1
|
Guo JY, Xu K, Wang XH, Li XM, Ku YP, Zeng L, Wan B, Yang GY, Wang J, Chu BB, Pan JJ, Hao WB. Host factor DIAPH1 regulates pseudorabivirus replication by modulating the dynamics of cytoskeleton. Int J Biol Macromol 2025; 298:140112. [PMID: 39842589 DOI: 10.1016/j.ijbiomac.2025.140112] [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: 11/07/2024] [Revised: 01/05/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
As obligate parasites, viruses exploit host cell organelles and molecular components to complete their life cycle. Among which, viruses firstly hijack the cytoskeleton of host cells to ensure their efficiently cell entry and replication. Although formin family members play a key role in both microfilament and microtubule cytoskeletal remodeling, few studies addressed the detailed function and mechanism of formins in the process of viral infection. Here, we showed that sus scrofa DIAPH1 was involved in the regulation of cytoskeletal dynamics during PRV replication. Firstly, we found that DIAPH1 showed significant changes in the expression level and intracellular localization during PRV infection of PK-15 cells. Next, inhibition of DIAPH1 by RNA interference or small molecular inhibitor SMIFH2 was found to diminish the outcome of PRV infection. Besides, DIAPH1 partially co-localized with actin and tubulin in PRV-infected cells. Cross-talk occurred between microfilaments and microfilaments, which also had an influence on the intracellular localization of DIAPH1. What's more, inhibition of DIAPH1 induced the reorganization of microfilament and the stability of microtubule. These results suggested that DIAPH1 regulated PRV infection by remodeling microfilament and microtubule cytoskeletal dynamics.
Collapse
Affiliation(s)
- Jie-Yuan Guo
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China
| | - Kun Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China
| | - Xiao-Han Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China
| | - Xin-Man Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China
| | - Yan-Pei Ku
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China
| | - Lei Zeng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China
| | - Bo Wan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China; Henan University of Animal Husbandry and Economy, Zhengzhou 450047, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China
| | - Jia-Jia Pan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China.
| | - Wen-Bo Hao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China.
| |
Collapse
|
2
|
McCaig CD. Electrical Forces Regulate Single-Cell Wound Healing. Rev Physiol Biochem Pharmacol 2025; 187:103-113. [PMID: 39838011 DOI: 10.1007/978-3-031-68827-0_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Damage to the cell membrane can be life threatening for single-celled organisms. Several mechanisms of single-cell wound healing occur and aspects of these are regulated by electrical forces.
Collapse
Affiliation(s)
- Colin D McCaig
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| |
Collapse
|
3
|
Nandi M, Shekhar S, Choubey S. A generalized theoretical framework to investigate multicomponent actin dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627743. [PMID: 39713386 PMCID: PMC11661301 DOI: 10.1101/2024.12.10.627743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The length of actin filaments is regulated by the combined action of hundreds of actin-binding proteins. While the roles of individual proteins are well understood, how they combine to regulate actin dynamics in vivo remains unclear. Recent advances in microscopy have enabled precise, high-throughput measurements of filament lengths over time. However, the absence of a unified theoretical framework has hindered a mechanistic understanding of the multicomponent regulation of actin dynamics. To address this, we propose a general kinetic model that captures the combined effects of multiple regulatory proteins on actin dynamics. We provide closed-form expressions for both time-dependent and steady-state moments of the filament length distribution. Our framework not only differentiates between various regulatory mechanisms but also serves as a powerful tool for interpreting current data and driving future experiments.
Collapse
Affiliation(s)
- Mintu Nandi
- Department of Chemistry, Indian Institute of Engineering Science and Technology, Shibpur, Howrah 711103, India
| | - Shashank Shekhar
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, Georgia 30322, USA
| | - Sandeep Choubey
- The Institute of Mathematical Sciences, CIT Campus, Taramani, Chennai 600113, India and Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| |
Collapse
|
4
|
Hollander S, Guo Y, Wolfenson H, Zaritsky A. Spatiotemporal analysis of F-actin polymerization with micropillar arrays reveals synchronization between adhesion sites. Mol Biol Cell 2024; 35:br23. [PMID: 39441710 PMCID: PMC11656478 DOI: 10.1091/mbc.e24-06-0276] [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: 06/25/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
We repurposed micropillar arrays to quantify spatiotemporal inter-adhesion communication. Following the observation that integrin adhesions formed around pillar tops we relied on the precise repetitive spatial control of the pillars to reliably monitor F-actin dynamics in mouse embryonic fibroblasts as a model for spatiotemporal adhesion-related intracellular signaling. Using correlation-based analyses, we revealed localized information flows propagating between adjacent pillars that were integrated over space and time to synchronize the adhesion dynamics within the entire cell. Probing the mechanical regulation, we discovered that stiffer pillars or partial actomyosin contractility inhibition enhances inter-adhesion F-actin synchronization, and that inhibition of Arp2/3, but not formin, reduces synchronization. Our results suggest that adhesions can communicate and highlight the potential of using micropillar arrays as a tool to measure spatiotemporal intracellular signaling.
Collapse
Affiliation(s)
- Sarit Hollander
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Yuanning Guo
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Assaf Zaritsky
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
5
|
Nama K, Su B, Marquez J, Khokha MK, Habas R. The dishevelled associated activator of morphogenesis protein 2 (Daam2) regulates neural tube closure. Dev Dyn 2024; 253:1130-1146. [PMID: 38877839 PMCID: PMC11611695 DOI: 10.1002/dvdy.720] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 12/05/2024] Open
Abstract
BACKGROUND The Wnt signaling pathway is highly conserved in metazoans and regulates a large array of cellular processes including motility, polarity and fate determination, and stem cell homeostasis. Modulation of the actin cytoskeleton via the non-canonical Wnt pathway regulate cell polarity and cell migration that are required for proper vertebrate gastrulation and subsequent neurulation. However, the mechanism(s) of how the non-canonical pathway mediates actin cytoskeleton modulation is not fully understood. RESULTS Herein, we characterize the role of the Formin-homology protein; dishevelled associated activator of morphogenesis 2 (Daam2) protein in the Wnt signaling pathway. Co-immunoprecipitation assays confirm the binding of Daam2 to dishevelled2 (Dvl2) as well as the domains within these proteins required for interaction; additionally, the interaction between Daam2 and Dvl2 was Wnt-regulated. Sub-cellular localization studies reveal Daam2 is cytoplasmic and regulates the cellular actin cytoskeleton by modulating actin filament formation. During Xenopus development, a knockdown or loss of Daam2 specifically produces neural tube closure defects indicative of a role in non-canonical signaling. Additionally, our studies did not identify any role for Daam2 in canonical Wnt signaling in mammalian culture cells or the Xenopus embryo. CONCLUSIONS Our studies together identify Daam2 as a component of the non-canonical Wnt pathway and Daam2 is a regulator of neural tube morphogenesis during vertebrate development.
Collapse
Affiliation(s)
- Kaushik Nama
- Department of Biology, Temple University, Philadelphia, PA 19122
| | - Baihao Su
- Department of Biology, Temple University, Philadelphia, PA 19122
| | - Jonathan Marquez
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Mustapha K Khokha
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Raymond Habas
- Department of Biology, Temple University, Philadelphia, PA 19122
| |
Collapse
|
6
|
Zhu L, Wang Y, Shan L, Xue Y, Schett G, Herrmann M, Liu L. Colchicine inhibits monosodium urate crystal-mediated inflammation by influencing F-actin formation. Biochim Biophys Acta Mol Basis Dis 2024; 1871:167602. [PMID: 39626855 DOI: 10.1016/j.bbadis.2024.167602] [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: 06/27/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/08/2024]
Abstract
OBJECTIVES To understand the mechanism by which colchicine inhibits the inflammatory properties of monosodium urate (MSU) crystal deposits and tophi. METHODS We investigated the effects of colchicine on the inflammatory properties of monosodium urate (MSU) crystal deposits in several models: (i) In vitro tophus formation by MSU and neutrophils; (ii) MSU-induced peritonitis model; (iii) Alpha-1-antitrypsin-induced peritoneal MSU flare model; (iv) MSU-induced arthritis model. We measured neutrophil numbers, NET formation, IL-1β production and F-actin generation by MSU crystals. In addition, we tested the effect of actin inhibitors SMIFH2, Cytochalasin B and Latrunculin B in the models. RESULTS Colchicine did not affect neutrophil numbers in all these models. However, colchicine was highly effective to inhibit NET formation, IL-1β production and F-actin generation indicating less pronounced tophus formation, lower inflammatory properties of tophi and reduced conversion from G-actin into F-actin, respectively. F-actin was shown to accumulate in tophi without presence of colchicine and being resistant to degradation by DNase I. Actin inhibitors SMIFH2 and Cytochalasin B significantly reduced IL-1β and neutrophil elastase levels and mitigated MSU-induced arthritis. CONCLUSION Colchicine effects on gout flares are not based on reducing neutrophil numbers but on changing the functional properties of tophi by reducing their DNase-resistant F-actin concentrations and thereby reducing the negative impact of NETs on IL-1β production and the pro-inflammatory state of tophi. Actin inhibitors may be interesting tools to convey anti-inflammatory properties and reduction of flares in gout patients.
Collapse
Affiliation(s)
- Lingjiang Zhu
- Department of Rheumatology, The Second affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang 310009, PR China
| | - Yuqi Wang
- Department of Rheumatology, The Second affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang 310009, PR China
| | - Lizhen Shan
- Department of Endocrinology, The Second affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang 310009, PR China
| | - Yu Xue
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 90154 Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 90154 Erlangen, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 90154 Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 90154 Erlangen, Germany
| | - Lei Liu
- Department of Rheumatology, The Second affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang 310009, PR China.
| |
Collapse
|
7
|
Ro N, Oh H, Ko HC, Yi J, Na YW, Haile M. Exploring Genomic Regions Associated with Fruit Traits in Pepper: Insights from Multiple GWAS Models. Int J Mol Sci 2024; 25:11836. [PMID: 39519386 PMCID: PMC11546569 DOI: 10.3390/ijms252111836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
This study utilized 303 pepper accessions from diverse Capsicum species to explore fruit traits, including length, width, wall thickness, and weight. Descriptive statistics revealed a mean fruit length of 66.19 mm, width of 23.48 mm, wall thickness of 1.89 mm, and weight of 15.29 g, with significant variability, particularly in fruit weight. Correlation analysis demonstrated strong positive relationships between fruit width, weight, and fruit wall thickness (r = 0.89 and r = 0.86, respectively), while fruit length showed weaker correlations with these traits. Analysis of fruit positions revealed that the majority of accessions had a pendent fruit position (156), followed by erect (85) and intermediate (8). In terms of fruit shape, triangular and narrow triangular shapes were the most common, observed in 102 and 98 accessions, respectively. Genome-wide association studies (GWAS) identified significant single nucleotide polymorphisms (SNPs) associated with fruit traits across four models (Blink, FarmCPU, MLM, MLMM). The number of significantly associated SNPs were as follows: fruit length (89), fruit width (55), fruit weight (63), fruit wall thickness (48), fruit shape (151), and fruit position (51). Several genes were also identified where the SNPs are located or adjacent to, providing candidate genes for further exploration of the genetic basis of fruit morphology. Notably, genes such as E3 ubiquitin-protein ligase RGLG1 (associated with fruit width), Homeobox-leucine zipper protein HDG11 (involved in fruit width), Auxin response factor 23 (linked to fruit shape), and ATP-dependent zinc metalloprotease FtsH (related to fruit weight) were identified. These findings enhance our understanding of the genetic basis of fruit morphology in Capsicum, offering valuable insights for breeding and agricultural practices.
