1
|
Ishida H, Woodman AG, Kitada N, Aizawa T, Vogel HJ. The Dictyostelium discoideum FimA protein, unlike yeast and plant fimbrins, is regulated by calcium similar to mammalian plastins. Sci Rep 2023; 13:16208. [PMID: 37758724 PMCID: PMC10533516 DOI: 10.1038/s41598-023-42682-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Plastins, also known as fimbrins, are highly conserved eukaryotic multidomain proteins that are involved in actin-bundling. They all contain four independently folded Calponin Homology-domains and an N-terminal headpiece that is comprised of two calcium-binding EF-hand motifs. Since calcium-binding has been shown to be integral to regulating the activity of the three mammalian plastin proteins, we decided to study the properties of the headpiece regions of fimbrins from the model plant Arabidopsis thaliana, the yeasts Saccharomyces cerevisiae and Schizosaccharomyces pombe and the amoeba Dictyostelium discoideum. Of these protein domains only the FimA headpiece from the amoeba protein possesses calcium binding properties. Structural characterization of this protein domain by multidimensional NMR and site-directed mutagenesis studies indicates that this EF-hand region of FimA also contains a regulatory 'switch helix' that is essential to regulating the activity of the human L-plastin protein. Interestingly this regulatory helical region seems to be lacking in the plant and yeast proteins and in fimbrins from all other nonmotile systems. Typical calmodulin antagonists can displace the switch-helix from the FimA headpiece, suggesting that such drugs can deregulate the Ca2+-regulation of the actin-bunding in the amoeba, thereby making it a useful organism for drug screening against mammalian plastins.
Collapse
Affiliation(s)
- Hiroaki Ishida
- Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Andrew G Woodman
- Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Naoya Kitada
- Faculty of Advanced Life Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Tomoyasu Aizawa
- Faculty of Advanced Life Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Hans J Vogel
- Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
2
|
Aoki K, Yamamoto K, Ohkuma M, Sugita T, Tanaka N, Takashima M. Hyphal Growth in Trichosporon asahii Is Accelerated by the Addition of Magnesium. Microbiol Spectr 2023; 11:e0424222. [PMID: 37102973 PMCID: PMC10269644 DOI: 10.1128/spectrum.04242-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 04/10/2023] [Indexed: 04/28/2023] Open
Abstract
Fungal dimorphism involves two morphologies: a unicellular yeast cell and a multicellular hyphal form. Invasion of hyphae into human cells causes severe opportunistic infections. The transition between yeast and hyphal forms is associated with the virulence of fungi; however, the mechanism is poorly understood. Therefore, we aimed to identify factors that induce hyphal growth of Trichosporon asahii, a dimorphic basidiomycete that causes trichosporonosis. T. asahii showed poor growth and formed small cells containing large lipid droplets and fragmented mitochondria when cultivated for 16 h in a nutrient-deficient liquid medium. However, these phenotypes were suppressed via the addition of yeast nitrogen base. When T. asahii cells were cultivated in the presence of different compounds present in the yeast nitrogen base, we found that magnesium sulfate was a key factor for inducing cell elongation, and its addition dramatically restored hyphal growth in T. asahii. In T. asahii hyphae, vacuoles were enlarged, the size of lipid droplets was decreased, and mitochondria were distributed throughout the cell cytoplasm and adjacent to the cell walls. Additionally, hyphal growth was disrupted due to treatment with an actin inhibitor. The actin inhibitor latrunculin A disrupted the mitochondrial distribution even in hyphal cells. Furthermore, magnesium sulfate treatment accelerated hyphal growth in T. asahii for 72 h when the cells were cultivated in a nutrient-deficient liquid medium. Collectively, our results suggest that an increase in magnesium levels triggers the transition from the yeast to hyphal form in T. asahii. These findings will support studies on the pathogenesis of fungi and aid in developing treatments. IMPORTANCE Understanding the mechanism underlying fungal dimorphism is crucial to discern its invasion into human cells. Invasion is caused by the hyphal form rather than the yeast form; therefore, it is important to understand the mechanism of transition from the yeast to hyphal form. To study the transition mechanism, we utilized Trichosporon asahii, a dimorphic basidiomycete that causes severe trichosporonosis since there are fewer studies on T. asahii than on ascomycetes. This study suggests that an increase in Mg2+, the most abundant mineral in living cells, triggers growth of filamentous hyphae and increases the distribution of mitochondria throughout the cell cytoplasm and adjacent to the cell walls in T. asahii. Understanding the mechanism of hyphal growth triggered by Mg2+ increase will provide a model system to explore fungal pathogenicity in the future.
Collapse
Affiliation(s)
- Keita Aoki
- Laboratory of Yeast Systematics, Tokyo NODAI Research Institute, Tokyo University of Agriculture, Setagaya, Tokyo, Japan
| | - Kosuke Yamamoto
- Department of Molecular Microbiology, Faculty of Life Sciences, Tokyo University of Agriculture, Setagaya, Tokyo, Japan
| | - Moriya Ohkuma
- Japan Collection of Microorganisms, RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
| | - Takashi Sugita
- Department of Microbiology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Naoto Tanaka
- Department of Molecular Microbiology, Faculty of Life Sciences, Tokyo University of Agriculture, Setagaya, Tokyo, Japan
| | - Masako Takashima
- Laboratory of Yeast Systematics, Tokyo NODAI Research Institute, Tokyo University of Agriculture, Setagaya, Tokyo, Japan
| |
Collapse
|
3
|
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
|
4
|
Lehne F, Bogdan S. Getting cells into shape by calcium-dependent actin cross-linking proteins. Front Cell Dev Biol 2023; 11:1171930. [PMID: 37025173 PMCID: PMC10070769 DOI: 10.3389/fcell.2023.1171930] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
The actin cytoskeleton represents a highly dynamic filament system providing cell structure and mechanical forces to drive a variety of cellular processes. The dynamics of the actin cytoskeleton are controlled by a number of conserved proteins that maintain the pool of actin monomers, promote actin nucleation, restrict the length of actin filaments and cross-link filaments into networks or bundles. Previous work has been established that cytoplasmic calcium is an important signal to rapidly relay information to the actin cytoskeleton, but the underlying mechanisms remain poorly understood. Here, we summarize new recent perspectives on how calcium fluxes are transduced to the actin cytoskeleton in a physiological context. In this mini-review we will focus on three calcium-binding EF-hand-containing actin cross-linking proteins, α-actinin, plastin and EFHD2/Swiprosin-1, and how these conserved proteins affect the cell's actin reorganization in the context of cell migration and wound closure in response to calcium.
