1
|
Guérin C, N'Diaye AB, Gressin L, Mogilner A, Théry M, Blanchoin L, Colin A. Balancing limited resources in actin network competition. Curr Biol 2025; 35:500-513.e5. [PMID: 39793569 DOI: 10.1016/j.cub.2024.11.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/05/2024] [Accepted: 11/26/2024] [Indexed: 01/13/2025]
Abstract
In cells, multiple actin networks coexist in a dynamic manner. These networks compete for a common pool of actin monomers and actin-binding proteins. Interestingly, all of these networks manage to coexist despite the strong competition for resources. Moreover, the coexistence of networks with various strengths is key to cell adaptation to external changes. However, a comprehensive view of how these networks coexist in this competitive environment, where resources are limited, is still lacking. To address this question, we used a reconstituted system, in closed microwells, consisting of beads propelled by actin polymerization or micropatterns functionalized with lipids capable of initiating polymerization close to a membrane. This system enabled us to build dynamic actin architectures, competing for a limited pool of proteins, over a period of hours. We demonstrated the importance of protein turnover for the coexistence of actin networks, showing that it ensures resource distribution between weak and strong networks. However, when competition becomes too intense, turnover alone is insufficient, leading to a selection process that favors the strongest networks. Consequently, we emphasize the importance of competition strength, which is defined by the turnover rate, the amount of available protein, and the number of competing structures. More generally, this work illustrates how turnover allows biological populations with various competition strengths to coexist despite resource constraints.
Collapse
Affiliation(s)
- Christophe Guérin
- Cytomorpholab, Laboratoire de Physiologie Cellulaire and Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, 17 avenue des Martyrs, 38054 Grenoble, France
| | - Anne-Betty N'Diaye
- Cytomorpholab, Laboratoire de Physiologie Cellulaire and Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, 17 avenue des Martyrs, 38054 Grenoble, France
| | - Laurène Gressin
- Cytomorpholab, Laboratoire de Physiologie Cellulaire and Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, 17 avenue des Martyrs, 38054 Grenoble, France
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences and Department of Biology, New York University, 251 Mercer Street, New York, NY 10012, USA
| | - Manuel Théry
- Cytomorpholab, Institut Chimie Biologie Innovation, Institut Pierre-Gilles de Gennes, Université Paris Sciences et Lettres, CEA, ESPCI, 6 rue Jean Calvin, 75005 Paris, France.
| | - Laurent Blanchoin
- Cytomorpholab, Laboratoire de Physiologie Cellulaire and Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, 17 avenue des Martyrs, 38054 Grenoble, France; Cytomorpholab, Institut Chimie Biologie Innovation, Institut Pierre-Gilles de Gennes, Université Paris Sciences et Lettres, CEA, ESPCI, 6 rue Jean Calvin, 75005 Paris, France.
| | - Alexandra Colin
- Cytomorpholab, Laboratoire de Physiologie Cellulaire and Végétale, Interdisciplinary Research Institute of Grenoble, University of Grenoble-Alpes, CEA, CNRS, INRA, 17 avenue des Martyrs, 38054 Grenoble, France.
| |
Collapse
|
2
|
Gadsby JR, Ioannou PS, Butler R, Mason J, Smith AJ, Dobramysl U, Chin SE, Dobson C, Gallop JL. The open to closed D-loop conformational switch determines length in filopodia-like actin bundles. Biochem J 2024; 481:1977-1995. [PMID: 39621444 DOI: 10.1042/bcj20240367] [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: 06/27/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/24/2024]
Abstract
Filopodia, microspikes and cytonemes are implicated in sensing the environment and in dissemination of morphogens, organelles and pathogens across tissues. Their major structural component is parallel bundles of actin filaments that assemble from the cell membrane. Whilst the length of filopodia is central to their function, it is not known how their lengths are determined by actin bundle dynamics. Here, we identified a set of monoclonal antibodies that lengthen filopodia-like structures formed in a cell-free reconstitution system, and used them to uncover a key molecular switch governing length regulation. Using immunolabelling, enzyme-linked immunosorbent assays, immunoprecipitation and immunoblock experiments, we identified four antibodies that lengthen actin bundles by selectively binding the open DNase 1-binding loop (D-loop) of actin filaments. The antibodies inhibit actin disassembly and their effects can be alleviated by providing additional actin or cofilin. This work indicates that maintaining an open state of the actin filament D-loop is a mechanism of generating long filopodia-like actin bundles.
Collapse
Affiliation(s)
- Jonathan R Gadsby
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Pantelis Savvas Ioannou
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Richard Butler
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
| | - Julia Mason
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Alison J Smith
- Biologics Engineering, Oncology R&D, AstraZeneca, Cambridge, U.K
| | - Ulrich Dobramysl
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, U.K
| | - Stacey E Chin
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, U.K
| | - Claire Dobson
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, U.K
| | - Jennifer L Gallop
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, U.K
| |
Collapse
|
3
|
Li T, Song Y, Wei L, Song X, Duan R. Disulfidptosis: a novel cell death modality induced by actin cytoskeleton collapse and a promising target for cancer therapeutics. Cell Commun Signal 2024; 22:491. [PMID: 39394612 PMCID: PMC11470700 DOI: 10.1186/s12964-024-01871-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024] Open
Abstract
Disulfidptosis is a novel discovered form of programmed cell death (PCD) that diverges from apoptosis, necroptosis, ferroptosis, and cuproptosis, stemming from disulfide stress-induced cytoskeletal collapse. In cancer cells exhibiting heightened expression of the solute carrier family 7 member 11 (SLC7A11), excessive cystine importation and reduction will deplete nicotinamide adenine dinucleotide phosphate (NADPH) under glucose deprivation, followed by an increase in intracellular disulfide stress and aberrant disulfide bond formation within actin networks, ultimately culminating in cytoskeletal collapse and disulfidptosis. Disulfidptosis involves crucial physiological processes in eukaryotic cells, such as cystine and glucose uptake, NADPH metabolism, and actin dynamics. The Rac1-WRC pathway-mediated actin polymerization is also implicated in this cell death due to its contribution to disulfide bond formation. However, the precise mechanisms underlying disulfidptosis and its role in tumors are not well understood. This is probably due to the multifaceted functionalities of SLC7A11 within cells and the complexities of the downstream pathways driving disulfidptosis. This review describes the critical roles of SLC7A11 in cells and summarizes recent research advancements in the potential pathways of disulfidptosis. Moreover, the less-studied aspects of this newly discovered cell death process are highlighted to stimulate further investigations in this field.
Collapse
Affiliation(s)
- Tianyi Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Song
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Lijuan Wei
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Xiangyi Song
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Ruifeng Duan
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China.
| |
Collapse
|
4
|
Lambert C, Karger M, Steffen A, Tang Y, Döring H, Stradal TEB, Lappalainen P, Faix J, Bieling P, Rottner K. Differential interference with actin-binding protein function by acute Cytochalasin B. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.611976. [PMID: 39372773 PMCID: PMC11451763 DOI: 10.1101/2024.09.11.611976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Dynamic actin filament remodeling is crucial for a plethora of fundamental cell biological processes, ranging from cell division and migration to cell communication, intracellular trafficking or tissue development. Cytochalasin B and -D are fungal secondary metabolites frequently used for interference with such processes. Although generally assumed to block actin filament polymerization at their rapidly growing barbed ends and compete with regulators at these sites, our molecular understanding of their precise effects in dynamic actin structures is scarce. Here we combine live cell imaging and analysis of fluorescent actin-binding protein dynamics with acute treatment of lamellipodia in migrating cells with cytochalasin B. Our results show that in spite of an abrupt halt of lamellipodium protrusion, cytochalasin B affects various actin filament barbed end-binding proteins in a differential fashion. Cytochalasin B enhances instead of diminishes the accumulation of prominent barbed end-binding factors such as Ena/VASP family proteins and heterodimeric capping protein (CP) in the lamellipodium. Similar results were obtained with cytochalasin D. All these effects are highly specific, as cytochalasin-induced VASP accumulation requires the presence of CP, but not vice versa , and coincides with abrogation of both actin and VASP turnover. Cytochalasin B can also increase apparent barbed end interactions with the actin-binding β-tentacle of CP and partially mimic its Arp2/3 complex-promoting activity in the lamellipodium. In conclusion, our results reveal a new spectrum of cytochalasin activities on barbed end-binding factors, with important implications for the interpretation of their effects on dynamic actin structures.
Collapse
|
5
|
Chang CY, Pearce G, Betaneli V, Kapustsenka T, Hosseini K, Fischer-Friedrich E, Corbeil D, Karbanová J, Taubenberger A, Dahncke B, Rauner M, Furesi G, Perner S, Rost F, Jessberger R. The F-actin bundler SWAP-70 promotes tumor metastasis. Life Sci Alliance 2024; 7:e202302307. [PMID: 38760173 PMCID: PMC11101836 DOI: 10.26508/lsa.202302307] [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: 08/07/2023] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/19/2024] Open
Abstract
Dynamic rearrangements of the F-actin cytoskeleton are a hallmark of tumor metastasis. Thus, proteins that govern F-actin rearrangements are of major interest for understanding metastasis and potential therapies. We hypothesized that the unique F-actin binding and bundling protein SWAP-70 contributes importantly to metastasis. Orthotopic, ectopic, and short-term tail vein injection mouse breast and lung cancer models revealed a strong positive dependence of lung and bone metastasis on SWAP-70. Breast cancer cell growth, migration, adhesion, and invasion assays revealed SWAP-70's key role in these metastasis-related cell features and the requirement for SWAP-70 to bind F-actin. Biophysical experiments showed that tumor cell stiffness and deformability are negatively modulated by SWAP-70. Together, we present a hitherto undescribed, unique F-actin modulator as an important contributor to tumor metastasis.
