1
|
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: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
2
|
Alampally H, Chandrashekar C, Rodrigues G, Carnelio S. Fascin in tooth germs: an immunohistochemical analysis. J Histotechnol 2018. [DOI: 10.1080/01478885.2017.1404286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Harishanker Alampally
- Department of Oral and Maxillofacial Pathology, Manipal College of Dental Sciences, Manipal University, Manipal, India
| | - Chethana Chandrashekar
- Department of Oral and Maxillofacial Pathology, Manipal College of Dental Sciences, Manipal University, Manipal, India
| | - Gabriel Rodrigues
- Department of General Surgery, Kasturba Medical College, Manipal University, Manipal, India
| | - Sunitha Carnelio
- Department of Oral and Maxillofacial Pathology, Manipal College of Dental Sciences, Manipal University, Manipal, India
| |
Collapse
|
3
|
Biochemistry of Drebrin and Its Binding to Actin Filaments. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1006:37-47. [DOI: 10.1007/978-4-431-56550-5_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
4
|
Wang X, Nichols L, Grunz-Borgmann EA, Sun Z, Meininger GA, Domeier TL, Baines CP, Parrish AR. Fascin2 regulates cisplatin-induced apoptosis in NRK-52E cells. Toxicol Lett 2016; 266:56-64. [PMID: 27989596 DOI: 10.1016/j.toxlet.2016.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 11/01/2016] [Accepted: 11/27/2016] [Indexed: 01/09/2023]
Abstract
Previous studies have shown that the aging kidney has a marked loss of α(E)-catenin in proximal tubular epithelium. α-Catenin, a key regulator of the actin cytoskeleton, interacts with a variety of actin-binding proteins. Cisplatin-induced loss of fascin2, an actin bundling protein, was observed in cells with a stable knockdown of α(E)-catenin (C2 cells), as well as in aging (24 mon), but not young (4 mon), kidney. Fascin2 co-localized with α-catenin and the actin cytoskeleton in NRK-52E cells. Knockdown of fascin2 increased the susceptibility of tubular epithelial cells to cisplatin-induced injury. Overexpression of fascin2 in C2 cells restored actin stress fibers and attenuated the increased sensitivity of C2 cells to cisplatin-induced apoptosis. Interestingly, fascin2 overexpression attenuated cisplatin-induced mitochondrial dysfunction and oxidative stress in C2 cells. These data demonstrate that fascin2, a putative target of α(E)-catenin, may play important role in preventing cisplatin-induced acute kidney injury.
Collapse
Affiliation(s)
- Xinhui Wang
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - LaNita Nichols
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Elizabeth A Grunz-Borgmann
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Zhe Sun
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States; Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Gerald A Meininger
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States; Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Christopher P Baines
- Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO 65212, United States; Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Alan R Parrish
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States.
| |
Collapse
|
5
|
Dierks S, von Hardenberg S, Schmidt T, Bremmer F, Burfeind P, Kaulfuß S. Leupaxin stimulates adhesion and migration of prostate cancer cells through modulation of the phosphorylation status of the actin-binding protein caldesmon. Oncotarget 2016; 6:13591-606. [PMID: 26079947 PMCID: PMC4537036 DOI: 10.18632/oncotarget.3792] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/18/2015] [Indexed: 12/15/2022] Open
Abstract
The focal adhesion protein leupaxin (LPXN) is overexpressed in a subset of prostate cancers (PCa) and is involved in the progression of PCa. In the present study, we analyzed the LPXN-mediated adhesive and cytoskeletal changes during PCa progression. We identified an interaction between the actin-binding protein caldesmon (CaD) and LPXN and this interaction is increased during PCa cell migration. Furthermore, knockdown of LPXN did not affect CaD expression but reduced CaD phosphorylation. This is known to destabilize the affinity of CaD to F-actin, leading to dynamic cell structures that enable cell motility. Thus, downregulation of CaD increased migration and invasion of PCa cells. To identify the kinase responsible for the LPXN-mediated phosphorylation of CaD, we used data from an antibody array, which showed decreased expression of TGF-beta-activated kinase 1 (TAK1) after LPXN knockdown in PC-3 PCa cells. Subsequent analyses of the downstream kinases revealed the extracellular signal-regulated kinase (ERK) as an interaction partner of LPXN that facilitates CaD phosphorylation during LPXN-mediated PCa cell migration. In conclusion, we demonstrate that LPXN directly influences cytoskeletal dynamics via interaction with the actin-binding protein CaD and regulates CaD phosphorylation by recruiting ERK to highly dynamic structures within PCa cells.
Collapse
Affiliation(s)
- Sascha Dierks
- Institute of Human Genetics, University Medical Center Göttingen, Germany
| | - Sandra von Hardenberg
- Institute of Human Genetics, University Medical Center Göttingen, Germany.,Center of Pharmacology and Toxicology, Hannover Medical School, Germany
| | - Thomas Schmidt
- Institute of Human Genetics, University Medical Center Göttingen, Germany.,Department of Anatomy, University of Witten/Herdecke, Witten, Germany
| | - Felix Bremmer
- Institute of Pathology, University Medical Center Göttingen, Germany
| | - Peter Burfeind
- Institute of Human Genetics, University Medical Center Göttingen, Germany
| | - Silke Kaulfuß
- Institute of Human Genetics, University Medical Center Göttingen, Germany
| |
Collapse
|
6
|
Miao Y, Han X, Zheng L, Xie Y, Mu Y, Yates JR, Drubin DG. Fimbrin phosphorylation by metaphase Cdk1 regulates actin cable dynamics in budding yeast. Nat Commun 2016; 7:11265. [PMID: 27068241 PMCID: PMC4832064 DOI: 10.1038/ncomms11265] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 03/07/2016] [Indexed: 12/15/2022] Open
Abstract
Actin cables, composed of actin filament bundles nucleated by formins, mediate intracellular transport for cell polarity establishment and maintenance. We previously observed that metaphase cells preferentially promote actin cable assembly through cyclin-dependent kinase 1 (Cdk1) activity. However, the relevant metaphase Cdk1 targets were not known. Here we show that the highly conserved actin filament crosslinking protein fimbrin is a critical Cdk1 target for actin cable assembly regulation in budding yeast. Fimbrin is specifically phosphorylated on threonine 103 by the metaphase cyclin–Cdk1 complex, in vivo and in vitro. On the basis of conformational simulations, we suggest that this phosphorylation stabilizes fimbrin's N-terminal domain, and modulates actin filament binding to regulate actin cable assembly and stability in cells. Overall, this work identifies fimbrin as a key target for cell cycle regulation of actin cable assembly in budding yeast, and suggests an underlying mechanism. Metaphase cells preferentially promote actin cable assembly through cyclin-dependent kinase 1 (Cdk1) activity. Here the authors identify fimbrin as one of the main metaphase Cdk1 targets for cell cycle regulation of actin cable assembly in budding yeast.
Collapse
Affiliation(s)
- Yansong Miao
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3202, USA.,School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.,School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Xuemei Han
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Liangzhen Zheng
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Ying Xie
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - David G Drubin
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3202, USA
| |
Collapse
|
7
|
Wu X, Cao MP, Shen YY, Chu KP, Tao WB, Song WT, Liu LP, Wang XH, Zheng YF, Chen SD, Zeng QL, Xia RH. Weak power frequency magnetic field acting similarly to EGF stimulation, induces acute activations of the EGFR sensitive actin cytoskeleton motility in human amniotic cells. PLoS One 2014; 9:e87626. [PMID: 24505297 PMCID: PMC3914819 DOI: 10.1371/journal.pone.0087626] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 12/16/2013] [Indexed: 12/30/2022] Open
Abstract
In this article, we have examined the motility-related effects of weak power frequency magnetic fields (MFs) on the epidermal growth factor receptor (EGFR)-sensitive motility mechanism, including the F-actin cytoskeleton, growth of invasive protrusions and the levels of signal molecules in human amniotic epithelial (FL) cells. Without extracellular EGF stimulation, the field stimulated a large growth of new protrusions, especially filopodia and lamellipodia, an increased population of vinculin-associated focal adhesions. And, an obvious reduction of stress fiber content in cell centers was found, corresponding to larger cell surface areas and decreased efficiency of actin assembly of FL cells in vitro, which was associated with a decrease in overall F-actin content and special distributions. These effects were also associated with changes in protein content or distribution patterns of the EGFR downstream motility-related signaling molecules. All of these effects are similar to those following epidermal growth factor (EGF) stimulation of the cells and are time dependent. These results suggest that power frequency MF exposure acutely affects the migration/motility-related actin cytoskeleton reorganization that is regulated by the EGFR-cytoskeleton signaling pathway. Therefore, upon the MF exposure, cells are likely altered to be ready to transfer into a state of migration in response to the stimuli.
