1
|
Zhang X, Baumann C, De La Fuente R. Fluo-Cast-Bright: a deep learning pipeline for the non-invasive prediction of chromatin structure and developmental potential in live oocytes. Commun Biol 2025; 8:141. [PMID: 39880880 PMCID: PMC11779945 DOI: 10.1038/s42003-025-07568-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
In mammalian oocytes, large-scale chromatin organization regulates transcription, nuclear architecture, and maintenance of chromosome stability in preparation for meiosis onset. Pre-ovulatory oocytes with distinct chromatin configurations exhibit profound differences in metabolic and transcriptional profiles that ultimately determine meiotic competence and developmental potential. Here, we developed a deep learning pipeline for the non-invasive prediction of chromatin structure and developmental potential in live mouse oocytes. Our Fluorescence prediction and Classification on Bright-field (Fluo-Cast-Bright) pipeline achieved 91.3% accuracy in the classification of chromatin state in fixed oocytes and 85.7% accuracy in live oocytes. Importantly, transcriptome analysis following non-invasive selection revealed that meiotically competent oocytes exhibit a higher expression of transcripts associated with RNA and protein nuclear export, maternal mRNA deadenylation, histone modifications, chromatin remodeling and signaling pathways regulating microtubule dynamics during the metaphase-I to metaphase-II transition. Fluo-Cast-Bright provides fast and non-invasive selection of meiotically competent oocytes for downstream research and clinical applications.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Claudia Baumann
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Rabindranath De La Fuente
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA.
- Regenerative Bioscience Center (RBC), University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
2
|
Alderfer S, Sun J, Tahtamouni L, Prasad A. Morphological signatures of actin organization in single cells accurately classify genetic perturbations using CNNs with transfer learning. SOFT MATTER 2022; 18:8342-8354. [PMID: 36222484 DOI: 10.1039/d2sm01000c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The actin cytoskeleton plays essential roles in countless cell processes, from cell division to migration to signaling. In cancer cells, cytoskeletal dynamics, cytoskeletal filament organization, and overall cell morphology are known to be altered substantially. We hypothesize that actin fiber organization and cell shape may carry specific signatures of genetic or signaling perturbations. We used convolutional neural networks (CNNs) on a small fluorescence microscopy image dataset of retinal pigment epithelial (RPE) cells and triple-negative breast cancer (TNBC) cells for identifying morphological signatures in cancer cells. Using a transfer learning approach, CNNs could be trained to accurately distinguish between normal and oncogenically transformed RPE cells with an accuracy of about 95% or better at the single cell level. Furthermore, CNNs could distinguish transformed cell lines differing by an oncogenic mutation from each other and could also detect knockdown of cofilin in TNBC cells, indicating that each single oncogenic mutation or cytoskeletal perturbation produces a unique signature in actin morphology. Application of the Local Interpretable Model-Agnostic Explanations (LIME) method for visually interpreting the CNN results revealed features of the global actin structure relevant for some cells and classification tasks. Interestingly, many of these features were supported by previous biological observation. Actin fiber organization is thus a sensitive marker for cell identity, and identification of its perturbations could be very useful for assaying cell phenotypes, including disease states.
Collapse
Affiliation(s)
- Sydney Alderfer
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA.
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Jiangyu Sun
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Lubna Tahtamouni
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
- Department of Biology and Biotechnology, The Hashemite University, Zarqa, Jordan
| | - Ashok Prasad
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA.
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
3
|
Devi SS, Yadav R, Mashangva F, Chaudhary P, Sharma S, Arya R. Generation and Characterization of a Skeletal Muscle Cell-Based Model Carrying One Single Gne Allele: Implications in Actin Dynamics. Mol Neurobiol 2021; 58:6316-6334. [PMID: 34510381 DOI: 10.1007/s12035-021-02549-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 08/28/2021] [Indexed: 12/13/2022]
Abstract
UDP-N-Acetyl glucosamine-2 epimerase/N-acetyl mannosamine kinase (GNE) catalyzes key enzymatic reactions in the biosynthesis of sialic acid. Mutation in GNE gene causes GNE myopathy (GNEM) characterized by adult-onset muscle weakness and degeneration. However, recent studies propose alternate roles of GNE in other cellular processes beside sialic acid biosynthesis, particularly interaction of GNE with α-actinin 1 and 2. Lack of appropriate model system limits drug and treatment options for GNEM as GNE knockout was found to be embryonically lethal. In the present study, we have generated L6 rat skeletal muscle myoblast cell-based model system carrying one single Gne allele where GNE gene is knocked out at exon-3 using AAV mediated SEPT homology recombination (SKM-GNEHz). The cell line was heterozygous for GNE gene with one wild type and one truncated allele as confirmed by sequencing. The phenotype showed reduced GNE epimerase activity with little reduction in sialic acid content. In addition, the heterozygous GNE knockout cells revealed altered cytoskeletal organization with disrupted actin filament. Further, we observed increased levels of RhoA leading to reduced cofilin activity and causing reduced F-actin polymerization. The disturbed signaling cascade resulted in reduced migration of SKM-GNEHz cells. Our study indicates possible role of GNE in regulating actin dynamics and cell migration of skeletal muscle cell. The skeletal muscle cell-based system offers great potential in understanding pathomechanism and target identification for GNEM.
Collapse
Affiliation(s)
| | - Rashmi Yadav
- School of Biotechnology, Jawaharlal Nehru University, 110067, New Delhi, India
| | | | - Priyanka Chaudhary
- School of Biotechnology, Jawaharlal Nehru University, 110067, New Delhi, India
| | - Shweta Sharma
- School of Biotechnology, Jawaharlal Nehru University, 110067, New Delhi, India
| | - Ranjana Arya
- School of Biotechnology, Jawaharlal Nehru University, 110067, New Delhi, India. .,Special Centre for Systems Medicine (Concurrent Faculty), Jawaharlal Nehru University, New Mehrauli Road, 110067, New Delhi, India.
| |
Collapse
|
4
|
Bozal-Basterra L, Gonzalez-Santamarta M, Muratore V, Martín-Martín N, Ercilla A, Rodríguez JA, Carracedo A, Sutherland JD, Barrio R. LUZP1 Controls Cell Division, Migration and Invasion Through Regulation of the Actin Cytoskeleton. Front Cell Dev Biol 2021; 9:624089. [PMID: 33869174 PMCID: PMC8049182 DOI: 10.3389/fcell.2021.624089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/03/2021] [Indexed: 12/21/2022] Open
Abstract
LUZP1 is a centrosomal and actin cytoskeleton-localizing protein that regulates both ciliogenesis and actin filament bundling. As the cytoskeleton and cilia are implicated in metastasis and tumor suppression, we examined roles for LUZP1 in the context of cancer. Here we show that LUZP1 exhibits frequent genomic aberrations in cancer, with a predominance of gene deletions. Furthermore, we demonstrate that CRISPR/Cas9-mediated loss of Luzp1 in mouse fibroblasts promotes cell migration and invasion features, reduces cell viability, and increases cell apoptosis, centriole numbers, and nuclear size while altering the actin cytoskeleton. Loss of Luzp1 also induced changes to ACTR3 (Actin Related Protein 3, also known as ARP3) and phospho-cofilin ratios, suggesting regulatory roles in actin polymerization, beyond its role in filament bundling. Our results point to an unprecedented role for LUZP1 in the regulation of cancer features through the control of actin cytoskeleton.
Collapse
Affiliation(s)
- Laura Bozal-Basterra
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance, Bizkaia Technology Park, Derio, Spain
| | - María Gonzalez-Santamarta
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance, Bizkaia Technology Park, Derio, Spain
| | - Veronica Muratore
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance, Bizkaia Technology Park, Derio, Spain
| | - Natalia Martín-Martín
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance, Bizkaia Technology Park, Derio, Spain
| | - Amaia Ercilla
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance, Bizkaia Technology Park, Derio, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose A Rodríguez
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Spain
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance, Bizkaia Technology Park, Derio, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain.,Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - James D Sutherland
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance, Bizkaia Technology Park, Derio, Spain
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance, Bizkaia Technology Park, Derio, Spain
| |
Collapse
|
5
|
Lazki-Hagenbach P, Klein O, Sagi-Eisenberg R. The actin cytoskeleton and mast cell function. Curr Opin Immunol 2021; 72:27-33. [PMID: 33765561 DOI: 10.1016/j.coi.2021.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/11/2021] [Accepted: 03/02/2021] [Indexed: 10/21/2022]
Abstract
The application of high and super-resolution microscopy techniques has extended the possibilities of studying actin dynamics in mast cells (MCs). These studies demonstrated the close correlation between actin-driven changes in cell morphology and the functions that MC perform during their life cycle. Dynamic conversions between actin polymerization and depolymerization support MC degranulation and leading to the release of the preformed, secretory granule (SG)-contained, inflammatory mediators. Cell flattening inflicting an actin porous geometry and clearing of cortical actin, characterize the secretory actin phenotype. In contrast, pericentral actin clusters, that entrap the SGs, characterize the migratory actin phenotype, which supports MC migration, but restricts MC degranulation. Multiple actin binding and actin interacting proteins regulate these actin rearrangements, in compliance with the signals elicited by the respective activating receptors. Here, we review recent findings on the interplay between the actin cytoskeleton and MC migration and degranulation.
Collapse
Affiliation(s)
- Pia Lazki-Hagenbach
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ofir Klein
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ronit Sagi-Eisenberg
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
6
|
Ruggiero C, Lalli E. Targeting the cytoskeleton against metastatic dissemination. Cancer Metastasis Rev 2021; 40:89-140. [PMID: 33471283 DOI: 10.1007/s10555-020-09936-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/08/2020] [Indexed: 02/08/2023]
Abstract
Cancer is a pathology characterized by a loss or a perturbation of a number of typical features of normal cell behaviour. Indeed, the acquisition of an inappropriate migratory and invasive phenotype has been reported to be one of the hallmarks of cancer. The cytoskeleton is a complex dynamic network of highly ordered interlinking filaments playing a key role in the control of fundamental cellular processes, like cell shape maintenance, motility, division and intracellular transport. Moreover, deregulation of this complex machinery contributes to cancer progression and malignancy, enabling cells to acquire an invasive and metastatic phenotype. Metastasis accounts for 90% of death from patients affected by solid tumours, while an efficient prevention and suppression of metastatic disease still remains elusive. This results in the lack of effective therapeutic options currently available for patients with advanced disease. In this context, the cytoskeleton with its regulatory and structural proteins emerges as a novel and highly effective target to be exploited for a substantial therapeutic effort toward the development of specific anti-metastatic drugs. Here we provide an overview of the role of cytoskeleton components and interacting proteins in cancer metastasis with a special focus on small molecule compounds interfering with the actin cytoskeleton organization and function. The emerging involvement of microtubules and intermediate filaments in cancer metastasis is also reviewed.
Collapse
Affiliation(s)
- Carmen Ruggiero
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, CNRS, 660 route des Lucioles-Sophia Antipolis, 06560, Valbonne, France.