Collapse
Affiliation(s)
- Chao-Yuan Chang
- Institute for Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Glen Pearce
- Institute for Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Viktoria Betaneli
- Institute for Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Tatsiana Kapustsenka
- Institute for Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kamran Hosseini
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | | | - Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Dresden, Germany
- Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jana Karbanová
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Dresden, Germany
- Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anna Taubenberger
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Dresden, Germany
- Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Björn Dahncke
- Institute for Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III and Center for Healthy Aging, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Giulia Furesi
- Department of Medicine III and Center for Healthy Aging, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sven Perner
- Institute of Pathology, University of Lübeck and University Hospital Schleswig-Holstein, Lübeck, Germany
- Institute of Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Fabian Rost
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Rolf Jessberger
- Institute for Physiological Chemistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
6
|
Xie K, Wang B, Pang P, Li G, Yang Q, Fang C, Jiang W, Feng Y, Ma H. A novel disulfidptosis-related prognostic gene signature and experimental validation identify ACTN4 as a novel therapeutic target in lung adenocarcinoma. Cancer Biomark 2024:CBM230276. [PMID: 38517776 DOI: 10.3233/cbm-230276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a prevalent form of malignancy globally. Disulfidptosis is novel programmed cell death pathway based on disulfide proteins, may have a positive impact on the development of LUAD treatment strategies. OBJECTIVE To investigate the impact of disulfidptosis-related genes (DRGs) on the prognosis of LUAD, developed a risk model to facilitate the diagnosis and prognostication of patients. We also explored ACTN4 (DRGs) as a new therapeutic biomarker for LUAD. METHODS We investigated the expression patterns of DRGs in both LUAD and noncancerous tissues. To assess the prognostic value of the DRGs, we developed risk models through univariate Cox analysis and lasso regression. The expression and function of ACTN4 was evaluated by qRT-PCR, immunohistochemistry and in vitro experiments. The TIMER examined the association between ACTN4 expression and immune infiltration in LUAD. RESULTS Ten differentially expressed DRGs were identified. And ACTN4 was identified as potential risk factors through univariate Cox regression analysis (P< 0.05). ACTN4 expression and riskscore were used to construct a risk model to predict overall survival in LUAD, and high-risk demonstrated a significantly higher mortality rate compared to the low-risk cohort. qRT-PCR and immunohistochemistry assays indicated ACTN4 was upregulated in LUAD, and the upregulation was associated with clinicopathologic features. In vitro experiments showed the knockdown of ACTN4 expression inhibited the proliferation in LUAD cells. The TIMER analysis demonstrated a correlation between the expression of ACTN4 and the infiltration of diverse immune cells. Elevated ACTN4 expression was associated with a reduction in memory B cell count. Additionally, the ACTN4 expression was associated with m6A modification genes. CONCLUSIONS Our study introduced a prognostic model based on DRGs, which could forecast the prognosis of patients with LUAD. The biomarker ACTN4 exhibits promise for the diagnosis and management of LUAD, given its correlation with tumor immune infiltration and m6A modification.
Collapse
Affiliation(s)
- Kai Xie
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bin Wang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Pei Pang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Guangbin Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qianqian Yang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chen Fang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wei Jiang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yu Feng
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Haitao Ma
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
7
|
Goode BL, Eskin J, Shekhar S. Mechanisms of actin disassembly and turnover. J Cell Biol 2023; 222:e202309021. [PMID: 37948068 PMCID: PMC10638096 DOI: 10.1083/jcb.202309021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Cellular actin networks exhibit a wide range of sizes, shapes, and architectures tailored to their biological roles. Once assembled, these filamentous networks are either maintained in a state of polarized turnover or induced to undergo net disassembly. Further, the rates at which the networks are turned over and/or dismantled can vary greatly, from seconds to minutes to hours or even days. Here, we review the molecular machinery and mechanisms employed in cells to drive the disassembly and turnover of actin networks. In particular, we highlight recent discoveries showing that specific combinations of conserved actin disassembly-promoting proteins (cofilin, GMF, twinfilin, Srv2/CAP, coronin, AIP1, capping protein, and profilin) work in concert to debranch, sever, cap, and depolymerize actin filaments, and to recharge actin monomers for new rounds of assembly.
Collapse
Affiliation(s)
- Bruce L. Goode
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA
| | - Julian Eskin
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA
| | - Shashank Shekhar
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, GA, USA
| |
Collapse
|
8
|
Suarez C, Winkelman JD, Harker AJ, Ye HJ, McCall PM, Morganthaler AN, Gardel ML, Kovar DR. Reconstitution of the transition from a lamellipodia- to filopodia-like actin network with purified proteins. Eur J Cell Biol 2023; 102:151367. [PMID: 37890285 DOI: 10.1016/j.ejcb.2023.151367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
How cells utilize complex mixtures of actin binding proteins to assemble and maintain functionally diverse actin filament networks with distinct architectures and dynamics within a common cytoplasm is a longstanding question in cell biology. A compelling example of complex and specialized actin structures in cells are filopodia which sense extracellular chemical and mechanical signals to help steer motile cells. Filopodia have distinct actin architecture, composed of long, parallel actin filaments bundled by fascin, which form finger-like membrane protrusions. Elongation of the parallel actin filaments in filopodia can be mediated by two processive actin filament elongation factors, formin and Ena/VASP, which localize to the tips of filopodia. There remains debate as to how the architecture of filopodia are generated, with one hypothesis proposing that filopodia are generated from the lamellipodia, which consists of densely packed, branched actin filaments nucleated by Arp2/3 complex and kept short by capping protein. It remains unclear if different actin filament elongation factors are necessary and sufficient to facilitate the emergence of filopodia with diverse characteristics from a highly dense network of short-branched capped filaments. To address this question, we combined bead motility and micropatterning biomimetic assays with multi-color Total Internal Reflection Fluorescence microscopy imaging, to successfully reconstitute the formation of filopodia-like networks (FLN) from densely-branched lamellipodia-like networks (LLN) with eight purified proteins (actin, profilin, Arp2/3 complex, Wasp pWA, fascin, capping protein, VASP and formin mDia2). Saturating capping protein concentrations inhibit FLN assembly, but the addition of either formin or Ena/VASP differentially rescues the formation of FLN from LLN. Specifically, we found that formin/mDia2-generated FLNs are relatively long and lack capping protein, whereas VASP-generated FLNs are comparatively short and contain capping protein, indicating that the actin elongation factor can affect the architecture and composition of FLN emerging from LLN. Our biomimetic reconstitution systems reveal that formin or VASP are necessary and sufficient to induce the transition from a LLN to a FLN, and establish robust in vitro platforms to investigate FLN assembly mechanisms.
Collapse
Affiliation(s)
- Cristian Suarez
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA.
| | - Jonathan D Winkelman
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Alyssa J Harker
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Hannah J Ye
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Patrick M McCall
- Department of Physics, The University of Chicago, Chicago, IL 60637, USA; James Franck Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Alisha N Morganthaler
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Margaret L Gardel
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Department of Physics, The University of Chicago, Chicago, IL 60637, USA; James Franck Institute, The University of Chicago, Chicago, IL 60637, USA; Pritzker School for Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
9
|
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
|
10
|
Sirotkin V. Cappin' or formin': Formin and capping protein competition for filament ends shapes actin networks. J Cell Biol 2023; 222:e202302009. [PMID: 36928466 PMCID: PMC10039713 DOI: 10.1083/jcb.202302009] [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] [Indexed: 03/18/2023] Open
Abstract
How cells assemble distinct actin networks from shared cytoplasmic components remains an important unresolved question. In this issue, Wirshing et al. (2023. J. Cell Biol.https://doi.org/10.1083/jcb.202209105) demonstrate how capping protein and formin competition for actin filament barbed ends controls the assembly of branched and linear actin networks.
Collapse
Affiliation(s)
- Vladimir Sirotkin
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
11
|
Hein JI, Scholz J, Körber S, Kaufmann T, Faix J. Unleashed Actin Assembly in Capping Protein-Deficient B16-F1 Cells Enables Identification of Multiple Factors Contributing to Filopodium Formation. Cells 2023; 12:cells12060890. [PMID: 36980231 PMCID: PMC10047565 DOI: 10.3390/cells12060890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
Background: Filopodia are dynamic, finger-like actin-filament bundles that overcome membrane tension by forces generated through actin polymerization at their tips to allow extension of these structures a few microns beyond the cell periphery. Actin assembly of these protrusions is regulated by accessory proteins including heterodimeric capping protein (CP) or Ena/VASP actin polymerases to either terminate or promote filament growth. Accordingly, the depletion of CP in B16-F1 melanoma cells was previously shown to cause an explosive formation of filopodia. In Ena/VASP-deficient cells, CP depletion appeared to result in ruffling instead of inducing filopodia, implying that Ena/VASP proteins are absolutely essential for filopodia formation. However, this hypothesis was not yet experimentally confirmed. Methods: Here, we used B16-F1 cells and CRISPR/Cas9 technology to eliminate CP either alone or in combination with Ena/VASP or other factors residing at filopodia tips, followed by quantifications of filopodia length and number. Results: Unexpectedly, we find massive formations of filopodia even in the absence of CP and Ena/VASP proteins. Notably, combined inactivation of Ena/VASP, unconventional myosin-X and the formin FMNL3 was required to markedly impair filopodia formation in CP-deficient cells. Conclusions: Taken together, our results reveal that, besides Ena/VASP proteins, numerous other factors contribute to filopodia formation.
Collapse
Affiliation(s)
| | | | | | | | - Jan Faix
- Correspondence: ; Tel.: +49-511-532-2928
| |
Collapse
|
12
|
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
|
13
|
Bieling P, Rottner K. From WRC to Arp2/3: Collective molecular mechanisms of branched actin network assembly. Curr Opin Cell Biol 2023; 80:102156. [PMID: 36868090 DOI: 10.1016/j.ceb.2023.102156] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 03/05/2023]
Abstract
Branched actin networks have emerged as major force-generating structures driving the protrusions in various distinct cell types and processes, ranging from lamellipodia operating in mesenchymal and epithelial cell migration or tails pushing intracellular pathogens and vesicles to developing spine heads on neurons. Many key molecular features are conserved among all those Arp2/3 complex-containing, branched actin networks. Here, we will review recent progress in our molecular understanding of the core biochemical machinery driving branched actin nucleation, from the generation of filament primers to Arp2/3 activator recruitment, regulation and turnover. Due to the wealth of information on distinct, Arp2/3 network-containing structures, we are largely focusing-in an exemplary fashion-on canonical lamellipodia of mesenchymal cells, which are regulated by Rac GTPases, their downstream effector WAVE Regulatory Complex and its target Arp2/3 complex. Novel insight additionally confirms that WAVE and Arp2/3 complexes regulate or are themselves tuned by additional prominent actin regulatory factors, including Ena/VASP family members and heterodimeric capping protein. Finally, we are considering recent insights into effects exerted by mechanical force, both at the branched network and individual actin regulator level.
Collapse
Affiliation(s)
- Peter Bieling
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany.
