1
|
Inagaki T, Kim J, Eijiro M, Matsumoto T. Macroscopic creep behavior of spheroids derived from mesenchymal stem cells under compression. J Mech Behav Biomed Mater 2025; 161:106816. [PMID: 39549472 DOI: 10.1016/j.jmbbm.2024.106816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/18/2024]
Abstract
Spheroid culture, where cells are aggregated three-dimensionally, is expected to have applications as a model that better recapitulates invivo environment beyond two-dimensional environments. When human mesenchymal stem cells are subjected to spheroid culture in the presence of osteogenesis supplements, the gene expression of osteocyte differentiation marker is greatly increased within a short period compared to two-dimensional culture. However, how such alterations may be reflected to mechanical properties of the spheroid remains unknown. In this study, using a uniaxial compression system, we evaluated the macroscopic mechanical properties of human mesenchymal stem cell-derived spheroids including viscoelastic behavior. The Young's modulus of spheroids cultured for 2 days was about 18 kPa, whereas that of individual cells is around 1-10 kPa. We also found that creep behavior of the spheroid was greater in 50% strain compression beyond 10 or 30% strain, indicating that they are viscoelastic materials. Upon release from compression, the spheroids tended to revert to their original shape through elastic deformation. However, spheroids in which actin filament formation was inhibited exhibited a remarkably greater plastic deformation, suggesting that the actin filaments play a crucial role in the elastic behavior of spheroids. By understanding the mechanical properties and behavior of spheroids, it provides a framework for predicting and manipulating the development of tissues and organs in the field of morphogenesis.
Collapse
Affiliation(s)
- Takashi Inagaki
- Department of Mechanical Systems Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Jeonghyun Kim
- Department of Mechanical Systems Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan.
| | - Maeda Eijiro
- Department of Mechanical Systems Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Takeo Matsumoto
- Department of Mechanical Systems Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| |
Collapse
|
2
|
Li Y, Zhang H, Yang F, Zhu D, Chen S, Wang Z, Wei Z, Yang Z, Jia J, Zhang Y, Wang D, Ma M, Kang X. Mechanisms and therapeutic potential of disulphidptosis in cancer. Cell Prolif 2025; 58:e13752. [PMID: 39354653 DOI: 10.1111/cpr.13752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/30/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
SLC7A11 plays a pivotal role in tumour development by facilitating cystine import to enhance glutathione synthesis and counteract oxidative stress. Disulphidptosis, an emerging form of cell death observed in cells with high expression of SLC7A11 under glucose deprivation, is regulated through reduction-oxidation reactions and disulphide bond formation. This process leads to contraction and collapse of the F-actin cytoskeleton from the plasma membrane, ultimately resulting in cellular demise. Compared to other forms of cell death, disulphidptosis exhibits distinctive characteristics and regulatory mechanisms. This mechanism provides novel insights and innovative strategies for cancer treatment while also inspiring potential therapeutic approaches for other diseases. Our review focuses on elucidating the molecular mechanism underlying disulphidptosis and its connection with the actin cytoskeleton, identifying alternative metabolic forms of cell death, as well as offering insights into disulphidptosis-based cancer therapy. A comprehensive understanding of disulphidptosis will contribute to our knowledge about fundamental cellular homeostasis and facilitate the development of groundbreaking therapies for disease treatment.
Collapse
Affiliation(s)
- Yanhu Li
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Haijun Zhang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
- The Second People's Hospital of Gansu Province, Lanzhou, PR China
| | - Fengguang Yang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Daxue Zhu
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Shijie Chen
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Zhaoheng Wang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Ziyan Wei
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Zhili Yang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Jingwen Jia
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Yizhi Zhang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Dongxin Wang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Mingdong Ma
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Xuewen Kang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| |
Collapse
|
3
|
Gadsby JR, Ioannou PS, Butler R, Mason J, Smith AJ, Dobramysl U, Chin SE, Dobson C, Gallop JL. The open to closed D-loop conformational switch determines length in filopodia-like actin bundles. Biochem J 2024; 481:1977-1995. [PMID: 39621444 DOI: 10.1042/bcj20240367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/24/2024]
Abstract
Filopodia, microspikes and cytonemes are implicated in sensing the environment and in dissemination of morphogens, organelles and pathogens across tissues. Their major structural component is parallel bundles of actin filaments that assemble from the cell membrane. Whilst the length of filopodia is central to their function, it is not known how their lengths are determined by actin bundle dynamics. Here, we identified a set of monoclonal antibodies that lengthen filopodia-like structures formed in a cell-free reconstitution system, and used them to uncover a key molecular switch governing length regulation. Using immunolabelling, enzyme-linked immunosorbent assays, immunoprecipitation and immunoblock experiments, we identified four antibodies that lengthen actin bundles by selectively binding the open DNase 1-binding loop (D-loop) of actin filaments. The antibodies inhibit actin disassembly and their effects can be alleviated by providing additional actin or cofilin. This work indicates that maintaining an open state of the actin filament D-loop is a mechanism of generating long filopodia-like actin bundles.
Collapse
Affiliation(s)
- Jonathan R Gadsby
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Pantelis Savvas Ioannou
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Richard Butler
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
| | - Julia Mason
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Alison J Smith
- Biologics Engineering, Oncology R&D, AstraZeneca, Cambridge, U.K
| | - Ulrich Dobramysl
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, U.K
| | - Stacey E Chin
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, U.K
| | - Claire Dobson
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, U.K
| | - Jennifer L Gallop
- Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, U.K
| |
Collapse
|
4
|
Rötte M, Höhne MY, Klug D, Ramlow K, Zedler C, Lehne F, Schneider M, Bischoff MC, Bogdan S. CYRI controls epidermal wound closure and cohesion of invasive border cell cluster in Drosophila. J Cell Biol 2024; 223:e202310153. [PMID: 39453414 PMCID: PMC11519390 DOI: 10.1083/jcb.202310153] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 07/19/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024] Open
Abstract
Cell motility is crucial for many biological processes including morphogenesis, wound healing, and cancer invasion. The WAVE regulatory complex (WRC) is a central Arp2/3 regulator driving cell motility downstream of activation by Rac GTPase. CYFIP-related Rac1 interactor (CYRI) proteins are thought to compete with WRC for interaction with Rac1 in a feedback loop regulating lamellipodia dynamics. However, the physiological role of CYRI proteins in vivo in healthy tissues is unclear. Here, we used Drosophila as a model system to study CYRI function at the cellular and organismal levels. We found that CYRI is not only a potent WRC regulator in single macrophages that controls lamellipodial spreading but also identified CYRI as a molecular brake on the Rac-WRC-Arp2/3 pathway to slow down epidermal wound healing. In addition, we found that CYRI limits invasive border cell migration by controlling cluster cohesion and migration. Thus, our data highlight CYRI as an important regulator of cellular and epithelial tissue dynamics conserved across species.