Collapse
Affiliation(s)
- Nayoung Ro
- National Agrobiodiversity Center, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea; (H.O.); (H.-C.K.); (J.Y.); (Y.-W.N.)
| | | | | | | | | | - Mesfin Haile
- National Agrobiodiversity Center, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea; (H.O.); (H.-C.K.); (J.Y.); (Y.-W.N.)
| |
Collapse
|
8
|
Zeni C, Komiya Y, Habas R. Formin Binding Protein 1 (FNBP1) regulates non-canonical Wnt signaling and vertebrate gastrulation. Dev Biol 2024; 515:18-29. [PMID: 38945423 PMCID: PMC11317212 DOI: 10.1016/j.ydbio.2024.06.019] [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: 11/03/2023] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/02/2024]
Abstract
The Formin protein Daam1 is required for Wnt-induced cytoskeletal changes during gastrulation, though how it accomplishes this remains unresolved. Here we report the characterization of Formin Binding Protein 1 (FNBP1) as a binding partner of Daam1. The interaction of Daam1 with FNBP1 and its domains required for this interaction were delineated. Immunofluorescence studies showed FNBP1 co-localizes with Daam1, and is an integral component of the actin cytoskeletal complex that is responsive to Wnt stimulation. Specifically, FNBP1 can induce intracellular tubule-like structures and localize to focal adhesions suggesting a role for FNBP1 in cell migration. Functional FNBP1 studies in Xenopus embryos uncover a critical role for FNBP1 in regulating vertebrate gastrulation. Additionally, suboptimal doses of Daam1 and FNBP1 synergize to produce severe gastrulation defects, indicating FNBP1 and Daam1 may function within the same signaling pathway. These results together show FNBP1 is an integral component of Daam1-regulated non-canonical Wnt signaling required for vertebrate gastrulation.
Collapse
Affiliation(s)
- Courtney Zeni
- Department of Biology, Temple University, Philadelphia, PA, 19122, USA
| | - Yuko Komiya
- Department of Biology, Temple University, Philadelphia, PA, 19122, USA
| | - Raymond Habas
- Department of Biology, Temple University, Philadelphia, PA, 19122, USA.
| |
Collapse
|
9
|
Mezzacappa C, Komiya Y, Habas R. Reversion induced LIM domain protein (RIL) is a Daam1-interacting protein and regulator of the actin cytoskeleton during non-canonical Wnt signaling. Dev Biol 2024; 515:46-58. [PMID: 38968989 PMCID: PMC11321505 DOI: 10.1016/j.ydbio.2024.06.022] [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: 11/03/2023] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 07/07/2024]
Abstract
The Daam1 protein regulates Wnt-induced cytoskeletal changes during vertebrate gastrulation though its full mode of action and binding partners remain unresolved. Here we identify Reversion Induced LIM domain protein (RIL) as a new interacting protein of Daam1. Interaction studies uncover binding of RIL to the C-terminal actin-nucleating portion of Daam1 in a Wnt-responsive manner. Immunofluorescence studies showed subcellular localization of RIL to actin fibers and co-localization with Daam1 at the plasma membrane. RIL gain- and loss-of-function approaches in Xenopus produced severe gastrulation defects in injected embryos. Additionally, a simultaneous loss of Daam1 and RIL synergized to produce severe gastrulation defects indicating RIL and Daam1 may function in the same signaling pathway. RIL further synergizes with another novel Daam1-interacting protein, Formin Binding Protein 1 (FNBP1), to regulate gastrulation. Our studies altogether show RIL mediates Daam1-regulated non-canonical Wnt signaling that is required for vertebrate gastrulation.
Collapse
Affiliation(s)
| | - Yuko Komiya
- Department of Biology, Temple University, Philadelphia, PA, 19122, USA
| | - Raymond Habas
- Department of Biology, Temple University, Philadelphia, PA, 19122, USA.
| |
Collapse
|
10
|
Malik S, Ali SA, Mehdi AM, Raza A, Bashir S, Baig DN. A pilot study: Examining cytoskeleton gene expression profiles in Pakistani children with autism spectrum disorder. Int J Dev Neurosci 2024; 84:769-778. [PMID: 39285780 DOI: 10.1002/jdn.10372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Finding effective pharmacological interventions to address the complex array of neurodevelopmental disorders is currently an urgent imperative within the scientific community as these conditions present significant challenges for patients and their families, often impacting cognitive, emotional, and social development. In this study, we aimed to explore non-invasive method to diagnose autism spectrum disorders (ASD) within Pakistan children population and to identify clinical drugs for its treatment. AIMS The current report outlines a comprehensive bidirectional investigation showcasing the successful utilization of saliva samples to quantify the expression patterns of profilins (PFN1, 2, and 3); and ERM (ezrin, radixin, and moesin) proteins; and additionally moesin pseudogene 1 and moesin pseudogene 1 antisense (MSNP1AS). Subsequently, these expression profiles were employed to forecast interactions between drugs and genes in children diagnosed with ASD. METHODS This study sought to delve into the intricate gene expression profiles using qualitative polymerase chain reaction of profilin isoforms (PFN1, 2, and 3) and ERM genes extracted from saliva samples obtained from children diagnosed with ASD. Through this analysis, we aimed to elucidate potential molecular mechanisms underlying ASD pathogenesis, shedding light on novel biomarkers and therapeutic targets for this complex neurological condition. (n = 22). Subsequently, we implemented a diagnostic model utilizing sparse partial least squares discriminant analysis (sPLS-DA) to predict drugs against our genes of interest. Furthermore, connectivity maps were developed to illustrate the predicted associations of 24 drugs with the genes expression. RESULTS Our study results showed varied expression profile of cytoskeleton linked genes. Similarly, sPLS-DA model precisely predicted drug to genes response. Sixteen of the examined drugs had significant positive correlations with the expression of the targeted genes whereas eight of the predicted drugs had shown negative correlations. CONCLUSION Here we report the role of cytoskeleton linked genes (PFN and ERM) in co-relation to ASD. Furthermore, variable yet significant quantitative expression of these genes successfully predicted drug-gene interactions as shown with the help of connectivity maps that can be used to support the clinical use of these drugs to treat individuals with ASD in future studies.
Collapse
Affiliation(s)
- Sana Malik
- Kauser Abdullah Malik School of Life Sciences, Forman Christian College (A Chartered University) Lahore, Lahore, Pakistan
| | - Syed Aoun Ali
- Australian Institute of Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Ahmed Murtaza Mehdi
- Diamantina Institute, Faculty of Medicine, Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Amir Raza
- Department of Biotechnology, Knowledge Unit of Science, University of Management and Technology (Sialkot Campus), Sialkot, Pakistan
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Deeba Noreen Baig
- Kauser Abdullah Malik School of Life Sciences, Forman Christian College (A Chartered University) Lahore, Lahore, Pakistan
- University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
11
|
Wang W, Rui M. Advances in understanding the roles of actin scaffolding and membrane trafficking in dendrite development. J Genet Genomics 2024; 51:1151-1161. [PMID: 38925347 DOI: 10.1016/j.jgg.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
Dendritic morphology is typically highly branched, and the branching and synaptic abundance of dendrites can enhance the receptive range of neurons and the diversity of information received, thus providing the basis for information processing in the nervous system. Once dendritic development is aberrantly compromised or damaged, it may lead to abnormal connectivity of the neural network, affecting the function and stability of the nervous system and ultimately triggering a series of neurological disorders. Research on the regulation of dendritic developmental processes has flourished, and much progress is now being made in its regulatory mechanisms. Noteworthily, dendrites are characterized by an extremely complex dendritic arborization that cannot be attributed to individual protein functions alone, requiring a systematic analysis of the intrinsic and extrinsic signals and the coordinated roles among them. Actin cytoskeleton organization and membrane vesicle trafficking are required during dendrite development, with actin providing tracks for vesicles and vesicle trafficking in turn providing material for actin assembly. In this review, we focus on these two basic biological processes and discuss the molecular mechanisms and their synergistic effects underlying the morphogenesis of neuronal dendrites. We also offer insights and discuss strategies for the potential preventive and therapeutic treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Wanting Wang
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210031, China
| | - Menglong Rui
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210031, China.
| |
Collapse
|
12
|
Sigler AL, Thompson SB, Ellwood-Digel L, Kandasamy A, Michaels MJ, Thumkeo D, Narumiya S, Del Alamo JC, Jacobelli J. FMNL1 and mDia1 promote efficient T cell migration through complex environments via distinct mechanisms. Front Immunol 2024; 15:1467415. [PMID: 39430739 PMCID: PMC11486666 DOI: 10.3389/fimmu.2024.1467415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
Lymphocyte trafficking and migration through tissues is critical for adaptive immune function and, to perform their roles, T cells must be able to navigate through diverse tissue environments that present a range of mechanical challenges. T cells predominantly express two members of the formin family of actin effectors, Formin-like 1 (FMNL1) and mammalian diaphanous-related formin 1 (mDia1). While both FMNL1 and mDia1 have been studied individually, they have not been directly compared to determine functional differences in promoting T cell migration. Through in vivo analysis and the use of in vitro 2D and 3D model environments, we demonstrate that FMNL1 and mDia1 are both required for effective T cell migration, but they have different localization and roles in T cells, with specific environment-dependent functions. We found that mDia1 promotes general motility in 3D environments in conjunction with Myosin-II activity. We also show that, while mDia1 is almost entirely in the cytoplasmic compartment, a portion of FMNL1 physically associates with the nucleus. Furthermore, FMNL1 localizes to the rear of migrating T cells and contributes to efficient migration by promoting deformation of the rigid T cell nucleus in confined environments. Overall, our data indicates that while FMNL1 and mDia1 have similar mechanisms of actin polymerization, they have distinct roles in promoting T cell migration. This suggests that differential modulation of FMNL1 and mDia1 can be an attractive therapeutic route to fine-tune T cell migration behavior.
Collapse
Affiliation(s)
- Ashton L. Sigler
- Department of Immunology & Microbiology and Barbara Davis Research Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Scott B. Thompson
- Department of Immunology & Microbiology and Barbara Davis Research Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Logan Ellwood-Digel
- Department of Immunology & Microbiology and Barbara Davis Research Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Adithan Kandasamy
- Department of Mechanical Engineering, University of Washington, Seattle, WA, United States
| | - Mary J. Michaels
- Department of Immunology & Microbiology and Barbara Davis Research Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Dean Thumkeo
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Juan C. Del Alamo
- Department of Mechanical Engineering, University of Washington, Seattle, WA, United States
- Division of Cardiology, University of Washington, Seattle, WA, United States
| | - Jordan Jacobelli
- Department of Immunology & Microbiology and Barbara Davis Research Center, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
13
|
Pimm ML, Haarer BK, Nobles AD, Haney LM, Marcin AG, Alcaide Eligio M, Henty-Ridilla JL. Coordination of actin plus-end dynamics by IQGAP1, formin, and capping protein. J Cell Biol 2024; 223:e202305065. [PMID: 38787349 PMCID: PMC11117073 DOI: 10.1083/jcb.202305065] [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: 05/16/2023] [Revised: 04/01/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Cell processes require precise regulation of actin polymerization that is mediated by plus-end regulatory proteins. Detailed mechanisms that explain plus-end dynamics involve regulators with opposing roles, including factors that enhance assembly, e.g., the formin mDia1, and others that stop growth (capping protein, CP). We explore IQGAP1's roles in regulating actin filament plus-ends and the consequences of perturbing its activity in cells. We confirm that IQGAP1 pauses elongation and interacts with plus ends through two residues (C756 and C781). We directly visualize the dynamic interplay between IQGAP1 and mDia1, revealing that IQGAP1 displaces the formin to influence actin assembly. Using four-color TIRF, we show that IQGAP1's displacement activity extends to formin-CP "decision complexes," promoting end-binding protein turnover at plus-ends. Loss of IQGAP1 or its plus-end activities disrupts morphology and migration, emphasizing its essential role. These results reveal a new role for IQGAP1 in promoting protein turnover on filament ends and provide new insights into how plus-end actin assembly is regulated in cells.
