building, CH-1015 Lausanne, Switzerland
| |
Collapse
|
15
|
Das S, Zhang Z, Kalvakota S, Soto R, Phillips ML, Terman JR, Reisler E. Parallel actin monomers in the 8S complex of actin-INF2. J Biomol Struct Dyn 2022; 41:3295-3304. [PMID: 35343388 PMCID: PMC10368088 DOI: 10.1080/07391102.2022.2050947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Polymerization and depolymerization of actin play an essential role in eukaryotic cells. Actin exists in cells in both monomeric (G-actin) and filamentous (polymer, F-actin) forms. Actin binding proteins (ABPs) facilitate the transition between these two states, and their interactions with these two states of actin are critical for actin-based cellular processes. Rapid depolymerization of actin is assisted in the brain and/or other cells by its oxidation by the enzyme Mical (yielding Mox-actin), and/or by the binding of Inverted Formin 2 (INF2) - which can also accelerate filaments formation. At their stoichiometric molar ratio INF2 and actin yield the 8S complex (consisting of 4 actin monomers: 2 INF2 dimer molecules). Using biochemical and biophysical methods, we investigate the structural arrangement of actin in the 8S particles and the interaction of INF2 with actin and Mox-actin. To that end, we show 2 D class averages of 8S particles obtained by negative staining electron microscopy. We also show that: (i) 8S particles can seed rapid actin assembly; (ii) Mox-actin and INF2 form 8S particles at proteins ratios similar to those of unoxidized actin; (iii) chemical crosslinkings suggest that actin monomers are in a parallel orientation in the 8S particles of both actin and Mox-actin; and (iv) INF2 accelerates the disassembly of Mox-F-actin. Our results provide better understanding of actin's arrangement in the 8S particles formed during actin depolymerization and in the early polymerization stages of both actin and Mox-actin.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sanchaita Das
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Zixin Zhang
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Saichandra Kalvakota
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Raul Soto
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Martin L Phillips
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Jonathan R Terman
- Departments of Neuroscience and Pharmacology, The University of Texas of Southwestern Medical Center, Dallas, TX, USA
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA.,Molecular Biology Institute, University of California, Los Angeles, CA, USA
| |
Collapse
|
16
|
Boiero Sanders M, Toret CP, Guillotin A, Antkowiak A, Vannier T, Robinson RC, Michelot A. Specialization of actin isoforms derived from the loss of key interactions with regulatory factors. EMBO J 2022; 41:e107982. [PMID: 35178724 PMCID: PMC8886540 DOI: 10.15252/embj.2021107982] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 11/09/2022] Open
Abstract
A paradox of eukaryotic cells is that while some species assemble a complex actin cytoskeleton from a single ortholog, other species utilize a greater diversity of actin isoforms. The physiological consequences of using different actin isoforms, and the molecular mechanisms by which highly conserved actin isoforms are segregated into distinct networks, are poorly known. Here, we sought to understand how a simple biological system, composed of a unique actin and a limited set of actin‐binding proteins, reacts to a switch to heterologous actin expression. Using yeast as a model system and biomimetic assays, we show that such perturbation causes drastic reorganization of the actin cytoskeleton. Our results indicate that defective interaction of a heterologous actin for important regulators of actin assembly limits certain actin assembly pathways while reinforcing others. Expression of two heterologous actin variants, each specialized in assembling a different network, rescues cytoskeletal organization and confers resistance to external perturbation. Hence, while species using a unique actin have homeostatic actin networks, actin assembly pathways in species using several actin isoforms may act more independently.
Collapse
Affiliation(s)
| | - Christopher P Toret
- CNRS, IBDM, Turing Centre for Living Systems, Aix Marseille Univ, Marseille, France
| | - Audrey Guillotin
- CNRS, IBDM, Turing Centre for Living Systems, Aix Marseille Univ, Marseille, France
| | - Adrien Antkowiak
- CNRS, IBDM, Turing Centre for Living Systems, Aix Marseille Univ, Marseille, France
| | - Thomas Vannier
- CNRS, IBDM, Turing Centre for Living Systems, Aix Marseille Univ, Marseille, France
| | - Robert C Robinson
- Research Institute for Interdisciplinary Science (RIIS), Okayama University, Okayama, Japan.,School of Biomolecular Science and Engineering (BSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, Thailand
| | - Alphée Michelot
- CNRS, IBDM, Turing Centre for Living Systems, Aix Marseille Univ, Marseille, France
| |
Collapse
|
17
|
Mehidi A, Kage F, Karatas Z, Cercy M, Schaks M, Polesskaya A, Sainlos M, Gautreau AM, Rossier O, Rottner K, Giannone G. Forces generated by lamellipodial actin filament elongation regulate the WAVE complex during cell migration. Nat Cell Biol 2021; 23:1148-1162. [PMID: 34737443 DOI: 10.1038/s41556-021-00786-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Actin filaments generate mechanical forces that drive membrane movements during trafficking, endocytosis and cell migration. Reciprocally, adaptations of actin networks to forces regulate their assembly and architecture. Yet, a demonstration of forces acting on actin regulators at actin assembly sites in cells is missing. Here we show that local forces arising from actin filament elongation mechanically control WAVE regulatory complex (WRC) dynamics and function, that is, Arp2/3 complex activation in the lamellipodium. Single-protein tracking revealed WRC lateral movements along the lamellipodium tip, driven by elongation of actin filaments and correlating with WRC turnover. The use of optical tweezers to mechanically manipulate functional WRC showed that piconewton forces, as generated by single-filament elongation, dissociated WRC from the lamellipodium tip. WRC activation correlated with its trapping, dwell time and the binding strength at the lamellipodium tip. WRC crosslinking, hindering its mechanical dissociation, increased WRC dwell time and Arp2/3-dependent membrane protrusion. Thus, forces generated by individual actin filaments on their regulators can mechanically tune their turnover and hence activity during cell migration.
Collapse
Affiliation(s)
- Amine Mehidi
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Frieda Kage
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Zeynep Karatas
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Maureen Cercy
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Matthias Schaks
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Anna Polesskaya
- CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Matthieu Sainlos
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Alexis M Gautreau
- CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Olivier Rossier
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Grégory Giannone
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France.
| |
Collapse
|
18
|
Zweifel ME, Sherer LA, Mahanta B, Courtemanche N. Nucleation limits the lengths of actin filaments assembled by formin. Biophys J 2021; 120:4442-4456. [PMID: 34506773 DOI: 10.1016/j.bpj.2021.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/06/2021] [Accepted: 09/02/2021] [Indexed: 10/24/2022] Open
Abstract
Formins stimulate actin polymerization by promoting both filament nucleation and elongation. Because nucleation and elongation draw upon a common pool of actin monomers, the rate at which each reaction proceeds influences the other. This interdependent mechanism determines the number of filaments assembled over the course of a polymerization reaction, as well as their equilibrium lengths. In this study, we used kinetic modeling and in vitro polymerization reactions to dissect the contributions of filament nucleation and elongation to the process of formin-mediated actin assembly. We found that the rates of nucleation and elongation evolve over the course of a polymerization reaction. The period over which each process occurs is a key determinant of the total number of filaments that are assembled, as well as their average lengths at equilibrium. Inclusion of formin in polymerization reactions speeds filament nucleation, thus increasing the number and shortening the lengths of filaments that are assembled over the course of the reaction. Modulation of the elongation rate produces modest changes in the equilibrium lengths of formin-bound filaments. However, the dependence of filament length on the elongation rate is limited by the number of filament ends generated via formin's nucleation activity. Sustained elongation of small numbers of formin-bound filaments, therefore, requires inhibition of nucleation via monomer sequestration and a low concentration of activated formin. Our results underscore the mechanistic advantage for keeping formin's nucleation efficiency relatively low in cells, where unregulated actin assembly would produce deleterious effects on cytoskeletal dynamics. Under these conditions, differences in the elongation rates mediated by formin isoforms are most likely to impact the kinetics of actin assembly.