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany; Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany.
| |
Collapse
|
14
|
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
|
15
|
Myers KR, Fan Y, McConnell P, Cooper JA, Zheng JQ. Actin capping protein regulates postsynaptic spine development through CPI-motif interactions. Front Mol Neurosci 2022; 15:1020949. [PMID: 36245917 PMCID: PMC9557104 DOI: 10.3389/fnmol.2022.1020949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022] Open
Abstract
Dendritic spines are small actin-rich protrusions essential for the formation of functional circuits in the mammalian brain. During development, spines begin as dynamic filopodia-like protrusions that are then replaced by relatively stable spines containing an expanded head. Remodeling of the actin cytoskeleton plays a key role in the formation and modification of spine morphology, however many of the underlying regulatory mechanisms remain unclear. Capping protein (CP) is a major actin regulating protein that caps the barbed ends of actin filaments, and promotes the formation of dense branched actin networks. Knockdown of CP impairs the formation of mature spines, leading to an increase in the number of filopodia-like protrusions and defects in synaptic transmission. Here, we show that CP promotes the stabilization of dendritic protrusions, leading to the formation of stable mature spines. However, the localization and function of CP in dendritic spines requires interactions with proteins containing a capping protein interaction (CPI) motif. We found that the CPI motif-containing protein Twinfilin-1 (Twf1) also localizes to spines where it plays a role in CP spine enrichment. The knockdown of Twf1 leads to an increase in the density of filopodia-like protrusions and a decrease in the stability of dendritic protrusions, similar to CP knockdown. Finally, we show that CP directly interacts with Shank and regulates its spine accumulation. These results suggest that spatiotemporal regulation of CP in spines not only controls the actin dynamics underlying the formation of stable postsynaptic spine structures, but also plays an important role in the assembly of the postsynaptic apparatus underlying synaptic function.
Collapse
Affiliation(s)
- Kenneth R. Myers
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Yanjie Fan
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Patrick McConnell
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO, United States
| | - John A. Cooper
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO, United States
| | - James Q. Zheng
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
16
|
Fox S, Tran A, Trinkle-Mulcahy L, Copeland JW. Cooperative assembly of filopodia by the formin FMNL2 and I-BAR domain protein IRTKS. J Biol Chem 2022; 298:102512. [PMID: 36259517 PMCID: PMC9579038 DOI: 10.1016/j.jbc.2022.102512] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
Filopodia are long finger-like actin-based structures that project out from the plasma membrane as cells navigate and explore their extracellular environment. The initiation of filopodia formation requires release of tension at the plasma membrane followed by the coordinated assembly of long unbranched actin filaments. Filopodia growth is maintained by a tip complex that promotes actin polymerization and protects the growing barbed ends of the actin fibers from capping proteins. Filopodia growth also depends on additional F-actin bundling proteins to stiffen the actin filaments as well as extension of the membrane sheath projecting from the cell periphery. These activities can be provided by a number of actin-binding and membrane-binding proteins including formins such as formin-like 2 (FMNL2) and FMNL3, and Inverse-Bin-Amphiphysin-Rvs (I-BAR) proteins such as IRTKS and IRSp53, but the specific requirement for these proteins in filopodia assembly is not clear. We report here that IRTKS and IRSp53 are FMNL2-binding proteins. Coexpression of FMNL2 with either I-BAR protein promotes cooperative filopodia assembly. We find IRTKS, but not IRSp53, is required for FMNL2-induced filopodia assembly, and FMNL2 and IRTKS are mutually dependent cofactors in this process. Our results suggest that the primary function for FMNL2 during filopodia assembly is binding to the plasma membrane and that regulation of actin dynamics by its formin homology 2 domain is secondary. From these results, we conclude that FMNL2 initiates filopodia assembly via an unexpected novel mechanism, by bending the plasma membrane to recruit IRTKS and thereby nucleate filopodia assembly.
Collapse
|
17
|
Glutamate Signaling and Filopodiagenesis of Astrocytoma Cells in Brain Cancers: Survey and Questions. Cells 2022; 11:cells11172657. [PMID: 36078065 PMCID: PMC9454653 DOI: 10.3390/cells11172657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022] Open
Abstract
Astrocytes are non-excitable cells in the CNS that can cause life-threatening astrocytoma tumors when they transform to cancerous cells. Perturbed homeostasis of the neurotransmitter glutamate is associated with astrocytoma tumor onset and progression, but the factors that govern this phenomenon are less known. Herein, we review possible mechanisms by which glutamate may act in facilitating the growth of projections in astrocytic cells. This review discusses the similarities and differences between the morphology of astrocytes and astrocytoma cells, and the role that dysregulation in glutamate and calcium signaling plays in the aberrant morphology of astrocytoma cells. Converging reports suggest that ionotropic glutamate receptors and voltage-gated calcium channels expressed in astrocytes may be responsible for the abnormal filopodiagenesis or process extension leading to astrocytoma cells’ infiltration throughout the brain.
Collapse
|
18
|
Song J, Liu C, Li B, Liu L, Zeng L, Ye Z, Mao T, Wu W, Hu B. Tunable Cellular Localization and Extensive Cytoskeleton-Interplay of Reflectins. Front Cell Dev Biol 2022; 10:862011. [PMID: 35813206 PMCID: PMC9259870 DOI: 10.3389/fcell.2022.862011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Reflectin proteins are natural copolymers consisting of repeated canonical domains. They are located in a biophotonic system called Bragg lamellae and manipulate the dynamic structural coloration of iridocytes. Their biological functions are intriguing, but the underlying mechanism is not fully understood. Reflectin A1, A2, B1, and C were found to present distinguished cyto-/nucleoplasmic localization preferences in the work. Comparable intracellular localization was reproduced by truncated reflectin variants, suggesting a conceivable evolutionary order among reflectin proteins. The size-dependent access of reflectin variants into the nucleus demonstrated a potential model of how reflectins get into Bragg lamellae. Moreover, RfA1 was found to extensively interact with the cytoskeleton, including its binding to actin and enrichment at the microtubule organizing center. This implied that the cytoskeleton system plays a fundamental role during the organization and transportation of reflectin proteins. The findings presented here provide evidence to get an in-depth insight into the evolutionary processes and working mechanisms of reflectins, as well as novel molecular tools to achieve tunable intracellular transportation.
Collapse
Affiliation(s)
- Junyi Song
- College of Liberal Arts Science, National University of Defense Technology, Changsha, China
| | - Chuanyang Liu
- College of Liberal Arts Science, National University of Defense Technology, Changsha, China
| | - Baoshan Li
- College of Liberal Arts Science, National University of Defense Technology, Changsha, China
| | - Liangcheng Liu
- College of Liberal Arts Science, National University of Defense Technology, Changsha, China
| | - Ling Zeng
- College of Liberal Arts Science, National University of Defense Technology, Changsha, China
| | - Zonghuang Ye
- College of Liberal Arts Science, National University of Defense Technology, Changsha, China
| | - Ting Mao
- Logistics Center, National University of Defense Technology, Changsha, China
| | - Wenjian Wu
- College of Liberal Arts Science, National University of Defense Technology, Changsha, China
| | - Biru Hu
- College of Liberal Arts Science, National University of Defense Technology, Changsha, China
| |
Collapse
|
19
|
Bashirzadeh Y, Moghimianavval H, Liu AP. Encapsulated actomyosin patterns drive cell-like membrane shape changes. iScience 2022; 25:104236. [PMID: 35521522 PMCID: PMC9061794 DOI: 10.1016/j.isci.2022.104236] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/26/2022] [Accepted: 04/07/2022] [Indexed: 11/21/2022] Open
Abstract
Cell shape changes from locomotion to cytokinesis are, to a large extent, driven by myosin-driven remodeling of cortical actin patterns. Passive crosslinkers such as α-actinin and fascin as well as actin nucleator Arp2/3 complex largely determine actin network architecture and, consequently, membrane shape changes. Here we reconstitute actomyosin networks inside cell-sized lipid bilayer vesicles and show that depending on vesicle size and concentrations of α-actinin and fascin actomyosin networks assemble into ring and aster-like patterns. Anchoring actin to the membrane does not change actin network architecture yet exerts forces and deforms the membrane when assembled in the form of a contractile ring. In the presence of α-actinin and fascin, an Arp2/3 complex-mediated actomyosin cortex is shown to assemble a ring-like pattern at the equatorial cortex followed by myosin-driven clustering and consequently blebbing. An active gel theory unifies a model for the observed membrane shape changes induced by the contractile cortex.
Collapse
Affiliation(s)
- Yashar Bashirzadeh
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Allen P. Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
20
|
Vibration, a treatment for migraine, linked to calpain driven changes in actin cytoskeleton. PLoS One 2022; 17:e0262058. [PMID: 35482731 PMCID: PMC9049534 DOI: 10.1371/journal.pone.0262058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/04/2022] [Indexed: 11/19/2022] Open
Abstract
Understanding how a human cell reacts to external physical stimuli is essential to understanding why vibration can elicit localized pain reduction. Stimulation of epithelial cells with external vibration forces has been shown to change cell shape, particularly in regards to structures involved in non-muscle cell motility. We hypothesized that epithelial cells respond to vibration transduction by altering proteins involved in remodeling cytoskeleton. Epithelial cells were exposed to vibration and assessed by microscopy, cytoskeletal staining, immunoblotting and quantitative RT-PCR. Here, we report that epithelial cell lines exposed to 15 minutes of vibration retract filopodia and concentrate actin at the periphery of the cell. In particular, we show an increased expression of the calcium-dependent, cysteine protease, calpain. The discovery that cell transitions are induced by limited exposure to natural forces, such as vibration, provides a foundation to explain how vibrational treatment helps migraine patients.
Collapse
|
21
|
Structure and function of an atypical homodimeric actin capping protein from the malaria parasite. Cell Mol Life Sci 2022; 79:125. [PMID: 35132495 PMCID: PMC8821504 DOI: 10.1007/s00018-021-04032-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/28/2021] [Accepted: 11/09/2021] [Indexed: 11/27/2022]
Abstract
Apicomplexan parasites, such as Plasmodium spp., rely on an unusual actomyosin motor, termed glideosome, for motility and host cell invasion. The actin filaments are maintained by a small set of essential regulators, which provide control over actin dynamics in the different stages of the parasite life cycle. Actin filament capping proteins (CPs) are indispensable heterodimeric regulators of actin dynamics. CPs have been extensively characterized in higher eukaryotes, but their role and functional mechanism in Apicomplexa remain enigmatic. Here, we present the first crystal structure of a homodimeric CP from the malaria parasite and compare the homo- and heterodimeric CP structures in detail. Despite retaining several characteristics of a canonical CP, the homodimeric Plasmodium berghei (Pb)CP exhibits crucial differences to the canonical heterodimers. Both homo- and heterodimeric PbCPs regulate actin dynamics in an atypical manner, facilitating rapid turnover of parasite actin, without affecting its critical concentration. Homo- and heterodimeric PbCPs show partially redundant activities, possibly to rescue actin filament capping in life cycle stages where the β-subunit is downregulated. Our data suggest that the homodimeric PbCP also influences actin kinetics by recruiting lateral actin dimers. This unusual function could arise from the absence of a β-subunit, as the asymmetric PbCP homodimer lacks structural elements essential for canonical barbed end interactions suggesting a novel CP binding mode. These findings will facilitate further studies aimed at elucidating the precise actin filament capping mechanism in Plasmodium.
