1
|
Hardin KR, Penas AB, Joubert S, Ye C, Myers KR, Zheng JQ. A Critical Role for the Fascin Family of Actin Bundling Proteins in Axon Development, Brain Wiring and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.21.639554. [PMID: 40027761 PMCID: PMC11870622 DOI: 10.1101/2025.02.21.639554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Actin-based cell motility drives many neurodevelopmental events including guided axonal growth. Fascin is a major family of F-actin bundling proteins, but its role in axon development in vivo and brain wiring remains unclear. Here, we report that fascin is required for axon development, brain wiring and function. We show that fascin is enriched in the motile filopodia of axonal growth cones and its inhibition impairs axonal extension and branching of hippocampal neurons in culture. We next provide evidence that fascin is essential for axon development and brain wiring in vivo using Drosophila melanogaster as a model. Drosophila expresses a single ortholog of mammalian fascin called Singed (SN), which is highly expressed in the mushroom body (MB) of the central nervous system. We observe that loss of SN results in drastic MB disruption, highlighted by α- and β-lobe defects that are consistent with altered axonal guidance. SN-null flies also exhibit defective sensorimotor behaviors as assessed by the negative geotaxis assay. MB- specific expression of SN in SN-null flies rescues MB structure and sensorimotor deficits, indicating that SN functions autonomously in MB neurons. Together, our data from primary neuronal culture and in vivo models highlight a critical role for fascin in brain development and function. Highlights Fascin regulates axonal growth and branching of hippocampal neurons in culture.Singed, Drosophila fascin, is enriched specifically in mushroom body (MB) axons.Singed loss causes axon guidance defects and sensorimotor issues in flies.MB-specific Singed re-expression rescues MB structure and behavior in flies.
Collapse
|
2
|
Pan J, Chen K, Lin L, Xu LY, Li EM, Dong G. Exploring the Allosteric Response of Fascin to Its Inhibitor. J Phys Chem B 2024; 128:12050-12058. [PMID: 39621550 DOI: 10.1021/acs.jpcb.4c04813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Fascin is a major actin-binding protein (ABP) for stabilizing filopodia to support efficient adhesion and migration of cancer cells. Fascin is also highly expressed in metastatic tumors. Disrupting the actin-binding site (ABS) on fascin constitutes a critical approach to hindering tumor metastasis. The G2 series of small molecules was formulated with the specific purpose of obstructing the binding pocket of fascin. The determination of inhibitor-induced structural dynamics in fascin is crucial for a comprehensive of its biological functions and the strategic development of pharmacological interventions. In this study, we utilized both equilibrium and dynamical-nonequilibrium molecular dynamics (D-NEMD) to elucidate the molecular mechanisms responsible for transmitting structural changes when removing the G2 inhibitor, in both the wild type (WT) and its variants. Our findings indicate that when G2 is removed, structural dynamics in fascin originate from the G2 binding pocket of fascin and propagate signals through the conformational transformation that spans all four β-trefoil domains. Although different mutant variants demonstrated comparable conformational networks, they showed varying response times. However, the signaling pathways in mutants remained consistent in comparison to the WT fascin. This study provides valuable insights into the structural features and communication pathways of fascin and provides avenues for the development of targeted inhibitors with promising prospects in cancer therapy.
Collapse
Affiliation(s)
- Jinmei Pan
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, PR China
| | - Kai Chen
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, PR China
| | - Lirui Lin
- Department of Bioinformatics, Shantou University Medical College, Shantou 515041, PR China
- Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Shantou University Medical College, Shantou 515041, PR China
| | - Li-Yan Xu
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, PR China
- Cancer Research Center, Shantou University Medical College, Shantou 515041, PR China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, PR China
- Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Shantou University Medical College, Shantou 515041, PR China
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, PR China
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou 515041, PR China
- Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Shantou University Medical College, Shantou 515041, PR China
| | - Geng Dong
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, PR China
- Medical Informatics Research Center, Shantou University Medical College, Shantou 515041, PR China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, PR China
- Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Shantou University Medical College, Shantou 515041, PR China
| |
Collapse
|
3
|
Remsburg CM, Konrad KD, Testa MD, Stepicheva N, Lee K, Choe LH, Polson S, Bhavsar J, Huang H, Song JL. miR-31-mediated local translation at the mitotic spindle is important for early development. Development 2024; 151:dev202619. [PMID: 39250531 PMCID: PMC11423917 DOI: 10.1242/dev.202619] [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: 12/15/2023] [Accepted: 07/17/2024] [Indexed: 09/11/2024]
Abstract
miR-31 is a highly conserved microRNA that plays crucial roles in cell proliferation, migration and differentiation. We discovered that miR-31 and some of its validated targets are enriched on the mitotic spindle of the dividing sea urchin embryo and mammalian cells. Using the sea urchin embryo, we found that miR-31 inhibition led to developmental delay correlated with increased cytoskeletal and chromosomal defects. We identified miR-31 to directly suppress several actin remodeling transcripts, including β-actin, Gelsolin, Rab35 and Fascin. De novo translation of Fascin occurs at the mitotic spindle of sea urchin embryos and mammalian cells. Importantly, miR-31 inhibition leads to a significant a increase of newly translated Fascin at the spindle of dividing sea urchin embryos. Forced ectopic localization of Fascin transcripts to the cell membrane and translation led to significant developmental and chromosomal segregation defects, highlighting the importance of the regulation of local translation by miR-31 at the mitotic spindle to ensure proper cell division. Furthermore, miR-31-mediated post-transcriptional regulation at the mitotic spindle may be an evolutionarily conserved regulatory paradigm of mitosis.
Collapse
Affiliation(s)
- Carolyn M. Remsburg
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Kalin D. Konrad
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Michael D. Testa
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Nadezda Stepicheva
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Kelvin Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
- National Institute for Innovation in Manufacturing Biopharmaceuticals, Newark, DE 19716, USA
| | - Leila H. Choe
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
- National Institute for Innovation in Manufacturing Biopharmaceuticals, Newark, DE 19716, USA
| | - Shawn Polson
- Department of Computer and Informational Sciences; Plant & Soil Sciences; Biological Sciences, CBCB Bioinformatics Core Facility; Bioinformatics, Healthcare Informatics, and Data Science Network of Delaware, University of Delaware, Newark, DE 19716, USA
| | - Jaysheel Bhavsar
- Department of Computer and Informational Sciences, University of Delaware, DE 19716, USA
| | - Hongzhan Huang
- Department of Computer and Informational Sciences, University of Delaware, DE 19716, USA
| | - Jia L. Song
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
4
|
Kulkarni S, Alampally H, Guddattu V, Rodrigues G, Carnelio S. Expression of Fascin and SALL4 in odontogenic cysts and tumors: an immunohistochemical appraisal. F1000Res 2024; 11:1578. [PMID: 38895097 PMCID: PMC11184278 DOI: 10.12688/f1000research.126091.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 06/21/2024] Open
Abstract
Background Various stemness markers (SOX2, OCT4, and NANOG) have been studied in odontogenic cysts and tumors. However, studies on SALL4 having similar properties of stemness has not been documented. Additionally, insight into fascin as a migratory molecule is less explored. In this study, the expression of SALL4 and fascin were evaluated in ameloblastoma, adenomatoid odontogenic tumor (AOT), odontogenic keratocyst (OKC), dentigerous cyst (DC), radicular cyst (RC), and calcifying odontogenic cyst (COC). Methods Semi-quantitative analysis of fascin and SALL4 immuno-positive cells was done in a total of 40 cases of ameloblastoma (11 plexiform, 12 follicular, 12 unicystic, and 5 desmoplastic) variants, 6 cases of AOT, 15 each of OKC, DC, RC and 5 of COC. Chi-square test was applied to evaluate the association between SALL4 and fascin expression in odontogenic cysts and tumors. Results Fascin immunopositivity was observed in peripheral ameloblast-like cells, and the expression was weak or absent in stellate reticulum-like cells. A moderate to weak immune-reactivity to SALL4 was observed in the cytoplasm of ameloblastoma, epithelial cells of dentigerous and radicular cysts, having a marked inflammatory infiltrate, which was an interesting observation. COC and AOT had negative to weak expressions. No recurrence has been reported. Conclusions Expression of fascin in ameloblastomas elucidate their role in motility and localized invasion. Its expression in less aggressive lesions like DC, COC, AOT will incite to explore the other functional properties of fascin. SALL4 expression in the cytoplasm of odontogenic cysts and tumors may represent inactive or mutant forms which requires further validation.
Collapse
Affiliation(s)
- Spoorti Kulkarni
- Oral Pathology and Microbiology, Manipal College of Dental Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Harishanker Alampally
- Oral Pathology and Microbiology, Manipal College of Dental Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Vasudev Guddattu
- Department of Data Science, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Gabriel Rodrigues
- Department of General Surgery, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sunitha Carnelio
- Oral Pathology and Microbiology, Manipal College of Dental Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| |
Collapse
|
5
|
Isogai T, Murali VS, Zhou F, Wang X, Rajendran D, Perez-Castro L, Venkateswaran N, Conacci-Sorrell M, Danuser G. Anchorage-independent cell proliferation promoted by fascin's F-actin bundling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.04.592404. [PMID: 38746129 PMCID: PMC11092747 DOI: 10.1101/2024.05.04.592404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The actin filament (F-actin) bundling protein fascin-1 is highly enriched in many metastatic cancers. Fascin's contribution to metastasis have been ascribed to its enhancement of cell migration and invasion. However, mouse genetic studies clearly point to functions also in tumorigenesis, yet without mechanistic underpinnings. Here, we show that fascin expression promotes the formation of a non-canonical signaling complex that enables anchorage-independent proliferation. This complex shares similarities to focal adhesions and we refer to them as pseudo-adhesion signaling scaffolds (PASS). PASS are enriched with tyrosine phosphorylated proteins and require fascin's F-actin-bundling activity for its assembly. PASS serve as hubs for the Rac1/PAK/JNK proliferation signaling axis, driven by PASS-associated Rac-specific GEFs. Experimental disruption of either fascin or RacGEF function abrogates sustained proliferation of aggressive cancers in vitro and in vivo . These results add a new molecular element to the growing arsenal of metabolic and oncogenic signaling programs regulated by the cytoskeleton architecture.
Collapse
|
6
|
Liu H, Hao W, Wang X, Zhang Y, He L, Xue X, Yang J, Zhang C. Identification of novel molecules and pathways associated with fascin actin‑bundling protein 1 in laryngeal squamous cell carcinoma through comprehensive transcriptome analysis. Int J Mol Med 2024; 53:39. [PMID: 38426543 PMCID: PMC10914310 DOI: 10.3892/ijmm.2024.5363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/14/2024] [Indexed: 03/02/2024] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a common malignant tumor with a poor prognosis. Fascin actin‑bundling protein 1 (FSCN1) has been reported to play a crucial role in the development and progression of LSCC; however, the underlying molecular mechanisms remain unknown. Herein, a whole transcriptome microarray analysis was performed to screen for differentially expressed genes (DEGs) in cells in which FSCN1 was knocked down. A total of 462 up and 601 downregulated mRNA transcripts were identified. Functional annotation analysis revealed that these DEGs were involved in multiple biological functions, such as transcriptional regulation, response to radiation, focal adhesion, extracellular matrix‑receptor interaction, steroid biosynthesis and others. Through co‑expression and protein‑protein interaction analysis, FSCN1 was linked to novel functions, including defense response to virus and steroid biosynthesis. Furthermore, crosstalk analysis with FSCN1‑interacting proteins revealed seven DEGs, identified as FSCN1‑interacting partners, in LSCC cells, three of which were selected for further validation. Co‑immunoprecipitation validation confirmed that FSCN1 interacted with prostaglandin reductase 1 and 24‑dehydrocholesterol reductase (DHCR24). Of note, DHCR24 is a key enzyme involved in cholesterol biosynthesis, and its overexpression promotes the proliferation and migration of LSCC cells. These findings suggest that DHCR24 is a novel molecule associated with FSCN1 in LSCC, and that the FSCN1‑DHCR24 interaction may promote LSCC progression by regulating cholesterol metabolism‑related signaling pathways.