Collapse
Affiliation(s)
- Marvin Rötte
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Mila Y. Höhne
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Dennis Klug
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Kirsten Ramlow
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Caroline Zedler
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Franziska Lehne
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Meike Schneider
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Maik C. Bischoff
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| | - Sven Bogdan
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
5
|
Boiero Sanders M, Oosterheert W, Hofnagel O, Bieling P, Raunser S. Phalloidin and DNase I-bound F-actin pointed end structures reveal principles of filament stabilization and disassembly. Nat Commun 2024; 15:7969. [PMID: 39261469 PMCID: PMC11390976 DOI: 10.1038/s41467-024-52251-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Actin filament turnover involves subunits binding to and dissociating from the filament ends, with the pointed end being the primary site of filament disassembly. Several molecules modulate filament turnover, but the underlying mechanisms remain incompletely understood. Here, we present three cryo-EM structures of the F-actin pointed end in the presence and absence of phalloidin or DNase I. The two terminal subunits at the undecorated pointed end adopt a twisted conformation. Phalloidin can still bind and bridge these subunits, inducing a conformational shift to a flattened, F-actin-like state. This explains how phalloidin prevents depolymerization at the pointed end. Interestingly, two DNase I molecules simultaneously bind to the phalloidin-stabilized pointed end. In the absence of phalloidin, DNase I binding would disrupt the terminal actin subunit packing, resulting in filament disassembly. Our findings uncover molecular principles of pointed end regulation and provide structural insights into the kinetic asymmetry between the actin filament ends.
Collapse
Affiliation(s)
- Micaela Boiero Sanders
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Wout Oosterheert
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Oliver Hofnagel
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Peter Bieling
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany.
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany.
| |
Collapse
|
6
|
Limatola N, Chun JT, Schmitt JL, Lehn JM, Santella L. The Effect of Synthetic Polyamine BPA-C8 on the Fertilization Process of Intact and Denuded Sea Urchin Eggs. Cells 2024; 13:1477. [PMID: 39273047 PMCID: PMC11394060 DOI: 10.3390/cells13171477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Sea urchin eggs are covered with layers of extracellular matrix, namely, the vitelline layer (VL) and jelly coat (JC). It has been shown that sea urchin eggs' JC components serve as chemoattractants or ligands for the receptor on the fertilizing sperm to promote the acrosome reaction. Moreover, the egg's VL provides receptors for conspecific sperm to bind, and, to date, at least two sperm receptors have been identified on the surface of sea urchin eggs. Interestingly, however, according to our previous work, denuded sea urchin eggs devoid of the JC and VL do not fail to become fertilized by sperm. Instead, they are bound and penetratedby multiple sperm, raising the possibility that an alternative pathway independent of the VL-residing sperm receptor may be at work. In this research, we studied the roles of the JC and VL using intact and denuded eggs and the synthetic polyamine BPA-C8. BPA-C8 is known to bind to the negatively charged macromolecular complexes in the cells, such as the JC, VL, and the plasma membrane of echinoderm eggs, as well as to the actin filaments in fibroblasts. Our results showed that, when added to seawater, BPA-C8 significantly repressed the Ca2+ wave in the intact P. lividus eggs at fertilization. In eggs deprived of the VL and JC, BPA-C8 binds to the plasma membrane and increases fibrous structures connecting microvilli, thereby allowing the denuded eggs to revert towards monospermy at fertilization. However, the reduced Ca2+ signal in denuded eggs was nullified compared to the intact eggs because removing the JC and VL already decreased the Ca2+ wave. BPA-C8 does not cross the VL and the cell membrane of unfertilized sea urchin eggs to diffuse into the cytoplasm at variance with the fibroblasts. Indeed, the jasplakinolide-induced polymerization of subplasmalemmal actin filaments was inhibited in the eggs microinjected with BPA-C8, but not in the ones bath-incubated with the same dose of BPA-C8.
Collapse
Affiliation(s)
- Nunzia Limatola
- Department of Research Infrastructures for Marine Biological Resources, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| | - Jong Tai Chun
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| | - Jean-Louis Schmitt
- Laboratory of Supramolecular Chemistry, Institut de Science et d'Ingénierie Supramoléculaires ISIS, Université de Strasbourg, 8 Allée Gaspard Monge, 67000 Strasbourg, France
| | - Jean-Marie Lehn
- Laboratory of Supramolecular Chemistry, Institut de Science et d'Ingénierie Supramoléculaires ISIS, Université de Strasbourg, 8 Allée Gaspard Monge, 67000 Strasbourg, France
| | - Luigia Santella
- Department of Research Infrastructures for Marine Biological Resources, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy
| |
Collapse
|
7
|
Gallo G. The Axonal Actin Filament Cytoskeleton: Structure, Function, and Relevance to Injury and Degeneration. Mol Neurobiol 2024; 61:5646-5664. [PMID: 38216856 DOI: 10.1007/s12035-023-03879-7] [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: 10/26/2023] [Accepted: 12/13/2023] [Indexed: 01/14/2024]
Abstract
Early investigations of the neuronal actin filament cytoskeleton gave rise to the notion that, although growth cones exhibit high levels of actin filaments, the axon shaft exhibits low levels of actin filaments. With the development of new tools and imaging techniques, the axonal actin filament cytoskeleton has undergone a renaissance and is now an active field of research. This article reviews the current state of knowledge about the actin cytoskeleton of the axon shaft. The best understood forms of actin filament organization along axons are axonal actin patches and a submembranous system of rings that endow the axon with protrusive competency and structural integrity, respectively. Additional forms of actin filament organization along the axon have also been described and their roles are being elucidated. Extracellular signals regulate the axonal actin filament cytoskeleton and our understanding of the signaling mechanisms involved is being elaborated. Finally, recent years have seen advances in our perspective on how the axonal actin cytoskeleton is impacted by, and contributes to, axon injury and degeneration. The work to date has opened new venues and future research will undoubtedly continue to provide a richer understanding of the axonal actin filament cytoskeleton.
Collapse
Affiliation(s)
- Gianluca Gallo
- Department of Neural Sciences, Shriners Pediatric Research Center, Lewis Katz School of Medicine at Temple University, 3500 North Broad St, Philadelphia, PA, 19140, USA.
| |
Collapse
|
8
|
Fu F, Yu Y, Zou B, Long Y, Wu L, Yin J, Zhou Q. Role of actin-binding proteins in prostate cancer. Front Cell Dev Biol 2024; 12:1430386. [PMID: 39055653 PMCID: PMC11269120 DOI: 10.3389/fcell.2024.1430386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
The molecular mechanisms driving the onset and metastasis of prostate cancer remain poorly understood. Actin, under the control of actin-binding proteins (ABPs), plays a crucial role in shaping the cellular cytoskeleton, which in turn supports the morphological alterations in normal cells, as well as the invasive spread of tumor cells. Previous research indicates that ABPs of various types serve distinct functions, and any disruptions in their activities could predispose individuals to prostate cancer. These ABPs are intricately implicated in the initiation and advancement of prostate cancer through a complex array of intracellular processes, such as severing, linking, nucleating, inducing branching, assembling, facilitating actin filament elongation, terminating elongation, and promoting actin molecule aggregation. As such, this review synthesizes existing literature on several ABPs linked to prostate cancer, including cofilin, filamin A, and fascin, with the aim of shedding light on the molecular mechanisms through which ABPs influence prostate cancer development and identifying potential therapeutic targets. Ultimately, this comprehensive examination seeks to contribute to the understanding and management of prostate diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qing Zhou
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
9
|
Müller L, Keil R, Glaß M, Hatzfeld M. Plakophilin 4 controls the spatio-temporal activity of RhoA at adherens junctions to promote cortical actin ring formation and tissue tension. Cell Mol Life Sci 2024; 81:291. [PMID: 38970683 PMCID: PMC11335210 DOI: 10.1007/s00018-024-05329-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/17/2024] [Accepted: 06/18/2024] [Indexed: 07/08/2024]
Abstract
Plakophilin 4 (PKP4) is a component of cell-cell junctions that regulates intercellular adhesion and Rho-signaling during cytokinesis with an unknown function during epidermal differentiation. Here we show that keratinocytes lacking PKP4 fail to develop a cortical actin ring, preventing adherens junction maturation and generation of tissue tension. Instead, PKP4-depleted cells display increased stress fibers. PKP4-dependent RhoA localization at AJs was required to activate a RhoA-ROCK2-MLCK-MLC2 axis and organize actin into a cortical ring. AJ-associated PKP4 provided a scaffold for the Rho activator ARHGEF2 and the RhoA effectors MLCK and MLC2, facilitating the spatio-temporal activation of RhoA signaling at cell junctions to allow cortical ring formation and actomyosin contraction. In contrast, association of PKP4 with the Rho suppressor ARHGAP23 reduced ARHGAP23 binding to RhoA which prevented RhoA activation in the cytoplasm and stress fiber formation. These data identify PKP4 as an AJ component that transduces mechanical signals into cytoskeletal organization.