Collapse
Affiliation(s)
- Xia Wu
- Physics Department, East China Normal University, Shanghai, China
- State Key Laboratory of Precision Spectroscopy, East China Normal University, Shanghai, China
| | - Mei-Ping Cao
- Physics Department, East China Normal University, Shanghai, China
| | - Yun-Yun Shen
- Bioelectromagnetics Laboratory, Zhejiang University, Hangzhou, China
| | - Ke-Ping Chu
- Physics Department, East China Normal University, Shanghai, China
| | - Wu-Bin Tao
- Physics Department, East China Normal University, Shanghai, China
| | - Wei-Tao Song
- Physics Department, East China Normal University, Shanghai, China
| | - Li-Ping Liu
- School of Life Sciences, Fudan University, Shanghai, China
| | - Xiang-Hui Wang
- Physics Department, East China Normal University, Shanghai, China
| | - Yu-Fang Zheng
- School of Life Sciences, Fudan University, Shanghai, China
| | - Shu-De Chen
- Physics Department, East China Normal University, Shanghai, China
| | - Qun-Li Zeng
- Bioelectromagnetics Laboratory, Zhejiang University, Hangzhou, China
| | - Ruo-Hong Xia
- Physics Department, East China Normal University, Shanghai, China
- State Key Laboratory of Precision Spectroscopy, East China Normal University, Shanghai, China
| |
Collapse
|
8
|
Lehman W, Li XE, Orzechowski M, Fischer S. The structural dynamics of α-tropomyosin on F-actin shape the overlap complex between adjacent tropomyosin molecules. Arch Biochem Biophys 2013; 552-553:68-73. [PMID: 24071513 DOI: 10.1016/j.abb.2013.09.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 08/31/2013] [Accepted: 09/13/2013] [Indexed: 12/15/2022]
Abstract
Coiled-coil tropomyosin, localized on actin filaments in virtually all eukaryotic cells, serves as a gatekeeper regulating access of the motor protein myosin and other actin-binding proteins onto the thin filament surface. Tropomyosin's modular pseudo-repeating pattern of approximately 39 amino acid residues is designed to allow binding of the coiled-coil to successive actin subunits along thin filaments. Even though different tropomyosin isoforms contain varying numbers of repeat modules, the pseudo-repeat length, in all cases, matches that of a single actin subunit. Thus, the seven pseudo-repeats of 42nm long muscle tropomyosin bind to seven successive actin subunits along thin filaments, while simultaneously bending into a super-helical conformation that is preshaped to the actin filament helix. In order to form a continuous cable on thin filaments that is free of gaps, adjacent tropomyosin molecules polymerize head-to-tail by means of a short (∼9 residue) overlap. Several laboratories have engineered peptides to mimic the N- and C-terminal tropomyosin association and to characterize the overlap structure. All overlapping domains examined show a compact N-terminal coiled-coil inserting into a partially opened C-terminal partner, where the opposing coiled-coils at the overlap junction face each other at up to ∼90° twist angles. Here, Molecular Dynamics (MD) simulations were carried out to determine constraints on the formation of the tropomyosin overlap complex and to assess the amount of twisting exhibited by full-length tropomyosin when bound to actin. With the exception of the last 20-40 C- and N-terminal residues, we find that the average tropomyosin structure closely resembles a "canonical" model proposed in the classic work of McLachlan and Stewart, displaying perfectly symmetrical supercoil geometry matching the F-actin helix with an integral number of coiled-coil turns, a coiled-coil helical pitch of 137Å, a superhelical pitch of 770Å, and no localized pseudo-rotation. Over the middle 70% of tropomyosin, the average twisting of the coiled-coil deviates only by 10° from the canonical model and the torsional freedom is very small (std. dev. of 7°). This small degree of twisting cannot yield the orthogonal N- and C-termini configuration observed experimentally. In marked contrast, considerable coiled-coil unfolding, splaying and twisting at N- and C-terminal ends is observed, providing the conformational plasticity needed for head-to-tail nexus formation.
Collapse
Affiliation(s)
- William Lehman
- Department of Physiology and Biophysics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA.
| | - Xiaochuan Edward Li
- Department of Physiology and Biophysics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA; Computational Biochemistry Group, IWR, University of Heidelberg, Im Neuenheimer Feld 368, Heidelberg D69120, Germany
| | - Marek Orzechowski
- Department of Physiology and Biophysics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA; Computational Biochemistry Group, IWR, University of Heidelberg, Im Neuenheimer Feld 368, Heidelberg D69120, Germany
| | - Stefan Fischer
- Computational Biochemistry Group, IWR, University of Heidelberg, Im Neuenheimer Feld 368, Heidelberg D69120, Germany.
| |
Collapse
|
9
|
Groen CM, Spracklen AJ, Fagan TN, Tootle TL. Drosophila Fascin is a novel downstream target of prostaglandin signaling during actin remodeling. Mol Biol Cell 2012; 23:4567-78. [PMID: 23051736 PMCID: PMC3510018 DOI: 10.1091/mbc.e12-05-0417] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Prostaglandins (PGs) regulate the actin cytoskeleton. However, their mechanisms of action are unknown. Use of Drosophila oogenesis—specifically nurse cell dumping—as a model shows that PGs regulate the actin bundler Fascin to control parallel actin filament bundle formation and cortical actin integrity. Although prostaglandins (PGs)—lipid signals produced downstream of cyclooxygenase (COX) enzymes—regulate actin cytoskeletal dynamics, their mechanisms of action are unknown. We previously established Drosophila oogenesis, in particular nurse cell dumping, as a new model to determine how PGs regulate actin remodeling. PGs, and thus the Drosophila COX-like enzyme Pxt, are required for both the parallel actin filament bundle formation and the cortical actin strengthening required for dumping. Here we provide the first link between Fascin (Drosophila Singed, Sn), an actin-bundling protein, and PGs. Loss of either pxt or fascin results in similar actin defects. Fascin interacts, both pharmacologically and genetically, with PGs, as reduced Fascin levels enhance the effects of COX inhibition and synergize with reduced Pxt levels to cause both parallel bundle and cortical actin defects. Conversely, overexpression of Fascin in the germline suppresses the effects of COX inhibition and genetic loss of Pxt. These data lead to the conclusion that PGs regulate Fascin to control actin remodeling. This novel interaction has implications beyond Drosophila, as both PGs and Fascin-1, in mammalian systems, contribute to cancer cell migration and invasion.
Collapse
Affiliation(s)
- Christopher M Groen
- Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
10
|
Nagano K, Akpan A, Warnasuriya G, Corless S, Totty N, Yang A, Stein R, Zvelebil M, Stensballe A, Burlingame A, Waterfield M, Cramer R, Timms JF, Naaby-Hansen S. Functional proteomic analysis of long-term growth factor stimulation and receptor tyrosine kinase coactivation in Swiss 3T3 fibroblasts. Mol Cell Proteomics 2012; 11:1690-708. [PMID: 22956732 DOI: 10.1074/mcp.m112.019778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In Swiss 3T3 fibroblasts, long-term stimulation with PDGF, but not insulin-like growth factor 1 (IGF-1) or EGF, results in the establishment of an elongated migratory phenotype, characterized by the formation of retractile dendritic protrusions and absence of actin stress fibers and focal adhesion complexes. To identify receptor tyrosine kinase-specific reorganization of the Swiss 3T3 proteome during phenotypic differentiation, we compared changes in the pattern of protein synthesis and phosphorylation during long-term exposure to PDGF, IGF-1, EGF, and their combinations using 2DE-based proteomics after (35)S- and (33)P-metabolic labeling. One hundred and five differentially regulated proteins were identified by mass spectrometry and some of these extensively validated. PDGF stimulation produced the highest overall rate of protein synthesis at any given time and induced the most sustained phospho-signaling. Simultaneous activation with two or three of the growth factors revealed both synergistic and antagonistic effects on protein synthesis and expression levels with PDGF showing dominance over both IGF-1 and EGF in generating distinct proteome compositions. Using signaling pathway inhibitors, PI3K was identified as an early site for signal diversification, with sustained activity of the PI3K/AKT pathway critical for regulating late protein synthesis and phosphorylation of target proteins and required for maintaining the PDGF-dependent motile phenotype. Several proteins were identified with novel PI3K/Akt-dependent synthesis and phosphorylations including eEF2, PRS7, RACK-1, acidic calponin, NAP1L1, Hsp73, and fascin. The data also reveal induction/suppression of key F-actin and actomyosin regulators and chaperonins that enable PDGFR to direct the assembly of a motile cytoskeleton, despite simultaneous antagonistic signaling activities. Together, the study demonstrates that long-term exposure to different growth factors results in receptor tyrosine kinase-specific regulation of relatively small subproteomes, and implies that the strength and longevity of receptor tyrosine kinase-specific signals are critical in defining the composition and functional activity of the resulting proteome.
Collapse
Affiliation(s)
- Kohji Nagano
- Discovery Research Department, Chugai Pharmaceutical Co. Ltd., Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Lehman W, Morgan KG. Structure and dynamics of the actin-based smooth muscle contractile and cytoskeletal apparatus. J Muscle Res Cell Motil 2012; 33:461-9. [PMID: 22311558 DOI: 10.1007/s10974-012-9283-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 01/16/2012] [Indexed: 01/04/2023]
Abstract
The thin filaments of differentiated smooth muscle cells are composed of actin and tropomyosin isoforms and numerous ancillary actin-binding proteins that assemble together into distinct thin filament classes. These different filament classes are segregated in smooth muscle cells into structurally and functionally separated contractile and cytoskeletal cellular domains. Typically, thin filaments in smooth muscle cells have been considered to be relatively stable structures like those in striated cells. However, recent efforts have shown that smooth muscle thin filaments indeed are dynamic and that remodeling of the actin cytoskeleton, in particular, regulates smooth muscle function. Thus, the cytoskeleton of differentiated smooth muscle cells appears to function midway between that of less dynamic striated muscle cells and that of very plastic proliferative cells such as fibroblasts. Michael and Kate Bárány keenly followed and participated in some of these studies, consistent with their broad interest in actin function and smooth muscle mechanisms. As a way of honoring the memory of these two pioneer members of the muscle research community, we review data on distribution and remodeling of thin filaments in smooth muscle cells, one of the many research topics that intrigued them.