Collapse
Affiliation(s)
- Morgan L. Pimm
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Brian K. Haarer
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Alexander D. Nobles
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Laura M. Haney
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Alexandra G. Marcin
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Marcela Alcaide Eligio
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jessica L. Henty-Ridilla
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
14
|
Shultz KD, Al Anbari YF, Wright NT. I told you to stop: obscurin's role in epithelial cell migration. Biochem Soc Trans 2024; 52:1947-1956. [PMID: 39051125 DOI: 10.1042/bst20240564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024]
Abstract
The giant cytoskeletal protein obscurin contains multiple cell signaling domains that influence cell migration. Here, we follow each of these pathways, examine how these pathways modulate epithelial cell migration, and discuss the cross-talk between these pathways. Specifically, obscurin uses its PH domain to inhibit phosphoinositide-3-kinase (PI3K)-dependent migration and its RhoGEF domain to activate RhoA and slow cell migration. While obscurin's effect on the PI3K pathway agrees with the literature, obscurin's effect on the RhoA pathway runs counter to most other RhoA effectors, whose activation tends to lead to enhanced motility. Obscurin also phosphorylates cadherins, and this may also influence cell motility. When taken together, obscurin's ability to modulate three independent cell migration pathways is likely why obscurin knockout cells experience enhanced epithelial to mesenchymal transition, and why obscurin is a frequently mutated gene in several types of cancer.
Collapse
Affiliation(s)
- Kamrin D Shultz
- Department of Chemistry and Biochemistry, James Madison University, 901 Carrier Dr., Harrisonburg, VA 22807, U.S.A
| | - Yasmin F Al Anbari
- Department of Chemistry and Biochemistry, James Madison University, 901 Carrier Dr., Harrisonburg, VA 22807, U.S.A
| | - Nathan T Wright
- Department of Chemistry and Biochemistry, James Madison University, 901 Carrier Dr., Harrisonburg, VA 22807, U.S.A
| |
Collapse
|
15
|
Schuldt C, Khudayberdiev S, Chandra BD, Linne U, Rust MB. Cyclase-associated protein (CAP) inhibits inverted formin 2 (INF2) to induce dendritic spine maturation. Cell Mol Life Sci 2024; 81:353. [PMID: 39154297 PMCID: PMC11335277 DOI: 10.1007/s00018-024-05393-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/09/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
The morphology of dendritic spines, the postsynaptic compartment of most excitatory synapses, decisively modulates the function of neuronal circuits as also evident from human brain disorders associated with altered spine density or morphology. Actin filaments (F-actin) form the backbone of spines, and a number of actin-binding proteins (ABP) have been implicated in shaping the cytoskeleton in mature spines. Instead, only little is known about the mechanisms that control the reorganization from unbranched F-actin of immature spines to the complex, highly branched cytoskeleton of mature spines. Here, we demonstrate impaired spine maturation in hippocampal neurons upon genetic inactivation of cyclase-associated protein 1 (CAP1) and CAP2, but not of CAP1 or CAP2 alone. We found a similar spine maturation defect upon overactivation of inverted formin 2 (INF2), a nucleator of unbranched F-actin with hitherto unknown synaptic function. While INF2 overactivation failed in altering spine density or morphology in CAP-deficient neurons, INF2 inactivation largely rescued their spine defects. From our data we conclude that CAPs inhibit INF2 to induce spine maturation. Since we previously showed that CAPs promote cofilin1-mediated cytoskeletal remodeling in mature spines, we identified them as a molecular switch that control transition from filopodia-like to mature spines.
Collapse
Affiliation(s)
- Cara Schuldt
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany
| | - Sharof Khudayberdiev
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany
| | - Ben-David Chandra
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany
| | - Uwe Linne
- Department of Chemistry, Philipps-University Marburg, 35032, Marburg, Germany
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032, Marburg, Germany.
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032, Marburg, Germany.
| |
Collapse
|
16
|
Azizoglu ZB, Babayeva R, Haskologlu ZS, Acar MB, Ayaz-Guner S, Okus FZ, Alsavaf MB, Can S, Basaran KE, Canatan MF, Ozcan A, Erkmen H, Leblebici CB, Yilmaz E, Karakukcu M, Kose M, Canoz O, Özen A, Karakoc-Aydiner E, Ceylaner S, Gümüş G, Per H, Gumus H, Canatan H, Ozcan S, Dogu F, Ikinciogullari A, Unal E, Baris S, Eken A. DIAPH1-Deficiency is Associated with Major T, NK and ILC Defects in Humans. J Clin Immunol 2024; 44:175. [PMID: 39120629 PMCID: PMC11315734 DOI: 10.1007/s10875-024-01777-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024]
Abstract
Loss of function mutations in Diaphanous related formin 1 (DIAPH1) are associated with seizures, cortical blindness, and microcephaly syndrome (SCBMS) and are recently linked to combined immunodeficiency. However, the extent of defects in T and innate lymphoid cells (ILCs) remain unexplored. Herein, we characterized the primary T, natural killer (NK) and helper ILCs of six patients carrying two novel loss of function mutation in DIAPH1 and Jurkat cells after DIAPH1 knockdown. Mutations were identified by whole exome sequencing. T-cell immunophenotyping, proliferation, migration, cytokine signaling, survival, and NK cell cytotoxicity were studied via flow cytometry-based assays, confocal microscopy, and real-time qPCR. CD4+ T cell proteome was analyzed by mass spectrometry. p.R351* and p.R322*variants led to a significant reduction in the DIAPH1 mRNA and protein levels. DIAPH1-deficient T cells showed proliferation, activation, as well as TCR-mediated signaling defects. DIAPH1-deficient PBMCs also displayed impaired transwell migration, defective STAT5 phosphorylation in response to IL-2, IL-7 and IL-15. In vitro generation/expansion of Treg cells from naïve T cells was significantly reduced. shRNA-mediated silencing of DIAPH1 in Jurkat cells reduced DIAPH1 protein level and inhibited T cell proliferation and IL-2/STAT5 axis. Additionally, NK cells from patients had diminished cytotoxic activity, function and IL-2/STAT5 axis. Lastly, DIAPH1-deficient patients' peripheral blood contained dramatically reduced numbers of all helper ILC subsets. DIAPH1 deficiency results in major functional defects in T, NK cells and helper ILCs underlining the critical role of formin DIAPH1 in the biology of those cell subsets.
Collapse
Affiliation(s)
- Zehra Busra Azizoglu
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye
| | - Royala Babayeva
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Zehra Sule Haskologlu
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | | | - Serife Ayaz-Guner
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Türkiye
| | - Fatma Zehra Okus
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
| | | | - Salim Can
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Kemal Erdem Basaran
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey
| | | | - Alper Ozcan
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey
| | - Hasret Erkmen
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Can Berk Leblebici
- Department of Medical Genetics, Ankara University Faculty of Medicine, Ankara, Türkiye
| | - Ebru Yilmaz
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey
| | - Musa Karakukcu
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey
| | - Mehmet Kose
- Division of Pediatric Pulmonology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
| | - Ozlem Canoz
- Department of Pathology, Faculty of Medicine, Erciyes University, 38039, Kayseri, Türkiye
| | - Ahmet Özen
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Elif Karakoc-Aydiner
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Serdar Ceylaner
- Intergen, Genetic, Rare and Undiagnosed Diseases, Diagnosis and Research Center, Ankara, Türkiye
| | - Gülsüm Gümüş
- Division of Pediatric Radiology, Department of Radiology, Erciyes University Faculty of Medicine, Kayseri, Türkiye
| | - Huseyin Per
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
| | - Hakan Gumus
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
| | - Halit Canatan
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye
| | - Servet Ozcan
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye
- Department of Biology, Faculty of Science, Erciyes University, Kayseri, 38039, Türkiye
| | - Figen Dogu
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Aydan Ikinciogullari
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Ekrem Unal
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey.
- School of Health Sciences, Hasan Kalyoncu University, Gaziantep, Türkiye.
- Medical Point Hospital, Pediatric Hematology Oncology and BMT Unit, Gaziantep, Türkiye.
| | - Safa Baris
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye.
| | - Ahmet Eken
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye.
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye.
| |
Collapse
|
17
|
Velle KB, Swafford AJM, Garner E, Fritz-Laylin LK. Actin network evolution as a key driver of eukaryotic diversification. J Cell Sci 2024; 137:jcs261660. [PMID: 39120594 DOI: 10.1242/jcs.261660] [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] [Indexed: 08/10/2024] Open
Abstract
Eukaryotic cells have been evolving for billions of years, giving rise to wildly diverse cell forms and functions. Despite their variability, all eukaryotic cells share key hallmarks, including membrane-bound organelles, heavily regulated cytoskeletal networks and complex signaling cascades. Because the actin cytoskeleton interfaces with each of these features, understanding how it evolved and diversified across eukaryotic phyla is essential to understanding the evolution and diversification of eukaryotic cells themselves. Here, we discuss what we know about the origin and diversity of actin networks in terms of their compositions, structures and regulation, and how actin evolution contributes to the diversity of eukaryotic form and function.
Collapse
Affiliation(s)
- Katrina B Velle
- Department of Biology, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA
| | | | - Ethan Garner
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | | |
Collapse
|
18
|
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
|
19
|
Zhao X, Fan C, Qie T, Fu X, Chen X, Wang Y, Wu Y, Fu X, Shi K, Yan W, Yu H. Diaph1 knockout inhibits mouse primordial germ cell proliferation and affects gonadal development. Reprod Biol Endocrinol 2024; 22:82. [PMID: 39010074 PMCID: PMC11247884 DOI: 10.1186/s12958-024-01257-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Exploring the molecular mechanisms of primordial germ cell (PGC) migration and the involvement of gonadal somatic cells in gonad development is valuable for comprehending the origins and potential treatments of reproductive-related diseases. METHODS Diaphanous related formin 1 (Diaph1, also known as mDia1) was screened by analyzing publicly available datasets (ATAC-seq, DNase-seq, and RNA-seq). Subsequently, the CRISPR-Cas9 technology was used to construct Diaph1 knockout mice to investigate the role of Diaph1 in gonad development. RESULTS Based on data from public databases, a differentially expressed gene Diaph1, was identified in the migration of mouse PGC. Additionally, the number of PGCs was significantly reduced in Diaph1 knockout mice compared to wild type mice, and the expression levels of genes related to proliferation (Dicer1, Mcm9), adhesion (E-cadherin, Cdh1), and migration (Cxcr4, Hmgcr, Dazl) were significantly decreased. Diaph1 knockout also inhibited Leydig cell proliferation and induced apoptosis in the testis, as well as granulosa cell apoptosis in the ovary. Moreover, the sperm count in the epididymal region and the count of ovarian follicles were significantly reduced in Diaph1 knockout mice, resulting in decreased fertility, concomitant with lowered levels of serum testosterone and estradiol. Further research found that in Diaph1 knockout mice, the key enzymes involved in testosterone synthesis (CYP11A1, 3β-HSD) were decreased in Leydig cells, and the estradiol-associated factor (FSH receptor, AMH) in granulosa cells were also downregulated. CONCLUSIONS Overall, our findings indicate that the knockout of Diaph1 can disrupt the expression of factors that regulate sex hormone production, leading to impaired secretion of sex hormones, ultimately resulting in damage to reproductive function. These results provide a new perspective on the molecular mechanisms underlying PGC migration and gonadal development, and offer valuable insights for further research on the causes, diagnosis, and treatment of related diseases.