Collapse
Affiliation(s)
- Lisa Müller
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120, Halle, Germany.
| | - René Keil
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120, Halle, Germany
| | - Markus Glaß
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120, Halle, Germany
| | - Mechthild Hatzfeld
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120, Halle, Germany
| |
Collapse
|
10
|
Chatterjee P, Mukherjee S, Majumder P. Shaping Drosophila eggs: unveiling the roles of Arpc1 and cpb in morphogenesis. Funct Integr Genomics 2024; 24:120. [PMID: 38960936 DOI: 10.1007/s10142-024-01396-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024]
Abstract
The Drosophila egg chamber (EC) starts as a spherical tissue at the beginning. With maturation, the outer follicle cells of EC collectively migrate in a direction perpendicular to the anterior-posterior axis, to shape EC from spherical to ellipsoidal. Filamentous actin (F-actin) plays a significant role in shaping individual migratory cells to the overall EC shape, like in every cell migration. The primary focus of this article is to unveil the function of different Actin Binding Proteins (ABPs) in regulating mature Drosophila egg shape. We have screened 66 ABPs, and the genetic screening data revealed that individual knockdown of Arp2/3 complex genes and the "capping protein β" (cpb) gene have severely altered the egg phenotype. Arpc1 and cpb RNAi mediated knockdown resulted in the formation of spherical eggs which are devoid of dorsal appendages. Studies also showed the role of Arpc1 and cpb on the number of laid eggs and follicle cell morphology. Furthermore, the depletion of Arpc1 and cpb resulted in a change in F-actin quantity. Together, the data indicate that Arpc1 and cpb regulate Drosophila egg shape, F-actin management, egg-laying characteristics and dorsal appendages formation.
Collapse
Affiliation(s)
- Poulami Chatterjee
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Sandipan Mukherjee
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Pralay Majumder
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India.
| |
Collapse
|
11
|
Cao R, Sun R, Ye Y, Tian P, Huang B, Ye H, Dai L, Lan Z, Liu J, Li L. Low shear stress-induced blockage of autophagic flux impairs endothelial barrier and facilitates atherosclerosis in mice. Exp Cell Res 2024; 439:114071. [PMID: 38729336 DOI: 10.1016/j.yexcr.2024.114071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/25/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024]
Abstract
Atherosclerosis preferentially occurs in areas with low shear stress (LSS) and oscillatory flow. LSS has been demonstrated to correlate with the development of atherosclerosis. The sphingosine 1-phosphate receptor 1 (S1PR1), involving intravascular blood flow sensing, regulates vascular development and vascular barrier function. However, whether LSS affects atherosclerosis via regulating S1PR1 remains incompletely clear. In this study, immunostaining results of F-actin, β-catenin, and VE-cadherin indicated that LSS impaired endothelial barrier function in human umbilical vein endothelial cells (HUVECs). Western blot analysis showed that LSS resulted in blockage of autophagic flux in HUVECs. In addition, autophagy agonist Rapamycin (Rapa) antagonized LSS-induced endothelial barrier dysfunction, whereas autophagic flux inhibitor Bafilomycin A1 (BafA1) exacerbated it, indicating that LSS promoted endothelial barrier dysfunction by triggering autophagic flux blockage. Notably, gene expression analysis revealed that LSS downregulated S1PR1 expression, which was antagonized by Rapa. Selective S1PR1 antagonist W146 impaired endothelial barrier function of HUVECs under high shear stress (HSS) conditions. Moreover, our data showed that expression of GAPARAPL2, a member of autophagy-related gene 8 (Atg8) proteins, was decreased in HUVECs under LSS conditions. Autophagic flux blockage induced by GAPARAPL2 knockdown inhibited S1PR1, aggravated endothelial barrier dysfunction of HUVECs in vitro, and promoted aortic atherosclerosis in ApoE-/- mice in vivo. Our study demonstrates that autophagic flux blockage induced by LSS downregulates S1PR1 expression and impairs endothelial barrier function. GABARAPL2 inhibition is involved in LSS-induced autophagic flux blockage, which impairs endothelial barrier function via downregulation of S1PR1.
Collapse
Affiliation(s)
- Ruhao Cao
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Ruxian Sun
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Yuanzhi Ye
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, China
| | - Pingge Tian
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Bin Huang
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Haowen Ye
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Libing Dai
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Zirong Lan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, China
| | - Jia Liu
- VIP Medical Service Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| | - Li Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China.
| |
Collapse
|
12
|
Zhuo L, Stöckl JB, Fröhlich T, Moser S, Vollmar AM, Zahler S. A Novel Interaction of Slug (SNAI2) and Nuclear Actin. Cells 2024; 13:696. [PMID: 38667311 PMCID: PMC11049500 DOI: 10.3390/cells13080696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Actin is a protein of central importance to many cellular functions. Its localization and activity are regulated by interactions with a high number of actin-binding proteins. In a yeast two-hybrid (Y2H) screening system, snail family transcriptional repressor 2 (SNAI2 or slug) was identified as a yet unknown potential actin-binding protein. We validated this interaction using immunoprecipitation and analyzed the functional relation between slug and actin. Since both proteins have been reported to be involved in DNA double-strand break (DSB) repair, we focused on their interaction during this process after treatment with doxorubicin or UV irradiation. Confocal microscopy elicits that the overexpression of actin fused to an NLS stabilizes complexes of slug and γH2AX, an early marker of DNA damage repair.
Collapse
Affiliation(s)
- Ling Zhuo
- Center for Drug Research, Ludwig-Maximilians-University Munich, Butenandtstr, 5-13, 81377 Munich, Germany; (L.Z.); (A.M.V.)
| | - Jan B. Stöckl
- Laboratory for Functional Genome Analysis, Gene Center Munich, Ludwig-Maximilians-University Munich, Feodor-Lynen-Str. 25, 81377 Munich, Germany; (J.B.S.); (T.F.)