Collapse
|
5
|
Abstract
To fulfill the cytoskeleton’s diverse functions in cell mechanics and motility, actin networks with specialized architectures are built by cross-linking proteins. How these cross-linkers specify cytoskeletal network geometry is poorly understood at the level of protein structure. Here, we introduce a machine-learning–enabled pipeline for visualizing cross-linkers bridging cytoskeletal filaments with cryogenic electron microscopy (cryo-EM). We apply our method to T-plastin, a member of the evolutionarily conserved plastin/fimbrin family, revealing a sequence of conformational changes that enables T-plastin to bridge pairs of actin filaments in both parallel and antiparallel orientations. This provides a structural framework for understanding how plastins can generate actin networks featuring mixed filament polarity. To orchestrate cell mechanics, trafficking, and motility, cytoskeletal filaments must assemble into higher-order networks whose local subcellular architecture and composition specify their functions. Cross-linking proteins bridge filaments at the nanoscale to control a network’s μm-scale geometry, thereby conferring its mechanical properties and functional dynamics. While these interfilament linkages are key determinants of cytoskeletal function, their structural mechanisms remain poorly understood. Plastins/fimbrins are an evolutionarily ancient family of tandem calponin-homology domain (CHD) proteins required to construct multiple classes of actin networks, which feature diverse geometries specialized to power cytokinesis, microvilli and stereocilia biogenesis, and persistent cell migration. Here, we focus on the structural basis of actin network assembly by human T-plastin, a ubiquitously expressed isoform necessary for the maintenance of stable cellular protrusions generated by actin polymerization forces. By implementing a machine-learning–enabled cryo-electron microscopy pipeline for visualizing cross-linkers bridging multiple filaments, we uncover a sequential bundling mechanism enabling T-plastin to bridge pairs of actin filaments in both parallel and antiparallel orientations. T-plastin populates distinct structural landscapes in these two bridging orientations that are selectively compatible with actin networks featuring divergent architectures and functions. Our structural, biochemical, and cell biological data highlight inter-CHD linkers as key structural elements underlying flexible but stable cross-linking that are likely to be disrupted by T-plastin mutations that cause hereditary bone diseases.
Collapse
|
6
|
Gao J, Ma Y, Yang G, Li G. Translationally controlled tumor protein: the mediator promoting cancer invasion and migration and its potential clinical prospects. J Zhejiang Univ Sci B 2022; 23:642-654. [PMID: 35953758 DOI: 10.1631/jzus.b2100910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Translationally controlled tumor protein (TCTP) is a highly conserved multifunctional protein localized in the cytoplasm and nucleus of eukaryotic cells. It is secreted through exosomes and its degradation is associated with the ubiquitin-proteasome system (UPS), heat shock protein 27 (Hsp27), and chaperone-mediated autophagy (CMA). Its structure contains three α-helices and eleven β-strands, and features a helical hairpin as its hallmark. TCTP shows a remarkable similarity to the methionine-R-sulfoxide reductase B (MsrB) and mammalian suppressor of Sec4 (Mss4/Dss4) protein families, which exerts guanine nucleotide exchange factor (GEF) activity on small guanosine triphosphatase (GTPase) proteins, suggesting that some functions of TCTP may at least depend on its GEF action. Indeed, TCTP exerts GEF activity on Ras homolog enriched in brain (Rheb) to boost the growth and proliferation of Drosophila cells. TCTP also enhances the expression of cell division control protein 42 homolog (Cdc42) to promote cancer cell invasion and migration. Moreover, TCTP regulates cytoskeleton organization by interacting with actin microfilament (MF) and microtubule (MT) proteins and inducing the epithelial-mesenchymal transition (EMT) process. In essence, TCTP promotes cancer cell movement. It is usually highly expressed in cancerous tissues and thus reduces patient survival; meanwhile, drugs can target TCTP to reduce this effect. In this review, we summarize the mechanisms of TCTP in promoting cancer invasion and migration, and describe the current inhibitory strategy to target TCTP in cancerous diseases.
Collapse
Affiliation(s)
- Junying Gao
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Yan Ma
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Guiwen Yang
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan 250014, China.
| | - Guorong Li
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan 250014, China. ,
| |
Collapse
|
7
|
Fischer L, Nosratlo M, Hast K, Karakaya E, Ströhlein N, Esser TU, Gerum R, Richter S, Engel FB, Detsch R, Fabry B, Thievessen I. Calcium supplementation of bioinks reduces shear stress-induced cell damage during bioprinting. Biofabrication 2022; 14. [PMID: 35896101 DOI: 10.1088/1758-5090/ac84af] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 07/27/2022] [Indexed: 11/12/2022]
Abstract
During bioprinting, cells are suspended in a viscous bioink and extruded under pressure through small diameter printing needles. The combination of high pressure and small needle diameter exposes cells to considerable shear stress, which can lead to cell damage and death. Approaches to monitor and control shear stress-induced cell damage are currently not well established. To visualize the effects of printing-induced shear stress on plasma membrane integrity, we add FM 1-43 to the bioink, a styryl dye that becomes fluorescent when bound to lipid membranes, such as the cellular plasma membrane. Upon plasma membrane disruption, the dye enters the cell and also stains intracellular membranes. Extrusion of alginate-suspended NIH/3T3 cells through a 200µm printing needle led to an increased FM 1-43 incorporation at high pressure, demonstrating that typical shear stresses during bioprinting can transiently damage the plasma membrane. Cell imaging in a microfluidic channel confirmed that FM 1-43 incorporation is caused by cell strain. Notably, high printing pressure also impaired cell survival in bioprinting experiments. Using cell types of different stiffnesses, we find that shear stress-induced cell strain, FM 1-43 incorporation and cell death were reduced in stiffer compared to softer cell types and demonstrate that cell damage and death correlate with shear stress-induced cell deformation. Importantly, supplementation of the suspension medium with physiological concentrations of CaCl2greatly reduced shear stress-induced cell damage and death but not cell deformation. As the sudden influx of calcium ions is known to induce rapid cellular vesicle exocytosis and subsequent actin polymerization in the cell cortex, we hypothesize that calcium supplementation facilitates the rapid resealing of plasma membrane damage sites. We recommend that bioinks should be routinely supplemented with physiological concentrations of calcium ions to reduce shear stress-induced cell damage and death during extrusion bioprinting.
Collapse
Affiliation(s)
- Lena Fischer
- Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Mojtaba Nosratlo
- Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Katharina Hast
- Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Emine Karakaya
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Nadine Ströhlein
- Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Tilman U Esser
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Richard Gerum
- Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany.,Department of Physics and Astronomy, York-University Toronto, Ontario, Canada
| | - Sebastian Richter
- Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - F B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Rainer Detsch
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ben Fabry
- Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ingo Thievessen
- Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
8
|
Linehan JB, Zepeda JL, Mitchell TA, LeClair EE. Follow that cell: leukocyte migration in L-plastin mutant zebrafish. Cytoskeleton (Hoboken) 2022; 79:26-37. [PMID: 35811499 DOI: 10.1002/cm.21717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/21/2022] [Accepted: 07/07/2022] [Indexed: 11/09/2022]
Abstract