Collapse
|
22
|
Carvalho AM, Soares da Costa D, Reis RL, Pashkuleva I. Influence of Hyaluronan Density on the Behavior of Breast Cancer Cells with Different CD44 Expression. Adv Healthc Mater 2022; 11:e2101309. [PMID: 34694735 DOI: 10.1002/adhm.202101309] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/18/2021] [Indexed: 11/10/2022]
Abstract
Molecular gradients are common in biosystems and play an essential role in physiological and pathological processes. During carcinogenesis, for example, hyaluronan (HA) homeostasis is dysregulated by cancer cells and the altered synthesis and degradation processes result in the formation of HA gradients within the tumor microenvironment. Herein, a platform is developed to study the biological role of HA gradient in breast cancer cells. Cells with different aggressiveness and expression of CD44-the main HA receptor usually overexpressed in breast cancers, are selected for this study. The developed platform is compatible with several imaging modalities and allows assessment of cell density, morphology, CD44 expression, and cell motility in a function of HA density. Using high-throughput analysis, it is shown that cells that do not express CD44 do not change along the gradient, while CD44 positive cells respond differently to the HA gradient depending on the level of CD44 expression and HA density. This different response is associated with the activation of different signaling pathways by the CD44-HA interactions.
Collapse
Affiliation(s)
- Ana M. Carvalho
- 3B's Research Group ‐ Biomaterials Biodegradable and Biomimetics Avepark ‐ Parque de Ciência e Tecnologia Zona Industrial da Gandra Barco 4805‐017 Portugal
- ICVS/3B's ‐ PT Government Associate Laboratory University of Minho Braga/Guimarães Portugal
| | - Diana Soares da Costa
- 3B's Research Group ‐ Biomaterials Biodegradable and Biomimetics Avepark ‐ Parque de Ciência e Tecnologia Zona Industrial da Gandra Barco 4805‐017 Portugal
- ICVS/3B's ‐ PT Government Associate Laboratory University of Minho Braga/Guimarães Portugal
| | - Rui L. Reis
- 3B's Research Group ‐ Biomaterials Biodegradable and Biomimetics Avepark ‐ Parque de Ciência e Tecnologia Zona Industrial da Gandra Barco 4805‐017 Portugal
- ICVS/3B's ‐ PT Government Associate Laboratory University of Minho Braga/Guimarães Portugal
| | - Iva Pashkuleva
- 3B's Research Group ‐ Biomaterials Biodegradable and Biomimetics Avepark ‐ Parque de Ciência e Tecnologia Zona Industrial da Gandra Barco 4805‐017 Portugal
- ICVS/3B's ‐ PT Government Associate Laboratory University of Minho Braga/Guimarães Portugal
| |
Collapse
|
23
|
Vyshnava SS, Kanderi DK, Dowlathabad MR. Confocal laser scanning microscopy study of intercellular events in filopodia using 3-mercaptopropoinc acid capped CdSe/ZnS quantum dots. Micron 2022; 153:103200. [PMID: 34973488 DOI: 10.1016/j.micron.2021.103200] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 11/18/2022]
Abstract
Physico-chemical mobility of cells in three dimensions is dependent on the development of filipodia, which is the fundamental instinct for survival and other cellular functions in live cells. Specifically, our present research paper describes the synthesis of 3-Mercaptopropoinc acid (MPA) capped CdSe/ZnS quantum dots (QDs), which are biocompatible and utilized for cellular bioimaging applications. Using the pancreatic cell lines BXCP3 cells, we successfully demonstrated the applicability of MPA-capped QDs for intercellular filopodia imaging. Employing these QDs, we examined the dynamics of filopodia formation in real-time along the Z-axis by using confocal laser microscopy.
Collapse
Affiliation(s)
| | - Dileep Kumar Kanderi
- Department of Microbiology, Sri Krishnadevaraya University, Anantapuram, A.P, India.
| | | |
Collapse
|
24
|
Mehidi A, Kage F, Karatas Z, Cercy M, Schaks M, Polesskaya A, Sainlos M, Gautreau AM, Rossier O, Rottner K, Giannone G. Forces generated by lamellipodial actin filament elongation regulate the WAVE complex during cell migration. Nat Cell Biol 2021; 23:1148-1162. [PMID: 34737443 DOI: 10.1038/s41556-021-00786-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Actin filaments generate mechanical forces that drive membrane movements during trafficking, endocytosis and cell migration. Reciprocally, adaptations of actin networks to forces regulate their assembly and architecture. Yet, a demonstration of forces acting on actin regulators at actin assembly sites in cells is missing. Here we show that local forces arising from actin filament elongation mechanically control WAVE regulatory complex (WRC) dynamics and function, that is, Arp2/3 complex activation in the lamellipodium. Single-protein tracking revealed WRC lateral movements along the lamellipodium tip, driven by elongation of actin filaments and correlating with WRC turnover. The use of optical tweezers to mechanically manipulate functional WRC showed that piconewton forces, as generated by single-filament elongation, dissociated WRC from the lamellipodium tip. WRC activation correlated with its trapping, dwell time and the binding strength at the lamellipodium tip. WRC crosslinking, hindering its mechanical dissociation, increased WRC dwell time and Arp2/3-dependent membrane protrusion. Thus, forces generated by individual actin filaments on their regulators can mechanically tune their turnover and hence activity during cell migration.
Collapse
Affiliation(s)
- Amine Mehidi
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Frieda Kage
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Zeynep Karatas
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Maureen Cercy
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Matthias Schaks
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Anna Polesskaya
- CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Matthieu Sainlos
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Alexis M Gautreau
- CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Olivier Rossier
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Grégory Giannone
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France.
| |
Collapse
|
25
|
Detoxification, Hydrogen Sulphide Metabolism and Wound Healing Are the Main Functions That Differentiate Caecum Protein Expression from Ileum of Week-Old Chicken. Animals (Basel) 2021; 11:ani11113155. [PMID: 34827887 PMCID: PMC8614574 DOI: 10.3390/ani11113155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Although the ileum and caecum represent adjacent parts of the gastrointestinal tract, both compartments differ by function as well as inner environment parameters such as oxygen availability or density of colonising microbiota. As the function of a particular tissue is generally reflected by protein expression, mass spectrometry proteomics was used to characterise expressed proteins of both segments of the gastrointestinal tract. Differentially expressed proteins were identified and grouped according to biological processes specific to both gut compartments. Abstract Sections of chicken gut differ in many aspects, e.g., the passage of digesta (continuous vs. discontinuous), the concentration of oxygen, and the density of colonising microbiota. Using an unbiased LC-MS/MS protocol, we compared protein expression in 18 ileal and 57 caecal tissue samples that originated from 7-day old ISA brown chickens. We found that proteins specific to the ileum were either structural (e.g., 3 actin isoforms, villin, or myosin 1A), or those required for nutrient digestion (e.g., sucrose isomaltase, maltase–glucoamylase, peptidase D) and absorption (e.g., fatty acid-binding protein 2 and 6 or bile acid–CoA:amino acid N-acyltransferase). On the other hand, proteins characteristic of the caecum were involved in sensing and limiting the consequences of oxidative stress (e.g., thioredoxin, peroxiredoxin 6), cell adhesion, and motility associated with wound healing (e.g., fibronectin 1, desmoyokin). These mechanisms are coupled with the activation of mechanisms suppressing the inflammatory response (galectin 1). Rather prominent were also expressions of proteins linked to hydrogen sulphide metabolism in caecum represented by cystathionin beta synthase, selenium-binding protein 1, mercaptopyruvate sulphurtransferase, and thiosulphate sulphurtransferase. Higher mRNA expression of nuclear factor, erythroid 2-like 2, the main oxidative stress transcriptional factor in caecum, further supported our observations.
Collapse
|
26
|
Cheok YY, Tan GMY, Fernandez KC, Chan YT, Lee CYQ, Cheong HC, Looi CY, Vadivelu J, Abdullah S, Wong WF. Podoplanin Drives Motility of Active Macrophage via Regulating Filamin C During Helicobacter pylori Infection. Front Immunol 2021; 12:702156. [PMID: 34707599 PMCID: PMC8543000 DOI: 10.3389/fimmu.2021.702156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/10/2021] [Indexed: 01/12/2023] Open
Abstract
Podoplanin (Pdpn) is a mucin-type transmembrane protein that has been implicated in multiple physiological settings including lymphangiogenesis, platelet aggregation, and cancer metastasis. Here, we reported an absence of Pdpn transcript expression in the resting mouse monocytic macrophages, RAW264.7 cells; intriguingly, a substantial upregulation of Pdpn was observed in activated macrophages following Helicobacter pylori or lipopolysaccharide stimulation. Pdpn-knockout macrophages demonstrated intact phagocytic and intracellular bactericidal activities comparable to wild type but exhibited impaired migration due to attenuated filopodia formation. In contrast, an ectopic expression of Pdpn augmented filopodia protrusion in activated macrophages. NanoString analysis uncovered a close dependency of Filamin C gene on the presence of Pdpn, highlighting an involvement of Filamin C in modulation of actin polymerization activity, which controls cell filopodia formation and migration. In addition, interleukin-1β production was significantly declined in the absence of Pdpn, suggesting a role of Pdpn in orchestrating inflammation during H. pylori infection besides cellular migration. Together, our findings unravel the Pdpn network that modulates movement of active macrophages.
Collapse
Affiliation(s)
- Yi Ying Cheok
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Grace Min Yi Tan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Keith Conrad Fernandez
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yee Teng Chan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Chalystha Yie Qin Lee
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Heng Choon Cheong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Chung Yeng Looi
- School of Bioscience, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Jamuna Vadivelu
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Suhailah Abdullah
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
27
|
Eddy CZ, Raposo H, Manchanda A, Wong R, Li F, Sun B. Morphodynamics facilitate cancer cells to navigate 3D extracellular matrix. Sci Rep 2021; 11:20434. [PMID: 34650167 PMCID: PMC8516896 DOI: 10.1038/s41598-021-99902-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 09/27/2021] [Indexed: 12/23/2022] Open
Abstract
Cell shape is linked to cell function. The significance of cell morphodynamics, namely the temporal fluctuation of cell shape, is much less understood. Here we study the morphodynamics of MDA-MB-231 cells in type I collagen extracellular matrix (ECM). We systematically vary ECM physical properties by tuning collagen concentrations, alignment, and gelation temperatures. We find that morphodynamics of 3D migrating cells are externally controlled by ECM mechanics and internally modulated by Rho/ROCK-signaling. We employ machine learning to classify cell shape into four different morphological phenotypes, each corresponding to a distinct migration mode. As a result, we map cell morphodynamics at mesoscale into the temporal evolution of morphological phenotypes. We characterize the mesoscale dynamics including occurrence probability, dwell time and transition matrix at varying ECM conditions, which demonstrate the complex phenotype landscape and optimal pathways for phenotype transitions. In light of the mesoscale dynamics, we show that 3D cancer cell motility is a hidden Markov process whereby the step size distributions of cell migration are coupled with simultaneous cell morphodynamics. Morphological phenotype transitions also facilitate cancer cells to navigate non-uniform ECM such as traversing the interface between matrices of two distinct microstructures. In conclusion, we demonstrate that 3D migrating cancer cells exhibit rich morphodynamics that is controlled by ECM mechanics, Rho/ROCK-signaling, and regulate cell motility. Our results pave the way to the functional understanding and mechanical programming of cell morphodynamics as a route to predict and control 3D cell motility.