Collapse
Affiliation(s)
- Hongliang Liu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Otolaryngology Head and Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Cell Biology, The Basic Medical School of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Wenjing Hao
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Cell Biology, The Basic Medical School of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xinfang Wang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Cell Biology, The Basic Medical School of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yuliang Zhang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Long He
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xuting Xue
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jiao Yang
- Department of Anatomy, The Basic Medical School of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Chunming Zhang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
- Department of Otolaryngology Head and Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
7
|
Brücker L, Becker SK, Maissl V, Harms G, Parsons M, May-Simera HL. The actin-bundling protein Fascin-1 modulates ciliary signalling. J Mol Cell Biol 2023; 15:mjad022. [PMID: 37015875 PMCID: PMC10485897 DOI: 10.1093/jmcb/mjad022] [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: 02/17/2022] [Revised: 12/22/2022] [Accepted: 01/25/2023] [Indexed: 04/06/2023] Open
Abstract
Primary cilia are microtubule-based cell organelles important for cellular communication. Since they are involved in the regulation of numerous signalling pathways, defects in cilia development or function are associated with genetic disorders, collectively called ciliopathies. Besides their ciliary functions, recent research has shown that several ciliary proteins are involved in the coordination of the actin cytoskeleton. Although ciliary and actin phenotypes are related, the exact nature of their interconnection remains incompletely understood. Here, we show that the protein BBS6, associated with the ciliopathy Bardet-Biedl syndrome, cooperates with the actin-bundling protein Fascin-1 in regulating filopodia and ciliary signalling. We found that loss of Bbs6 affects filopodia length potentially via attenuated interaction with Fascin-1. Conversely, loss of Fascin-1 leads to a ciliary phenotype, subsequently affecting ciliary Wnt signalling, possibly in collaboration with BBS6. Our data shed light on how ciliary proteins are involved in actin regulations and provide new insight into the involvement of the actin regulator Fascin-1 in ciliogenesis and cilia-associated signalling. Advancing our knowledge of the complex regulations between primary cilia and actin dynamics is important to understand the pathogenic consequences of ciliopathies.
Collapse
Affiliation(s)
- Lena Brücker
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Stefanie Kornelia Becker
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Vanessa Maissl
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Gregory Harms
- Imaging Core Facility, Cell Biology Unit, University Medical Centre, Johannes Gutenberg University Mainz, 55101 Mainz, Germany
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Helen Louise May-Simera
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| |
Collapse
|
8
|
Ruggiero C, Tamburello M, Rossini E, Zini S, Durand N, Cantini G, Cioppi F, Hantel C, Kiseljak-Vassiliades K, Wierman ME, Landwehr LS, Weigand I, Kurlbaum M, Zizioli D, Turtoi A, Yang S, Berruti A, Luconi M, Sigala S, Lalli E. FSCN1 as a new druggable target in adrenocortical carcinoma. Int J Cancer 2023; 153:210-223. [PMID: 36971100 DOI: 10.1002/ijc.34526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/02/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023]
Abstract
Adrenocortical carcinoma (ACC) is a rare endocrine malignancy with a high risk of relapse and metastatic spread. The actin-bundling protein fascin (FSCN1) is overexpressed in aggressive ACC and represents a reliable prognostic indicator. FSCN1 has been shown to synergize with VAV2, a guanine nucleotide exchange factor for the Rho/Rac GTPase family, to enhance the invasion properties of ACC cancer cells. Based on those results, we investigated the effects of FSCN1 inactivation by CRISPR/Cas9 or pharmacological blockade on the invasive properties of ACC cells, both in vitro and in an in vivo metastatic ACC zebrafish model. Here, we showed that FSCN1 is a transcriptional target for β-catenin in H295R ACC cells and that its inactivation resulted in defects in cell attachment and proliferation. FSCN1 knock-out modulated the expression of genes involved in cytoskeleton dynamics and cell adhesion. When Steroidogenic Factor-1 (SF-1) dosage was upregulated in H295R cells, activating their invasive capacities, FSCN1 knock-out reduced the number of filopodia, lamellipodia/ruffles and focal adhesions, while decreasing cell invasion in Matrigel. Similar effects were produced by the FSCN1 inhibitor G2-044, which also diminished the invasion of other ACC cell lines expressing lower levels of FSCN1 than H295R. In the zebrafish model, metastases formation was significantly reduced in FSCN1 knock-out cells and G2-044 significantly reduced the number of metastases formed by ACC cells. Our results indicate that FSCN1 is a new druggable target for ACC and provide the rationale for future clinical trials with FSCN1 inhibitors in patients with ACC.
Collapse
Affiliation(s)
- Carmen Ruggiero
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, 06560, Valbonne, France
- Université Côte d'Azur, 06560, Valbonne, France
| | - Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Elisa Rossini
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Silvia Zini
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Nelly Durand
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, 06560, Valbonne, France
- Université Côte d'Azur, 06560, Valbonne, France
| | - Giulia Cantini
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50134, Florence, Italy
| | - Francesca Cioppi
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50134, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134, Florence, Italy
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), 8091, Zürich, Switzerland
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, 01307, Dresden, Germany
| | - Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, 80045, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Affairs Medical Center, 80045, Aurora, Colorado, USA
| | - Margaret E Wierman
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, 80045, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Affairs Medical Center, 80045, Aurora, Colorado, USA
| | - Laura-Sophie Landwehr
- Division of Endocrinology and Diabetology-Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
| | - Isabel Weigand
- Division of Endocrinology and Diabetology-Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
- Department of Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, 81377, Munich, Germany
| | - Max Kurlbaum
- Division of Endocrinology and Diabetology-Department of Internal Medicine I, University Hospital, University of Würzburg, 97080, Würzburg, Germany
| | - Daniela Zizioli
- Section of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Andrei Turtoi
- Tumor Microenvironment and Resistance to Therapy Laboratory, Institut de Recherche en Cancérologie de Montpellier, Université de Montpellier-INSERM U1194, 34090, Montpellier, France
- Platform for Translational Oncometabolomics, Biocampus, CNRS-INSERM-Université de Montpellier, 34090, Montpellier, France
| | - Shengyu Yang
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, 17033, Hershey, Pennsylvania, USA
| | - Alfredo Berruti
- Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia and ASST Spedali Civili di Brescia, 25123, Brescia, Italy
| | - Michaela Luconi
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50134, Florence, Italy
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25124, Brescia, Italy
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, 06560, Valbonne, France
- Université Côte d'Azur, 06560, Valbonne, France
- Inserm, 06560, Valbonne, France
| |
Collapse
|
9
|
Zhang H, Fan J, Maclin JM, Wan LQ. The Actin Crosslinker Fascin Regulates Cell Chirality. Adv Biol (Weinh) 2023; 7:e2200240. [PMID: 36658789 PMCID: PMC10293081 DOI: 10.1002/adbi.202200240] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/27/2022] [Indexed: 01/21/2023]
Abstract
The left-right (L-R) asymmetry of the cells, or cell chirality, is a well-known intrinsic property derived from the dynamic organization of the actin cytoskeleton. Cell chirality can be regulated by actin-binding proteins such as α-actinin-1 and can also be mediated by certain signaling pathways, such as protein kinase C (PKC) signaling. Fascin, an actin crosslinker known to mediate parallel bundling of actin filaments, appears as a prominent candidate in cell chirality regulation, given its role in facilitating cell migration as an important PKC substrate. Here, it is shown that the chirality of NIH/3T3 cells can be altered by PKC activation and fascin manipulation. With either small-molecule drug inhibition or genetic knockdown of fascin, the chirality of 3T3 cells is reversed from a clockwise (CW) bias to a counterclockwise (CCW) bias on ring-shaped micropatterns, accompanied by the reversal in cell directional migration. The Ser-39 fascin-actin binding sites are further explored in cell chirality regulation. The findings of this study reveal the critical role of fascin as an important intermediator in cell chirality, shedding novel insights into the mechanisms of L-R asymmetric cell migration and multicellular morphogenesis.
Collapse
Affiliation(s)
- Haokang Zhang
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Jie Fan
- Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, MI 48128, USA
| | - Joshua M.A. Maclin
- Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Department of Biological Science, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Leo Q. Wan
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Department of Biological Science, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Modeling, Simulation and Imaging in Medicine, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
10
|
Rajan S, Kudryashov DS, Reisler E. Actin Bundles Dynamics and Architecture. Biomolecules 2023; 13:450. [PMID: 36979385 PMCID: PMC10046292 DOI: 10.3390/biom13030450] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/04/2023] Open
Abstract
Cells use the actin cytoskeleton for many of their functions, including their division, adhesion, mechanosensing, endo- and phagocytosis, migration, and invasion. Actin bundles are the main constituent of actin-rich structures involved in these processes. An ever-increasing number of proteins that crosslink actin into bundles or regulate their morphology is being identified in cells. With recent advances in high-resolution microscopy and imaging techniques, the complex process of bundles formation and the multiple forms of physiological bundles are beginning to be better understood. Here, we review the physiochemical and biological properties of four families of highly conserved and abundant actin-bundling proteins, namely, α-actinin, fimbrin/plastin, fascin, and espin. We describe the similarities and differences between these proteins, their role in the formation of physiological actin bundles, and their properties-both related and unrelated to their bundling abilities. We also review some aspects of the general mechanism of actin bundles formation, which are known from the available information on the activity of the key actin partners involved in this process.
Collapse
Affiliation(s)
- Sudeepa Rajan
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Dmitri S. Kudryashov
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH 43210, USA
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
11
|
Zhang N, Bian Q, Gao Y, Wang Q, Shi Y, Li X, Ma X, Chen H, Zhao Z, Yu H. The Role of Fascin-1 in Human Urologic Cancers: A Promising Biomarker or Therapeutic Target? Technol Cancer Res Treat 2023; 22:15330338231175733. [PMID: 37246525 PMCID: PMC10240877 DOI: 10.1177/15330338231175733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/04/2023] [Accepted: 04/24/2023] [Indexed: 05/30/2023] Open
Abstract
Human cancer statistics show that an increased incidence of urologic cancers such as bladder cancer, prostate cancer, and renal cell carcinoma. Due to the lack of early markers and effective therapeutic targets, their prognosis is poor. Fascin-1 is an actin-binding protein, which functions in the formation of cell protrusions by cross-linking with actin filaments. Studies have found that fascin-1 expression is elevated in most human cancers and is related to outcomes such as neoplasm metastasis, reduced survival, and increased aggressiveness. Fascin-1 has been considered as a potential therapeutic target for urologic cancers, but there is no comprehensive review to evaluate these studies. This review aimed to provide an enhanced literature review, outline, and summarize the mechanism of fascin-1 in urologic cancers and discuss the therapeutic potential of fascin-1 and the possibility of its use as a potential marker. We also focused on the correlation between the overexpression of fascin-1 and clinicopathological parameters. Mechanistically, fascin-1 is regulated by several regulators and signaling pathways (such as long noncoding RNA, microRNA, c-Jun N-terminal kinase, and extracellular regulated protein kinases). The overexpression of fascin-1 is related to clinicopathologic parameters such as pathological stage, bone or lymph node metastasis, and reduced disease-free survival. Several fascin-1 inhibitors (G2, NP-G2-044) have been evaluated in vitro and in preclinical models. The study proved the promising potential of fascin-1 as a newly developing biomarker and a potential therapeutic target that needs further investigation. The data also highlight the inadequacy of fascin-1 to serve as a novel biomarker for prostate cancer.