Actin assemblies are important in motile cells such as leukocytes which form dynamic plasma membrane extensions or podia. L-plastin (LCP1) is a leukocyte-specific calcium-dependent actin-bundling protein that, in mammals, is known to affect immune cell migration. Previously, we generated CRISPR/Cas9 engineered zebrafish lacking L-plastin (lcp1-/-) and reported that they had reduced survival to adulthood, suggesting that lack of this actin-bundler might negatively affect the immune system. To test this hypothesis, we examined the distribution and migration of neutrophils and macrophages in the transparent tail of early zebrafish larvae using cell-specific markers and an established wound-migration assay. Knockout larvae were similar to their heterozygous siblings in having equal body sizes and comparable numbers of neutrophils in caudal hematopoietic tissue at two days post-fertilization, indicating no gross defect in neutrophil production or developmental migration. When stimulated by a tail wound, all genotypes of neutrophils were equally migratory in a two-hour window. However for macrophages we observed both migration defects and morphological differences. L-plastin knockout macrophages (lcp1 -/-) still homed to wounds but were slower, less directional and had a star-like morphology with many leading and trailing projections. In contrast, heterozygous macrophages lcp1 (+/-) were faster, more directional, and had a streamlined, slug-like morphology. Overall, these findings show that in larval zebrafish L-plastin knockout primarily affects the macrophage response with possible consequences for organismal immunity. Consistent with our observations, we propose a model in which cytoplasmic L-plastin negatively regulates macrophage integrin adhesion by holding these transmembrane heterodimers in a 'clasped', inactive form and is a necessary part of establishing macrophage polarity during chemokine-induced motility. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- J B Linehan
- Department of Biological Sciences, DePaul University, USA
| | - J L Zepeda
- Department of Biological Sciences, DePaul University, USA
| | - T A Mitchell
- Department of Biological Sciences, DePaul University, USA
| | - E E LeClair
- Department of Biological Sciences, DePaul University, USA
| |
Collapse
|
9
|
Joshi H, Morley SC. Efficient T Cell Migration and Activation Require L-Plastin. Front Immunol 2022; 13:916137. [PMID: 35844504 PMCID: PMC9277003 DOI: 10.3389/fimmu.2022.916137] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/02/2022] [Indexed: 11/28/2022] Open
Abstract
Rapid re-organization of the actin cytoskeleton supports T-cell trafficking towards immune sites and interaction with antigen presenting cells (APCs). F-actin rearrangement enables T-cell trafficking by stabilizing adhesion to vascular endothelial cells and promoting transendothelial migration. T-cell/APC immune synapse (IS) maturation also relies upon f-actin-anchored LFA-1:ICAM-1 ligation. Therefore, efficient T-cell responses require tight regulation of f-actin dynamics. In this review, we summarize how the actin-bundling protein L-plastin (LPL) regulates T-cell activation and migration. LPL enhances f-actin polymerization and also directly binds to the β2 chain of the integrin LFA-1 to support intercellular adhesion and IS formation in human and murine T cells. LPL- deficient T cells migrate slowly in response to chemo-attractants such as CXCL12, CCL19, and poorly polarize towards ICAM-1. Loss of LPL impairs thymic egress and intranodal motility. LPL is also required for T-cell IS maturation with APCs, and therefore for efficient cytokine production and proliferation. LPL-/- mice are less susceptible to T-cell mediated pathologies, such as allograft rejection and experimental autoimmune encephalomyelitis (EAE). LPL activity is regulated by its N-terminal “headpiece”, which contains serine and threonine phosphorylation and calcium- and calmodulin-binding sites. LPL phosphorylation is required for lamellipodia formation during adhesion and migration, and also for LFA-1 clustering during IS formation. However, the precise molecular interactions by which LPL supports T-cell functional responses remain unclear. Future studies elucidating LPL-mediated regulation of T-cell migration and/or activation may illuminate pathways for therapeutic targeting in T-cell-mediated diseases.
Collapse
Affiliation(s)
- Hemant Joshi
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO, United States
| | - Sharon Celeste Morley
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Division of Immunobiology, Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO, United States
- *Correspondence: Sharon Celeste Morley,
| |
Collapse
|
10
|
Allosteric regulation controls actin-bundling properties of human plastins. Nat Struct Mol Biol 2022; 29:519-528. [PMID: 35589838 DOI: 10.1038/s41594-022-00771-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/05/2022] [Indexed: 11/08/2022]
Abstract
Plastins/fimbrins are conserved actin-bundling proteins contributing to motility, cytokinesis and other cellular processes by organizing strikingly different actin assemblies as in aligned bundles and branched networks. We propose that this ability of human plastins stems from an allosteric communication between their actin-binding domains (ABD1/2) engaged in a tight spatial association. Here we show that ABD2 can bind actin three orders of magnitude stronger than ABD1, unless the domains are involved in an equally strong inhibitory engagement. A mutation mimicking physiologically relevant phosphorylation at the ABD1-ABD2 interface greatly weakened their association, dramatically potentiating actin cross-linking. Cryo-EM reconstruction revealed the ABD1-actin interface and enabled modeling of the plastin bridge and domain separation in parallel bundles. We predict that a strong and tunable allosteric inhibition between the domains allows plastins to modulate the cross-linking strength, contributing to remodeling of actin assemblies of different morphologies defining the unique place of plastins in actin organization.
Collapse
|
11
|
Plastin 3 in health and disease: a matter of balance. Cell Mol Life Sci 2021; 78:5275-5301. [PMID: 34023917 PMCID: PMC8257523 DOI: 10.1007/s00018-021-03843-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/06/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023]
Abstract
For a long time, PLS3 (plastin 3, also known as T-plastin or fimbrin) has been considered a rather inconspicuous protein, involved in F-actin-binding and -bundling. However, in recent years, a plethora of discoveries have turned PLS3 into a highly interesting protein involved in many cellular processes, signaling pathways, and diseases. PLS3 is localized on the X-chromosome, but shows sex-specific, inter-individual and tissue-specific expression variability pointing towards skewed X-inactivation. PLS3 is expressed in all solid tissues but usually not in hematopoietic cells. When escaping X-inactivation, PLS3 triggers a plethora of different types of cancers. Elevated PLS3 levels are considered a prognostic biomarker for cancer and refractory response to therapies. When it is knocked out or mutated in humans and mice, it causes osteoporosis with bone fractures; it is the only protein involved in actin dynamics responsible for osteoporosis. Instead, when PLS3 is upregulated, it acts as a highly protective SMN-independent modifier in spinal muscular atrophy (SMA). Here, it seems to counteract reduced F-actin levels by restoring impaired endocytosis and disturbed calcium homeostasis caused by reduced SMN levels. In contrast, an upregulation of PLS3 on wild-type level might cause osteoarthritis. This emphasizes that the amount of PLS3 in our cells must be precisely balanced; both too much and too little can be detrimental. Actin-dynamics, regulated by PLS3 among others, are crucial in a lot of cellular processes including endocytosis, cell migration, axonal growth, neurotransmission, translation, and others. Also, PLS3 levels influence the infection with different bacteria, mycosis, and other pathogens.
Collapse
|
12
|
Anaya EP, Lin X, Todd EM, Szasz TP, Morley SC. Novel Mouse Model Reveals That Serine Phosphorylation of L-Plastin Is Essential for Effective Splenic Clearance of Pneumococcus. THE JOURNAL OF IMMUNOLOGY 2021; 206:2135-2145. [PMID: 33858961 DOI: 10.4049/jimmunol.2000899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 02/19/2021] [Indexed: 01/04/2023]
Abstract
Asplenia imparts susceptibility to life-threatening sepsis with encapsulated bacteria, such as the pneumococcus. However, the cellular components within the splenic environment that guard against pneumococcal bacteremia have not been defined. The actin-bundling protein L-plastin (LPL) is essential for the generation of marginal zone B cells and for anti-pneumococcal host defense, as revealed by a mouse model of genetic LPL deficiency. In independent studies, serine phosphorylation of LPL at residue 5 (S5) has been described as a key "switch" in regulating LPL actin binding and subsequent cell motility, although much of the data are correlative. To test the importance of S5 phosphorylation in LPL function, and to specifically assess the requirement of LPL S5 phosphorylation in anti-pneumococcal host defense, we generated the "S5A" mouse, expressing endogenous LPL bearing a serine-to-alanine mutation at this position. S5A mice were bred to homozygosity, and LPL was expressed at levels equivalent to wild-type, but S5 phosphorylation was absent. S5A mice exhibited specific impairment in clearance of pneumococci following i.v. challenge, with 10-fold-higher bacterial bloodstream burden 24 h after challenge compared with wild-type or fully LPL-deficient animals. Defective bloodstream clearance correlated with diminished population of marginal zone macrophages and with reduced phagocytic capacity of multiple innate immune cells. Development and function of other tested leukocyte lineages, such as T and B cell motility and activation, were normal in S5A mice. The S5A mouse thus provides a novel system in which to elucidate the precise molecular control of critical immune cell functions in specific host-pathogen defense interactions.