Collapse
Affiliation(s)
- Christopher Z Eddy
- Department of Physics, Oregon State University, Corvallis, OR, 97331, USA
| | - Helena Raposo
- Department of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR, 97331, USA
| | - Aayushi Manchanda
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, OR, 97331, USA
| | - Ryan Wong
- Department of Physics, Oregon State University, Corvallis, OR, 97331, USA
| | - Fuxin Li
- School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, OR, 97331, USA
| | - Bo Sun
- Department of Physics, Oregon State University, Corvallis, OR, 97331, USA.
| |
Collapse
|
28
|
Funk J, Merino F, Schaks M, Rottner K, Raunser S, Bieling P. A barbed end interference mechanism reveals how capping protein promotes nucleation in branched actin networks. Nat Commun 2021; 12:5329. [PMID: 34504078 PMCID: PMC8429771 DOI: 10.1038/s41467-021-25682-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 08/18/2021] [Indexed: 12/26/2022] Open
Abstract
Heterodimeric capping protein (CP/CapZ) is an essential factor for the assembly of branched actin networks, which push against cellular membranes to drive a large variety of cellular processes. Aside from terminating filament growth, CP potentiates the nucleation of actin filaments by the Arp2/3 complex in branched actin networks through an unclear mechanism. Here, we combine structural biology with in vitro reconstitution to demonstrate that CP not only terminates filament elongation, but indirectly stimulates the activity of Arp2/3 activating nucleation promoting factors (NPFs) by preventing their association to filament barbed ends. Key to this function is one of CP's C-terminal "tentacle" extensions, which sterically masks the main interaction site of the terminal actin protomer. Deletion of the β tentacle only modestly impairs capping. However, in the context of a growing branched actin network, its removal potently inhibits nucleation promoting factors by tethering them to capped filament ends. End tethering of NPFs prevents their loading with actin monomers required for activation of the Arp2/3 complex and thus strongly inhibits branched network assembly both in cells and reconstituted motility assays. Our results mechanistically explain how CP couples two opposed processes-capping and nucleation-in branched actin network assembly.
Collapse
Affiliation(s)
- Johanna Funk
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Felipe Merino
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany.,Department of Protein Evolution, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Matthias Schaks
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| | - Peter Bieling
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| |
Collapse
|
29
|
Kadzik RS, Homa KE, Kovar DR. F-Actin Cytoskeleton Network Self-Organization Through Competition and Cooperation. Annu Rev Cell Dev Biol 2021; 36:35-60. [PMID: 33021819 DOI: 10.1146/annurev-cellbio-032320-094706] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Many fundamental cellular processes such as division, polarization, endocytosis, and motility require the assembly, maintenance, and disassembly of filamentous actin (F-actin) networks at specific locations and times within the cell. The particular function of each network is governed by F-actin organization, size, and density as well as by its dynamics. The distinct characteristics of different F-actin networks are determined through the coordinated actions of specific sets of actin-binding proteins (ABPs). Furthermore, a cell typically assembles and uses multiple F-actin networks simultaneously within a common cytoplasm, so these networks must self-organize from a common pool of shared globular actin (G-actin) monomers and overlapping sets of ABPs. Recent advances in multicolor imaging and analysis of ABPs and their associated F-actin networks in cells, as well as the development of sophisticated in vitro reconstitutions of networks with ensembles of ABPs, have allowed the field to start uncovering the underlying principles by which cells self-organize diverse F-actin networks to execute basic cellular functions.
Collapse
Affiliation(s)
- Rachel S Kadzik
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois 60637, USA; , .,Department of Molecular BioSciences, Northwestern University, Evanston, Illinois 60208, USA;
| | - Kaitlin E Homa
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois 60637, USA; ,
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois 60637, USA; , .,Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois 60637, USA
| |
Collapse
|
30
|
Actin Cytoskeleton Role in the Maintenance of Neuronal Morphology and Long-Term Memory. Cells 2021; 10:cells10071795. [PMID: 34359964 PMCID: PMC8305626 DOI: 10.3390/cells10071795] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022] Open
Abstract
Evidence indicates that long-term memory formation creates long-lasting changes in neuronal morphology within a specific neuronal network that forms the memory trace. Dendritic spines, which include most of the excitatory synapses in excitatory neurons, are formed or eliminated by learning. These changes may be long-lasting and correlate with memory strength. Moreover, learning-induced changes in the morphology of existing spines can also contribute to the formation of the neuronal network that underlies memory. Altering spines morphology after memory consolidation can erase memory. These observations strongly suggest that learning-induced spines modifications can constitute the changes in synaptic connectivity within the neuronal network that form memory and that stabilization of this network maintains long-term memory. The formation and elimination of spines and other finer morphological changes in spines are mediated by the actin cytoskeleton. The actin cytoskeleton forms networks within the spine that support its structure. Therefore, it is believed that the actin cytoskeleton mediates spine morphogenesis induced by learning. Any long-lasting changes in the spine morphology induced by learning require the preservation of the spine actin cytoskeleton network to support and stabilize the spine new structure. However, the actin cytoskeleton is highly dynamic, and the turnover of actin and its regulatory proteins that determine and support the actin cytoskeleton network structure is relatively fast. Molecular models, suggested here, describe ways to overcome the dynamic nature of the actin cytoskeleton and the fast protein turnover and to support an enduring actin cytoskeleton network within the spines, spines stability and long-term memory. These models are based on long-lasting changes in actin regulatory proteins concentrations within the spine or the formation of a long-lasting scaffold and the ability for its recurring rebuilding within the spine. The persistence of the actin cytoskeleton network within the spine is suggested to support long-lasting spine structure and the maintenance of long-term memory.
Collapse
|
31
|
Pocaterra A, Scattolin G, Romani P, Ament C, Ribback S, Chen X, Evert M, Calvisi DF, Dupont S. Fascin1 empowers YAP mechanotransduction and promotes cholangiocarcinoma development. Commun Biol 2021; 4:763. [PMID: 34155338 PMCID: PMC8217270 DOI: 10.1038/s42003-021-02286-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 06/01/2021] [Indexed: 12/14/2022] Open
Abstract
Mechanical forces control cell behavior, including cancer progression. Cells sense forces through actomyosin to activate YAP. However, the regulators of F-actin dynamics playing relevant roles during mechanostransduction in vitro and in vivo remain poorly characterized. Here we identify the Fascin1 F-actin bundling protein as a factor that sustains YAP activation in response to ECM mechanical cues. This is conserved in the mouse liver, where Fascin1 regulates YAP-dependent phenotypes, and in human cholangiocarcinoma cell lines. Moreover, this is relevant for liver tumorigenesis, because Fascin1 is required in the AKT/NICD cholangiocarcinogenesis model and it is sufficient, together with AKT, to induce cholangiocellular lesions in mice, recapitulating genetic YAP requirements. In support of these findings, Fascin1 expression in human intrahepatic cholangiocarcinomas strongly correlates with poor patient prognosis. We propose that Fascin1 represents a pro-oncogenic mechanism that can be exploited during intrahepatic cholangiocarcinoma development to overcome a mechanical tumor-suppressive environment.
Collapse
Affiliation(s)
- Arianna Pocaterra
- Department of Molecular Medicine, University of Padua Medical School, Padua, Italy
| | - Gloria Scattolin
- Department of Molecular Medicine, University of Padua Medical School, Padua, Italy
| | - Patrizia Romani
- Department of Molecular Medicine, University of Padua Medical School, Padua, Italy
| | - Cindy Ament
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California, USA
| | - Matthias Evert
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Diego F Calvisi
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Sirio Dupont
- Department of Molecular Medicine, University of Padua Medical School, Padua, Italy.
| |
Collapse
|
32
|
Mylvaganam S, Freeman SA, Grinstein S. The cytoskeleton in phagocytosis and macropinocytosis. Curr Biol 2021; 31:R619-R632. [PMID: 34033794 DOI: 10.1016/j.cub.2021.01.036] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cells of the innate immune system, notably macrophages, neutrophils and dendritic cells, perform essential antimicrobial and homeostatic functions. These functions rely on the dynamic surveillance of the environment supported by the formation of elaborate membrane protrusions. Such protrusions - pseudopodia, lamellipodia and filopodia - facilitate the sampling of the surrounding fluid by macropinocytosis, as well as the engulfment of particulates by phagocytosis. Both processes entail extreme plasma membrane deformations that require the coordinated rearrangement of cytoskeletal polymers, which exert protrusive force and drive membrane coalescence and scission. The resulting vacuolar compartments undergo pronounced remodeling and ultimate resolution by mechanisms that also involve the cytoskeleton. Here, we describe the regulation and functions of cytoskeletal assembly and remodeling during macropinocytosis and phagocytosis.
Collapse
Affiliation(s)
- Sivakami Mylvaganam
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Spencer A Freeman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Sergio Grinstein
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
33
|
The multiple roles of actin-binding proteins at invadopodia. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021. [PMID: 33962752 DOI: 10.1016/bs.ircmb.2021.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Invadopodia are actin-rich membrane protrusions that facilitate cancer cell dissemination by focusing on proteolytic activity and clearing paths for migration through physical barriers, such as basement membranes, dense extracellular matrices, and endothelial cell junctions. Invadopodium formation and activity require spatially and temporally regulated changes in actin filament organization and dynamics. About three decades of research have led to a remarkable understanding of how these changes are orchestrated by sequential recruitment and coordinated activity of different sets of actin-binding proteins. In this chapter, we provide an update on the roles of the actin cytoskeleton during the main stages of invadopodium development with a particular focus on actin polymerization machineries and production of pushing forces driving extracellular matrix remodeling.