Collapse
Affiliation(s)
- Naibin Zhang
- Department of Biochemistry, Jining Medical University, Jining, Shandong, People's Republic of China
- Clinical Medical College, Jining Medical University, Jining, Shandong, People's Republic of China
| | - Qiang Bian
- Department of Biochemistry, Jining Medical University, Jining, Shandong, People's Republic of China
- Department of Pathophysiology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Yankun Gao
- Clinical Medical College, Jining Medical University, Jining, Shandong, People's Republic of China
| | - Qianqian Wang
- Department of Biochemistry, Jining Medical University, Jining, Shandong, People's Republic of China
| | - Ying Shi
- Department of Biochemistry, Jining Medical University, Jining, Shandong, People's Republic of China
| | - Xiangling Li
- Department of Biochemistry, Jining Medical University, Jining, Shandong, People's Republic of China
| | - Xiaolei Ma
- Department of Biochemistry, Jining Medical University, Jining, Shandong, People's Republic of China
| | - Huiyuan Chen
- College of Radiology, Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Zhankui Zhao
- The Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, People's Republic of China
| | - Honglian Yu
- Department of Biochemistry, Jining Medical University, Jining, Shandong, People's Republic of China
- Collaborative Innovation Center, Jining Medical University, Jining, Shandong, People's Republic of China
| |
Collapse
|
12
|
He Z, Wang J, Xu J, Jiang X, Liu X, Jiang J. Dynamic regulation of KIF15 phosphorylation and acetylation promotes focal adhesions disassembly in pancreatic cancer. Cell Death Dis 2022; 13:896. [PMID: 36280663 PMCID: PMC9592618 DOI: 10.1038/s41419-022-05338-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
Pancreatic cancer (PC) is prone to distant metastasis in the early stage, which is attributed to the strong migration ability of tumor cells. Focal adhesion turnover is essential for cancer cell metastasis, and the integrin recycling process is a key activation pathway for focal adhesion depolymerization. To identify the key motor protein involving in the integrin β1 recycling, we screened kinesin proteins involved in integrin β1 recycling using a kinesin family siRNA library and identified kinesin family 15 (KIF15) as a key regulator. KIF15 was upregulated in metastasis PC tissues and promoted PC cell migration and invasion. We identified KIF15 as a key component mediating integrin β1/FAK signaling that accelerated FA disassembly in a FAK-Y397-dependent manner. KIF15 recruited PI3K-C2α to promote integrin β1/FAK signaling and FA disassembly in a RAB11A-dependent manner. The C-terminal tail of KIF15 is required for the PI3K-C2α interaction and RAB11A activation. In addition, we also found that SIRT1-mediated acetylation of KIF15 is essential for KIF15 phosphorylation, which is the key activation event in motor protein function. Together, these findings indicate that KIF15 interacts with PI3K-C2α to promote FA turnover in PC cells by controlling the endosome recycling of integrin β1 in a SIRT1 acetylation modification-dependent manner, eventually promoting focal adhesions turnover and distant metastasis in PC.
Collapse
Affiliation(s)
- Zhiwei He
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jie Wang
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jian Xu
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xueyi Jiang
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xinyuan Liu
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jianxin Jiang
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| |
Collapse
|
13
|
Lawson CD, Peel S, Jayo A, Corrigan A, Iyer P, Baxter Dalrymple M, Marsh RJ, Cox S, Van Audenhove I, Gettemans J, Parsons M. Nuclear fascin regulates cancer cell survival. eLife 2022; 11:e79283. [PMID: 36039640 PMCID: PMC9427113 DOI: 10.7554/elife.79283] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Fascin is an important regulator of F-actin bundling leading to enhanced filopodia assembly. Fascin is also overexpressed in most solid tumours where it supports invasion through control of F-actin structures at the periphery and nuclear envelope. Recently, fascin has been identified in the nucleus of a broad range of cell types but the contributions of nuclear fascin to cancer cell behaviour remain unknown. Here, we demonstrate that fascin bundles F-actin within the nucleus to support chromatin organisation and efficient DDR. Fascin associates directly with phosphorylated Histone H3 leading to regulated levels of nuclear fascin to support these phenotypes. Forcing nuclear fascin accumulation through the expression of nuclear-targeted fascin-specific nanobodies or inhibition of Histone H3 kinases results in enhanced and sustained nuclear F-actin bundling leading to reduced invasion, viability, and nuclear fascin-specific/driven apoptosis. These findings represent an additional important route through which fascin can support tumourigenesis and provide insight into potential pathways for targeted fascin-dependent cancer cell killing.
Collapse
Affiliation(s)
- Campbell D Lawson
- Randall Centre for Cell and Molecular Biophysics, King’s College London, Guy’s CampusLondonUnited Kingdom
| | - Samantha Peel
- Discovery Sciences, R&D, AstraZeneca (United Kingdom)CambridgeUnited Kingdom
| | - Asier Jayo
- Randall Centre for Cell and Molecular Biophysics, King’s College London, Guy’s CampusLondonUnited Kingdom
| | - Adam Corrigan
- Discovery Sciences, R&D, AstraZeneca (United Kingdom)CambridgeUnited Kingdom
| | - Preeti Iyer
- Molecular AI, Discovery Sciences, R&D, AstraZeneca (Sweden)MölndalSweden
| | - Mabel Baxter Dalrymple
- Randall Centre for Cell and Molecular Biophysics, King’s College London, Guy’s CampusLondonUnited Kingdom
| | - Richard J Marsh
- Randall Centre for Cell and Molecular Biophysics, King’s College London, Guy’s CampusLondonUnited Kingdom
| | - Susan Cox
- Randall Centre for Cell and Molecular Biophysics, King’s College London, Guy’s CampusLondonUnited Kingdom
| | - Isabel Van Audenhove
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent UniversityGhentBelgium
| | - Jan Gettemans
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent UniversityGhentBelgium
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King’s College London, Guy’s CampusLondonUnited Kingdom
| |
Collapse
|
14
|
Cai H, Wang R, Tang Z, Lu T, Cui Y. FSCN1 Promotes Esophageal Carcinoma Progression Through Downregulating PTK6 via its RNA-Binding Protein Effect. Front Pharmacol 2022; 13:868296. [PMID: 35401239 PMCID: PMC8984143 DOI: 10.3389/fphar.2022.868296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Esophageal squamous cell carcinoma (ESCC) causes many deaths worldwide every year. Fascin actin-bundling protein 1(FSCN1) has been reported to be a promoter of ESCC via its actin-binding function, however, its new role as an RNA-binding protein (RBP) has not been investigated. Here, we explored the RBP role of FSCN1 in the development of ESCC. Methods: Whole-genome expression sequencing was performed to screen for altered genes after FSCN1 knockdown. RNA immunoprecipitation was performed to determine the target mRNA of FSCN1 as an RBP. In vitro experiments with ECA-109 and KYSE-150 and ex vivo experiments in tumor-bearing mice were performed to investigate the effects of FSCN1 and Protein Tyrosine Kinase 6 (PTK6) on ESCC progression. Results: FSCN1 could downregulate mRNA and the protein level of PTK6. The binding position of PTK6 (PTK6-T2) pre-mRNA to FSCN1 was determined. PTK6-T2 blocked the binding between FSCN1 and the pre-mRNA of PTK6, and thus reversed the promotion effect of FSCN1 on ESCC tumor progression via the AKT/GSK3β signaling pathway. Conclusion: A novel effect of FSCN1, RBP-binding with the pre-mRNA of PTK6, was confirmed to play an important role in ESCC progression. PTK6-T2, which is a specific inhibitor of FSCN1 binding to the pre-mRNA of PTK6, could impede the development of ESCC.
Collapse
Affiliation(s)
- Hongfei Cai
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Rui Wang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China.,Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Ze Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Tianyu Lu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Youbin Cui
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
15
|
Gupta I, Vranic S, Al-Thawadi H, Al Moustafa AE. Fascin in Gynecological Cancers: An Update of the Literature. Cancers (Basel) 2021; 13:cancers13225760. [PMID: 34830909 PMCID: PMC8616296 DOI: 10.3390/cancers13225760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Fascin, an actin-binding protein, is upregulated in different types of human cancers. It is reportedly responsible for increasing the invasive and metastatic ability of cancer cells by reducing cell–cell adhesions. This review provides a brief overview of fascin and its interactions with other genes and oncoviruses to induce the onset and progression of cancer. Abstract Fascin is an actin-binding protein that is encoded by the FSCN1 gene (located on chromosome 7). It triggers membrane projections and stimulates cell motility in cancer cells. Fascin overexpression has been described in different types of human cancers in which its expression correlated with tumor growth, migration, invasion, and metastasis. Moreover, overexpression of fascin was found in oncovirus-infected cells, such as human papillomaviruses (HPVs) and Epstein-Barr virus (EBV), disrupting the cell–cell adhesion and enhancing cancer progression. Based on these findings, several studies reported fascin as a potential biomarker and a therapeutic target in various cancers. This review provides a brief overview of the FSCN1 role in various cancers with emphasis on gynecological malignancies. We also discuss fascin interactions with other genes and oncoviruses through which it might induce cancer development and progression.
Collapse
Affiliation(s)
- Ishita Gupta
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (I.G.); (S.V.); (H.A.-T.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
| | - Semir Vranic
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (I.G.); (S.V.); (H.A.-T.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
| | - Hamda Al-Thawadi
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (I.G.); (S.V.); (H.A.-T.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
| | - Ala-Eddin Al Moustafa
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (I.G.); (S.V.); (H.A.-T.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
- Biomedical Research Centre, QU Health, Qatar University, Doha 2713, Qatar
- Correspondence: ; Tel.: +974-4403-7817
| |
Collapse
|
16
|
Lamb MC, Kaluarachchi CP, Lansakara TI, Mellentine SQ, Lan Y, Tivanski AV, Tootle TL. Fascin limits Myosin activity within Drosophila border cells to control substrate stiffness and promote migration. eLife 2021; 10:69836. [PMID: 34698017 PMCID: PMC8547955 DOI: 10.7554/elife.69836] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/11/2021] [Indexed: 12/24/2022] Open
Abstract
A key regulator of collective cell migrations, which drive development and cancer metastasis, is substrate stiffness. Increased substrate stiffness promotes migration and is controlled by Myosin. Using Drosophila border cell migration as a model of collective cell migration, we identify, for the first time, that the actin bundling protein Fascin limits Myosin activity in vivo. Loss of Fascin results in: increased activated Myosin on the border cells and their substrate, the nurse cells; decreased border cell Myosin dynamics; and increased nurse cell stiffness as measured by atomic force microscopy. Reducing Myosin restores on-time border cell migration in fascin mutant follicles. Further, Fascin’s actin bundling activity is required to limit Myosin activation. Surprisingly, we find that Fascin regulates Myosin activity in the border cells to control nurse cell stiffness to promote migration. Thus, these data shift the paradigm from a substrate stiffness-centric model of regulating migration, to uncover that collectively migrating cells play a critical role in controlling the mechanical properties of their substrate in order to promote their own migration. This understudied means of mechanical regulation of migration is likely conserved across contexts and organisms, as Fascin and Myosin are common regulators of cell migration.
Collapse
Affiliation(s)
- Maureen C Lamb
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, United States
| | | | | | - Samuel Q Mellentine
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, United States
| | - Yiling Lan
- Department of Chemistry, University of Iowa, Iowa City, United States
| | - Alexei V Tivanski
- Department of Chemistry, University of Iowa, Iowa City, United States
| | - Tina L Tootle
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, United States
| |
Collapse
|
17
|
Kashgari G, Venkatesh S, Refuerzo S, Pham B, Bayat A, Klein RH, Ramos R, Ta AP, Plikus MV, Wang PH, Andersen B. GRHL3 activates FSCN1 to relax cell-cell adhesions between migrating keratinocytes during wound reepithelialization. JCI Insight 2021; 6:e142577. [PMID: 34494554 PMCID: PMC8492311 DOI: 10.1172/jci.insight.142577] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 07/21/2021] [Indexed: 01/23/2023] Open
Abstract
The migrating keratinocyte wound front is required for skin wound closure. Despite significant advances in wound healing research, we do not fully understand the molecular mechanisms that orchestrate collective keratinocyte migration. Here, we show that, in the wound front, the epidermal transcription factor Grainyhead like-3 (GRHL3) mediates decreased expression of the adherens junction protein E-cadherin; this results in relaxed adhesions between suprabasal keratinocytes, thus promoting collective cell migration and wound closure. Wound fronts from mice lacking GRHL3 in epithelial cells (Grhl3-cKO) have lower expression of Fascin-1 (FSCN1), a known negative regulator of E-cadherin. Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) on wounded keratinocytes shows decreased wound-induced chromatin accessibility near the Fscn1 gene in Grhl3-cKO mice, a region enriched for GRHL3 motifs. These data reveal a wound-induced GRHL3/FSCN1/E-cadherin pathway that regulates keratinocyte-keratinocyte adhesion during wound-front migration; this pathway is activated in acute human wounds and is altered in diabetic wounds in mice, suggesting translational relevance.
