1
|
Martin C, Servais L. X-linked myotubular myopathy: an untreated treatable disease. Expert Opin Biol Ther 2025; 25:379-394. [PMID: 40042390 DOI: 10.1080/14712598.2025.2473430] [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: 10/31/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION X-linked myotubular myopathy (XLMTM) is a life-threatening congenital disorder characterized by severe respiratory and motor impairment. This disease presents significant therapeutic challenges, with various strategies being explored to address its underlying pathology. Among these approaches, gene replacement therapy has demonstrated substantial functional improvements in clinical trials. However, safety issues emerged across different therapeutic approaches, highlighting the need for further research. AREAS COVERED This review provides a comprehensive analysis of the data gathered from natural history studies, preclinical models and clinical trials, with a particular focus on gene replacement therapy for XLMTM. The different therapeutic strategies are addressed, including their outcomes and associated safety concerns. EXPERT OPINION Despite the encouraging potential of gene therapy for XLMTM, the occurrence of safety challenges emphasizes the urgent need for a more comprehensive understanding of the disease's complex phenotype. Enhancing preclinical models to more accurately mimic the full spectrum of disease manifestations will be crucial for optimizing therapeutic strategies and reducing risks in future clinical applications.
Collapse
Affiliation(s)
- Cristina Martin
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Laurent Servais
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Pediatrics, Neuromuscular Reference Center, University and University Hospital of Liège, Liège, Belgium
| |
Collapse
|
2
|
Tijoriwalla S, Liyanage T, Herath TUB, Lee N, Rehman A, Gianfelice A, Ireton K. The host GTPase Dynamin 2 modulates apical junction structure to control cell-to-cell spread of Listeria monocytogenes. Infect Immun 2024; 92:e0013624. [PMID: 39133017 PMCID: PMC11475654 DOI: 10.1128/iai.00136-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/16/2024] [Indexed: 08/13/2024] Open
Abstract
The food-borne pathogen Listeria monocytogenes uses actin-based motility to generate plasma membrane protrusions that mediate the spread of bacteria between host cells. In polarized epithelial cells, efficient protrusion formation by L. monocytogenes requires the secreted bacterial protein InlC, which binds to a carboxyl-terminal Src homology 3 (SH3) domain in the human scaffolding protein Tuba. This interaction antagonizes Tuba, thereby diminishing cortical tension at the apical junctional complex and enhancing L. monocytogenes protrusion formation and spread. Tuba contains five SH3 domains apart from the domain that interacts with InlC. Here, we show that human GTPase Dynamin 2 associates with two SH3 domains in the amino-terminus of Tuba and acts together with this scaffolding protein to control the spread of L. monocytogenes. Genetic or pharmacological inhibition of Dynamin 2 or knockdown of Tuba each restored normal protrusion formation and spread to a bacterial strain deleted for the inlC gene (∆inlC). Dynamin 2 localized to apical junctions in uninfected human cells and protrusions in cells infected with L. monocytogenes. Localization of Dynamin 2 to junctions and protrusions depended on Tuba. Knockdown of Dynamin 2 or Tuba diminished junctional linearity, indicating a role for these proteins in controlling cortical tension. Infection with L. monocytogenes induced InlC-dependent displacement of Dynamin 2 from junctions, suggesting a possible mechanism of antagonism of this GTPase. Collectively, our results show that Dynamin 2 cooperates with Tuba to promote intercellular tension that restricts the spread of ∆inlC Listeria. By expressing InlC, wild-type L. monocytogenes overcomes this restriction.
Collapse
Affiliation(s)
- Serena Tijoriwalla
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Thiloma Liyanage
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Thilina U. B. Herath
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Nicole Lee
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Attika Rehman
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Antonella Gianfelice
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Keith Ireton
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
3
|
Romero MD, Carabeo RA. Dynamin-dependent entry of Chlamydia trachomatis is sequentially regulated by the effectors TarP and TmeA. Nat Commun 2024; 15:4926. [PMID: 38858371 PMCID: PMC11164928 DOI: 10.1038/s41467-024-49350-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 05/30/2024] [Indexed: 06/12/2024] Open
Abstract
Chlamydia invasion of epithelial cells is a pathogen-driven process involving two functionally distinct effectors - TarP and TmeA. They collaborate to promote robust actin dynamics at sites of entry. Here, we extend studies on the molecular mechanism of invasion by implicating the host GTPase dynamin 2 (Dyn2) in the completion of pathogen uptake. Importantly, Dyn2 function is modulated by TarP and TmeA at the levels of recruitment and activation through oligomerization, respectively. TarP-dependent recruitment requires phosphatidylinositol 3-kinase and the small GTPase Rac1, while TmeA has a post-recruitment role related to Dyn2 oligomerization. This is based on the rescue of invasion duration and efficiency in the absence of TmeA by the Dyn2 oligomer-stabilizing small molecule activator Ryngo 1-23. Notably, Dyn2 also regulated turnover of TarP- and TmeA-associated actin networks, with disrupted Dyn2 function resulting in aberrant turnover dynamics, thus establishing the interdependent functional relationship between Dyn2 and the effectors TarP and TmeA.
Collapse
Affiliation(s)
- Matthew D Romero
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rey A Carabeo
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
4
|
Romero MD, Carabeo RA. Dynamin-dependent entry of Chlamydia trachomatis is sequentially regulated by the effectors TarP and TmeA. RESEARCH SQUARE 2023:rs.3.rs-3376558. [PMID: 37841835 PMCID: PMC10571596 DOI: 10.21203/rs.3.rs-3376558/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Chlamydia invasion of epithelial cells is a pathogen-driven process involving two functionally distinct effectors - TarP and TmeA. They collaborate to promote robust actin dynamics at sites of entry. Here, we extend studies on the molecular mechanism of invasion by implicating the host GTPase dynamin 2 (Dyn2) in the completion of pathogen uptake. Importantly, Dyn2 function is modulated by TarP and TmeA at the levels of recruitment and activation through oligomerization, respectively. TarP-dependent recruitment requires phosphatidylinositol 3-kinase and the small GTPase Rac1, while TmeA has a post-recruitment role related to Dyn2 oligomerization. This is based on the rescue of invasion duration and efficiency in the absence of TmeA by the Dyn2 oligomer-stabilizing small molecule activator Ryngo 1-23. Notably, Dyn2 also regulated turnover of TarP- and TmeA-associated actin networks, with disrupted Dyn2 function resulting in aberrant turnover dynamics, thus establishing the interdependent functional relationship between Dyn2 and the effectors TarP and TmeA.
Collapse
Affiliation(s)
- Matthew D. Romero
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Rey A. Carabeo
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
5
|
Arriagada-Diaz J, Flores-Muñoz C, Gómez-Soto B, Labraña-Allende M, Mattar-Araos M, Prado-Vega L, Hinostroza F, Gajardo I, Guerra-Fernández MJ, Bevilacqua JA, Cárdenas AM, Bitoun M, Ardiles AO, Gonzalez-Jamett AM. A centronuclear myopathy-causing mutation in dynamin-2 disrupts neuronal morphology and excitatory synaptic transmission in a murine model of the disease. Neuropathol Appl Neurobiol 2023; 49:e12918. [PMID: 37317811 DOI: 10.1111/nan.12918] [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: 09/12/2022] [Revised: 04/30/2023] [Accepted: 06/02/2023] [Indexed: 06/16/2023]
Abstract
AIMS Dynamin-2 is a large GTPase, a member of the dynamin superfamily that regulates membrane remodelling and cytoskeleton dynamics. Mutations in the dynamin-2 gene (DNM2) cause autosomal dominant centronuclear myopathy (CNM), a congenital neuromuscular disorder characterised by progressive weakness and atrophy of the skeletal muscles. Cognitive defects have been reported in some DNM2-linked CNM patients suggesting that these mutations can also affect the central nervous system (CNS). Here we studied how a dynamin-2 CNM-causing mutation influences the CNS function. METHODS Heterozygous mice harbouring the p.R465W mutation in the dynamin-2 gene (HTZ), the most common causing autosomal dominant CNM, were used as disease model. We evaluated dendritic arborisation and spine density in hippocampal cultured neurons, analysed excitatory synaptic transmission by electrophysiological field recordings in hippocampal slices, and evaluated cognitive function by performing behavioural tests. RESULTS HTZ hippocampal neurons exhibited reduced dendritic arborisation and lower spine density than WT neurons, which was reversed by transfecting an interference RNA against the dynamin-2 mutant allele. Additionally, HTZ mice showed defective hippocampal excitatory synaptic transmission and reduced recognition memory compared to the WT condition. CONCLUSION Our findings suggest that the dynamin-2 p.R465W mutation perturbs the synaptic and cognitive function in a CNM mouse model and support the idea that this GTPase plays a key role in regulating neuronal morphology and excitatory synaptic transmission in the hippocampus.
Collapse
Affiliation(s)
- Jorge Arriagada-Diaz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Magister en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Bárbara Gómez-Soto
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Magister en Ciencias Médicas, Mención Biología Celular y Molecular, Universidad de Valparaíso, Valparaíso, Chile
| | - Marjorie Labraña-Allende
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Magister en Ciencias Médicas, Mención Biología Celular y Molecular, Universidad de Valparaíso, Valparaíso, Chile
| | - Michelle Mattar-Araos
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Lorena Prado-Vega
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Magister en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Fernando Hinostroza
- Centro de Investigación de Estudios Avanzados del Maule, CIEAM, Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile
- Centro de Investigación en Neuropsicología y Neurociencias Cognitivas, Facultad de Ciencias de la Salud, Universidad Católica del Maule, Talca, Chile
- Escuela de Química y Farmacia, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
| | - Ivana Gajardo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Jorge A Bevilacqua
- Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ana M Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Marc Bitoun
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, F-75013, France
| | - Alvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Estudios en Salud, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar, Chile
| | - Arlek M Gonzalez-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
6
|
Joshi VB, Gutierrez Ruiz OL, Razidlo GL. The Cell Biology of Metastatic Invasion in Pancreatic Cancer: Updates and Mechanistic Insights. Cancers (Basel) 2023; 15:cancers15072169. [PMID: 37046830 PMCID: PMC10093482 DOI: 10.3390/cancers15072169] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related mortality worldwide. This is largely due to the lack of routine screening protocols, an absence of symptoms in early-stage disease leading to late detection, and a paucity of effective treatment options. Critically, the majority of patients either present with metastatic disease or rapidly develop metastatic disease. Thus, there is an urgent need to deepen our understanding of metastasis in PDAC. During metastasis, tumor cells escape from the primary tumor, enter the circulation, and travel to a distant site to form a secondary tumor. In order to accomplish this relatively rare event, tumor cells develop an enhanced ability to detach from the primary tumor, migrate into the surrounding matrix, and invade across the basement membrane. In addition, cancer cells interact with the various cell types and matrix proteins that comprise the tumor microenvironment, with some of these factors working to promote metastasis and others working to suppress it. In PDAC, many of these processes are not well understood. The purpose of this review is to highlight recent advances in the cell biology of the early steps of the metastatic cascade in pancreatic cancer. Specifically, we will examine the regulation of epithelial-to-mesenchymal transition (EMT) in PDAC and its requirement for metastasis, summarize our understanding of how PDAC cells invade and degrade the surrounding matrix, and discuss how migration and adhesion dynamics are regulated in PDAC to optimize cancer cell motility. In addition, the role of the tumor microenvironment in PDAC will also be discussed for each of these invasive processes.