Collapse
Affiliation(s)
- Edgar P Anaya
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO; and
| | - Xue Lin
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO; and
| | - Elizabeth M Todd
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO; and
| | - Taylor P Szasz
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO; and
| | - S Celeste Morley
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO; and .,Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
13
|
L-plastin Ser5 phosphorylation is modulated by the PI3K/SGK pathway and promotes breast cancer cell invasiveness. Cell Commun Signal 2021; 19:22. [PMID: 33618712 PMCID: PMC7898450 DOI: 10.1186/s12964-021-00710-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/18/2021] [Indexed: 01/15/2023] Open
Abstract
Background Metastasis is the predominant cause for cancer morbidity and mortality accounting for approximatively 90% of cancer deaths. The actin-bundling protein L-plastin has been proposed as a metastatic marker and phosphorylation on its residue Ser5 is known to increase its actin-bundling activity. We recently showed that activation of the ERK/MAPK signalling pathway leads to L-plastin Ser5 phosphorylation and that the downstream kinases RSK1 and RSK2 are able to directly phosphorylate Ser5. Here we investigate the involvement of the PI3K pathway in L-plastin Ser5 phosphorylation and the functional effect of this phosphorylation event in breast cancer cells. Methods To unravel the signal transduction network upstream of L-plastin Ser5 phosphorylation, we performed computational modelling based on immunoblot analysis data, followed by experimental validation through inhibition/overexpression studies and in vitro kinase assays. To assess the functional impact of L-plastin expression/Ser5 phosphorylation in breast cancer cells, we either silenced L-plastin in cell lines initially expressing endogenous L-plastin or neoexpressed L-plastin wild type and phosphovariants in cell lines devoid of endogenous L-plastin. The established cell lines were used for cell biology experiments and confocal microscopy analysis. Results Our modelling approach revealed that, in addition to the ERK/MAPK pathway and depending on the cellular context, the PI3K pathway contributes to L-plastin Ser5 phosphorylation through its downstream kinase SGK3. The results of the transwell invasion/migration assays showed that shRNA-mediated knockdown of L-plastin in BT-20 or HCC38 cells significantly reduced cell invasion, whereas stable expression of the phosphomimetic L-plastin Ser5Glu variant led to increased migration and invasion of BT-549 and MDA-MB-231 cells. Finally, confocal image analysis combined with zymography experiments and gelatin degradation assays provided evidence that L-plastin Ser5 phosphorylation promotes L-plastin recruitment to invadopodia, MMP-9 activity and concomitant extracellular matrix degradation. Conclusion Altogether, our results demonstrate that L-plastin Ser5 phosphorylation increases breast cancer cell invasiveness. Being a downstream molecule of both ERK/MAPK and PI3K/SGK pathways, L-plastin is proposed here as a potential target for therapeutic approaches that are aimed at blocking dysregulated signalling outcome of both pathways and, thus, at impairing cancer cell invasion and metastasis formation. Video abstract
Collapse
|
14
|
Schwebach CL, Kudryashova E, Kudryashov DS. Plastin 3 in X-Linked Osteoporosis: Imbalance of Ca 2+-Dependent Regulation Is Equivalent to Protein Loss. Front Cell Dev Biol 2021; 8:635783. [PMID: 33553175 PMCID: PMC7859272 DOI: 10.3389/fcell.2020.635783] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/28/2020] [Indexed: 12/14/2022] Open
Abstract
Osteogenesis imperfecta is a genetic disorder disrupting bone development and remodeling. The primary causes of osteogenesis imperfecta are pathogenic variants of collagen and collagen processing genes. However, recently variants of the actin bundling protein plastin 3 have been identified as another source of osteogenesis imperfecta. Plastin 3 is a highly conserved protein involved in several important cellular structures and processes and is controlled by intracellular Ca2+ which potently inhibits its actin-bundling activity. The precise mechanisms by which plastin 3 causes osteogenesis imperfecta remain unclear, but recent advances have contributed to our understanding of bone development and the actin cytoskeleton. Here, we review the link between plastin 3 and osteogenesis imperfecta highlighting in vitro studies and emphasizing the importance of Ca2+ regulation in the localization and functionality of plastin 3.
Collapse
Affiliation(s)
- Christopher L Schwebach
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, United States
| | - Elena Kudryashova
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, United States
| | - Dmitri S Kudryashov
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
15
|
Li X, Wang L, Huang B, Gu Y, Luo Y, Zhi X, Hu Y, Zhang H, Gu Z, Cui J, Cao L, Guo J, Wang Y, Zhou Q, Jiang H, Fang C, Weng W, Chen X, Chen X, Su J. Targeting actin-bundling protein L-plastin as an anabolic therapy for bone loss. SCIENCE ADVANCES 2020; 6:6/47/eabb7135. [PMID: 33208358 PMCID: PMC7673802 DOI: 10.1126/sciadv.abb7135] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 10/01/2020] [Indexed: 05/15/2023]
Abstract
The actin-bundling protein L-plastin (LPL) mediates the resorption activity of osteoclasts, but its therapeutic potential in pathological bone loss remains unexplored. Here, we report that LPL knockout mice show increased bone mass and cortical thickness with more mononuclear tartrate-resistant acid phosphatase-positive cells, osteoblasts, CD31hiEmcnhi endothelial vessels, and fewer multinuclear osteoclasts in the bone marrow and periosteum. LPL deletion impeded preosteoclasts fusion by inhibiting filopodia formation and increased the number of preosteoclasts, which release platelet-derived growth factor-BB to promote CD31hiEmcnhi vessel growth and bone formation. LPL expression is regulated by the phosphatidylinositol 3-kinase/AKT/specific protein 1 axis in response to receptor activator of nuclear factor-κB ligand. Furthermore, we identified an LPL inhibitor, oroxylin A, that could maintain bone mass in ovariectomy-induced osteoporosis and accelerate bone fracture healing in mice. In conclusion, we showed that LPL regulates osteoclasts fusion, and targeting LPL serves as a novel anabolic therapy for pathological bone loss.
Collapse
Affiliation(s)
- Xiaoqun Li
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
- Department of Orthopedics, No. 929 Hospital, Naval Medical University, Shanghai 200433, China
| | - Lipeng Wang
- Graduate Management Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Biaotong Huang
- Institute of translational medicine, Shanghai University, Shanghai 201900, China
| | - Yanqiu Gu
- Department of Pharmacy, Shanghai Ninth People's Hospital, School of Medicine of Shanghai Jiao Tong University, Shanghai 201999, China
- School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Ying Luo
- Central Laboratory, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xin Zhi
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan Hu
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Hao Zhang
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zhengrong Gu
- Department of Orthopedics, Shanghai Baoshan Luodian Hospital, Shanghai 201900, China
| | - Jin Cui
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Liehu Cao
- Department of Orthopedics, Shanghai Baoshan Luodian Hospital, Shanghai 201900, China
| | - Jiawei Guo
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yajun Wang
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Qirong Zhou
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Hao Jiang
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Chao Fang
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Weizong Weng
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xiaofei Chen
- School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| | - Xiao Chen
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
- Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Jiacan Su
- Department of Orthopedics, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
- Institute of translational medicine, Shanghai University, Shanghai 201900, China
| |
Collapse
|
16
|
Vinogradova EV, Zhang X, Remillard D, Lazar DC, Suciu RM, Wang Y, Bianco G, Yamashita Y, Crowley VM, Schafroth MA, Yokoyama M, Konrad DB, Lum KM, Simon GM, Kemper EK, Lazear MR, Yin S, Blewett MM, Dix MM, Nguyen N, Shokhirev MN, Chin EN, Lairson LL, Melillo B, Schreiber SL, Forli S, Teijaro JR, Cravatt BF. An Activity-Guided Map of Electrophile-Cysteine Interactions in Primary Human T Cells. Cell 2020; 182:1009-1026.e29. [PMID: 32730809 PMCID: PMC7775622 DOI: 10.1016/j.cell.2020.07.001] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/14/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022]
Abstract
Electrophilic compounds originating from nature or chemical synthesis have profound effects on immune cells. These compounds are thought to act by cysteine modification to alter the functions of immune-relevant proteins; however, our understanding of electrophile-sensitive cysteines in the human immune proteome remains limited. Here, we present a global map of cysteines in primary human T cells that are susceptible to covalent modification by electrophilic small molecules. More than 3,000 covalently liganded cysteines were found on functionally and structurally diverse proteins, including many that play fundamental roles in immunology. We further show that electrophilic compounds can impair T cell activation by distinct mechanisms involving the direct functional perturbation and/or degradation of proteins. Our findings reveal a rich content of ligandable cysteines in human T cells and point to electrophilic small molecules as a fertile source for chemical probes and ultimately therapeutics that modulate immunological processes and their associated disorders.