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560, Valbonne, France.
| | - Enzo Lalli
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
- Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des Lucioles - Sophia Antipolis, 06560, Valbonne, France
| |
Collapse
|
7
|
Werle SD, Schwab JD, Tatura M, Kirchhoff S, Szekely R, Diels R, Ikonomi N, Sipos B, Sperveslage J, Gress TM, Buchholz M, Kestler HA. Unraveling the Molecular Tumor-Promoting Regulation of Cofilin-1 in Pancreatic Cancer. Cancers (Basel) 2021; 13:725. [PMID: 33578795 PMCID: PMC7916621 DOI: 10.3390/cancers13040725] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/26/2021] [Accepted: 02/07/2021] [Indexed: 12/24/2022] Open
Abstract
Cofilin-1 (CFL1) overexpression in pancreatic cancer correlates with high invasiveness and shorter survival. Besides a well-documented role in actin remodeling, additional cellular functions of CFL1 remain poorly understood. Here, we unraveled molecular tumor-promoting functions of CFL1 in pancreatic cancer. For this purpose, we first show that a knockdown of CFL1 results in reduced growth and proliferation rates in vitro and in vivo, while apoptosis is not induced. By mechanistic modeling we were able to predict the underlying regulation. Model simulations indicate that an imbalance in actin remodeling induces overexpression and activation of CFL1 by acting on transcription factor 7-like 2 (TCF7L2) and aurora kinase A (AURKA). Moreover, we could predict that CFL1 impacts proliferation and apoptosis via the signal transducer and activator of transcription 3 (STAT3). These initial model-based regulations could be substantiated by studying protein levels in pancreatic cancer cell lines and human datasets. Finally, we identified the surface protein CD44 as a promising therapeutic target for pancreatic cancer patients with high CFL1 expression.
Collapse
Affiliation(s)
- Silke D. Werle
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (S.D.W.); (J.D.S.); (R.S.); (N.I.)
| | - Julian D. Schwab
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (S.D.W.); (J.D.S.); (R.S.); (N.I.)
| | - Marina Tatura
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, 35043 Marburg, Germany; (M.T.); (S.K.); (R.D.); (T.M.G.); (M.B.)
| | - Sandra Kirchhoff
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, 35043 Marburg, Germany; (M.T.); (S.K.); (R.D.); (T.M.G.); (M.B.)
| | - Robin Szekely
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (S.D.W.); (J.D.S.); (R.S.); (N.I.)
| | - Ramona Diels
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, 35043 Marburg, Germany; (M.T.); (S.K.); (R.D.); (T.M.G.); (M.B.)
| | - Nensi Ikonomi
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (S.D.W.); (J.D.S.); (R.S.); (N.I.)
| | - Bence Sipos
- Institute of Pathology, University of Tübingen, 72076 Tübingen, Germany; (B.S.); (J.S.)
| | - Jan Sperveslage
- Institute of Pathology, University of Tübingen, 72076 Tübingen, Germany; (B.S.); (J.S.)
| | - Thomas M. Gress
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, 35043 Marburg, Germany; (M.T.); (S.K.); (R.D.); (T.M.G.); (M.B.)
| | - Malte Buchholz
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, 35043 Marburg, Germany; (M.T.); (S.K.); (R.D.); (T.M.G.); (M.B.)
| | - Hans A. Kestler
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (S.D.W.); (J.D.S.); (R.S.); (N.I.)
| |
Collapse
|
8
|
Luo Y, Hu J, Liu Y, Li L, Li Y, Sun B, Kong R. Invadopodia: A potential target for pancreatic cancer therapy. Crit Rev Oncol Hematol 2021; 159:103236. [PMID: 33482351 DOI: 10.1016/j.critrevonc.2021.103236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 01/05/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023] Open
Abstract
Dissemination of cancer cells is an intricate multistep process that represents the most deadly aspect of cancer. Cancer cells form F-actin-rich protrusions known as invadopodia to invade surrounding tissues, blood vessels and lymphatics. A number of studies have demonstrated the significant roles of invadopodia in cancer. Therefore, the specific cells and molecules involved in invadopodia activity can provide as therapeutic targets. In this review, we included a thorough overview of studies in invadopodia and discussed their functions in cancer metastasis. We then presented the specific cells and molecules involved in invadopodia activity in pancreatic cancer and analyzed their suitability to be effective therapeutic targets. Currently, drugs targeting invadopodia and relevant clinical trials are negligible. Here, we highlighted the significance of potential drugs and discussed future obstacles in implementing clinical trials. This review presents a new perspective on invadopodia-induced pancreatic cancer metastasis and may prosper the development of targeted therapeutics against pancreatic cancer.
Collapse
Affiliation(s)
- Yan Luo
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jisheng Hu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Kong
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
9
|
Regulation of actin dynamics in dendritic spines: Nanostructure, molecular mobility, and signaling mechanisms. Mol Cell Neurosci 2020; 109:103564. [DOI: 10.1016/j.mcn.2020.103564] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/04/2020] [Indexed: 12/16/2022] Open
|
10
|
Lee S, Kumar S. Cofilin is required for polarization of tension in stress fiber networks during migration. J Cell Sci 2020; 133:jcs243873. [PMID: 32501289 PMCID: PMC7358140 DOI: 10.1242/jcs.243873] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/11/2020] [Indexed: 01/04/2023] Open
Abstract
Cell migration is associated with the establishment of defined leading and trailing edges, which in turn requires polarization of contractile forces. While the actomyosin stress fiber (SF) network plays a critical role in enforcing this polarity, precisely how this asymmetry is established remains unclear. Here, we provide evidence for a model in which the actin-severing protein cofilin (specifically cofilin-1) participates in symmetry breakage by removing low-tension actomyosin filaments during transverse arc assembly. Cofilin knockdown (KD) produces a non-polarized SF architecture that cannot be rescued with chemokines or asymmetric matrix patterns. Whereas cofilin KD increases whole-cell prestress, it decreases prestress within single SFs, implying an accumulation of low-tension SFs. This notion is supported by time-lapse imaging, which reveals weakly contractile and incompletely fused transverse arcs. Confocal and super-resolution imaging further associate this failed fusion with the presence of crosslinker-rich, tropomyosin-devoid nodes at the junctions of multiple transverse arc fragments and dorsal SFs. These results support a model in which cofilin facilitates the formation of high-tension transverse arcs, thereby promoting mechanical asymmetry.
Collapse
Affiliation(s)
- Stacey Lee
- UC Berkeley-UCSF Graduate Program in Bioengineering, USA
- UC Berkeley Department of Bioengineering, UC Berkeley, CA, USA
| | - Sanjay Kumar
- UC Berkeley-UCSF Graduate Program in Bioengineering, USA
- UC Berkeley Department of Bioengineering, UC Berkeley, CA, USA
- UC Berkeley Department of Chemical and Biomolecular Engineering, 274A Stanley Hall #1762, UC Berkeley, Berkeley, CA 94720-1762, UC Berkeley, CA, USA
| |
Collapse
|
11
|
Arzash S, McCall PM, Feng J, Gardel ML, MacKintosh FC. Stress relaxation in F-actin solutions by severing. SOFT MATTER 2019; 15:6300-6307. [PMID: 31342050 DOI: 10.1039/c9sm01263j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Networks of filamentous actin (F-actin) are important for the mechanics of most animal cells. These cytoskeletal networks are highly dynamic, with a variety of actin-associated proteins that control cross-linking, polymerization and force generation in the cytoskeleton. Inspired by recent rheological experiments on reconstituted solutions of dynamic actin filaments, we report a theoretical model that describes stress relaxation behavior of these solutions in the presence of severing proteins. We show that depending on the kinetic rates of assembly, disassembly, and severing, one can observe both length-dependent and length-independent relaxation behavior.
Collapse
Affiliation(s)
- Sadjad Arzash
- Department of Chemical & Biomolecular Engineering, Rice University, Houston, TX 77005, USA. and Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA
| | - Patrick M McCall
- Department of Physics, University of Chicago, Chicago, IL 60637, USA and James Franck Institute, University of Chicago, Chicago, IL 60637, USA and Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany and Max Planck Institute for the Physics of Complex Systems, Nöthnitzerstraße 38, 01187 Dresden, Germany and Center for Systems Biology Dresden, Pfotenhauerstraße 108, 01307, Dresden, Germany
| | - Jingchen Feng
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA
| | - Margaret L Gardel
- Department of Physics, University of Chicago, Chicago, IL 60637, USA and James Franck Institute, University of Chicago, Chicago, IL 60637, USA and Institute for Biophysical Dynamics, University of Chicago, IL 60637, USA
| | - Fred C MacKintosh
- Department of Chemical & Biomolecular Engineering, Rice University, Houston, TX 77005, USA. and Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA and Department of Chemistry, Rice University, Houston, TX 77005, USA and Department of Physics & Astronomy, Rice University, Houston, TX 77005, USA
| |
Collapse
|
12
|
Gulvady AC, Forsythe IJ, Turner CE. Hic-5 regulates Src-induced invadopodia rosette formation and organization. Mol Biol Cell 2019; 30:1298-1313. [PMID: 30893012 PMCID: PMC6724605 DOI: 10.1091/mbc.e18-10-0629] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fibroblasts transformed by the proto-oncogene Src form individual invadopodia that can spontaneously self-organize into large matrix-degrading superstructures called rosettes. However, the mechanisms by which the invadopodia can spatiotemporally reorganize their architecture is not well understood. Here, we show that Hic-5, a close relative of the scaffold protein paxillin, is essential for the formation and organization of rosettes in active Src-transfected NIH3T3 fibroblasts and cancer-associated fibroblasts. Live cell imaging, combined with domain-mapping analysis of Hic-5, identified critical motifs as well as phosphorylation sites that are required for the formation and dynamics of rosettes. Using pharmacological inhibition and mutant expression, we show that FAK kinase activity, along with its proximity to and potential interaction with the LD2,3 motifs of Hic-5, is necessary for rosette formation. Invadopodia dynamics and their coalescence into rosettes were also dependent on Rac1, formin, and myosin II activity. Superresolution microscopy revealed the presence of formin FHOD1 and INF2-mediated unbranched radial F-actin fibers emanating from invadopodia and rosettes, which may facilitate rosette formation. Collectively, our data highlight a novel role for Hic-5 in orchestrating the organization of invadopodia into higher-order rosettes, which may promote the localized matrix degradation necessary for tumor cell invasion.
Collapse
Affiliation(s)
- Anushree C Gulvady
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Ian J Forsythe
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Christopher E Turner
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
13
|
The actin cytoskeleton is important for rotavirus internalization and RNA genome replication. Virus Res 2019; 263:27-33. [PMID: 30639190 PMCID: PMC7173133 DOI: 10.1016/j.virusres.2019.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/13/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023]
Abstract
Different stages of the rotavirus lifecycle depend on the dynamics of the actin cytoskeleton. Alpha-actinin, Diaph, and the GTPase Cdc42 are important for virus entry. The GTPAse Rac1 is required for maximal viral RNA synthesis.
Numerous host factors are required for the efficient replication of rotavirus, including the activation and inactivation of several cell signaling pathways. One of the cellular structures that are reorganized during rotavirus infection is the actin cytoskeleton. In this work, we report that the dynamics of the actin microfilaments are important at different stages of the virus life cycle, specifically, during virus internalization and viral RNA synthesis at 6 h post-infection. Our results show that the actin-binding proteins alpha-actinin 4 and Diaph, as well as the Rho-family small GTPase Cdc42 are necessary for an efficient virus entry, while GTPase Rac1 is required for maximal viral RNA synthesis.