Collapse
Affiliation(s)
- William Lehman
- Department of Physiology and Biophysics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA.
| | | |
Collapse
|
12
|
Rao JN, Rivera-Santiago R, Li XE, Lehman W, Dominguez R. Structural analysis of smooth muscle tropomyosin α and β isoforms. J Biol Chem 2011; 287:3165-74. [PMID: 22119916 DOI: 10.1074/jbc.m111.307330] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A large number of tropomyosin (Tm) isoforms function as gatekeepers of the actin filament, controlling the spatiotemporal access of actin-binding proteins to specialized actin networks. Residues ∼40-80 vary significantly among Tm isoforms, but the impact of sequence variation on Tm structure and interactions with actin is poorly understood, because structural studies have focused on skeletal muscle Tmα. We describe structures of N-terminal fragments of smooth muscle Tmα and Tmβ (sm-Tmα and sm-Tmβ). The 2.0-Å structure of sm-Tmα81 (81-aa) resembles that of skeletal Tmα, displaying a similar super-helical twist matching the contours of the actin filament. The 1.8-Å structure of sm-Tmα98 (98-aa) unexpectedly reveals an antiparallel coiled coil, with the two chains staggered by only 4 amino acids and displaying hydrophobic core interactions similar to those of the parallel dimer. In contrast, the 2.5-Å structure of sm-Tmβ98, containing Gly-Ala-Ser at the N terminus to mimic acetylation, reveals a parallel coiled coil. None of the structures contains coiled-coil stabilizing elements, favoring the formation of head-to-tail overlap complexes in four of five crystallographically independent parallel dimers. These complexes show similarly arranged 4-helix bundles stabilized by hydrophobic interactions, but the extent of the overlap varies between sm-Tmβ98 and sm-Tmα81 from 2 to 3 helical turns. The formation of overlap complexes thus appears to be an intrinsic property of the Tm coiled coil, with the specific nature of hydrophobic contacts determining the extent of the overlap. Overall, the results suggest that sequence variation among Tm isoforms has a limited effect on actin binding but could determine its gatekeeper function.
Collapse
Affiliation(s)
- Jampani Nageswara Rao
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
13
|
Nie S, Kee Y, Bronner-Fraser M. Caldesmon regulates actin dynamics to influence cranial neural crest migration in Xenopus. Mol Biol Cell 2011; 22:3355-65. [PMID: 21795398 PMCID: PMC3172261 DOI: 10.1091/mbc.e11-02-0165] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
A nonmuscle caldesmon (CaD) is highly expressed in premigratory and migrating Xenopus cranial neural crest cells. A loss-of-function approach shows that CaD is critical for neural crest migration. The results further suggest that CaD influences cell morphology and motility by modulating actin dynamics in neural crest cells. Caldesmon (CaD) is an important actin modulator that associates with actin filaments to regulate cell morphology and motility. Although extensively studied in cultured cells, there is little functional information regarding the role of CaD in migrating cells in vivo. Here we show that nonmuscle CaD is highly expressed in both premigratory and migrating cranial neural crest cells of Xenopus embryos. Depletion of CaD with antisense morpholino oligonucleotides causes cranial neural crest cells to migrate a significantly shorter distance, prevents their segregation into distinct migratory streams, and later results in severe defects in cartilage formation. Demonstrating specificity, these effects are rescued by adding back exogenous CaD. Interestingly, CaD proteins with mutations in the Ca2+-calmodulin–binding sites or ErK/Cdk1 phosphorylation sites fail to rescue the knockdown phenotypes, whereas mutation of the PAK phosphorylation site is able to rescue them. Analysis of neural crest explants reveals that CaD is required for the dynamic arrangements of actin and, thus, for cell shape changes and process formation. Taken together, these results suggest that the actin-modulating activity of CaD may underlie its critical function and is regulated by distinct signaling pathways during normal neural crest migration.
Collapse
Affiliation(s)
- Shuyi Nie
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | |
Collapse
|
14
|
Abstract
Viral infection converts the normal functions of a cell to optimize viral replication and virion production. One striking observation of this conversion is the reconfiguration and reorganization of cellular actin, affecting every stage of the viral life cycle, from entry through assembly to egress. The extent and degree of cytoskeletal reorganization varies among different viral infections, suggesting the evolution of myriad viral strategies. In this Review, we describe how the interaction of viral proteins with the cell modulates the structure and function of the actin cytoskeleton to initiate, sustain and spread infections. The molecular biology of such interactions continues to engage virologists in their quest to understand viral replication and informs cell biologists about the role of the cytoskeleton in the uninfected cell.
Collapse
Affiliation(s)
- Matthew P Taylor
- Department of Molecular Biology, Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544, USA
| | | | | |
Collapse
|
15
|
Hashimoto Y, Kim DJ, Adams JC. The roles of fascins in health and disease. J Pathol 2011; 224:289-300. [DOI: 10.1002/path.2894] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 03/02/2011] [Accepted: 03/04/2011] [Indexed: 02/06/2023]
|
16
|
Lees JG, Bach CTT, O'Neill GM. Interior decoration: tropomyosin in actin dynamics and cell migration. Cell Adh Migr 2011; 5:181-6. [PMID: 21173575 DOI: 10.4161/cam.5.2.14438] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cell migration and invasion requires the precise temporal and spatial orchestration of a variety of biological processes. Filaments of polymerized actin are critical players in these diverse processes, including the regulation of cell anchorage points (both cell-cell and cell-extracellular matrix), the uptake and delivery of molecules via endocytic pathways and the generation of force for both membrane protrusion and retraction. How the actin filaments are specialized for each of these discrete functions is yet to be comprehensively elucidated. The cytoskeletal tropomyosins are a family of actin associating proteins that form head-to-tail polymers which lay in the major groove of polymerized actin filaments. In the present review we summarize the emerging isoform-specific functions of tropomyosins in cell migration and invasion and discuss their potential roles in the specialization of actin filaments for the diverse cellular processes that together regulate cell migration and invasion.
Collapse
Affiliation(s)
- Justin G Lees
- Children's Cancer Research Unit, Kids Research Institute, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | | | | |
Collapse
|
17
|
Mayanagi T, Sobue K. Diversification of caldesmon-linked actin cytoskeleton in cell motility. Cell Adh Migr 2011; 5:150-9. [PMID: 21350330 DOI: 10.4161/cam.5.2.14398] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The actin cytoskeleton plays a key role in regulating cell motility. Caldesmon (CaD) is an actin-linked regulatory protein found in smooth muscle and non-muscle cells that is conserved among a variety of vertebrates. It binds and stabilizes actin filaments, as well as regulating actin-myosin interaction in a calcium (Ca2+)/calmodulin (CaM)- and/or phosphorylation-dependent manner. CaD function is regulated qualitatively by Ca2+/CaM and by its phosphorylation state and quantitatively at the mRNA level, by three different transcriptional regulation of the CALD1 gene. CaD has numerous functions in cell motility, such as migration, invasion, and proliferation, exerted via the reorganization of the actin cytoskeleton. Here we will outline recent findings regarding CaD's structural features and functions.
Collapse
Affiliation(s)
- Taira Mayanagi
- Department of Neuroscience, Osaka University Graduate School of Medicine, Osaka, Japan
| | | |
Collapse
|
18
|
Yamakita Y, Matsumura F, Lipscomb MW, Chou PC, Werlen G, Burkhardt JK, Yamashiro S. Fascin1 promotes cell migration of mature dendritic cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:2850-9. [PMID: 21263068 DOI: 10.4049/jimmunol.1001667] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Dendritic cells (DCs) play central roles in innate and adaptive immunity. Upon maturation, DCs assemble numerous veil-like membrane protrusions, disassemble podosomes, and travel from the peripheral tissues to lymph nodes to present Ags to T cells. These alterations in morphology and motility are closely linked to the primary function of DCs, Ag presentation. However, it is unclear how and what cytoskeletal proteins control maturation-associated alterations, in particular, the change in cell migration. Fascin1, an actin-bundling protein, is specifically and greatly induced upon maturation, suggesting a unique role for fascin1 in mature DCs. To determine the physiological roles of fascin1, we characterized bone marrow-derived, mature DCs from fascin1 knockout mice. We found that fascin1 is critical for cell migration: fascin1-null DCs exhibit severely decreased membrane protrusive activity. Importantly, fascin1-null DCs have lower chemotactic activity toward CCL19 (a chemokine for mature DCs) in vitro, and in vivo, Langerhans cells show reduced emigration into draining lymph nodes. Morphologically, fascin1-null mature DCs are flatter and fail to disassemble podosomes, a specialized structure for cell-matrix adhesion. Expression of exogenous fascin1 in fascin1-null DCs rescues the defects in membrane protrusive activity, as well as in podosome disassembly. These results indicate that fascin1 positively regulates migration of mature DCs into lymph nodes, most likely by increasing dynamics of membrane protrusions, as well as by disassembling podosomes.
Collapse
Affiliation(s)
- Yoshihiko Yamakita
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Upregulated fascin1 in non-small cell lung cancer promotes the migration and invasiveness, but not proliferation. Cancer Lett 2010; 290:238-47. [DOI: 10.1016/j.canlet.2009.09.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Revised: 09/17/2009] [Accepted: 09/17/2009] [Indexed: 01/07/2023]
|
20
|
Wang CLA, Coluccio LM. New insights into the regulation of the actin cytoskeleton by tropomyosin. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2010; 281:91-128. [PMID: 20460184 PMCID: PMC2923581 DOI: 10.1016/s1937-6448(10)81003-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The actin cytoskeleton is regulated by a variety of actin-binding proteins including those constituting the tropomyosin family. Tropomyosins are coiled-coil dimers that bind along the length of actin filaments. In muscles, tropomyosin regulates the interaction of actin-containing thin filaments with myosin-containing thick filaments to allow contraction. In nonmuscle cells where multiple tropomyosin isoforms are expressed, tropomyosins participate in a number of cellular events involving the cytoskeleton. This chapter reviews the current state of the literature regarding tropomyosin structure and function and discusses the evidence that tropomyosins play a role in regulating actin assembly.