Collapse
Affiliation(s)
- Mark E Zweifel
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
| | - Laura A Sherer
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
| | - Biswaprakash Mahanta
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
| | - Naomi Courtemanche
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
19
|
Pan MH, Wan X, Wang HH, Pan ZN, Zhang Y, Sun SC. FMNL3 regulates FASCIN for actin-mediated spindle migration and cytokinesis in mouse oocytes†. Biol Reprod 2021; 102:1203-1212. [PMID: 32167535 DOI: 10.1093/biolre/ioaa033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/23/2020] [Accepted: 03/12/2020] [Indexed: 11/14/2022] Open
Abstract
Formin-like 3 (FMNL3) is a member of the formin-likes (FMNLs), which belong to the formin family. As an F-actin nucleator, FMNL3 is essential for several cellular functions, such as polarity control, invasion, and migration. However, the roles of FMNL3 during oocytes meiosis remain unclear. In this study, we investigated the functions of FMNL3 during mouse oocyte maturation. Our results showed that FMNL3 mainly concentrated in the oocyte cortex and spindle periphery. Depleting FMNL3 led to the failure of polar body extrusion, and we also found large polar bodies in the FMNL3-deleted oocytes, indicating the occurrence of symmetric meiotic division. There was no effect of FMNL3 on spindle organization; however, we observed spindle migration defects at late metaphase I, which might be due to the decreased cytoplasmic actin. Microinjecting Fmnl3-EGFP mRNA into Fmnl3-depleted oocytes significantly rescued these defects. In addition, the results of co-immunoprecipitation and the perturbation of protein expression experiments suggested that FMNL3 interacted with the actin-binding protein FASCIN for the regulation of actin filaments in oocytes. Thus, our results provide the evidence that FMNL3 regulates FASCIN for actin-mediated spindle migration and cytokinesis during mouse oocyte meiosis.
Collapse
Affiliation(s)
- Meng-Hao Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, China
| | - Xiang Wan
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, China
| | - Hong-Hui Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, China
| | - Zhen-Nan Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, China
| | - Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, China
| |
Collapse
|
20
|
Qianqian Z, Wei X, Chuang L, Zhenliang C, Qiaoli W, Mingzhi L, Longyan W, Rui B, Jianhui T, Junjie L, Shiqiao W. MicroRNAs are potential regulators of the timed artificial insemination effect in gilt endometrium. Anim Reprod Sci 2021; 233:106837. [PMID: 34517227 DOI: 10.1016/j.anireprosci.2021.106837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 11/20/2022]
Abstract
To determine effects of timed artificial insemination (TAI) hormonal treatments on reproductive performance of gilts/sows and explore molecular mechanisms, gilts (TAI: 90; Control:149; Total: 239) and sows (TAI: 370; Control: 492) were utilized. Results indicated the estrus/farrowing rate and number of piglets born alive and weaned in the TAI group were greater than in the control group for both gilts and sows. To explore the molecular mechanism for TAI hormonal effects, the small RNA of the gilt endometrium at 16 and 25 of gestation were sampled and sequenced to determine potential functions of microRNA (MiRNA); 358 known and 142 novel MiRNAs were detected. With comparison of TAI and control groups, there were 54 differentially abundant MiRNAs, and functional analysis results indicated "binding," "protein/ion binding," and "immune response" were mostly enriched. In addition, representative MiRNAs were selected based on criteria including being regulated on both day 16 and 25 of gestation (ssc-miR-10a-5p, ssc-miR-345-5p, ssc-miR-370) along with reproduction-related target genes (ssc-miR-424-5p, ssc-miR-142-5p). Furthermore, target genes of selected MiRNAs were screened, and functional enrichment of those genes also indicated that the "binding" and "immune response" were mainly enriched. Results from the present study confirmed TAI-hormonal treatments improved estrous/farrowing rate and number of piglets born alive/weaned of gilts/sows and that hormonal treatment regimens leading to behavioral estrus at timed artificial insemination in gilts results in microRNA patterns in the endometrium that are more supportive of pregnancy. Results contribute valuable information for future studies of effects of TAI hormonal treatments on pig reproductive performance.
Collapse
Affiliation(s)
- Zhao Qianqian
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Xia Wei
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China; College of Life Science and Technology, Southwest Minzu University, Chengdu 610041, China; Hebei Technology Innovation Center of Cattle and Sheep Embryo, Baoding 071000, China
| | - Liu Chuang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Cui Zhenliang
- Ningbo Sansheng Biotechnology Co., Ltd,Ningbo 315100, China
| | - Wei Qiaoli
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Liu Mingzhi
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Wang Longyan
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Bai Rui
- College of Life Science and Technology, Southwest Minzu University, Chengdu 610041, China
| | - Tian Jianhui
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Li Junjie
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China; Hebei Technology Innovation Center of Cattle and Sheep Embryo, Baoding 071000, China.
| | - Weng Shiqiao
- Ningbo Sansheng Biotechnology Co., Ltd,Ningbo 315100, China.
| |
Collapse
|
21
|
Krüger LK, Gélin M, Ji L, Kikuti C, Houdusse A, Théry M, Blanchoin L, Tran PT. Kinesin-6 Klp9 orchestrates spindle elongation by regulating microtubule sliding and growth. eLife 2021; 10:67489. [PMID: 34080538 PMCID: PMC8205488 DOI: 10.7554/elife.67489] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/02/2021] [Indexed: 11/13/2022] Open
Abstract
Mitotic spindle function depends on the precise regulation of microtubule dynamics and microtubule sliding. Throughout mitosis, both processes have to be orchestrated to establish and maintain spindle stability. We show that during anaphase B spindle elongation in Schizosaccharomyces pombe, the sliding motor Klp9 (kinesin-6) also promotes microtubule growth in vivo. In vitro, Klp9 can enhance and dampen microtubule growth, depending on the tubulin concentration. This indicates that the motor is able to promote and block tubulin subunit incorporation into the microtubule lattice in order to set a well-defined microtubule growth velocity. Moreover, Klp9 recruitment to spindle microtubules is dependent on its dephosphorylation mediated by XMAP215/Dis1, a microtubule polymerase, creating a link between the regulation of spindle length and spindle elongation velocity. Collectively, we unravel the mechanism of anaphase B, from Klp9 recruitment to the motors dual-function in regulating microtubule sliding and microtubule growth, allowing an inherent coordination of both processes.
Collapse
Affiliation(s)
- Lara Katharina Krüger
- Institut Curie, PSL Research University, Sorbonne Université CNRS, UMR 144, Paris, France
| | - Matthieu Gélin
- Institut de Recherche Saint Louis,U976 Human Immunology Pathophysiology Immunotherapy (HIPI), CytoMorpho Lab, University of Paris, INSERM, CEA, Paris, France
| | - Liang Ji
- Institut Curie, PSL Research University, Sorbonne Université CNRS, UMR 144, Paris, France
| | - Carlos Kikuti
- Institut Curie, PSL Research University, Sorbonne Université CNRS, UMR 144, Paris, France
| | - Anne Houdusse
- Institut Curie, PSL Research University, Sorbonne Université CNRS, UMR 144, Paris, France
| | - Manuel Théry
- Institut de Recherche Saint Louis,U976 Human Immunology Pathophysiology Immunotherapy (HIPI), CytoMorpho Lab, University of Paris, INSERM, CEA, Paris, France.,Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, CytoMorpho Lab, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, Paris, France
| | - Laurent Blanchoin
- Institut de Recherche Saint Louis,U976 Human Immunology Pathophysiology Immunotherapy (HIPI), CytoMorpho Lab, University of Paris, INSERM, CEA, Paris, France.,Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, CytoMorpho Lab, University of Grenoble-Alpes, CEA, CNRS, INRA, Grenoble, Paris, France
| | - Phong T Tran
- Institut Curie, PSL Research University, Sorbonne Université CNRS, UMR 144, Paris, France.,Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
22
|
Inhibition of polar actin assembly by astral microtubules is required for cytokinesis. Nat Commun 2021; 12:2409. [PMID: 33893302 PMCID: PMC8065111 DOI: 10.1038/s41467-021-22677-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 03/19/2021] [Indexed: 02/07/2023] Open
Abstract
During cytokinesis, the actin cytoskeleton is partitioned into two spatially distinct actin isoform specific networks: a β-actin network that generates the equatorial contractile ring, and a γ-actin network that localizes to the cell cortex. Here we demonstrate that the opposing regulation of the β- and γ-actin networks is required for successful cytokinesis. While activation of the formin DIAPH3 at the cytokinetic furrow underlies β-actin filament production, we show that the γ-actin network is specifically depleted at the cell poles through the localized deactivation of the formin DIAPH1. During anaphase, CLIP170 is delivered by astral microtubules and displaces IQGAP1 from DIAPH1, leading to formin autoinhibition, a decrease in cortical stiffness and localized membrane blebbing. The contemporaneous production of a β-actin contractile ring at the cell equator and loss of γ-actin from the poles is required to generate a stable cytokinetic furrow and for the completion of cell division. During cell division, the actin cytoskeletal network at both the equatorial contractile ring and cell cortex are known to play a role, but the regulation of γ-actin during cytokinesis is less well understood. Here, the authors show that recruitment of β-actin to the contractile ring and loss of γ-actin from the cell poles is required for completion of cell division.