Collapse
|
14
|
Islam SMA, Patel R, Bommareddy RR, Khalid KM, Acevedo-Duncan M. The modulation of actin dynamics via atypical Protein Kinase-C activated Cofilin regulates metastasis of colorectal cancer cells. Cell Adh Migr 2018; 13:106-120. [PMID: 30417717 PMCID: PMC6527392 DOI: 10.1080/19336918.2018.1546513] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in the United States. The exact mechanism of CRC cells metastasis is poorly understood. Actin polymerization is thought to be an initial step in the cancer cell motility cycle which drives the formation of cell protrusions and defines the direction of migration. Cofilin, a significant actin-regulating molecule, regulates the migration of cancer cells by the formation of lamellipodia and filopodia, however, little is known about the upstream regulation of cofilin. In this study, the effect of atypical Protein Kinase C (atypical PKC) on Cofilin activity in CRC was studied. This study demonstrates that the atypical PKC inhibition impedes the metastasis of CRC cells by increasing phospho-Cofilin (S3) and changing actin organization.
Collapse
Affiliation(s)
- S M Anisul Islam
- a Department of Chemistry , University of South Florida , Tampa , FL , USA
| | - Rekha Patel
- a Department of Chemistry , University of South Florida , Tampa , FL , USA
| | | | | | | |
Collapse
|
15
|
Jin ZL, Jo YJ, Namgoong S, Kim NH. CAP1 mediated actin cycling via ADF/cofilin is essential for asymmetric division in mouse oocytes. J Cell Sci 2018; 131:jcs.222356. [DOI: 10.1242/jcs.222356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 10/23/2018] [Indexed: 11/20/2022] Open
Abstract
Dynamic reorganization of the actin cytoskeleton is fundamental to a number of cellular events, and various actin-regulatory proteins modulate actin polymerization and depolymerization. Cyclase-associated proteins (CAPs), highly conserved actin monomer-binding proteins, have been known to promote actin disassembly by enhancing the actin-severing activity of ADF/cofilin. In this study, we found that CAP1 regulated actin remodeling during mouse oocyte maturation. Efficient actin disassembly during oocyte maturation is essential for asymmetric division and cytokinesis. CAP1 knockdown impaired meiotic spindle migration and asymmetric division, and it resulted in an accumulation of excessive actin filaments near the spindles. In contrast, CAP1 overexpression reduced actin mesh levels. CAP1 knockdown also rescued the decrease in cofilin overexpression-mediated actin levels, and simultaneous expression of human CAP1 (hCAP1) and cofilin synergistically decreased cytoplasmic actin levels. Overexpression of hCAP1 decreased the amount of phosphorylated cofilin, indicating that CAP1 facilitated actin depolymerization via interaction with ADF/cofilin during mouse oocyte maturation. Taken together, our results provide evidence of the importance of dynamic actin recycling by CAP1 and cofilin in the asymmetric division of mouse female gametes.
Collapse
Affiliation(s)
- Zhe-Long Jin
- Department of Animal Sciences, Chungbuk National University, Cheongju 361-763, Korea
| | - Yu-Jin Jo
- Department of Animal Sciences, Chungbuk National University, Cheongju 361-763, Korea
| | - Suk Namgoong
- Department of Animal Sciences, Chungbuk National University, Cheongju 361-763, Korea
| | - Nam-Hyung Kim
- Department of Animal Sciences, Chungbuk National University, Cheongju 361-763, Korea
| |
Collapse
|
16
|
Yuan X, Wang X, Gu B, Ma Y, Liu Y, Sun M, Kong J, Sun W, Wang H, Zhou F, Gao S. Directional Migration in Esophageal Squamous Cell Carcinoma (ESCC) is Epigenetically Regulated by SET Nuclear Oncogene, a Member of the Inhibitor of Histone Acetyltransferase Complex. Neoplasia 2017; 19:868-884. [PMID: 28938158 PMCID: PMC5608591 DOI: 10.1016/j.neo.2017.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/04/2017] [Accepted: 08/04/2017] [Indexed: 11/26/2022] Open
Abstract
Directional cell migration is of fundamental importance to a variety of biological events, including metastasis of malignant cells. Herein, we specifically investigated SET oncoprotein, a subunit of the recently identified inhibitor of acetyltransferases (INHAT) complex and identified its role in the establishment of front-rear cell polarity and directional migration in Esophageal Squamous Cell Carcinoma (ESCC). We further define the molecular circuits that govern these processes by showing that SET modulated DOCK7/RAC1 and cofilin signaling events. Moreover, a detailed analysis of the spatial distribution of RAC1 and cofilin allowed us to decipher the synergistical contributions of the two in coordinating the advancing dynamics by measuring architectures, polarities, and cytoskeletal organizations of the lamellipodia leading edges. In further investigations in vivo, we identified their unique role at multiple levels of the invasive cascade for SET cell and indicate the necessity for their functional balance to enable efficient invasion as well. Additionally, SET epigenetically repressed miR-30c expression by deacetylating histones H2B and H4 on its promoter, which was functionally important for the biological effects of SET in our cell-context. Finally, we corroborated our findings in vivo by evaluating the clinical relevance of SET signaling in the metastatic burden in mice and a large series of patients with ESCC at diagnosis, observing it's significance in predicting metastasis formation. Our findings uncovered a novel signaling network initiated by SET that epigenetically modulated ESCC properties and suggest that targeting the regulatory axis might be a promising strategy to inhibit migration and metastasis.
Collapse
Affiliation(s)
- Xiang Yuan
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China, 471003; Department of Medical Oncology, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China, 471003
| | - Xinshuai Wang
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China, 471003; Department of Medical Oncology, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China, 471003
| | - Bianli Gu
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China, 471003
| | - Yingjian Ma
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China, 471003
| | - Yiwen Liu
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China, 471003
| | - Man Sun
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China, 471003
| | - Jinyu Kong
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China, 471003
| | - Wei Sun
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China, 471003
| | - Huizhi Wang
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Room 263D, 501 South Preston Street, Louisville, KY 40202, USA
| | - Fuyou Zhou
- Department of Thoracic Surgery, Anyang Tumor Hospital, Anyang, China, 455000
| | - Shegan Gao
- Henan Key Laboratory of Cancer Epigenetics; Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China, 471003; Department of Medical Oncology, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China, 471003.
| |
Collapse
|
17
|
Bagnato P, Castagnino A, Cortese K, Bono M, Grasso S, Bellese G, Daniele T, Lundmark R, Defilippi P, Castagnola P, Tacchetti C. Cooperative but distinct early co-signaling events originate from ERBB2 and ERBB1 receptors upon trastuzumab treatment in breast cancer cells. Oncotarget 2017; 8:60109-60122. [PMID: 28947957 PMCID: PMC5601125 DOI: 10.18632/oncotarget.17686] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 04/03/2017] [Indexed: 12/19/2022] Open
Abstract
ERBB2 receptor belongs to the ERBB tyrosine kinase receptor family. At variance to the other family members, ERBB2 is a constitutively active orphan receptor. Upon ligand binding and activation, ERBB receptors form homo- or hetero-dimers with the other family members, including ERBB2, promoting an intracellular signaling cascade. ERBB2 is the preferred dimerization partner and ERBB2 heterodimers signaling is stronger and longer acting compared to heterodimers between other ERBB members. The specific contribution of ERBB2 in heterodimer signaling is still undefined. Here we report the formation of circular dorsal ruffles (CDRs) upon treatment of the ERBB2-overexpressing breast cancer cell lines SK-BR-3 and ZR751 with Trastuzumab, a therapeutic humanized monoclonal antibody directed against ERBB2. We found that in SK-BR-3 cells Trastuzumab leads to surface redistribution of ERBB2 and ERBB1 in CDRs, and that the ERBB2-dependent ERK1/2 phosphorylation and ERBB1 expression are both required for CDR formation. In particular, in these cells CDR formation requires activation of both the protein regulator of actin polymerization N-WASP, mediated by ERK1/2, and of the actin depolymerizing protein cofilin, mediated by ERBB1. Furthermore, we suggest that this latter event may be inhibited by the negative cell motility regulator p140Cap, as we found that p140Cap overexpression led to cofilin deactivation and inhibition of CDR formation. In conclusion, here we show for the first time an ERBB2-specific signaling contribution to an ERBB2/ERBB1 heterodimer, in the activation of a complex biological process such as the formation of CDRs.