Collapse
Affiliation(s)
- Vidhu B Joshi
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Omar L Gutierrez Ruiz
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Gina L Razidlo
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
7
|
Abouelezz A, Almeida-Souza L. The mammalian endocytic cytoskeleton. Eur J Cell Biol 2022; 101:151222. [DOI: 10.1016/j.ejcb.2022.151222] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 12/27/2022] Open
|
8
|
Gómez-Oca R, Cowling BS, Laporte J. Common Pathogenic Mechanisms in Centronuclear and Myotubular Myopathies and Latest Treatment Advances. Int J Mol Sci 2021; 22:11377. [PMID: 34768808 PMCID: PMC8583656 DOI: 10.3390/ijms222111377] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/18/2021] [Indexed: 01/18/2023] Open
Abstract
Centronuclear myopathies (CNM) are rare congenital disorders characterized by muscle weakness and structural defects including fiber hypotrophy and organelle mispositioning. The main CNM forms are caused by mutations in: the MTM1 gene encoding the phosphoinositide phosphatase myotubularin (myotubular myopathy), the DNM2 gene encoding the mechanoenzyme dynamin 2, the BIN1 gene encoding the membrane curvature sensing amphiphysin 2, and the RYR1 gene encoding the skeletal muscle calcium release channel/ryanodine receptor. MTM1, BIN1, and DNM2 proteins are involved in membrane remodeling and trafficking, while RyR1 directly regulates excitation-contraction coupling (ECC). Several CNM animal models have been generated or identified, which confirm shared pathological anomalies in T-tubule remodeling, ECC, organelle mispositioning, protein homeostasis, neuromuscular junction, and muscle regeneration. Dynamin 2 plays a crucial role in CNM physiopathology and has been validated as a common therapeutic target for three CNM forms. Indeed, the promising results in preclinical models set up the basis for ongoing clinical trials. Another two clinical trials to treat myotubular myopathy by MTM1 gene therapy or tamoxifen repurposing are also ongoing. Here, we review the contribution of the different CNM models to understanding physiopathology and therapy development with a focus on the commonly dysregulated pathways and current therapeutic targets.
Collapse
Affiliation(s)
- Raquel Gómez-Oca
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67400 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67400 Illkirch, France
- Strasbourg University, 67081 Strasbourg, France
- Dynacure, 67400 Illkirch, France;
| | | | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67400 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67400 Illkirch, France
- Strasbourg University, 67081 Strasbourg, France
| |
Collapse
|
9
|
Trochet D, Bitoun M. A review of Dynamin 2 involvement in cancers highlights a promising therapeutic target. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:238. [PMID: 34294140 PMCID: PMC8296698 DOI: 10.1186/s13046-021-02045-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/15/2021] [Indexed: 12/23/2022]
Abstract
Dynamin 2 (DNM2) is an ubiquitously expressed large GTPase well known for its role in vesicle formation in endocytosis and intracellular membrane trafficking also acting as a regulator of cytoskeletons. During the last two decades, DNM2 involvement, through mutations or overexpression, emerged in an increasing number of cancers and often associated with poor prognosis. A wide panel of DNM2-dependent processes was described in cancer cells which explains DNM2 contribution to cancer pathomechanisms. First, DNM2 dysfunction may promote cell migration, invasion and metastasis. Second, DNM2 acts on intracellular signaling pathways fostering tumor cell proliferation and survival. Relative to these roles, DNM2 was demonstrated as a therapeutic target able to reduce cell proliferation, induce apoptosis, and reduce the invasive phenotype in a wide range of cancer cells in vitro. Moreover, proofs of concept of therapy by modulation of DNM2 expression was also achieved in vivo in several animal models. Consequently, DNM2 appears as a promising molecular target for the development of anti-invasive agents and the already provided proofs of concept in animal models represent an important step of preclinical development.
Collapse
Affiliation(s)
- Delphine Trochet
- Centre de Recherche en Myologie, Sorbonne Université, Inserm, UMRS 974, Institut de Myologie, F-75013, Paris, France
| | - Marc Bitoun
- Centre de Recherche en Myologie, Sorbonne Université, Inserm, UMRS 974, Institut de Myologie, F-75013, Paris, France.
| |
Collapse
|
10
|
Lynn KS, Easley KF, Martinez FJ, Reed RC, Schlingmann B, Koval M. Asymmetric distribution of dynamin-2 and β-catenin relative to tight junction spikes in alveolar epithelial cells. Tissue Barriers 2021; 9:1929786. [PMID: 34107845 DOI: 10.1080/21688370.2021.1929786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Tight junctions between lung alveolar epithelial cells maintain an air-liquid barrier necessary for healthy lung function. Previously, we found that rearrangement of tight junctions from a linear, cortical orientation into perpendicular protrusions (tight junction spikes) is associated with a decrease in alveolar barrier function, especially in alcoholic lung syndrome. Using quantitative super-resolution microscopy, we found that spikes in control cells were enriched for claudin-18 as compared with alcohol-exposed cells. Moreover, using an in situ method to measure barrier function, tight junction spikes were not associated with localized increases in permeability. This suggests that tight junction spikes have a regulatory role as opposed to causing a physical weakening of the epithelial barrier. We found that tight junction spikes form at cell-cell junctions oriented away from pools of β-catenin associated with actin filaments, suggesting that adherens junctions determine the directionality of tight junction spikes. Dynamin-2 was associated with junctional claudin-18 and ZO-1, but showed little localization with β-catenin and tight junction spikes. Treatment with Dynasore decreased the number of tight junction spikes/cell, increased tight junction spike length, and stimulated actin to redistribute to cortical tight junctions. By contrast, Dynole 34-2 and MiTMAB altered β-catenin localization, and reduced tight junction spike length. These data suggest a novel role for dynamin-2 in tight junction spike formation by reorienting junction-associated actin. Moreover, the greater spatial separation of adherens and tight junctions in squamous alveolar epithelial cells as compared with columnar epithelial cells facilitates analysis of molecular regulation of the apical junctional complex.
Collapse
Affiliation(s)
- K Sabrina Lynn
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, USA
| | - Kristen F Easley
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, USA
| | - Francisco J Martinez
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, USA
| | - Ryan C Reed
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, USA
| | - Barbara Schlingmann
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, USA
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, USA.,Department of Cell Biology, Emory University School of Medicine, Atlanta, USA
| |
Collapse
|
11
|
Kim J, Cooper JA. Junctional Localization of Septin 2 Is Required for Organization of Junctional Proteins in Static Endothelial Monolayers. Arterioscler Thromb Vasc Biol 2021; 41:346-359. [PMID: 33147991 PMCID: PMC7769918 DOI: 10.1161/atvbaha.120.315472] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Septin 2 is localized at junctions in human microvascular endothelial monolayers. The junctional localization of septin 2 is necessary for organization of cell-cell adhesion proteins of endothelial cells. Approach and Results: Septin 2 was depleted at junctions by suppression of expression using shRNA, treatment with inflammatory cytokine, TNF (tumor necrosis factor)-α, and ectopic overexpression of septin 2 phosphatidylinositol 4,5-bisphosphate binding mutant defect in interaction with plasma membrane. Under those conditions, organizations and expression levels of various junctional proteins were analyzed. Confocal images of immunofluorescence staining showed substantial disorganization of adherens junctional proteins, nectin-2 and afadin, TJP (tight junction protein), ZO (zonula occludens)-1, and intercellular adhesion protein, PECAM-1 (platelet-endothelial cell adhesion molecule-1). Immunoblots for those proteins did not show significant changes in expression except for nectin-2 that highly increased in expression. Significant differential gene expression profiles and biological pathway analysis by septin 2 suppression and by TNF-α treatment using RNA-seq showed common overlapping pathways. The commonalities in expression may be consistent with the similar effects on the overall organization of cell-cell adhesion proteins. CONCLUSIONS Localization of septin 2 at cell junctions are required for the arrangement of junctional proteins and the integrity of the barrier formed by endothelial monolayers.
Collapse
Affiliation(s)
- Joanna Kim
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, USA
| | - John A. Cooper
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
12
|
Arriagada-Diaz J, Prado-Vega L, Cárdenas Díaz AM, Ardiles AO, Gonzalez-Jamett AM. Dynamin Superfamily at Pre- and Postsynapses: Master Regulators of Synaptic Transmission and Plasticity in Health and Disease. Neuroscientist 2020; 28:41-58. [PMID: 33300419 DOI: 10.1177/1073858420974313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Dynamin superfamily proteins (DSPs) comprise a large group of GTP-ases that orchestrate membrane fusion and fission, and cytoskeleton remodeling in different cell-types. At the central nervous system, they regulate synaptic vesicle recycling and signaling-receptor turnover, allowing the maintenance of synaptic transmission. In the presynapses, these GTP-ases control the recycling of synaptic vesicles influencing the size of the ready-releasable pool and the release of neurotransmitters from nerve terminals, whereas in the postsynapses, they are involved in AMPA-receptor trafficking to and from postsynaptic densities, supporting excitatory synaptic plasticity, and consequently learning and memory formation. In agreement with these relevant roles, an important number of neurological disorders are associated with mutations and/or dysfunction of these GTP-ases. Along the present review we discuss the importance of DSPs at synapses and their implication in different neuropathological contexts.
Collapse
Affiliation(s)
- Jorge Arriagada-Diaz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Magister en Ciencias, mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Lorena Prado-Vega
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Magister en Ciencias, mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Ana M Cárdenas Díaz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Alvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Estudios en Salud, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar, Chile
| | - Arlek M Gonzalez-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
13
|
Lin SS, Hsieh TL, Liou GG, Li TN, Lin HC, Chang CW, Wu HY, Yao CK, Liu YW. Dynamin-2 Regulates Postsynaptic Cytoskeleton Organization and Neuromuscular Junction Development. Cell Rep 2020; 33:108310. [DOI: 10.1016/j.celrep.2020.108310] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/23/2020] [Accepted: 10/05/2020] [Indexed: 11/30/2022] Open
|
14
|
Dynamin regulates the dynamics and mechanical strength of the actin cytoskeleton as a multifilament actin-bundling protein. Nat Cell Biol 2020; 22:674-688. [PMID: 32451441 PMCID: PMC7953826 DOI: 10.1038/s41556-020-0519-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/07/2020] [Indexed: 01/28/2023]
Abstract
The dynamin GTPase is known to bundle actin filaments, but the underlying molecular mechanism and physiological relevance remain unclear. Our genetic analyses revealed a function of dynamin in propelling invasive membrane protrusions during myoblast fusion in vivo. Using biochemistry, total internal reflection fluorescence microscopy, electron microscopy and cryo-electron tomography, we show that dynamin bundles actin while forming a helical structure. At its full capacity, each dynamin helix captures 12-16 actin filaments on the outer rim of the helix. GTP hydrolysis by dynamin triggers disassembly of fully assembled dynamin helices, releasing free dynamin dimers/tetramers and facilitating Arp2/3-mediated branched actin polymerization. The assembly/disassembly cycles of dynamin promote continuous actin bundling to generate mechanically stiff actin super-bundles. Super-resolution and immunogold platinum replica electron microscopy revealed dynamin along actin bundles at the fusogenic synapse. These findings implicate dynamin as a unique multifilament actin-bundling protein that regulates the dynamics and mechanical strength of the actin cytoskeletal network.