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis, Gene Center Munich, Ludwig-Maximilians-University Munich, Feodor-Lynen-Str. 25, 81377 Munich, Germany; (J.B.S.); (T.F.)
| | - Simone Moser
- Department of Pharmacognosy, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria;
| | - Angelika M. Vollmar
- Center for Drug Research, Ludwig-Maximilians-University Munich, Butenandtstr, 5-13, 81377 Munich, Germany; (L.Z.); (A.M.V.)
| | - Stefan Zahler
- Center for Drug Research, Ludwig-Maximilians-University Munich, Butenandtstr, 5-13, 81377 Munich, Germany; (L.Z.); (A.M.V.)
| |
Collapse
|
13
|
De Franceschi N, Hoogenberg B, Katan A, Dekker C. Engineering ssRNA tile filaments for (dis)assembly and membrane binding. NANOSCALE 2024; 16:4890-4899. [PMID: 38323489 DOI: 10.1039/d3nr06423a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Cytoskeletal protein filaments such as actin and microtubules confer mechanical support to cells and facilitate many cellular functions such as motility and division. Recent years have witnessed the development of a variety of molecular scaffolds that mimic such filaments. Indeed, filaments that are programmable and compatible with biological systems may prove useful in studying or substituting such proteins. Here, we explore the use of ssRNA tiles to build and modify filaments in vitro. We engineer a number of functionalities that are crucial to the function of natural proteins filaments into the ssRNA tiles, including the abilities to assemble or disassemble filaments, to tune the filament stiffness, to induce membrane binding, and to bind proteins. This work paves the way for building dynamic cytoskeleton-mimicking systems made out of rationally designed ssRNA tiles that can be transcribed in natural or synthetic cells.
Collapse
Affiliation(s)
- Nicola De Franceschi
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, The Netherlands.
| | - Baukje Hoogenberg
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, The Netherlands.
| | - Allard Katan
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, The Netherlands.
| | - Cees Dekker
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, The Netherlands.
| |
Collapse
|
14
|
Qiao Y, Ji X, Guo H, Zheng W, Yao W. Complementary transcriptomic and proteomic analyses elucidate the toxicological molecular mechanisms of deoxynivalenol-induced contractile dysfunction in enteric smooth muscle cells. Food Chem Toxicol 2024; 186:114545. [PMID: 38403181 DOI: 10.1016/j.fct.2024.114545] [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: 01/02/2024] [Revised: 02/18/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
Deoxynivalenol (DON) is one of the frequent Fusarium mycotoxins and poses a serious threat to public health worldwide. DON-induced weight loss is tightly connected with its ability to decrease feed intake by influencing gastrointestinal tract (GIT) motility. Our previous reports indicated that DON interfered with intestinal motility by injuring the contractility of enteric smooth muscle cells (SMC). Here, we further explored the potential mechanisms by employing a complementary method of transcriptomics and proteomics using the porcine enteric smooth muscle cell line (PISMC) as an experimental model. The transcriptomic and proteomic data uncover that the expression of numerous extracellular matrix (ECM) proteins and multiple integrin subunits were downregulated in PISMC under DON exposure, suppressing the ECM-integrin receptor interaction and its mediated signaling. Furthermore, DON treatment could depress actin polymerization, as reflected by the upregulated expression of Rho GTPase-activating proteins and cofilin in PISMC. Meanwhile, the expression levels of downstream contractile apparatus genes were significantly inhibited after challenge with DON. Taken together, the current results suggest that DON inhibits enteric SMC contractility by regulating the ECM-integrin-actin polymerization signaling pathway. Our findings provide novel insights into the potential mechanisms behind the DON toxicological effects in the GIT of humans and animals.
Collapse
Affiliation(s)
- Yu Qiao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, 230031, China
| | - Xu Ji
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, 230031, China
| | - Huiduo Guo
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212018, China
| | - Weijiang Zheng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wen Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Key Lab of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
15
|
Zhao X, Zhang Y, Wu F, Li X, Guo S, Li X. MeCP2-Induced Alternations of Transcript Levels and m6A Methylation in Human Retinal Pigment Epithelium Cells. ACS OMEGA 2023; 8:47964-47973. [PMID: 38144074 PMCID: PMC10734004 DOI: 10.1021/acsomega.3c06610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/05/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023]
Abstract
MeCP2 is a transcriptional regulator that is involved in epithelial-mesenchymal transition (EMT) and is highly expressed in proliferative vitreoretinopathy. m6A methylation is a critical post-transcriptional regulation in eukaryotic cells. However, the connection between MeCP2 and m6A methylation has not been revealed in retinal pigment epithelium (RPE), and the regulatory role of MeCP2 at the post-transcriptional level in an m6A-dependent manner is rarely investigated. In this study, we used sequencing to reveal differences in transcript levels and m6A abundance of individual genes in RPE cells after treatment with human recombinant protein MeCP2. The biological functions and processes of differential genes were further analyzed by bioinformatics. The results exhibited that after MeCP2 treatment, 65 genes were up-regulated and 43 genes were down-regulated at the transcription level, and 4 peaks were hypermethylated and 9,041 peaks were hypomethylated at the m6A modification level. Enrichment analysis found that differentially expressed genes were associated with organic acid metabolism, melanogenesis, and vascular smooth muscle contraction. In addition, differentially methylated genes were related to cell junction, RNA processing and metabolism, cell activity, actin cytoskeleton, and several signaling pathways associated with EMT. Further conjoint analysis indicated that the transcription and m6A levels of the EGR1, ELOVL2, and SFR1 genes were altered, and EGR1 is an essential transcription factor in the EMT process. The RNA levels and m6A levels of the three genes were verified by qPCR and m6A-IP-qPCR, respectively. Overall, this study preliminarily revealed the differential mapping of MeCP2-induced m6A modifications, which contributes to the study of the epigenetic and EMT mechanism in RPE cells.
Collapse
Affiliation(s)
- Xueru Zhao
- Henan
Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Zhengzhou
University People’s Hospital, 450000 Zhengzhou, China
- People’s
Hospital of Henan University, 450003 Zhengzhou, China
- Eye
Institute, Henan Academy of Innovations
in Medical Science, 450000 Zhengzhou, China
| | - Yongya Zhang
- Henan
Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Zhengzhou
University People’s Hospital, 450000 Zhengzhou, China
| | - Fei Wu
- Henan
Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Zhengzhou
University People’s Hospital, 450000 Zhengzhou, China
| | - Xue Li
- Henan
Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Zhengzhou
University People’s Hospital, 450000 Zhengzhou, China
- People’s
Hospital of Henan University, 450003 Zhengzhou, China
- Eye
Institute, Henan Academy of Innovations
in Medical Science, 450000 Zhengzhou, China
| | - Sibei Guo
- Henan
Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Xinxiang
Medical University Henan Provincial People’s Hospital, 453003 Xinxiang, China
| | - Xiaohua Li
- Henan
Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Zhengzhou
University People’s Hospital, 450000 Zhengzhou, China
- People’s
Hospital of Henan University, 450003 Zhengzhou, China
- Eye
Institute, Henan Academy of Innovations
in Medical Science, 450000 Zhengzhou, China
| |
Collapse
|
16
|
Loescher CM, Freundt JK, Unger A, Hessel AL, Kühn M, Koser F, Linke WA. Titin governs myocardial passive stiffness with major support from microtubules and actin and the extracellular matrix. NATURE CARDIOVASCULAR RESEARCH 2023; 2:991-1002. [PMID: 39196092 PMCID: PMC11358001 DOI: 10.1038/s44161-023-00348-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 09/19/2023] [Indexed: 08/29/2024]
Abstract
Myocardial passive stiffness is crucial for the heart's pump function and is determined by mechanical elements, including the extracellular matrix and cytoskeletal filaments; however, their individual contributions are controversially discussed and difficult to quantify. In this study, we targeted the cytoskeletal filaments in a mouse model, which enables the specific, acute and complete cleavage of the sarcomeric titin springs. We show in vitro that each cytoskeletal filament's stiffness contribution varies depending on whether the elastic or the viscous forces are considered and on strain level. Titin governs myocardial elastic forces, with the largest contribution provided at both low and high strain. Viscous force contributions are more uniformly distributed among the microtubules, titin and actin. The extracellular matrix contributes at high strain. The remaining forces after total target element disruption are likely derived from desmin filaments. Our findings answer longstanding questions about cardiac mechanical architecture and allow better targeting of passive myocardial stiffness in heart failure.