Collapse
Affiliation(s)
| | | | | | - Brandon Pham
- Department of Biological Chemistry, School of Medicine
| | - Anita Bayat
- Department of Biological Chemistry, School of Medicine
| | | | - Raul Ramos
- Department of Developmental & Cell Biology, School of Biological Sciences, and
| | - Albert Paul Ta
- Department of Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine (UCI), California, USA
| | - Maksim V Plikus
- Department of Developmental & Cell Biology, School of Biological Sciences, and
| | - Ping H Wang
- Department of Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine (UCI), California, USA
| | - Bogi Andersen
- Department of Biological Chemistry, School of Medicine.,Department of Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine (UCI), California, USA
| |
Collapse
|
18
|
Abstract
Actin filaments and microtubules are cytoskeletal polymers that participate in many vital cell functions including division, morphogenesis, phagocytosis, and motility. Despite the persistent dogma that actin filament and microtubule networks are distinct in localization, structure, and function, a growing body of evidence shows that these elements are choreographed through intricate mechanisms sensitive to either polymer. Many proteins and cellular signals that mediate actin–microtubule interactions have already been identified. However, the impact of these regulators is typically assessed with actin filament or microtubule polymers alone, independent of the other system. Further, unconventional modes and regulators coordinating actin–microtubule interactions are still being discovered. Here we examine several methods of actin–microtubule crosstalk with an emphasis on the molecular links between both polymer systems and their higher-order interactions.
Collapse
Affiliation(s)
- Morgan L Pimm
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - Jessica L Henty-Ridilla
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210.,Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
19
|
Castillo-Badillo JA, Gautam N. An optogenetic model reveals cell shape regulation through FAK and fascin. J Cell Sci 2021; 134:269115. [PMID: 34114634 DOI: 10.1242/jcs.258321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/01/2021] [Indexed: 12/17/2022] Open
Abstract
Cell shape regulation is important, but the mechanisms that govern shape are not fully understood, in part due to limited experimental models in which cell shape changes and underlying molecular processes can be rapidly and non-invasively monitored in real time. Here, we used an optogenetic tool to activate RhoA in the middle of mononucleated macrophages to induce contraction, resulting in a side with the nucleus that retained its shape and a non-nucleated side that was unable to maintain its shape and collapsed. In cells overexpressing focal adhesion kinase (FAK; also known as PTK2), the non-nucleated side exhibited a wide flat morphology and was similar in adhesion area to the nucleated side. In cells overexpressing fascin, an actin-bundling protein, the non-nucleated side assumed a spherical shape and was similar in height to the nucleated side. This effect of fascin was also observed in fibroblasts even without inducing furrow formation. Based on these results, we conclude that FAK and fascin work together to maintain cell shape by regulating adhesion area and height, respectively, in different cell types. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Jean A Castillo-Badillo
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - N Gautam
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA.,Department of Genetics, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
20
|
Liu H, Zhang Y, Li L, Cao J, Guo Y, Wu Y, Gao W. Fascin actin-bundling protein 1 in human cancer: promising biomarker or therapeutic target? Mol Ther Oncolytics 2021; 20:240-264. [PMID: 33614909 PMCID: PMC7873579 DOI: 10.1016/j.omto.2020.12.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fascin actin-bundling protein 1 (FSCN1) is a highly conserved actin-bundling protein that cross links F-actin microfilaments into tight, parallel bundles. Elevated FSCN1 levels have been reported in many types of human cancers and have been correlated with aggressive clinical progression, poor prognosis, and survival outcomes. The overexpression of FSCN1 in cancer cells has been associated with tumor growth, migration, invasion, and metastasis. Currently, FSCN1 is recognized as a candidate biomarker for multiple cancer types and as a potential therapeutic target. The aim of this study was to provide a brief overview of the FSCN1 gene and protein structure and elucidate on its actin-bundling activity and physiological functions. The main focus was on the role of FSCN1 and its upregulatory mechanisms and significance in cancer cells. Up-to-date studies on FSCN1 as a novel biomarker and therapeutic target for human cancers are reviewed. It is shown that FSCN1 is an unusual biomarker and a potential therapeutic target for cancer.
Collapse
Affiliation(s)
- Hongliang Liu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Yu Zhang
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Li Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Jimin Cao
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Yujia Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Yongyan Wu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Wei Gao
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| |
Collapse
|
21
|
Lamb MC, Tootle TL. Fascin in Cell Migration: More Than an Actin Bundling Protein. BIOLOGY 2020; 9:biology9110403. [PMID: 33212856 PMCID: PMC7698196 DOI: 10.3390/biology9110403] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022]
Abstract
Simple Summary Cell migration is an essential biological process that regulates both development and diseases, such as cancer metastasis. Therefore, understanding the factors that promote cell migration is crucial. One of the factors known to regulate cell migration is the actin-binding protein, Fascin. Fascin is typically thought to promote cell migration through bundling actin to form migratory structures such as filopodia and invadapodia. However, Fascin has many other functions in the cell that may contribute to cell migration. How these novel functions promote cell migration and are regulated is still not well understood. Here, we review the structure of Fascin, the many functions of Fascin and how they may promote cell migration, how Fascin is regulated, and Fascin’s role in diseases such as cancer metastasis. Abstract Fascin, an actin-binding protein, regulates many developmental migrations and contributes to cancer metastasis. Specifically, Fascin promotes cell motility, invasion, and adhesion by forming filopodia and invadopodia through its canonical actin bundling function. In addition to bundling actin, Fascin has non-canonical roles in the cell that are thought to promote cell migration. These non-canonical functions include regulating the activity of other actin-binding proteins, binding to and regulating microtubules, mediating mechanotransduction to the nucleus via interaction with the Linker of the Nucleoskeleton and Cytoskeleton (LINC) Complex, and localizing to the nucleus to regulate nuclear actin, the nucleolus, and chromatin modifications. The many functions of Fascin must be coordinately regulated to control cell migration. While much remains to be learned about such mechanisms, Fascin is regulated by post-translational modifications, prostaglandin signaling, protein–protein interactions, and transcriptional means. Here, we review the structure of Fascin, the various functions of Fascin and how they contribute to cell migration, the mechanisms regulating Fascin, and how Fascin contributes to diseases, specifically cancer metastasis.
Collapse
|
22
|
Abstract
Simple Summary Cell migration is an essential process from embryogenesis to cell death. This is tightly regulated by numerous proteins that help in proper functioning of the cell. In diseases like cancer, this process is deregulated and helps in the dissemination of tumor cells from the primary site to secondary sites initiating the process of metastasis. For metastasis to be efficient, cytoskeletal components like actin, myosin, and intermediate filaments and their associated proteins should co-ordinate in an orderly fashion leading to the formation of many cellular protrusions-like lamellipodia and filopodia and invadopodia. Knowledge of this process is the key to control metastasis of cancer cells that leads to death in 90% of the patients. The focus of this review is giving an overall understanding of these process, concentrating on the changes in protein association and regulation and how the tumor cells use it to their advantage. Since the expression of cytoskeletal proteins can be directly related to the degree of malignancy, knowledge about these proteins will provide powerful tools to improve both cancer prognosis and treatment. Abstract Successful metastasis depends on cell invasion, migration, host immune escape, extravasation, and angiogenesis. The process of cell invasion and migration relies on the dynamic changes taking place in the cytoskeletal components; actin, tubulin and intermediate filaments. This is possible due to the plasticity of the cytoskeleton and coordinated action of all the three, is crucial for the process of metastasis from the primary site. Changes in cellular architecture by internal clues will affect the cell functions leading to the formation of different protrusions like lamellipodia, filopodia, and invadopodia that help in cell migration eventually leading to metastasis, which is life threatening than the formation of neoplasms. Understanding the signaling mechanisms involved, will give a better insight of the changes during metastasis, which will eventually help targeting proteins for treatment resulting in reduced mortality and longer survival.
Collapse
|
23
|
Fox EF, Lamb MC, Mellentine SQ, Tootle TL. Prostaglandins regulate invasive, collective border cell migration. Mol Biol Cell 2020; 31:1584-1594. [PMID: 32432969 PMCID: PMC7521797 DOI: 10.1091/mbc.e19-10-0578] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
While prostaglandins (PGs), short-range lipid signals, regulate single cell migration, their roles in collective migration remain unclear. To address this, we use Drosophila border cell migration, an invasive, collective migration that occurs during Stage 9 of oogenesis. Pxt is the Drosophila cyclooxygenase-like enzyme responsible for PG synthesis. Loss of Pxt results in both delayed border cell migration and elongated clusters, whereas somatic Pxt knockdown causes delayed migration and compacted clusters. These findings suggest PGs act in both the border cells and nurse cells, the substrate on which the border cells migrate. As PGs regulate the actin bundler Fascin, and Fascin is required for on-time migration, we assessed whether PGs regulate Fascin to promote border cell migration. Coreduction of Pxt and Fascin results in delayed migration and elongated clusters. The latter may be due to altered cell adhesion, as loss of Pxt or Fascin, or coreduction of both, decreases integrin levels on the border cell membranes. Conversely, integrin localization is unaffected by somatic knockdown of Pxt. Together these data lead to the model that PG signaling controls Fascin in the border cells to promote migration and in the nurse cells to maintain cluster cohesion.
Collapse
Affiliation(s)
- Emily F Fox
- Department of Anatomy and Cell Biology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Maureen C Lamb
- Department of Anatomy and Cell Biology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Samuel Q Mellentine
- Department of Anatomy and Cell Biology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Tina L Tootle
- Department of Anatomy and Cell Biology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
24
|
Lamb MC, Anliker KK, Tootle TL. Fascin regulates protrusions and delamination to mediate invasive, collective cell migration in vivo. Dev Dyn 2020; 249:961-982. [PMID: 32352613 DOI: 10.1002/dvdy.186] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/26/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The actin bundling protein Fascin is essential for developmental cell migrations and promotes cancer metastasis. In addition to bundling actin, Fascin has several actin-independent roles; how these other functions contribute to cell migration remains unclear. Border cell migration during Drosophila oogenesis provides an excellent model to study Fascin's various roles during invasive, collective cell migration. RESULTS On-time border cell migration during Stage 9 requires Fascin (Drosophila Singed). Fascin functions not only within the migrating border cells, but also within the nurse cells, the substrate for this migration. Fascin genetically interacts with the actin elongation factor Enabled to promote on-time Stage 9 migration and overexpression of Enabled suppresses the defects seen with loss of Fascin. Loss of Fascin results in increased, shorter and mislocalized protrusions during migration. Additionally, loss of Fascin inhibits border cell delamination and increases E-Cadherin (Drosophila Shotgun) adhesions on both the border cells and nurse cells. CONCLUSIONS Overall, Fascin promotes on-time border cell migration during Stage 9 and contributes to multiple aspects of this invasive, collective cell migration, including both protrusion dynamics and delamination. These findings have implications beyond Drosophila, as border cell migration has emerged as a model to study mechanisms mediating cancer metastasis.
Collapse
Affiliation(s)
- Maureen C Lamb
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Kelsey K Anliker
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Tina L Tootle
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
25
|
Pfisterer K, Levitt J, Lawson CD, Marsh RJ, Heddleston JM, Wait E, Ameer-Beg SM, Cox S, Parsons M. FMNL2 regulates dynamics of fascin in filopodia. J Cell Biol 2020; 219:e201906111. [PMID: 32294157 PMCID: PMC7199847 DOI: 10.1083/jcb.201906111] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/30/2019] [Accepted: 02/20/2020] [Indexed: 12/31/2022] Open
Abstract
Filopodia are peripheral F-actin-rich structures that enable cell sensing of the microenvironment. Fascin is an F-actin-bundling protein that plays a key role in stabilizing filopodia to support efficient adhesion and migration. Fascin is also highly up-regulated in human cancers, where it increases invasive cell behavior and correlates with poor patient prognosis. Previous studies have shown that fascin phosphorylation can regulate F-actin bundling, and that this modification can contribute to subcellular fascin localization and function. However, the factors that regulate fascin dynamics within filopodia remain poorly understood. In the current study, we used advanced live-cell imaging techniques and a fascin biosensor to demonstrate that fascin phosphorylation, localization, and binding to F-actin are highly dynamic and dependent on local cytoskeletal architecture in cells in both 2D and 3D environments. Fascin dynamics within filopodia are under the control of formins, and in particular FMNL2, that binds directly to dephosphorylated fascin. Our data provide new insight into control of fascin dynamics at the nanoscale and into the mechanisms governing rapid cytoskeletal adaptation to environmental changes. This filopodia-driven exploration stage may represent an essential regulatory step in the transition from static to migrating cancer cells.