Collapse
|
23
|
A M, Latario CJ, Pickrell LE, Higgs HN. Lysine acetylation of cytoskeletal proteins: Emergence of an actin code. J Biophys Biochem Cytol 2020; 219:211455. [PMID: 33044556 PMCID: PMC7555357 DOI: 10.1083/jcb.202006151] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/26/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
Reversible lysine acetylation of nuclear proteins such as histones is a long-established important regulatory mechanism for chromatin remodeling and transcription. In the cytoplasm, acetylation of a number of cytoskeletal proteins, including tubulin, cortactin, and the formin mDia2, regulates both cytoskeletal assembly and stability. More recently, acetylation of actin itself was revealed to regulate cytoplasmic actin polymerization through the formin INF2, with downstream effects on ER-to-mitochondrial calcium transfer, mitochondrial fission, and vesicle transport. This finding raises the possibility that actin acetylation, along with other post-translational modifications to actin, might constitute an "actin code," similar to the "histone code" or "tubulin code," controlling functional shifts to these central cellular proteins. Given the multiple roles of actin in nuclear functions, its modifications might also have important roles in gene expression.
Collapse
|
24
|
Chakraborty S, Jasnin M, Baumeister W. Three-dimensional organization of the cytoskeleton: A cryo-electron tomography perspective. Protein Sci 2020; 29:1302-1320. [PMID: 32216120 PMCID: PMC7255506 DOI: 10.1002/pro.3858] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 01/01/2023]
Abstract
Traditionally, structures of cytoskeletal components have been studied ex situ, that is, with biochemically purified materials. There are compelling reasons to develop approaches to study them in situ in their native functional context. In recent years, cryo-electron tomography emerged as a powerful method for visualizing the molecular organization of unperturbed cellular landscapes with the potential to attain near-atomic resolution. Here, we review recent works on the cytoskeleton using cryo-electron tomography, demonstrating the power of in situ studies. We also highlight the potential of this method in addressing important questions pertinent to the field of cytoskeletal biomechanics.
Collapse
Affiliation(s)
- Saikat Chakraborty
- Department of Molecular Structural BiologyMax Planck Institute of BiochemistryMartinsriedGermany
| | - Marion Jasnin
- Department of Molecular Structural BiologyMax Planck Institute of BiochemistryMartinsriedGermany
| | - Wolfgang Baumeister
- Department of Molecular Structural BiologyMax Planck Institute of BiochemistryMartinsriedGermany
| |
Collapse
|
25
|
Merino F, Pospich S, Raunser S. Towards a structural understanding of the remodeling of the actin cytoskeleton. Semin Cell Dev Biol 2019; 102:51-64. [PMID: 31836290 PMCID: PMC7221352 DOI: 10.1016/j.semcdb.2019.11.018] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/03/2022]
Abstract
Actin filaments (F-actin) are a key component of eukaryotic cells. Whether serving as a scaffold for myosin or using their polymerization to push onto cellular components, their function is always related to force generation. To control and fine-tune force production, cells have a large array of actin-binding proteins (ABPs) dedicated to control every aspect of actin polymerization, filament localization, and their overall mechanical properties. Although great advances have been made in our biochemical understanding of the remodeling of the actin cytoskeleton, the structural basis of this process is still being deciphered. In this review, we summarize our current understanding of this process. We outline how ABPs control the nucleation and disassembly, and how these processes are affected by the nucleotide state of the filaments. In addition, we highlight recent advances in the understanding of actomyosin force generation, and describe recent advances brought forward by the developments of electron cryomicroscopy.
Collapse
Affiliation(s)
- Felipe Merino
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Sabrina Pospich
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| |
Collapse
|
26
|
de Macêdo Mendes C, Teixeira DG, Lima JPMS, Lanza DCF. Characterization of putative proteins encoded by variable ORFs in white spot syndrome virus genome. BMC STRUCTURAL BIOLOGY 2019; 19:8. [PMID: 30999895 PMCID: PMC6474068 DOI: 10.1186/s12900-019-0106-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/28/2019] [Indexed: 01/07/2023]
Abstract
Background White Spot Syndrome Virus (WSSV) is an enveloped double-stranded DNA virus which causes mortality of several species of shrimp, being considered one of the main pathogens that affects global shrimp farming. This virus presents a complex genome of ~ 300 kb and viral isolates that present genomes with great identity. Despite this conservation, some variable regions in the WSSV genome occur in coding regions, and these putative proteins may have some relationship with viral adaptation and virulence mechanisms. Until now, the functions of these proteins were little studied. In this work, sequences and putative proteins encoded by WSSV variable regions were characterized in silico. Results The in silico approach enabled determining the variability of some sequences, as well as the identification of some domains resembling the Formin homology 2, RNA recognition motif, Xeroderma pigmentosum group D repair helicase, Hemagglutinin and Ankyrin motif. The information obtained from the sequences and the analysis of secondary and tertiary structure models allow to infer that some of these proteins possibly have functions related to protein modulation/degradation, intracellular transport, recombination and endosome fusion events. Conclusions The bioinformatics approaches were efficient in generating three-dimensional models and to identify domains, thereby enabling to propose possible functions for the putative polypeptides produced by the ORFs wsv129, wsv178, wsv249, wsv463a, wsv477, wsv479, wsv492, and wsv497. Electronic supplementary material The online version of this article (10.1186/s12900-019-0106-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cayro de Macêdo Mendes
- Applied Molecular Biology Lab - LAPLIC, Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil.,Postgraduate Program in Bioinformatics, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Diego Gomes Teixeira
- Postgraduate Program in Biochemistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - João Paulo Matos Santos Lima
- Postgraduate Program in Bioinformatics, Federal University of Rio Grande do Norte, Natal, RN, Brazil.,Postgraduate Program in Biochemistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Daniel Carlos Ferreira Lanza
- Applied Molecular Biology Lab - LAPLIC, Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil. .,Postgraduate Program in Bioinformatics, Federal University of Rio Grande do Norte, Natal, RN, Brazil. .,Postgraduate Program in Biochemistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| |
Collapse
|
27
|
Dasbiswas K, Hu S, Schnorrer F, Safran SA, Bershadsky AD. Ordering of myosin II filaments driven by mechanical forces: experiments and theory. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0114. [PMID: 29632266 DOI: 10.1098/rstb.2017.0114] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2017] [Indexed: 12/27/2022] Open
Abstract
Myosin II filaments form ordered superstructures in both cross-striated muscle and non-muscle cells. In cross-striated muscle, myosin II (thick) filaments, actin (thin) filaments and elastic titin filaments comprise the stereotypical contractile units of muscles called sarcomeres. Linear chains of sarcomeres, called myofibrils, are aligned laterally in registry to form cross-striated muscle cells. The experimentally observed dependence of the registered organization of myofibrils on extracellular matrix elasticity has been proposed to arise from the interactions of sarcomeric contractile elements (considered as force dipoles) through the matrix. Non-muscle cells form small bipolar filaments built of less than 30 myosin II molecules. These filaments are associated in registry forming superstructures ('stacks') orthogonal to actin filament bundles. Formation of myosin II filament stacks requires the myosin II ATPase activity and function of the actin filament crosslinking, polymerizing and depolymerizing proteins. We propose that the myosin II filaments embedded into elastic, intervening actin network (IVN) function as force dipoles that interact attractively through the IVN. This is in analogy with the theoretical picture developed for myofibrils where the elastic medium is now the actin cytoskeleton itself. Myosin stack formation in non-muscle cells provides a novel mechanism for the self-organization of the actin cytoskeleton at the level of the entire cell.This article is part of the theme issue 'Self-organization in cell biology'.
Collapse
Affiliation(s)
- Kinjal Dasbiswas
- James Franck Institute, University of Chicago, Chicago, IL 60637, USA
| | - Shiqiong Hu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, 13288 Marseille, France
| | - Samuel A Safran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alexander D Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore .,Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
28
|
Abstract
Formin homology proteins (formins) are a highly conserved family of cytoskeletal remodeling proteins that are involved in a diverse array of cellular functions. Formins are best known for their ability to regulate actin dynamics, but the same functional domains also govern stability and organization of microtubules. It is thought that this dual activity allows them to coordinate the activity of these two major cytoskeletal networks and thereby influence cellular architecture. Golgi ribbon assembly is dependent upon cooperative interactions between actin filaments and cytoplasmic microtubules originating both at the Golgi itself and from the centrosome. Similarly, centrosome assembly, centriole duplication, and centrosome positioning are also reliant on a dialogue between both cytoskeletal networks. As presented in this chapter, a growing body of evidence suggests that multiple formin proteins play essential roles in these central cellular processes.