Collapse
Affiliation(s)
| | - Xiaoyu Zhang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - David Remillard
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Daniel C Lazar
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Radu M Suciu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yujia Wang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Giulia Bianco
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yu Yamashita
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA; Medicinal Chemistry Research Laboratories, New Drug Research Division, Otsuka Pharmaceutical Co., Ltd., 463-10 Kawauchi-cho, Tokushima 771-0192, Japan
| | - Vincent M Crowley
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michael A Schafroth
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Minoru Yokoyama
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - David B Konrad
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kenneth M Lum
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Gabriel M Simon
- Vividion Therapeutics, 5820 Nancy Ridge Drive, San Diego, CA 92121, USA
| | - Esther K Kemper
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michael R Lazear
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sifei Yin
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Megan M Blewett
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Melissa M Dix
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nhan Nguyen
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Maxim N Shokhirev
- Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Emily N Chin
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Luke L Lairson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bruno Melillo
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA; Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02138, USA
| | - Stuart L Schreiber
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA 02138, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Stefano Forli
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - John R Teijaro
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
17
|
Schaffner-Reckinger E, Machado RAC. The actin-bundling protein L-plastin-A double-edged sword: Beneficial for the immune response, maleficent in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 355:109-154. [PMID: 32859369 DOI: 10.1016/bs.ircmb.2020.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The dynamic organization of the actin cytoskeleton into bundles and networks is orchestrated by a large variety of actin-binding proteins. Among them, the actin-bundling protein L-plastin is normally expressed in hematopoietic cells, where it is involved in the immune response. However, L-plastin is also often ectopically expressed in malignant cancer cells of non-hematopoietic origin and is even considered as a marker for cancer progression. Post-translational modification modulates L-plastin activity. In particular, L-plastin Ser5 phosphorylation has been shown to be important for the immune response in leukocytes as well as for invasion and metastasis formation of carcinoma cells. This chapter discusses the physiological and pathological role of L-plastin with a special focus on the importance of L-plastin Ser5 phosphorylation for the protein functions. The potential use of Ser5 phosphorylated L-plastin as a biomarker and/or therapeutic target will be evoked.
Collapse
Affiliation(s)
- Elisabeth Schaffner-Reckinger
- Cancer Cell Biology and Drug Discovery Group, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Raquel A C Machado
- Cancer Cell Biology and Drug Discovery Group, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
18
|
Schwebach CL, Kudryashova E, Zheng W, Orchard M, Smith H, Runyan LA, Egelman EH, Kudryashov DS. Osteogenesis imperfecta mutations in plastin 3 lead to impaired calcium regulation of actin bundling. Bone Res 2020; 8:21. [PMID: 32509377 PMCID: PMC7244493 DOI: 10.1038/s41413-020-0095-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 02/06/2020] [Accepted: 03/23/2020] [Indexed: 12/11/2022] Open
Abstract
Mutations in actin-bundling protein plastin 3 (PLS3) emerged as a cause of congenital osteoporosis, but neither the role of PLS3 in bone development nor the mechanisms underlying PLS3-dependent osteoporosis are understood. Of the over 20 identified osteoporosis-linked PLS3 mutations, we investigated all five that are expected to produce full-length protein. One of the mutations distorted an actin-binding loop in the second actin-binding domain of PLS3 and abolished F-actin bundling as revealed by cryo-EM reconstruction and protein interaction assays. Surprisingly, the remaining four mutants fully retained F-actin bundling ability. However, they displayed defects in Ca2+ sensitivity: two of the mutants lost the ability to be inhibited by Ca2+, while the other two became hypersensitive to Ca2+. Each group of the mutants with similar biochemical properties showed highly characteristic cellular behavior. Wild-type PLS3 was distributed between lamellipodia and focal adhesions. In striking contrast, the Ca2+-hyposensitive mutants were not found at the leading edge but localized exclusively at focal adhesions/stress fibers, which displayed reinforced morphology. Consistently, the Ca2+-hypersensitive PLS3 mutants were restricted to lamellipodia, while chelation of Ca2+ caused their redistribution to focal adhesions. Finally, the bundling-deficient mutant failed to co-localize with any F-actin structures in cells despite a preserved F-actin binding through a non-mutation-bearing actin-binding domain. Our findings revealed that severe osteoporosis can be caused by a mutational disruption of the Ca2+-controlled PLS3's cycling between adhesion complexes and the leading edge. Integration of the structural, biochemical, and cell biology insights enabled us to propose a molecular mechanism of plastin activity regulation by Ca2+.
Collapse
Affiliation(s)
- Christopher L. Schwebach
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
- Molecular Cellular and Developmental Biology graduate program, The Ohio State University, Columbus, OH 43210 USA
| | - Elena Kudryashova
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
| | - Weili Zheng
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908 USA
| | - Matthew Orchard
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
| | - Harper Smith
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
- Biophysics graduate program, The Ohio State University, Columbus, OH 43210 USA
| | - Lucas A. Runyan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
| | - Edward H. Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908 USA
| | - Dmitri S. Kudryashov
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
- Molecular Cellular and Developmental Biology graduate program, The Ohio State University, Columbus, OH 43210 USA
- Biophysics graduate program, The Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
19
|
Yokoi K, Balachandran C, Umezawa M, Tsuchiya K, Mitrić A, Aoki S. Amphiphilic Cationic Triscyclometalated Iridium(III) Complex-Peptide Hybrids Induce Paraptosis-like Cell Death of Cancer Cells via an Intracellular Ca 2+-Dependent Pathway. ACS OMEGA 2020; 5:6983-7001. [PMID: 32258934 PMCID: PMC7114882 DOI: 10.1021/acsomega.0c00337] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 02/27/2020] [Indexed: 06/11/2023]
Abstract
We report on the design and synthesis of a green-emitting iridium complex-peptide hybrid (IPH) 4, which has an electron-donating hydroxyacetic acid (glycolic acid) moiety between the Ir core and the peptide part. It was found that 4 is selectively cytotoxic against cancer cells, and the dead cells showed a green emission. Mechanistic studies of cell death indicate that 4 induces a paraptosis-like cell death through the increase in mitochondrial Ca2+ concentrations via direct Ca2+ transfer from ER to mitochondria, the loss of mitochondrial membrane potential (ΔΨm), and the vacuolization of cytoplasm and intracellular organelle. Although typical paraptosis and/or autophagy markers were upregulated by 4 through the mitogen-activated protein kinase (MAPK) signaling pathway, as confirmed by Western blot analysis, autophagy is not the main pathway in 4-induced cell death. The degradation of actin, which consists of a cytoskeleton, is also induced by high concentrations of Ca2+, as evidenced by costaining experiments using a specific probe. These results will be presented and discussed.