Collapse
|
34
|
Goodman KE, Hare JT, Khamis ZI, Hua T, Sang QXA. Exposure of Human Lung Cells to Polystyrene Microplastics Significantly Retards Cell Proliferation and Triggers Morphological Changes. Chem Res Toxicol 2021; 34:1069-1081. [PMID: 33720697 DOI: 10.1021/acs.chemrestox.0c00486] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Microplastics in the environment produced by decomposition of globally increasing waste plastics have become a dominant component of both water and air pollution. To examine the potential toxicological effects of microplastics on human cells, the cultured human alveolar A549 cells were exposed to polystyrene microplastics (PS-MPs) of 1 and 10 μm diameter as a model of the environmental contaminants. Both sizes caused a significant reduction in cell proliferation but exhibited little cytotoxicity, as measured by the maintenance of cell viabilities determined by trypan blue staining and by Calcein-AM staining. The cell viabilities did not drop below 93% even at concentrations of PS-MPs as high as 100 μg/mL. Despite these high viabilities, further assays revealed a population level decrease in metabolic activity parallel in time with a dramatic decrease in proliferation rate in PS-MP exposed cells. Furthermore, phase contrast imaging of live cells at 72 h revealed major changes in the morphology of cells exposed to microplastics, as well as the uptake of multiple 1 μm PS-MPs into the cells. Confocal fluorescent microscopy at 24 h of exposure confirmed the incorporation of 1 μm PS-MPs. These disturbances at the proliferative and cytoskeletal levels of human cells lead us to propose that airborne polystyrene microplastics may have toxicologic consequences. This is the first report of exposure of human cells to an environmental contaminant resulting in the dual effects of inhibition of cell proliferation and major changes in cell morphology. Our results make clear that human exposure to microplastic pollution has significant consequence and potential for harm to humans.
Collapse
Affiliation(s)
- Kerestin E Goodman
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Joan T Hare
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Zahraa I Khamis
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States.,Department of Chemistry and Biochemistry, Lebanese University, Faculty of Sciences, Hadath-Beirut, Lebanon
| | - Timothy Hua
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States.,Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida 32306, United States
| |
Collapse
|
35
|
Takeda S, Koike R, Fujiwara I, Narita A, Miyata M, Ota M, Maéda Y. Structural Insights into the Regulation of Actin Capping Protein by Twinfilin C-terminal Tail. J Mol Biol 2021; 433:166891. [PMID: 33639213 DOI: 10.1016/j.jmb.2021.166891] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 12/19/2022]
Abstract
Twinfilin is a conserved actin regulator that interacts with actin capping protein (CP) via C terminus residues (TWtail) that exhibits sequence similarity with the CP interaction (CPI) motif of CARMIL. Here we report the crystal structure of TWtail in complex with CP. Our structure showed that although TWtail and CARMIL CPI bind CP to an overlapping surface via their middle regions, they exhibit different CP-binding modes at both termini. Consequently, TWtail and CARMIL CPI restrict the CP in distinct conformations of open and closed forms, respectively. Interestingly, V-1, which targets CP away from the TWtail binding site, also favors the open-form CP. Consistently, TWtail forms a stable ternary complex with CP and V-1, a striking contrast to CARMIL CPI, which rapidly dissociates V-1 from CP. Our results demonstrate that TWtail is a unique CP-binding motif that regulates CP in a manner distinct from CARMIL CPI.
Collapse
Affiliation(s)
- Shuichi Takeda
- Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan.
| | - Ryotaro Koike
- Graduate School of Informatics, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Ikuko Fujiwara
- Graduate School of Science, Osaka City University, Osaka, Osaka 558-8585, Japan
| | - Akihiro Narita
- Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Makoto Miyata
- Graduate School of Science, Osaka City University, Osaka, Osaka 558-8585, Japan; The OCU Advanced Research Institute for Natural Science and Technology (OCARINA), Osaka City University, Osaka, Osaka 558-8585, Japan
| | - Motonori Ota
- Graduate School of Informatics, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Yuichiro Maéda
- Graduate School of Informatics, Nagoya University, Nagoya, Aichi 464-8601, Japan
| |
Collapse
|
36
|
Twinfilin uncaps filament barbed ends to promote turnover of lamellipodial actin networks. Nat Cell Biol 2021; 23:147-159. [PMID: 33558729 DOI: 10.1038/s41556-020-00629-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 12/21/2020] [Indexed: 01/18/2023]
Abstract
Coordinated polymerization of actin filaments provides force for cell migration, morphogenesis and endocytosis. Capping protein (CP) is a central regulator of actin dynamics in all eukaryotes. It binds to actin filament (F-actin) barbed ends with high affinity and slow dissociation kinetics to prevent filament polymerization and depolymerization. However, in cells, CP displays remarkably rapid dynamics within F-actin networks, but the underlying mechanism remains unclear. Here, we report that the conserved cytoskeletal regulator twinfilin is responsible for CP's rapid dynamics and specific localization in cells. Depletion of twinfilin led to stable association between CP and cellular F-actin arrays, as well as to its retrograde movement throughout leading-edge lamellipodia. These were accompanied by diminished F-actin turnover rates. In vitro single-filament imaging approaches revealed that twinfilin directly promotes dissociation of CP from filament barbed ends, while enabling subsequent filament depolymerization. These results uncover a bipartite mechanism that controls how actin cytoskeleton-mediated forces are generated in cells.
Collapse
|
37
|
Liu S, Trupiano MX, Simon J, Guo J, Anton ES. The essential role of primary cilia in cerebral cortical development and disorders. Curr Top Dev Biol 2021; 142:99-146. [PMID: 33706927 DOI: 10.1016/bs.ctdb.2020.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Primary cilium, first described in the 19th century in different cell types and organisms by Alexander Ecker, Albert Kolliker, Aleksandr Kowalevsky, Paul Langerhans, and Karl Zimmermann (Ecker, 1844; Kolliker, 1854; Kowalevsky, 1867; Langerhans, 1876; Zimmermann, 1898), play an essential modulatory role in diverse aspects of nervous system development and function. The primary cilium, sometimes referred to as the cell's 'antennae', can receive wide ranging inputs from cellular milieu, including morphogens, growth factors, neuromodulators, and neurotransmitters. Its unique structural and functional organization bequeaths it the capacity to hyper-concentrate signaling machinery in a restricted cellular domain approximately one-thousandth the volume of cell soma. Thus enabling it to act as a signaling hub that integrates diverse developmental and homestatic information from cellular milieu to regulate the development and function of neural cells. Dysfunction of primary cilia contributes to the pathophysiology of several brain malformations, intellectual disabilities, epilepsy, and psychiatric disorders. This review focuses on the most essential contributions of primary cilia to cerebral cortical development and function, in the context of neurodevelopmental disorders and malformations. It highlights the recent progress made in identifying the mechanisms underlying primary cilia's role in cortical progenitors, neurons and glia, in health and disease. A future challenge will be to translate these insights and advances into effective clinical treatments for ciliopathies.
Collapse
Affiliation(s)
- Siling Liu
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Mia X Trupiano
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Jeremy Simon
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Jiami Guo
- Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States.
| |
Collapse
|
38
|
Bellani C, Yue K, Flaig F, Hébraud A, Ray P, Annabi N, Selistre de Araújo HS, Branciforti MC, Minarelli Gaspar AM, Shin SR, Khademhosseini A, Schlatter G. Suturable elastomeric tubular grafts with patterned porosity for rapid vascularization of 3D constructs. Biofabrication 2021; 13. [PMID: 33482658 DOI: 10.1088/1758-5090/abdf1d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/22/2021] [Indexed: 12/11/2022]
Abstract
Vascularization is considered to be one of the key challenges in engineering functional 3D tissues. Engineering suturable vascular grafts containing pores with diameter of several tens of microns in tissue engineered constructs may provide an instantaneous blood perfusion through the grafts improving cell infiltration and thus, allowing rapid vascularization and vascular branching. The aim of this work was to develop suturable tubular scaffolds to be integrated in biofabricated constructs, enabling the direct connection of the biofabricated construct with the host blood stream, providing an immediate blood flow inside the construct. Here, tubular grafts with customizable shapes (tubes, Y-shape capillaries) and controlled diameter ranging from several hundreds of microns to few mm are fabricated based on poly(glycerol sebacate) (PGS) / poly(vinyl alcohol) (PVA) electrospun scaffolds. Furthermore, a network of pore channels of diameter in the order of 100 µm was machined by laser femtosecond ablation in the tube wall. Both non-machined and laser machined tubular scaffolds elongated more than 100% of their original size have shown suture retention, being 5.85 and 3.96 N/mm2 respectively. To demonstrate the potential of application, the laser machined porous grafts were embedded in gelatin methacryloyl (GelMA) hydrogels, resulting in elastomeric porous tubular graft/GelMA 3D constructs. These constructs were then co-seeded with osteoblast-like cells (MG-63) at the external side of the graft and endothelial cells (HUVEC) inside, forming a bone osteon model. The laser machined pore network allowed an immediate endothelial cell flow towards the osteoblasts enabling the osteoblasts and endothelial cells to interact and form 3D structures. This rapid vascularization approach could be applied, not only for bone tissue regeneration, but also for a variety of tissues and organs.
Collapse
Affiliation(s)
- Caroline Bellani
- University of Sao Paulo, AVENIDA TRABALHADOR SÃO-CARLENSE, 400, Sao Carlos, São Paulo, 13566-590, BRAZIL
| | - Kan Yue
- South China Advanced Institute for Soft Matter Science and Technology, South China University of Technology, 381 Wushan Rd, Guangzhou, Guangdong, 510641, CHINA
| | - Florence Flaig
- ICPEES, University of Strasbourg, 25 rue Bécquerel, Strasbourg, 67087, FRANCE
| | - Anne Hébraud
- ICPEES, 25 rue Bécquerel, Strasbourg, 67087, FRANCE
| | - Pengfei Ray
- Division of Health Sciences and Technology, MIT, 45 Carleton Street, Cambridge, Massachusetts, 02142, UNITED STATES
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, UCLA, 5531 Boelter Hall, Los Angeles, California, CA 90095, UNITED STATES
| | | | - Marcia Cristina Branciforti
- Depatament of Materials Engineering, University of Sao Paulo, AVENIDA TRABALHADOR SÃO-CARLENSE, 400, ARNOLD SCHMITED, SAO CARLOS, Sao Paulo, SAO PAULO, 13566-590, BRAZIL
| | - Ana Maria Minarelli Gaspar
- Department of Morphology, School of Dentistry at Araraquara, Sao Paulo State University Julio de Mesquita Filho, R. Humaitá, 1680, Araraquara, SP, 14801-385, BRAZIL
| | - Su Ryon Shin
- Medicine, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, MA 02115, UNITED STATES
| | - Ali Khademhosseini
- Department of Chemical and Biomolecular Engineering, UCLA, 5531 Boelter Hall, Los Angeles, California, CA 90095, UNITED STATES
| | - Guy Schlatter
- ICPEES, University of Strasbourg, 25 rue Bécquerel, Strasbourg, 67087, FRANCE
| |
Collapse
|
39
|
Role of Actin Cytoskeleton in E-cadherin-Based Cell–Cell Adhesion Assembly and Maintenance. J Indian Inst Sci 2021. [DOI: 10.1007/s41745-020-00214-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
40
|
Pereira De Carvalho B, Chern YJ, He J, Chan CH. The ubiquitin ligase RNF8 regulates Rho GTPases and promotes cytoskeletal changes and motility in triple-negative breast cancer cells. FEBS Lett 2020; 595:241-252. [PMID: 33205415 PMCID: PMC7898409 DOI: 10.1002/1873-3468.13999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/28/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022]
Abstract
The ubiquitin ligase RNF8 is known to induce epithelial-to-mesenchymal (EMT) transition and metastasis in triple-negative breast cancer (TNBC). Besides EMT, Rho GTPases have been shown as key regulators in metastasis. In this study, we investigated the role of RNF8 in regulating Rho GTPases and cell motility. We find that RNF8 knockdown in TNBC cells attenuates the protein and mRNA levels of Ras homolog family member A (RHOA) and cell division cycle 42 (CDC42). We show that the formation of filopodia, focal adhesions, and the association of focal adhesions to stress fibers is impaired upon RNF8 knockdown. Cell migration is significantly inhibited by RNF8 knockdown. Our study suggests a potential novel role for RNF8 in mediating cell migration in TNBC through regulation of the Rho GTPases RHOA and CDC42.