Collapse
Affiliation(s)
- Karin Pfisterer
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - James Levitt
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
- Microscopy Innovation Centre, King's College London, Guy's Campus, London, UK
| | - Campbell D. Lawson
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - Richard J. Marsh
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - John M. Heddleston
- Advanced Imaging Centre, Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA
| | - Eric Wait
- Advanced Imaging Centre, Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA
| | - Simon Morris Ameer-Beg
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, London, UK
| | - Susan Cox
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| |
Collapse
|
26
|
Barnawi R, Al-Khaldi S, Bakheet T, Fallatah M, Alaiya A, Ghebeh H, Al-Alwan M. Fascin Activates β-Catenin Signaling and Promotes Breast Cancer Stem Cell Function Mainly Through Focal Adhesion Kinase (FAK): Relation With Disease Progression. Front Oncol 2020; 10:440. [PMID: 32373510 PMCID: PMC7186340 DOI: 10.3389/fonc.2020.00440] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/12/2020] [Indexed: 12/28/2022] Open
Abstract
Cancer stem cells (CSCs), a rare population of tumor cells with high self-renewability potential, have gained increasing attention due to their contribution to chemoresistance and metastasis. We have previously demonstrated a critical role for the actin-bundling protein (fascin) in mediating breast cancer chemoresistance through activation of focal adhesion kinase (FAK). The latter is known to trigger the β-catenin signaling pathway. Whether fascin activation of FAK would ultimately trigger β-catenin signaling pathway has not been elucidated. Here, we assessed the effect of fascin manipulation in breast cancer cells on triggering β-catenin downstream targets and its dependence on FAK. Gain and loss of fascin expression showed its direct effect on the constitutive expression of β-catenin downstream targets and enhancement of self-renewability. In addition, fascin was essential for glycogen synthase kinase 3β inhibitor-mediated inducible expression and function of the β-catenin downstream targets. Importantly, fascin-mediated constitutive and inducible expression of β-catenin downstream targets, as well as its subsequent effect on CSC function, was at least partially FAK dependent. To assess the clinical relevance of the in vitro findings, we evaluated the consequence of fascin, FAK, and β-catenin downstream target coexpression on the outcome of breast cancer patient survival. Patients with coexpression of fascinhigh and FAKhigh or high β-catenin downstream targets showed the worst survival outcome, whereas in fascinlow, patient coexpression of FAKhigh or high β-catenin targets had less significant effect on the survival. Altogether, our data demonstrated the critical role of fascin-mediated β-catenin activation and its dependence on intact FAK signaling to promote breast CSC function. These findings suggest that targeting of fascin-FAK-β-catenin axis may provide a novel therapeutic approach for eradication of breast cancer from the root.
Collapse
Affiliation(s)
- Rayanah Barnawi
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Samiyah Al-Khaldi
- National Center for Stem Cells, Life Science and Environment Research Institute, King Abdulaziz City for Sciences and Technology, Riyadh, Saudi Arabia
| | - Tala Bakheet
- Molecular Biomedicine Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mohannad Fallatah
- National Center for Stem Cells, Life Science and Environment Research Institute, King Abdulaziz City for Sciences and Technology, Riyadh, Saudi Arabia
| | - Ayodele Alaiya
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hazem Ghebeh
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Collage of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| | - Monther Al-Alwan
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Collage of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| |
Collapse
|
27
|
Yu FL, Miao H, Xia J, Jia F, Wang H, Xu F, Guo L. Proteomics Analysis Identifies IRSp53 and Fascin as Critical for PRV Egress and Direct Cell-Cell Transmission. Proteomics 2019; 19:e1900009. [PMID: 31531927 DOI: 10.1002/pmic.201900009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 07/29/2019] [Indexed: 12/23/2022]
Abstract
Pseudorabies virus (PRV) has been widely used as a live trans-synaptic tracer for mapping neuronal circuits. Systematically identifying mature PRV virion proteomes and defining co-purified host proteins are necessary to fully understand the detailed mechanism underlying PRV transmission processes. Here, a PRV virion purification strategy based on sorting with flow cytometry is developed and the mature extracellular and intracellular PRV virion proteomes using LC coupled with MS/MS are characterized. In addition to viral proteins, a large number of host proteins are also identified, including proteins related to actin cytoskeletal dynamics and membrane protrusion. How many of these host proteins are true virion components are unknown and the majority of these may not be. Through functional analysis, it is found that IRSp53 and fascin are critical for the egress process and play a role in direct cell-cell transmission. Moreover, it is shown that CDC42 and Rac1 are also involved in the production of mature extracellular virions. The results suggest that the formation of the filopodia-like cytoskeleton and the rearrangement of the membrane, which are both associated with IRSp53 and fascin, may be important for the transmission of viruses used in neuronal tracing.
Collapse
Affiliation(s)
- Fei-Long Yu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Huan Miao
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Jinjin Xia
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Fan Jia
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Huadong Wang
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China
| | - Fuqiang Xu
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, China.,Center for Excellence in Brian Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Lin Guo
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Liu H, Cui J, Zhang Y, Niu M, Xue X, Yin H, Tang Y, Dai L, Dai F, Guo Y, Wu Y, Gao W. Mass spectrometry‐based proteomic analysis of FSCN1‐interacting proteins in laryngeal squamous cell carcinoma cells. IUBMB Life 2019; 71:1771-1784. [DOI: 10.1002/iub.2121] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/02/2019] [Indexed: 11/12/2022]
Affiliation(s)
- Hongliang Liu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck CancerShanxi Medical University Taiyuan China
- Otolaryngology Head & Neck Surgery Research InstituteShanxi Medical University Taiyuan China
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi ProvinceShanxi Medical University Taiyuan China
| | - Jiajia Cui
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck CancerShanxi Medical University Taiyuan China
- Otolaryngology Head & Neck Surgery Research InstituteShanxi Medical University Taiyuan China
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi ProvinceShanxi Medical University Taiyuan China
| | - Yuliang Zhang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck CancerShanxi Medical University Taiyuan China
- Otolaryngology Head & Neck Surgery Research InstituteShanxi Medical University Taiyuan China
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi ProvinceShanxi Medical University Taiyuan China
| | - Min Niu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck CancerShanxi Medical University Taiyuan China
- Otolaryngology Head & Neck Surgery Research InstituteShanxi Medical University Taiyuan China
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi ProvinceShanxi Medical University Taiyuan China
| | - Xuting Xue
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck CancerShanxi Medical University Taiyuan China
- Otolaryngology Head & Neck Surgery Research InstituteShanxi Medical University Taiyuan China
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi ProvinceShanxi Medical University Taiyuan China
| | - Hongyu Yin
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck CancerShanxi Medical University Taiyuan China
- Otolaryngology Head & Neck Surgery Research InstituteShanxi Medical University Taiyuan China
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi ProvinceShanxi Medical University Taiyuan China
| | - Yemei Tang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck CancerShanxi Medical University Taiyuan China
- Otolaryngology Head & Neck Surgery Research InstituteShanxi Medical University Taiyuan China
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi ProvinceShanxi Medical University Taiyuan China
| | - Li Dai
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck CancerShanxi Medical University Taiyuan China
- Otolaryngology Head & Neck Surgery Research InstituteShanxi Medical University Taiyuan China
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi ProvinceShanxi Medical University Taiyuan China
| | - Fengsheng Dai
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck CancerShanxi Medical University Taiyuan China
- Otolaryngology Head & Neck Surgery Research InstituteShanxi Medical University Taiyuan China
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi ProvinceShanxi Medical University Taiyuan China
| | - Yujia Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck CancerShanxi Medical University Taiyuan China
- Otolaryngology Head & Neck Surgery Research InstituteShanxi Medical University Taiyuan China
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi ProvinceShanxi Medical University Taiyuan China
| | - Yongyan Wu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck CancerShanxi Medical University Taiyuan China
- Otolaryngology Head & Neck Surgery Research InstituteShanxi Medical University Taiyuan China
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi ProvinceShanxi Medical University Taiyuan China
| | - Wei Gao
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck CancerShanxi Medical University Taiyuan China
- Otolaryngology Head & Neck Surgery Research InstituteShanxi Medical University Taiyuan China
- The Key Scientific and Technological Innovation Platform for Precision Diagnosis and Treatment of Head and Neck Cancer, Shanxi ProvinceShanxi Medical University Taiyuan China
| |
Collapse
|
29
|
Shiki A, Inoh Y, Yokawa S, Furuno T. Inhibition of degranulation in mast cells attached to a hydrogel through defective microtubule tracts. Exp Cell Res 2019; 381:248-255. [PMID: 31112735 DOI: 10.1016/j.yexcr.2019.05.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 01/07/2023]
Abstract
Mast cells (MCs) are important effectors of the immediate allergic response. MCs are distributed throughout various tissues and organs, and adhere to extracellular matrix (ECM) with broad stiffness in the body. Here we compared cellular responses following antigen stimulation in MCs on glass-base dishes with and without a hydrogel. We found that an antigen-induced increase in intracellular Ca2+ concentration was suppressed slightly in cells on hydrogel-coated dishes compared with those on non-coated dishes, whereas their subsequent degranulation was largely inhibited in cells adherent to the hydrogel. Focusing on focal adhesions (FAs), vinculin was distributed in a dot-like manner at the bottom of resting cells on non-coated dishes but not on hydrogel-coated dishes. According to antigen stimulation, phosphorylation of focal adhesion kinase and additive vinculin accumulation to FAs were promoted in cells on non-coated dishes, but were diminished on hydrogel-coated dishes. Moreover, microtubule reorganization and acetylation (which have important roles in MC degranulation) were also suppressed in activated MCs adherent to the hydrogel. These findings suggest that adhesion to a hydrogel led to failure of composition of functional FAs and microtubule tracts, which resulted in suppression of MC degranulation following antigen stimulation.
Collapse
Affiliation(s)
- Atsushi Shiki
- School of Pharmacy, Aichi Gakuin University, Nagoya, 464-8650, Japan
| | - Yoshikazu Inoh
- School of Pharmacy, Aichi Gakuin University, Nagoya, 464-8650, Japan
| | - Satoru Yokawa
- School of Pharmacy, Aichi Gakuin University, Nagoya, 464-8650, Japan
| | - Tadahide Furuno
- School of Pharmacy, Aichi Gakuin University, Nagoya, 464-8650, Japan.
| |
Collapse
|
30
|
The Cytoskeleton-A Complex Interacting Meshwork. Cells 2019; 8:cells8040362. [PMID: 31003495 PMCID: PMC6523135 DOI: 10.3390/cells8040362] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022] Open
Abstract
The cytoskeleton of animal cells is one of the most complicated and functionally versatile structures, involved in processes such as endocytosis, cell division, intra-cellular transport, motility, force transmission, reaction to external forces, adhesion and preservation, and adaptation of cell shape. These functions are mediated by three classical cytoskeletal filament types, as follows: Actin, microtubules, and intermediate filaments. The named filaments form a network that is highly structured and dynamic, responding to external and internal cues with a quick reorganization that is orchestrated on the time scale of minutes and has to be tightly regulated. Especially in brain tumors, the cytoskeleton plays an important role in spreading and migration of tumor cells. As the cytoskeletal organization and regulation is complex and many-faceted, this review aims to summarize the findings about cytoskeletal filament types, including substructures formed by them, such as lamellipodia, stress fibers, and interactions between intermediate filaments, microtubules and actin. Additionally, crucial regulatory aspects of the cytoskeletal filaments and the formed substructures are discussed and integrated into the concepts of cell motility. Even though little is known about the impact of cytoskeletal alterations on the progress of glioma, a final point discussed will be the impact of established cytoskeletal alterations in the cellular behavior and invasion of glioma.