Collapse
Affiliation(s)
- John Copeland
- Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
29
|
Courtemanche N. Mechanisms of formin-mediated actin assembly and dynamics. Biophys Rev 2018; 10:1553-1569. [PMID: 30392063 DOI: 10.1007/s12551-018-0468-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/18/2018] [Indexed: 12/14/2022] Open
Abstract
Cellular viability requires tight regulation of actin cytoskeletal dynamics. Distinct families of nucleation-promoting factors enable the rapid assembly of filament nuclei that elongate and are incorporated into diverse and specialized actin-based structures. In addition to promoting filament nucleation, the formin family of proteins directs the elongation of unbranched actin filaments. Processive association of formins with growing filament ends is achieved through continuous barbed end binding of the highly conserved, dimeric formin homology (FH) 2 domain. In cooperation with the FH1 domain and C-terminal tail region, FH2 dimers mediate actin subunit addition at speeds that can dramatically exceed the rate of spontaneous assembly. Here, I review recent biophysical, structural, and computational studies that have provided insight into the mechanisms of formin-mediated actin assembly and dynamics.
Collapse
Affiliation(s)
- Naomi Courtemanche
- Department of Genetics, Cell and Developmental Biology, University of Minnesota, 420 Washington Ave SE, 6-130 MCB, Minneapolis, MN, 55455, USA.
| |
Collapse
|
30
|
Zeng YF, Xiao YS, Liu Y, Luo XJ, Wen LD, Liu Q, Chen M. Formin-like 3 regulates RhoC/FAK pathway and actin assembly to promote cell invasion in colorectal carcinoma. World J Gastroenterol 2018; 24:3884-3897. [PMID: 30228782 PMCID: PMC6141330 DOI: 10.3748/wjg.v24.i34.3884] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/16/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To clarify the underlying mechanism of formin-like 3 (FMNL3) in the promotion of colorectal carcinoma (CRC) cell invasion.
METHODS The in vitro biological function analyses of FMNL3 were performed by gain- and loss-of function approaches. Changes in the F-actin cytoskeleton were detected by the technologies of phalloidin-TRITC labeling and confocal microscopy. The signaling pathway mediated by FMNL3 was explored by western blot, gelatin zymograph assay, co-immunoprecipitation (co-IP), immunofluorescence co-localization, and glutathione S-transferase (GST) pull-down assay.
RESULTS The in vitro experimental results showed that FMNL3 significantly promoted the proliferation, invasion, and migration of CRC cells (P < 0.05 and P < 0.01). Moreover, FMNL3 regulated the remodeling of actin-based protrusions such as filopodia and lamellipodia in a RhoC-dependent manner. The western blot and gelatin zymograph assay results indicated that FMNL3 was involved in the RhoC/ focal adhesion kinase (FAK) pathway and acted as an effector of RhoC to activate the downstream signaling of p-FAK as well as p-MAPK and p-AKT. This resulted in the increased expression of matrix metalloproteinase 2 (MMP2), matrix metalloproteinase 9 (MMP9) and vascular endothelial growth factor (VEGF), and the subsequent promotion of CRC cell invasion. The results of TAE226, U0126 or Ly294002 treatment confirmed an essential role of FMNL3 in activation of the RhoC/FAK pathway and the subsequent promotion of CRC invasion. Co-IP, co-localization and GST pull-down assays showed the direct interaction of FMNL3 with RhoC in vivo and in vitro.
CONCLUSION FMNL3 regulates the RhoC/FAK signaling pathway and RhoC-dependent remodeling of actin-based protrusions to promote CRC invasion.
Collapse
Affiliation(s)
- Yuan-Feng Zeng
- Department of Pathology, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Yi-Sheng Xiao
- Teaching and Researching Section of Morphology, College of Basic Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Yong Liu
- Department of Pathology, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Xiao-Jiang Luo
- Department of General Surgery, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Li-Dan Wen
- Clinical Medical Sciences Institute, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Qian Liu
- Department of Pathology, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Min Chen
- Department of Pathology, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
31
|
Aydin F, Courtemanche N, Pollard TD, Voth GA. Gating mechanisms during actin filament elongation by formins. eLife 2018; 7:37342. [PMID: 30035712 PMCID: PMC6056239 DOI: 10.7554/elife.37342] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 07/01/2018] [Indexed: 02/03/2023] Open
Abstract
Formins play an important role in the polymerization of unbranched actin filaments, and particular formins slow elongation by 5–95%. We studied the interactions between actin and the FH2 domains of formins Cdc12, Bni1 and mDia1 to understand the factors underlying their different rates of polymerization. All-atom molecular dynamics simulations revealed two factors that influence actin filament elongation and correlate with the rates of elongation. First, FH2 domains can sterically block the addition of new actin subunits. Second, FH2 domains flatten the helical twist of the terminal actin subunits, making the end less favorable for subunit addition. Coarse-grained simulations over longer time scales support these conclusions. The simulations show that filaments spend time in states that either allow or block elongation. The rate of elongation is a time-average of the degree to which the formin compromises subunit addition rather than the formin-actin complex literally being in ‘open’ or ‘closed’ states.
Collapse
Affiliation(s)
- Fikret Aydin
- Department of Chemistry, The University of Chicago, Chicago, United States.,Institute for Biophysical Dynamics, The University of Chicago, Chicago, United States.,James Franck Institute, The University of Chicago, Chicago, United States
| | - Naomi Courtemanche
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, United States.,Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, United States
| | - Thomas D Pollard
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, United States.,Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, United States
| | - Gregory A Voth
- Department of Chemistry, The University of Chicago, Chicago, United States.,Institute for Biophysical Dynamics, The University of Chicago, Chicago, United States.,James Franck Institute, The University of Chicago, Chicago, United States
| |
Collapse
|
32
|
Modulation of alternative splicing induced by paclitaxel in human lung cancer. Cell Death Dis 2018; 9:491. [PMID: 29706628 PMCID: PMC5924756 DOI: 10.1038/s41419-018-0539-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/25/2018] [Accepted: 03/27/2018] [Indexed: 12/25/2022]
Abstract
Paclitaxel is utilized as the first-line chemotherapeutic regimen for the majority of advanced non-small-cell lung carcinoma. However, whether paclitaxel could suppress cancer progression through modulating RNA alternative splicing remains largely unknown. Here, we demonstrated the effects of paclitaxel on cell proliferation inhibition, cell cycle arrest, and apoptosis. Mechanistically, paclitaxel leads to transcriptional alteration of networks involved in DNA replication and repair, chromosome segregation, chromatin silencing at rDNA, and mitosis at the transcriptional level. Moreover, paclitaxel regulates a number of cancer-associated RNA alternative splicing events, including genes involved in cellular response to DNA damage stimulus, preassembly of GPI anchor in ER membrane, transcription, and DNA repair. In particular, paclitaxel modulates the splicing of ECT2, a key factor involved in the regulation of cytokinesis. Briefly, paclitaxel favors the production of ECT2-S, the short splicing isoforms of ECT2, thereby inhibiting cancer cell proliferation. Our study provides mechanistic insights of paclitaxel on RNA alternative splicing regulation, thus to offer a potential novel route for paclitaxel to inhibit cancer progression.
Collapse
|
33
|
Kage F, Steffen A, Ellinger A, Ranftler C, Gehre C, Brakebusch C, Pavelka M, Stradal T, Rottner K. FMNL2 and -3 regulate Golgi architecture and anterograde transport downstream of Cdc42. Sci Rep 2017; 7:9791. [PMID: 28852060 PMCID: PMC5575334 DOI: 10.1038/s41598-017-09952-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/26/2017] [Indexed: 01/08/2023] Open
Abstract
The Rho-family small GTPase Cdc42 localizes at plasma membrane and Golgi complex and aside from protrusion and migration operates in vesicle trafficking, endo- and exocytosis as well as establishment and/or maintenance of cell polarity. The formin family members FMNL2 and -3 are actin assembly factors established to regulate cell edge protrusion during migration and invasion. Here we report these formins to additionally accumulate and function at the Golgi apparatus. As opposed to lamellipodia, Golgi targeting of these proteins required both their N-terminal myristoylation and the interaction with Cdc42. Moreover, Golgi association of FMNL2 or -3 induced a phalloidin-detectable actin meshwork around the Golgi. Importantly, functional interference with FMNL2/3 formins by RNAi or CRISPR/Cas9-mediated gene deletion invariably induced Golgi fragmentation in different cell lines. Furthermore, absence of these proteins led to enlargement of endosomes as well as defective maturation and/or sorting into late endosomes and lysosomes. In line with Cdc42 - recently established to regulate anterograde transport through the Golgi by cargo sorting and carrier formation - FMNL2/3 depletion also affected anterograde trafficking of VSV-G from the Golgi to the plasma membrane. Our data thus link FMNL2/3 formins to actin assembly-dependent functions of Cdc42 in anterograde transport through the Golgi apparatus.