Collapse
Affiliation(s)
| | - Yi-Jye Chern
- Department of Pharmacological Sciences, Stony Brook University, NY, USA
| | - Jiabei He
- Department of Pharmacological Sciences, Stony Brook University, NY, USA
| | - Chia-Hsin Chan
- Department of Pharmacological Sciences, Stony Brook University, NY, USA.,Stony Brook Cancer Center, Stony Brook University, NY, USA
| |
Collapse
|
41
|
Ferraris S, Spriano S, Scalia AC, Cochis A, Rimondini L, Cruz-Maya I, Guarino V, Varesano A, Vineis C. Topographical and Biomechanical Guidance of Electrospun Fibers for Biomedical Applications. Polymers (Basel) 2020; 12:E2896. [PMID: 33287236 PMCID: PMC7761715 DOI: 10.3390/polym12122896] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023] Open
Abstract
Electrospinning is gaining increasing interest in the biomedical field as an eco-friendly and economic technique for production of random and oriented polymeric fibers. The aim of this review was to give an overview of electrospinning potentialities in the production of fibers for biomedical applications with a focus on the possibility to combine biomechanical and topographical stimuli. In fact, selection of the polymer and the eventual surface modification of the fibers allow selection of the proper chemical/biological signal to be administered to the cells. Moreover, a proper design of fiber orientation, dimension, and topography can give the opportunity to drive cell growth also from a spatial standpoint. At this purpose, the review contains a first introduction on potentialities of electrospinning for the obtainment of random and oriented fibers both with synthetic and natural polymers. The biological phenomena which can be guided and promoted by fibers composition and topography are in depth investigated and discussed in the second section of the paper. Finally, the recent strategies developed in the scientific community for the realization of electrospun fibers and for their surface modification for biomedical application are presented and discussed in the last section.
Collapse
Affiliation(s)
- Sara Ferraris
- Department of Applied Science and Technology, Politecnico di Torino, 10129 Torino, Italy;
| | - Silvia Spriano
- Department of Applied Science and Technology, Politecnico di Torino, 10129 Torino, Italy;
| | - Alessandro Calogero Scalia
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases–CAAD, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (A.C.S.); (A.C.); (L.R.)
| | - Andrea Cochis
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases–CAAD, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (A.C.S.); (A.C.); (L.R.)
| | - Lia Rimondini
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases–CAAD, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (A.C.S.); (A.C.); (L.R.)
| | - Iriczalli Cruz-Maya
- Institute for Polymers, Composites and Biomaterials (IPCB), National Research Council of Italy, Mostra d’Oltremare, Pad. 20, V. le J.F. Kennedy 54, 80125 Napoli, Italy; (I.C.-M.); (V.G.)
| | - Vincenzo Guarino
- Institute for Polymers, Composites and Biomaterials (IPCB), National Research Council of Italy, Mostra d’Oltremare, Pad. 20, V. le J.F. Kennedy 54, 80125 Napoli, Italy; (I.C.-M.); (V.G.)
| | - Alessio Varesano
- Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing (STIIMA), National Research Council of Italy (CNR), Corso Giuseppe Pella 16, 13900 Biella, Italy; (A.V.); (C.V.)
| | - Claudia Vineis
- Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing (STIIMA), National Research Council of Italy (CNR), Corso Giuseppe Pella 16, 13900 Biella, Italy; (A.V.); (C.V.)
| |
Collapse
|
42
|
Ali M, Zuzga DS, Pitari GM. Differential Ser phosphorylation of vasodilator-stimulated phosphoprotein regulates colon tumor formation and growth. Life Sci 2020; 264:118671. [PMID: 33129878 DOI: 10.1016/j.lfs.2020.118671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 10/17/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023]
Abstract
AIMS Vasodilator-stimulated phosphoprotein (VASP) controls actin dynamics associated with the malignant phenotype of colorectal tumors. Oncogenic VASP function, in turn, is finely regulated by cyclic nucleotide-dependent phosphorylation of serine (Ser) residues 157 and 239, whose differential expression determines cell survival behavior in colon cancer. However, the role of differential VASP Ser phosphorylation in colorectal carcinogenesis remains unclear. MAIN METHODS Specific VASP phosphomutant constructs were employed to selectively silence Ser157 or Ser239 phosphorylation in human colon carcinoma cells. Cyclic nucleotide-dependent manipulation of VASP Ser phosphorylation was performed with 8-bromoadenosine 3',5'-cyclic adenosine monophosphate (8-Br-cAMP) or 8-chlorophenylthio 3',5'-cyclic guanosine monophosphate (8-CPT-cGMP). Tumorigenic and locomotory phenotypes were examined in vitro with clonogenic and wound healing assays, respectively. Finally, tumor formation and growth were investigated in vivo employing two distinct xenograft models of colorectal cancer. KEY FINDINGS Disruption of VASP Ser157 phosphorylation weakened the clonogenic and migratory abilities of human colon cancer cells, effects mimicked by 8-CPT-cGMP-dependent regulation of VASP Ser239. In contrast, inhibition of VASP Ser239 phosphorylation enhanced cell clonogenicity and migration and was phenocopied by 8-Br-cAMP-dependent regulation of VASP Ser157. Importantly, cancer cells bearing the phosphomutant construct targeting VASP Ser157 decreased, while those with the phosphomutation at Ser239 improved their abilities to establish productive tumor colonies and grow in the peritoneal cavity or subcutaneous tissues of nude mice. SIGNIFICANCE Together, present observations suggest differential VASP Ser phosphorylation is a relevant, targetable molecular event underlying tumor formation and progression in colon cancer.
Collapse
Affiliation(s)
- Mehboob Ali
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA; Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wexner Medical College, The Ohio State University, OH, USA.
| | - David S Zuzga
- Department of Biology, La Salle University, Philadelphia, PA, USA; BioDetego LLC, Philadelphia, PA, USA
| | - Giovanni M Pitari
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA; BioDetego LLC, Philadelphia, PA, USA
| |
Collapse
|
43
|
Santio NM, Vainio V, Hoikkala T, Mung KL, Lång M, Vahakoski R, Zdrojewska J, Coffey ET, Kremneva E, Rainio EM, Koskinen PJ. PIM1 accelerates prostate cancer cell motility by phosphorylating actin capping proteins. Cell Commun Signal 2020; 18:121. [PMID: 32771000 PMCID: PMC7414696 DOI: 10.1186/s12964-020-00618-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/27/2020] [Indexed: 12/22/2022] Open
Abstract
Background The PIM family kinases promote cancer cell survival and motility as well as metastatic growth in various types of cancer. We have previously identified several PIM substrates, which support cancer cell migration and invasiveness. However, none of them are known to regulate cellular movements by directly interacting with the actin cytoskeleton. Here we have studied the phosphorylation-dependent effects of PIM1 on actin capping proteins, which bind as heterodimers to the fast-growing actin filament ends and stabilize them. Methods Based on a phosphoproteomics screen for novel PIM substrates, we have used kinase assays and fluorescence-based imaging techniques to validate actin capping proteins as PIM1 substrates and interaction partners. We have analysed the functional consequences of capping protein phosphorylation on cell migration and adhesion by using wound healing and real-time impedance-based assays. We have also investigated phosphorylation-dependent effects on actin polymerization by analysing the protective role of capping protein phosphomutants in actin disassembly assays. Results We have identified capping proteins CAPZA1 and CAPZB2 as PIM1 substrates, and shown that phosphorylation of either of them leads to increased adhesion and migration of human prostate cancer cells. Phosphorylation also reduces the ability of the capping proteins to protect polymerized actin from disassembly. Conclusions Our data suggest that PIM kinases are able to induce changes in actin dynamics to support cell adhesion and movement. Thus, we have identified a novel mechanism through which PIM kinases enhance motility and metastatic behaviour of cancer cells. Video abstract
Graphical abstract ![]()
Collapse
Affiliation(s)
- Niina M Santio
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Veera Vainio
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Tuuli Hoikkala
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Kwan Long Mung
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Mirka Lång
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Riitta Vahakoski
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Justyna Zdrojewska
- Turku Bioscience, University of Turku and Åbo Akademi University, 20520, Turku, Finland
| | - Eleanor T Coffey
- Turku Bioscience, University of Turku and Åbo Akademi University, 20520, Turku, Finland
| | - Elena Kremneva
- Institute of Biotechnology, University of Helsinki, 00014, Helsinki, Finland
| | - Eeva-Marja Rainio
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland
| | - Päivi J Koskinen
- Section of Physiology and Genetics, Department of Biology, University of Turku, Vesilinnantie 5, FI-20500, Turku, Finland.
| |
Collapse
|
44
|
Alexandrova AY, Chikina AS, Svitkina TM. Actin cytoskeleton in mesenchymal-to-amoeboid transition of cancer cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:197-256. [PMID: 33066874 DOI: 10.1016/bs.ircmb.2020.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During development of metastasis, tumor cells migrate through different tissues and encounter different extracellular matrices. An ability of cells to adapt mechanisms of their migration to these diverse environmental conditions, called migration plasticity, gives tumor cells an advantage over normal cells for long distant dissemination. Different modes of individual cell motility-mesenchymal and amoeboid-are driven by different molecular mechanisms, which largely depend on functions of the actin cytoskeleton that can be modulated in a wide range by cellular signaling mechanisms in response to environmental conditions. Various triggers can switch one motility mode to another, but regulations of these transitions are incompletely understood. However, understanding of the mechanisms driving migration plasticity is instrumental for finding anti-cancer treatment capable to stop cancer metastasis. In this review, we discuss cytoskeletal features, which allow the individually migrating cells to switch between mesenchymal and amoeboid migrating modes, called mesenchymal-to-amoeboid transition (MAT). We briefly describe main characteristics of different cell migration modes, and then discuss the triggering factors that initiate MAT with special attention to cytoskeletal features essential for migration plasticity.