Collapse
Affiliation(s)
- Xin Zhao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Chunbiao Fan
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Tongtong Qie
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Xinrui Fu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Xiaoshuang Chen
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Yujia Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Yuan Wu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Xinyao Fu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Kesong Shi
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Wenlong Yan
- School of Biology and Agriculture, Shaoguan University, Shaoguan, 512005, Guangdong Province, China.
| | - Haiquan Yu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China.
| |
Collapse
|
20
|
Sakamoto R, Murrell MP. Composite branched and linear F-actin maximize myosin-induced membrane shape changes in a biomimetic cell model. Commun Biol 2024; 7:840. [PMID: 38987288 PMCID: PMC11236970 DOI: 10.1038/s42003-024-06528-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
The architecture of the actin cortex determines the generation and transmission of stresses, during key events from cell division to migration. However, its impact on myosin-induced cell shape changes remains unclear. Here, we reconstitute a minimal model of the actomyosin cortex with branched or linear F-actin architecture within giant unilamellar vesicles (GUVs, liposomes). Upon light activation of myosin, neither the branched nor linear F-actin architecture alone induces significant liposome shape changes. The branched F-actin network forms an integrated, membrane-bound "no-slip boundary" -like cortex that attenuates actomyosin contractility. By contrast, the linear F-actin network forms an unintegrated "slip boundary" -like cortex, where actin asters form without inducing membrane deformations. Notably, liposomes undergo significant deformations at an optimized balance of branched and linear F-actin networks. Our findings highlight the pivotal roles of branched F-actin in force transmission and linear F-actin in force generation to yield membrane shape changes.
Collapse
Affiliation(s)
- Ryota Sakamoto
- Department of Biomedical Engineering, Yale University, 10 Hillhouse Avenue, New Haven, CT, USA
- Systems Biology Institute, 850 West Campus Drive, West Haven, CT, USA
| | - Michael P Murrell
- Department of Biomedical Engineering, Yale University, 10 Hillhouse Avenue, New Haven, CT, USA.
- Systems Biology Institute, 850 West Campus Drive, West Haven, CT, USA.
- Department of Physics, Yale University, 217 Prospect Street, New Haven, CT, USA.
| |
Collapse
|
21
|
Cvrčková F, Ghosh R, Kočová H. Transmembrane formins as active cargoes of membrane trafficking. JOURNAL OF EXPERIMENTAL BOTANY 2024; 75:3668-3684. [PMID: 38401146 PMCID: PMC11194305 DOI: 10.1093/jxb/erae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/23/2024] [Indexed: 02/26/2024]
Abstract
Formins are a large, evolutionarily old family of cytoskeletal regulators whose roles include actin capping and nucleation, as well as modulation of microtubule dynamics. The plant class I formin clade is characterized by a unique domain organization, as most of its members are transmembrane proteins with possible cell wall-binding motifs exposed to the extracytoplasmic space-a structure that appears to be a synapomorphy of the plant kingdom. While such transmembrane formins are traditionally considered mainly as plasmalemma-localized proteins contributing to the organization of the cell cortex, we review, from a cell biology perspective, the growing evidence that they can also, at least temporarily, reside (and in some cases also function) in endomembranes including secretory and endocytotic pathway compartments, the endoplasmic reticulum, the nuclear envelope, and the tonoplast. Based on this evidence, we propose that class I formins may thus serve as 'active cargoes' of membrane trafficking-membrane-embedded proteins that modulate the fate of endo- or exocytotic compartments while being transported by them.
Collapse
Affiliation(s)
- Fatima Cvrčková
- Department of Experimental Plant Biology, Faculty of Science, Charles University, Viničná 5, CZ 128 43 Praha 2, Czechia
| | - Rajdeep Ghosh
- Department of Experimental Plant Biology, Faculty of Science, Charles University, Viničná 5, CZ 128 43 Praha 2, Czechia
| | - Helena Kočová
- Department of Experimental Plant Biology, Faculty of Science, Charles University, Viničná 5, CZ 128 43 Praha 2, Czechia
| |
Collapse
|
22
|
Fox S, Gaudreau-LaPierre A, Reshke R, Podinic I, Gibbings DJ, Trinkle-Mulcahy L, Copeland JW. Identification of an FMNL2 Interactome by Quantitative Mass Spectrometry. Int J Mol Sci 2024; 25:5686. [PMID: 38891874 PMCID: PMC11171801 DOI: 10.3390/ijms25115686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Formin Homology Proteins (Formins) are a highly conserved family of cytoskeletal regulatory proteins that participate in a diverse range of cellular processes. FMNL2 is a member of the Diaphanous-Related Formin sub-group, and previous reports suggest FMNL2's role in filopodia assembly, force generation at lamellipodia, subcellular trafficking, cell-cell junction assembly, and focal adhesion formation. How FMNL2 is recruited to these sites of action is not well understood. To shed light on how FMNL2 activity is partitioned between subcellular locations, we used biotin proximity labeling and proteomic analysis to identify an FMNL2 interactome. The interactome identified known and new FMNL2 interacting proteins with functions related to previously described FMNL2 activities. In addition, our interactome predicts a novel connection between FMNL2 and extracellular vesicle assembly. We show directly that FMNL2 protein is present in exosomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - John W. Copeland
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (S.F.)
| |
Collapse
|
23
|
Pan MH, Zhang KH, Wu SL, Pan ZN, Sun MH, Li XH, Ju JQ, Luo SM, Ou XH, Sun SC. FMNL2 regulates actin for endoplasmic reticulum and mitochondria distribution in oocyte meiosis. eLife 2024; 12:RP92732. [PMID: 38747713 PMCID: PMC11095938 DOI: 10.7554/elife.92732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024] Open
Abstract
During mammalian oocyte meiosis, spindle migration and asymmetric cytokinesis are unique steps for the successful polar body extrusion. The asymmetry defects of oocytes will lead to the failure of fertilization and embryo implantation. In present study, we reported that an actin nucleating factor Formin-like 2 (FMNL2) played critical roles in the regulation of spindle migration and organelle distribution in mouse and porcine oocytes. Our results showed that FMNL2 mainly localized at the oocyte cortex and periphery of spindle. Depletion of FMNL2 led to the failure of polar body extrusion and large polar bodies in oocytes. Live-cell imaging revealed that the spindle failed to migrate to the oocyte cortex, which caused polar body formation defects, and this might be due to the decreased polymerization of cytoplasmic actin by FMNL2 depletion in the oocytes of both mice and pigs. Furthermore, mass spectrometry analysis indicated that FMNL2 was associated with mitochondria and endoplasmic reticulum (ER)-related proteins, and FMNL2 depletion disrupted the function and distribution of mitochondria and ER, showing with decreased mitochondrial membrane potential and the occurrence of ER stress. Microinjecting Fmnl2-EGFP mRNA into FMNL2-depleted oocytes significantly rescued these defects. Thus, our results indicate that FMNL2 is essential for the actin assembly, which further involves into meiotic spindle migration and ER/mitochondria functions in mammalian oocytes.
Collapse
Affiliation(s)
- Meng-Hao Pan
- College of Animal Science and Technology, Nanjing Agricultural UniversityNanjingChina
- College of Veterinary Medicine, Northwest A&F UniversityShaanxiChina
| | - Kun-Huan Zhang
- College of Animal Science and Technology, Nanjing Agricultural UniversityNanjingChina
| | - Si-Le Wu
- College of Animal Science and Technology, Nanjing Agricultural UniversityNanjingChina
| | - Zhen-Nan Pan
- College of Animal Science and Technology, Nanjing Agricultural UniversityNanjingChina
| | - Ming-Hong Sun
- College of Animal Science and Technology, Nanjing Agricultural UniversityNanjingChina
| | - Xiao-Han Li
- College of Animal Science and Technology, Nanjing Agricultural UniversityNanjingChina
| | - Jia-Qian Ju
- College of Animal Science and Technology, Nanjing Agricultural UniversityNanjingChina
| | - Shi-Ming Luo
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General HospitalGuangzhouChina
| | - Xiang-Hong Ou
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General HospitalGuangzhouChina
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural UniversityNanjingChina
| |
Collapse
|
24
|
Coscia SM, Moore AS, Thompson CP, Tirrito CF, Ostap EM, Holzbaur ELF. An interphase actin wave promotes mitochondrial content mixing and organelle homeostasis. Nat Commun 2024; 15:3793. [PMID: 38714822 PMCID: PMC11076292 DOI: 10.1038/s41467-024-48189-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 04/22/2024] [Indexed: 05/10/2024] Open
Abstract
Across the cell cycle, mitochondrial dynamics are regulated by a cycling wave of actin polymerization/depolymerization. In metaphase, this wave induces actin comet tails on mitochondria that propel these organelles to drive spatial mixing, resulting in their equitable inheritance by daughter cells. In contrast, during interphase the cycling actin wave promotes localized mitochondrial fission. Here, we identify the F-actin nucleator/elongator FMNL1 as a positive regulator of the wave. FMNL1-depleted cells exhibit decreased mitochondrial polarization, decreased mitochondrial oxygen consumption, and increased production of reactive oxygen species. Accompanying these changes is a loss of hetero-fusion of wave-fragmented mitochondria. Thus, we propose that the interphase actin wave maintains mitochondrial homeostasis by promoting mitochondrial content mixing. Finally, we investigate the mechanistic basis for the observation that the wave drives mitochondrial motility in metaphase but mitochondrial fission in interphase. Our data indicate that when the force of actin polymerization is resisted by mitochondrial tethering to microtubules, as in interphase, fission results.
Collapse
Affiliation(s)
- Stephen M Coscia
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Andrew S Moore
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | - Cameron P Thompson
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Christian F Tirrito
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - E Michael Ostap
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Sakamoto R, Murrell MP. F-actin architecture determines the conversion of chemical energy into mechanical work. Nat Commun 2024; 15:3444. [PMID: 38658549 PMCID: PMC11043346 DOI: 10.1038/s41467-024-47593-x] [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: 10/13/2023] [Accepted: 04/03/2024] [Indexed: 04/26/2024] Open
Abstract
Mechanical work serves as the foundation for dynamic cellular processes, ranging from cell division to migration. A fundamental driver of cellular mechanical work is the actin cytoskeleton, composed of filamentous actin (F-actin) and myosin motors, where force generation relies on adenosine triphosphate (ATP) hydrolysis. F-actin architectures, whether bundled by crosslinkers or branched via nucleators, have emerged as pivotal regulators of myosin II force generation. However, it remains unclear how distinct F-actin architectures impact the conversion of chemical energy to mechanical work. Here, we employ in vitro reconstitution of distinct F-actin architectures with purified components to investigate their influence on myosin ATP hydrolysis (consumption). We find that F-actin bundles composed of mixed polarity F-actin hinder network contraction compared to non-crosslinked network and dramatically decelerate ATP consumption rates. Conversely, linear-nucleated networks allow network contraction despite reducing ATP consumption rates. Surprisingly, branched-nucleated networks facilitate high ATP consumption without significant network contraction, suggesting that the branched network dissipates energy without performing work. This study establishes a link between F-actin architecture and myosin energy consumption, elucidating the energetic principles underlying F-actin structure formation and the performance of mechanical work.
Collapse
Affiliation(s)
- Ryota Sakamoto
- Department of Biomedical Engineering, Yale University, 10 Hillhouse Avenue, New Haven, CT, USA
- Systems Biology Institute, 850 West Campus Drive, West Haven, CT, USA
| | - Michael P Murrell
- Department of Biomedical Engineering, Yale University, 10 Hillhouse Avenue, New Haven, CT, USA.
- Systems Biology Institute, 850 West Campus Drive, West Haven, CT, USA.
- Department of Physics, Yale University, 217 Prospect Street, New Haven, CT, USA.
| |
Collapse
|
26
|
Pimm ML, Haarer BK, Nobles AD, Haney LM, Marcin AG, Marcela Alcaide Eligio, Henty-Ridilla JL. Coordination of actin plus-end dynamics by IQGAP1, formin, and capping protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.04.539490. [PMID: 37205555 PMCID: PMC10187324 DOI: 10.1101/2023.05.04.539490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Cell processes require precise regulation of actin polymerization that is mediated by plus-end regulatory proteins. Detailed mechanisms that explain plus-end dynamics involve regulators with opposing roles, including factors that enhance assembly, e.g., the formin mDia1, and others that stop growth (Capping Protein, CPz). We explore IQGAP1's roles regulating actin filament plus-ends and the consequences of perturbing its activity in cells. We confirm that IQGAP1 pauses elongation and interacts with plus ends through two residues (C756 and C781). We directly visualize the dynamic interplay between IQGAP1 and mDia1, revealing that IQGAP1 displaces the formin to influence actin assembly. Using four-color TIRF we show that IQGAP1's displacement activity extends to formin-CPz 'decision complexes', promoting end-binding protein turnover at plus-ends. Loss of IQGAP1 or its plus-end activities disrupts morphology and migration, emphasizing its essential role. These results reveal a new role for IQGAP1 in promoting protein turnover on filament ends and provide new insights into how plus-end actin assembly is regulated in cells.