Collapse
|
15
|
Burton KM, Cao H, Chen J, Qiang L, Krueger EW, Johnson KM, Bamlet WR, Zhang L, McNiven MA, Razidlo GL. Dynamin 2 interacts with α-actinin 4 to drive tumor cell invasion. Mol Biol Cell 2020; 31:439-451. [PMID: 31967944 PMCID: PMC7185896 DOI: 10.1091/mbc.e19-07-0395] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/23/2019] [Accepted: 01/15/2020] [Indexed: 12/21/2022] Open
Abstract
The large GTPase Dynamin 2 (Dyn2) is known to increase the invasiveness of pancreatic cancer tumor cells, but the mechanisms by which Dyn2 regulates changes in the actin cytoskeleton to drive cell migration are still unclear. Here we report that a direct interaction between Dyn2 and the actin-bundling protein alpha-actinin (α-actinin) 4 is critical for tumor cell migration and remodeling of the extracellular matrix in pancreatic ductal adenocarcinoma (PDAC) cells. The direct interaction is mediated through the C-terminal tails of both Dyn2 and α-actinin 4, and these proteins interact at invasive structures at the plasma membrane. While Dyn2 binds directly to both α-actinin 1 and α-actinin 4, only the interaction with α-actinin 4 is required to promote tumor cell invasion. Specific disruption of the Dyn2-α-actinin 4 interaction blocks the ability of PDAC cells to migrate in either two dimensions or invade through extracellular matrix as a result of impaired invadopodia stability. Analysis of human PDAC tumor tissue additionally reveals that elevated α-actinin 4 or Dyn2 expression are predictive of poor survival. Overall, these data demonstrate that Dyn2 regulates cytoskeletal dynamics, in part, by interacting with the actin-binding protein α-actinin 4 during tumor cell invasion.
Collapse
Affiliation(s)
- Kevin M. Burton
- Mayo Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905
| | - Hong Cao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Jing Chen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Li Qiang
- Mayo Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - Eugene W. Krueger
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | | | - William R. Bamlet
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905
| | - Lizhi Zhang
- Department of Anatomic Pathology, Mayo Clinic, Rochester, MN 55905
| | - Mark A. McNiven
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - Gina L. Razidlo
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
16
|
Ali T, Bednarska J, Vassilopoulos S, Tran M, Diakonov IA, Ziyadeh-Isleem A, Guicheney P, Gorelik J, Korchev YE, Reilly MM, Bitoun M, Shevchuk A. Correlative SICM-FCM reveals changes in morphology and kinetics of endocytic pits induced by disease-associated mutations in dynamin. FASEB J 2019; 33:8504-8518. [PMID: 31017801 PMCID: PMC6593877 DOI: 10.1096/fj.201802635r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Dynamin 2 (DNM2) is a GTP-binding protein that controls endocytic vesicle scission and defines a whole class of dynamin-dependent endocytosis, including clathrin-mediated endocytosis by caveoli. It has been suggested that mutations in the DNM2 gene, associated with 3 inherited diseases, disrupt endocytosis. However, how exactly mutations affect the nanoscale morphology of endocytic machinery has never been studied. In this paper, we used live correlative scanning ion conductance microscopy (SICM) and fluorescence confocal microscopy (FCM) to study how disease-associated mutations affect the morphology and kinetics of clathrin-coated pits (CCPs) by directly following their dynamics of formation, maturation, and internalization in skin fibroblasts from patients with centronuclear myopathy (CNM) and in Cos-7 cells expressing corresponding dynamin mutants. Using SICM-FCM, which we have developed, we show how p.R465W mutation disrupts pit structure, preventing its maturation and internalization, and significantly increases the lifetime of CCPs. Differently, p.R522H slows down the formation of CCPs without affecting their internalization. We also found that CNM mutations in DNM2 affect the distribution of caveoli and reduce dorsal ruffling in human skin fibroblasts. Collectively, our SICM-FCM findings at single CCP level, backed up by electron microscopy data, argue for the impairment of several forms of endocytosis in DNM2-linked CNM.-Ali, T., Bednarska, J., Vassilopoulos, S., Tran, M., Diakonov, I. A., Ziyadeh-Isleem, A., Guicheney, P., Gorelik, J., Korchev, Y. E., Reilly, M. M., Bitoun, M., Shevchuk, A. Correlative SICM-FCM reveals changes in morphology and kinetics of endocytic pits induced by disease-associated mutations in dynamin.
Collapse
Affiliation(s)
- Tayyibah Ali
- Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Joanna Bednarska
- Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Stéphane Vassilopoulos
- Research Center for Myology, Institut de Myologie, UMRS 974, INSERM, Sorbonne Université, Paris, France
| | - Martin Tran
- Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Ivan A Diakonov
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Azza Ziyadeh-Isleem
- UMRS 1166, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France.,UMRS 1166, Sorbonne Universités-Pierre and Marie Curie University (UPMC), Paris, France
| | - Pascale Guicheney
- UMRS 1166, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France.,UMRS 1166, Sorbonne Universités-Pierre and Marie Curie University (UPMC), Paris, France
| | - Julia Gorelik
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Yuri E Korchev
- Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Mary M Reilly
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square London, United Kingdom
| | - Marc Bitoun
- Research Center for Myology, Institut de Myologie, UMRS 974, INSERM, Sorbonne Université, Paris, France
| | - Andrew Shevchuk
- Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
17
|
Jimah JR, Hinshaw JE. Structural Insights into the Mechanism of Dynamin Superfamily Proteins. Trends Cell Biol 2019; 29:257-273. [DOI: 10.1016/j.tcb.2018.11.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/30/2018] [Accepted: 11/02/2018] [Indexed: 12/28/2022]
|
18
|
Markwell SM, Ammer AG, Interval ET, Allen JL, Papenberg BW, Hames RA, Castaño JE, Schafer DA, Weed SA. Cortactin Phosphorylation by Casein Kinase 2 Regulates Actin-Related Protein 2/3 Complex Activity, Invadopodia Function, and Tumor Cell Invasion. Mol Cancer Res 2019; 17:987-1001. [PMID: 30610108 DOI: 10.1158/1541-7786.mcr-18-0391] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 09/18/2018] [Accepted: 12/12/2018] [Indexed: 12/13/2022]
Abstract
Malregulation of the actin cytoskeleton enhances tumor cell motility and invasion. The actin-binding protein cortactin facilitates branched actin network formation through activation of the actin-related protein (Arp) 2/3 complex. Increased cortactin expression due to gene amplification is observed in head and neck squamous cell carcinoma (HNSCC) and other cancers, corresponding with elevated tumor progression and poor patient outcome. Arp2/3 complex activation is responsible for driving increased migration and extracellular matrix (ECM) degradation by governing invadopodia formation and activity. Although cortactin-mediated activation of Arp2/3 complex and invadopodia regulation has been well established, signaling pathways responsible for governing cortactin binding to Arp2/3 are unknown and potentially present a new avenue for anti-invasive therapeutic targeting. Here we identify casein kinase (CK) 2α phosphorylation of cortactin as a negative regulator of Arp2/3 binding. CK2α directly phosphorylates cortactin at a conserved threonine (T24) adjacent to the canonical Arp2/3 binding motif. Phosphorylation of cortactin T24 by CK2α impairs the ability of cortactin to bind Arp2/3 and activate actin nucleation. Decreased invadopodia activity is observed in HNSCC cells with expression of CK2α phosphorylation-null cortactin mutants, shRNA-mediated CK2α knockdown, and with the CK2α inhibitor Silmitasertib. Silmitasertib inhibits HNSCC collective invasion in tumor spheroids and orthotopic tongue tumors in mice. Collectively these data suggest that CK2α-mediated cortactin phosphorylation at T24 is critical in regulating cortactin binding to Arp2/3 complex and pro-invasive activity, identifying a potential targetable mechanism for impairing HNSCC invasion. IMPLICATIONS: This study identifies a new signaling pathway that contributes to enhancing cancer cell invasion.Visual Overview: http://mcr.aacrjournals.org/content/molcanres/17/4/987/F1.large.jpg.
Collapse
Affiliation(s)
- Steven M Markwell
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, West Virginia
| | - Amanda G Ammer
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, West Virginia
| | - Erik T Interval
- Department of Otolaryngology, Head and Neck Surgery, West Virginia University, Morgantown, West Virginia
| | - Jessica L Allen
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, West Virginia
| | - Brenen W Papenberg
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, West Virginia
| | - River A Hames
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, West Virginia
| | - Johnathan E Castaño
- Department of Otolaryngology, Head and Neck Surgery, West Virginia University, Morgantown, West Virginia
| | - Dorothy A Schafer
- Department of Biology, University of Virginia, Charlottesville, Virginia
| | - Scott A Weed
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, West Virginia.
| |
Collapse
|
19
|
Dunn VK, Gleason E. Inhibition of endocytosis suppresses the nitric oxide-dependent release of Cl- in retinal amacrine cells. PLoS One 2018; 13:e0201184. [PMID: 30044876 PMCID: PMC6059450 DOI: 10.1371/journal.pone.0201184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/10/2018] [Indexed: 11/18/2022] Open
Abstract
Our lab has previously shown that nitric oxide (NO) can alter the synaptic response properties of amacrine cells by releasing Cl- from internal acidic compartments. This alteration in the Cl- gradient brings about a positive shift in the reversal potential of the GABA-gated current, which can convert inhibitory synapses into excitatory synapses. Recently, we have shown that the cystic fibrosis transmembrane regulator (CFTR) Cl- channel is involved in the Cl- release. Here, we test the hypothesis that (acidic) synaptic vesicles are a source of NO-releasable Cl- in chick retinal amacrine cells. If SVs are a source of Cl-, then depleting synaptic vesicles should decrease the nitric oxide-dependent shift in the reversal potential of the GABA-gated current. The efficacy of four inhibitors of dynamin (dynasore, Dyngo 4a, Dynole 34-2, and MiTMAB) were evaluated. In order to deplete synaptic vesicles, voltage-steps were used to activate V-gated Ca2+ channels and stimulate the synaptic vesicle cycle either under control conditions or after treatment with the dynamin inhibitors. Voltage-ramps were used to measure the NO-dependent shift in the reversal potential of the GABA-gated currents under both conditions. Our results reveal that activating the synaptic vesicle cycle in the presence of dynasore or Dyngo 4a blocked the NO-dependent shift in EGABA. However, we also discovered that some dynamin inhibitors reduced Ca2+ signaling and L-type Ca2+ currents. Conversely, dynasore also increased neurotransmitter release at autaptic sites. To further resolve the mechanism underlying the inhibition of the NO-dependent shift in the reversal potential for the GABA-gated currents, we also tested the effects of the clathrin assembly inhibitor Pitstop 2 and found that this compound also inhibited the shift. These data provide evidence that dynamin inhibitors have multiple effects on amacrine cell synaptic transmission. These data also suggest that inhibition of endocytosis disrupts the ability of NO to elicit Cl- release from internal stores which may in part be due to depletion of synaptic vesicles.
Collapse
Affiliation(s)
- Vernon K. Dunn
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Evanna Gleason
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| |
Collapse
|
20
|
Kim J, Cooper JA. Septins regulate junctional integrity of endothelial monolayers. Mol Biol Cell 2018; 29:1693-1703. [PMID: 29771630 PMCID: PMC6080707 DOI: 10.1091/mbc.e18-02-0136] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/25/2018] [Accepted: 05/09/2018] [Indexed: 01/03/2023] Open
Abstract
Junctional integrity of endothelial monolayers is crucial to control movement of molecules and cells across the endothelium. Examining the structure and dynamics of cell junctions in endothelial monolayers, we discovered a role for septins. Contacts between adjacent endothelial cells were dynamic, with protrusions extending above or below neighboring cells. Vascular endothelial cadherin (VE-cadherin) was present at cell junctions, with a membrane-associated layer of F-actin. Septins localized at cell-junction membranes, in patterns distinct from VE-cadherin and F-actin. Septins assumed curved and scallop-shaped patterns at junctions, especially in regions of positive membrane curvature associated with actin-rich membrane protrusions. Depletion of septins led to disrupted morphology of VE-cadherin junctions and increased expression of VE-cadherin. In videos, septin-depleted cells displayed remodeling at cell junctions; regions with VE-cadherin were broader, and areas with membrane ruffling were wider. Septin depletion and junction disruption led to functional loss of junctional integrity, revealed by decreased transendothelial electric resistance and increased transmigration of immune cells. We conclude that septins, as cytoskeletal elements associated with the plasma membrane, are important for cell junctions and junctional integrity of endothelial monolayers, functioning at regions of positive curvature in support of actin-rich protrusions to promote cadherin-based cell junctions.