Collapse
Affiliation(s)
- Paola Bagnato
- DIMES, Dipartimento di Medicina Sperimentale, Anatomia Umana, Università di Genova, Genova, Italy
| | - Alessia Castagnino
- DIMES, Dipartimento di Medicina Sperimentale, Anatomia Umana, Università di Genova, Genova, Italy
| | - Katia Cortese
- DIMES, Dipartimento di Medicina Sperimentale, Anatomia Umana, Università di Genova, Genova, Italy
| | - Maria Bono
- DIMES, Dipartimento di Medicina Sperimentale, Anatomia Umana, Università di Genova, Genova, Italy
| | - Silvia Grasso
- Molecular Biotechnology Centre and Department of Genetics, Biology and Biochemistry, Torino, Italy
| | - Grazia Bellese
- DIMES, Dipartimento di Medicina Sperimentale, Anatomia Umana, Università di Genova, Genova, Italy
| | - Tiziana Daniele
- San Raffaele Scientific Institute, Experimental Imaging Centre, Milan, Italy
| | - Richard Lundmark
- Department of Medical Biochemistry and Biophysics, Umea University, Umea, Sweden
| | - Paola Defilippi
- Molecular Biotechnology Centre and Department of Genetics, Biology and Biochemistry, Torino, Italy
| | - Patrizio Castagnola
- Department of Integrated Oncological Therapies, IRCCS AOU - San Martino - IST, Largo Rosanna Benzi, Genova, Italy
| | - Carlo Tacchetti
- DIMES, Dipartimento di Medicina Sperimentale, Anatomia Umana, Università di Genova, Genova, Italy.,San Raffaele Scientific Institute, Experimental Imaging Centre, Milan, Italy
| |
Collapse
|
18
|
Christensen JR, Hocky GM, Homa KE, Morganthaler AN, Hitchcock-DeGregori SE, Voth GA, Kovar DR. Competition between Tropomyosin, Fimbrin, and ADF/Cofilin drives their sorting to distinct actin filament networks. eLife 2017; 6. [PMID: 28282023 PMCID: PMC5404920 DOI: 10.7554/elife.23152] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 03/09/2017] [Indexed: 12/15/2022] Open
Abstract
The fission yeast actin cytoskeleton is an ideal, simplified system to investigate fundamental mechanisms behind cellular self-organization. By focusing on the stabilizing protein tropomyosin Cdc8, bundling protein fimbrin Fim1, and severing protein coffin Adf1, we examined how their pairwise and collective interactions with actin filaments regulate their activity and segregation to functionally diverse F-actin networks. Utilizing multi-color TIRF microscopy of in vitro reconstituted F-actin networks, we observed and characterized two distinct Cdc8 cables loading and spreading cooperatively on individual actin filaments. Furthermore, Cdc8, Fim1, and Adf1 all compete for association with F-actin by different mechanisms, and their cooperative association with actin filaments affects their ability to compete. Finally, competition between Fim1 and Adf1 for F-actin synergizes their activities, promoting rapid displacement of Cdc8 from a dense F-actin network. Our findings reveal that competitive and cooperative interactions between actin binding proteins help define their associations with different F-actin networks. DOI:http://dx.doi.org/10.7554/eLife.23152.001 Cells use a protein called actin to provide shape, to generate the forces needed for cells to divide, and for many other essential processes. Inside a cell, individual actin proteins join up to form long filaments. These actin filaments are organized in different ways to make networks that have distinct properties, each tailored for a specific process. For instance, bundles of straight actin filaments help a cell to divide, whereas a network of branched actin filaments allows cells to move. The different proteins that bind to actin filaments influence how quickly actin filaments are assembled and organized into networks. Therefore, many of the properties of an actin filament network are due to the actin binding proteins that are associated with it. Two actin binding proteins called fimbrin and cofilin associate with a type of actin filament network known as the actin patch. A third actin binding protein called tropomyosin associates with a different network that forms a ring. It is not known how particular actin binding proteins choose to associate with one actin network instead of another. Christensen et al. used a fluorescence microscopy technique to study how fimbrin, cofilin and tropomyosin associate with different actin networks in a single-celled organism called fission yeast. This technique involved incubating actin and actin binding proteins together in a microscope chamber. The experiments show that some actin binding proteins, like tropomyosin, cooperate to bind to actin. Individual tropomyosin molecules find it difficult to bind actin filaments on their own, but once one tropomyosin molecule is attached to the filament, others rapidly join to coat the filament. On the other hand, some actin-binding proteins compete for binding to filaments. For example, the binding of fimbrin to actin filaments causes tropomyosin to be removed from the actin network. Further experiments revealed that fimbrin and cofilin work with each other to rapidly generate a dense actin network and displace tropomyosin. Together, the findings of Christensen et al. suggest that competitions between actin binding proteins determine which actin binding proteins are associated with an actin network. The next challenge is to understand how the most competitive actin-binding proteins are kept off actin networks where they do not belong. Further studies will shed light on how these interactions cause large changes in how the cell is organized. DOI:http://dx.doi.org/10.7554/eLife.23152.002
Collapse
Affiliation(s)
- Jenna R Christensen
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, United States
| | - Glen M Hocky
- Department of Chemistry, The University of Chicago, Chicago, United States.,James Franck Institute, The University of Chicago, Chicago, United States
| | - Kaitlin E Homa
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, United States
| | - Alisha N Morganthaler
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, United States
| | - Sarah E Hitchcock-DeGregori
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, United States
| | - Gregory A Voth
- Department of Chemistry, The University of Chicago, Chicago, United States.,James Franck Institute, The University of Chicago, Chicago, United States.,Computation Institute, The University of Chicago, Chicago, United States.,Institute for Biophysical Dynamics, The University of Chicago, Chicago, United States
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, United States.,Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, United States
| |
Collapse
|
19
|
Abstract
Drebrin is a family of actin-binding proteins with two known members called drebrin A and E. Apart from the ability to stabilize F-actin microfilaments via their actin-binding domains near the N-terminus, drebrin also regulates multiple cellular functions due to its unique ability to recruit multiple binding partners to a specific cellular domain, such as the seminiferous epithelium during the epithelial cycle of spermatogenesis. Recent studies have illustrated the role of drebrin E in the testis during spermatogenesis in particular via its ability to recruit branched actin polymerization protein known as actin-related protein 3 (Arp3), illustrating its involvement in modifying the organization of actin microfilaments at the ectoplasmic specialization (ES) which includes the testis-specific anchoring junction at the Sertoli-spermatid (apical ES) interface and at the Sertoli cell-cell (basal ES) interface. These data are carefully evaluated in light of other recent findings herein regarding the role of drebrin in actin filament organization at the ES. We also provide the hypothetical model regarding its involvement in germ cell transport during the epithelial cycle in the seminiferous epithelium to support spermatogenesis.
Collapse
Affiliation(s)
- Haiqi Chen
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave., New York, NY, 10065, USA
| | - Michelle W M Li
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave., New York, NY, 10065, USA
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave., New York, NY, 10065, USA.
| |
Collapse
|
20
|
Parreno J, Nabavi Niaki M, Andrejevic K, Jiang A, Wu PH, Kandel RA. Interplay between cytoskeletal polymerization and the chondrogenic phenotype in chondrocytes passaged in monolayer culture. J Anat 2016; 230:234-248. [PMID: 27807861 DOI: 10.1111/joa.12554] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2016] [Indexed: 12/19/2022] Open
Abstract
Tubulin and actin exist as monomeric units that polymerize to form either microtubules or filamentous actin. As the polymerization status (monomeric/polymeric ratio) of tubulin and/or actin have been shown to be important in regulating gene expression and phenotype in non-chondrocyte cells, the objective of this study was to examine the role of cytoskeletal polymerization on the chondrocyte phenotype. We hypothesized that actin and/or tubulin polymerization status modulates the chondrocyte phenotype during monolayer culture as well as in 3D culture during redifferentiation. To test this hypothesis, articular chondrocytes were grown and passaged in 2D monolayer culture. Cell phenotype was investigated by assessing cell morphology (area and circularity), actin/tubulin content, organization and polymerization status, as well as by determination of proliferation, fibroblast and cartilage matrix gene expression with passage number. Bovine chondrocytes became larger, more elongated, and had significantly (P < 0.05) increased gene expression of proliferation-associated molecules (cyclin D1 and ki67), as well as significantly (P < 0.05) decreased cartilage matrix (type II collagen and aggrecan) and increased fibroblast-like matrix, type I collagen (COL1), gene expression by passage 2 (P2). Although tubulin polymerization status was not significantly (P > 0.05) modulated, actin polymerization was increased in bovine P2 cells. Actin depolymerization, but not tubulin depolymerization, promoted the chondrocyte phenotype by inducing cell rounding, increasing aggrecan and reducing COL1 expression. Knockdown of actin depolymerization factor, cofilin, in these cells induced further P2 cell actin polymerization and increased COL1 gene expression. To confirm that actin status regulated COL1 gene expression in human P2 chondrocytes, human P2 chondrocytes were exposed to cytochalasin D. Cytochalasin D decreased COL1 gene expression in human passaged chondrocytes. Furthermore, culture of bovine P2 chondrocytes in 3D culture on porous bone substitute resulted in actin depolymerization, which correlated with decreased expression of COL1 and proliferation molecules. In 3D cultures, aggrecan gene expression was increased by cytochalasin D treatment and COL1 was further decreased. These results reveal that actin polymerization status regulates chondrocyte dedifferentiation. Reorganization of the cytoskeleton by actin depolymerization appears to be an active regulatory mechanism for redifferentiation of passaged chondrocytes.
Collapse
Affiliation(s)
- Justin Parreno
- CIHR-BioEngineering of Skeletal Tissues Team, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Mortah Nabavi Niaki
- CIHR-BioEngineering of Skeletal Tissues Team, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Katarina Andrejevic
- CIHR-BioEngineering of Skeletal Tissues Team, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Amy Jiang
- CIHR-BioEngineering of Skeletal Tissues Team, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Po-Han Wu
- CIHR-BioEngineering of Skeletal Tissues Team, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Rita A Kandel
- CIHR-BioEngineering of Skeletal Tissues Team, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada
| |
Collapse
|
21
|
Chai X, Frotscher M. How does Reelin signaling regulate the neuronal cytoskeleton during migration? NEUROGENESIS 2016; 3:e1242455. [PMID: 28265585 DOI: 10.1080/23262133.2016.1242455] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/15/2016] [Accepted: 09/25/2016] [Indexed: 01/17/2023]
Abstract
Neuronal migration is an essential step in the formation of laminated brain structures. In the developing cerebral cortex, pyramidal neurons migrate toward the Reelin-containing marginal zone. Reelin is an extracellular matrix protein synthesized by Cajal-Retzius cells. In this review, we summarize our recent results and hypotheses on how Reelin might regulate neuronal migration by acting on the actin and microtubule cytoskeleton. By binding to ApoER2 receptors on the migrating neurons, Reelin induces stabilization of the leading processes extending toward the marginal zone, which involves Dab1 phosphorylation, adhesion molecule expression, cofilin phosphorylation and inhibition of tau phosphorylation. By binding to VLDLR and integrin receptors, Reelin interacts with Lis1 and induces nuclear translocation, accompanied by the ubiquitination of phosphorylated Dab1. Eventually Reelin induces clustering of its receptors resulting in the endocytosis of a Reelin/receptor complex (particularly VLDLR). The resulting decrease in Reelin contributes to neuronal arrest at the marginal zone.