Collapse
Affiliation(s)
- Morgan L Pimm
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Brian K Haarer
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Alexander D Nobles
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Laura M Haney
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Alexandra G Marcin
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Marcela Alcaide Eligio
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Jessica L Henty-Ridilla
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- Department of Neuroscience & Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| |
Collapse
|
27
|
Boussaty EC, Ninoyu Y, Andrade LR, Li Q, Takeya R, Sumimoto H, Ohyama T, Wahlin KJ, Manor U, Friedman RA. Altered Fhod3 expression involved in progressive high-frequency hearing loss via dysregulation of actin polymerization stoichiometry in the cuticular plate. PLoS Genet 2024; 20:e1011211. [PMID: 38498576 PMCID: PMC10977885 DOI: 10.1371/journal.pgen.1011211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 03/28/2024] [Accepted: 03/05/2024] [Indexed: 03/20/2024] Open
Abstract
Age-related hearing loss (ARHL) is a common sensory impairment with complex underlying mechanisms. In our previous study, we performed a meta-analysis of genome-wide association studies (GWAS) in mice and identified a novel locus on chromosome 18 associated with ARHL specifically linked to a 32 kHz tone burst stimulus. Consequently, we investigated the role of Formin Homology 2 Domain Containing 3 (Fhod3), a newly discovered candidate gene for ARHL based on the GWAS results. We observed Fhod3 expression in auditory hair cells (HCs) primarily localized at the cuticular plate (CP). To understand the functional implications of Fhod3 in the cochlea, we generated Fhod3 overexpression mice (Pax2-Cre+/-; Fhod3Tg/+) (TG) and HC-specific conditional knockout mice (Atoh1-Cre+/-; Fhod3fl/fl) (KO). Audiological assessments in TG mice demonstrated progressive high-frequency hearing loss, characterized by predominant loss of outer hair cells, and a decreased phalloidin intensities of CP. Ultrastructural analysis revealed loss of the shortest row of stereocilia in the basal turn of the cochlea, and alterations in the cuticular plate surrounding stereocilia rootlets. Importantly, the hearing and HC phenotype in TG mice phenocopied that of the KO mice. These findings suggest that balanced expression of Fhod3 is critical for proper CP and stereocilia structure and function. Further investigation of Fhod3 related hearing impairment mechanisms may lend new insight towards the myriad mechanisms underlying ARHL, which in turn could facilitate the development of therapeutic strategies for ARHL.
Collapse
Affiliation(s)
- Ely Cheikh Boussaty
- Department of Otolaryngology–Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States of America
| | - Yuzuru Ninoyu
- Department of Otolaryngology–Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States of America
| | - Leonardo R. Andrade
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Qingzhong Li
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Ryu Takeya
- Department of Pharmacology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hideki Sumimoto
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Takahiro Ohyama
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Karl J. Wahlin
- Shiley Eye Institute, University of California, San Diego, San Diego, California, United States of America
| | - Uri Manor
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, California, United States of America
- Department of Cell & Developmental Biology, School of Biological Sciences, University of California, San Diego, United States of America
| | - Rick A. Friedman
- Department of Otolaryngology–Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States of America
| |
Collapse
|
28
|
Xu L, Cao L, Li J, Staiger CJ. Cooperative actin filament nucleation by the Arp2/3 complex and formins maintains the homeostatic cortical array in Arabidopsis epidermal cells. THE PLANT CELL 2024; 36:764-789. [PMID: 38057163 PMCID: PMC10896301 DOI: 10.1093/plcell/koad301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/17/2023] [Accepted: 11/19/2023] [Indexed: 12/08/2023]
Abstract
Precise control over how and where actin filaments are created leads to the construction of unique cytoskeletal arrays within a common cytoplasm. Actin filament nucleators are key players in this activity and include the conserved actin-related protein 2/3 (Arp2/3) complex as well as a large family of formins. In some eukaryotic cells, these nucleators compete for a common pool of actin monomers and loss of one favors the activity of the other. To test whether this mechanism is conserved, we combined the ability to image single filament dynamics in the homeostatic cortical actin array of living Arabidopsis (Arabidopsis thaliana) epidermal cells with genetic and/or small molecule inhibitor approaches to stably or acutely disrupt nucleator activity. We found that Arp2/3 mutants or acute CK-666 treatment markedly reduced the frequency of side-branched nucleation events as well as overall actin filament abundance. We also confirmed that plant formins contribute to side-branched filament nucleation in vivo. Surprisingly, simultaneous inhibition of both classes of nucleator increased overall actin filament abundance and enhanced the frequency of de novo nucleation events by an unknown mechanism. Collectively, our findings suggest that multiple actin nucleation mechanisms cooperate to generate and maintain the homeostatic cortical array of plant epidermal cells.
Collapse
Affiliation(s)
- Liyuan Xu
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Lingyan Cao
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jiejie Li
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Christopher J Staiger
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN 47907, USA
- EMBRIO Institute, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
29
|
Rajan S, Aguirre R, Hong Zhou Z, Hauser P, Reisler E. Drebrin Protects Assembled Actin from INF2-FFC-mediated Severing and Stabilizes Cell Protrusions. J Mol Biol 2024; 436:168421. [PMID: 38158176 DOI: 10.1016/j.jmb.2023.168421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Highly specialized cells, such as neurons and podocytes, have arborized morphologies that serve their specific functions. Actin cytoskeleton and its associated proteins are responsible for the distinctive shapes of cells. The mechanism of their cytoskeleton regulation - contributing to cell shape maintenance - is yet to be fully clarified. Inverted formin 2 (INF2), one of the modulators of the cytoskeleton, is an atypical formin that can both polymerize and depolymerize actin filaments depending on its molar ratio to actin. Prior work has established that INF2 binds to the sides of actin filaments and severs them. Drebrin is another actin-binding protein that also binds filaments laterally and stabilizes them, but the interplay between drebrin and INF2 on actin filament stabilization is not well understood. Here, we have used biochemical assays, electron microscopy, and total internal reflection fluorescence microscopy imaging to show that drebrin protects actin filaments from severing by INF2 without inhibiting its polymerization activity. Notably, truncated drebrin - DrbA1-300 - is sufficient for this protection, though not as effective as the full-length protein. INF2 and drebrin are abundantly expressed in highly specialized cells and are crucial for the temporal regulation of their actin cytoskeleton, consistent with their involvement in peripheral neuropathy.
Collapse
Affiliation(s)
- Sudeepa Rajan
- Department of Chemistry and Biochemistry, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Roman Aguirre
- Department of Chemistry and Biochemistry, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA
| | - Z Hong Zhou
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA
| | - Peter Hauser
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA 91344, USA; Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Molecular Biology Institute, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
30
|
Casanova AG, Roth GS, Hausmann S, Lu X, Bischoff LJM, Froeliger EM, Belmudes L, Bourova-Flin E, Flores NM, Benitez AM, Chasan T, Caporicci M, Vayr J, Blanchet S, Ielasi F, Rousseaux S, Hainaut P, Gozani O, Le Romancer M, Couté Y, Palencia A, Mazur PK, Reynoird N. Cytoskeleton remodeling induced by SMYD2 methyltransferase drives breast cancer metastasis. Cell Discov 2024; 10:12. [PMID: 38296970 PMCID: PMC10830559 DOI: 10.1038/s41421-023-00644-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 12/13/2023] [Indexed: 02/02/2024] Open
Abstract
Malignant forms of breast cancer refractory to existing therapies remain a major unmet health issue, primarily due to metastatic spread. A better understanding of the mechanisms at play will provide better insights for alternative treatments to prevent breast cancer cell dispersion. Here, we identify the lysine methyltransferase SMYD2 as a clinically actionable master regulator of breast cancer metastasis. While SMYD2 is overexpressed in aggressive breast cancers, we notice that it is not required for primary tumor growth. However, mammary-epithelium specific SMYD2 ablation increases mouse overall survival by blocking the primary tumor cell ability to metastasize. Mechanistically, we identify BCAR3 as a genuine physiological substrate of SMYD2 in breast cancer cells. BCAR3 monomethylated at lysine K334 (K334me1) is recognized by a novel methyl-binding domain present in FMNLs proteins. These actin cytoskeleton regulators are recruited at the cell edges by the SMYD2 methylation signaling and modulate lamellipodia properties. Breast cancer cells with impaired BCAR3 methylation lose migration and invasiveness capacity in vitro and are ineffective in promoting metastases in vivo. Remarkably, SMYD2 pharmacologic inhibition efficiently impairs the metastatic spread of breast cancer cells, PDX and aggressive mammary tumors from genetically engineered mice. This study provides a rationale for innovative therapeutic prevention of malignant breast cancer metastatic progression by targeting the SMYD2-BCAR3-FMNL axis.
Collapse
Affiliation(s)
- Alexandre G Casanova
- Grenoble Alpes University, CNRS UMR 5309, INSERM U 1209, Institute for Advanced Biosciences, Grenoble, France
| | - Gael S Roth
- Grenoble Alpes University, CNRS UMR 5309, INSERM U 1209, Institute for Advanced Biosciences, Grenoble, France
- Clinique Universitaire d'Hépato-gastroentérologie et Oncologie digestive, CHU Grenoble Alpes, Grenoble, France
| | - Simone Hausmann
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoyin Lu
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ludivine J M Bischoff
- Grenoble Alpes University, CNRS UMR 5309, INSERM U 1209, Institute for Advanced Biosciences, Grenoble, France
| | - Emilie M Froeliger
- Grenoble Alpes University, CNRS UMR 5309, INSERM U 1209, Institute for Advanced Biosciences, Grenoble, France
| | - Lucid Belmudes
- Grenoble Alpes University, CEA, INSERM, UA13 BGE, CNRS CEA, FR2048, Grenoble, France
| | - Ekaterina Bourova-Flin
- Grenoble Alpes University, CNRS UMR 5309, INSERM U 1209, Institute for Advanced Biosciences, Grenoble, France
| | - Natasha M Flores
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ana Morales Benitez
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tourkian Chasan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marcello Caporicci
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jessica Vayr
- Grenoble Alpes University, CNRS UMR 5309, INSERM U 1209, Institute for Advanced Biosciences, Grenoble, France
| | - Sandrine Blanchet
- Grenoble Alpes University, CNRS UMR 5309, INSERM U 1209, Institute for Advanced Biosciences, Grenoble, France
| | - Francesco Ielasi
- Grenoble Alpes University, CNRS UMR 5309, INSERM U 1209, Institute for Advanced Biosciences, Grenoble, France
| | - Sophie Rousseaux
- Grenoble Alpes University, CNRS UMR 5309, INSERM U 1209, Institute for Advanced Biosciences, Grenoble, France
| | - Pierre Hainaut
- Grenoble Alpes University, CNRS UMR 5309, INSERM U 1209, Institute for Advanced Biosciences, Grenoble, France
| | - Or Gozani
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Muriel Le Romancer
- Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR5286, Lyon, France
| | - Yohann Couté
- Grenoble Alpes University, CEA, INSERM, UA13 BGE, CNRS CEA, FR2048, Grenoble, France
| | - Andres Palencia
- Grenoble Alpes University, CNRS UMR 5309, INSERM U 1209, Institute for Advanced Biosciences, Grenoble, France
| | - Pawel K Mazur
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Nicolas Reynoird
- Grenoble Alpes University, CNRS UMR 5309, INSERM U 1209, Institute for Advanced Biosciences, Grenoble, France.
| |
Collapse
|
31
|
Bremer KV, Wu C, Patel AA, He KL, Grunfeld AM, Chanfreau GF, Quinlan ME. Formin tails act as a switch, inhibiting or enhancing processive actin elongation. J Biol Chem 2024; 300:105557. [PMID: 38097186 PMCID: PMC10797183 DOI: 10.1016/j.jbc.2023.105557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 01/04/2024] Open
Abstract
Formins are large, multidomain proteins that nucleate new actin filaments and accelerate elongation through a processive interaction with the barbed ends of filaments. Their actin assembly activity is generally attributed to their eponymous formin homology (FH) 1 and 2 domains; however, evidence is mounting that regions outside of the FH1FH2 stretch also tune actin assembly. Here, we explore the underlying contributions of the tail domain, which spans the sequence between the FH2 domain and the C terminus of formins. Tails vary in length from ∼0 to >200 residues and contain a number of recognizable motifs. The most common and well-studied motif is the ∼15-residue-long diaphanous autoregulatory domain. This domain mediates all or nothing regulation of actin assembly through an intramolecular interaction with the diaphanous inhibitory domain in the N-terminal half of the protein. Multiple reports demonstrate that the tail can enhance both nucleation and processivity. In this study, we provide a high-resolution view of the alternative splicing encompassing the tail in the formin homology domain (Fhod) family of formins during development. While four distinct tails are predicted, we found significant levels of only two of these. We characterized the biochemical effects of the different tails. Surprisingly, the two highly expressed Fhod-tails inhibit processive elongation and diminish nucleation, while a third supports activity. These findings demonstrate a new mechanism of modulating actin assembly by formins and support a model in which splice variants are specialized to build distinct actin structures during development.