Collapse
Affiliation(s)
- Joanna Kim
- Departments of Biochemistry & Molecular Biophysics and Cell Biology & Physiology, Washington University, St. Louis, MO 63110
| | - John A. Cooper
- Departments of Biochemistry & Molecular Biophysics and Cell Biology & Physiology, Washington University, St. Louis, MO 63110
| |
Collapse
|
21
|
DePasquale JA. Apical surface ring formation in
Cyprinus carpio
scale epidermis. ACTA ZOOL-STOCKHOLM 2018. [DOI: 10.1111/azo.12256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
22
|
Local actin polymerization during endocytic carrier formation. Biochem Soc Trans 2018; 46:565-576. [DOI: 10.1042/bst20170355] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/06/2018] [Accepted: 03/16/2018] [Indexed: 12/20/2022]
Abstract
Extracellular macromolecules, pathogens and cell surface proteins rely on endocytosis to enter cells. Key steps of endocytic carrier formation are cargo molecule selection, plasma membrane folding and detachment from the cell surface. While dedicated proteins mediate each step, the actin cytoskeleton contributes to all. However, its role can be indirect to the actual molecular events driving endocytosis. Here, we review our understanding of the molecular steps mediating local actin polymerization during the formation of endocytic carriers. Clathrin-mediated endocytosis is the least reliant on local actin polymerization, as it is only engaged to counter forces induced by membrane tension or cytoplasmic pressure. Two opposite situations are coated pit formation in yeast and at the basolateral surface of polarized mammalian cells which are, respectively, dependent and independent on actin polymerization. Conversely, clathrin-independent endocytosis forming both nanometer [CLIC (clathrin-independent carriers)/GEEC (glycosylphosphatidylinositol (GPI)-anchored protein enriched endocytic compartments), caveolae, FEME (fast endophilin-mediated endocytosis) and IL-2β (interleukin-2β) uptake] and micrometer carriers (macropinocytosis) are dependent on actin polymerization to power local membrane deformation and carrier budding. A variety of endocytic adaptors can recruit and activate the Cdc42/N-WASP or Rac1/WAVE complexes, which, in turn, engage the Arp2/3 complex, thereby mediating local actin polymerization at the membrane. However, the molecular steps for RhoA and formin-mediated actin bundling during endocytic pit formation remain unclear.
Collapse
|
23
|
Single Intramuscular Injection of AAV-shRNA Reduces DNM2 and Prevents Myotubular Myopathy in Mice. Mol Ther 2018; 26:1082-1092. [PMID: 29506908 DOI: 10.1016/j.ymthe.2018.02.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 02/02/2018] [Accepted: 02/09/2018] [Indexed: 12/28/2022] Open
Abstract
Myotubular myopathy, or X-linked centronuclear myopathy, is a severe muscle disorder representing a significant burden for patients and their families. It is clinically characterized by neonatal and severe muscle weakness and atrophy. Mutations in the myotubularin (MTM1) gene cause myotubular myopathy, and no specific curative treatment is available. We previously found that dynamin 2 (DNM2) is upregulated in both Mtm1 knockout and patient muscle samples, whereas its reduction through antisense oligonucleotides rescues the clinical and histopathological features of this myopathy in mice. Here, we propose a novel approach targeting Dnm2 mRNA. We screened and validated in vitro and in vivo several short hairpin RNA (shRNA) sequences that efficiently target Dnm2 mRNA. A single intramuscular injection of AAV-shDnm2 resulted in long-term reduction of DNM2 protein level and restored muscle force, mass, histology, and myofiber ultrastructure and prevented molecular defects linked to the disease. Our results demonstrate a robust DNM2 knockdown and provide an alternative strategy based on reduction of DNM2 to treat myotubular myopathy.
Collapse
|
24
|
Yin M, Ma W, An L. Cortactin in cancer cell migration and invasion. Oncotarget 2017; 8:88232-88243. [PMID: 29152154 PMCID: PMC5675706 DOI: 10.18632/oncotarget.21088] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 08/29/2017] [Indexed: 12/20/2022] Open
Abstract
Cortactin, a substrate of sarcoma (Src) kinases, is an actin-binding protein that is involved in cytoskeletal regulation, and is frequently overexpressed in cancer cells. Binding to the actin related protein 2/3 (Arp2/3) complex stimulates cortactin activity, which promotes F-actin nucleation and assembly. Cortactin promotes cancer cell migration and invasion, and plays a pivotal role in invadopodia formation and extra cellular matrix degradation. Overexpression of cortactin, by amplification of the chromosomal band 11q13, increases tumor aggressiveness. In this review, we report on the current knowledge and potential mechanisms of action of cortactin as a critical mediator of cancer cell migration and invasion.
Collapse
Affiliation(s)
- Miao Yin
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Wenqing Ma
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Liguo An
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
25
|
González-Jamett AM, Baez-Matus X, Olivares MJ, Hinostroza F, Guerra-Fernández MJ, Vasquez-Navarrete J, Bui MT, Guicheney P, Romero NB, Bevilacqua JA, Bitoun M, Caviedes P, Cárdenas AM. Dynamin-2 mutations linked to Centronuclear Myopathy impair actin-dependent trafficking in muscle cells. Sci Rep 2017; 7:4580. [PMID: 28676641 PMCID: PMC5496902 DOI: 10.1038/s41598-017-04418-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 05/16/2017] [Indexed: 12/20/2022] Open
Abstract
Dynamin-2 is a ubiquitously expressed GTP-ase that mediates membrane remodeling. Recent findings indicate that dynamin-2 also regulates actin dynamics. Mutations in dynamin-2 cause dominant centronuclear myopathy (CNM), a congenital myopathy characterized by progressive weakness and atrophy of skeletal muscles. However, the muscle-specific roles of dynamin-2 affected by these mutations remain elusive. Here we show that, in muscle cells, the GTP-ase activity of dynamin-2 is involved in de novo actin polymerization as well as in actin-mediated trafficking of the glucose transporter GLUT4. Expression of dynamin-2 constructs carrying CNM-linked mutations disrupted the formation of new actin filaments as well as the stimulus-induced translocation of GLUT4 to the plasma membrane. Similarly, mature muscle fibers isolated from heterozygous knock-in mice that harbor the dynamin-2 mutation p.R465W, an animal model of CNM, exhibited altered actin organization, reduced actin polymerization and impaired insulin-induced translocation of GLUT4 to the sarcolemma. Moreover, GLUT4 displayed aberrant perinuclear accumulation in biopsies from CNM patients carrying dynamin-2 mutations, further suggesting trafficking defects. These results suggest that dynamin-2 is a key regulator of actin dynamics and GLUT4 trafficking in muscle cells. Our findings also support a model in which impairment of actin-dependent trafficking contributes to the pathological mechanism in dynamin-2-associated CNM.
Collapse
Affiliation(s)
- Arlek M González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile. .,Programa de Farmacología Molecular y Clinica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Ximena Baez-Matus
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - María José Olivares
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Fernando Hinostroza
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Doctorado en Ciencias, mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Maria José Guerra-Fernández
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jacqueline Vasquez-Navarrete
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Mai Thao Bui
- Université Sorbonne, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris, France.,Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière, Paris, France
| | - Pascale Guicheney
- INSERM, UMR_S1166, Sorbonne Universités, UPMC Univ Paris 06, UMR_S1166, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Norma Beatriz Romero
- Université Sorbonne, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris, France.,Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière, Paris, France
| | - Jorge A Bevilacqua
- Programa de Anatomía y Biología del Desarrollo, ICBM, Facultad de Medicina, Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Marc Bitoun
- Research Center for Myology, UPMC Univ Paris 06 and INSERM UMRS 974, Institute of Myology, Paris, France
| | - Pablo Caviedes
- Programa de Farmacología Molecular y Clinica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Ana M Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
26
|
González-Jamett AM, Guerra MJ, Olivares MJ, Haro-Acuña V, Baéz-Matus X, Vásquez-Navarrete J, Momboisse F, Martinez-Quiles N, Cárdenas AM. The F-Actin Binding Protein Cortactin Regulates the Dynamics of the Exocytotic Fusion Pore through its SH3 Domain. Front Cell Neurosci 2017; 11:130. [PMID: 28522963 PMCID: PMC5415606 DOI: 10.3389/fncel.2017.00130] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/18/2017] [Indexed: 11/20/2022] Open
Abstract
Upon cell stimulation, the network of cortical actin filaments is rearranged to facilitate the neurosecretory process. This actin rearrangement includes both disruption of the preexisting actin network and de novo actin polymerization. However, the mechanism by which a Ca2+ signal elicits the formation of new actin filaments remains uncertain. Cortactin, an actin-binding protein that promotes actin polymerization in synergy with the nucleation promoting factor N-WASP, could play a key role in this mechanism. We addressed this hypothesis by analyzing de novo actin polymerization and exocytosis in bovine adrenal chromaffin cells expressing different cortactin or N-WASP domains, or cortactin mutants that fail to interact with proline-rich domain (PRD)-containing proteins, including N-WASP, or to be phosphorylated by Ca2+-dependent kinases, such as ERK1/2 and Src. Our results show that the activation of nicotinic receptors in chromaffin cells promotes cortactin translocation to the cell cortex, where it colocalizes with actin filaments. We further found that, in association with PRD-containing proteins, cortactin contributes to the Ca2+-dependent formation of F-actin, and regulates fusion pore dynamics and the number of exocytotic events induced by activation of nicotinic receptors. However, whereas the actions of cortactin on the fusion pore dynamics seems to depend on the availability of monomeric actin and its phosphorylation by ERK1/2 and Src kinases, cortactin regulates the extent of exocytosis by a mechanism independent of actin polymerization. Together our findings point out a role for cortactin as a critical modulator of actin filament formation and exocytosis in neuroendocrine cells.
Collapse
Affiliation(s)
- Arlek M González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de ValparaísoValparaíso, Chile
| | - María J Guerra
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de ValparaísoValparaíso, Chile
| | - María J Olivares
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de ValparaísoValparaíso, Chile
| | - Valentina Haro-Acuña
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de ValparaísoValparaíso, Chile
| | - Ximena Baéz-Matus
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de ValparaísoValparaíso, Chile
| | - Jacqueline Vásquez-Navarrete
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de ValparaísoValparaíso, Chile
| | - Fanny Momboisse
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de ValparaísoValparaíso, Chile
| | - Narcisa Martinez-Quiles
- Departamento de Microbiología (Inmunología), Facultad de Medicina, Universidad Complutense de MadridMadrid, Spain
| | - Ana M Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de ValparaísoValparaíso, Chile
| |
Collapse
|
27
|
Edwards BS, Clay CM, Ellsworth BS, Navratil AM. Functional Role of Gonadotrope Plasticity and Network Organization. Front Endocrinol (Lausanne) 2017; 8:223. [PMID: 28936197 PMCID: PMC5595155 DOI: 10.3389/fendo.2017.00223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/16/2017] [Indexed: 11/18/2022] Open
Abstract
Gonadotrope cells of the anterior pituitary are characterized by their ability to mount a cyclical pattern of gonadotropin secretion to regulate gonadal function and fertility. Recent in vitro and in vivo evidence suggests that gonadotropes exhibit dramatic remodeling of the actin cytoskeleton following gonadotropin-releasing hormone (GnRH) exposure. GnRH engagement of actin is critical for gonadotrope function on multiple levels. First, GnRH-induced cell movements lead to spatial repositioning of the in vivo gonadotrope network toward vascular endothelium, presumably to access the bloodstream for effective hormone release. Interestingly, these plasticity changes can be modified depending on the physiological status of the organism. Additionally, GnRH-induced actin assembly appears to be fundamental to gonadotrope signaling at the level of extracellular signal-regulated kinase (ERK) activation, which is a well-known regulator of luteinizing hormone (LH) β-subunit synthesis. Last, GnRH-induced cell membrane projections are capable of concentrating LHβ-containing vesicles and disruption of the actin cytoskeleton reduces LH secretion. Taken together, gonadotrope network positioning and LH synthesis and secretion are linked to GnRH engagement of the actin cytoskeleton. In this review, we will cover the dynamics and organization of the in vivo gonadotrope cell network and the mechanisms of GnRH-induced actin-remodeling events important in ERK activation and subsequently hormone secretion.