Collapse
Affiliation(s)
- Antonina Y Alexandrova
- Laboratory of Mechanisms of Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia.
| | - Aleksandra S Chikina
- Cell Migration and Invasion and Spatio-Temporal Regulation of Antigen Presentation teams, UMR144/U932 Institut Curie, Paris, France
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
45
|
Cheng KW, Mullins RD. Initiation and disassembly of filopodia tip complexes containing VASP and lamellipodin. Mol Biol Cell 2020; 31:2021-2034. [PMID: 32579429 PMCID: PMC7543071 DOI: 10.1091/mbc.e20-04-0270] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The shapes of many eukaryotic cells depends on the actin cytoskeleton, and changes in actin assembly dynamics underlie many changes in cell shape. Ena/VASP-family actin polymerases, for example, modulate cell shape by accelerating actin filament assembly locally and slowing filament capping. When concentrated into discrete foci at the leading edge, VASP promotes filopodia assembly and forms part of a poorly understood molecular complex that remains associated with growing filopodia tips. Here we identify precursors of this filopodia tip complex in migrating B16F1 cells: small leading-edge clusters of the adaptor protein lamellipodin (Lpd) that subsequently recruit VASP and initiate filopodia formation. Dimerization, membrane association, and VASP binding are all required for lamellipodin to incorporate into filopodia tip complexes, and overexpression of monomeric, membrane-targeted lamellipodin mutants disrupts tip complex assembly. Once formed, tip complexes containing VASP and lamellipodin grow by fusing with each other, but their growth is limited by a size-dependent dynamic instability. Our results demonstrate that assembly and disassembly dynamics of filopodia tip complexes are determined, in part, by a network of multivalent interactions between Ena/VASP proteins, EVH1 ligands, and actin filaments.
Collapse
Affiliation(s)
- Karen W Cheng
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94143
| | - R Dyche Mullins
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94143.,Howard Hughes Medical Institute, University of California, San Francisco, CA 94143
| |
Collapse
|
46
|
Primary Cilia Signaling Promotes Axonal Tract Development and Is Disrupted in Joubert Syndrome-Related Disorders Models. Dev Cell 2020; 51:759-774.e5. [PMID: 31846650 DOI: 10.1016/j.devcel.2019.11.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/08/2019] [Accepted: 11/10/2019] [Indexed: 12/18/2022]
Abstract
Appropriate axonal growth and connectivity are essential for functional wiring of the brain. Joubert syndrome-related disorders (JSRD), a group of ciliopathies in which mutations disrupt primary cilia function, are characterized by axonal tract malformations. However, little is known about how cilia-driven signaling regulates axonal growth and connectivity. We demonstrate that the deletion of related JSRD genes, Arl13b and Inpp5e, in projection neurons leads to de-fasciculated and misoriented axonal tracts. Arl13b deletion disrupts the function of its downstream effector, Inpp5e, and deregulates ciliary-PI3K/AKT signaling. Chemogenetic activation of ciliary GPCR signaling and cilia-specific optogenetic modulation of downstream second messenger cascades (PI3K, AKT, and AC3) commonly regulated by ciliary signaling receptors induce rapid changes in axonal dynamics. Further, Arl13b deletion leads to changes in transcriptional landscape associated with dysregulated PI3K/AKT signaling. These data suggest that ciliary signaling acts to modulate axonal connectivity and that impaired primary cilia signaling underlies axonal tract defects in JSRD.
Collapse
|
47
|
Huang Y, Mao X, van Jaarsveld RH, Shu L, Terhal PA, Jia Z, Xi H, Peng Y, Yan H, Yuan S, Li Q, Wang H, Bellen HJ. Variants in CAPZA2, a member of an F-actin capping complex, cause intellectual disability and developmental delay. Hum Mol Genet 2020; 29:1537-1546. [PMID: 32338762 PMCID: PMC7268783 DOI: 10.1093/hmg/ddaa078] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/29/2020] [Accepted: 04/20/2020] [Indexed: 11/13/2022] Open
Abstract
The actin cytoskeleton is regulated by many proteins including capping proteins that stabilize actin filaments (F-actin) by inhibiting actin polymerization and depolymerization. Here, we report two pediatric probands who carry damaging heterozygous de novo mutations in CAPZA2 (HGNC: 1490) and exhibit neurological symptoms with shared phenotypes including global motor development delay, speech delay, intellectual disability, hypotonia and a history of seizures. CAPZA2 encodes a subunit of an F-actin-capping protein complex (CapZ). CapZ is an obligate heterodimer consisting of α and β heterodimer conserved from yeast to human. Vertebrate genomes contain three α subunits encoded by three different genes and CAPZA2 encodes the α2 subunit. The single orthologue of CAPZA genes in Drosophila is cpa. Loss of cpa leads to lethality in early development and expression of the human reference; CAPZA2 rescues this lethality. However, the two CAPZA2 variants identified in the probands rescue this lethality at lower efficiency than the reference. Moreover, expression of the CAPZA2 variants affects bristle morphogenesis, a process that requires extensive actin polymerization and bundling during development. Taken together, our findings suggest that variants in CAPZA2 lead to a non-syndromic neurodevelopmental disorder in children.
Collapse
Affiliation(s)
- Yan Huang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiao Mao
- National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, China
| | | | - Li Shu
- National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, China
| | - Paulien A Terhal
- Department of Genetics, University Medical Center Utrecht, Utrecht CX 3584, The Netherlands
| | - Zhengjun Jia
- National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, China
| | - Hui Xi
- National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, China
| | - Ying Peng
- National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, China
| | - Huiming Yan
- National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, China
| | - Shan Yuan
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, China
| | - Qibin Li
- Clabee Genomics, Shenzhen, Guangdong 518000, China
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Hua Wang
- National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, China
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
48
|
Gallop J. Filopodia and their links with membrane traffic and cell adhesion. Semin Cell Dev Biol 2020; 102:81-89. [DOI: 10.1016/j.semcdb.2019.11.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 11/14/2019] [Accepted: 11/28/2019] [Indexed: 01/24/2023]
|
49
|
Skruber K, Warp PV, Shklyarov R, Thomas JD, Swanson MS, Henty-Ridilla JL, Read TA, Vitriol EA. Arp2/3 and Mena/VASP Require Profilin 1 for Actin Network Assembly at the Leading Edge. Curr Biol 2020; 30:2651-2664.e5. [PMID: 32470361 DOI: 10.1016/j.cub.2020.04.085] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/09/2020] [Accepted: 04/29/2020] [Indexed: 12/27/2022]
Abstract
Cells have many types of actin structures, which must assemble from a common monomer pool. Yet, it remains poorly understood how monomers are distributed to and shared between different filament networks. Simplified model systems suggest that monomers are limited and heterogeneous, which alters actin network assembly through biased polymerization and internetwork competition. However, less is known about how monomers influence complex actin structures, where different networks competing for monomers overlap and are functionally interdependent. One example is the leading edge of migrating cells, which contains filament networks generated by multiple assembly factors. The leading edge dynamically switches between the formation of different actin structures, such as lamellipodia or filopodia, by altering the balance of these assembly factors' activities. Here, we sought to determine how the monomer-binding protein profilin 1 (PFN1) controls the assembly and organization of actin in mammalian cells. Actin polymerization in PFN1 knockout cells was severely disrupted, particularly at the leading edge, where both Arp2/3 and Mena/VASP-based filament assembly was inhibited. Further studies showed that in the absence of PFN1, Arp2/3 no longer localizes to the leading edge and Mena/VASP is non-functional. Additionally, we discovered that discrete stages of internetwork competition and collaboration between Arp2/3 and Mena/VASP networks exist at different PFN1 concentrations. Low levels of PFN1 caused filopodia to form exclusively at the leading edge, while higher concentrations inhibited filopodia and favored lamellipodia and pre-filopodia bundles. These results demonstrate that dramatic changes to actin architecture can be made simply by modifying PFN1 availability.
Collapse
Affiliation(s)
- Kristen Skruber
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Peyton V Warp
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Rachael Shklyarov
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - James D Thomas
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics, and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics, and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Jessica L Henty-Ridilla
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, NY 13210, USA
| | - Tracy-Ann Read
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Eric A Vitriol
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, Gainesville, FL 32610, USA.
| |
Collapse
|
50
|
Moodley S, Lian EY, Crupi MJF, Hyndman BD, Mulligan LM. RET isoform-specific interaction with scaffold protein Ezrin promotes cell migration and chemotaxis in lung adenocarcinoma. Lung Cancer 2020; 142:123-131. [PMID: 32146264 DOI: 10.1016/j.lungcan.2020.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/09/2020] [Accepted: 02/11/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Increased expression of REarranged during Transfection (RET) kinase is reported in 10-20 % of lung adenocarcinomas (LUAD) and is associated with metastasis and reduced survival. Ezrin is a scaffold protein that promotes protein interactions with the actin cytoskeleton to regulate cell migration and is also associated with invasion and metastasis in cancers. RET isoforms interact with unique combinations of scaffold proteins to promote distinct signaling pathways. We hypothesized that RET isoforms associate distinctly with Ezrin for cytoskeletal reorganization and LUAD cell migration processes. METHODS HCC1833 and A549 LUAD, SH-SY5Y neuroblastoma or HEK-293 cells expressing RET and Ezrin were stimulated with the RET ligand glial cell line-derived neurotrophic factor (GDNF) and treated with RET, Ezrin or Src inhibitors. Co-immunoprecipitation or pull-down assays coupled to immunoblotting were used to investigate protein activation and interactions. Immunofluorescence confocal microscopy assessed LUAD cytoskeletal reorganization and colocalization of RET and Ezrin. Live-cell fluorescence imaging was used to measure cell migration and chemotaxis. RESULTS GDNF promoted activation, interaction and colocalization of RET51 isoform and Ezrin. Inhibition of RET or Src impaired Ezrin interactions with RET and Src. GDNF stimulation enhanced the formation of actin-rich filopodia, in which both RET and Ezrin were enriched, and promoted chemotaxis in LUAD cells. However, inhibition of RET, Src or Ezrin suppressed filopodia formation, reduced colocalization of Ezrin with RET, and impaired cell migration and/ or chemotaxis. We further showed that GDNF-mediated activation of RET and Ezrin promoted RhoA-GTPase activity and signaling of ROCK1 and ROCK2 in LUAD cells. CONCLUSIONS Expression and activation of RET51 mediates unique protein interactions with Ezrin to promote LUAD cell chemotaxis for cancer cell dissemination, which may have implications in LUAD metastatic progression.
Collapse
Affiliation(s)
- Serisha Moodley
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Eric Y Lian
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Mathieu J F Crupi
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Brandy D Hyndman
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Lois M Mulligan
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada.
| |
Collapse
|