Collapse
Affiliation(s)
- Kathryn V Bremer
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California, USA
| | - Carolyn Wu
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California, USA
| | - Aanand A Patel
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California, USA
| | - Kevin L He
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California, USA
| | - Alex M Grunfeld
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California, USA
| | - Guillaume F Chanfreau
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California, USA; Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, USA
| | - Margot E Quinlan
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California, USA; Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, USA.
| |
Collapse
|
32
|
Li Z, Su M, Xie X, Wang P, Bi H, Li E, Ren K, Dong L, Lv Z, Ma X, Liu Y, Zhao B, Peng Y, Liu J, Liu L, Yang J, Ji P, Mei Y. mDia formins form hetero-oligomers and cooperatively maintain murine hematopoiesis. PLoS Genet 2023; 19:e1011084. [PMID: 38157491 PMCID: PMC10756686 DOI: 10.1371/journal.pgen.1011084] [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: 05/12/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024] Open
Abstract
mDia formin proteins regulate the dynamics and organization of the cytoskeleton through their linear actin nucleation and polymerization activities. We previously showed that mDia1 deficiency leads to aberrant innate immune activation and induces myelodysplasia in a mouse model, and mDia2 regulates enucleation and cytokinesis of erythroblasts and the engraftment of hematopoietic stem and progenitor cells (HSPCs). However, whether and how mDia formins interplay and regulate hematopoiesis under physiological and stress conditions remains unknown. Here, we found that both mDia1 and mDia2 are required for HSPC regeneration under stress, such as serial plating, aging, and reconstitution after myeloid ablation. We showed that mDia1 and mDia2 form hetero-oligomers through the interactions between mDia1 GBD-DID and mDia2 DAD domains. Double knockout of mDia1 and mDia2 in hematopoietic cells synergistically impaired the filamentous actin network and serum response factor-involved transcriptional signaling, which led to declined HSPCs, severe anemia, and significant mortality in neonates and newborn mice. Our data demonstrate the potential roles of mDia hetero-oligomerization and their non-rodent functions in the regulation of HSPCs activity and orchestration of hematopoiesis.
Collapse
Affiliation(s)
- Zhaofeng Li
- Hunan Provincial Key Laboratory of Animal Model and Molecular Medicine, Hunan University, Changsha, China
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Meng Su
- Hunan Provincial Key Laboratory of Animal Model and Molecular Medicine, Hunan University, Changsha, China
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Xinshu Xie
- Hunan Provincial Key Laboratory of Animal Model and Molecular Medicine, Hunan University, Changsha, China
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Pan Wang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Honghao Bi
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Ermin Li
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Kehan Ren
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Lili Dong
- Hunan Provincial Key Laboratory of Animal Model and Molecular Medicine, Hunan University, Changsha, China
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Zhiyi Lv
- Hunan Provincial Key Laboratory of Animal Model and Molecular Medicine, Hunan University, Changsha, China
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Xuezhen Ma
- Hunan Provincial Key Laboratory of Animal Model and Molecular Medicine, Hunan University, Changsha, China
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Yijie Liu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Baobing Zhao
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yuanliang Peng
- Department of Hematology, the Second Xiangya Hospital; Molecular Biology Research Center, School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University; Changsha, China
| | - Jing Liu
- Department of Hematology, the Second Xiangya Hospital; Molecular Biology Research Center, School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University; Changsha, China
| | - Lu Liu
- Hunan Provincial Key Laboratory of Animal Model and Molecular Medicine, Hunan University, Changsha, China
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Jing Yang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Peng Ji
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Yang Mei
- Hunan Provincial Key Laboratory of Animal Model and Molecular Medicine, Hunan University, Changsha, China
- School of Biomedical Sciences, Hunan University, Changsha, China
| |
Collapse
|
33
|
Lopes Dos Santos R, Malo M, Campillo C. Spatial Control of Arp2/3-Induced Actin Polymerization on Phase-Separated Giant Unilamellar Vesicles. ACS Synth Biol 2023; 12:3267-3274. [PMID: 37909673 DOI: 10.1021/acssynbio.3c00268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Deciphering the physical mechanisms underlying cell shape changes, while avoiding the cellular interior's complexity, involves the development of controlled basic biomimetic systems that imitate cell functions. In particular, the reconstruction of cytoskeletal dynamics on cell-sized giant unilamellar vesicles (GUVs) has allowed for the reconstituting of some cell-like processes in vitro. In fact, such a bottom-up strategy could be the basis for forming protocells able to reorganize or even move autonomously. However, reconstituting the subtle and controlled dynamics of the cytoskeleton-membrane interface in vitro remains an experimental challenge. Taking advantage of the lipid-induced segregation of an actin polymerization activator, we present a system that targets actin polymerization in specific domains of phase-separated GUVs. We observe actin networks localized on Lo, Ld, or on both types of domains and the actin-induced deformation or reorganization of these domains. These results suggest that the system we have developed here could pave the way for future experiments further detailing the interplay between actin dynamics and membrane heterogeneities.
Collapse
Affiliation(s)
- Rogério Lopes Dos Santos
- Université Paris-Saclay, Univ Evry, CY Cergy Paris Université, CNRS, LAMBE, 91025 Evry, Courcouronnes, France
| | - Michel Malo
- Université Paris-Saclay, Univ Evry, CY Cergy Paris Université, CNRS, LAMBE, 91025 Evry, Courcouronnes, France
| | - Clément Campillo
- Université Paris-Saclay, Univ Evry, CY Cergy Paris Université, CNRS, LAMBE, 91025 Evry, Courcouronnes, France
- Institut Universitaire de France (IUF), 75005 Paris, France
| |
Collapse
|
34
|
Belian S, Korenkova O, Zurzolo C. Actin-based protrusions at a glance. J Cell Sci 2023; 136:jcs261156. [PMID: 37987375 DOI: 10.1242/jcs.261156] [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] [Indexed: 11/22/2023] Open
Abstract
Actin-based protrusions are at the base of many fundamental cellular processes, such as cell adhesion, migration and intercellular communication. In recent decades, the discovery of new types of actin-based protrusions with unique functions has enriched our comprehension of cellular processes. However, as the repertoire of protrusions continues to expand, the rationale behind the classification of newly identified and previously known structures becomes unclear. Although current nomenclature allows good categorization of protrusions based on their functions, it struggles to distinguish them when it comes to structure, composition or formation mechanisms. In this Cell Science at a Glance article, we discuss the different types of actin-based protrusions, focusing on filopodia, cytonemes and tunneling nanotubes, to help better distinguish and categorize them based on their structural and functional differences and similarities.
Collapse
Affiliation(s)
- Sevan Belian
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, F-75015 Paris, France
- Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Olga Korenkova
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, F-75015 Paris, France
- Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Chiara Zurzolo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, F-75015 Paris, France
| |
Collapse
|
35
|
Chen H, Hu Y, Yang G, Li P, Yin J, Feng X, Wu Q, Zhang J, Xiao B, Sui Z. Macropinocytosis in Gracilariopsis lemaneiformis (Rhodophyta). FRONTIERS IN PLANT SCIENCE 2023; 14:1225675. [PMID: 37822336 PMCID: PMC10562585 DOI: 10.3389/fpls.2023.1225675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/05/2023] [Indexed: 10/13/2023]
Abstract
Macropinocytosis is an endocytic process that plays an important role in animal development and disease occurrence but until now has been rarely reported in organisms with cell walls. We investigated the properties of endocytosis in a red alga, Gracilariopsis lemaneiformis. The cells non-selectively internalized extracellular fluid into large-scale endocytic vesicles (1.94 ± 0.51 μm), and this process could be inhibited by 5-(N-ethyl-N-isopropyl) amiloride, an macropinocytosis inhibitor. Moreover, endocytosis was driven by F-actin, which promotes formation of ruffles and cups from the cell surface and facilitates formation of endocytotic vesicles. After vesicle formation, endocytic vesicles could be acidified and acquire digestive function. These results indicated macropinocytosis in G. lemaneiformis. Abundant phosphatidylinositol kinase and small GTPase encoding genes were found in the genome of this alga, while PI3K, Ras, and Rab5, the important participators of traditional macropinocytosis, seem to be lacked. Such findings provide a new insight into endocytosis in organisms with cell walls and facilitate further research into the core regulatory mechanisms and evolution of macropinocytosis.
Collapse
Affiliation(s)
- Haihong Chen
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, China
| | - Yiyi Hu
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
| | - Guanpin Yang
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
- Institutes of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Pingping Li
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
| | - Jingru Yin
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
| | - Xiaoqing Feng
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
| | - Qiong Wu
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
| | - Jingyu Zhang
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
| | - Baoheng Xiao
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
| | - Zhenghong Sui
- Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, China
| |
Collapse
|
36
|
Belliveau NM, Footer MJ, Akdoǧan E, van Loon AP, Collins SR, Theriot JA. Whole-genome screens reveal regulators of differentiation state and context-dependent migration in human neutrophils. Nat Commun 2023; 14:5770. [PMID: 37723145 PMCID: PMC10507112 DOI: 10.1038/s41467-023-41452-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 08/31/2023] [Indexed: 09/20/2023] Open
Abstract
Neutrophils are the most abundant leukocyte in humans and provide a critical early line of defense as part of our innate immune system. We perform a comprehensive, genome-wide assessment of the molecular factors critical to proliferation, differentiation, and cell migration in a neutrophil-like cell line. Through the development of multiple migration screen strategies, we specifically probe directed (chemotaxis), undirected (chemokinesis), and 3D amoeboid cell migration in these fast-moving cells. We identify a role for mTORC1 signaling in cell differentiation, which influences neutrophil abundance, survival, and migratory behavior. Across our individual migration screens, we identify genes involved in adhesion-dependent and adhesion-independent cell migration, protein trafficking, and regulation of the actomyosin cytoskeleton. This genome-wide screening strategy, therefore, provides an invaluable approach to the study of neutrophils and provides a resource that will inform future studies of cell migration in these and other rapidly migrating cells.