Collapse
|
31
|
Barnawi R, Al-Khaldi S, Colak D, Tulbah A, Al-Tweigeri T, Fallatah M, Monies D, Ghebeh H, Al-Alwan M. β1 Integrin is essential for fascin-mediated breast cancer stem cell function and disease progression. Int J Cancer 2019; 145:830-841. [PMID: 30719702 PMCID: PMC6593770 DOI: 10.1002/ijc.32183] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 12/19/2018] [Accepted: 01/28/2019] [Indexed: 12/19/2022]
Abstract
Breast cancer remains the second cause of tumor‐related mortality in women worldwide mainly due to chemoresistance and metastasis. The chemoresistance and metastasis are attributed to a rare subpopulation with enriched stem‐like characteristics, thus called Cancer Stem Cells (CSCs). We have previously reported aberrant expression of the actin‐bundling protein (fascin) in breast cancer cells, which enhances their chemoresistance, metastasis and enriches CSC population. The intracellular mechanisms that link fascin with its downstream effectors are not fully elucidated. Here, loss and gain of function approaches in two different breast cancer models were used to understand how fascin promotes disease progression. Importantly, findings were aligned with expression data from actual breast cancer patients. Expression profiling of a large breast cancer dataset (TCGA, 530 patients) showed statistically significant correlation between fascin expression and a key adherence molecule, β1 integrin (ITGB1). In vitro manipulation of fascin expression in breast cancer cells exhibited its direct effect on ITGB1 expression. Fascin‐mediated regulation of ITGB1 was critical for several breast cancer cell functions including adhesion to different extracellular matrix, self‐renewability and chemoresistance. Importantly, there was a significant relationship between fascin and ITGB1 co‐expression and short disease‐free as well as overall survival in chemo‐treated breast cancer patients. This novel role of fascin effect on ITGB1 expression and its outcome on cell self‐renewability and chemoresistance strongly encourages for dual targeting of fascin‐ITGB1 axis as a therapeutic approach to halt breast cancer progression and eradicate it from the root. What's new? Residual cancer stem cells (CSCs) have the ability to regrow tumors and to metastasize to distant organs, resulting in disease relapse and increased cancer mortality. In breast cancer, CSC populations are enriched by aberrant expression of the actin‐bundling protein fascin, induction of which is also associated with chemoresistance and metastasis. In this study, fascin was found to upregulate β1 integrin (ITGB1) expression, an effect that proved critical to breast cancer cell adhesion and self‐renewal. Coexpression of fascin and ITGB1 was associated with decreased survival in chemotherapy‐treated breast cancer patients. The findings identify the fascin‐ITGB1 axis as a potential therapeutic target.
Collapse
Affiliation(s)
- Rayanah Barnawi
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Samiyah Al-Khaldi
- National Center for Stem Cells, Life Science and Environment Research Institute, King Abdulaziz City for Sciences and Technology, Riyadh, Saudi Arabia
| | - Dilek Colak
- Department of Biostatistics, Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Asma Tulbah
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Taher Al-Tweigeri
- Department of Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mohannad Fallatah
- National Center for Stem Cells, Life Science and Environment Research Institute, King Abdulaziz City for Sciences and Technology, Riyadh, Saudi Arabia
| | - Dorota Monies
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hazem Ghebeh
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Collage of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| | - Monther Al-Alwan
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Collage of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| |
Collapse
|
32
|
van Vuuren RJ, Botes M, Jurgens T, Joubert AM, van den Bout I. Novel sulphamoylated 2-methoxy estradiol derivatives inhibit breast cancer migration by disrupting microtubule turnover and organization. Cancer Cell Int 2019; 19:1. [PMID: 30622437 PMCID: PMC6317210 DOI: 10.1186/s12935-018-0719-4] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/21/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The estrogen metabolite 2-methoxyestradiol (2ME2) and a number of synthesised derivatives have been shown to bind to microtubules thereby arresting cancer cells in mitosis which leads to apoptosis. In interphase cells, microtubules play an important role in the delivery of proteins to subcellular locations including the focal adhesions. In fact, focal adhesion dynamics and cell migration are in part regulated by microtubules. We hypothesised that novel 2ME2 derivatives can alter cell migration by influencing microtubule dynamics in interphase cells. In this report we describe 2ME2 derivatives that display anti-migratory capabilities in a metastatic breast cancer cell line through their effects on the microtubule network resulting in altered focal adhesion signalling and RhoA activity. METHODS Cell migration was assayed using wound healing assays. To eliminate mitosis blockage and cell rounding as a confounding factor cell migration was also assessed in interphase blocked cells. Fluorescence confocal microscopy was used to visualise microtubule dynamics and actin cytoskeleton organisation while western blot analysis was performed to analyse focal adhesion signalling and RhoA activation. RESULTS 2ME2 derivatives, ESE-one and ESE-15-one, inhibited cell migration in cycling cells as expected but equally diminished migration in cells blocked in interphase. While no significant effects were observed on the actin cytoskeleton, focal adhesion kinase activity was increased while RhoA GTPase activity was inhibited after exposure to either compound. Microtubule stability was increased as evidenced by the increased length and number of detyrosinated microtubules while at the same time clear disorganisation of the normal radial microtubule organisation was observed including multiple foci. CONCLUSIONS ESE-15-one and ESE-one are potent migration inhibitors of metastatic breast cancer cells. This ability is coupled to alterations in focal adhesion signalling but more importantly is associated with severe disorganisation of microtubule dynamics and polarity. Therefore, these compounds may offer potential as anti-metastatic therapies.
Collapse
Affiliation(s)
| | - Mandie Botes
- Department of Physiology, University of Pretoria, Pretoria, 0084 South Africa
- Centre for Neuroendocrinology, University of Pretoria, Pretoria, 0084 South Africa
| | - Tamarin Jurgens
- Department of Physiology, University of Pretoria, Pretoria, 0084 South Africa
| | | | - Iman van den Bout
- Department of Physiology, University of Pretoria, Pretoria, 0084 South Africa
- Centre for Neuroendocrinology, University of Pretoria, Pretoria, 0084 South Africa
| |
Collapse
|
33
|
Chen T, Lien W, Cheng H, Kuan T, Sheu S, Wang C. Chloroquine inhibits human retina pigmented epithelial cell growth and microtubule nucleation by downregulating p150
glued. J Cell Physiol 2018; 234:10445-10457. [DOI: 10.1002/jcp.27712] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/15/2018] [Indexed: 01/11/2023]
Affiliation(s)
- Ting‐Yu Chen
- Department of Cell Biology and Anatomy College of Medicine, National Cheng Kung University Tainan Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University Tainan Taiwan
| | - Wei‐Chih Lien
- Department of Cell Biology and Anatomy College of Medicine, National Cheng Kung University Tainan Taiwan
- Department of Physical Medicine and Rehabilitation National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University Tainan Taiwan
| | - Hui‐Ling Cheng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University Tainan Taiwan
| | - Ta‐Shen Kuan
- Department of Physical Medicine and Rehabilitation National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University Tainan Taiwan
- Department of Physical Medicine and Rehabilitation College of Medicine, National Cheng Kung University Tainan Taiwan
| | - Shi‐Yuan Sheu
- School of Medicine, Chung Shan Medical University Taichung Taiwan
- Department of Integrated Chinese and Western Medicine Chung Shan Medical University Hospital Taichung Taiwan
| | - Chia‐Yih Wang
- Department of Cell Biology and Anatomy College of Medicine, National Cheng Kung University Tainan Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University Tainan Taiwan
| |
Collapse
|
34
|
Beghein E, Devriese D, Van Hoey E, Gettemans J. Cortactin and fascin-1 regulate extracellular vesicle release by controlling endosomal trafficking or invadopodia formation and function. Sci Rep 2018; 8:15606. [PMID: 30353022 PMCID: PMC6199335 DOI: 10.1038/s41598-018-33868-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/06/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer cell-derived extracellular vesicles (EVs) are increasingly being recognized as genuine invasive structures as they contribute to many aspects of invasion and metastasis. Unfortunately, the mechanisms underlying EV biogenesis or release are still poorly understood. Recent reports however indicate a role of the actin cytoskeleton in this process. In this study, we have exploited thoroughly characterized camelid nanobodies against actin binding proteins cortactin and fascin-1, a branched actin regulator and actin bundler, respectively, in order to assess their roles in EV biogenesis or release. Using this strategy, we demonstrate a role of the cortactin NTA and SH3 domains in EV release. Fascin-1 also regulates EV release, independently of its actin-bundling activity. We show a contribution of these protein domains in endosomal trafficking, a crucial step in EV biogenesis, and we confirm that EVs are preferentially released at invadopodia, the latter being actin-rich invasive cell protrusions in which cortactin and fascin-1 perform essential roles. Accordingly, EVs are enriched with invadopodial proteins such as the matrix metalloproteinase MT1-MMP and exert gelatinolytic activity. Based on our findings, we report that both cortactin and fascin-1 play key roles in EV release by regulating endosomal trafficking or invadopodia formation and function.
Collapse
Affiliation(s)
- Els Beghein
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Delphine Devriese
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Evy Van Hoey
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Jan Gettemans
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium.
| |
Collapse
|
35
|
Wei Z, Jiang W, Wang H, Li H, Tang B, Liu B, Jiang H, Sun X. The IL-6/STAT3 pathway regulates adhesion molecules and cytoskeleton of endothelial cells in thromboangiitis obliterans. Cell Signal 2018; 44:118-126. [PMID: 29339086 DOI: 10.1016/j.cellsig.2018.01.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/14/2017] [Accepted: 01/11/2018] [Indexed: 12/21/2022]
Abstract
Thromboangiitis obliterans (TAO) (also known as Buerger's disease) is an inflammatory vascular disease that predominantly affects small- and medium-sized blood vessels of extremities. Endothelial cells play critical roles in the initiation and progression of this disease, but the underlying mechanisms remain unclear. In the present study, we demonstrate that patients with TAO had significantly higher levels of interleukin-6 (IL-6), soluble intercellular adhesion molecule-1 (sICAM-1) and soluble vascular cell adhesion molecule-1 (sVCAM-1) in their plasmas, and the involved arterial tissues expressed higher levels of phosphorylated signal transducer and activator of transcription 3 (p-STAT3), ICAM-1 and VCAM-1. In exploring the molecular mechanisms with human aortic endothelial cells (HAECs), we found that recombinant IL-6 activated the STAT3 pathway, leading to the upregulation and overproduction of ICAM-1 and VCAM-1. RhoA (Ras homolog family member A), eNOS (endothelial nitric oxide synthase) and MMP-9 (matrix metalloproteinase-9) participated in this cellular signaling, and their interaction regulated the expression of ICAM-1 and VCAM-1. The activated STAT3 pathway by IL-6 also modulated the cytoskeleton of HAECs by regulating phosphorylation of focal adhesion kinase (FAK) and acetylation of α-tubulin through interplaying with RhoA. In summary, the present results indicate that activation of the IL-6/STAT3 pathway contributes to the pathogenesis of TAO by regulating cellular adhesion molecules and cytoskeleton of vascular endothelial cells, suggesting that targeting this pathway may provide a potential approach for the management of TAO.
Collapse
Affiliation(s)
- Zheng Wei
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Wenjing Jiang
- Key Laboratory of Hepatosplenic Surgery, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Hengzhen Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Hali Li
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Bo Tang
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Bing Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Hongchi Jiang
- Key Laboratory of Hepatosplenic Surgery, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xueying Sun
- Key Laboratory of Hepatosplenic Surgery, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
36
|
Pike R, Ortiz-Zapater E, Lumicisi B, Santis G, Parsons M. KIF22 coordinates CAR and EGFR dynamics to promote cancer cell proliferation. Sci Signal 2018; 11:11/515/eaaq1060. [PMID: 29382784 DOI: 10.1126/scisignal.aaq1060] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The coxsackievirus and adenovirus receptor (CAR) is a transmembrane receptor that plays a key role in cell-cell adhesion. CAR is found in normal epithelial cells and is increased in abundance in various human tumors, including lung carcinomas. We investigated the potential mechanisms by which CAR contributes to cancer cell growth and found that depletion of CAR in human lung cancer cells reduced anchorage-independent growth, epidermal growth factor (EGF)-dependent proliferation, and tumor growth in vivo. EGF induced the phosphorylation of CAR and its subsequent relocalization to cell junctions through the activation of the kinase PKCδ. EGF promoted the binding of CAR to the chromokinesin KIF22. KIF22-dependent regulation of microtubule dynamics led to delayed EGFR internalization, enhanced EGFR signaling, and coordination of CAR dynamics at cell-cell junctions. These data suggest a role for KIF22 in the coordination of membrane receptors and provide potential new therapeutic strategies to combat lung tumor growth.