Collapse
Affiliation(s)
- Kenta Yokoi
- Faculty of Pharmaceutical
Sciences, Tokyo University of Science, 2641 Yamazaki,
Noda, Chiba 278-8510, Japan
| | - Chandrasekar Balachandran
- Faculty of Pharmaceutical
Sciences, Tokyo University of Science, 2641 Yamazaki,
Noda, Chiba 278-8510, Japan
| | - Masakazu Umezawa
- Research Institute for Science and Technology (RIST), Tokyo University of Science, 2641 Yamazaki,
Noda, Chiba 278-8510, Japan
| | - Koji Tsuchiya
- Research Institute for Science and Technology (RIST), Tokyo University of Science, 2641 Yamazaki,
Noda, Chiba 278-8510, Japan
| | - Aleksandra Mitrić
- Faculty of Pharmaceutical
Sciences, Tokyo University of Science, 2641 Yamazaki,
Noda, Chiba 278-8510, Japan
- Faculty of Technology and Metallurgy, University of Belgrade, 4 Karnegijeva Street, Belgrade 11000, Serbia
| | - Shin Aoki
- Faculty of Pharmaceutical
Sciences, Tokyo University of Science, 2641 Yamazaki,
Noda, Chiba 278-8510, Japan
- Research Institute for Science and Technology (RIST), Tokyo University of Science, 2641 Yamazaki,
Noda, Chiba 278-8510, Japan
| |
Collapse
|
20
|
Spatial oxidation of L-plastin downmodulates actin-based functions of tumor cells. Nat Commun 2019; 10:4073. [PMID: 31501427 PMCID: PMC6733871 DOI: 10.1038/s41467-019-11909-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 08/06/2019] [Indexed: 01/15/2023] Open
Abstract
Several antitumor therapies work by increasing reactive oxygen species (ROS) within the tumor micromilieu. Here, we reveal that L-plastin (LPL), an established tumor marker, is reversibly regulated by ROS-induced thiol oxidation on Cys101, which forms a disulfide bridge with Cys42. LPL reduction is mediated by the Thioredoxin1 (TRX1) system, as shown by TRX1 trapping, TRX1 knockdown and blockade of Thioredoxin1 reductase (TRXR1) with auranofin. LPL oxidation diminishes its actin-bundling capacity. Ratiometric imaging using an LPL-roGFP-Orp1 fusion protein and a dimedone-based proximity ligation assay (PLA) reveal that LPL oxidation occurs primarily in actin-based cellular extrusions and strongly inhibits cell spreading and filopodial extension formation in tumor cells. This effect is accompanied by decreased tumor cell migration, invasion and extracellular matrix (ECM) degradation. Since LPL oxidation occurs following treatment of tumors with auranofin or γ-irradiation, it may be a molecular mechanism contributing to the effectiveness of tumor treatment with redox-altering therapies. The actin-remodelling protein L-plastin promotes tumour migration and invasion. Here, the authors show that L-plastin is regulated spatially by ROS-induced thiol oxidation which inhibits its actin-bundling function and cell spreading and filopodial extension formation in tumor cells.
Collapse
|
21
|
Freudenmann LK, Mayer C, Rodemann HP, Dittmann K. Reduced exosomal L-Plastin is responsible for radiation-induced bystander effect. Exp Cell Res 2019; 383:111498. [PMID: 31302031 DOI: 10.1016/j.yexcr.2019.111498] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 01/21/2023]
Abstract
Radiation-induced bystander effects (RIBE) are discussed as relevant processes during radiotherapy. Irradiated cells are suggested to release growth-inhibitory/DNA-damaging factors transported to non-irradiated cells. However, the molecular nature of this phenomenon has not yet been resolved. We aimed at identifying the growth-inhibitory factor(s) transmitted to non-irradiated cells. RIBE-competent PC3 cells were used to produce conditioned medium (CM) after exposure to ionizing radiation. Indicator cells were incubated with CM and clonogenic survival as well as cell proliferation were determined as endpoints. A549 indicator cells exhibited a bystander effect upon incubation with CM from irradiated PC3 cells. This bystander effect was not due to DNA-damaging factors, but a radiation-triggered reduction of mitogenic/clonogenic activity present in CM. Several tumor cells, but not normal fibroblasts secrete this factor, whose release is reduced by irradiation. We identified L-Plastin to be responsible for the mitogenic/clonogenic activity. Removal of L-Plastin from CM by immunoprecipitation or siRNA-mediated knockdown of L-Plastin expression resulted in loss or reduction of mitogenic/clonogenic activity transmitted via CM, respectively. Exosome-transported L-Plastin was constitutively Ser5-phosphorylated, indicative of its bioactive conformation. In summary, we observed production and exosomal secretion of L-Plastin by cancer cells. Via exosome-transmitted L-Plastin, tumors induce clonogenic and mitogenic activity in cancer and normal cells of the tumor microenvironment. Irradiation inhibits L-Plastin production targeting both cancer cells and the tumor niche and may explain the high impact of radiotherapy in tumor control.
Collapse
Affiliation(s)
- Lena Katharina Freudenmann
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tübingen, Germany; DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), Germany
| | - Claus Mayer
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tübingen, Germany; DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), Germany
| | - H Peter Rodemann
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tübingen, Germany; DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), Germany
| | - Klaus Dittmann
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tübingen, Germany; DKFZ Partner Site Tübingen, German Cancer Consortium (DKTK), Germany.
| |
Collapse
|
22
|
Takahashi D, Suzuki K, Sakamoto T, Iwamoto T, Murata T, Sakane F. Crystal structure and calcium-induced conformational changes of diacylglycerol kinase α EF-hand domains. Protein Sci 2019; 28:694-706. [PMID: 30653270 DOI: 10.1002/pro.3572] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/10/2019] [Accepted: 01/10/2019] [Indexed: 01/07/2023]
Abstract
Diacylglycerol kinases (DGKs) are multi-domain lipid kinases that phosphorylate diacylglycerol into phosphatidic acid, modulating the levels of these key signaling lipids. Recently, increasing attention has been paid to DGKα isozyme as a potential target for cancer immunotherapy. We have previously shown that DGKα is positively regulated by Ca2+ binding to its N-terminal EF-hand domains (DGKα-EF). However, little progress has been made for the structural biology of mammalian DGKs and the molecular mechanism underlying the Ca2+ -triggered activation remains unclear. Here we report the first crystal structure of Ca2+ -bound DGKα-EF and analyze the structural changes upon binding to Ca2+ . DGKα-EF adopts a canonical EF-hand fold, but unexpectedly, has an additional α-helix (often called a ligand mimic [LM] helix), which is packed into the hydrophobic core. Biophysical and biochemical analyses reveal that DGKα-EF adopts a protease-susceptible "open" conformation without Ca2+ that tends to form a dimer. Cooperative binding of two Ca2+ ions dissociates the dimer into a well-folded monomer, which resists to proteolysis. Taken together, our results provide experimental evidence that Ca2+ binding induces substantial conformational changes in DGKα-EF, which likely regulates intra-molecular interactions responsible for the activation of DGKα and suggest a possible role of the LM helix for the Ca2+ -induced conformational changes. SIGNIFICANCE STATEMENT: Diacylglycerol kinases (DGKs), which modulates the levels of two lipid second messengers, diacylglycerol and phosphatidic acid, is still structurally enigmatic enzymes since its first identification in 1959. We here present the first crystal structure of EF-hand domains of diacylglycerol kinase α in its Ca2+ bound form and characterize Ca2+ -induced conformational changes, which likely regulates intra-molecular interactions. Our study paves the way for future studies to understand the structural basis of DGK isozymes.