Collapse
Affiliation(s)
- Nathan M Belliveau
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Matthew J Footer
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Emel Akdoǧan
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Aaron P van Loon
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Sean R Collins
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, 95616, USA
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
37
|
Casanova AG, Roth GS, Hausmann S, Lu X, Belmudes L, Bourova-Flin E, Flores NM, Benitez AM, Caporicci M, Vayr J, Blanchet S, Ielasi F, Rousseaux S, Hainaut P, Gozani O, Couté Y, Palencia A, Mazur PK, Reynoird N. Cytoskeleton remodeling induced by SMYD2 methyltransferase drives breast cancer metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.558201. [PMID: 37790557 PMCID: PMC10542120 DOI: 10.1101/2023.09.18.558201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Malignant forms of breast cancer refractory to existing therapies remain a major unmet health issue, primarily due to metastatic spread. A better understanding of the mechanisms at play will provide better insights for alternative treatments to prevent breast cancer cells dispersion. Here, we identify the lysine methyltransferase SMYD2 as a clinically actionable master regulator of breast cancer metastasis. While SMYD2 is overexpressed in aggressive breast cancers, we notice that it is not required for primary tumor growth. However, mammary-epithelium specific SMYD2 ablation increases mouse overall survival by blocking the primary tumor cells ability to metastasize. Mechanistically, we identify BCAR3 as a genuine physiological substrate of SMYD2 in breast cancer cells. BCAR3 monomethylated at lysine K334 (K334me1) is recognized by a novel methyl-binding domain present in FMNLs proteins. These actin cytoskeleton regulators are recruited at the cell edges by the SMYD2 methylation signaling and modulates lamellipodia properties. Breast cancer cells with impaired BCAR3 methylation loose migration and invasiveness capacity in vitro and are ineffective in promoting metastases in vivo . Remarkably, SMYD2 pharmacologic inhibition efficiently impairs the metastatic spread of breast cancer cells, PDX and aggressive mammary tumors from genetically engineered mice. This study provides a rationale for innovative therapeutic prevention of malignant breast cancer metastatic progression by targeting the SMYD2-BCAR3-FMNL axis.
Collapse
|
38
|
Bai Y, Zhao F, Wu T, Chen F, Pang X. Actin polymerization and depolymerization in developing vertebrates. Front Physiol 2023; 14:1213668. [PMID: 37745245 PMCID: PMC10515290 DOI: 10.3389/fphys.2023.1213668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Development is a complex process that occurs throughout the life cycle. F-actin, a major component of the cytoskeleton, is essential for the morphogenesis of tissues and organs during development. F-actin is formed by the polymerization of G-actin, and the dynamic balance of polymerization and depolymerization ensures proper cellular function. Disruption of this balance results in various abnormalities and defects or even embryonic lethality. Here, we reviewed recent findings on the structure of G-actin and F-actin and the polymerization of G-actin to F-actin. We also focused on the functions of actin isoforms and the underlying mechanisms of actin polymerization/depolymerization in cellular and organic morphogenesis during development. This information will extend our understanding of the role of actin polymerization in the physiologic or pathologic processes during development and may open new avenues for developing therapeutics for embryonic developmental abnormalities or tissue regeneration.
Collapse
Affiliation(s)
- Yang Bai
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Feng Zhao
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tingting Wu
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Fangchun Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xiaoxiao Pang
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
39
|
Vasalou V, Kotidis E, Tatsis D, Boulogeorgou K, Grivas I, Koliakos G, Cheva A, Ioannidis O, Tsingotjidou A, Angelopoulos S. The Effects of Tissue Healing Factors in Wound Repair Involving Absorbable Meshes: A Narrative Review. J Clin Med 2023; 12:5683. [PMID: 37685753 PMCID: PMC10488606 DOI: 10.3390/jcm12175683] [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: 06/21/2023] [Revised: 08/17/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
Wound healing is a complex and meticulously orchestrated process involving multiple phases and cellular interactions. This narrative review explores the intricate mechanisms behind wound healing, emphasizing the significance of cellular processes and molecular factors. The phases of wound healing are discussed, focusing on the roles of immune cells, growth factors, and extracellular matrix components. Cellular shape alterations driven by cytoskeletal modulation and the influence of the 'Formin' protein family are highlighted for their impact on wound healing processes. This review delves into the use of absorbable meshes in wound repair, discussing their categories and applications in different surgical scenarios. Interleukins (IL-2 and IL-6), CD31, CD34, platelet rich plasma (PRP), and adipose tissue-derived mesenchymal stem cells (ADSCs) are discussed in their respective roles in wound healing. The interactions between these factors and their potential synergies with absorbable meshes are explored, shedding light on how these combinations might enhance the healing process. Recent advances and challenges in the field are also presented, including insights into mesh integration, biocompatibility, infection prevention, and postoperative complications. This review underscores the importance of patient-specific factors and surgical techniques in optimizing mesh placement and healing outcomes. As wound healing remains a dynamic field, this narrative review provides a comprehensive overview of the current understanding and potential avenues for future research and clinical applications.
Collapse
Affiliation(s)
- Varvara Vasalou
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
- Andreas Syggros Hospital, 11528 Athens, Greece
| | - Efstathios Kotidis
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
| | - Dimitris Tatsis
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
- Oral and Maxillofacial Surgery Department, School of Dentistry, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
| | - Kassiani Boulogeorgou
- Department of Pathology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.B.)
| | - Ioannis Grivas
- Laboratory of Anatomy, Histology & Embryology, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Georgios Koliakos
- Department of Biochemistry, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Angeliki Cheva
- Department of Pathology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.B.)
| | - Orestis Ioannidis
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
| | - Anastasia Tsingotjidou
- Laboratory of Anatomy, Histology & Embryology, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Stamatis Angelopoulos
- Fourth Surgical Department, School of Medicine, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece
| |
Collapse
|
40
|
Boussaty EC, Ninoyu Y, Andrade L, Li Q, Takeya R, Sumimoto H, Ohyama T, Wahlin KJ, Manor U, Friedman RA. Altered Fhod3 Expression Involved in Progressive High-Frequency Hearing Loss via Dysregulation of Actin Polymerization Stoichiometry in The Cuticular Plate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.20.549974. [PMID: 37546952 PMCID: PMC10401921 DOI: 10.1101/2023.07.20.549974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Age-related hearing loss (ARHL) is a common sensory impairment with comlex underlying mechanisms. In our previous study, we performed a meta-analysis of genome-wide association studies (GWAS) in mice and identified a novel locus on chromosome 18 associated with ARHL specifically linked to a 32 kHz tone burst stimulus. Consequently, we investigated the role of Formin Homology 2 Domain Containing 3 (Fhod3), a newly discovered candidate gene for ARHL based on the GWAS results. We observed Fhod3 expression in auditory hair cells (HCs) and primarily localized at the cuticular plate (CP). To understand the functional implications of Fhod3 in the cochlea, we generated Fhod3 overexpression mice (Pax2-Cre+/-; Fhod3Tg/+) (TG) and HC-specific conditional knockout mice (Atoh1-Cre+/-; Fhod3fl/fl) (KO). Audiological assessments in TG mice demonstrated progressive high-frequency hearing loss, characterized by predominant loss of outer HCs and decrease phalloidin intensities of CP. Ultrastructural analysis revealed shortened stereocilia in the basal turn cochlea. Importantly, the hearing and HC phenotype in TG mice were replicated in KO mice. These findings indicate that Fhod3 plays a critical role in regulating actin dynamics in CP and stereocilia. Further investigation of Fhod3-related hearing impairment mechanisms may facilitate the development of therapeutic strategies for ARHL in humans.
Collapse
|
41
|
Zhang Q, Wan M, Kudryashova E, Kudryashov DS, Mao Y. Membrane-dependent actin polymerization mediated by the Legionella pneumophila effector protein MavH. PLoS Pathog 2023; 19:e1011512. [PMID: 37463171 PMCID: PMC10381072 DOI: 10.1371/journal.ppat.1011512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023] Open
Abstract
L. pneumophila propagates in eukaryotic cells within a specialized niche, the Legionella-containing vacuole (LCV). The infection process is controlled by over 330 effector proteins delivered through the type IV secretion system. In this study, we report that the Legionella MavH effector localizes to endosomes and remodels host actin cytoskeleton in a phosphatidylinositol 3-phosphate (PI(3)P) dependent manner when ectopically expressed. We show that MavH recruits host actin capping protein (CP) and actin to the endosome via its CP-interacting (CPI) motif and WH2-like actin-binding domain, respectively. In vitro assays revealed that MavH stimulates actin assembly on PI(3)P-containing liposomes causing their tubulation. In addition, the recruitment of CP by MavH negatively regulates F-actin density at the membrane. We further show that, in L. pneumophila-infected cells, MavH appears around the LCV at the very early stage of infection and facilitates bacterium entry into the host. Together, our results reveal a novel mechanism of membrane tubulation induced by membrane-dependent actin polymerization catalyzed by MavH that contributes to the early stage of L. pneumophila infection by regulating host actin dynamics.
Collapse
Affiliation(s)
- Qing Zhang
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, United States of America
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Min Wan
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, United States of America
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Elena Kudryashova
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Dmitri S Kudryashov
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Yuxin Mao
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, United States of America
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
42
|
Li D, Yang Y, Lv C, Wang Y, Chao X, Huang J, Singh SP, Yuan Y, Zhang C, Lou J, Gao P, Huang S, Li B, Cai H. GxcM-Fbp17/RacC-WASP signaling regulates polarized cortex assembly in migrating cells via Arp2/3. J Cell Biol 2023; 222:e202208151. [PMID: 37010470 PMCID: PMC10072221 DOI: 10.1083/jcb.202208151] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 02/02/2023] [Accepted: 03/17/2023] [Indexed: 04/04/2023] Open
Abstract
The actin-rich cortex plays a fundamental role in many cellular processes. Its architecture and molecular composition vary across cell types and physiological states. The full complement of actin assembly factors driving cortex formation and how their activities are spatiotemporally regulated remain to be fully elucidated. Using Dictyostelium as a model for polarized and rapidly migrating cells, we show that GxcM, a RhoGEF localized specifically in the rear of migrating cells, functions together with F-BAR protein Fbp17, a small GTPase RacC, and the actin nucleation-promoting factor WASP to coordinately promote Arp2/3 complex-mediated cortical actin assembly. Overactivation of this signaling cascade leads to excessive actin polymerization in the rear cortex, whereas its disruption causes defects in cortical integrity and function. Therefore, apart from its well-defined role in the formation of the protrusions at the cell front, the Arp2/3 complex-based actin carries out a previously unappreciated function in building the rear cortical subcompartment in rapidly migrating cells.
Collapse
Affiliation(s)
- Dong Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yihong Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Chenglin Lv
- Department of Engineering Mechanics, Applied Mechanics Laboratory, Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing, China
| | - Yingjie Wang
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiaoting Chao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jiafeng Huang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | - Ye Yuan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Chengyu Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jizhong Lou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Pu Gao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shanjin Huang
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Bo Li
- Department of Engineering Mechanics, Applied Mechanics Laboratory, Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing, China
| | - Huaqing Cai
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
43
|
Campellone KG, Lebek NM, King VL. Branching out in different directions: Emerging cellular functions for the Arp2/3 complex and WASP-family actin nucleation factors. Eur J Cell Biol 2023; 102:151301. [PMID: 36907023 DOI: 10.1016/j.ejcb.2023.151301] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 02/07/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The actin cytoskeleton impacts practically every function of a eukaryotic cell. Historically, the best-characterized cytoskeletal activities are in cell morphogenesis, motility, and division. The structural and dynamic properties of the actin cytoskeleton are also crucial for establishing, maintaining, and changing the organization of membrane-bound organelles and other intracellular structures. Such activities are important in nearly all animal cells and tissues, although distinct anatomical regions and physiological systems rely on different regulatory factors. Recent work indicates that the Arp2/3 complex, a broadly expressed actin nucleator, drives actin assembly during several intracellular stress response pathways. These newly described Arp2/3-mediated cytoskeletal rearrangements are coordinated by members of the Wiskott-Aldrich Syndrome Protein (WASP) family of actin nucleation-promoting factors. Thus, the Arp2/3 complex and WASP-family proteins are emerging as crucial players in cytoplasmic and nuclear activities including autophagy, apoptosis, chromatin dynamics, and DNA repair. Characterizations of the functions of the actin assembly machinery in such stress response mechanisms are advancing our understanding of both normal and pathogenic processes, and hold great promise for providing insights into organismal development and interventions for disease.