Collapse
Affiliation(s)
- Frieda Kage
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Anika Steffen
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Adolf Ellinger
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | - Carmen Ranftler
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | - Christian Gehre
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Cord Brakebusch
- Biomedical Institute, BRIC, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Margit Pavelka
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | - Theresia Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany. .,Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany.
| |
Collapse
|
34
|
Vig AT, Földi I, Szikora S, Migh E, Gombos R, Tóth MÁ, Huber T, Pintér R, Talián GC, Mihály J, Bugyi B. The activities of the C-terminal regions of the formin protein disheveled-associated activator of morphogenesis (DAAM) in actin dynamics. J Biol Chem 2017. [PMID: 28642367 DOI: 10.1074/jbc.m117.799247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Disheveled-associated activator of morphogenesis (DAAM) is a diaphanous-related formin protein essential for the regulation of actin cytoskeleton dynamics in diverse biological processes. The conserved formin homology 1 and 2 (FH1-FH2) domains of DAAM catalyze actin nucleation and processively mediate filament elongation. These activities are indirectly regulated by the N- and C-terminal regions flanking the FH1-FH2 domains. Recently, the C-terminal diaphanous-autoregulatory domain (DAD) and the C terminus (CT) of formins have also been shown to regulate actin assembly by directly interacting with actin. Here, to better understand the biological activities of DAAM, we studied the role of DAD-CT regions of Drosophila DAAM in its interaction with actin with in vitro biochemical and in vivo genetic approaches. We found that the DAD-CT region binds actin in vitro and that its main actin-binding element is the CT region, which does not influence actin dynamics on its own. However, we also found that it can tune the nucleating activity and the filament end-interaction properties of DAAM in an FH2 domain-dependent manner. We also demonstrate that DAD-CT makes the FH2 domain more efficient in antagonizing with capping protein. Consistently, in vivo data suggested that the CT region contributes to DAAM-mediated filopodia formation and dynamics in primary neurons. In conclusion, our results demonstrate that the CT region of DAAM plays an important role in actin assembly regulation in a biological context.
Collapse
Affiliation(s)
- Andrea Teréz Vig
- From the Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, Pécs H-7624
| | - István Földi
- the Biological Research Centre, Institute of Genetics, MTA-SZBK NAP B Axon Growth and Regeneration Group, Hungarian Academy of Sciences, Temesvári krt. 62, Szeged H-6726, and
| | - Szilárd Szikora
- the Biological Research Centre, Institute of Genetics, MTA-SZBK NAP B Axon Growth and Regeneration Group, Hungarian Academy of Sciences, Temesvári krt. 62, Szeged H-6726, and
| | - Ede Migh
- the Biological Research Centre, Institute of Genetics, MTA-SZBK NAP B Axon Growth and Regeneration Group, Hungarian Academy of Sciences, Temesvári krt. 62, Szeged H-6726, and
| | - Rita Gombos
- the Biological Research Centre, Institute of Genetics, MTA-SZBK NAP B Axon Growth and Regeneration Group, Hungarian Academy of Sciences, Temesvári krt. 62, Szeged H-6726, and
| | - Mónika Ágnes Tóth
- From the Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, Pécs H-7624
| | - Tamás Huber
- From the Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, Pécs H-7624
| | - Réka Pintér
- From the Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, Pécs H-7624
| | - Gábor Csaba Talián
- From the Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, Pécs H-7624
| | - József Mihály
- the Biological Research Centre, Institute of Genetics, MTA-SZBK NAP B Axon Growth and Regeneration Group, Hungarian Academy of Sciences, Temesvári krt. 62, Szeged H-6726, and
| | - Beáta Bugyi
- From the Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, Pécs H-7624, .,the Szentágothai Research Center, Ifjúság Str. 34, Pécs H-7624, Hungary
| |
Collapse
|
35
|
Thyssen GN, Fang DD, Turley RB, Florane CB, Li P, Mattison CP, Naoumkina M. A Gly65Val substitution in an actin, GhACT_LI1, disrupts cell polarity and F-actin organization resulting in dwarf, lintless cotton plants. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2017; 90:111-121. [PMID: 28078746 DOI: 10.1111/tpj.13477] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 12/16/2016] [Accepted: 01/03/2017] [Indexed: 06/06/2023]
Abstract
Actin polymerizes to form part of the cytoskeleton and organize polar growth in all eukaryotic cells. Species with numerous actin genes are especially useful for the dissection of actin molecular function due to redundancy and neofunctionalization. Here, we investigated the role of a cotton (Gossypium hirsutum) actin gene in the organization of actin filaments in lobed cotyledon pavement cells and the highly elongated single-celled trichomes that comprise cotton lint fibers. Using mapping-by-sequencing, virus-induced gene silencing, and molecular modeling, we identified the causative mutation of the dominant dwarf Ligon lintless Li1 short fiber mutant as a single Gly65Val amino acid substitution in a polymerization domain of an actin gene, GhACT_LI1 (Gh_D04G0865). We observed altered cell morphology and disrupted organization of F-actin in Li1 plant cells by confocal microscopy. Mutant leaf cells lacked interdigitation of lobes and F-actin did not uniformly decorate the nuclear envelope. While wild-type lint fiber trichome cells contained long longitudinal actin cables, the short Li1 fiber cells accumulated disoriented transverse cables. The polymerization-defective Gly65Val allele in Li1 plants likely disrupts processive elongation of F-actin, resulting in a disorganized cytoskeleton and reduced cell polarity, which likely accounts for the dominant gene action and diverse pleiotropic effects associated with the Li1 mutation. Lastly, we propose a model to account for these effects, and underscore the roles of actin organization in determining plant cell polarity, shape and plant growth.
Collapse
Affiliation(s)
- Gregory N Thyssen
- Cotton Fiber Bioscience Research Unit, United States Department of Agriculture (USDA), Agricultural Research Service (ARS), Southern Regional Research Center (SRRC), 1100 Robert E. Lee Blvd, New Orleans, LA, 70124, USA
- Cotton Chemistry and Utilization Research Unit, USDA-ARS-SRRC, 1100 Robert E. Lee Blvd, New Orleans, LA, 70124, USA
| | - David D Fang
- Cotton Fiber Bioscience Research Unit, United States Department of Agriculture (USDA), Agricultural Research Service (ARS), Southern Regional Research Center (SRRC), 1100 Robert E. Lee Blvd, New Orleans, LA, 70124, USA
| | - Rickie B Turley
- Crop Genetics Research Unit, USDA-ARS, 141 Experiment Station Road, Stoneville, MS, 38776, USA
| | - Christopher B Florane
- Cotton Fiber Bioscience Research Unit, United States Department of Agriculture (USDA), Agricultural Research Service (ARS), Southern Regional Research Center (SRRC), 1100 Robert E. Lee Blvd, New Orleans, LA, 70124, USA
| | - Ping Li
- Cotton Fiber Bioscience Research Unit, United States Department of Agriculture (USDA), Agricultural Research Service (ARS), Southern Regional Research Center (SRRC), 1100 Robert E. Lee Blvd, New Orleans, LA, 70124, USA
| | - Christopher P Mattison
- Food Processing and Sensory Quality Research Unit, USDA-ARS-SRRC, 1100 Robert E. Lee Blvd, New Orleans, LA, 70124, USA
| | - Marina Naoumkina
- Cotton Fiber Bioscience Research Unit, United States Department of Agriculture (USDA), Agricultural Research Service (ARS), Southern Regional Research Center (SRRC), 1100 Robert E. Lee Blvd, New Orleans, LA, 70124, USA
| |
Collapse
|
36
|
Li X, Mei Y, Yan B, Vitriol E, Huang S, Ji P, Qiu Y. Histone deacetylase 6 regulates cytokinesis and erythrocyte enucleation through deacetylation of formin protein mDia2. Haematologica 2017; 102:984-994. [PMID: 28255013 PMCID: PMC5451330 DOI: 10.3324/haematol.2016.161513] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/27/2017] [Indexed: 11/09/2022] Open
Abstract
The formin protein mDia2 plays a critical role in a number of cellular processes through its ability to promote nucleation and elongation of actin filaments. In erythroblasts, this includes control of cytokinesis and enucleation by regulating contractile actin ring formation. Here we report a novel mechanism of how mDia2 is regulated: through acetylation and deacetylation at lysine 970 in the formin homology 2 domain. Ectopic expression of an acetyl-mimic mDia2 mutant in mouse erythroblasts is sufficient to abolish contractile actin ring formation at the cleavage furrow and subsequent erythrocyte cytokinesis and enucleation. We also identified that class II histone deacetylase 6 deacetylates and subsequently activates mDia2. Knockdown or inhibition of histone deacetylase 6 impairs contractile actin ring formation, and expression of a non-acetyl-mimic mDia2 mutant restores the contractile actin ring and rescues the impairment of enucleation. In addition to revealing a new step in mDia2 regulation, this study may unveil a novel regulatory mechanism of formin-mediated actin assembly, since the K970 acetylation site is conserved among Dia proteins
Collapse
Affiliation(s)
- Xuehui Li
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yang Mei