Collapse
Affiliation(s)
- Rosemary Pike
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Elena Ortiz-Zapater
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK.,Division of Asthma, Allergy and Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Hospital Campus, London SE1 1UL, UK
| | - Brooke Lumicisi
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - George Santis
- Division of Asthma, Allergy and Lung Biology, King's College London, 5th Floor Tower Wing, Guy's Hospital Campus, London SE1 1UL, UK
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK.
| |
Collapse
|
37
|
Song G, Kwon CT, Kim SH, Shim Y, Lim C, Koh HJ, An G, Kang K, Paek NC. The Rice SPOTTED LEAF4 ( SPL4) Encodes a Plant Spastin That Inhibits ROS Accumulation in Leaf Development and Functions in Leaf Senescence. FRONTIERS IN PLANT SCIENCE 2018; 9:1925. [PMID: 30666263 PMCID: PMC6330318 DOI: 10.3389/fpls.2018.01925] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/11/2018] [Indexed: 05/21/2023]
Abstract
Lesion mimic mutants (LMMs) are usually controlled by single recessive mutations that cause the formation of necrotic lesions without pathogen invasion. These genetic defects are useful to reveal the regulatory mechanisms of defense-related programmed cell death in plants. Molecular evidence has been suggested that some of LMMs are closely associated with the regulation of leaf senescence in rice (Oryza sativa). Here, we characterized the mutation underlying spotted leaf4 (spl4), which results in lesion formation and also affects leaf senescence in rice. Map-based cloning revealed that the γ ray-induced spl4-1 mutant has a single base substitution in the splicing site of the SPL4 locus, resulting in a 13-bp deletion within the encoded microtubule-interacting-and-transport (MIT) spastin protein containing an AAA-type ATPase domain. The T-DNA insertion spl4-2 mutant exhibited spontaneous lesions similar to those of the spl4-1 mutant, confirming that SPL4 is responsible for the LMM phenotype. In addition, both spl4 mutants exhibited delayed leaf yellowing during dark-induced or natural senescence. Western blot analysis of spl4 mutant leaves suggested possible roles for SPL4 in the degradation of photosynthetic proteins. Punctate signals of SPL4-fused fluorescent proteins were detected in the cytoplasm, similar to the cellular localization of animal spastin. Based on these findings, we propose that SPL4 is a plant spastin that is involved in multiple aspects of leaf development, including senescence.
Collapse
Affiliation(s)
- Giha Song
- Department of Plant Science, Plant Genomics and Breeding Institute, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Choon-Tak Kwon
- Department of Plant Science, Plant Genomics and Breeding Institute, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Suk-Hwan Kim
- Department of Plant Science, Plant Genomics and Breeding Institute, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Yejin Shim
- Department of Plant Science, Plant Genomics and Breeding Institute, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Chaemyeong Lim
- Department of Plant Science, Plant Genomics and Breeding Institute, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Hee-Jong Koh
- Department of Plant Science, Plant Genomics and Breeding Institute, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Gynheung An
- Department of Plant Molecular Systems Biotechnology, Crop Biotech Institute, Kyung Hee University, Seoul, South Korea
| | - Kiyoon Kang
- Department of Plant Science, Plant Genomics and Breeding Institute, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
- *Correspondence: Kiyoon Kang, Nam-Chon Paek,
| | - Nam-Chon Paek
- Department of Plant Science, Plant Genomics and Breeding Institute, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
- *Correspondence: Kiyoon Kang, Nam-Chon Paek,
| |
Collapse
|
38
|
Zheng HC, Zhao S. The meta and bioinformatics analysis of fascin expression in gastric cancer: a potential marker for aggressiveness and worse prognosis. Oncotarget 2017; 8:105574-105583. [PMID: 29285273 PMCID: PMC5739660 DOI: 10.18632/oncotarget.22325] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/08/2017] [Indexed: 01/09/2023] Open
Abstract
Fascin is a FSCN1-encoded actin bundling protein, and positively associated with proliferation, migration and metastasis of malignancies. Here, we performed a systematic meta and bioinformatics analysis through multiple online databases up to March 14, 2017. We found up-regulated fascin expression in gastric cancer, compared with normal mucosa (p<0.05). Fascin expression was positively with lymph node metastasis, TNM staging and worse prognosis of gastric cancer (p<0.05). According to bioinformatics database, FSCN1 mRNA expression was higher in gastric cancer than normal tissues (p<0.05). According to Kaplan-Meier plotter, we found that a higher FSCN1 expression was negatively correlated with overall and progression-free survival rates of all cancer patients, even stratified by aggressive parameters (p<0.05). These findings indicated that fascin expression might be employed as a potential marker to indicate gastric carcinogenesis and subsequent progression, even prognosis.
Collapse
Affiliation(s)
- Hua-Chuan Zheng
- Department of Experimental Oncology and Animal Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Shuang Zhao
- Department of Experimental Oncology and Animal Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
39
|
Strong fascin expression promotes metastasis independent of its F-actin bundling activity. Oncotarget 2017; 8:110077-110091. [PMID: 29299131 PMCID: PMC5746366 DOI: 10.18632/oncotarget.22249] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 09/27/2017] [Indexed: 11/25/2022] Open
Abstract
High expression of the actin bundling protein Fascin increases the malignancy of tumor cells. Here we show that fascin expression is up-regulated in more malignant sub-cell lines of MDA-MB-231 cells as compared to parental cells. Since also parental MDA-MB-231 cells exhibit high fascin levels, increased fascin expression was termed as “hyperexpression”. To examine the effect of fascin hyperexpression, fascin was hyperexpressed in parental MDA-MB-231 cells and metastasis was analyzed in NOD scid gamma (NSG) mice. In addition, the effect of fascin mutants with inactive or constitutively active actin bundling activity was examined. Unexpectedly, we found that hyperexpression of both, wildtype (wt) and mutant fascin strongly increased metastasis in vivo, showing that the effect of fascin hyperexpression did not depend on its actin bundling activity. Cellular assays revealed that hyperexpression of wt and mutant fascin increased adhesion of MDA-MB-231 cells while transmigration and proliferation were not affected. Since it has been shown that fascin controls adhesion by directly interacting with microtubules (MTs), we analyzed if fascin hyperexpression affects MT dynamics. We found that at high concentrations fascin significantly increased MT dynamics in cells and in cell-free approaches. In summary our data show that strong expression of fascin in breast cancer cells increases metastasis independent of its actin bundling activity. Thus, it seems that the mechanism of fascin-stimulated metastasis depends on its concentration.
Collapse
|
40
|
Teruel É, Rizkallah G, Journo C, Dutartre H. [The actin cytoskeleton goes retroviral]. Med Sci (Paris) 2017; 33:839-842. [PMID: 28994374 DOI: 10.1051/medsci/20173310009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Élodie Teruel
- Centre international de recherche en infectiologie, équipe oncogenèse rétrovirale, Inserm U1111 - université Claude Bernard Lyon 1, CNRS, UMR5308, École normale supérieure de Lyon, université de Lyon, 46, allée d'Italie, F-69007, Lyon, France ; équipe labellisée « Ligue nationale contre le cancer »
| | - Gerges Rizkallah
- Centre international de recherche en infectiologie, équipe oncogenèse rétrovirale, Inserm U1111 - université Claude Bernard Lyon 1, CNRS, UMR5308, École normale supérieure de Lyon, université de Lyon, 46, allée d'Italie, F-69007, Lyon, France ; équipe labellisée « Ligue nationale contre le cancer »
| | - Chloé Journo
- Centre international de recherche en infectiologie, équipe oncogenèse rétrovirale, Inserm U1111 - université Claude Bernard Lyon 1, CNRS, UMR5308, École normale supérieure de Lyon, université de Lyon, 46, allée d'Italie, F-69007, Lyon, France ; équipe labellisée « Ligue nationale contre le cancer »
| | - Hélène Dutartre
- Centre international de recherche en infectiologie, équipe oncogenèse rétrovirale, Inserm U1111 - université Claude Bernard Lyon 1, CNRS, UMR5308, École normale supérieure de Lyon, université de Lyon, 46, allée d'Italie, F-69007, Lyon, France ; équipe labellisée « Ligue nationale contre le cancer »
| |
Collapse
|
41
|
Kliewe F, Scharf C, Rogge H, Darm K, Lindenmeyer MT, Amann K, Cohen CD, Endlich K, Endlich N. Studying the role of fascin-1 in mechanically stressed podocytes. Sci Rep 2017; 7:9916. [PMID: 28855604 PMCID: PMC5577297 DOI: 10.1038/s41598-017-10116-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 08/04/2017] [Indexed: 11/09/2022] Open
Abstract
Glomerular hypertension causes glomerulosclerosis via the loss of podocytes, which are challenged by increased mechanical load. We have demonstrated that podocytes are mechanosensitive. However, the response of podocytes to mechanical stretching remains incompletely understood. Here we demonstrate that the actin-bundling protein fascin-1 plays an important role in podocytes that are exposed to mechanical stress. Immunofluorescence staining revealed colocalization of fascin-1 and nephrin in mouse kidney sections. In cultured mouse podocytes fascin-1 was localized along actin fibers and filopodia in stretched and unstretched podocytes. The mRNA and protein levels of fascin-1 were not affected by mechanical stress. By Western blot and 2D-gelelectrophoresis we observed that phospho-fascin-1 was significantly downregulated after mechanical stretching. It is known that phosphorylation at serine 39 (S39) regulates the bundling activity of fascin-1, e.g. required for filopodia formation. Podocytes expressing wild type GFP-fascin-1 and non-phosphorylatable GFP-fascin-1-S39A showed marked filopodia formation, being absent in podocytes expressing phosphomimetic GFP-fascin-1-S39D. Finally, the immunofluorescence signal of phosphorylated fascin-1 was strongly reduced in glomeruli of patients with diabetic nephropathy compared to healthy controls. In summary, mechanical stress dephosphorylates fascin-1 in podocytes in vitro and in vivo thereby fascin-1 may play an important role in the adaptation of podocytes to mechanical forces.
Collapse
Affiliation(s)
- Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Christian Scharf
- Department of Ear, Nose and Throat Diseases, University Medicine Greifswald, Greifswald, Germany
| | - Henrik Rogge
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Katrin Darm
- Department of Ear, Nose and Throat Diseases, University Medicine Greifswald, Greifswald, Germany
| | - Maja T Lindenmeyer
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Kerstin Amann
- Department of Nephropathology, University Medicine Erlangen, Erlangen, Germany
| | - Clemens D Cohen
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Karlhans Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
42
|
Rodrigues PC, Sawazaki-Calone I, Ervolino de Oliveira C, Soares Macedo CC, Dourado MR, Cervigne NK, Miguel MC, Ferreira do Carmo A, Lambert DW, Graner E, Daniela da Silva S, Alaoui-Jamali MA, Paes Leme AF, Salo TA, Coletta RD. Fascin promotes migration and invasion and is a prognostic marker for oral squamous cell carcinoma. Oncotarget 2017; 8:74736-74754. [PMID: 29088820 PMCID: PMC5650375 DOI: 10.18632/oncotarget.20360] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/12/2017] [Indexed: 01/06/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) prognosis is related to clinical stage and histological grade. However, this stratification needs to be refined. We conducted a comparative proteome study in microdissected samples from normal oral mucosa and OSCC to identify biomarkers for malignancy. Fascin and plectin were identified as differently expressed and both are implicated in several malignancies, but the clinical impacts of aberrant fascin and plectin expression in OSCCs remains largely unknown. Immunohistochemistry and real-time quantitative PCR were carried out in ex vivo OSCC samples and cell lines. A loss-of-function strategy using shRNA targeting fascin was employed to investigate in vitro and in vivo the fascin role on oral tumorigenesis. Transfections of microRNA mimics were performed to determine whether the fascin overexpression is regulated by miR-138 and miR-145. We found that fascin and plectin are frequently upregulated in OSCC samples and cell lines, but only fascin overexpression is an independent unfavorable prognostic indicator of disease-specific survival. In combination with advanced T stage, high fascin level is also an independent factor of disease-free survival. Knockdown of fascin in OSCC cells promoted cell adhesion and inhibited migration, invasion and EMT, and forced expression of miR-138 in OSCC cells significantly decreased the expression of fascin. In addition, fascin downregulation leads to reduced filopodia formation and decrease on paxillin expression. The subcutaneous xenograft model showed that tumors formed in the presence of low levels of fascin were significantly smaller compared to those formed with high fascin levels. Collectively, our findings suggest that fascin expression correlates with disease progression and may serve as a prognostic marker and therapeutic target for patients with OSCC.