Collapse
Affiliation(s)
| | - Johanna K Freundt
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Andreas Unger
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Anthony L Hessel
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Michel Kühn
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Franziska Koser
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Wolfgang A Linke
- Institute of Physiology II, University of Muenster, Muenster, Germany.
| |
Collapse
|
17
|
Oosterheert W, Blanc FEC, Roy A, Belyy A, Sanders MB, Hofnagel O, Hummer G, Bieling P, Raunser S. Molecular mechanisms of inorganic-phosphate release from the core and barbed end of actin filaments. Nat Struct Mol Biol 2023; 30:1774-1785. [PMID: 37749275 PMCID: PMC10643162 DOI: 10.1038/s41594-023-01101-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/18/2023] [Indexed: 09/27/2023]
Abstract
The release of inorganic phosphate (Pi) from actin filaments constitutes a key step in their regulated turnover, which is fundamental to many cellular functions. The mechanisms underlying Pi release from the core and barbed end of actin filaments remain unclear. Here, using human and bovine actin isoforms, we combine cryo-EM with molecular-dynamics simulations and in vitro reconstitution to demonstrate how actin releases Pi through a 'molecular backdoor'. While constantly open at the barbed end, the backdoor is predominantly closed in filament-core subunits and opens only transiently through concerted amino acid rearrangements. This explains why Pi escapes rapidly from the filament end but slowly from internal subunits. In a nemaline-myopathy-associated actin variant, the backdoor is predominantly open in filament-core subunits, resulting in accelerated Pi release and filaments with drastically shortened ADP-Pi caps. Our results provide the molecular basis for Pi release from actin and exemplify how a disease-linked mutation distorts the nucleotide-state distribution and atomic structure of the filament.
Collapse
Affiliation(s)
- Wout Oosterheert
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Florian E C Blanc
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Ankit Roy
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Alexander Belyy
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Micaela Boiero Sanders
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Oliver Hofnagel
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
- Institute for Biophysics, Goethe University, Frankfurt am Main, Germany.
| | - Peter Bieling
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| |
Collapse
|
18
|
The great escape - how inorganic phosphate is released from actin filaments. Nat Struct Mol Biol 2023; 30:1621-1622. [PMID: 37775570 DOI: 10.1038/s41594-023-01102-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2023]
|
19
|
Quadri R, Rotondo G, Sertic S, Pozzi S, dell’Oca MC, Guerrini L, Muzi-Falconi M. A Haspin-ARHGAP11A axis regulates epithelial morphogenesis through Rho-ROCK dependent modulation of LIMK1-Cofilin. iScience 2023; 26:108011. [PMID: 37841592 PMCID: PMC10570125 DOI: 10.1016/j.isci.2023.108011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/20/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
Throughout mitosis, a plethora of processes must be efficiently concerted to ensure cell proliferation and tissue functionality. The mitotic spindle does not only mediate chromosome segregation, but also defines the axis of cellular division, thus determining tissue morphology. Functional spindle orientation relies on precise actin dynamics, shaped in mitosis by the LIMK1-Cofilin axis. The kinase Haspin acts as a guardian of faithful chromosome segregation that ensures amphitelic chromosome attachment and prevents unscheduled cohesin cleavage. Here, we report an unprecedented role for Haspin in the determination of spindle orientation in mitosis. We show that, during mitosis, Haspin regulates Rho-ROCK activity through ARHGAP11A, a poorly characterized GAP, and that ROCK is in turn responsible for the mitotic activation of LIMK1 and stabilization of the actin cytoskeleton, thus supporting a functional spindle orientation. By exploiting 3D cell cultures, we show that this pathway is pivotal for the establishment of a morphologically functional tissue.
Collapse
Affiliation(s)
- Roberto Quadri
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Giuseppe Rotondo
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Sarah Sertic
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Sara Pozzi
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | | | - Luisa Guerrini
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Marco Muzi-Falconi
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| |
Collapse
|
20
|
Huang D, Chen S, Xiong D, Wang H, Zhu L, Wei Y, Li Y, Zou S. Mitochondrial Dynamics: Working with the Cytoskeleton and Intracellular Organelles to Mediate Mechanotransduction. Aging Dis 2023; 14:1511-1532. [PMID: 37196113 PMCID: PMC10529762 DOI: 10.14336/ad.2023.0201] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/01/2023] [Indexed: 05/19/2023] Open
Abstract
Cells are constantly exposed to various mechanical environments; therefore, it is important that they are able to sense and adapt to changes. It is known that the cytoskeleton plays a critical role in mediating and generating extra- and intracellular forces and that mitochondrial dynamics are crucial for maintaining energy homeostasis. Nevertheless, the mechanisms by which cells integrate mechanosensing, mechanotransduction, and metabolic reprogramming remain poorly understood. In this review, we first discuss the interaction between mitochondrial dynamics and cytoskeletal components, followed by the annotation of membranous organelles intimately related to mitochondrial dynamic events. Finally, we discuss the evidence supporting the participation of mitochondria in mechanotransduction and corresponding alterations in cellular energy conditions. Notable advances in bioenergetics and biomechanics suggest that the mechanotransduction system composed of mitochondria, the cytoskeletal system, and membranous organelles is regulated through mitochondrial dynamics, which may be a promising target for further investigation and precision therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuyu Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shujuan Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Bai Y, Zhao F, Wu T, Chen F, Pang X. Actin polymerization and depolymerization in developing vertebrates. Front Physiol 2023; 14:1213668. [PMID: 37745245 PMCID: PMC10515290 DOI: 10.3389/fphys.2023.1213668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Development is a complex process that occurs throughout the life cycle. F-actin, a major component of the cytoskeleton, is essential for the morphogenesis of tissues and organs during development. F-actin is formed by the polymerization of G-actin, and the dynamic balance of polymerization and depolymerization ensures proper cellular function. Disruption of this balance results in various abnormalities and defects or even embryonic lethality. Here, we reviewed recent findings on the structure of G-actin and F-actin and the polymerization of G-actin to F-actin. We also focused on the functions of actin isoforms and the underlying mechanisms of actin polymerization/depolymerization in cellular and organic morphogenesis during development. This information will extend our understanding of the role of actin polymerization in the physiologic or pathologic processes during development and may open new avenues for developing therapeutics for embryonic developmental abnormalities or tissue regeneration.