Collapse
Affiliation(s)
- Xuejun Chai
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg (ZMNH) , Hamburg, Germany
| | - Michael Frotscher
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg (ZMNH) , Hamburg, Germany
| |
Collapse
|
22
|
Zhou T, Wang CH, Yan H, Zhang R, Zhao JB, Qian CF, Xiao H, Liu HY. Inhibition of the Rac1-WAVE2-Arp2/3 signaling pathway promotes radiosensitivity via downregulation of cofilin-1 in U251 human glioma cells. Mol Med Rep 2016; 13:4414-20. [PMID: 27052944 DOI: 10.3892/mmr.2016.5088] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 03/16/2016] [Indexed: 11/05/2022] Open
Abstract
The Ras-related C3 botulinum toxin substrate 1 (Rac1)-WASP-family verprolin-homologous protein-2 (WAVE2)-actin-related protein 2/3 (Arp2/3) signaling pathway has been identified to be involved in cell migration and invasion in various types of cancer cell. Cofilin‑1 (CFL‑1), which is regulated by the Rac1‑WAVE2‑Arp2/3 signaling pathway, may promote radioresistance in glioma. Therefore, the present study aimed to investigate the potential role of the Rac1‑WAVE2‑Arp2/3 signaling pathway in radioresistance in U251 human glioma cells and elucidate its affect on CFL‑1 expression. Western blot analysis was performed to evaluate the protein expression of CFL‑1. In the present study, Rac1 was inhibited by NSC 23766, WAVE2 was inhibited by transfection with short hairpin (sh)RNA‑WAVE2 using Lipofectamine™ 2000 and Arp2/3 was inhibited by CK‑666. Cell viability was measured using the 3‑(4,5‑dimethylthiazol‑2‑yl)-2,5‑diphenyltetrazolium bromide assay, the cell migration ability was examined by a wound‑healing assay, and the cell invasion ability was assessed using a Transwell culture chamber system. The results showed that inhibition of the Rac1‑WAVE2‑Arp2/3 signaling pathway using NSC 23766, shRNA‑WAVE2 or CK‑666 reduced the cell viability, migration and invasion abilities in U251 human glioma cells, concordant with a reduced expression of CFL‑1. Furthermore, the expression of CFL‑1 was significantly increased in radioresistant U251 glioma cells when compared with normal U251 human glioma cells. These findings indicate that inhibition of the Rac1‑WAVE2‑Arp2/3 signaling pathway may promote radiosensitivity, which may partially result from the downregulation of CFL‑1 in U251 human glioma cells.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Neurosurgery, Nanjing Medical University, Affiliated Nanjing Brain Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Chen-Han Wang
- Department of Neurosurgery, Nanjing Medical University, Affiliated Nanjing Brain Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Hua Yan
- Department of Neurosurgery, Nanjing Medical University, Affiliated Nanjing Brain Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Rui Zhang
- Department of Neurosurgery, Nanjing Medical University, Affiliated Nanjing Brain Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Jin-Bing Zhao
- Department of Neurosurgery, Nanjing Medical University, Affiliated Nanjing Brain Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Chun-Fa Qian
- Department of Neurosurgery, Nanjing Medical University, Affiliated Nanjing Brain Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Hong Xiao
- Neuropsychiatric Institute, Nanjing Medical University, Affiliated Nanjing Brain Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Hong-Yi Liu
- Department of Neurosurgery, Nanjing Medical University, Affiliated Nanjing Brain Hospital, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
23
|
Chai X, Zhao S, Fan L, Zhang W, Lu X, Shao H, Wang S, Song L, Failla AV, Zobiak B, Mannherz HG, Frotscher M. Reelin and cofilin cooperate during the migration of cortical neurons: a quantitative morphological analysis. Development 2016; 143:1029-40. [PMID: 26893343 DOI: 10.1242/dev.134163] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/29/2016] [Indexed: 12/19/2022]
Abstract
In reeler mutant mice, which are deficient in reelin (Reln), the lamination of the cerebral cortex is disrupted. Reelin signaling induces phosphorylation of LIM kinase 1, which phosphorylates the actin-depolymerizing protein cofilin in migrating neurons. Conditional cofilin mutants show neuronal migration defects. Thus, both reelin and cofilin are indispensable during cortical development. To analyze the effects of cofilin phosphorylation on neuronal migration we used in utero electroporation to transfect E14.5 wild-type cortical neurons with pCAG-EGFP plasmids encoding either a nonphosphorylatable form of cofilin 1 (cofilin(S3A)), a pseudophosphorylated form (cofilin(S3E)) or wild-type cofilin 1 (cofilin(WT)). Wild-type controls and reeler neurons were transfected with pCAG-EGFP. Real-time microscopy and histological analyses revealed that overexpression of cofilin(WT) and both phosphomutants induced migration defects and morphological abnormalities of cortical neurons. Of note, reeler neurons and cofilin(S3A)- and cofilin(S3E)-transfected neurons showed aberrant backward migration towards the ventricular zone. Overexpression of cofilin(S3E), the pseudophosphorylated form, partially rescued the migration defect of reeler neurons, as did overexpression of Limk1. Collectively, the results indicate that reelin and cofilin cooperate in controlling cytoskeletal dynamics during neuronal migration.
Collapse
Affiliation(s)
- Xuejun Chai
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Shanting Zhao
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany College of Veterinary Medicine, Northwest A&F University, 712100 Yangling, People's Republic of China
| | - Li Fan
- Institute of Zoology, School of Life Science, Lanzhou University, 730000 Lanzhou, People's Republic of China
| | - Wei Zhang
- College of Veterinary Medicine, Northwest A&F University, 712100 Yangling, People's Republic of China
| | - Xi Lu
- College of Veterinary Medicine, Northwest A&F University, 712100 Yangling, People's Republic of China
| | - Hong Shao
- Institute of Zoology, School of Life Science, Lanzhou University, 730000 Lanzhou, People's Republic of China
| | - Shaobo Wang
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Lingzhen Song
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Antonio Virgilio Failla
- UKE Microscopy Imaging Facility (UMIF), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Bernd Zobiak
- UKE Microscopy Imaging Facility (UMIF), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Hans G Mannherz
- Institute of Anatomy and Molecular Embryology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Michael Frotscher
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
24
|
Zhao P, Xu Y, Wei Y, Qiu Q, Chew TL, Kang Y, Cheng C. The CD44s splice isoform is a central mediator for invadopodia activity. J Cell Sci 2016; 129:1355-65. [PMID: 26869223 DOI: 10.1242/jcs.171959] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 02/04/2016] [Indexed: 01/04/2023] Open
Abstract
The ability for tumor cells to spread and metastasize to distant organs requires proteolytic degradation of extracellular matrix (ECM). This activity is mediated by invadopodia, actin-rich membrane protrusions that are enriched for proteases. However, the mechanisms underlying invadopodia activity are not fully understood. Here, we report that a specific CD44 splice isoform, CD44s, is an integral component in invadopodia. We show that CD44s, but not another splice isoform CD44v, is localized in invadopodia. Small hairpin (sh)RNA-mediated depletion of CD44s abolishes invadopodia activity, prevents matrix degradation and decreases tumor cell invasiveness. Our results suggest that CD44s promotes cortactin phosphorylation and recruits MT1-MMP (also known as MMP14) to sites of matrix degradation, which are important activities for invadopodia function. Importantly, we show that depletion of CD44s inhibits breast cancer cell metastasis to the lung in animals. These findings suggest a crucial mechanism underlying the role of the CD44s splice isoform in breast cancer metastasis.
Collapse
Affiliation(s)
- Pu Zhao
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yilin Xu
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yong Wei
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Qiong Qiu
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Teng-Leong Chew
- Cell Imaging Facility & Nikon Imaging Center, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Chonghui Cheng
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
25
|
Rotty JD, Bear JE. Competition and collaboration between different actin assembly pathways allows for homeostatic control of the actin cytoskeleton. BIOARCHITECTURE 2015; 5:27-34. [PMID: 26430713 DOI: 10.1080/19490992.2015.1090670] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Tremendous insight into actin-associated proteins has come from careful biochemical and cell biological characterization of their activities and regulation. However, many studies of their cellular behavior have only considered each in isolation. Recent efforts reveal that assembly factors compete for polymerization-competent actin monomers, suggesting that actin is homeostatically regulated. It seems that a major regulatory component is competition between Arp2/3-activating nucleation promoting factors and profilin for actin monomers. The result is differential delivery of actin to different pathways, allowing for simultaneous assembly of competing F-actin structures and collaborative building of higher order cellular structures. Although there are likely to be additional factors that regulate actin homeostasis, especially in a cell type-dependent fashion, we advance the notion that competition between actin assembly factors results in a tunable system that can be adjusted according to extracellular and intracellular cues.
Collapse
Affiliation(s)
- Jeremy D Rotty
- a UNC Lineberger Comprehensive Cancer Center; University of North Carolina at Chapel Hill ; Chapel Hill , NC USA.,b Department of Cell Biology and Physiology; University of North Carolina at Chapel Hill ; Chapel Hill , NC USA
| | - James E Bear
- a UNC Lineberger Comprehensive Cancer Center; University of North Carolina at Chapel Hill ; Chapel Hill , NC USA.,b Department of Cell Biology and Physiology; University of North Carolina at Chapel Hill ; Chapel Hill , NC USA
| |
Collapse
|
26
|
Li L, Zhang W, Chai X, Zhang Q, Xie J, Chen S, Zhao S. Neuronal maturation and laminar formation in the chicken optic tectum are accompanied by the transition of phosphorylated cofilin from cytoplasm to nucleus. Gene Expr Patterns 2014; 16:75-85. [PMID: 25290739 DOI: 10.1016/j.gep.2014.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 09/23/2014] [Accepted: 09/25/2014] [Indexed: 11/17/2022]
Abstract
Laminar formation in the chicken optic tectum requires processes that coordinate proliferation, migration and differentiation of neurons, in which the dynamics of actin filaments are crucial. Cofilin plays pivotal roles in regulating actin arrangement via its phosphorylation on Ser3. Given poor studies on the profile of phosphorylated cofilin (p-cofilin) in the developing tectum, we investigated its expression pattern. As determined by immunofluorescence histochemistry and western blotting, p-cofilin could be detected in most tectal layers except for the neural epithelium. In addition, we found p-cofilin was expressed both in the cytoplasm and the nucleus. During development, the expression of the cytoplasmic p-cofilin was decreasing and the nuclear p-cofilin was gradually increasing, but the total level of p-cofilin was down regulated. Double-labeling experiments revealed that the nuclear p-cofilin could be labeled in mature neurons but undetected in immature neurons. Furthermore, the number of cells co-stained with nuclear p-cofilin and NeuN was up-regulated during lamination and 60% cells were detected to be mature neurons that can express nuclear p-cofilin just at the first appearance of completed laminae. Our results demonstrate that the maturation of neurons is accompanied by this cytoplasm-to-nucleus transition of p-cofilin, and the nuclear p-cofilin can work effectively as a marker in the laminar formation of the chicken optic tectum.
Collapse
Affiliation(s)
- Lingling Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Wei Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Xuejun Chai
- Institute for Structural Neurobiology, Center for Molecular Neurobiology Hamburg (ZMNH), Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Qi Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Jiongfang Xie
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Shulin Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100 Shaanxi, China.
| |
Collapse
|
27
|
Beaty BT, Condeelis J. Digging a little deeper: the stages of invadopodium formation and maturation. Eur J Cell Biol 2014; 93:438-44. [PMID: 25113547 DOI: 10.1016/j.ejcb.2014.07.003] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 07/10/2014] [Accepted: 07/10/2014] [Indexed: 01/09/2023] Open
Abstract
Invadopodia are actin-rich protrusions that degrade the extracellular matrix and are required for penetration through the basement membrane, stromal invasion and intravasation. Invadopodia are enriched in actin regulators, such as cortactin, cofilin, N-WASp, Arp2/3 and fascin. Much of the work to date has centered around identifying the proteins involved in regulating actin polymerization and matrix degradation. Recently, there have been significant advances in characterization of the very early stages of invadopodium precursor assembly and the role of adhesion proteins, such as β1 integrin, talin, FAK and Hic-5, in promoting invadopodium maturation. This review summarizes these findings in the context of our current model of invadopodial function and highlights some of the important unanswered questions in the field.
Collapse
Affiliation(s)
- Brian T Beaty
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, United States.
| | - John Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, United States; Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, United States.
| |
Collapse
|
28
|
Tania N, Condeelis J, Edelstein-Keshet L. Modeling the synergy of cofilin and Arp2/3 in lamellipodial protrusive activity. Biophys J 2014; 105:1946-55. [PMID: 24209839 DOI: 10.1016/j.bpj.2013.09.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 09/10/2013] [Accepted: 09/16/2013] [Indexed: 12/13/2022] Open
Abstract
Rapid polymerization of actin filament barbed ends generates protrusive forces at the cell edge, leading to cell migration. Two important regulators of free barbed ends, cofilin and Arp2/3, have been shown to work in synergy (net effect greater than additive). To explore this synergy, we model the dynamics of F-actin at the leading edge, motivated by data from EGF-stimulated mammary carcinoma cells. We study how synergy depends on the localized rates and relative timing of cofilin and Arp2/3 activation at the cell edge. The model incorporates diffusion of cofilin, membrane protrusion, F-actin capping, aging, and severing by cofilin and branch nucleation by Arp2/3 (but not G-actin recycling). In a well-mixed system, cofilin and Arp2/3 can each generate a large pulse of barbed ends on their own, but have little synergy; high synergy occurs only at low activation rates, when few barbed ends are produced. In the full spatially distributed model, both synergy and barbed-end production are significant over a range of activation rates. Furthermore, barbed-end production is greatest when Arp2/3 activation is delayed relative to cofilin. Our model supports a direct role for cofilin-mediated actin polymerization in stimulated cell migration, including chemotaxis and cancer invasion.