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Bowen Yan
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Eric Vitriol
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Suming Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA.,Macau Institute for Applied Research in Medicine and Health, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau
| | - Peng Ji
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yi Qiu
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
37
|
Wu Y, Shen Z, Wang K, Ha Y, Lei H, Jia Y, Ding R, Wu D, Gan S, Li R, Luo B, Jiang H, Jie W. High FMNL3 expression promotes nasopharyngeal carcinoma cell metastasis: role in TGF-β1-induced epithelia-to-mesenchymal transition. Sci Rep 2017; 7:42507. [PMID: 28198387 PMCID: PMC5309845 DOI: 10.1038/srep42507] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 01/09/2017] [Indexed: 12/16/2022] Open
Abstract
Formin-like 3 (FMNL3) plays a crucial role in cytoskeletal mediation and is potentially a biomarker for cell migration; however, its role in cancer metastasis remains unknown. In this study, we found elevated FMNL3 protein expression in clinical nasopharyngeal carcinoma (NPC) tissues. FMNL3 expression positively correlated to the clinical stage, T (tumour), N (lymph node metastasis) and M (distant metastasis) classification of NPC patients. Moreover, FMNL3 positively correlated to Vimentin expression and negatively correlated to E-cadherin expression in clinical NPC samples. In vitro experiments showed that FMNL3 expression was inversely related to NPC cell differentiation status. Overexpression of FMNL3 led to epithelial-to-mesenchymal transition (EMT) in well differentiated CNE1 cells. TGF-β1-treated poorly differentiated CNE2 cells showed changes in EMT accompanied by enhanced FMNL3 expression and cell migration. On the contrary, knockdown of FMNL3 partially attenuated the TGF-β1-promoted CNE2 cell migration, together with associated changes in EMT markers. Finally, knockdown of FMNL3 also weakened EMT in tumours in xenographs. Our study indicates for the first time that TGF-β1/FMNL3 signalling may be a novel mechanism mediating EMT in NPC, which is closely associated with NPC metastasis.
Collapse
Affiliation(s)
- Yanxia Wu
- Department of Pathology, Guangdong Medical University, Zhanjiang 524023, China
| | - Zhihua Shen
- Department of Pathophysiology, Guangdong Medical University, Zhanjiang 524023, China
| | - Keke Wang
- Department of Pathology, Guangdong Medical University, Zhanjiang 524023, China
| | - Yanping Ha
- Department of Pathology, Guangdong Medical University, Zhanjiang 524023, China
| | - Hong Lei
- Department of Pathology, Guangdong Medical University, Zhanjiang 524023, China
| | - Yanan Jia
- Department of Pathophysiology, Guangdong Medical University, Zhanjiang 524023, China
| | - Ranran Ding
- Department of Pathology, Guangdong Medical University, Zhanjiang 524023, China.,Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dongmei Wu
- Department of Pathology, Guangdong Medical University, Zhanjiang 524023, China
| | - Siyuan Gan
- Department of Pathology, Guangdong Medical University, Zhanjiang 524023, China
| | - Rujia Li
- Department of Pathology, Guangdong Medical University, Zhanjiang 524023, China
| | - Botao Luo
- Department of Pathology, Guangdong Medical University, Zhanjiang 524023, China
| | - Hanguo Jiang
- Department of Pathology, Guangdong Medical University, Zhanjiang 524023, China
| | - Wei Jie
- Department of Pathology, Guangdong Medical University, Zhanjiang 524023, China
| |
Collapse
|
38
|
Lee CY, Lou J, Wen KK, McKane M, Eskin SG, Rubenstein PA, Chien S, Ono S, Zhu C, McIntire LV. Regulation of actin catch-slip bonds with a RhoA-formin module. Sci Rep 2016; 6:35058. [PMID: 27731359 PMCID: PMC5059732 DOI: 10.1038/srep35058] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 09/21/2016] [Indexed: 11/23/2022] Open
Abstract
The dynamic turnover of the actin cytoskeleton is regulated cooperatively by force and biochemical signaling. We previously demonstrated that actin depolymerization under force is governed by catch-slip bonds mediated by force-induced K113:E195 salt-bridges. Yet, the biochemical regulation as well as the functional significance of actin catch bonds has not been elucidated. Using AFM force-clamp experiments, we show that formin controlled by RhoA switches the actin catch-slip bonds to slip-only bonds. SMD simulations reveal that the force does not induce the K113:E195 interaction when formin binds to actin K118 and E117 residues located at the helical segment extending to K113. Actin catch-slip bonds are suppressed by single residue replacements K113E and E195K that interrupt the force-induced K113:E195 interaction; and this suppression is rescued by a K113E/E195K double mutant (E/K) restoring the interaction in the opposite orientation. These results support the biological significance of actin catch bonds, as they corroborate reported observations that RhoA and formin switch force-induced actin cytoskeleton alignment and that either K113E or E195K induces yeast cell growth defects rescued by E/K. Our study demonstrates how the mechano-regulation of actin dynamics is modulated by biochemical signaling molecules, and suggests that actin catch bonds may be important in cell functions.
Collapse
Affiliation(s)
- Cho-yin Lee
- Wallace H Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Taiwan
| | - Jizhong Lou
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Kuo-Kuang Wen
- Department of Biochemistry, University of Iowa, Iowa City, IA, USA
| | - Melissa McKane
- Department of Biochemistry, University of Iowa, Iowa City, IA, USA
| | - Suzanne G. Eskin
- Wallace H Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | | | - Shu Chien
- Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Shoichiro Ono
- Department of Pathology, Emory University, Atlanta, GA, USA
| | - Cheng Zhu
- Wallace H Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Larry V. McIntire
- Wallace H Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| |
Collapse
|
39
|
Jepsen L, Kruth KA, Rubenstein PA, Sept D. Two Deafness-Causing Actin Mutations (DFNA20/26) Have Allosteric Effects on the Actin Structure. Biophys J 2016; 111:323-332. [PMID: 27463135 PMCID: PMC4968419 DOI: 10.1016/j.bpj.2016.06.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/03/2016] [Accepted: 06/09/2016] [Indexed: 12/21/2022] Open
Abstract
Point mutations in γ-cytoplasmic actin have been shown to result in autosomal-dominant, nonsyndromic, early-onset deafness. Two mutations at the same site, K118M and K118N, provide a unique opportunity to compare the effects of two dissimilar amino acid substitutions that produce a similar phenotype in humans. K118 resides in a helix that runs from K113 to T126, and mutations that alter the position, dynamics, and/or biochemistry of this helix can result in a wide range of pathologies. Using a combination of computational and experimental studies, both employing yeast actin, we find that these mutations at K118 result in changes in the structure and dynamics of the DNase-I loop, alterations in the structure of the H73 loop as well as the side-chain orientations of W79 and W86, changes in nucleotide exchange rates, and significant shifts in the twist of the actin monomer. Interestingly, in the case of K118N, the twist of the monomer is nearly identical to that of the F-actin protomer, and in vitro polymerization assays show that this mutation results in faster polymerization. Taken together, these results indicate that mutations at this site give rise to a series of small changes that can be tolerated in vivo but result in misregulation of actin assembly and dynamics.
Collapse
Affiliation(s)
- Lauren Jepsen
- Bioinformatics Graduate Program, University of Michigan, Ann Arbor, Michigan; Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan; Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Karina A Kruth
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Peter A Rubenstein
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - David Sept
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan; Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
40
|
Shekhar S, Kerleau M, Kühn S, Pernier J, Romet-Lemonne G, Jégou A, Carlier MF. Formin and capping protein together embrace the actin filament in a ménage à trois. Nat Commun 2015; 6:8730. [PMID: 26564775 PMCID: PMC4660058 DOI: 10.1038/ncomms9730] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 09/24/2015] [Indexed: 11/09/2022] Open
Abstract
Proteins targeting actin filament barbed ends play a pivotal role in motile processes. While formins enhance filament assembly, capping protein (CP) blocks polymerization. On their own, they both bind barbed ends with high affinity and very slow dissociation. Their barbed-end binding is thought to be mutually exclusive. CP has recently been shown to be present in filopodia and controls their morphology and dynamics. Here we explore how CP and formins may functionally coregulate filament barbed-end assembly. We show, using kinetic analysis of individual filaments by microfluidics-assisted fluorescence microscopy, that CP and mDia1 formin are able to simultaneously bind barbed ends. This is further confirmed using single-molecule imaging. Their mutually weakened binding enables rapid displacement of one by the other. We show that formin FMNL2 behaves similarly, thus suggesting that this is a general property of formins. Implications in filopodia regulation and barbed-end structural regulation are discussed.