Collapse
Affiliation(s)
- Daisuke Takahashi
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Kano Suzuki
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| | - Taiichi Sakamoto
- Department of Life Science, Faculty of Advanced Engineering, Chiba Institute of Technology, Chiba, Japan
| | - Takeo Iwamoto
- Division of Molecular Cell Biology, Core Research Facilities for Basic Science, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba, Japan
| | - Takeshi Murata
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan.,Molecular Chirality Research Center, Chiba University, Chiba, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba, Japan
| |
Collapse
|
23
|
Khassan O, Jensen KV, Woodman AG, Vogel HJ, Ishida H. Characterization of the EF-Hand Calcium-Binding Domains of Human Plastins. Methods Mol Biol 2019; 1929:245-260. [PMID: 30710278 DOI: 10.1007/978-1-4939-9030-6_16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The three human plastins (L-plastin, T-plastin, and I-plastin) are important regulatory Ca2+-binding proteins that belong to the family of actin-binding proteins. Plastins are involved in the regulation of the actin cytoskeleton as well as the cross-linking of actin filaments. In addition to four calponin-homology (CH) domains, all three plastins contain two N-terminal EF-hand Ca2+-binding motifs which together are homologous to a single lobe of the well-known calcium-regulatory protein calmodulin. This part of the protein allows for the regulation of the actin bundling activity in response to elevated calcium levels. In this protocol, we describe the purification of the EF-hand headpiece domains of all three plastins, as well as SPR studies, ITC studies, and NMR interaction studies with different peptides and calcium. In combination, these three experimental techniques provide detailed insights into a novel regulatory mechanism, involving the linker region between the EF-hand domain and the first CH domain of the plastins.
Collapse
Affiliation(s)
- Oleg Khassan
- Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Katharine V Jensen
- Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Andrew G Woodman
- Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Hans J Vogel
- Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, AB, Canada.
| | - Hiroaki Ishida
- Biochemistry Research Group, Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
24
|
Ngo VA, Fanning JK, Noskov SY. Comparative Analysis of Protein Hydration from MD simulations with Additive and Polarizable Force Fields. ADVANCED THEORY AND SIMULATIONS 2018. [DOI: 10.1002/adts.201800106] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Van A. Ngo
- Department of Biological SciencesCentre for Molecular Simulation and Biochemistry Research ClusterUniversity of Calgary Calgary Alberta T2N 1N4 Canada
| | - John Keenan Fanning
- Department of Biological SciencesCentre for Molecular Simulation and Biochemistry Research ClusterUniversity of Calgary Calgary Alberta T2N 1N4 Canada
| | - Sergei Yu Noskov
- Department of Biological SciencesCentre for Molecular Simulation and Biochemistry Research ClusterUniversity of Calgary Calgary Alberta T2N 1N4 Canada
| |
Collapse
|
25
|
Direct effects of Ca2+/calmodulin on actin filament formation. Biochem Biophys Res Commun 2018; 506:355-360. [DOI: 10.1016/j.bbrc.2018.07.159] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/31/2018] [Indexed: 01/06/2023]
|
26
|
Bosseler M, Marani V, Broukou A, Lequeux A, Kaoma T, Schlesser V, François JH, Palissot V, Berchem GJ, Aouali N, Janji B. Inhibition of HIF1α-Dependent Upregulation of Phospho-l-Plastin Resensitizes Multiple Myeloma Cells to Frontline Therapy. Int J Mol Sci 2018; 19:ijms19061551. [PMID: 29882856 PMCID: PMC6032243 DOI: 10.3390/ijms19061551] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/08/2018] [Accepted: 05/12/2018] [Indexed: 12/18/2022] Open
Abstract
The introduction of novel frontline agents in multiple myeloma (MM), like immunomodulatory drugs and proteasome inhibitors, has improved the overall survival of patients. Yet, MM is still not curable, and drug resistance (DR) remains the main challenge. To improve the understanding of DR in MM, we established a resistant cell line (MOLP8/R). The exploration of DR mechanisms yielded an overexpression of HIF1α, due to impaired proteasome activity of MOLP8/R. We show that MOLP8/R, like other tumor cells, overexpressing HIF1α, have an increased resistance to the immune system. By exploring the main target genes regulated by HIF1α, we could not show an overexpression of these targets in MOLP8/R. We, however, show that MOLP8/R cells display a very high overexpression of LCP1 gene (l-Plastin) controlled by HIF1α, and that this overexpression also exists in MM patient samples. The l-Plastin activity is controlled by its phosphorylation in Ser5. We further show that the inhibition of l-Plastin phosphorylation restores the sensitivity of MOLP8/R to immunomodulatory drugs (IMiDs) and proteasome inhibitors (PIs). Our results reveal a new target gene of DR, controlled by HIF1α.
Collapse
Affiliation(s)
- Manon Bosseler
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Vanessa Marani
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Angelina Broukou
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Amandine Lequeux
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Tony Kaoma
- Bioinformatics and Modelling, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Vincent Schlesser
- Laboratory of Hematology, Centre Hospitalier de Luxembourg (CHL), L-1526 Luxembourg City, Luxembourg.
| | - Jean-Hugues François
- Laboratory of Hematology, Centre Hospitalier de Luxembourg (CHL), L-1526 Luxembourg City, Luxembourg.
| | - Valérie Palissot
- Laboratory of Oncolytic-Virus-Immuno-Therapeutics, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Guy J Berchem
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
- Laboratory of Hematology, Centre Hospitalier de Luxembourg (CHL), L-1526 Luxembourg City, Luxembourg.
| | - Nasséra Aouali
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| | - Bassam Janji
- Laboratory of Experimental Cancer Research, Luxembourg Institute of Health (LIH), L-1526 Luxembourg City, Luxembourg.
| |
Collapse
|
27
|
Targeted deletion of the zebrafish actin-bundling protein L-plastin (lcp1). PLoS One 2018; 13:e0190353. [PMID: 29293625 PMCID: PMC5749806 DOI: 10.1371/journal.pone.0190353] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 12/13/2017] [Indexed: 01/09/2023] Open
Abstract
Regulation of the cytoskeleton is essential for cell migration in health and disease. Lymphocyte cytosolic protein 1 (lcp1, also called L-plastin) is a hematopoietic-specific actin-bundling protein that is highly conserved in zebrafish, mice and humans. In addition, L-plastin expression is documented as both a genetic marker and a cellular mechanism contributing to the invasiveness of tumors and transformed cell lines. Despite L-plastin’s role in both immunity and cancer, in zebrafish there are no direct studies of its function, and no mutant, knockout or reporter lines available. Using CRISPR-Cas9 genome editing, we generated null alleles of zebrafish lcp1 and examined the phenotypes of these fish throughout the life cycle. Our editing strategy used gRNA to target the second exon of lcp1, producing F0 mosaic fish that were outcrossed to wild types to confirm germline transmission. F1 heterozygotes were then sequenced to identify three unique null alleles, here called ‘Charlie’, ‘Foxtrot’ and ‘Lima’. In silico, each allele truncates the endogenous protein to less than 5% normal size and removes both essential actin-binding domains (ABD1 and ABD2). Although none of the null lines express detectable LCP1 protein, homozygous mutant zebrafish (-/-) can develop and reproduce normally, a finding consistent with that of the L-plastin null mouse (LPL -/-). However, such mice do have a profound immune defect when challenged by lung bacteria. Interestingly, we observed reduced long-term survival of zebrafish lcp1 -/- homozygotes (~30% below the expected numbers) in all three of our knockout lines, with greatest mortality corresponding to the period (4–6 weeks post-fertilization) when the innate immune system is functional, but the adaptive immune system is not yet mature. This suggests that null zebrafish may have reduced capacity to combat opportunistic infections, which are more easily transmissible in the aquatic environment. Overall, our novel mutant lines establish a sound genetic model and an enhanced platform for further studies of L-plastin gene function in hematopoiesis and cancer.