Collapse
Affiliation(s)
- Nessy Tania
- Department of Mathematics, University of British Columbia, Vancouver, Canada; Department of Mathematics and Statistics, Smith College, Northampton, Massachusetts
| | | | | |
Collapse
|
29
|
Beaty BT, Wang Y, Bravo-Cordero JJ, Sharma VP, Miskolci V, Hodgson L, Condeelis J. Talin regulates moesin-NHE-1 recruitment to invadopodia and promotes mammary tumor metastasis. J Cell Biol 2014; 205:737-51. [PMID: 24891603 PMCID: PMC4050723 DOI: 10.1083/jcb.201312046] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 04/28/2014] [Indexed: 02/08/2023] Open
Abstract
Invadopodia are actin-rich protrusions that degrade the extracellular matrix and are required for stromal invasion, intravasation, and metastasis. The role of the focal adhesion protein talin in regulating these structures is not known. Here, we demonstrate that talin is required for invadopodial matrix degradation and three-dimensional extracellular matrix invasion in metastatic breast cancer cells. The sodium/hydrogen exchanger 1 (NHE-1) is linked to the cytoskeleton by ezrin/radixin/moesin family proteins and is known to regulate invadopodium-mediated matrix degradation. We show that the talin C terminus binds directly to the moesin band 4.1 ERM (FERM) domain to recruit a moesin-NHE-1 complex to invadopodia. Silencing talin resulted in a decrease in cytosolic pH at invadopodia and blocked cofilin-dependent actin polymerization, leading to impaired invadopodium stability and matrix degradation. Furthermore, talin is required for mammary tumor cell motility, intravasation, and spontaneous lung metastasis in vivo. Thus, our findings provide a novel understanding of how intracellular pH is regulated and a molecular mechanism by which talin enhances tumor cell invasion and metastasis.
Collapse
Affiliation(s)
- Brian T Beaty
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Yarong Wang
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Jose Javier Bravo-Cordero
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Ved P Sharma
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Veronika Miskolci
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Louis Hodgson
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - John Condeelis
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| |
Collapse
|
30
|
Al Haj A, Mazur AJ, Buchmeier S, App C, Theiss C, Silvan U, Schoenenberger CA, Jockusch BM, Hannappel E, Weeds AG, Mannherz HG. Thymosin beta4 inhibits ADF/cofilin stimulated F-actin cycling and hela cell migration: Reversal by active Arp2/3 complex. Cytoskeleton (Hoboken) 2013; 71:95-107. [DOI: 10.1002/cm.21128] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 04/17/2013] [Accepted: 12/10/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Abdulatif Al Haj
- Department of Anatomy and Molecular Embryology; Ruhr-University; Bochum Germany
| | | | - Sabine Buchmeier
- Cell Biology Group; Institute of Zoology; Technical University of Braunschweig; Germany
| | - Christine App
- Institute of Biochemistry; University of Erlangen; Erlangen Germany
| | | | - Unai Silvan
- Maurice E. Müller Institute for Structural Biology, Biocenter; Basel Switzerland
| | | | - Brigitte M. Jockusch
- Cell Biology Group; Institute of Zoology; Technical University of Braunschweig; Germany
| | - Ewald Hannappel
- Institute of Biochemistry; University of Erlangen; Erlangen Germany
| | - Alan G. Weeds
- MRC Laboratory of Molecular Biology and Trinity College; Cambridge United Kingdom
| | - Hans Georg Mannherz
- Department of Anatomy and Molecular Embryology; Ruhr-University; Bochum Germany
- Department of Physical Biochemistry; Max-Planck-Institute of Molecular Physiology; Dortmund Germany
| |
Collapse
|
31
|
Vitriol EA, Wise AL, Berginski ME, Bamburg JR, Zheng JQ. Instantaneous inactivation of cofilin reveals its function of F-actin disassembly in lamellipodia. Mol Biol Cell 2013; 24:2238-47. [PMID: 23676663 PMCID: PMC3708729 DOI: 10.1091/mbc.e13-03-0156] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/01/2013] [Accepted: 05/08/2013] [Indexed: 12/23/2022] Open
Abstract
Cofilin is a key regulator of the actin cytoskeleton. It can sever actin filaments, accelerate filament disassembly, act as a nucleation factor, recruit or antagonize other actin regulators, and control the pool of polymerization-competent actin monomers. In cells these actions have complex functional outputs. The timing and localization of cofilin activity are carefully regulated, and thus global, long-term perturbations may not be sufficient to probe its precise function. To better understand cofilin's spatiotemporal action in cells, we implemented chromophore-assisted laser inactivation (CALI) to instantly and specifically inactivate it. In addition to globally inhibiting actin turnover, CALI of cofilin generated several profound effects on the lamellipodia, including an increase of F-actin, a rearward expansion of the actin network, and a reduction in retrograde flow speed. These results support the hypothesis that the principal role of cofilin in lamellipodia at steady state is to break down F-actin, control filament turnover, and regulate the rate of retrograde flow.
Collapse
Affiliation(s)
- Eric A. Vitriol
- Departments of Cell Biology and Neurology, Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
| | - Ariel L. Wise
- Departments of Cell Biology and Neurology, Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
| | - Mathew E. Berginski
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - James R. Bamburg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523
| | - James Q. Zheng
- Departments of Cell Biology and Neurology, Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
32
|
Bravo-Cordero JJ, Magalhaes MAO, Eddy RJ, Hodgson L, Condeelis J. Functions of cofilin in cell locomotion and invasion. Nat Rev Mol Cell Biol 2013; 14:405-15. [PMID: 23778968 DOI: 10.1038/nrm3609] [Citation(s) in RCA: 380] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recently, a consensus has emerged that cofilin severing activity can generate free actin filament ends that are accessible for F-actin polymerization and depolymerization without changing the rate of G-actin association and dissociation at either filament end. The structural basis of actin filament severing by cofilin is now better understood. These results have been integrated with recently discovered mechanisms for cofilin activation in migrating cells, which led to new models for cofilin function that provide insights into how cofilin regulation determines the temporal and spatial control of cell behaviour.
Collapse
Affiliation(s)
- Jose Javier Bravo-Cordero
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, New York 10461, USA.
| | | | | | | | | |
Collapse
|
33
|
Bravo-Cordero JJ, Sharma VP, Roh-Johnson M, Chen X, Eddy R, Condeelis J, Hodgson L. Spatial regulation of RhoC activity defines protrusion formation in migrating cells. J Cell Sci 2013; 126:3356-69. [PMID: 23704350 DOI: 10.1242/jcs.123547] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Protrusion formation is the first step that precedes cell movement of motile cells. Spatial control of actin polymerization is necessary to achieve directional protrusion during cell migration. Here we show that the spatial coordinators p190RhoGEF and p190RhoGAP regulate actin polymerization during leading edge protrusions by regulating the actin barbed end distribution and amplitude. The distribution of RhoC activity and proper balance of cofilin activation achieved by p190RhoGEF and p190RhoGAP determines the direction of final protrusive activity. These findings provide a new insight into the dynamic plasticity in the amplitude and distribution of barbed ends, which can be modulated by fine-tuning RhoC activity by upstream GEFs and GAPs for directed cell motility.
Collapse
Affiliation(s)
- Jose Javier Bravo-Cordero
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Hirahara Y, Matsuda KI, Liu YF, Yamada H, Kawata M, Boggs JM. 17β-Estradiol and 17α-estradiol induce rapid changes in cytoskeletal organization in cultured oligodendrocytes. Neuroscience 2013; 235:187-99. [PMID: 23337538 DOI: 10.1016/j.neuroscience.2012.12.070] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 11/22/2012] [Accepted: 12/15/2012] [Indexed: 12/12/2022]
Abstract
Dramatic changes in the cytoskeleton and the morphology of oligodendrocytes (OLs) occur during various stages of the myelination process. OLs in culture produce large membrane sheets containing cytoskeletal veins of microtubules and actin filaments. We recently showed that estrogen receptors (ER) related to ERα/β were expressed in the membrane sheets of mature OLs in culture. Ligation of these or other membrane ERs in OLs with both 17β- and 17α-estradiol mediated rapid non-genomic signaling. Here, we show that estrogens also mediate rapid non-genomic remodeling of the cytoskeleton in mature OLs in culture. 17β-Estradiol caused a rapid loss of microtubules and the actin cytoskeleton in the OL membrane sheets. It also increased phosphorylation of the actin filament-severing protein cofilin, thus inactivating it. Staining for actin barbed ends with rhodamine-actin showed that it decreased the amount of actin barbed ends. 17α-Estradiol, on the other hand, increased the percentage of cells with abundant staining of actin filaments and actin barbed ends, suggesting that it stabilized and/or increased the dynamics of the actin cytoskeleton. The specific ERα and ERβ agonists, 4,4',4″-(4-propyl-(1H)-pyrazole-1,3,5-triyl) trisphenol (PPT) and diarylpropionitrile 2,3-bis(4-hydroxy-phenyl)-propionitrile (DPN), respectively, also caused the rapid phosphorylation of cofilin. Estrogen-induced phosphorylation of cofilin was inhibited by Y-27632, a specific inhibitor of the Rho-associated protein serine/threonine kinase (ROCK). The Rho/ROCK/cofilin pathway is therefore implicated in actin rearrangement via estrogen ligation of membrane ERs, which may include forms of ERα and ERβ. These results indicate a role for estrogens in modulation of the cytoskeleton in mature OLs, and thus in various processes required for myelinogenesis.
Collapse
Affiliation(s)
- Y Hirahara
- Department of Anatomy and Cell Science, Kansai Medical University, Moriguchi-City, 570-8506 Osaka, Japan
| | | | | | | | | | | |
Collapse
|
35
|
Ueda H, Matsunaga H, Olaposi OI, Nagai J. Lysophosphatidic acid: Chemical signature of neuropathic pain. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:61-73. [DOI: 10.1016/j.bbalip.2012.08.014] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 08/21/2012] [Accepted: 08/21/2012] [Indexed: 02/07/2023]
|
36
|
Pharmacological Inhibition of Actin Assembly to Target Tumor Cell Motility. Rev Physiol Biochem Pharmacol 2013; 166:23-42. [DOI: 10.1007/112_2013_16] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
37
|
Actin cytoskeleton manipulation by effector proteins secreted by diarrheagenic Escherichia coli pathotypes. BIOMED RESEARCH INTERNATIONAL 2012; 2013:374395. [PMID: 23509714 PMCID: PMC3591105 DOI: 10.1155/2013/374395] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 10/22/2012] [Indexed: 11/18/2022]
Abstract
The actin cytoskeleton is a dynamic structure necessary for cell and tissue organization, including the maintenance of epithelial barriers. Disruption of the epithelial barrier coincides with alterations of the actin cytoskeleton in several disease states. These disruptions primarily affect the paracellular space, which is normally regulated by tight junctions. Thereby, the actin cytoskeleton is a common and recurring target of bacterial virulence factors. In order to manipulate the actin cytoskeleton, bacteria secrete and inject toxins and effectors to hijack the host cell machinery, which interferes with host-cell pathways and with a number of actin binding proteins. An interesting model to study actin manipulation by bacterial effectors is Escherichia coli since due to its genome plasticity it has acquired diverse genetic mobile elements, which allow having different E. coli varieties in one bacterial species. These E. coli pathotypes, including intracellular and extracellular bacteria, interact with epithelial cells, and their interactions depend on a specific combination of virulence factors. In this paper we focus on E. coli effectors that mimic host cell proteins to manipulate the actin cytoskeleton. The study of bacterial effector-cytoskeleton interaction will contribute not only to the comprehension of the molecular causes of infectious diseases but also to increase our knowledge of cell biology.