Collapse
|
21
|
The Shape and Flexibility of Tropomyosin Coiled Coils: Implications for Actin Filament Assembly and Regulation. J Mol Biol 2010; 395:327-39. [DOI: 10.1016/j.jmb.2009.10.060] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 10/19/2009] [Accepted: 10/27/2009] [Indexed: 12/18/2022]
|
22
|
Lin JJ, Li Y, Eppinga RD, Wang Q, Jin J. Chapter 1 Roles of Caldesmon in Cell Motility and Actin Cytoskeleton Remodeling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2009; 274:1-68. [DOI: 10.1016/s1937-6448(08)02001-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
23
|
Gestalt-binding of tropomyosin to actin filaments. J Muscle Res Cell Motil 2008; 29:213-9. [PMID: 19116763 DOI: 10.1007/s10974-008-9157-6] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Accepted: 12/01/2008] [Indexed: 01/26/2023]
Abstract
We argue that the overall behavior of tropomyosin on F-actin cannot be easily discerned by examining thin filaments reduced to their smallest interacting units. In isolation, the individual interactions of actin and tropomyosin, by themselves, are too weak to account for the specificity of the system. Instead the association of tropomyosin on actin can only be fully explained after considering the concerted action of the entire acto-tropomyosin system. We propose that the low K ( a ) describing tropomyosin:actin interaction, when taken together with the form-fitting complementarity of tropomyosin strands on F-actin and the tendency for tropomyosin to polymerize end-to-end, make possible unique thin filament functions both locally and at higher levels of filament organization.
Collapse
|
24
|
Kudryashova TV, Rutkevich PN, Shevelev AY, Vlasik TN, Vorotnikov AV. Caldesmon affects actin organization at the leading edge and inhibits cell migration. Biophysics (Nagoya-shi) 2008. [DOI: 10.1134/s0006350908060110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
25
|
Wang CLA. Caldesmon and the regulation of cytoskeletal functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 644:250-72. [PMID: 19209827 PMCID: PMC2975104 DOI: 10.1007/978-0-387-85766-4_19] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Caldesmon (CaD) is an extraordinary actin-binding protein, because in addition to actin, it also bindsmyosin, calmodulin and tropomyosin. As a component of the smoothmuscle and nonmuscle contractile apparatus CaD inhibits the actomyosin ATPase activity and its inhibitory action is modulated by both Ca2+ and phosphorylation. The multiplicity of binding partners and diverse biochemical properties suggest CaD is a potent and versatile regulatory protein both in contractility and cell motility. However, after decades ofinvestigation in numerous laboratories, hard evidence is still lacking to unequivocally identify its in vivo functions, although indirect evidence is mounting to support an important role in connection with the actin cytoskeleton. This chapter reviews the highlights of the past findings and summarizes the current views on this protein, with emphasis of its interaction with tropomyosin.
Collapse
Affiliation(s)
- C L Albert Wang
- Boston Biomedical Research Institute, 64 Grove Street, Watertown, MA 02472, USA.
| |
Collapse
|
26
|
Schüler H, Peti W. Structure-function analysis of the filamentous actin binding domain of the neuronal scaffolding protein spinophilin. FEBS J 2008; 275:59-68. [PMID: 18028445 PMCID: PMC2927859 DOI: 10.1111/j.1742-4658.2007.06171.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Spinophilin, a neuronal scaffolding protein, is essential for synaptic transmission, and functions to target protein phosphatase-1 to distinct subcellular locations in dendritic spines. It is vital for the regulation of dendritic spine formation and motility, and functions by regulating glutamatergic receptors and binding to filamentous actin. To investigate its role in regulating actin cytoskeletal structure, we initiated structural studies of the actin binding domain of spinophilin. We demonstrate that the spinophilin actin binding domain is intrinsically unstructured, and that, with increasing C-terminal length, the domain shows augmented secondary structure content. Further characterization confirmed the previously known crosslinking activity and uncovered a novel filamentous actin pointed-end capping activity. Both of these functions seem to be fully contained within residues 1-154 of spinophilin.
Collapse
Affiliation(s)
- Herwig Schüler
- Max Delbrück Center for Molecular Medicine, Berlin-Buch, Germany.
| | | |
Collapse
|
27
|
Hashimoto Y, Parsons M, Adams JC. Dual actin-bundling and protein kinase C-binding activities of fascin regulate carcinoma cell migration downstream of Rac and contribute to metastasis. Mol Biol Cell 2007; 18:4591-602. [PMID: 17855511 PMCID: PMC2043557 DOI: 10.1091/mbc.e07-02-0157] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2007] [Revised: 08/29/2007] [Accepted: 08/31/2007] [Indexed: 01/08/2023] Open
Abstract
Recurrence of carcinomas due to cells that migrate away from the primary tumor is a major problem in cancer treatment. Immunohistochemical analyses of human carcinomas have consistently correlated up-regulation of the actin-bundling protein fascin with a clinically aggressive phenotype and poor prognosis. To understand the functional and mechanistic contributions of fascin, we undertook inducible short hairpin RNA (shRNA) knockdown of fascin in human colon carcinoma cells derived from an aggressive primary tumor. Fascin-depletion led to decreased numbers of filopodia and altered morphology of cell protrusions, decreased Rac-dependent migration on laminin, decreased turnover of focal adhesions, and, in vivo, decreased xenograft tumor development and metastasis. cDNA rescue of fascin shRNA-knockdown cells with wild-type green fluorescent protein-fascin or fascins mutated at the protein kinase C (PKC) phosphorylation site revealed that both the actin-bundling and active PKC-binding activities of fascin are required for the organization of filopodial protrusions, Rac-dependent migration, and tumor metastasis. Thus, fascin contributes to carcinoma migration and metastasis through dual pathways that impact on multiple subcellular structures needed for cell migration.
Collapse
Affiliation(s)
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, King's College London, Guy's Campus, London SE1 1UL, United Kingdom
| | - Josephine C. Adams
- *Department of Cell Biology, Lerner Research Institute, and
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, The Cleveland Clinic, Cleveland, OH 44195; and
| |
Collapse
|
28
|
Tropomyosins as interpreters of the signalling environment to regulate the local cytoskeleton. Semin Cancer Biol 2007; 18:35-44. [PMID: 17942320 DOI: 10.1016/j.semcancer.2007.08.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Accepted: 08/28/2007] [Indexed: 01/11/2023]
Abstract
A key regulator of cell morphology is the actin cytoskeleton and it has long been appreciated that the cytoskeleton is characteristically altered in cancer. Actin is organized into polymeric structures with distinct dynamics which in turn participate in a wide variety of cell processes including adhesion, migration, cell division and apoptosis. Despite displaying an altered actin cytoskeleton, transformed cells retain--and in many cases increase--their ability to adhere, move, divide and respond to apoptotic stimuli. Thus cancer cells maintain responsive actin cytoskeletons. Actin dynamics are regulated by numerous actin-binding proteins and chief among these are the tropomyosins which are core components of the microfilament. Recent advances in genomic and proteomic profiling confirm that Tm expression profiles are profoundly changed in transformed cells. It is therefore timely to review the role of Tms in the regulation of actin dynamics that pertain to crucial phenotypic changes in cancer. In this review we discuss how actin filaments containing different Tm isoforms respond to the activation of cell signalling pathways and consider the implications of this for cancer progression and therapy.
Collapse
|
29
|
Ishikawa R, Katoh K, Takahashi A, Xie C, Oseki K, Watanabe M, Igarashi M, Nakamura A, Kohama K. Drebrin attenuates the interaction between actin and myosin-V. Biochem Biophys Res Commun 2007; 359:398-401. [PMID: 17543276 DOI: 10.1016/j.bbrc.2007.05.123] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Accepted: 05/20/2007] [Indexed: 01/22/2023]
Abstract
Drebrin-A is an actin-binding protein localized in the dendritic spines of mature neurons, and has been suggested to affect spine morphology [K. Hayashi, T. Shirao, Change in the shape of dendritic spines caused by overexpression of drebrin in cultured cortical neurons, J. Neurosci. 19 (1999) 3918-3925]. However, no biochemical analysis of drebrin-A has yet been reported. In this study, we purified drebrin-A using a bacterial expression system, and characterized it in vitro. Drebrin-A bound to actin filaments with a stoichiometry of one drebrin molecule to 5-6 actin molecules. Furthermore, drebrin-A decreased the Mg-ATPase activity of myosin V. In vitro motility assay revealed that the attachment of F-actin to glass surface coated with myosin-V was decreased by drebrin-A, but once F-actin attached to the surface, the sliding speed of F-actin was unaffected by the presence of drebrin A. These findings suggest that drebrin-A may affect spine dynamics, vesicle transport, and other myosin-V-driven motility in neurons through attenuating the interaction between actin and myosin-V.
Collapse
Affiliation(s)
- Ryoki Ishikawa
- Department of Molecular and Cellular Pharmacology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Han IS, Seo TB, Kim KH, Yoon JH, Yoon SJ, Namgung U. Cdc2-mediated Schwann cell migration during peripheral nerve regeneration. J Cell Sci 2007; 120:246-55. [PMID: 17200138 DOI: 10.1242/jcs.03322] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Schwann cell migration facilitates peripheral nerve regeneration after injury. We have recently found increased activation of Cdc2 kinase in regenerating sciatic nerves. Here we show that Cdc2 phosphorylation of caldesmon regulates Schwann cell migration and nerve regeneration. A robust but transient increase in Cdc2 expression was found in cultured Schwann cells prepared from the sciatic nerve in rats that had undergone crush injury for 7 days. These `injury-preconditioned' Schwann cells exhibited enhanced migration compared with non-preconditioned control cells and treatment with the cdk inhibitor roscovitine prevented cell migration. After transduction with recombinant Cdc2 DNA adenoviral vectors, Schwann cells were implanted into sciatic nerves; those expressing wild-type Cdc2 migrated further in the distal direction than those expressing dominant-negative Cdc2. We identified caldesmon as a downstream substrate of Cdc2 in Schwann cells and its phosphorylation by Cdc2 changed its subcellular localization. Overexpression of dominant-negative caldesmon significantly counteracted the migration effect caused by Cdc2. Finally, neurite outgrowth of cultured DRG sensory neurons, facilitated by co-culture with injury-preconditioned Schwann cells, was suppressed by roscovitine treatment. The results indicate that activation of the Cdc2-caldesmon pathway is necessary for Schwann cell migration and suggest a role for this pathway in peripheral axonal growth.