Collapse
Affiliation(s)
- Brian S. Edwards
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| | - Colin M. Clay
- Department of Biomedical Science, Colorado State University, Fort Collins, CO, United States
| | - Buffy S. Ellsworth
- Department of Physiology, Southern Illinois University Carbondale, Carbondale, IL, United States
| | - Amy M. Navratil
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
- *Correspondence: Amy M. Navratil,
| |
Collapse
|
28
|
Okon IS, Ding Y, Coughlan KA, Wang Q, Song P, Benbrook DM, Zou MH. Aberrant NRP-1 expression serves as predicator of metastatic endometrial and lung cancers. Oncotarget 2016; 7:7970-8. [PMID: 26701889 PMCID: PMC4884968 DOI: 10.18632/oncotarget.6699] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 11/25/2015] [Indexed: 01/13/2023] Open
Abstract
Neuropilin-1 (NRP-1) has emerged as an important driver of tumor-promoting phenotypes of human malignancies. However, incomplete knowledge exists as to how this single-pass transmembrane receptor mediates pleiotropic tumor-promoting functions. The purpose of this study was to evaluate NRP-1 expression and metastatic properties in 94 endometrial cancer and matching serum specimens and in a lung cancer cell line. We found that NRP-1 expression significantly correlated with increased tumoral expression of vascular endothelial growth factor 2 (VEGFR2) and serum levels of hepatocyte growth factor (HGF) and cell growth-stimulating factor (C-GSF). Tumoral NRP-1 also was positively associated with expression of NEDD9, a pro-metastatic protein. In the highly metastatic lung cancer cell line (H1792), stable LKB1 depletion caused increased migration in vitro and accentuated NRP-1 and NEDD9 expression in vivo. Our findings demonstrate that perturbed expression of these targets correlate with metastatic potential of endometrial and lung tumors, providing clinically-relevant biomarker applications for diagnostic and therapeutic targeting.
Collapse
Affiliation(s)
- Imoh S Okon
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30302, USA
| | - Ye Ding
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30302, USA
| | | | - Qiongxin Wang
- Section of Molecular Medicine, Oklahoma City, OK 73104, USA
| | - Ping Song
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30302, USA
| | - Doris M Benbrook
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK 73104, USA
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30302, USA
| |
Collapse
|
29
|
Zhang Y, Nolan M, Yamada H, Watanabe M, Nasu Y, Takei K, Takeda T. Dynamin2 GTPase contributes to invadopodia formation in invasive bladder cancer cells. Biochem Biophys Res Commun 2016; 480:409-414. [DOI: 10.1016/j.bbrc.2016.10.063] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 10/19/2016] [Indexed: 02/04/2023]
|
30
|
Chalick M, Jacobi O, Pichinuk E, Garbar C, Bensussan A, Meeker A, Ziv R, Zehavi T, Smorodinsky NI, Hilkens J, Hanisch FG, Rubinstein DB, Wreschner DH. MUC1-ARF-A Novel MUC1 Protein That Resides in the Nucleus and Is Expressed by Alternate Reading Frame Translation of MUC1 mRNA. PLoS One 2016; 11:e0165031. [PMID: 27768738 PMCID: PMC5074479 DOI: 10.1371/journal.pone.0165031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 10/05/2016] [Indexed: 01/26/2023] Open
Abstract
Translation of mRNA in alternate reading frames (ARF) is a naturally occurring process heretofore underappreciated as a generator of protein diversity. The MUC1 gene encodes MUC1-TM, a signal-transducing trans-membrane protein highly expressed in human malignancies. Here we show that an AUG codon downstream to the MUC1-TM initiation codon initiates an alternate reading frame thereby generating a novel protein, MUC1-ARF. MUC1-ARF, like its MUC1-TM 'parent’ protein, contains a tandem repeat (VNTR) domain. However, the amino acid sequence of the MUC1-ARF tandem repeat as well as N- and C- sequences flanking it differ entirely from those of MUC1-TM. In vitro protein synthesis assays and extensive immunohistochemical as well as western blot analyses with MUC1-ARF specific monoclonal antibodies confirmed MUC1-ARF expression. Rather than being expressed at the cell membrane like MUC1-TM, immunostaining showed that MUC1-ARF protein localizes mainly in the nucleus: Immunohistochemical analyses of MUC1-expressing tissues demonstrated MUC1-ARF expression in the nuclei of secretory luminal epithelial cells. MUC1-ARF expression varies in different malignancies. While the malignant epithelial cells of pancreatic cancer show limited expression, in breast cancer tissue MUC1-ARF demonstrates strong nuclear expression. Proinflammatory cytokines upregulate expression of MUC1-ARF protein and co-immunoprecipitation analyses demonstrate association of MUC1-ARF with SH3 domain-containing proteins. Mass spectrometry performed on proteins coprecipitating with MUC1-ARF demonstrated Glucose-6-phosphate 1-dehydrogenase (G6PD) and Dynamin 2 (DNM2). These studies not only reveal that the MUC1 gene generates a previously unidentified MUC1-ARF protein, they also show that just like its ‘parent’ MUC1-TM protein, MUC1-ARF is apparently linked to signaling and malignancy, yet a definitive link to these processes and the roles it plays awaits a precise identification of its molecular functions. Comprising at least 524 amino acids, MUC1-ARF is, furthermore, the longest ARF protein heretofore described.
Collapse
Affiliation(s)
- Michael Chalick
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Oded Jacobi
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Edward Pichinuk
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Christian Garbar
- Department of Biopathology, Institut Jean-Godinot, Reims Cedex, France
| | | | - Alan Meeker
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ravit Ziv
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Tania Zehavi
- Department of Pathology, Meir Medical Center, Kfar Saba, Israel
| | | | - John Hilkens
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Franz-Georg Hanisch
- Institute of Biochemistry II, Medical Faculty, University of Cologne, Köln, Germany
| | | | - Daniel H. Wreschner
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
- * E-mail:
| |
Collapse
|
31
|
Hatch AL, Ji WK, Merrill RA, Strack S, Higgs HN. Actin filaments as dynamic reservoirs for Drp1 recruitment. Mol Biol Cell 2016; 27:3109-3121. [PMID: 27559132 PMCID: PMC5063618 DOI: 10.1091/mbc.e16-03-0193] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/19/2016] [Indexed: 11/11/2022] Open
Abstract
Drp1 is a dynamin-family GTPase recruited to mitochondria and peroxisomes, where it oligomerizes and drives membrane fission. Regulation of mitochondrial Drp1 recruitment is not fully understood. We previously showed that Drp1 binds actin filaments directly, and actin polymerization is necessary for mitochondrial Drp1 oligomerization in mammals. Here we show the Drp1/actin interaction displays unusual properties that are influenced by several factors. At saturation, only a fraction Drp1 binds actin filaments, and the off-rate of actin-bound Drp1 is significantly increased by unbound Drp1. GDP and GTP accelerate and decelerate Drp1/actin binding dynamics, respectively. Actin has a biphasic effect on Drp1 GTP hydrolysis, increasing at low actin:Drp1 ratio but returning to baseline at high ratio. Drp1 also bundles filaments. Bundles have reduced dynamics but follow the same trends as single filaments. Drp1 preferentially incorporates into bundles at higher ionic strength. We measure Drp1 concentration to be ∼0.5 μM in U2OS cell cytosol, suggesting the actin-binding affinity measured here (Kd = 0.6 μM) is in the physiologically relevant range. The ability of Drp1 to bind actin filaments in a highly dynamic manner provides potential for actin filaments to serve as reservoirs of oligomerization-competent Drp1 that can be accessed for mitochondrial fission.
Collapse
Affiliation(s)
- Anna L Hatch
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755
| | - Wei-Ke Ji
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755
| | - Ronald A Merrill
- Department of Pharmacology, Carver School of Medicine, University of Iowa, Iowa City, IA 52242
| | - Stefan Strack
- Department of Pharmacology, Carver School of Medicine, University of Iowa, Iowa City, IA 52242
| | - Henry N Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755
| |
Collapse
|
32
|
Yamada H, Takeda T, Michiue H, Abe T, Takei K. Actin bundling by dynamin 2 and cortactin is implicated in cell migration by stabilizing filopodia in human non-small cell lung carcinoma cells. Int J Oncol 2016; 49:877-86. [PMID: 27572123 PMCID: PMC4948956 DOI: 10.3892/ijo.2016.3592] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 05/25/2016] [Indexed: 11/06/2022] Open
Abstract
The endocytic protein dynamin participates in the formation of actin-based membrane protrusions such as podosomes, pseudopodia, and invadopodia, which facilitate cancer cell migration, invasion, and metastasis. However, the role of dynamin in the formation of actin-based membrane protrusions at the leading edge of cancer cells is unclear. In this study, we demonstrate that the ubiquitously expressed dynamin 2 isoform facilitates cell migration by stabilizing F-actin bundles in filopodia of the lung cancer cell line H1299. Pharmacological inhibition of dynamin 2 decreased cell migration and filopodial formation. Furthermore, dynamin 2 and cortactin mostly colocalized along F-actin bundles in filopodia of serum-stimulated H1299 cells by immunofluorescent and immunoelectron microscopy. Knockdown of dynamin 2 or cortactin inhibited the formation of filopodia in serum-stimulated H1299 cells, concomitant with a loss of F-actin bundles. Expression of wild-type cortactin rescued the punctate-like localization of dynamin 2 and filopodial formation. The incubation of dynamin 2 and cortactin with F-actin induced the formation of long and thick actin bundles, with these proteins colocalizing at F-actin bundles. A depolymerization assay revealed that dynamin 2 and cortactin increased the stability of F-actin bundles. These results indicate that dynamin 2 and cortactin participate in cell migration by stabilizing F-actin bundles in filopodia. Taken together, these findings suggest that dynamin might be a possible molecular target for anticancer therapy.