Collapse
|
38
|
Liu WM, Zhang F, Moshiach S, Zhou B, Huang C, Srinivasan K, Khurana S, Zheng Y, Lahti JM, Zhang XA. Tetraspanin CD82 inhibits protrusion and retraction in cell movement by attenuating the plasma membrane-dependent actin organization. PLoS One 2012; 7:e51797. [PMID: 23251627 PMCID: PMC3522597 DOI: 10.1371/journal.pone.0051797] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 11/12/2012] [Indexed: 11/18/2022] Open
Abstract
To determine how tetraspanin KAI1/CD82, a tumor metastasis suppressor, inhibits cell migration, we assessed which cellular events critical for motility are altered by KAI1/CD82 and how KAI1/CD82 regulates these events. We found that KAI1/CD82-expressing cells typically exhibited elongated cellular tails and diminished lamellipodia. Live imaging demonstrated that the polarized protrusion and retraction of the plasma membrane became deficient upon KAI1/CD82 expression. The deficiency in developing these motility-related cellular events was caused by poor formations of actin cortical network and stress fiber and by aberrant dynamics in actin organization. Rac1 activity was reduced by KAI1/CD82, consistent with the diminution of lamellipodia and actin cortical network; while the growth factor-stimulated RhoA activity was blocked by KAI1/CD82, consistent with the loss of stress fiber and attenuation in cellular retraction. Upon KAI1/CD82 expression, Rac effector cofilin was not enriched at the cell periphery to facilitate lamellipodia formation while Rho kinase exhibited a significantly lower activity leading to less retraction. Phosphatidylinositol 4, 5-biphosphate, which initiates actin polymerization from the plasma membrane, became less detectable at the cell periphery in KAI1/CD82-expressing cells. Moreover, KAI1/CD82-induced phenotypes likely resulted from the suppression of multiple signaling pathways such as integrin and growth factor signaling. In summary, at the cellular level KAI1/CD82 inhibited polarized protrusion and retraction events by disrupting actin reorganization; at the molecular level, KAI1/CD82 deregulated Rac1, RhoA, and their effectors cofilin and Rho kinase by perturbing the plasma membrane lipids.
Collapse
Affiliation(s)
- Wei M. Liu
- Vascular Biology and Cancer Centers and Departments of Medicine and Molecular Science, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Feng Zhang
- Vascular Biology and Cancer Centers and Departments of Medicine and Molecular Science, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Simon Moshiach
- Department of Genetics and Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Bin Zhou
- Vascular Biology and Cancer Centers and Departments of Medicine and Molecular Science, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Chao Huang
- Vascular Biology and Cancer Centers and Departments of Medicine and Molecular Science, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Kamalakkannan Srinivasan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Seema Khurana
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Yi Zheng
- Division of Experimental Hematology, Cincinnati Children's Hospital, Cincinnati, Ohio, United States of America
| | - Jill M. Lahti
- Department of Genetics and Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Xin A. Zhang
- Vascular Biology and Cancer Centers and Departments of Medicine and Molecular Science, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
39
|
Taulet N, Delorme-Walker VD, DerMardirossian C. Reactive oxygen species regulate protrusion efficiency by controlling actin dynamics. PLoS One 2012; 7:e41342. [PMID: 22876286 PMCID: PMC3410878 DOI: 10.1371/journal.pone.0041342] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 06/20/2012] [Indexed: 12/31/2022] Open
Abstract
Productive protrusions allowing motile cells to sense and migrate toward a chemotactic gradient of reactive oxygen species (ROS) require a tight control of the actin cytoskeleton. However, the mechanisms of how ROS affect cell protrusion and actin dynamics are not well elucidated yet. We show here that ROS induce the formation of a persistent protrusion. In migrating epithelial cells, protrusion of the leading edge requires the precise regulation of the lamellipodium and lamella F-actin networks. Using fluorescent speckle microscopy, we showed that, upon ROS stimulation, the F-actin retrograde flow is enhanced in the lamellipodium. This event coincides with an increase of cofilin activity, free barbed ends formation, Arp2/3 recruitment, and ERK activity at the cell edge. In addition, we observed an acceleration of the F-actin flow in the lamella of ROS-stimulated cells, which correlates with an enhancement of the cell contractility. Thus, this study demonstrates that ROS modulate both the lamellipodium and the lamella networks to control protrusion efficiency.
Collapse
Affiliation(s)
- Nicolas Taulet
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Violaine D. Delorme-Walker
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Céline DerMardirossian
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
40
|
Jang I, Jeon BT, Jeong EA, Kim EJ, Kang D, Lee JS, Jeong BG, Kim JH, Choi BH, Lee JE, Kim JW, Choi JY, Roh GS. Pak1/LIMK1/Cofilin Pathway Contributes to Tumor Migration and Invasion in Human Non-Small Cell Lung Carcinomas and Cell Lines. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2012; 16:159-65. [PMID: 22802696 PMCID: PMC3394917 DOI: 10.4196/kjpp.2012.16.3.159] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 04/20/2012] [Accepted: 05/12/2012] [Indexed: 01/16/2023]
Abstract
Squamous cell carcinoma (SCC) and adenocarcinoma (AC) are the major histological types of non-small cell lung carcinoma (NSCLC). Although both SCCs and ACs have been characterized histologically and clinically, the precise mechanisms underlying their migration and invasion are not yet known. Here, we address the involvement in NSCLC of the p21-associated kinase1 (Pak1)/LIM kinase1 (LIMK1)/cofilin pathway, which recently has been reported to play a critical role in tumor migration and invasion. The Pak1/LIMK1/cofilin pathway was evaluated in tumors from SCC (n=35) and AC (n=35) patients and in SCC- and AC-type cell lines by western blotting, immunohistochemistry, and in vitro migration and invasion assays. The levels of phosphorylated Pak1, LIMK1, and cofilin in lung tumor tissues from SCC patients were increased as compared to normal tissues. In addition, immunohistochemistry showed greater expression of phosphorylated cofilin in SCC tissues. Expression of phosphorylated Pak1 and LIMK1 proteins was also significantly higher in SCC-type cells than in AC-type cells. Moreover, migration and invasion assays revealed that a higher percentage of SCC type cells exhibited migration and invasion compared to AC type cells. Migration was also decreased in LIMK1 knockdown SK-MES-1 cells. These findings suggest that the activation of the Pak1/LIMK1/cofilin pathway could preferentially contribute to greater tumor migration and invasion in SCC, relative to that in AC.
Collapse
Affiliation(s)
- Inseok Jang
- Department of Thoracic and Cardiovascular Surgery, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-290, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
This review focuses on basic principles of motility in different cell types, formation of the specific cell structures that enable directed migration, and how external signals are transduced into cells and coupled to the motile machinery. Feedback mechanisms and their potential role in maintenance of internal chemotactic gradients and persistence of directed migration are highlighted.
Collapse
Affiliation(s)
- A V Vorotnikov
- Department of Biochemistry and Molecular Medicine, Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
42
|
Lou D, Sun B, Wei H, Deng X, Chen H, Xu D, Li G, Xu H, Wang Y. Spatiotemporal Expression of Testicular Protein Kinase 1 After Rat Sciatic Nerve Injury. J Mol Neurosci 2012; 47:180-91. [DOI: 10.1007/s12031-012-9712-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 01/20/2012] [Indexed: 11/28/2022]
|
43
|
Magalhaes MAO, Larson DR, Mader CC, Bravo-Cordero JJ, Gil-Henn H, Oser M, Chen X, Koleske AJ, Condeelis J. Cortactin phosphorylation regulates cell invasion through a pH-dependent pathway. ACTA ACUST UNITED AC 2011; 195:903-20. [PMID: 22105349 PMCID: PMC3257566 DOI: 10.1083/jcb.201103045] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Invadopodia are invasive protrusions with proteolytic activity uniquely found in tumor cells. Cortactin phosphorylation is a key step during invadopodia maturation, regulating Nck1 binding and cofilin activity. The precise mechanism of cortactin-dependent cofilin regulation and the roles of this pathway in invadopodia maturation and cell invasion are not fully understood. We provide evidence that cortactin-cofilin binding is regulated by local pH changes at invadopodia that are mediated by the sodium-hydrogen exchanger NHE1. Furthermore, cortactin tyrosine phosphorylation mediates the recruitment of NHE1 to the invadopodium compartment, where it locally increases the pH to cause the release of cofilin from cortactin. We show that this mechanism involving cortactin phosphorylation, local pH increase, and cofilin activation regulates the dynamic cycles of invadopodium protrusion and retraction and is essential for cell invasion in 3D. Together, these findings identify a novel pH-dependent regulation of cell invasion.
Collapse
Affiliation(s)
- Marco A O Magalhaes
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Tania N, Prosk E, Condeelis J, Edelstein-Keshet L. A temporal model of cofilin regulation and the early peak of actin barbed ends in invasive tumor cells. Biophys J 2011; 100:1883-92. [PMID: 21504724 DOI: 10.1016/j.bpj.2011.02.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 01/27/2011] [Accepted: 02/22/2011] [Indexed: 10/18/2022] Open
Abstract
Cofilin is an important regulator of actin polymerization, cell migration, and chemotaxis. Recent experimental data on mammary carcinoma cells reveal that stimulation by epidermal growth factor (EGF) generates a pool of active cofilin that results in a peak of actin filament barbed ends on the timescale of 1 min. Here, we present results of a mathematical model for the dynamics of cofilin and its transition between several pools in response to EGF stimulation. We describe the interactions of phospholipase C, membrane lipids (PIP(2)), and cofilin bound to PIP(2) and to F-actin, as well as diffusible cofilin in active G-actin-monomer-bound or phosphorylated states. We consider a simplified representation in which the thin cell edge (lamellipod) and the cell interior are represented by two compartments that are linked by diffusion. We demonstrate that a high basal level of active cofilin stored by binding to PIP(2), as well as the highly enriched local milieu of F-actin at the cell edge, is essential to capture the EGF-induced barbed-end amplification observed experimentally.
Collapse
Affiliation(s)
- Nessy Tania
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | |
Collapse
|
45
|
Abstract
Viral infection converts the normal functions of a cell to optimize viral replication and virion production. One striking observation of this conversion is the reconfiguration and reorganization of cellular actin, affecting every stage of the viral life cycle, from entry through assembly to egress. The extent and degree of cytoskeletal reorganization varies among different viral infections, suggesting the evolution of myriad viral strategies. In this Review, we describe how the interaction of viral proteins with the cell modulates the structure and function of the actin cytoskeleton to initiate, sustain and spread infections. The molecular biology of such interactions continues to engage virologists in their quest to understand viral replication and informs cell biologists about the role of the cytoskeleton in the uninfected cell.