Collapse
Affiliation(s)
- Shashank Shekhar
- Cytoskeleton Dynamics and Cell Motility, Department of Biochemistry, Biophysics and Structural Biology, I2BC, CNRS, 91198 Gif-sur-Yvette, France
| | - Mikael Kerleau
- Cytoskeleton Dynamics and Cell Motility, Department of Biochemistry, Biophysics and Structural Biology, I2BC, CNRS, 91198 Gif-sur-Yvette, France
| | - Sonja Kühn
- Cytoskeleton Dynamics and Cell Motility, Department of Biochemistry, Biophysics and Structural Biology, I2BC, CNRS, 91198 Gif-sur-Yvette, France
| | - Julien Pernier
- Cytoskeleton Dynamics and Cell Motility, Department of Biochemistry, Biophysics and Structural Biology, I2BC, CNRS, 91198 Gif-sur-Yvette, France
| | - Guillaume Romet-Lemonne
- Cytoskeleton Dynamics and Cell Motility, Department of Biochemistry, Biophysics and Structural Biology, I2BC, CNRS, 91198 Gif-sur-Yvette, France
| | - Antoine Jégou
- Cytoskeleton Dynamics and Cell Motility, Department of Biochemistry, Biophysics and Structural Biology, I2BC, CNRS, 91198 Gif-sur-Yvette, France
| | - Marie-France Carlier
- Cytoskeleton Dynamics and Cell Motility, Department of Biochemistry, Biophysics and Structural Biology, I2BC, CNRS, 91198 Gif-sur-Yvette, France
| |
Collapse
|
41
|
Gunning PW, Ghoshdastider U, Whitaker S, Popp D, Robinson RC. The evolution of compositionally and functionally distinct actin filaments. J Cell Sci 2015; 128:2009-19. [DOI: 10.1242/jcs.165563] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
ABSTRACT
The actin filament is astonishingly well conserved across a diverse set of eukaryotic species. It has essentially remained unchanged in the billion years that separate yeast, Arabidopsis and man. In contrast, bacterial actin-like proteins have diverged to the extreme, and many of them are not readily identified from sequence-based homology searches. Here, we present phylogenetic analyses that point to an evolutionary drive to diversify actin filament composition across kingdoms. Bacteria use a one-filament-one-function system to create distinct filament systems within a single cell. In contrast, eukaryotic actin is a universal force provider in a wide range of processes. In plants, there has been an expansion of the number of closely related actin genes, whereas in fungi and metazoa diversification in tropomyosins has increased the compositional variety in actin filament systems. Both mechanisms dictate the subset of actin-binding proteins that interact with each filament type, leading to specialization in function. In this Hypothesis, we thus propose that different mechanisms were selected in bacteria, plants and metazoa, which achieved actin filament compositional variation leading to the expansion of their functional diversity.
Collapse
Affiliation(s)
- Peter W. Gunning
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Umesh Ghoshdastider
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore 138673
| | - Shane Whitaker
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - David Popp
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore 138673
| | - Robert C. Robinson
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore 138673
- Department of Biochemistry, National University of Singapore, 8 Medical Drive, Singapore 117597
| |
Collapse
|
42
|
The structure of FMNL2-Cdc42 yields insights into the mechanism of lamellipodia and filopodia formation. Nat Commun 2015; 6:7088. [PMID: 25963737 PMCID: PMC4432619 DOI: 10.1038/ncomms8088] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 03/31/2015] [Indexed: 11/08/2022] Open
Abstract
Formins are actin polymerization factors that elongate unbranched actin filaments at the barbed end. Rho family GTPases activate Diaphanous-related formins through the relief of an autoregulatory interaction. The crystal structures of the N-terminal domains of human FMNL1 and FMNL2 in complex with active Cdc42 show that Cdc42 mediates contacts with all five armadillo repeats of the formin with specific interactions formed by the Rho-GTPase insert helix. Mutation of three residues within Rac1 results in a gain-of-function mutation for FMNL2 binding and reconstitution of the Cdc42 phenotype in vivo. Dimerization of FMNL1 through a parallel coiled coil segment leads to formation of an umbrella-shaped structure that—together with Cdc42—spans more than 15 nm in diameter. The two interacting FMNL–Cdc42 heterodimers expose six membrane interaction motifs on a convex protein surface, the assembly of which may facilitate actin filament elongation at the leading edge of lamellipodia and filopodia. FMNL formins polymerize actin filaments to generate cellular protrusions such as lamellipodia and filopodia at the leading edge of a cell. Here the authors provide detailed mechanistic insights into the formation of actin-based protrusions through GTPase dependent activation and membrane localization of FMNL1 and FMNL2.
Collapse
|
43
|
Cdc42 mediates Bmp-induced sprouting angiogenesis through Fmnl3-driven assembly of endothelial filopodia in zebrafish. Dev Cell 2015; 32:109-22. [PMID: 25584797 DOI: 10.1016/j.devcel.2014.11.024] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 07/05/2014] [Accepted: 11/13/2014] [Indexed: 02/07/2023]
Abstract
During angiogenesis in vivo, endothelial cells (ECs) at the tips of vascular sprouts actively extend filopodia that are filled with bundles of linear actin filaments. To date, signaling pathways involved in the formation of endothelial filopodia have been studied using in-vitro-cultured ECs that behave differently from those in vivo. Herein, we have delineated a signaling pathway that governs the assembly of endothelial filopodia during angiogenic sprouting of the caudal vein plexus (CVP) in zebrafish. During CVP formation, bone morphogenetic protein induces the extension of endothelial filopodia and their migration via Arhgef9b-mediated activation of Cdc42. Active Cdc42 binds to and stimulates Formin-like 3, an actin-regulatory protein of the formin family, which, in turn, promotes the extension of endothelial filopodia to facilitate angiogenic sprouting of the CVP. Thus, this study has elucidated molecular mechanisms underlying the formation of endothelial filopodia and their role in angiogenesis in vivo.
Collapse
|
44
|
Vizcarra CL, Bor B, Quinlan ME. The role of formin tails in actin nucleation, processive elongation, and filament bundling. J Biol Chem 2014; 289:30602-30613. [PMID: 25246531 DOI: 10.1074/jbc.m114.588368] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Formins are multidomain proteins that assemble actin in a wide variety of biological processes. They both nucleate and remain processively associated with growing filaments, in some cases accelerating filament growth. The well conserved formin homology 1 and 2 domains were originally thought to be solely responsible for these activities. Recently a role in nucleation was identified for the Diaphanous autoinhibitory domain (DAD), which is C-terminal to the formin homology 2 domain. The C-terminal tail of the Drosophila formin Cappuccino (Capu) is conserved among FMN formins but distinct from other formins. It does not have a DAD domain. Nevertheless, we find that Capu-tail plays a role in filament nucleation similar to that described for mDia1 and other formins. Building on this, replacement of Capu-tail with DADs from other formins tunes nucleation activity. Capu-tail has low-affinity interactions with both actin monomers and filaments. Removal of the tail reduces actin filament binding and bundling. Furthermore, when the tail is removed, we find that processivity is compromised. Despite decreased processivity, the elongation rate of filaments is unchanged. Again, replacement of Capu-tail with DADs from other formins tunes the processive association with the barbed end, indicating that this is a general role for formin tails. Our data show a role for the Capu-tail domain in assembling the actin cytoskeleton, largely mediated by electrostatic interactions. Because of its multifunctionality, the formin tail is a candidate for regulation by other proteins during cytoskeletal rearrangements.