Collapse
Affiliation(s)
- Hiroshi Yamada
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| | - Tetsuya Takeda
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| | - Hiroyuki Michiue
- Department of Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| | - Tadashi Abe
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| | - Kohji Takei
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| |
Collapse
|
33
|
Yamada H, Kobayashi K, Zhang Y, Takeda T, Takei K. Expression of a dynamin 2 mutant associated with Charcot-Marie-Tooth disease leads to aberrant actin dynamics and lamellipodia formation. Neurosci Lett 2016; 628:179-85. [PMID: 27328317 DOI: 10.1016/j.neulet.2016.06.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 06/03/2016] [Accepted: 06/16/2016] [Indexed: 10/21/2022]
Abstract
Specific mutations in dynamin 2 are linked to Charcot-Marie-Tooth disease (CMT), an inherited peripheral neuropathy. However, the effects of these mutations on dynamin function, particularly in relation to the regulation of the actin cytoskeleton remain unclear. Here, selected CMT-associated dynamin mutants were expressed to examine their role in the pathogenesis of CMT in U2OS cells. Ectopic expression of the dynamin CMT mutants 555Δ3 and K562E caused an approximately 50% decrease in serum stimulation-dependent lamellipodia formation; however, only K562E caused aberrations in the actin cytoskeleton. Immunofluorescence analysis showed that the K562E mutation resulted in the disappearance of radially aligned actin bundles and the simultaneous appearance of F-actin clusters. Live-cell imaging analyses showed F-actin polymers of decreased length assembled into immobile clusters in K562E-expressing cells. The K562E dynamin mutant colocalized with the F-actin clusters, whereas its colocalization with clathrin-coated pit marker proteins was decreased. Essentially the same results were obtained using another cell line, HeLa and NG108-15 cells. The present study is the first to show the association of dynamin CMT mutations with aberrant actin dynamics and lamellipodia, which may contribute to defective endocytosis and myelination in Schwann cells in CMT.
Collapse
Affiliation(s)
- Hiroshi Yamada
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan; CREST, Japan Science and Technology Agency, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kinue Kobayashi
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Yubai Zhang
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Tetsuya Takeda
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan; CREST, Japan Science and Technology Agency, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kohji Takei
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan; CREST, Japan Science and Technology Agency, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| |
Collapse
|
34
|
Edwards BS, Dang AK, Murtazina DA, Dozier MG, Whitesell JD, Khan SA, Cherrington BD, Amberg GC, Clay CM, Navratil AM. Dynamin Is Required for GnRH Signaling to L-Type Calcium Channels and Activation of ERK. Endocrinology 2016; 157:831-43. [PMID: 26696122 PMCID: PMC4733113 DOI: 10.1210/en.2015-1575] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have shown that GnRH-mediated engagement of the cytoskeleton induces cell movement and is necessary for ERK activation. It also has previously been established that a dominant negative form of the mechano-GTPase dynamin (K44A) attenuates GnRH activation of ERK. At present, it is not clear at what level these cellular events might be linked. To explore this, we used live cell imaging in the gonadotrope-derived αT3-1 cell line to determine that dynamin-green fluorescent protein accumulated in GnRH-induced lamellipodia and plasma membrane protrusions. Coincident with translocation of dynamin-green fluorescent protein to the plasma membrane, we demonstrated that dynamin colocalizes with the actin cytoskeleton and the actin binding protein, cortactin at the leading edge of the plasma membrane. We next wanted to assess the physiological significance of these findings by inhibiting dynamin GTPase activity using dynasore. We find that dynasore suppresses activation of ERK, but not c-Jun N-terminal kinase, after exposure to GnRH agonist. Furthermore, exposure of αT3-1 cells to dynasore inhibited GnRH-induced cyto-architectural rearrangements. Recently it has been discovered that GnRH induced Ca(2+) influx via the L-type Ca(2+) channels requires an intact cytoskeleton to mediate ERK phosphorylation. Interestingly, not only does dynasore attenuate GnRH-mediated actin reorganization, it also suppresses Ca(2+) influx through L-type Ca(2+) channels visualized in living cells using total internal reflection fluorescence microscopy. Collectively, our data suggest that GnRH-induced membrane remodeling events are mediated in part by the association of dynamin and cortactin engaging the actin cytoskeleton, which then regulates Ca(2+) influx via L-type channels to facilitate ERK phosphorylation.
Collapse
Affiliation(s)
- Brian S Edwards
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - An K Dang
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Dilyara A Murtazina
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Melissa G Dozier
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Jennifer D Whitesell
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Shaihla A Khan
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Brian D Cherrington
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Gregory C Amberg
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Colin M Clay
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Amy M Navratil
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| |
Collapse
|
35
|
Ji WK, Hatch AL, Merrill RA, Strack S, Higgs HN. Actin filaments target the oligomeric maturation of the dynamin GTPase Drp1 to mitochondrial fission sites. eLife 2015; 4:e11553. [PMID: 26609810 PMCID: PMC4755738 DOI: 10.7554/elife.11553] [Citation(s) in RCA: 245] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/25/2015] [Indexed: 12/19/2022] Open
Abstract
While the dynamin GTPase Drp1 plays a critical role during mitochondrial fission, mechanisms controlling its recruitment to fission sites are unclear. A current assumption is that cytosolic Drp1 is recruited directly to fission sites immediately prior to fission. Using live-cell microscopy, we find evidence for a different model, progressive maturation of Drp1 oligomers on mitochondria through incorporation of smaller mitochondrially-bound Drp1 units. Maturation of a stable Drp1 oligomer does not forcibly lead to fission. Drp1 oligomers also translocate directionally along mitochondria. Ionomycin, a calcium ionophore, causes rapid mitochondrial accumulation of actin filaments followed by Drp1 accumulation at the fission site, and increases fission rate. Inhibiting actin polymerization, myosin IIA, or the formin INF2 reduces both un-stimulated and ionomycin-induced Drp1 accumulation and mitochondrial fission. Actin filaments bind purified Drp1 and increase GTPase activity in a manner that is synergistic with the mitochondrial protein Mff, suggesting a role for direct Drp1/actin interaction. We propose that Drp1 is in dynamic equilibrium on mitochondria in a fission-independent manner, and that fission factors such as actin filaments target productive oligomerization to fission sites. DOI:http://dx.doi.org/10.7554/eLife.11553.001 Inside cells, structures called mitochondria supply the energy needed to carry out the processes that sustain life. Mitochondria constantly divide (a process known as fission) or fuse together, which helps to keep them in good working condition and well distributed around the cell. Several neurological disorders, including Parkinson’s disease and Alzheimer’s, are associated with problems that affect mitochondrial fission. Many different molecules work together to help mitochondria divide, including a protein called Drp1. A number of Drp1 molecules can associate with each other to form an “oligomer” in the shape of a ring around a mitochondrion. The ring then constricts to split the mitochondrion in two. It is often assumed that Drp1 molecules are recruited to the mitochondria immediately before fission and then form the oligomer ring. However, by using microscopy to track the movement of fluorescently labeled Drp1 molecules in human cells, Ji, Hatch et al. now suggest that Drp1 is continuously binding to and releasing from mitochondria, regardless of the need for fission. The experiments showed that when bound to surface of the mitochondrion, Drp1 switches between assembling and disassembling the oligomer ring. This process of Drp1 assembly and oligomerization on mitochondria is called maturation. Specific signals for fission can push Drp1 toward maturation, which then leads to fission. Ji, Hatch et al. found that one such signal is the assembly of filaments of a protein called actin. Preventing actin filaments from forming reduced the amount of Drp1 that accumulated at mitochondria, and resulted in the mitochondria dividing less frequently. Further biochemical experiments also revealed that actin interacts directly with Drp1 and stimulates Drp1 activity, helping the ring to organize and assist mitochondrial fission. The formation of actin filaments is not the only mechanism that can recruit Drp1 to mitochondria. Future work should investigate whether other mechanisms work with actin to recruit Drp1. As with actin filaments, other signals might be predicted to influence the balance of maturation and disassembly of Drp1 oligomers. DOI:http://dx.doi.org/10.7554/eLife.11553.002
Collapse
Affiliation(s)
- Wei-ke Ji
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, United States
| | - Anna L Hatch
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, United States
| | - Ronald A Merrill
- Department of Pharmacology, The University of Iowa, Iowa City, United States
| | - Stefan Strack
- Department of Pharmacology, The University of Iowa, Iowa City, United States
| | - Henry N Higgs
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, United States
| |
Collapse
|
36
|
Wang J, Liu M, Chen L, Chan THM, Jiang L, Yuan YF, Guan XY. Overexpression of N-terminal kinase like gene promotes tumorigenicity of hepatocellular carcinoma by regulating cell cycle progression and cell motility. Oncotarget 2015; 6:1618-30. [PMID: 25575811 PMCID: PMC4359319 DOI: 10.18632/oncotarget.2730] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/08/2014] [Indexed: 11/25/2022] Open
Abstract
Amplification and overexpression of CHD1L is one of the most frequent genetic alterations in hepatocellular carcinoma (HCC). Here we found that one of CHD1L downstream targets, NTKL, was frequently upregulated in HCC, which was significantly correlated with vascular invasion (P = 0.012) and poor prognosis (P = 0.050) of HCC. ChIP assay demonstrated the binding of CHD1L to the promoter region of NTKL. QRT-PCR study showed that the expression of NTKL positively correlated with CHD1L expression in both clinical samples and cell lines. Functional study found that NTKL had strong oncogenic roles, including increased cell growth, colony formation in soft agar, and tumor formation in nude mice. Further study found that NTKL could promote G1/S transition by decreasing P53 and increasing CyclinD1 expressions. NTKL overexpression could accelerate the mitotic exit and chromosome segregation, which led to the cytokinesis failure and subsequently induced apoptosis. NTKL also regulated cell motility by facilitating philopodia and lamellipodia formation through regulating F-actin reorganization and the phosphorylation of small GTPase Rac1/cdc42. Using co-IP and mass spectrometry approach, we identified the large GTPase dynamin2 as an interacting protein of NTKL, which might be responsible for the phenotype alterations caused by NTKL overexpression, such as cytokinesis failure, increased cell motility and abnormal of cell division.
Collapse
Affiliation(s)
- Jian Wang
- Departments of Clinical Oncology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China.,Center for Cancer Research, The University of Hong Kong, Hong Kong, China
| | - Ming Liu
- Departments of Clinical Oncology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China.,Center for Cancer Research, The University of Hong Kong, Hong Kong, China
| | - Leilei Chen
- Departments of Clinical Oncology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Tim Hon Man Chan
- Departments of Clinical Oncology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Lingxi Jiang
- Departments of Clinical Oncology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Yun-Fei Yuan
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin-Yuan Guan
- Departments of Clinical Oncology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China.,Center for Cancer Research, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
37
|
Preta G, Cronin JG, Sheldon IM. Dynasore - not just a dynamin inhibitor. Cell Commun Signal 2015; 13:24. [PMID: 25889964 PMCID: PMC4396812 DOI: 10.1186/s12964-015-0102-1] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/26/2015] [Indexed: 12/23/2022] Open
Abstract
Dynamin is a GTPase protein that is essential for membrane fission during clathrin-mediated endocytosis in eukaryotic cells. Dynasore is a GTPase inhibitor that rapidly and reversibly inhibits dynamin activity, which prevents endocytosis. However, comparison between cells treated with dynasore and RNA interference of genes encoding dynamin, reveals evidence that dynasore reduces labile cholesterol in the plasma membrane, and disrupts lipid raft organization, in a dynamin-independent manner. To explore the role of dynamin it is important to use multiple dynamin inhibitors, alongside the use of dynamin mutants and RNA interference targeting genes encoding dynamin. On the other hand, dynasore provides an interesting tool to explore the regulation of cholesterol in plasma membranes.