Collapse
Affiliation(s)
- Yang Bai
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Feng Zhao
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tingting Wu
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Fangchun Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xiaoxiao Pang
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
22
|
Steffensen KE, Dawson JF. Actin's C-terminus coordinates actin structural changes and functions. Cytoskeleton (Hoboken) 2023; 80:313-329. [PMID: 37036084 DOI: 10.1002/cm.21757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/17/2023] [Accepted: 03/30/2023] [Indexed: 04/11/2023]
Abstract
Actin is essential to eukaryotic cellular processes. Actin's C-terminus appears to play a direct role in modulating actin's structure and properties, facilitating the binding and function of actin-binding proteins (ABPs). The structural and functional characterization of filamentous actin's C-terminus has been impeded by its inherent flexibility, as well as actin's resistance to crystallization for x-ray diffraction and the historical resolution constraints associated with electron microscopy. Many biochemical studies have established that actin's C-terminus must retain its flexibility and structural integrity to modulate actin's structure and functions. For example, C-terminal structural changes are known to affect nucleotide binding and exchange, as well as propagate actin structural changes throughout extensive allosteric networks, facilitating the binding and function of ABPs. Advances in electron microscopy have resulted in high-resolution structures of filamentous actin, providing insights into subtle structural changes that are mediated by actin's C-terminus. Here, we review existing knowledge establishing the importance of actin's C-terminus within actin structural changes and functions and discuss how modern structural characterization techniques provide the tools to understand the role of actin's C-terminus in cellular processes.
Collapse
Affiliation(s)
- Karl E Steffensen
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - John F Dawson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
23
|
Ahmad M, Weiswald LB, Poulain L, Denoyelle C, Meryet-Figuiere M. Involvement of lncRNAs in cancer cells migration, invasion and metastasis: cytoskeleton and ECM crosstalk. J Exp Clin Cancer Res 2023; 42:173. [PMID: 37464436 PMCID: PMC10353155 DOI: 10.1186/s13046-023-02741-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023] Open
Abstract
Cancer is the main cause of death worldwide and metastasis is a major cause of poor prognosis and cancer-associated mortality. Metastatic conversion of cancer cells is a multiplex process, including EMT through cytoskeleton remodeling and interaction with TME. Tens of thousands of putative lncRNAs have been identified, but the biological functions of most are still to be identified. However, lncRNAs have already emerged as key regulators of gene expression at transcriptional and post-transcriptional level to control gene expression in a spatio-temporal fashion. LncRNA-dependent mechanisms can control cell fates during development and their perturbed expression is associated with the onset and progression of many diseases including cancer. LncRNAs have been involved in each step of cancer cells metastasis through different modes of action. The investigation of lncRNAs different roles in cancer metastasis could possibly lead to the identification of new biomarkers and innovative cancer therapeutic options.
Collapse
Affiliation(s)
- Mohammad Ahmad
- (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Baclesse Cancer Centre, Université de Caen Normandie Inserm Anticipe UMR 1086, Normandie Univ, Research Building, F-14000 François 3 Avenue Général Harris, BP 45026, 14 076, cedex 05, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
- Biochemistry Division, Chemistry Department, Faculty of Science, Damanhour University, Damanhour, 14000, Egypt
| | - Louis-Bastien Weiswald
- (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Baclesse Cancer Centre, Université de Caen Normandie Inserm Anticipe UMR 1086, Normandie Univ, Research Building, F-14000 François 3 Avenue Général Harris, BP 45026, 14 076, cedex 05, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Laurent Poulain
- (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Baclesse Cancer Centre, Université de Caen Normandie Inserm Anticipe UMR 1086, Normandie Univ, Research Building, F-14000 François 3 Avenue Général Harris, BP 45026, 14 076, cedex 05, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Christophe Denoyelle
- (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Baclesse Cancer Centre, Université de Caen Normandie Inserm Anticipe UMR 1086, Normandie Univ, Research Building, F-14000 François 3 Avenue Général Harris, BP 45026, 14 076, cedex 05, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Matthieu Meryet-Figuiere
- (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Baclesse Cancer Centre, Université de Caen Normandie Inserm Anticipe UMR 1086, Normandie Univ, Research Building, F-14000 François 3 Avenue Général Harris, BP 45026, 14 076, cedex 05, Caen, France.
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France.
| |
Collapse
|
24
|
Jin L, Ma J, Chen Z, Wang F, Li Z, Shang Z, Dong J. Osteoarthritis related epigenetic variations in miRNA expression and DNA methylation. BMC Med Genomics 2023; 16:163. [PMID: 37434153 PMCID: PMC10337191 DOI: 10.1186/s12920-023-01597-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 07/01/2023] [Indexed: 07/13/2023] Open
Abstract
Osteoarthritis (OA) is chronic arthritis characterized by articular cartilage degradation. However, a comprehensive regulatory network for OA-related microRNAs and DNA methylation modifications has yet to be established. Thus, we aimed to identify epigenetic changes in microRNAs and DNA methylation and establish the regulatory network between miRNAs and DNA methylation. The mRNA, miRNA, and DNA methylation expression profiles of healthy or osteoarthritis articular cartilage samples were downloaded from Gene Expression Omnibus (GEO) database, including GSE169077, GSE175961, and GSE162484. The differentially expressed genes (DEGs), differentially expressed miRNAs (DEMs), and differentially methylated genes (DMGs) were analyzed by the online tool GEO2R. DAVID and STRING databases were applied for functional enrichment analysis and protein-protein interaction (PPI) network. Potential therapeutic compounds for the treatment of OA were identified by Connectivity map (CMap) analysis. A total of 1424 up-regulated DEGs, 1558 down-regulated DEGs, 5 DEMs with high expression, 6 DEMs with low expression, 1436 hypermethylated genes, and 455 hypomethylated genes were selected. A total of 136 up-regulated and 65 downregulated genes were identified by overlapping DEGs and DEMs predicted target genes which were enriched in apoptosis and circadian rhythm. A total of 39 hypomethylated and 117 hypermethylated genes were obtained by overlapping DEGs and DMGs, which were associated with ECM receptor interactions and cellular metabolic processes, cell connectivity, and transcription. Moreover, The PPI network showed COL5A1, COL6A1, LAMA4, T3GAL6A, and TP53 were the most connective proteins. After overlapping of DEGs, DMGs and DEMs predicted targeted genes, 4 up-regulated genes and 11 down-regulated genes were enriched in the Axon guidance pathway. The top ten genes ranked by PPI network connectivity degree in the up-regulated and downregulated overlapping genes of DEGs and DMGs were further analyzed by the CMap database, and nine chemicals were predicted as potential drugs for the treatment of OA. In conclusion, TP53, COL5A1, COL6A1, LAMA4, and ST3GAL6 may play important roles in OA genesis and development.
Collapse
Affiliation(s)
- Lingpeng Jin
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Jun Ma
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Zhen Chen
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Fei Wang
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Zhikuan Li
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Ziqi Shang
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Jiangtao Dong
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China.
| |
Collapse
|
25
|
Voronkina A, Romanczuk-Ruszuk E, Przekop RE, Lipowicz P, Gabriel E, Heimler K, Rogoll A, Vogt C, Frydrych M, Wienclaw P, Stelling AL, Tabachnick K, Tsurkan D, Ehrlich H. Honeycomb Biosilica in Sponges: From Understanding Principles of Unique Hierarchical Organization to Assessing Biomimetic Potential. Biomimetics (Basel) 2023; 8:234. [PMID: 37366830 DOI: 10.3390/biomimetics8020234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Structural bioinspiration in modern material science and biomimetics represents an actual trend that was originally based on the bioarchitectural diversity of invertebrate skeletons, specifically, honeycomb constructs of natural origin, which have been in humanities focus since ancient times. We conducted a study on the principles of bioarchitecture regarding the unique biosilica-based honeycomb-like skeleton of the deep-sea glass sponge Aphrocallistes beatrix. Experimental data show, with compelling evidence, the location of actin filaments within honeycomb-formed hierarchical siliceous walls. Principles of the unique hierarchical organization of such formations are discussed. Inspired by poriferan honeycomb biosilica, we designed diverse models, including 3D printing, using PLA-, resin-, and synthetic-glass-prepared corresponding microtomography-based 3D reconstruction.