Collapse
Affiliation(s)
- Matthew P Taylor
- Department of Molecular Biology, Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544, USA
| | | | | |
Collapse
|
46
|
Oser M, Mader CC, Gil-Henn H, Magalhaes M, Bravo-Cordero JJ, Koleske AJ, Condeelis J. Specific tyrosine phosphorylation sites on cortactin regulate Nck1-dependent actin polymerization in invadopodia. J Cell Sci 2011; 123:3662-73. [PMID: 20971703 DOI: 10.1242/jcs.068163] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Invadopodia are matrix-degrading membrane protrusions in invasive carcinoma cells enriched in proteins that regulate actin polymerization. The on-off regulatory switch that initiates actin polymerization in invadopodia requires phosphorylation of tyrosine residues 421, 466, and 482 on cortactin. However, it is unknown which of these cortactin tyrosine phosphorylation sites control actin polymerization. We investigated the contribution of individual tyrosine phosphorylation sites (421, 466, and 482) on cortactin to the regulation of actin polymerization in invadopodia. We provide evidence that the phosphorylation of tyrosines 421 and 466, but not 482, is required for the generation of free actin barbed ends in invadopodia. In addition, these same phosphotyrosines are important for Nck1 recruitment to invadopodia via its SH2 domain, for the direct binding of Nck1 to cortactin in vitro, and for the FRET interaction between Nck1 and cortactin in invadopodia. Furthermore, matrix proteolysis-dependent tumor cell invasion is dramatically inhibited in cells expressing a mutation in phosphotyrosine 421 or 466. Together, these results identify phosphorylation of tyrosines 421 and 466 on cortactin as the crucial residues that regulate Nck1-dependent actin polymerization in invadopodia and tumor cell invasion, and suggest that specifically blocking either tyrosine 421 or 466 phosphorylation might be effective at inhibiting tumor cell invasion in vivo.
Collapse
Affiliation(s)
- Matthew Oser
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Kim JH, Cho A, Yin H, Schafer DA, Mouneimne G, Simpson KJ, Nguyen KV, Brugge JS, Montell DJ. Psidin, a conserved protein that regulates protrusion dynamics and cell migration. Genes Dev 2011; 25:730-41. [PMID: 21406550 DOI: 10.1101/gad.2028611] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Dynamic assembly and disassembly of actin filaments is a major driving force for cell movements. Border cells in the Drosophila ovary provide a simple and genetically tractable model to study the mechanisms regulating cell migration. To identify new genes that regulate cell movement in vivo, we screened lethal mutations on chromosome 3R for defects in border cell migration and identified two alleles of the gene psidin (psid). In vitro, purified Psid protein bound F-actin and inhibited the interaction of tropomyosin with F-actin. In vivo, psid mutations exhibited genetic interactions with the genes encoding tropomyosin and cofilin. Border cells overexpressing Psid together with GFP-actin exhibited altered protrusion/retraction dynamics. Psid knockdown in cultured S2 cells reduced, and Psid overexpression enhanced, lamellipodial dynamics. Knockdown of the human homolog of Psid reduced the speed and directionality of migration in wounded MCF10A breast epithelial monolayers, whereas overexpression of the protein increased migration speed and altered protrusion dynamics in EGF-stimulated cells. These results indicate that Psid is an actin regulatory protein that plays a conserved role in protrusion dynamics and cell migration.
Collapse
Affiliation(s)
- Ji Hoon Kim
- Department of Biological Chemistry, Center for Cell Dynamics, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Zhang L, Luo J, Wan P, Wu J, Laski F, Chen J. Regulation of cofilin phosphorylation and asymmetry in collective cell migration during morphogenesis. Development 2011; 138:455-64. [PMID: 21205790 DOI: 10.1242/dev.046870] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
During Drosophila oogenesis, two actin dynamics regulators, cofilin and Rac, are required for the collective migration of a coherent cluster of cells called border cells. Cell culture data have shown that Rac and cofilin are both essential for lamellipodium formation, but Rac signaling results in phosphorylation and hence inactivation of cofilin. So it remains unclear whether cofilin phosphorylation plays a promoting or inhibitory role during cell migration. We show here that cofilin is required for F-actin turnover and lamellipodial protrusion in the border cells. Interestingly, reducing the dosage of cofilin by half or expressing a phospho-mimetic mutant form, S3E, partially rescues the migration and protrusion defects of Rac-deficient border cells. Moreover, cofilin exhibits moderate accumulation in border cells at the migratory front of the cluster, whereas phospho-cofilin has a robust and uniform distribution pattern in all the outer border cells. Blocking or overactivating Rac signaling in border cells greatly reduces or increases cofilin phosphorylation, respectively, and each abolishes cell migration. Furthermore, Rac may signal through Pak and LIMK to result in uniform phosphorylation of cofilin in all the outer border cells, whereas the guidance receptor Pvr (PDGF/VEGF receptor) mediates the asymmetric localization of cofilin in the cluster but does not affect its phosphorylation. Our study provides one of the first models of how cofilin functions and is regulated in the collective migration of a group of cells in vivo.
Collapse
Affiliation(s)
- Lijun Zhang
- Model Animal Research Center, and MOE Key Laboratory of Model Animals for Disease Study, Nanjing University, Nanjing, China
| | | | | | | | | | | |
Collapse
|
49
|
Martin-Martin B, Tovell V, Dahlmann-Noor AH, Khaw PT, Bailly M. The effect of MMP inhibitor GM6001 on early fibroblast-mediated collagen matrix contraction is correlated to a decrease in cell protrusive activity. Eur J Cell Biol 2011; 90:26-36. [PMID: 21040999 PMCID: PMC7611814 DOI: 10.1016/j.ejcb.2010.09.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Revised: 09/14/2010] [Accepted: 09/20/2010] [Indexed: 12/20/2022] Open
Abstract
Although fibroblasts play an essential part during the wound healing response, the mechanisms by which they mediate tissue remodelling and contraction are still unclear. Using live cell and matrix imaging within 3D free-floating fibroblast-populated collagen lattices as a model for tissue contraction, we compared the behaviour of a range of fibroblasts with low and high contraction abilities and analysed the effect of the broad spectrum MMP-inhibitor GM6001 on cell behaviour and matrix contraction. We identified two mechanisms underlying matrix contraction, one via direct cell-mediated contractile activity, the second through matrix degradation. These appear to be linked to cell morphology and regulated by the collagen concentration within the matrix. Cells with a rounded morphology proliferated in the matrix but did not remodel it efficiently, resulting in a poor ability to contract matrices. Cells with an elongated morphology showed higher levels of protrusive activity, leading to efficient matrix remodelling and contraction. GM6001 inhibited week-long matrix contraction to various extents with the different cell lines. However, quantitative analysis of the cell protrusive activity showed that GM6001 consistently decreased cell dynamics in 3D by about 20%, and this was correlated with a significant reduction in early matrix contraction. Overall our results suggest that although fibroblast-mediated matrix contraction depends on both cell dynamics and MMP-mediated matrix degradation, the efficiency of GM6001 treatment in preventing contraction might be linked to a direct effect on cell dynamics.
Collapse
Affiliation(s)
- Belen Martin-Martin
- Department of Cell Biology, Therapeutics and Moorfields Eye Hospital/UCL Institute of Ophthalmology NIHR Biomedical Research Centre, UCL Institute of Ophthalmology, 11-43 Bath St., London EC1V 9EL, UK
| | - Victoria Tovell
- Department of Cell Biology, Therapeutics and Moorfields Eye Hospital/UCL Institute of Ophthalmology NIHR Biomedical Research Centre, UCL Institute of Ophthalmology, 11-43 Bath St., London EC1V 9EL, UK
| | - Annegret H. Dahlmann-Noor
- Department of Ocular Biology and Therapeutics and Moorfields Eye Hospital/UCL Institute of Ophthalmology NIHR Biomedical Research Centre, UCL Institute of Ophthalmology, 11-43 Bath St., London EC1V 9EL, UK
| | - Peng T Khaw
- Department of Ocular Biology and Therapeutics and Moorfields Eye Hospital/UCL Institute of Ophthalmology NIHR Biomedical Research Centre, UCL Institute of Ophthalmology, 11-43 Bath St., London EC1V 9EL, UK
| | - Maryse Bailly
- Department of Cell Biology, Therapeutics and Moorfields Eye Hospital/UCL Institute of Ophthalmology NIHR Biomedical Research Centre, UCL Institute of Ophthalmology, 11-43 Bath St., London EC1V 9EL, UK
| |
Collapse
|
50
|
Garg P, Verma R, Cook L, Soofi A, Venkatareddy M, George B, Mizuno K, Gurniak C, Witke W, Holzman LB. Actin-depolymerizing factor cofilin-1 is necessary in maintaining mature podocyte architecture. J Biol Chem 2010; 285:22676-88. [PMID: 20472933 PMCID: PMC2903407 DOI: 10.1074/jbc.m110.122929] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2010] [Revised: 04/30/2010] [Indexed: 12/23/2022] Open
Abstract
Actin dynamics determines podocyte morphology during development and in response to podocyte injury and might be necessary for maintaining normal podocyte morphology. Because podocyte intercellular junction receptor Nephrin plays a role in regulating actin dynamics, and given the described role of cofilin in actin filament polymerization and severing, we hypothesized that cofilin-1 activity is regulated by Nephrin and is necessary in normal podocyte actin dynamics. Nephrin activation induced cofilin dephosphorylation via intermediaries that include phosphatidylinositol 3-kinase, SSH1, 14-3-3, and LIMK in a cell culture model. This Nephrin-induced cofilin activation required a direct interaction between Nephrin and the p85 subunit of phosphatidylinositol 3-kinase. In a similar fashion, cofilin-1 dephosphorylation was observed in a rat model of podocyte injury at a time when foot process spreading is initially observed. To investigate the necessity of cofilin-1 in the glomerulus, podocyte-specific Cfl1 null mice were generated. Cfl1 null podocytes developed normally. However, these mice developed persistent proteinuria by 3 months of age, although they did not exhibit foot process spreading until 8 months, when the rate of urinary protein excretion became more exaggerated. In a mouse model of podocyte injury, protamine sulfate perfusion of the Cfl1 mutant mouse induced a broadened and flattened foot process morphology that was distinct from that observed following perfusion of control kidneys, and mutant podocytes did not recover normal structure following additional perfusion with heparin sulfate. We conclude that cofilin-1 is necessary for maintenance of normal podocyte architecture and for actin structural changes that occur during induction and recovery from podocyte injury.
Collapse
Affiliation(s)
- Puneet Garg
- From the Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Rakesh Verma
- From the Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Leslie Cook
- From the Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Abdul Soofi
- From the Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Madhusudan Venkatareddy
- From the Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Britta George
- the Department of Veterans Affairs, Philadelphia, Pennsylvania 19104
| | - Kensaku Mizuno
- the Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578 Japan
| | - Christine Gurniak
- the Institute of Genetics, University of Bonn, Bonn 53117, Germany, and
| | - Walter Witke
- the Institute of Genetics, University of Bonn, Bonn 53117, Germany, and
| | - Lawrence B. Holzman
- the Department of Veterans Affairs, Philadelphia, Pennsylvania 19104
- the Division of Nephrology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104
| |
Collapse
|