Collapse
Affiliation(s)
- Giulio Preta
- Institute of Life Science, College of Medicine, Swansea University, Swansea, SA2 8PP, UK.
| | - James G Cronin
- Institute of Life Science, College of Medicine, Swansea University, Swansea, SA2 8PP, UK.
| | - I Martin Sheldon
- Institute of Life Science, College of Medicine, Swansea University, Swansea, SA2 8PP, UK.
| |
Collapse
|
38
|
Role of dynamin in elongated cell migration in a 3D matrix. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:611-8. [DOI: 10.1016/j.bbamcr.2014.12.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/04/2014] [Accepted: 12/05/2014] [Indexed: 11/18/2022]
|
39
|
Mooren OL, Li J, Nawas J, Cooper JA. Endothelial cells use dynamic actin to facilitate lymphocyte transendothelial migration and maintain the monolayer barrier. Mol Biol Cell 2014; 25:4115-29. [PMID: 25355948 PMCID: PMC4263454 DOI: 10.1091/mbc.e14-05-0976] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Actin assembly downstream of WAVE2 in endothelial cells is necessary to engage transmigrating lymphocytes, promote the transcellular route of migration, and close junctional pores after the lymphocyte moves away. In addition, WAVE2 is necessary for endothelial monolayer integrity. The vascular endothelium is a highly dynamic structure, and the integrity of its barrier function is tightly regulated. Normally impenetrable to cells, the endothelium actively assists lymphocytes to exit the bloodstream during inflammation. The actin cytoskeleton of the endothelial cell (EC) is known to facilitate transmigration, but the cellular and molecular mechanisms are not well understood. Here we report that actin assembly in the EC, induced by Arp2/3 complex under control of WAVE2, is important for several steps in the process of transmigration. To begin transmigration, ECs deploy actin-based membrane protrusions that create a cup-shaped docking structure for the lymphocyte. We found that docking structure formation involves the localization and activation of Arp2/3 complex by WAVE2. The next step in transmigration is creation of a migratory pore, and we found that endothelial WAVE2 is needed for lymphocytes to follow a transcellular route through an EC. Later, ECs use actin-based protrusions to close the gap behind the lymphocyte, which we discovered is also driven by WAVE2. Finally, we found that ECs in resting endothelial monolayers use lamellipodial protrusions dependent on WAVE2 to form and maintain contacts and junctions between cells.
Collapse
Affiliation(s)
- Olivia L Mooren
- Department of Cell Biology and Physiology, Washington University, St. Louis, MO 63110
| | - Jinmei Li
- Department of Cell Biology and Physiology, Washington University, St. Louis, MO 63110
| | - Julie Nawas
- Department of Cell Biology and Physiology, Washington University, St. Louis, MO 63110
| | - John A Cooper
- Department of Cell Biology and Physiology, Washington University, St. Louis, MO 63110
| |
Collapse
|
40
|
Shin NY, Choi H, Neff L, Wu Y, Saito H, Ferguson SM, De Camilli P, Baron R. Dynamin and endocytosis are required for the fusion of osteoclasts and myoblasts. ACTA ACUST UNITED AC 2014; 207:73-89. [PMID: 25287300 PMCID: PMC4195819 DOI: 10.1083/jcb.201401137] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dynamin function is essential for cell–cell fusion in both osteoclast precursors and myoblasts in part through its effects on endocytosis. Cell–cell fusion is an evolutionarily conserved process that leads to the formation of multinucleated myofibers, syncytiotrophoblasts and osteoclasts, allowing their respective functions. Although cell–cell fusion requires the presence of fusogenic membrane proteins and actin-dependent cytoskeletal reorganization, the precise machinery allowing cells to fuse is still poorly understood. Using an inducible knockout mouse model to generate dynamin 1– and 2–deficient primary osteoclast precursors and myoblasts, we found that fusion of both cell types requires dynamin. Osteoclast and myoblast cell–cell fusion involves the formation of actin-rich protrusions closely associated with clathrin-mediated endocytosis in the apposed cell. Furthermore, impairing endocytosis independently of dynamin also prevented cell–cell fusion. Since dynamin is involved in both the formation of actin-rich structures and in endocytosis, our results indicate that dynamin function is central to the osteoclast precursors and myoblasts fusion process, and point to an important role of endocytosis in cell–cell fusion.
Collapse
Affiliation(s)
- Nah-Young Shin
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Hyewon Choi
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Lynn Neff
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Yumei Wu
- Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510 Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510
| | - Hiroaki Saito
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Shawn M Ferguson
- Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510 Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510
| | - Pietro De Camilli
- Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510 Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510 Howard Hughes Medical Institute, Chevy Chase, MD 20815
| | - Roland Baron
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| |
Collapse
|
41
|
Helgeson LA, Prendergast JG, Wagner AR, Rodnick-Smith M, Nolen BJ. Interactions with actin monomers, actin filaments, and Arp2/3 complex define the roles of WASP family proteins and cortactin in coordinately regulating branched actin networks. J Biol Chem 2014; 289:28856-69. [PMID: 25160634 DOI: 10.1074/jbc.m114.587527] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Arp2/3 complex is an important actin filament nucleator that creates branched actin filament networks required for formation of lamellipodia and endocytic actin structures. Cellular assembly of branched actin networks frequently requires multiple Arp2/3 complex activators, called nucleation promoting factors (NPFs). We recently presented a mechanism by which cortactin, a weak NPF, can displace a more potent NPF, N-WASP, from nascent branch junctions to synergistically accelerate nucleation. The distinct roles of these NPFs in branching nucleation are surprising given their similarities. We biochemically dissected these two classes of NPFs to determine how their Arp2/3 complex and actin interacting segments modulate their influences on branched actin networks. We find that the Arp2/3 complex-interacting N-terminal acidic sequence (NtA) of cortactin has structural features distinct from WASP acidic regions (A) that are required for synergy between the two NPFs. Our mutational analysis shows that differences between NtA and A do not explain the weak intrinsic NPF activity of cortactin, but instead that cortactin is a weak NPF because it cannot recruit actin monomers to Arp2/3 complex. We use TIRF microscopy to show that cortactin bundles branched actin filaments using actin filament binding repeats within a single cortactin molecule, but that N-WASP antagonizes cortactin-mediated bundling. Finally, we demonstrate that multiple WASP family proteins synergistically activate Arp2/3 complex and determine the biochemical requirements in WASP proteins for synergy. Our data indicate that synergy between WASP proteins and cortactin may play a general role in assembling diverse actin-based structures, including lamellipodia, podosomes, and endocytic actin networks.
Collapse
Affiliation(s)
- Luke A Helgeson
- From the Institute of Molecular Biology and Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon 97403-1229
| | - Julianna G Prendergast
- From the Institute of Molecular Biology and Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon 97403-1229
| | - Andrew R Wagner
- From the Institute of Molecular Biology and Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon 97403-1229
| | - Max Rodnick-Smith
- From the Institute of Molecular Biology and Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon 97403-1229
| | - Brad J Nolen
- From the Institute of Molecular Biology and Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon 97403-1229
| |
Collapse
|
42
|
Chou AM, Sem KP, Wright GD, Sudhaharan T, Ahmed S. Dynamin1 is a novel target for IRSp53 protein and works with mammalian enabled (Mena) protein and Eps8 to regulate filopodial dynamics. J Biol Chem 2014; 289:24383-96. [PMID: 25031323 DOI: 10.1074/jbc.m114.553883] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Filopodia are dynamic actin-based structures that play roles in processes such as cell migration, wound healing, and axonal guidance. Cdc42 induces filopodial formation through IRSp53, an Inverse-Bin-Amphiphysins-Rvs (I-BAR) domain protein. Previous work from a number of laboratories has shown that IRSp53 generates filopodia by coupling membrane protrusion with actin dynamics through its Src homology 3 domain binding partners. Here, we show that dynamin1 (Dyn1), the large guanosine triphosphatase, is an interacting partner of IRSp53 through pulldown and Förster resonance energy transfer analysis, and we explore its role in filopodial formation. In neuroblastoma cells, Dyn1 localizes to filopodia, associated tip complexes, and the leading edge just behind the anti-capping protein mammalian enabled (Mena). Dyn1 knockdown reduces filopodial formation, which can be rescued by overexpressing wild-type Dyn1 but not the GTPase mutant Dyn1-K44A and the loss-of-function actin binding domain mutant Dyn1-K/E. Interestingly, dynasore, an inhibitor of Dyn GTPase, also reduced filopodial number and increased their lifetime. Using rapid time-lapse total internal reflection fluorescence microscopy, we show that Dyn1 and Mena localize to filopodia only during initiation and assembly. Dyn1 actin binding domain mutant inhibits filopodial formation, suggesting a role in actin elongation. In contrast, Eps8, an actin capping protein, is seen most strongly at filopodial tips during disassembly. Taken together, the results suggest IRSp53 partners with Dyn1, Mena, and Eps8 to regulate filopodial dynamics.
Collapse
Affiliation(s)
- Ai Mei Chou
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Kai Ping Sem
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Graham Daniel Wright
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Thankiah Sudhaharan
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Sohail Ahmed
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| |
Collapse
|
43
|
Menon M, Askinazi OL, Schafer DA. Dynamin2 organizes lamellipodial actin networks to orchestrate lamellar actomyosin. PLoS One 2014; 9:e94330. [PMID: 24710573 PMCID: PMC3978067 DOI: 10.1371/journal.pone.0094330] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/14/2014] [Indexed: 12/02/2022] Open
Abstract
Actin networks in migrating cells exist as several interdependent structures: sheet-like networks of branched actin filaments in lamellipodia; arrays of bundled actin filaments co-assembled with myosin II in lamellae; and actin filaments that engage focal adhesions. How these dynamic networks are integrated and coordinated to maintain a coherent actin cytoskeleton in migrating cells is not known. We show that the large GTPase dynamin2 is enriched in the distal lamellipod where it regulates lamellipodial actin networks as they form and flow in U2-OS cells. Within lamellipodia, dynamin2 regulated the spatiotemporal distributions of α-actinin and cortactin, two actin-binding proteins that specify actin network architecture. Dynamin2's action on lamellipodial F-actin influenced the formation and retrograde flow of lamellar actomyosin via direct and indirect interactions with actin filaments and a finely tuned GTP hydrolysis activity. Expression in dynamin2-depleted cells of a mutant dynamin2 protein that restores endocytic activity, but not activities that remodel actin filaments, demonstrated that actin filament remodeling by dynamin2 did not depend of its functions in endocytosis. Thus, dynamin2 acts within lamellipodia to organize actin filaments and regulate assembly and flow of lamellar actomyosin. We hypothesize that through its actions on lamellipodial F-actin, dynamin2 generates F-actin structures that give rise to lamellar actomyosin and for efficient coupling of F-actin at focal adhesions. In this way, dynamin2 orchestrates the global actin cytoskeleton.
Collapse
Affiliation(s)
- Manisha Menon
- Department of Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Olga L. Askinazi
- Department of Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Dorothy A. Schafer
- Department of Biology, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
44
|
Huh YH, Kim SH, Chung KH, Oh S, Kwon MS, Choi HW, Rhee S, Ryu JH, Park ZY, Jun CD, Song WK. Swiprosin-1 modulates actin dynamics by regulating the F-actin accessibility to cofilin. Cell Mol Life Sci 2014; 70:4841-54. [PMID: 23959172 PMCID: PMC3830201 DOI: 10.1007/s00018-013-1447-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 07/31/2013] [Accepted: 08/02/2013] [Indexed: 11/27/2022]
Abstract
Membrane protrusions, like lamellipodia, and cell movement are dependent on actin dynamics, which are regulated by a variety of actin-binding proteins acting cooperatively to reorganize actin filaments. Here, we provide evidence that Swiprosin-1, a newly identified actin-binding protein, modulates lamellipodial dynamics by regulating the accessibility of F-actin to cofilin. Overexpression of Swiprosin-1 increased lamellipodia formation in B16F10 melanoma cells, whereas knockdown of Swiprosin-1 inhibited EGF-induced lamellipodia formation, and led to a loss of actin stress fibers at the leading edges of cells but not in the cell cortex. Swiprosin-1 strongly facilitated the formation of entangled or clustered F-actin, which remodeled the structural organization of actin filaments making them in accessible to cofilin. EGF-induced phosphorylation of Swiprosin-1 at Ser183, a phosphorylation site newly identified using mass spectrometry, effectively inhibited clustering of actin filaments and permitted cofilin access to F-actin, resulting in actin depolymerization. Cells over expressing a Swiprosin-1 phosphorylation-mimicking mutant or a phosphorylation-deficient mutant exhibited irregular membrane dynamics during the protrusion and retraction cycles of lamellipodia. Taken together, these findings suggest that dynamic exchange of Swiprosin-1 phosphorylation and dephosphorylation is a novel mechanism that regulates actin dynamics by modulating the pattern of cofilin activity at the leading edges of cells.