Collapse
Affiliation(s)
- In Sun Han
- Department of Oriental Medicine, Daejeon University, Daejeon 300-716, Korea
| | | | | | | | | | | |
Collapse
|
31
|
Eppinga RD, Li Y, Lin JLC, Lin JJC. Tropomyosin and caldesmon regulate cytokinesis speed and membrane stability during cell division. Arch Biochem Biophys 2006; 456:161-74. [PMID: 16854366 DOI: 10.1016/j.abb.2006.06.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Revised: 06/14/2006] [Accepted: 06/15/2006] [Indexed: 01/11/2023]
Abstract
The contractile ring and the cell cortex generate force to divide the cell while maintaining symmetrical shape. This requires temporal and spatial regulation of the actin cytoskeleton at these areas. We force-expressed misregulated versions of actin-binding proteins, tropomyosin and caldesmon, into cells and analyzed their effects on cell division. Cells expressing proteins that increase actomyosin ATPase, such as human tropomyosin chimera (hTM5/3), significantly speed up division, whereas cells expressing proteins that inhibit actomyosin, such as caldesmon mutants defective in Ca(2+)/calmodulin binding (CaD39-AB) and in cdk1 phosphorylation sites (CaD39-6F), divide slowly. hTM5 and hTM5/3-expressing cells lift one daughter cell off the substrate and twist. Furthermore, CaD39-AB- and CaD39-6F-expressing cells are sensitive to hypotonic swelling and show severe blebbing during division, whereas hTM5/3-expressing cells are resistant to hypotonic swelling and produce membrane bulges. These results support a model where Ca(2+)/calmodulin and cdk1 dynamically control caldesmon inhibition of tropomyosin-activated actomyosin to regulate division speed and to suppress membrane blebs.
Collapse
Affiliation(s)
- Robbin D Eppinga
- Department of Biological Sciences, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
32
|
Vignjevic D, Kojima SI, Aratyn Y, Danciu O, Svitkina T, Borisy GG. Role of fascin in filopodial protrusion. ACTA ACUST UNITED AC 2006; 174:863-75. [PMID: 16966425 PMCID: PMC2064340 DOI: 10.1083/jcb.200603013] [Citation(s) in RCA: 399] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In this study, the mechanisms of actin-bundling in filopodia were examined. Analysis of cellular localization of known actin cross-linking proteins in mouse melanoma B16F1 cells revealed that fascin was specifically localized along the entire length of all filopodia, whereas other actin cross-linkers were not. RNA interference of fascin reduced the number of filopodia, and remaining filopodia had abnormal morphology with wavy and loosely bundled actin organization. Dephosphorylation of serine 39 likely determined cellular filopodia frequency. The constitutively active fascin mutant S39A increased the number and length of filopodia, whereas the inactive fascin mutant S39E reduced filopodia frequency. Fluorescence recovery after photobleaching of GFP-tagged wild-type and S39A fascin showed that dephosphorylated fascin underwent rapid cycles of association to and dissociation from actin filaments in filopodia, with t1/2 < 10 s. We propose that fascin is a key specific actin cross-linker, providing stiffness for filopodial bundles, and that its dynamic behavior allows for efficient coordination between elongation and bundling of filopodial actin filaments.
Collapse
Affiliation(s)
- Danijela Vignjevic
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Grosheva I, Vittitow JL, Goichberg P, Gabelt BT, Kaufman PL, Borrás T, Geiger B, Bershadsky AD. Caldesmon effects on the actin cytoskeleton and cell adhesion in cultured HTM cells. Exp Eye Res 2006; 82:945-58. [PMID: 16679125 DOI: 10.1016/j.exer.2006.01.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2005] [Revised: 01/17/2006] [Accepted: 01/17/2006] [Indexed: 01/27/2023]
Abstract
Caldesmon is a multifunctional ubiquitous regulator of the actin cytoskeleton, which can affect both actomyosin contractility and actin polymerization. Previous studies showed that caldesmon over-expression in cultured fibroblasts produces effects that resemble those of chemical inhibitors of cellular contractility. Since these inhibitors (H-7, Y-27632, etc.) have been shown to lower intraocular pressure and increase outflow facility from the anterior chamber of the eye, we proposed that caldesmon might be used for gene therapy of glaucoma. In the present study we examined the effects of expression of adenovirus-delivered rat non-muscle caldesmon fused with green fluorescent protein (AdCaldGFP) on the actin cytoskeleton and matrix adhesions in cultured human trabecular meshwork (HTM) cells. In addition, we assessed the effect of caldesmon on the stability of cell-cell junctions in kidney epithelial MDCK cells. Cultured HTM cells demonstrate a well-developed actin cytoskeleton, comprising mainly arrays of parallel actomyosin bundles (stress fibers). Lamellipodial protrusions containing dense actin networks are also observed. Cell-matrix adhesions are dominated by focal adhesions (FAs) associated with the ends of the stress fibers, focal complexes in lamellipodia, and fibrillar adhesions in the central part of the spread cells. Treatment of HTM cells with AdCaldGFP resulted in dose-dependent morphological changes within 24-48 hr post-infection. Cells expressing moderate levels of caldesmon exhibited straight bundles containing actin and myosin II, which were considerably shorter than those in control cells. Short filament bundles in caldesmon over-expressing cells formed arrays consisting of triangular actin structures with small vinculin-positive FAs at their vertices. In addition, the fraction of cells displaying large lamellipodia increased. About 40-50% of the population of caldesmon-expressing cells demonstrated high levels of GFP-caldesmon expression and severe changes in the actin cytoskeleton, manifested by the disappearance of stress fibers and the formation of curved actin- and myosin-containing bundles. These bundles formed together a dynamic network consisting of pulsating loops filling the entire cytoplasm. Addition of thapsigargin, which increases intracellular Ca++ concentration, resulted in a straightening of the curved bundles. Another type of novel actin structures induced by caldesmon over-expression were highly dynamic circular waves that propagated over the affected cells with a velocity about 10 microm min. In cells with disrupted stress fibers, vinculin-containing FAs and tensin-rich fibrillar adhesions had also essentially vanished. However, phosphotyrosine-positive focal complexes were still prominent throughout the lamellipodia of these cells. Over-expression of caldesmon in MDCK cells reduced, in a dose dependent manner, the beta-catenin content at cell-cell adherens junctions and in some cases led to physical disruption of adherens junctions. Thus, caldesmon over-expression induces unique reorganization of the actin cytoskeleton in affected cells, accompanied by disruption of focal and fibrillar cell-matrix adhesions, and destabilization of cell-cell adherens junctions. Inducing such changes in the contractility and actin cytoskeleton of HTM cells in glaucomatous eyes in vivo could produce a therapeutically useful increase in outflow facility.
Collapse
Affiliation(s)
- Inna Grosheva
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Eves R, Webb BA, Zhou S, Mak AS. Caldesmon is an integral component of podosomes in smooth muscle cells. J Cell Sci 2006; 119:1691-702. [PMID: 16595550 DOI: 10.1242/jcs.02881] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Podosomes are highly dynamic actin-based structures commonly found in motile and invasive cells such as macrophages, osteoclasts and vascular smooth muscle cells. Here, we have investigated the role of caldesmon, an actin-binding protein, in the formation of podosomes in aortic smooth muscle A7r5 cells induced by the phorbol ester PDBu. We found that endogenous low molecular weight caldesmon (l-caldesmon), which was normally localised to actin-stress fibres and membrane ruffles, was recruited to the actin cores of PDBu-induced podosomes. Overexpression of l-caldesmon in A7r5 cells caused dissociation of actin-stress fibres and disruption of focal adhesion complexes, and significantly reduced the ability of PDBu to induce podosome formation. By contrast, siRNA interference of caldesmon expression enhanced PDBu-induced formation of podosomes. The N-terminal fragment of l-caldesmon, CaD40, which contains the myosin-binding site, did not label stress fibres and was not translocated to PDBu-induced podosomes. Cad39, the C-terminal fragment housing the binding sites for actin, tropomyosin and calmodulin, was localised to stress fibres and was translocated to podosomes induced by PDBu. The caldesmon mutant, CadCamAB, which does not interact with Ca2+/calmodulin, was not recruited to PDBu-induced podosomes. These results show that (1) l-caldesmon is an integral part of the actin-rich core of the podosome; (2) overexpression of l-caldesmon suppresses podosome formation, whereas siRNA knock-down of l-caldesmon facilitates its formation; and (3) the actin-binding and calmodulin-binding sites on l-caldesmon are essential for the translocation of l-caldesmon to the podosomes. In summary, this data suggests that caldesmon may play a role in the regulation of the dynamics of podosome assembly and that Ca2+/calmodulin may be part of a regulatory mechanism in podosome formation.