Collapse
Affiliation(s)
- Priscila Campioni Rodrigues
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil.,Unit of Cancer Research and Translational Medicine, Faculty of Medicine and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Iris Sawazaki-Calone
- Oral Pathology and Oral Medicine, Dentistry School, Western Paraná State University, Cascavel, PR, Brazil
| | | | | | - Mauricio Rocha Dourado
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil.,Unit of Cancer Research and Translational Medicine, Faculty of Medicine and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Nilva K Cervigne
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil.,Current/Present address: Clinical Department, Faculty of Medicine of Jundiai, Jundiai, SP, Brazil
| | - Marcia Costa Miguel
- Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Andreia Ferreira do Carmo
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil.,Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Daniel W Lambert
- Integrated Biosciences, School of Clinical Dentistry and Sheffield Cancer Centre, University of Sheffield, Sheffield, United Kingdom
| | - Edgard Graner
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil
| | - Sabrina Daniela da Silva
- Departments of Medicine, Oncology, Pharmacology and Therapeutics, Segal Cancer Centre and Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, Montreal, Quebec, Canada.,Otolaryngology-Head and Neck Surgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Moulay A Alaoui-Jamali
- Departments of Medicine, Oncology, Pharmacology and Therapeutics, Segal Cancer Centre and Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, Montreal, Quebec, Canada.,Otolaryngology-Head and Neck Surgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | | | - Tuula A Salo
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil.,Unit of Cancer Research and Translational Medicine, Faculty of Medicine and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland.,Institute of Oral and Maxillofacial Disease, University of Helsinki, and HUSLAB, Department of Pathology, Helsinki University Hospital, Helsinki, Finland
| | - Ricardo D Coletta
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil
| |
Collapse
|
43
|
Routray S, Kheur S, Chougule HM, Mohanty N, Dash R. Establishing Fascin over-expression as a strategic regulator of neoplastic aggression and lymph node metastasis in oral squamous cell carcinoma tumor microenvironment. Ann Diagn Pathol 2017; 30:36-41. [PMID: 28965626 DOI: 10.1016/j.anndiagpath.2017.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 05/15/2017] [Accepted: 05/22/2017] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Oral squamous cell carcinoma (OSCC) has an aggressive behaviour with high incidence of nodal metastasis, even in the early stages, leading to poor prognosis. For progression and metastasis of cancers, the dominant element considered is cell motility. Fascin, an actin-binding protein has emerged as a protein of general importance for a diverse set of cell protrusions with functions in cell adhesion, cell interactions, and cell migration. The role of Fascin in various carcinomas, including aggressive behaviour in OSCC has been documented, but its role as a key regulator in lymph nodes metastasis is yet to be validated. AIM This study was piloted to evaluate and correlate Fascin expression in OSCC lymph nodes and understand the role of Fascin in contemptuous Lesional tissue, as a predictor of survival. A retrospective study designed with 40 archival OSCC cases was included as sample, 20 each of both lymph node metastasis +ve (Group 1) and -ve (Group 2) groups. All the participants were smokeless tobacco user and had tumor located at gingivo-buccal complex. RESULTS We established that Fascin over-expression in lymph nodes were significantly associated with clinico-histopathological parameters like staging (p=0.01), tumor size (cT) (p=0.03) and differentiation; and furthermore it was highly significant in correlation to nodal status (cN) (*p≤0.001). Fascin over-expression in lymph node metastasis positive cases correlated with that of Fascin expression in contemptuous Lesional tissue signifying its role in promoting aggressive progression and metastasis. This association was found to be statistically significant (p value=0.05). Overall Survival Analysis of both lymph node metastasis +ve and -ve groups assessed by Kaplan-Meier analysis (taking death and recurrence into consideration) showed patients with high Fascin expression (in lymph node and Lesional tissue) had shorter overall survival than patients who had no to weak Fascin expression. CONCLUSION Our findings thereby establish Fascin expression as a regulator of metastasis in OSCC tumor microenvironment and predictor of survival.
Collapse
Affiliation(s)
- Samapika Routray
- Department of Oral Pathology and Microbiology, Institute of Dental Sciences, Siksha 'O'Anusandhan University, Bhubaneswar 751030, India; Department of Dental Surgery, All India Institute of Medical Sciences, Bhubaneswar-751019.
| | - Supriya Kheur
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College & Hospital, D. Y. Patil Vidyapeeth, Pimri, Pune 411081, India
| | - Hemlata M Chougule
- Department of Oral Pathology and Microbiology, Dr. D. Y. Patil Dental College & Hospital, D. Y. Patil Vidyapeeth, Pimri, Pune 411081, India
| | - Neeta Mohanty
- Department of Oral Pathology and Microbiology, Institute of Dental Sciences, Siksha 'O'Anusandhan University, Bhubaneswar 751030, India
| | - Rupesh Dash
- Scientist-D, Institute of Life Sciences, Bhubaneswar 751023, India
| |
Collapse
|
44
|
Tang DD, Gerlach BD. The roles and regulation of the actin cytoskeleton, intermediate filaments and microtubules in smooth muscle cell migration. Respir Res 2017; 18:54. [PMID: 28390425 PMCID: PMC5385055 DOI: 10.1186/s12931-017-0544-7] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 04/05/2017] [Indexed: 02/06/2023] Open
Abstract
Smooth muscle cell migration has been implicated in the development of respiratory and cardiovascular systems; and airway/vascular remodeling. Cell migration is a polarized cellular process involving a protrusive cell front and a retracting trailing rear. There are three cytoskeletal systems in mammalian cells: the actin cytoskeleton, the intermediate filament network, and microtubules; all of which regulate all or part of the migrated process. The dynamic actin cytoskeleton spatially and temporally regulates protrusion, adhesions, contraction, and retraction from the cell front to the rear. c-Abl tyrosine kinase plays a critical role in regulating actin dynamics and migration of airway smooth muscle cells and nonmuscle cells. Recent studies suggest that intermediate filaments undergo reorganization during migration, which coordinates focal adhesion dynamics, cell contraction, and nucleus rigidity. In particular, vimentin intermediate filaments undergo phosphorylation and reorientation in smooth muscle cells, which may regulate cell contraction and focal adhesion assembly/disassembly. Motile cells are characterized by a front-rear polarization of the microtubule framework, which regulates all essential processes leading to cell migration through its role in cell mechanics, intracellular trafficking, and signaling. This review recapitulates our current knowledge how the three cytoskeletal systems spatially and temporally modulate the migratory properties of cells. We also summarize the potential role of migration-associated biomolecules in lung and vascular diseases.
Collapse
Affiliation(s)
- Dale D Tang
- Department of Molecular and Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, MC-8, Albany, NY, 12208, USA.
| | - Brennan D Gerlach
- Department of Molecular and Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, MC-8, Albany, NY, 12208, USA
| |
Collapse
|
45
|
The Tax-Inducible Actin-Bundling Protein Fascin Is Crucial for Release and Cell-to-Cell Transmission of Human T-Cell Leukemia Virus Type 1 (HTLV-1). PLoS Pathog 2016; 12:e1005916. [PMID: 27776189 PMCID: PMC5077169 DOI: 10.1371/journal.ppat.1005916] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/05/2016] [Indexed: 01/07/2023] Open
Abstract
The delta-retrovirus Human T-cell leukemia virus type 1 (HTLV-1) preferentially infects CD4+ T-cells via cell-to-cell transmission. Viruses are transmitted by polarized budding and by transfer of viral biofilms at the virological synapse (VS). Formation of the VS requires the viral Tax protein and polarization of the host cytoskeleton, however, molecular mechanisms of HTLV-1 cell-to-cell transmission remain incompletely understood. Recently, we could show Tax-dependent upregulation of the actin-bundling protein Fascin (FSCN-1) in HTLV-1-infected T-cells. Here, we report that Fascin contributes to HTLV-1 transmission. Using single-cycle replication-dependent HTLV-1 reporter vectors, we found that repression of endogenous Fascin by short hairpin RNAs and by Fascin-specific nanobodies impaired gag p19 release and cell-to-cell transmission in 293T cells. In Jurkat T-cells, Tax-induced Fascin expression enhanced virus release and Fascin-dependently augmented cell-to-cell transmission to Raji/CD4+ B-cells. Repression of Fascin in HTLV-1-infected T-cells diminished virus release and gag p19 transfer to co-cultured T-cells. Spotting the mechanism, flow cytometry and automatic image analysis showed that Tax-induced T-cell conjugate formation occurred Fascin-independently. However, adhesion of HTLV-1-infected MT-2 cells in co-culture with Jurkat T-cells was reduced upon knockdown of Fascin, suggesting that Fascin contributes to dissemination of infected T-cells. Imaging of chronically infected MS-9 T-cells in co-culture with Jurkat T-cells revealed that Fascin’s localization at tight cell-cell contacts is accompanied by gag polarization suggesting that Fascin directly affects the distribution of gag to budding sites, and therefore, indirectly viral transmission. In detail, we found gag clusters that are interspersed with Fascin clusters, suggesting that Fascin makes room for gag in viral biofilms. Moreover, we observed short, Fascin-containing membrane extensions surrounding gag clusters and clutching uninfected T-cells. Finally, we detected Fascin and gag in long-distance cellular protrusions. Taken together, we show for the first time that HTLV-1 usurps the host cell factor Fascin to foster virus release and cell-to-cell transmission. Human T-cell leukemia virus type 1 (HTLV-1) is the only human retrovirus causing cancer and is transmitted via breast feeding, sexual intercourse, and cell-containing blood products. Efficient infection of CD4+ T-cells occurs via polarized budding of virions or via cell surface transfer of viral biofilms at a tight, specialized cell-cell contact, the virological synapse (VS). The viral protein Tax and polarization of the host cell cytoskeleton are crucial for formation of the VS, however, only little is known about the link between Tax and remodeling of the cytoskeleton to foster viral spread. The actin-bundling protein Fascin has evolved as a therapeutic target in several types of cancer. Here, we show that Fascin is also crucial for release and transmission of the tumorvirus HTLV-1. Since Fascin is a transcriptional target gene of Tax in T-cells, our work provides a link between Tax’s activity and virus transmission. Visualization of cell-cell contacts between infected and uninfected T-cells suggests a role of Fascin in viral transmission potentially by facilitating the transport of viral proteins to budding sites. Thus, Fascin is not only crucial for metastasis of tumors, but also for transmission of HTLV-1 and is a new cellular target to counteract HTLV-1.
Collapse
|
46
|
Byron A, Frame MC. Adhesion protein networks reveal functions proximal and distal to cell-matrix contacts. Curr Opin Cell Biol 2016; 39:93-100. [PMID: 26930633 PMCID: PMC5094910 DOI: 10.1016/j.ceb.2016.02.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 02/09/2016] [Accepted: 02/09/2016] [Indexed: 12/15/2022]
Abstract
Cell adhesion to the extracellular matrix is generally mediated by integrin receptors, which bind to intracellular adhesion proteins that form multi-molecular scaffolding and signalling complexes. The networks of proteins, and their interactions, are dynamic, mechanosensitive and extremely complex. Recent efforts to characterise adhesions using a variety of technologies, including imaging, proteomics and bioinformatics, have provided new insights into their composition, organisation and how they are regulated, and have also begun to reveal unexpected roles for so-called adhesion proteins in other cellular compartments (for example, the nucleus or centrosomes) in diseases such as cancer. We believe this is opening a new chapter on understanding the wider functions of adhesion proteins, both proximal and distal to cell-matrix contacts.
Collapse
Affiliation(s)
- Adam Byron
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom.
| | - Margaret C Frame
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|