Collapse
Affiliation(s)
- Yun Hyun Huh
- Bio Imaging and Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 500712 Korea
| | - So Hee Kim
- Bio Imaging and Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 500712 Korea
| | - Kyoung-Hwun Chung
- Bio Imaging and Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 500712 Korea
| | - Sena Oh
- Bio Imaging and Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 500712 Korea
| | - Min-Sung Kwon
- Bio Imaging and Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 500712 Korea
| | - Hyun-Woo Choi
- Bio Imaging and Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 500712 Korea
| | - Sangmyung Rhee
- School of Biological Sciences, Joong Ang University, Seoul, 156756 Korea
| | - Je-Hwang Ryu
- Research Center for Biomineralization Disorders and Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, 500757 Korea
| | - Zee Yong Park
- Bio Imaging and Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 500712 Korea
| | - Chang-Duk Jun
- Bio Imaging and Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 500712 Korea
| | - Woo Keun Song
- Bio Imaging and Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 500712 Korea
| |
Collapse
|
45
|
Ivanov AI. Pharmacological inhibitors of exocytosis and endocytosis: novel bullets for old targets. Methods Mol Biol 2014; 1174:3-18. [PMID: 24947371 DOI: 10.1007/978-1-4939-0944-5_1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Pharmacological inhibitors of vesicle trafficking possess great promise as valuable analytical tools for the study of a variety of biological processes and as potential therapeutic agents to fight microbial infections and cancer. However, many commonly used trafficking inhibitors are characterized by poor selectivity that diminishes their use in solving basic problems of cell biology or drug development. Recent high-throughput chemical screens intensified the search for novel modulators of vesicle trafficking, and successfully identified a number of small molecules that inhibit exocytosis and endocytosis in different types of mammalian cells. This chapter provides a systematic overview of recently discovered inhibitors of vesicle trafficking. It describes cellular effects and mechanisms of action of novel inhibitors of exocytosis and endocytosis. Furthermore, it pays special attention to the selectivity and possible off-target effects of these inhibitors.
Collapse
Affiliation(s)
- Andrei I Ivanov
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Virginia Commonwealth University, Goodwin Laboratory, 401 College Street, 980035, Richmond, VA, 23298, USA,
| |
Collapse
|
46
|
Destaing O, Ferguson SM, Grichine A, Oddou C, De Camilli P, Albiges-Rizo C, Baron R. Essential function of dynamin in the invasive properties and actin architecture of v-Src induced podosomes/invadosomes. PLoS One 2013; 8:e77956. [PMID: 24348990 PMCID: PMC3857171 DOI: 10.1371/journal.pone.0077956] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 09/06/2013] [Indexed: 01/07/2023] Open
Abstract
The large GTPase dynamin plays a key role in endocytosis but is also localized at numerous actin rich sites. We investigated dynamin functions at podosomes/invadosomes, actin-based cellular adhesion structures implicated in tissue invasion. Podosomes/invadosomes are constituted of long F-actin bundles perpendicular to the substratum (actin cores), connected to randomly arranged F-actin fibers parallel to the substratum (actin cloud). We show here that dynamin depletion in v-Src-transformed fibroblasts triggers a massive disorganization of podosomes/invadosomes (isolated or in rosettes), with a corresponding inhibition of their invasive properties. The action of dynamin at podosomes/invadosomes requires a functional full-length protein, suggesting that the effects of dynamin at these sites and in membrane remodelling during endocytosis are mediated by similar mechanisms. In order to determine direct effect of dynamin depletion on invadosome, an optogenetic approach based on the photosensitizer KillerRed was developed. Acute dynamin photo-inactivation leads to a very rapid disorganization of invadosome without affecting focal adhesions. Dynamin therefore is a key regulator of the architecture of actin in podosomes/invadosomes.
Collapse
Affiliation(s)
- Olivier Destaing
- Institut Albert Bonniot, Université Joseph Fourier; Université Joseph Fourier site Santé, Grenoble cedex, France
- Department of Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| | - Shawn M. Ferguson
- Department of Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Alexei Grichine
- Institut Albert Bonniot, Université Joseph Fourier; Université Joseph Fourier site Santé, Grenoble cedex, France
| | - Christiane Oddou
- Institut Albert Bonniot, Université Joseph Fourier; Université Joseph Fourier site Santé, Grenoble cedex, France
| | - Pietro De Camilli
- Department of Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, United States of America
| | - Corinne Albiges-Rizo
- Institut Albert Bonniot, Université Joseph Fourier; Université Joseph Fourier site Santé, Grenoble cedex, France
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, United States of America
| | - Roland Baron
- Department of Medicine, Harvard Medical School, Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
47
|
N'-[4-(dipropylamino)benzylidene]-2-hydroxybenzohydrazide is a dynamin GTPase inhibitor that suppresses cancer cell migration and invasion by inhibiting actin polymerization. Biochem Biophys Res Commun 2013; 443:511-7. [PMID: 24316215 DOI: 10.1016/j.bbrc.2013.11.118] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 11/29/2013] [Indexed: 11/24/2022]
Abstract
Dynasore, a specific dynamin GTPase inhibitor, suppresses lamellipodia formation and cancer cell invasion by destabilizing actin filaments. In search for novel dynamin inhibitors that suppress actin dynamics more efficiently, dynasore analogues were screened. N'-[4-(dipropylamino)benzylidene]-2-hydroxybenzohydrazide (DBHA) markedly reduced in vitro actin polymerization, and dose-dependently inhibited phosphatidylserine-stimulated dynamin GTPase activity. DBHA significantly suppressed both the recruitment of dynamin 2 to the leading edge in U2OS cells and ruffle formation in H1299 cells. Furthermore, DBHA suppressed both the migration and invasion of H1299 cells by approximately 70%. Furthermore, intratumoral DBHA delivery significantly repressed tumor growth. DBHA was much less cytotoxic than dynasore. These results strongly suggest that DBHA inhibits dynamin-dependent actin polymerization by altering the interactions between dynamin and lipid membranes. DBHA and its derivative may be potential candidates for potent anti-cancer drugs.
Collapse
|
48
|
Abstract
Recent evidence suggests that endocytosis, not exocytosis, can be rate limiting for neurotransmitter release at excitatory CNS synapses during sustained activity and therefore may be a principal determinant of synaptic fatigue. At low stimulation frequencies, the probability of synaptic release is linked to the probability of synaptic retrieval such that evoked release results in proportional retrieval even for release of single synaptic vesicles. The exact mechanism by which the retrieval rates are coupled to release rates, known as compensatory endocytosis, remains unknown. Here we show that inactivation of presynaptic myosin II (MII) decreases the probability of synaptic retrieval. To be able to differentiate between the presynaptic and postsynaptic functions of MII, we developed a live cell substrate patterning technique to create defined neural circuits composed of small numbers of embryonic mouse hippocampal neurons and physically isolated from the surrounding culture. Acute application of blebbistatin to inactivate MII in circuits strongly inhibited evoked release but not spontaneous release. In circuits incorporating both control and MIIB knock-out cells, loss of presynaptic MIIB function correlated with a large decrease in the amplitude of evoked release. Using activity-dependent markers FM1-43 and horseradish peroxidase, we found that MII inactivation greatly slowed vesicular replenishment of the recycling pool but did not impede synaptic release. These results indicate that MII-driven tension or actin dynamics regulate the major pathway for synaptic vesicle retrieval. Changes in retrieval rates determine the size of the recycling pool. The resulting effect on release rates, in turn, brings about changes in synaptic strength.
Collapse
|
49
|
Kapus A, Janmey P. Plasma membrane--cortical cytoskeleton interactions: a cell biology approach with biophysical considerations. Compr Physiol 2013; 3:1231-81. [PMID: 23897686 DOI: 10.1002/cphy.c120015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
From a biophysical standpoint, the interface between the cell membrane and the cytoskeleton is an intriguing site where a "two-dimensional fluid" interacts with an exceedingly complex three-dimensional protein meshwork. The membrane is a key regulator of the cytoskeleton, which not only provides docking sites for cytoskeletal elements through transmembrane proteins, lipid binding-based, and electrostatic interactions, but also serves as the source of the signaling events and molecules that control cytoskeletal organization and remolding. Conversely, the cytoskeleton is a key determinant of the biophysical and biochemical properties of the membrane, including its shape, tension, movement, composition, as well as the mobility, partitioning, and recycling of its constituents. From a cell biological standpoint, the membrane-cytoskeleton interplay underlies--as a central executor and/or regulator--a multitude of complex processes including chemical and mechanical signal transduction, motility/migration, endo-/exo-/phagocytosis, and other forms of membrane traffic, cell-cell, and cell-matrix adhesion. The aim of this article is to provide an overview of the tight structural and functional coupling between the membrane and the cytoskeleton. As biophysical approaches, both theoretical and experimental, proved to be instrumental for our understanding of the membrane/cytoskeleton interplay, this review will "oscillate" between the cell biological phenomena and the corresponding biophysical principles and considerations. After describing the types of connections between the membrane and the cytoskeleton, we will focus on a few key physical parameters and processes (force generation, curvature, tension, and surface charge) and will discuss how these contribute to a variety of fundamental cell biological functions.
Collapse
Affiliation(s)
- András Kapus
- Keenan Research Center, Li Ka Shing Knowledge Institute, St. Michael's Hospital and Department of Surgery, University of Toronto, Ontario, Canada.
| | | |
Collapse
|
50
|
Zebrafish Dynamin is required for maintenance of enveloping layer integrity and the progression of epiboly. Dev Biol 2013; 385:52-66. [PMID: 24161849 DOI: 10.1016/j.ydbio.2013.10.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 10/14/2013] [Accepted: 10/16/2013] [Indexed: 11/21/2022]
Abstract
Epiboly, the first morphogenetic cell movement that occurs in the zebrafish embryo, is the process by which the blastoderm thins and spreads to engulf the yolk cell. This process requires the concerted actions of the deep cells, the enveloping layer (EVL) and the extra-embryonic yolk syncytial layer (YSL). The EVL is mechanically coupled to the YSL which acts as an epiboly motor, generating the force necessary to draw the blastoderm towards the vegetal pole though actomyosin flow and contraction of the actomyosin ring. However, it has been proposed that the endocytic removal of yolk cell membrane just ahead of the advancing blastoderm may also play a role. To assess the contribution of yolk cell endocytosis in driving epiboly movements, we used a combination of drug- and dominant-negative-based approaches to inhibit Dynamin, a large GTPase with a well-characterized role in vesicle scission. We show that Dynamin-dependent endocytosis in the yolk cell is dispensable for epiboly of the blastoderm. However, global inhibition of Dynamin function revealed that Dynamin plays a fundamental role within the blastoderm during epiboly, where it maintains epithelial integrity and the transmission of tension across the EVL. The epithelial defects were associated with disrupted tight junctions and a striking reduction of cortically localized phosphorylated ezrin/radixin/moesin (P-ERM), key regulators of epithelial integrity in other systems. Furthermore, we show that Dynamin maintains EVL and promotes epiboly progression by antagonizing Rho A activity.
Collapse
|