Collapse
Affiliation(s)
- Alona Voronkina
- Pharmacy Department, National Pirogov Memorial Medical University, Vinnytsya, Pyrogov str. 56, 21018 Vinnytsia, Ukraine
- Institute of Electronics and Sensor Materials, TU Bergakademie Freiberg, Gustav-Zeuner Str. 3, 09599 Freiberg, Germany
| | - Eliza Romanczuk-Ruszuk
- Faculty of Mechanical Engineering, Institute of Biomedical Engineering, Bialystok University of Technology, Wiejska Str. 45C, 15-351 Bialystok, Poland
| | - Robert E Przekop
- Center for Advanced Technology, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 10, 61-614 Poznan, Poland
| | - Pawel Lipowicz
- Faculty of Mechanical Engineering, Institute of Biomedical Engineering, Bialystok University of Technology, Wiejska Str. 45C, 15-351 Bialystok, Poland
| | - Ewa Gabriel
- Center for Advanced Technology, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 10, 61-614 Poznan, Poland
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, 8 Uniwersytetu Poznańskiego, 61-614 Poznan, Poland
| | - Korbinian Heimler
- Institute of Analytical Chemistry, TU Bergakademie Freiberg, Leipziger Str. 29, 09599 Freiberg, Germany
| | - Anika Rogoll
- Institute of Analytical Chemistry, TU Bergakademie Freiberg, Leipziger Str. 29, 09599 Freiberg, Germany
| | - Carla Vogt
- Institute of Analytical Chemistry, TU Bergakademie Freiberg, Leipziger Str. 29, 09599 Freiberg, Germany
| | - Milosz Frydrych
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, 8 Uniwersytetu Poznańskiego, 61-614 Poznan, Poland
| | - Pawel Wienclaw
- Faculty of Physics, University of Warsaw, Pasteura 7, 02-093 Warsaw, Poland
| | - Allison L Stelling
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080, USA
| | - Konstantin Tabachnick
- International Institute of Biomineralogy GmbH, Am St.-Niclas Schacht 13, 09599 Freiberg, Germany
| | - Dmitry Tsurkan
- Institute of Electronics and Sensor Materials, TU Bergakademie Freiberg, Gustav-Zeuner Str. 3, 09599 Freiberg, Germany
| | - Hermann Ehrlich
- Center for Advanced Technology, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 10, 61-614 Poznan, Poland
| |
Collapse
|
26
|
Ostalé CM, Vega-Cuesta P, González T, López-Varea A, de Celis JF. RNAi screen in the Drosophila wing of genes encoding proteins related to cytoskeleton organization and cell division. Dev Biol 2023; 498:61-76. [PMID: 37015290 DOI: 10.1016/j.ydbio.2023.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/04/2023]
Abstract
Cell division and cytoskeleton organization are fundamental processes participating in the development of Drosophila imaginal discs. In this manuscript we describe the phenotypes in the adult fly wing generated by knockdowns of 85% of Drosophila genes encoding proteins likely related to the regulation of cell division and cytoskeleton organization. We also compile a molecular classification of these proteins into classes that describe their expected or known main biochemical characteristics, as well as mRNA expression in the wing disc and likely protein subcellular localization for a subset of these genes. Finally, we analyze in more detail one protein family of cytoskeleton genes (Arp2/3 complex), and define the consequences of interfering with cell division for wing growth and patterning.
Collapse
|
27
|
Nakamura M, Hui J, Parkhurst SM. Bending actin filaments: twists of fate. Fac Rev 2023; 12:7. [PMID: 37081903 PMCID: PMC10111394 DOI: 10.12703/r/12-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023] Open
Abstract
In many cellular contexts, intracellular actomyosin networks must generate directional forces to carry out cellular tasks such as migration and endocytosis, which play important roles during normal developmental processes. A number of different actin binding proteins have been identified that form linear or branched actin, and that regulate these filaments through activities such as bundling, crosslinking, and depolymerization to create a wide variety of functional actin assemblies. The helical nature of actin filaments allows them to better accommodate tensile stresses by untwisting, as well as to bend to great curvatures without breaking. Interestingly, this latter property, the bending of actin filaments, is emerging as an exciting new feature for determining dynamic actin configurations and functions. Indeed, recent studies using in vitro assays have found that proteins including IQGAP, Cofilin, Septins, Anillin, α-Actinin, Fascin, and Myosins-alone or in combination-can influence the bending or curvature of actin filaments. This bending increases the number and types of dynamic assemblies that can be generated, as well as the spectrum of their functions. Intriguingly, in some cases, actin bending creates directionality within a cell, resulting in a chiral cell shape. This actin-dependent cell chirality is highly conserved in vertebrates and invertebrates and is essential for cell migration and breaking L-R symmetry of tissues/organs. Here, we review how different types of actin binding protein can bend actin filaments, induce curved filament geometries, and how they impact on cellular functions.
Collapse
Affiliation(s)
- Mitsutoshi Nakamura
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA 98109
| | - Justin Hui
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA 98109
| | - Susan M Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA 98109
| |
Collapse
|
28
|
Biomolecular condensation involving the cytoskeleton. Brain Res Bull 2023; 194:105-117. [PMID: 36690162 DOI: 10.1016/j.brainresbull.2023.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/07/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Biomolecular condensation of proteins contributes to the organization of the cytoplasm and nucleoplasm. A number of condensation processes appear to be directly involved in regulating the structure, function and dynamics of the cytoskeleton. Liquid-liquid phase separation of cytoskeleton proteins, together with polymerization modulators, promotes cytoskeletal fiber nucleation and branching. Furthermore, the attachment of protein condensates to the cytoskeleton can contribute to cytoskeleton stability and organization, regulate transport, create patterns of functional reaction containers, and connect the cytoskeleton with membranes. Surface-bound condensates can exert and buffer mechanical forces that give stability and flexibility to the cytoskeleton, thus, may play a large role in cell biology. In this review, we introduce the concept and role of cellular biomolecular condensation, explain its special function on cytoskeletal fiber surfaces, and point out potential definition and experimental caveats. We review the current literature on protein condensation processes related to the actin, tubulin, and intermediate filament cytoskeleton, and discuss some of them in the context of neurobiology. In summary, we provide an overview about biomolecular condensation in relation to cytoskeleton structure and function, which offers a base for the exploration and interpretation of cytoskeletal condensates in neurobiology.