Collapse
Affiliation(s)
- Robert Eves
- Department of Biochemistry, and Protein Function Discovery Program, Queen's University, Kingston, ON K7L 3N6, Canada
| | | | | | | |
Collapse
|
35
|
Russ M, Croft D, Ali O, Martinez R, Steimle P. Myosin heavy-chain kinase A from Dictyostelium possesses a novel actin-binding domain that cross-links actin filaments. Biochem J 2006; 395:373-83. [PMID: 16372899 PMCID: PMC1422765 DOI: 10.1042/bj20051376] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Myosin heavy-chain kinase A (MHCK A) catalyses the disassembly of myosin II filaments in Dictyostelium cells via myosin II heavy-chain phosphorylation. MHCK A possesses a 'coiled-coil'-enriched domain that mediates the oligomerization, cellular localization and actin-binding activities of the kinase. F-actin (filamentous actin) binding by the coiled-coil domain leads to a 40-fold increase in MHCK A activity. In the present study we examined the actin-binding characteristics of the coiled-coil domain as a means of identifying mechanisms by which MHCK A-mediated disassembly of myosin II filaments can be regulated in the cell. Co-sedimentation assays revealed that the coiled-coil domain of MHCK A binds co-operatively to F-actin with an apparent K(D) of approx. 0.5 muM and a stoichiometry of approx. 5:1 [actin/C(1-498)]. Further analyses indicate that the coiled-coil domain binds along the length of the actin filament and possesses at least two actin-binding regions. Quite surprisingly, we found that the coiled-coil domain cross-links actin filaments into bundles, indicating that MHCK A can affect the cytoskeleton in two important ways: (1) by driving myosin II-filament disassembly via myosin II heavy-chain phosphorylation, and (2) by cross-linking/bundling actin filaments. This discovery, along with other supporting data, suggests a model in which MHCK A-mediated bundling of actin filaments plays a central role in the recruitment and activation of the kinase at specific sites in the cell. Ultimately this provides a means for achieving the robust and highly localized disruption of myosin II filaments that facilitates polarized changes in cell shape during processes such as chemotaxis, cytokinesis and multicellular development.
Collapse
Affiliation(s)
- Misty Russ
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27402, U.S.A
| | - Daniel Croft
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27402, U.S.A
| | - Omar Ali
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27402, U.S.A
| | - Raquel Martinez
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27402, U.S.A
| | - Paul A. Steimle
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27402, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
36
|
Hashimoto Y, Skacel M, Adams JC. Roles of fascin in human carcinoma motility and signaling: prospects for a novel biomarker? Int J Biochem Cell Biol 2005; 37:1787-804. [PMID: 16002322 DOI: 10.1016/j.biocel.2005.05.004] [Citation(s) in RCA: 212] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2005] [Revised: 04/27/2005] [Accepted: 05/03/2005] [Indexed: 12/20/2022]
Abstract
Fascin is a globular actin cross-linking protein that has a major function in forming parallel actin bundles in cell protrusions that are key specialisations of the plasma membrane for environmental guidance and cell migration. Fascin is widely expressed in mesenchymal tissues and the nervous system and is low or absent in adult epithelia. Recent data from a number of laboratories have highlighted that fascin is up-regulated in many human carcinomas and, in individual tissues, correlates with the clinical aggressiveness of tumours and poor patient survival. In cell culture, over-expression or depletion of fascin modulates cell migration and alters cytoskeletal organisation. The identification of biomarkers to provide more effective early diagnosis of potentially aggressive tumours, or identify tumours susceptible to targeted therapies, is an important goal in clinical research. Here, we discuss the evidence that fascin is upregulated in carcinomas, its contributions to carcinoma cell behaviour and its potential as a candidate novel biomarker or therapeutic target.
Collapse
Affiliation(s)
- Yosuke Hashimoto
- Department of Cell Biology, Lerner Research Institute, NC1-10, Cleveland Clinic Foundation, OH 44195, USA
| | | | | |
Collapse
|
37
|
Cuomo ME, Knebel A, Platt G, Morrice N, Cohen P, Mittnacht S. Regulation of microfilament organization by Kaposi sarcoma-associated herpes virus-cyclin.CDK6 phosphorylation of caldesmon. J Biol Chem 2005; 280:35844-58. [PMID: 16115893 DOI: 10.1074/jbc.m503877200] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Kaposi sarcoma-associated herpes virus (KSHV) encodes a D-like cyclin (K-cyclin) that is thought to contribute to the viral oncogenicity. K-cyclin activates cellular cyclin-dependent kinases (CDK) 4 and 6, generating enzymes with a substrate selectivity deviant from CDK4 and CDK6 activated by D-type cyclins, suggesting different biochemical and biological functions. Here we report the identification of the actin- and calmodulin-binding protein caldesmon (CALD1) as a novel K-cyclin.CDK substrate, which is not phosphorylated by D.CDK. CALD1 plays a central role in the regulation of microfilament organization, consequently controlling cell shape, adhesion, cytokinesis and motility. K-cyclin.CDK6 specifically phosphorylates four Ser/Thr sites in the human CALD1 carboxyl terminus, abolishing CALD1 binding to its effector protein, actin, and its regulator protein, calmodulin. CALD1 is hyperphosphorylated in cells following K-cyclin expression and in KSHV-transformed lymphoma cells. Moreover, expression of exogenous K-cyclin results in microfilament loss and changes in cell morphology; both effects are reliant on CDK catalysis and can be reversed by the expression of a phosphorylation defective CALD1. Together, these data strongly suggest that K-cyclin expression modulates the activity of caldesmon and through this the microfilament functions in cells. These results establish a novel link between KSHV infection and the regulation of the actin cytoskeleton.
Collapse
MESH Headings
- Actins/chemistry
- Animals
- Calmodulin-Binding Proteins/chemistry
- Calmodulin-Binding Proteins/metabolism
- Catalysis
- Chromatography, Affinity
- Cloning, Molecular
- Cyclin-Dependent Kinase 4/metabolism
- Cyclin-Dependent Kinase 6/metabolism
- Cytoskeleton/metabolism
- Electrophoresis, Polyacrylamide Gel
- Gene Expression Regulation, Viral
- HeLa Cells
- Herpesvirus 8, Human/metabolism
- Humans
- Mass Spectrometry
- Mice
- Microscopy, Fluorescence
- NIH 3T3 Cells
- Peptides/chemistry
- Phosphorylation
- Protein Binding
- Protein Structure, Tertiary
- RNA, Small Interfering/metabolism
- Recombinant Proteins/chemistry
- Retinoblastoma Protein/metabolism
- Sepharose/chemistry
- Serine/chemistry
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Substrate Specificity
- Threonine/chemistry
- Time Factors
Collapse
Affiliation(s)
- Maria Emanuela Cuomo
- Cancer Research UK Centre for Cell and Molecular Biology, Chester Beatty Laboratories, The Institute of Cancer Research, 237 Fulham Road, SW3 6JB London, United Kingdom
| | | | | | | | | | | |
Collapse
|
38
|
Li Y, Lin JLC, Reiter RS, Daniels K, Soll DR, Lin JJC. Caldesmon mutant defective in Ca2+-calmodulin binding interferes with assembly of stress fibers and affects cell morphology, growth and motility. J Cell Sci 2004; 117:3593-604. [PMID: 15226374 DOI: 10.1242/jcs.01216] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Despite intensive in vitro studies, little is known about the regulation of caldesmon (CaD) by Ca2+-calmodulin (Ca2+-CaM) in vivo. To investigate this regulation, a mutant was generated of the C-terminal fragment of human fibroblast CaD, termed CaD39-AB, in which two crucial tryptophan residues involved in Ca2+-CaM binding were each replaced with alanine. The mutation abolished most CaD39-AB binding to Ca2+-CaM in vitro but had little effect on in vitro binding to actin filaments and the ability to inhibit actin/tropomyosin-activated heavy meromyosin ATPase. To study the functional consequences of these mutations in vivo, we transfected an expression plasmid carrying CaD39-AB cDNA into Chinese hamster ovary (CHO) cells and isolated several clones expressing various amounts of CaD39-AB. Immunofluorescence microscopy revealed that mutant CaD39-AB was distributed diffusely throughout the cytoplasm but also concentrated at membrane ruffle regions. Stable expression of CaD39-AB in CHO cells disrupted assembly of stress fibers and focal adhesions, altered cell morphology, and slowed cell cycle progression. Moreover, CaD39-AB-expressing cells exhibited motility defects in a wound-healing assay, in both velocity and the persistence of translocation, suggesting a role for CaD regulation by Ca2+-CaM in cell migration. Together, these results demonstrate that CaD plays a crucial role in mediating the effects of Ca2+-CaM on the dynamics of the actin cytoskeleton during cell migration.
Collapse
Affiliation(s)
- Yan Li
- Department of Biological Sciences, University of Iowa, 340 Biology Building East, Iowa City, IA 52242-1324, USA
| | | | | | | | | | | |
Collapse
|
39
|
Peraud A, Mondal S, Hawkins C, Mastronardi M, Bailey K, Rutka JT. Expression of fascin, an actin-bundling protein, in astrocytomas of varying grades. Brain Tumor Pathol 2004; 20:53-8. [PMID: 14756441 DOI: 10.1007/bf02483447] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Malignant astrocytomas are highly infiltrative neoplasms that invade readily into regions of normal brain. On a cellular basis, the motility and invasiveness of human cancers can be ascribed in part to complex rearrangements of the actin cytoskeleton that are governed by several actinbinding proteins. One such actin-binding protein that has been linked to the invasive behavior of carcinomas is fascin, which serves to aggregate F actin into bundles. In this study, we examined the expression of fascin in a series of human malignant astrocytomas (WHO grades I-IV). Five grade I, 5 grade II, 10 grade III, and 26 grade IV human astrocytomas were examined for fascin and glial fibrillary acidic protein (GFAP) expression by double immunofluorescence confocal microscopy. Expression of fascin and GFAP was also determined by Western blot analysis. Fascin expression increased with increasing WHO grade of astrocytoma. This is in marked contrast to GFAP expression, which decreased with increasing WHO grade. In grades I and II neoplasms, and within non-neoplastic brain, fascin and GFAP were expressed diffusely within regions examined. However, in the higher-grade astrocytomas (grades III and IV), fascin and GFAP were expressed regionally in distinctly separate tumor cell populations. This is the first study to demonstrate the expression of fascin in human astrocytic neoplasms. The role that fascin plays in contributing to the invasive phenotype of anaplastic astrocytomas awaits further study and investigation.