Collapse
Affiliation(s)
- Kenneth G Campellone
- Department of Molecular and Cell Biology, Institute for Systems Genomics; University of Connecticut; Storrs, CT, USA.
| | - Nadine M Lebek
- Department of Molecular and Cell Biology, Institute for Systems Genomics; University of Connecticut; Storrs, CT, USA
| | - Virginia L King
- Department of Molecular and Cell Biology, Institute for Systems Genomics; University of Connecticut; Storrs, CT, USA
| |
Collapse
|
44
|
Xu J, Sang M, Cheng J, Luo C, Shi J, Sun F. Knockdown of disheveled-associated activator of morphogenesis 2 disrupts cytoskeletal organization and phagocytosis in rat Sertoli cells. Mol Cell Endocrinol 2023; 563:111867. [PMID: 36681175 DOI: 10.1016/j.mce.2023.111867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023]
Abstract
Disheveled-associated activator of morphogenesis 2 (DAAM2) regulates actin polymerization and cell motility. In this study, we investigated the role of DAAM2 in the cytoskeleton and phagocytosis of rat Sertoli cells in vitro and in vivo through siRNA transfection and intratesticular injection. We found that knockdown of DAAM2 significantly attenuated cytoskeletal and tight junction marker expression and reduced the integrity of the Sertoli cell monolayer. In rats, loss of DAAM2 induced disarrangement and deformation of sperms and promoted accumulation of apoptotic sperms in the testis, accompanied by morphological abnormalities in the blood-testis barrier. DAAM2 silencing also reduced the ability of Sertoli cells to engulf apoptotic spermatogenic cells and green fluorescence-labeled beads. RNA sequencing and bioinformatics analysis revealed that phagocytosis and cytoskeleton-related genes and pathways were significantly associated with DAAM2. Our study suggests that DAAM2 may be involved in spermatogenesis possibly by regulating cytoskeleton organization and phagocytosis of Sertoli cells.
Collapse
Affiliation(s)
- Junjie Xu
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, China; Department of Obstetrics and Gynecology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Mengmeng Sang
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, China
| | - Jinmei Cheng
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, China
| | - Chunhai Luo
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Jie Shi
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, China
| | - Fei Sun
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, China.
| |
Collapse
|
45
|
El-Mansi S, Robinson CL, Kostelnik KB, McCormack JJ, Mitchell TP, Lobato-Márquez D, Rajeeve V, Cutillas P, Cutler DF, Mostowy S, Nightingale TD. Proximity proteomics identifies septins and PAK2 as decisive regulators of actomyosin-mediated expulsion of von Willebrand factor. Blood 2023; 141:930-944. [PMID: 36564030 PMCID: PMC10023740 DOI: 10.1182/blood.2022017419] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/07/2022] [Accepted: 11/27/2022] [Indexed: 12/25/2022] Open
Abstract
In response to tissue injury, within seconds the ultra-large glycoprotein von Willebrand factor (VWF) is released from endothelial storage organelles (Weibel-Palade bodies) into the lumen of the blood vasculature, where it leads to the recruitment of platelets. The marked size of VWF multimers represents an unprecedented burden on the secretory machinery of endothelial cells (ECs). ECs have evolved mechanisms to overcome this, most notably an actomyosin ring that forms, contracts, and squeezes out its unwieldy cargo. Inhibiting the formation or function of these structures represents a novel therapeutic target for thrombotic pathologies, although characterizing proteins associated with such a dynamic process has been challenging. We have combined APEX2 proximity labeling with an innovative dual loss-of-function screen to identify proteins associated with actomyosin ring function. We show that p21 activated kinase 2 (PAK2) recruits septin hetero-oligomers, a molecular interaction that forms a ring around exocytic sites. This cascade of events controls actomyosin ring function, aiding efficient exocytic release. Genetic or pharmacological inhibition of PAK2 or septins led to inefficient release of VWF and a failure to form platelet-catching strings. This new molecular mechanism offers additional therapeutic targets for the control of thrombotic disease and is highly relevant to other secretory systems that employ exocytic actomyosin machinery.
Collapse
Affiliation(s)
- Sammy El-Mansi
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Christopher L. Robinson
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Katja B. Kostelnik
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Jessica J. McCormack
- MRC Laboratory of Molecular Cell Biology, University College London, London, United Kingdom
| | - Tom P. Mitchell
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Damián Lobato-Márquez
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Vinothini Rajeeve
- Cell Signalling & Proteomics Group, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Pedro Cutillas
- Cell Signalling & Proteomics Group, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Daniel F. Cutler
- MRC Laboratory of Molecular Cell Biology, University College London, London, United Kingdom
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Thomas D. Nightingale
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
46
|
Li Z, Wei X, Zhu Y. The prognostic value of DAAM2 in lower grade glioma, liver cancer, and breast cancer. Clin Transl Oncol 2023:10.1007/s12094-023-03111-x. [PMID: 36790676 DOI: 10.1007/s12094-023-03111-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023]
Abstract
PURPOSE Dishevelled-associated activator of morphogenesis 2 (DAAM2) is a formin protein and has a potential role in the tumor metastasis. The prognostic value of DAAM2 in pan-cancer is investigated in this study. METHODS TCGA and GTEx database were downloaded to perform bioinformatics analysis and ROC curves. Then we explored protein-protein interaction and GO-KEGG enrichment to figure out the protein pathways associated with DAAM2 and studied DAAM2-related immune infiltration and methylation. Fifteen pairs of BRCA clinical samples were enrolled to determine the expression and distribution of DAAM2 in BRCA sections by immunohistochemistry. Finally, BRCA cells were transfected with siRNA targeting DAAM2 and subsequently subject to cell proliferation, migration, and invasion assays. RESULTS DAAM2 was closely related to the diagnosis and clinical characteristics of lower grade glioma (LGG), liver hepatocellular carcinoma (LIHC), and breast cancer (BRCA). Survival curve analysis demonstrated DAAM2 served as a potential prognostic indicator of LGG and LIHC (P = 0.0029 and P = 0.025, respectively). DAAM2 was mainly participated in signaling pathways mediating cytoskeleton regulation and tumor development. The correlation of DAAM2 with tumor-infiltrating immune cells (TIICs) and methylation levels was conducive to the prediction of novel biomarkers of pan-carcinoma. DAAM2 was highly expressed in BRCA tissues than that in paracancerous tissues. The proliferation, invasion, and migration of BRCA cells were inhibited by DAAM2 siRNA. CONCLUSION DAAM2 had a specific value in foretelling the prognosis of LGG, LIHC, and BRCA. High expression level of DAAM2 has longer survival rates in LGG and LIHC. The knockdown of DAAM2 retards the proliferation, invasion, and migration of BRCA cells. This study provides a novel sight of DAAM2 into the exploration of a potential biomarker in pan-cancer.
Collapse
Affiliation(s)
- Zeying Li
- The First Clinical Medical College, Nanjing Medical University, Nanjing, 211166, China
| | - Xiao Wei
- The First Clinical Medical College, Nanjing Medical University, Nanjing, 211166, China
| | - Yichao Zhu
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
47
|
Wirshing AC, Rodriguez SG, Goode BL. Evolutionary tuning of barbed end competition allows simultaneous construction of architecturally distinct actin structures. J Cell Biol 2023; 222:213854. [PMID: 36729023 PMCID: PMC9929936 DOI: 10.1083/jcb.202209105] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/01/2022] [Accepted: 01/13/2023] [Indexed: 02/03/2023] Open
Abstract
How cells simultaneously assemble actin structures of distinct sizes, shapes, and filamentous architectures is still not well understood. Here, we used budding yeast as a model to investigate how competition for the barbed ends of actin filaments might influence this process. We found that while vertebrate capping protein (CapZ) and formins can simultaneously associate with barbed ends and catalyze each other's displacement, yeast capping protein (Cap1/2) poorly displaces both yeast and vertebrate formins. Consistent with these biochemical differences, in vivo formin-mediated actin cable assembly was strongly attenuated by the overexpression of CapZ but not Cap1/2. Multiwavelength live cell imaging further revealed that actin patches in cap2∆ cells acquire cable-like features over time, including recruitment of formins and tropomyosin. Together, our results suggest that the activities of S. cerevisiae Cap1/2 have been tuned across evolution to allow robust cable assembly by formins in the presence of high cytosolic levels of Cap1/2, which conversely limit patch growth and shield patches from formins.
Collapse
Affiliation(s)
- Alison C.E. Wirshing
- https://ror.org/05abbep66Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA
| | - Sofia Gonzalez Rodriguez
- https://ror.org/05abbep66Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA
| | - Bruce L. Goode
- https://ror.org/05abbep66Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA,Correspondence to Bruce L. Goode:
| |
Collapse
|
48
|
Zhang Q, Wan M, Mao Y. Membrane-dependent actin polymerization mediated by the Legionella pneumophila effector protein MavH. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525393. [PMID: 36747622 PMCID: PMC9900769 DOI: 10.1101/2023.01.24.525393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
L. pneumophila propagates in eukaryotic cells within a specialized niche, the Legionella -containing vacuole (LCV). The infection process is controlled by over 330 effector proteins delivered through the type IV secretion system. In this study, we report that the Legionella MavH effector harbors a lipid-binding domain that specifically recognizes PI(3)P (phosphatidylinositol 3-phosphate) and localizes to endosomes when ectopically expressed. We show that MavH recruits host actin capping proteins (CP) and actin to the endosome via its CP interacting (CPI) motif and WH2-like actin-binding domain, respectively. In vitro assays revealed that MavH stimulates robust actin polymerization only in the presence of PI(3)P-containing liposomes and the recruitment of CP by MavH negatively regulates F-actin density at the membrane. Furthermore, in L. pneumophila -infected cells, MavH can be detected around the LCV at the very early stage of infection. Together, our results reveal a novel mechanism of membrane-dependent actin polymerization catalyzed by MavH that may play a role at the early stage of L. pneumophila infection by regulating host actin dynamics.
Collapse
Affiliation(s)
- Qing Zhang
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Min Wan
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Yuxin Mao
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA.,Corresponding Author: , Telephone: 607-255-0783
| |
Collapse
|
49
|
Schneider F, Metz I, Rust MB. Regulation of actin filament assembly and disassembly in growth cone motility and axon guidance. Brain Res Bull 2023; 192:21-35. [PMID: 36336143 DOI: 10.1016/j.brainresbull.2022.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Directed outgrowth of axons is fundamental for the establishment of neuronal networks. Axon outgrowth is guided by growth cones, highly motile structures enriched in filamentous actin (F-actin) located at the axons' distal tips. Growth cones exploit F-actin-based protrusions to scan the environment for guidance cues, and they contain the sensory apparatus to translate guidance cue information into intracellular signaling cascades. These cascades act upstream of actin-binding proteins (ABP) and thereby control assembly and disassembly of F-actin. Spatiotemporally controlled F-actin dis-/assembly in growth cones steers the axon towards attractants and away from repellents, and it thereby navigates the axon through the developing nervous system. Hence, ABP that control F-actin dynamics emerged as critical regulators of neuronal network formation. In the present review article, we will summarize and discuss current knowledge of the mechanisms that control remodeling of the actin cytoskeleton in growth cones, focusing on recent progress in the field. Further, we will introduce tools and techniques that allow to study actin regulatory mechanism in growth cones.
Collapse
Affiliation(s)
- Felix Schneider
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany; Molecular Urooncology, Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Isabell Metz
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps-University of Marburg, 35032 Marburg, Germany; DFG Research Training Group 'Membrane Plasticity in Tissue Development and Remodeling', GRK 2213, Philipps-University of Marburg, 35032 Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, 35032 Marburg, Germany.
| |
Collapse
|
50
|
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: 1] [Impact Index Per Article: 0.3] [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
|