Collapse
Affiliation(s)
- Christina L Vizcarra
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California 90095
| | - Batbileg Bor
- Molecular Biology Interdepartmental Ph.D. Program, and University of California Los Angeles, Los Angeles, California 90095
| | - Margot E Quinlan
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California 90095; Molecular Biology Institute, University of California Los Angeles, Los Angeles, California 90095.
| |
Collapse
|
45
|
Sharma S, Grintsevich E, Woo J, Gurel PS, Higgs HN, Reisler E, Gimzewski JK. Nanostructured self-assembly of inverted formin 2 (INF2) and F-actin-INF2 complexes revealed by atomic force microscopy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:7533-7539. [PMID: 24915113 PMCID: PMC4082382 DOI: 10.1021/la501748x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 06/08/2014] [Indexed: 06/03/2023]
Abstract
Self-organization of cytoskeletal proteins such as actin and tubulin into filaments and microtubules is frequently assisted by the proteins binding to them. Formins are regulatory proteins that nucleate the formation of new filaments and are essential for a wide range of cellular functions. The vertebrate inverted formin 2 (INF2) has both actin filament nucleating and severing/depolymerizing activities connected to its ability to encircle actin filaments. Using atomic force microscopy, we report that a formin homology 2 (FH2) domain-containing construct of INF2 (INF2-FH1-FH2-C or INF2-FFC) self-assembles into nanoscale ringlike oligomeric structures in the absence of actin filaments, demonstrating an inherent ability to reorganize from a dimeric to an oligomeric state. A construct lacking the C-terminal region (INF2-FH1-FH2 or INF2-FF) also oligomerizes, confirming the dominant role of FH2-mediated interactions. Moreover, INF2-FFC domains were observed to organize into ringlike structures around single actin filaments. This is the first demonstration that formin FH2 domains can self-assemble into oligomers in the absence of filaments and has important implications for observing unaveraged decoration and/or remodeling of filaments by actin binding proteins.
Collapse
Affiliation(s)
- Shivani Sharma
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, California 90095, United States
- California
NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Elena
E. Grintsevich
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, California 90095, United States
| | - JungReem Woo
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, California 90095, United States
| | - Pinar S. Gurel
- Department
of Biochemistry, Geisel School of Medicine
at Dartmouth, Hanover, New Hampshire 03755, United States
| | - Henry N. Higgs
- Department
of Biochemistry, Geisel School of Medicine
at Dartmouth, Hanover, New Hampshire 03755, United States
| | - Emil Reisler
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, California 90095, United States
- Molecular
Biology Institute, University of California, Los Angeles, California 90095, United States
| | - James K. Gimzewski
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, California 90095, United States
- California
NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Jonsson
Comprehensive Cancer Center, University
of California, Los Angeles, California 90095, United States
- International
Center for Materials Nanoarchitectonics Satellite (MANA), National Institute for Materials Science (NIMS), Tsukuba, Japan
- Centre for
Nanoscience and Quantum Information, University
of Bristol, Bristol BS8 1TH, U.K.
| |
Collapse
|
46
|
Truong D, Copeland JW, Brumell JH. Bacterial subversion of host cytoskeletal machinery: hijacking formins and the Arp2/3 complex. Bioessays 2014; 36:687-96. [PMID: 24849003 DOI: 10.1002/bies.201400038] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The host actin nucleation machinery is subverted by many bacterial pathogens to facilitate their entry, motility, replication, and survival. The majority of research conducted in the past primarily focused on exploitation of a host actin nucleator, the Arp2/3 complex, by bacterial pathogens. Recently, new studies have begun to explore the role of formins, another family of host actin nucleators, in bacterial pathogenesis. This review provides an overview of recent advances in the study of the exploitation of the Arp2/3 complex and formins by bacterial pathogens. Secreted bacterial effector proteins seem to manipulate the regulation of these actin nucleators or functionally mimic them to drive bacterial entry, motility and survival within host cells. An enhanced understanding of how formins are exploited will provide us with greater insight into how a fundamental eurkaryotic cellular process is utilized by bacteria and will also advance our knowledge of host-pathogen interactions.
Collapse
Affiliation(s)
- Dorothy Truong
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | | | | |
Collapse
|
47
|
Rubenstein PA, Wen KK. Insights into the effects of disease-causing mutations in human actins. Cytoskeleton (Hoboken) 2014; 71:211-29. [PMID: 24574087 DOI: 10.1002/cm.21169] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 02/13/2013] [Accepted: 02/19/2014] [Indexed: 01/04/2023]
Abstract
Mutations in all six actins in humans have now been shown to cause diseases. However, a number of factors have made it difficult to gain insight into how the changes in actin functions brought about by these pathogenic mutations result in the disease phenotype. These include the presence of multiple actins in the same cell, limited accessibility to pure mutant material, and complexities associated with the structures and their component cells that manifest the diseases. To try to circumvent these difficulties, investigators have turned to the use of model systems. This review describes these various approaches, the initial results obtained using them, and the insight they have provided into allosteric mechanisms that govern actin function. Although results so far have not explained a particular disease phenotype at the molecular level, they have provided valuable insight into actin function at the mechanistic level which can be utilized in the future to delineate the molecular bases of these different actinopathies.
Collapse
Affiliation(s)
- Peter A Rubenstein
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | | |
Collapse
|
48
|
INF2-mediated severing through actin filament encirclement and disruption. Curr Biol 2014; 24:156-164. [PMID: 24412206 DOI: 10.1016/j.cub.2013.12.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/14/2013] [Accepted: 12/09/2013] [Indexed: 11/21/2022]
Abstract
BACKGROUND INF2 is a formin protein with the unique ability to accelerate both actin polymerization and depolymerization, the latter requiring filament severing. Mutations in INF2 lead to the kidney disease focal segmental glomerulosclerosis (FSGS) and the neurological disorder Charcot-Marie Tooth disease (CMTD). RESULTS Here, we compare the severing mechanism of INF2 with that of the well-studied severing protein cofilin. INF2, like cofilin, binds stoichiometrically to filament sides and severs in a manner that requires phosphate release from the filament. In contrast to cofilin, however, INF2 binds ADP and ADP-Pi filaments equally well. Furthermore, two-color total internal reflection fluorescence (TIRF) microscopy reveals that a low number of INF2 molecules, as few as a single INF2 dimer, are capable of severing, while measurable cofilin-mediated severing requires more extensive binding. Hence, INF2 is a more potent severing protein than cofilin. While a construct containing the FH1 and FH2 domains alone has some severing activity, addition of the C-terminal region increases severing potency by 40-fold, and we show that the WH2-resembling DAD motif is responsible for this increase. Helical 3D reconstruction from electron micrographs at 20 Å resolution provides a structure of filament-bound INF2, showing that the FH2 domain encircles the filament. CONCLUSIONS We propose a severing model in which FH2 binding and phosphate release causes local filament deformation, allowing the DAD to bind adjacent actin protomers, further disrupting filament structure.
Collapse
|
49
|
Zahm JA, Padrick SB, Chen Z, Pak CW, Yunus AA, Henry L, Tomchick DR, Chen Z, Rosen MK. The bacterial effector VopL organizes actin into filament-like structures. Cell 2013; 155:423-34. [PMID: 24120140 DOI: 10.1016/j.cell.2013.09.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 07/22/2013] [Accepted: 09/11/2013] [Indexed: 12/28/2022]
Abstract
VopL is an effector protein from Vibrio parahaemolyticus that nucleates actin filaments. VopL consists of a VopL C-terminal domain (VCD) and an array of three WASP homology 2 (WH2) motifs. Here, we report the crystal structure of the VCD dimer bound to actin. The VCD organizes three actin monomers in a spatial arrangement close to that found in the canonical actin filament. In this arrangement, WH2 motifs can be modeled into the binding site of each actin without steric clashes. The data suggest a mechanism of nucleation wherein VopL creates filament-like structures, organized by the VCD with monomers delivered by the WH2 array, that can template addition of new subunits. Similarities with Arp2/3 complex and formin proteins suggest that organization of monomers into filament-like structures is a general and central feature of actin nucleation.
Collapse
Affiliation(s)
- Jacob A Zahm
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Renault L, Deville C, van Heijenoort C. Structural features and interfacial properties of WH2, β-thymosin domains and other intrinsically disordered domains in the regulation of actin cytoskeleton dynamics. Cytoskeleton (Hoboken) 2013; 70:686-705. [DOI: 10.1002/cm.21140] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 08/28/2013] [Accepted: 09/01/2013] [Indexed: 01/12/2023]
Affiliation(s)
- Louis Renault
- Laboratoire d'Enzymologie et Biochimie Structurales; Centre de Recherche de Gif, CNRS; Gif-sur-Yvette France
| | - Célia Deville
- Laboratoire de Chimie et Biologie Structurales; Institut de Chimie des Substances Naturelles, Centre de Recherche de Gif, CNRS; Gif-sur-Yvette France
| | - Carine van Heijenoort
- Laboratoire de Chimie et Biologie Structurales; Institut de Chimie des Substances Naturelles, Centre de Recherche de Gif, CNRS; Gif-sur-Yvette France
| |
Collapse
|