Collapse
Affiliation(s)
- Aurelia Peraud
- Arthur and Sonia Labatt Brain Tumour Research Centre, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
40
|
Ishikawa R, Sakamoto T, Ando T, Higashi-Fujime S, Kohama K. Polarized actin bundles formed by human fascin-1: their sliding and disassembly on myosin II and myosin V in vitro. J Neurochem 2003; 87:676-85. [PMID: 14535950 DOI: 10.1046/j.1471-4159.2003.02058.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Fascin-1 is a putative bundling factor of actin filaments in the filopodia of neuronal growth cones. Here, we examined the structure of the actin bundle formed by human fascin-1 (actin/fascin bundle), and its mode of interaction with myosin in vitro. The distance between cross-linked filaments in the actin/bundle was 8-9 nm, and the bundle showed the transverse periodicity of 36 nm perpendicular to the bundle axis, which was confirmed by electron microscopy. Decoration of the actin/fascin bundle with heavy meromyosin revealed that the arrowheads of filaments in the bundle pointed in the same direction, indicating that the bundle has polarity. This result suggested that fascin-1 plays an essential role in polarity of actin bundles in filopodia. In the in vitro motility assay, actin/fascin bundles slid as fast as single actin filaments on myosin II and myosin V. When myosin was attached to the surface at high density, the actin/fascin bundle disassembled to single filaments at the pointed end of the bundle during sliding. These results suggest that myosins may drive filopodial actin bundles backward by interacting with actin filaments on the surface, and may induce disassembly of the bundle at the basal region of filopodia.
Collapse
Affiliation(s)
- Ryoki Ishikawa
- Department of Pharmacology, Gunma University School of Medicine, Maebashi, Japan.
| | | | | | | | | |
Collapse
|
41
|
Matsumoto S, Hanai T, Ohnishi N, Yamamoto K, Kurita T. Bladder smooth muscle cell phenotypic changes and implication of expression of contractile proteins (especially caldesmon) in rats after partial outlet obstruction. Int J Urol 2003; 10:339-45. [PMID: 12757606 DOI: 10.1046/j.1442-2042.2003.00637.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND The purpose of the present study was to investigate morphological changes in bladder smooth muscle of rats with partial outlet obstruction. We investigated smooth muscle cell phenotypic changes and implication of synthetic phenotype in contractility decrease and bladder compliance after bladder outlet obstruction. METHODS Partial bladder outlet obstruction was introduced in female rats. Bladder were removed at 1, 3, 6, 10 and 20 weeks after the obstruction. Temporal pattern of changes in bladder mass, light microscopic pathogenesis and phenotypic expression of the bladder smooth muscle cells in the electron micrographs were investigated. Expression of contractile protein was also investigated by the immunoblotting method. RESULTS Marked increase in bladder mass with marked thickening of smooth muscle layer was observed at 1 week after obstruction. The ratio of myocytes exhibiting contractile and synthetic phenotypes was almost constant until 6 weeks after the obstruction, but thereafter, synthetic phenotypes gradually increased and the ratio (synthetic/contractile phenotype) was 1.5-fold at 20 weeks after the obstruction. Caldesmon was most markedly expressed after the obstruction among contractile proteins examined by the immunoblotting method. CONCLUSION Phenotypic changes were confirmed in bladder smooth muscle, and the decrease of the ratio of contractile phenotype was observed after long-term obstruction of the bladder outlet. Among the contractile proteins in the bladder smooth muscle cell, caldesmon was considered a reliable marker for predicting the pathogenetic conditions of the bladder.
Collapse
|
42
|
Samstag Y, Eibert SM, Klemke M, Wabnitz GH. Actin cytoskeletal dynamics in T lymphocyte activation and migration. J Leukoc Biol 2003; 73:30-48. [PMID: 12525560 DOI: 10.1189/jlb.0602272] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Dynamic rearrangements of the actin cytoskeleton are crucial for the function of numerous cellular elements including T lymphocytes. They are required for migration of T lymphocytes through the body to scan for the presence of antigens, as well as for the formation and stabilization of the immunological synapse at the interface between antigen-presenting cells and T lymphocytes. Supramolecular activation clusters within the immunological synapse play an important role for the initiation of T cell responses and for the execution of T cell effector functions. In addition to the T cell receptor/CD3 induced actin nucleation via Wasp/Arp2/3-activation, signals through accessory receptors of the T cell (i.e., costimulation) regulate actin cytoskeletal dynamics. In this regard, the actin-binding proteins cofilin and L-plastin represent prominent candidates linking accessory receptor stimulation to the rearrangement of the actin cytoskeleton. Cofilin enhances actin polymerization via its actin-severing activity, and as a long-lasting effect, cofilin generates novel actin monomers through F-actin depolymerization. L-plastin stabilizes actin filament structures by means of its actin-bundling activity.
Collapse
Affiliation(s)
- Yvonne Samstag
- Institute for Immunology, Ruprecht-Karls-University, Im Neuenheimer Feld 305, D-69120 Heidelberg, Germany.
| | | | | | | |
Collapse
|
43
|
Abstract
BACKGROUND Fascin containing actin bundles provide mechanical support to cellular protrusions and stress fibers. In cancers, some malignant cells (e.g. subsets of breast and ovarian carcinomas) express fascin. In skin cancer, the role of fascin is unknown. METHODS Cases of 61 keratocytic neoplasms, 35 melanocytic neoplasms, nine extramammary Paget's disease (four with adenocarcinoma) and five sarcomas (angiosarcoma and atypical fibroxanthoma) were examined by immunohistochemistry, using monoclonal antihuman fascin antibody, clone 55 k-2 (Dako Corporation, Carpinteria, CA, USA). RESULTS Fascin labeled all sarcomas and all keratinocytic neoplasms except for pagetoid pattern Bowen's disease. The regions of most intense fascin labeling were seen in the basal cells of infiltrative tumor margins. A minority of Merkel cell carcinomas exhibited weak or absent immunoreactivity. All melanocytic nevi except for some junctional nests of dysplastic melanocytic nevi expressed fascin. However, pagetoid cells of melanoma in situ and epithelioid cells of invasive melanoma weakly expressed or did not express fascin, whereas melanoma cells exhibiting spindle cell morphologies labeled intensely with fascin. Lastly, all cells of extramammary Paget's disease and most associated adenocarcinomas cells did not or were faintly labeled by fascin antibodies. Decreased or absent fascin expression was significantly associated with skin cancers with a high risk for metastasis (e.g. melanoma) vs. those with a low risk (e.g. basal cell carcinoma) (24% vs. 100% with > 50% immunoreactivity; p = 0.0001, chi-squared test). CONCLUSION Fascin is expressed by skin tumors that locally infiltrate and replace surrounding tissues indicating a role for fascin in cell adhesion, cell motility and invasiveness. No or weak fascin expression is exhibited by cancers with pagetoid intraepidermal spread and by invasive tumors with a high risk of metastasis. Downregulation or loss of fascin's actin-bundling properties, probably associated with disorganization of cell-cell and cell-matrix contacts, may be a crucial step in the progression from locally invasive to widely disseminating cancers.
Collapse
|
44
|
Tseng Y, Schafer BW, Almo SC, Wirtz D. Functional synergy of actin filament cross-linking proteins. J Biol Chem 2002; 277:25609-16. [PMID: 12006593 DOI: 10.1074/jbc.m202609200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The organization of filamentous actin (F-actin) in resilient networks is coordinated by various F-actin cross-linking proteins. The relative tolerance of cells to null mutations of genes that code for a single actin cross-linking protein suggests that the functions of those proteins are highly redundant. This apparent functional redundancy may, however, reflect the limited resolution of available assays in assessing the mechanical role of F-actin cross-linking/bundling proteins. Using reconstituted F-actin networks and rheological methods, we demonstrate how alpha-actinin and fascin, two F-actin cross-linking/bundling proteins that co-localize along stress fibers and in lamellipodia, could synergistically enhance the resilience of F-actin networks in vitro. These two proteins can generate microfilament arrays that "yield" at a strain amplitude that is much larger than each one of the proteins separately. F-actin/alpha-actinin/fascin networks display strain-induced hardening, whereby the network "stiffens" under shear deformations, a phenomenon that is non-existent in F-actin/fascin networks and much weaker in F-actin/alpha-actinin networks. Strain-hardening is further enhanced at high rates of deformation and high concentrations of actin cross-linking proteins. A simplified model suggests that the optimum results of the competition between the increased stiffness of bundles and their decreased density of cross-links. Our studies support a re-evaluation of the notion of functional redundancy among cytoskeletal regulatory proteins.