Collapse
|
28
|
Schenk LK, Möller-Kerutt A, Klosowski R, Wolters D, Schaffner-Reckinger E, Weide T, Pavenstädt H, Vollenbröker B. Angiotensin II regulates phosphorylation of actin-associated proteins in human podocytes. FASEB J 2017; 31:5019-5035. [PMID: 28768720 DOI: 10.1096/fj.201700142r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/17/2017] [Indexed: 02/02/2023]
Abstract
Within the kidney, angiotensin II (AngII) targets different cell types in the vasculature, tubuli, and glomeruli. An important part of the renal filtration barrier is composed of podocytes with their actin-rich foot processes. In this study, we used stable isotope labeling with amino acids in cell culture coupled to mass spectrometry to characterize relative changes in the phosphoproteome of human podocytes in response to short-term treatment with AngII. In 4 replicates, we identified a total of 17,956 peptides that were traceable to 2081 distinct proteins. Bioinformatic analyses revealed that among the increasingly phosphorylated peptides are predominantly peptides that are related to actin filaments, cytoskeleton, lamellipodia, mammalian target of rapamycin, and MAPK signaling. Among others, this screening approach highlighted the increased phosphorylation of actin-bundling protein, l-plastin (LCP1). AngII-dependent phosphorylation of LCP1 in cultured podocytes was mediated by the kinases ERK, p90 ribosomal S6 kinase, PKA, or PKC. LCP1 phosphorylation increased filopodia formation. In addition, treatment with AngII led to LCP1 redistribution to the cell margins, membrane ruffling, and formation of lamellipodia. Our data highlight the importance of AngII-triggered actin cytoskeleton-associated signal transduction in podocytes.-Schenk, L. K., Möller-Kerutt, A., Klosowski, R., Wolters, D., Schaffner-Reckinger, E., Weide, T., Pavenstädt, H., Vollenbröker, B. Angiotensin II regulates phosphorylation of actin-associated proteins in human podocytes.
Collapse
Affiliation(s)
- Laura K Schenk
- Medizinischen Klinik und Poliklinik D, Universitätsklinikum Münster, Munster, Germany
| | - Annika Möller-Kerutt
- Medizinischen Klinik und Poliklinik D, Universitätsklinikum Münster, Munster, Germany
| | - Rafael Klosowski
- Analytische Chemie, Biomolekulare Massenspektrometrie, Ruhr-Universität Bochum, Bochum, Germany
| | - Dirk Wolters
- Analytische Chemie, Biomolekulare Massenspektrometrie, Ruhr-Universität Bochum, Bochum, Germany
| | - Elisabeth Schaffner-Reckinger
- Laboratory of Cytoskeleton and Cell Plasticity, Life Sciences Research Unit, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Thomas Weide
- Medizinischen Klinik und Poliklinik D, Universitätsklinikum Münster, Munster, Germany
| | - Hermann Pavenstädt
- Medizinischen Klinik und Poliklinik D, Universitätsklinikum Münster, Munster, Germany
| | - Beate Vollenbröker
- Medizinischen Klinik und Poliklinik D, Universitätsklinikum Münster, Munster, Germany;
| |
Collapse
|
29
|
Schwebach CL, Agrawal R, Lindert S, Kudryashova E, Kudryashov DS. The Roles of Actin-Binding Domains 1 and 2 in the Calcium-Dependent Regulation of Actin Filament Bundling by Human Plastins. J Mol Biol 2017; 429:2490-2508. [PMID: 28694070 DOI: 10.1016/j.jmb.2017.06.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/30/2017] [Accepted: 06/30/2017] [Indexed: 01/04/2023]
Abstract
The actin cytoskeleton is a complex network controlled by a vast array of intricately regulated actin-binding proteins. Human plastins (PLS1, PLS2, and PLS3) are evolutionary conserved proteins that non-covalently crosslink actin filaments into tight bundles. Through stabilization of such bundles, plastins contribute, in an isoform-specific manner, to the formation of kidney and intestinal microvilli, inner ear stereocilia, immune synapses, endocytic patches, adhesion contacts, and invadosomes of immune and cancer cells. All plastins comprise an N-terminal Ca2+-binding regulatory headpiece domain followed by two actin-binding domains (ABD1 and ABD2). Actin bundling occurs due to simultaneous binding of both ABDs to separate actin filaments. Bundling is negatively regulated by Ca2+, but the mechanism of this inhibition remains unknown. In this study, we found that the bundling abilities of PLS1 and PLS2 were similarly sensitive to Ca2+ (pCa50 ~6.4), whereas PLS3 was less sensitive (pCa50 ~5.9). At the same time, all three isoforms bound to F-actin in a Ca2+-independent manner, suggesting that binding of only one of the ABDs is inhibited by Ca2+. Using limited proteolysis and mass spectrometry, we found that in the presence of Ca2+ the EF-hands of human plastins bound to an immediately adjacent sequence homologous to canonical calmodulin-binding peptides. Furthermore, our data from differential centrifugation, Förster resonance energy transfer, native electrophoresis, and chemical crosslinking suggest that Ca2+ does not affect ABD1 but inhibits the ability of ABD2 to interact with actin. A structural mechanism of signal transmission from Ca2+ to ABD2 through EF-hands remains to be established.
Collapse
Affiliation(s)
- Christopher L Schwebach
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA; Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH 43210, USA
| | - Richa Agrawal
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Steffen Lindert
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Elena Kudryashova
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Dmitri S Kudryashov
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA; Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
30
|
Kubiak JZ, Kloc M. Elusive Role of TCTP Protein and mRNA in Cell Cycle and Cytoskeleton Regulation. Results Probl Cell Differ 2017; 64:217-225. [PMID: 29149411 DOI: 10.1007/978-3-319-67591-6_11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Translationally Controlled Tumor-associated Protein (TCTP) is a small, 23 kDa multifunctional and ubiquitous protein localized both in the cytoplasm and in the nucleus of eukaryotic cells. It is evolutionarily highly conserved. Certain aspects of its structure show remarkable similarities to guanine nucleotide-free chaperons Mss4 and Dss4 suggesting that at least some functions of TCTP may depend on its chaperon-like action on other proteins. Besides other functions, TCTP is clearly involved in cell cycle regulation. It is also regulated in a cell-cycle-dependent manner suggesting a reciprocal interaction between this protein and the cell cycle-regulating machinery. TCTP also interacts with the cytoskeleton, mostly with actin microfilaments (MFs) and microtubules (MTs). It regulates the cytoskeleton organization and through this action it also influences cell shape and motility. The exact role of TCTP in cell cycle and cytoskeleton regulation is certainly not fully understood. In this chapter, we summarize recent data on cell cycle and cytoskeletal aspects of TCTP regulatory role.
Collapse
Affiliation(s)
- Jacek Z Kubiak
- Institute of Genetics and Development of Rennes (IGDR), Cell Cycle Group, CNRS, UMR 6290, 35043, Rennes, France.
- Faculty of Medicine, University Rennes 1, UEB, IFR 140, 35043, Rennes, France.
- Laboratory of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology (WIHE), Warsaw, Poland.
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgery, The Houston Methodist Hospital, Houston, TX, USA
- University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|