Collapse
Affiliation(s)
- Yiider Tseng
- Department of Chemical Engineering and Graduate Program in Molecular Biophysics, The Johns Hopkins University, Baltimore, Maryland 21218, USA
| | | | | | | |
Collapse
|
45
|
Qian Y, Baisden JM, Cherezova L, Summy JM, Guappone-Koay A, Shi X, Mast T, Pustula J, Zot HG, Mazloum N, Lee MY, Flynn DC. PC phosphorylation increases the ability of AFAP-110 to cross-link actin filaments. Mol Biol Cell 2002; 13:2311-22. [PMID: 12134071 PMCID: PMC117315 DOI: 10.1091/mbc.e01-12-0148] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2001] [Revised: 04/02/2002] [Accepted: 04/12/2002] [Indexed: 11/11/2022] Open
Abstract
The actin filament-associated protein and Src-binding partner, AFAP-110, is an adaptor protein that links signaling molecules to actin filaments. AFAP-110 binds actin filaments directly and multimerizes through a leucine zipper motif. Cellular signals downstream of Src(527F) can regulate multimerization. Here, we determined recombinant AFAP-110 (rAFAP-110)-bound actin filaments cooperatively, through a lateral association. We demonstrate rAFAP-110 has the capability to cross-link actin filaments, and this ability is dependent on the integrity of the carboxy terminal actin binding domain. Deletion of the leucine zipper motif or PKC phosphorylation affected AFAP-110's conformation, which correlated with changes in multimerization and increased the capability of rAFAP-110 to cross-link actin filaments. AFAP-110 is both a substrate and binding partner of PKC. On PKC activation, stress filament organization is lost, motility structures form, and AFAP-110 colocalizes strongly with motility structures. Expression of a deletion mutant of AFAP-110 that is unable to bind PKC blocked the effect of PMA on actin filaments. We hypothesize that upon PKC activation, AFAP-110 can be cooperatively recruited to newly forming actin filaments, like those that exist in cell motility structures, and that PKC phosphorylation effects a conformational change that may enable AFAP-110 to promote actin filament cross-linking at the cell membrane.
Collapse
Affiliation(s)
- Yong Qian
- The Mary Babb Randolph Cancer Center and the Department of Microbiology and Immunology, West Virginia University, Morgantown, West Virginia 26506-9300, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kureishy N, Sapountzi V, Prag S, Anilkumar N, Adams JC. Fascins, and their roles in cell structure and function. Bioessays 2002; 24:350-61. [PMID: 11948621 DOI: 10.1002/bies.10070] [Citation(s) in RCA: 260] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The fascins are a structurally unique and evolutionarily conserved group of actin cross-linking proteins. Fascins function in the organisation of two major forms of actin-based structures: dynamic, cortical cell protrusions and cytoplasmic microfilament bundles. The cortical structures, which include filopodia, spikes, lamellipodial ribs, oocyte microvilli and the dendrites of dendritic cells, have roles in cell-matrix adhesion, cell interactions and cell migration, whereas the cytoplasmic actin bundles appear to participate in cell architecture. We discuss the current understanding of the cellular mechanisms that regulate the binding of fascin to actin and how these processes contribute to the organisation or disassembly of cell protrusions. Although the in vivo roles of fascin have been studied principally in Drosophila, several human diseases are associated with inherited or acquired alterations in the expression of fascins. Strategies to modulate fascin-containing protrusions and thereby cell adhesive and migratory behaviour could have potential for therapeutic intervention in these conditions. The supplementary material referred to in this section can be found at http://www.interscience.wiley.com/jpages/0265-9247/suppmat/2002/v24.350.html
Collapse
Affiliation(s)
- Nina Kureishy
- MRC Laboratory for Molecular Cell Biology and Department of Biochemistry and Molecular Biology, University College London
| | | | | | | | | |
Collapse
|
47
|
Alexanian AR, Bamburg JR, Hidaka H, Mornet D. Calcium-dependent regulation of interactions of caldesmon with calcium-binding proteins found in growth cones of chick forebrain neurons. Cell Mol Neurobiol 2001; 21:437-51. [PMID: 11860183 PMCID: PMC11533846 DOI: 10.1023/a:1013885404738] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
1. This study was undertaken to determine if caldesmon, calmodulin, S100beta, and neurocalcin delta were present in chick forebrain neurons, and if so, to investigate the interactions of these proteins in the presence of different concentrations of calcium. 2. Immunocytochemistry was used to determine the presence and localization of these proteins in cultured forebrain neurons. Western blotting, gel electrophoresis in the presence of different concentrations of calcium, chemical cross-linking, and affinity chromatography were used to investigate the interactions of these proteins with each other. 3. Our data show that caldesmon and three calcium-binding proteins (S100beta, calmodulin, and neurocalcin 3) are localized in growth cones and neurites of chick forebrain neurons in culture. In the presence of different concentration of calcium, these calcium-binding proteins have different affinities to caldesmon and to each other. S100beta binds with greater affinity than calmodulin to caldesmon, and its ability to bind to caldesmon is regulated by neurocalcin delta. 4. These findings suggest a specific calcium-dependent regulatory pathway for modulating actomyosin during growth cone motility.
Collapse
Affiliation(s)
- A R Alexanian
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, USA.
| | | | | | | |
Collapse
|
48
|
Baisden JM, Qian Y, Zot HM, Flynn DC. The actin filament-associated protein AFAP-110 is an adaptor protein that modulates changes in actin filament integrity. Oncogene 2001; 20:6435-47. [PMID: 11607843 DOI: 10.1038/sj.onc.1204784] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The actin filament-associated protein of 110 kDa (AFAP-110) was first identified as an SH3/SH2 binding partner for the nonreceptor tyrosine kinase, Src. Subsequent data have demonstrated that AFAP-110 can interact with other Src family members. AFAP-110 contains additional protein binding modules including two pleckstrin homology domains, a leucine zipper motif and a target sequence for serine/threonine phosphorylation. AFAP-110 interacts with actin filaments directly via a carboxy terminal actin-binding domain. Thus AFAP-110 may function as an adaptor protein by linking Src family members and/or other signaling proteins to actin filaments. AFAP-110 also has an intrinsic capability to alter actin filament integrity that can be revealed upon conformational changes associated with phosphorylation or mutagenesis. Recent data has indicated that AFAP-110 may also serve to activate cSrc in response to this conformational change as well. Thus, AFAP-110 may function in several ways by (1) acting as an adaptor protein that links signaling molecules to actin filaments, (2) serving as a platform for the construction of larger signaling complexes, (3) serving as an activator of Src family kinases in response to cellular signals that alter its conformation and (4) directly effecting actin filament organization as an actin filament cross-linking protein. Here, we will review the structure and function of AFAP-110 as well as potential binding partners and effectors of AFAP-110's ability to alter actin filament integrity.
Collapse
Affiliation(s)
- J M Baisden
- Department Microbiology & Immunology, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, West Virginia, WV 26506-9300 USA
| | | | | | | |
Collapse
|
49
|
Sasaki Y, Ohsawa K, Kanazawa H, Kohsaka S, Imai Y. Iba1 is an actin-cross-linking protein in macrophages/microglia. Biochem Biophys Res Commun 2001; 286:292-7. [PMID: 11500035 DOI: 10.1006/bbrc.2001.5388] [Citation(s) in RCA: 263] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Iba1 is a 17-kDa EF hand protein that is specifically expressed in macrophages/microglia and is upregulated during the activation of these cells. When exposed to macrophage colony-stimulating factor (M-CSF), microglia cell line MG5 immediately produces intense membrane ruffles in which Iba1 accumulates together with filamentous actin. In this study, we investigated the physical interaction between Iba1 and actin by centrifugation assay and electron microscopic examination and showed that Iba1 possesses actin-binding and -cross-linking activities. Inhibitory mutant Iba1 that suppresses M-CSF-induced membrane ruffling had lost the actin-cross-linking activity, and it inhibited the cross-linking activity of intact Iba1. These results indicate that Iba1 is a macrophage/microglia-specific actin-cross-linking protein essential for M-CSF-induced membrane ruffling.
Collapse
Affiliation(s)
- Y Sasaki
- Department of Neurochemistry, National Institute of Neuroscience, 4-1-1 Ogawahigashi, Kodaira, Tokyo, 187-8502, Japan
| | | | | | | | | |
Collapse
|
50
|
Tseng Y, Fedorov E, McCaffery JM, Almo SC, Wirtz D. Micromechanics and ultrastructure of actin filament networks crosslinked by human fascin: a comparison with alpha-actinin. J Mol Biol 2001; 310:351-66. [PMID: 11428894 DOI: 10.1006/jmbi.2001.4716] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fascin is an actin crosslinking protein that organizes actin filaments into tightly packed bundles believed to mediate the formation of cellular protrusions and to provide mechanical support to stress fibers. Using quantitative rheological methods, we studied the evolution of the mechanical behavior of filamentous actin (F-actin) networks assembled in the presence of human fascin. The mechanical properties of F-actin/fascin networks were directly compared with those formed by alpha-actinin, a prototypical actin filament crosslinking/bundling protein. Gelation of F-actin networks in the presence of fascin (fascin to actin molar ratio >1:50) exhibits a non-monotonic behavior characterized by a burst of elasticity followed by a slow decline over time. Moreover, the rate of gelation shows a non-monotonic dependence on fascin concentration. In contrast, alpha-actinin increased the F-actin network elasticity and the rate of gelation monotonically. Time-resolved multiple-angle light scattering and confocal and electron microscopies suggest that this unique behavior is due to competition between fascin-mediated crosslinking and side-branching of actin filaments and bundles, on the one hand, and delayed actin assembly and enhanced network micro-heterogeneity, on the other hand. The behavior of F-actin/fascin solutions under oscillatory shear of different frequencies, which mimics the cell's response to forces applied at different rates, supports a key role for fascin-mediated F-actin side-branching. F-actin side-branching promotes the formation of interconnected networks, which completely inhibits the motion of actin filaments and bundles. Our results therefore show that despite sharing seemingly similar F-actin crosslinking/bundling activity, alpha-actinin and fascin display completely different mechanical behavior. When viewed in the context of recent microrheological measurements in living cells, these results provide the basis for understanding the synergy between multiple crosslinking proteins, and in particular the complementary mechanical roles of fascin and alpha-actinin in vivo.
Collapse
Affiliation(s)
- Y Tseng
- Department of Chemical Engineering, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| | | | | | | | | |
Collapse
|