Collapse
|
29
|
Deciphering the molecular mechanisms of actin cytoskeleton regulation in cell migration using cryo-EM. Biochem Soc Trans 2023; 51:87-99. [PMID: 36695514 PMCID: PMC9987995 DOI: 10.1042/bst20220221] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/26/2023]
Abstract
The actin cytoskeleton plays a key role in cell migration and cellular morphodynamics in most eukaryotes. The ability of the actin cytoskeleton to assemble and disassemble in a spatiotemporally controlled manner allows it to form higher-order structures, which can generate forces required for a cell to explore and navigate through its environment. It is regulated not only via a complex synergistic and competitive interplay between actin-binding proteins (ABP), but also by filament biochemistry and filament geometry. The lack of structural insights into how geometry and ABPs regulate the actin cytoskeleton limits our understanding of the molecular mechanisms that define actin cytoskeleton remodeling and, in turn, impact emerging cell migration characteristics. With the advent of cryo-electron microscopy (cryo-EM) and advanced computational methods, it is now possible to define these molecular mechanisms involving actin and its interactors at both atomic and ultra-structural levels in vitro and in cellulo. In this review, we will provide an overview of the available cryo-EM methods, applicable to further our understanding of the actin cytoskeleton, specifically in the context of cell migration. We will discuss how these methods have been employed to elucidate ABP- and geometry-defined regulatory mechanisms in initiating, maintaining, and disassembling cellular actin networks in migratory protrusions.
Collapse
|
30
|
Rajan S, Terman JR, Reisler E. MICAL-mediated oxidation of actin and its effects on cytoskeletal and cellular dynamics. Front Cell Dev Biol 2023; 11:1124202. [PMID: 36875759 PMCID: PMC9982024 DOI: 10.3389/fcell.2023.1124202] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/02/2023] [Indexed: 02/19/2023] Open
Abstract
Actin and its dynamic structural remodelings are involved in multiple cellular functions, including maintaining cell shape and integrity, cytokinesis, motility, navigation, and muscle contraction. Many actin-binding proteins regulate the cytoskeleton to facilitate these functions. Recently, actin's post-translational modifications (PTMs) and their importance to actin functions have gained increasing recognition. The MICAL family of proteins has emerged as important actin regulatory oxidation-reduction (Redox) enzymes, influencing actin's properties both in vitro and in vivo. MICALs specifically bind to actin filaments and selectively oxidize actin's methionine residues 44 and 47, which perturbs filaments' structure and leads to their disassembly. This review provides an overview of the MICALs and the impact of MICAL-mediated oxidation on actin's properties, including its assembly and disassembly, effects on other actin-binding proteins, and on cells and tissue systems.
Collapse
Affiliation(s)
- Sudeepa Rajan
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jonathan R. Terman
- Departments of Neuroscience and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Emil Reisler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
31
|
Zhang X, Han J, Fan D, Wang J, Lin X, Zhang H, Zhang C, Bai J, Huang H, Gu Y. Lysine-40 succinylation of TAGLN2 induces glioma angiogenesis and tumor growth through regulating TMSB4X. Cancer Gene Ther 2023; 30:172-181. [PMID: 36131066 DOI: 10.1038/s41417-022-00534-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/12/2022] [Accepted: 09/06/2022] [Indexed: 01/19/2023]
Abstract
Protein lysine succinylation (Ksucc) represents an important regulatory mechanism of tumor development. In this work, the difference of protein Ksucc between HCMEC/D3 co-cultured with U87 (glioma endothelia cells, GEC) and without U87 (normal endothelia cells, NEC) was investigated using TMT labeling and affinity enrichment followed by high-resolution LC-MS/MS analysis. Interestingly, TAGLN2 was highly succinylated at K40 in GEC (15.36 folds vs. NEC). Compared to the Vector group, TAGLN2WT and a succinylation-mimetic TAGLN2K40E greatly promoted the angiogenesis of glioma in vitro and in vivo. Furthermore, the adhesion and metastasis of U87 co-cultured with GEC in the TAGLN2WT or TAGLN2K40E group were also significantly promoted. This was consistent with the increased expression of VE-cadherin and actin cytoskeleton remodeling induced by TAGLN2 K40succ in GEC. In addition, high K40succ of TAGLN2 was associated with poor prognosis in patients with glioma. Overexpression of TAGLN2K40E also markedly promoted the proliferation and migration of glioma cells, further analysis of in vivo xenograft tumors showed that there was a significant decrease in tumor size and angiogenesis in the TAGLN2K40R group. Notably, the co-localization of TMSB4X and TAGLN2 mainly in the nucleus and cytoplasm of glioma cells was detected by immunofluorescence staining. We identified TMSB4X as a potential target of TAGLN2, which was proved to interact with TAGLN2WT rather than TAGLN2K40A. And the inhibition of TMSB4X could markedly attenuate the proliferation and migration of glioma cells induced by TAGLN2 K40succ. The results revealed K40succ of TAGLN2 could be a novelty diagnosis and therapeutic target for gliomas.
Collapse
Affiliation(s)
- Xiaoyi Zhang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Jin Han
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Di Fan
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, P. R. China
| | - Jiahong Wang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Xiangdan Lin
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, P. R. China
| | - Hong Zhang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Cai Zhang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Jialing Bai
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Hailan Huang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China
| | - Yanting Gu
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, P. R. China.
| |
Collapse
|
32
|
Jeong M, Jung E, Oh S, Shin SY. Homeobox Protein PROX1 Expression is Negatively Regulated by Histone Deacetylase 1 and c-JUN Complex in MDA-MB-231 Human Breast Cancer Cells. Folia Biol (Praha) 2023; 69:81-90. [PMID: 38206773 DOI: 10.14712/fb2023069030081] [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] [Indexed: 01/13/2024]
Abstract
Prospero homeobox 1 (PROX1) is a member of the homeobox transcription factor family that plays a critical role in the development of multiple tissues and specification of cell fate. PROX1 expression is differentially regulated based on the cellular context and plays an antagonistic role as a tumour promoter or suppressor in different tumour types. In human breast cancer, PROX1 expression is suppress-ed; however, the molecular mechanism by which it is down-regulated remains poorly understood. Here, we show that ectopic expression of PROX1 reduces the motility and invasiveness of MDA-MB-231 human breast cancer cells, suggesting that PROX1 functions as a negative regulator of tumour invasion in MDA-MB-231 cells. Treatment with histone deacetylase (HDAC) inhibitors up-regulates PROX1 mRNA and protein expression levels. Knockdown of HDAC1 using short hairpin RNA also up-regulates PROX1 mRNA and protein expression levels. We found that HDAC1 interacted with c-JUN at the activator protein (AP)-1-binding site located at -734 to -710 in the PROX1 promoter region to suppress PROX1 expression. In addition, c-JUN N-terminal kinase-mediated c-JUN phosphorylation was found to be crucial for silencing PROX1 expression. In conclusion, PROX1 expression can be silenced by the epigenetic mechanism involved in the complex formation of HDAC1 and c-JUN at the AP-1 site in the PROX1 promoter region in MDA-MB-231 human breast cancer cells. Therefore, this study revealed the epigenetic regulatory mechanism involved in the suppression of PROX1 expression in breast cancer cells.
Collapse
Affiliation(s)
- Munki Jeong
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Euitaek Jung
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Sukjin Oh
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Soon Young Shin
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
33
|
Li Y, Wang D, Ge H, Güngör C, Gong X, Chen Y. Cytoskeletal and Cytoskeleton-Associated Proteins: Key Regulators of Cancer Stem Cell Properties. Pharmaceuticals (Basel) 2022; 15:1369. [PMID: 36355541 PMCID: PMC9698833 DOI: 10.3390/ph15111369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/02/2022] [Accepted: 11/06/2022] [Indexed: 08/08/2023] Open
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer cells possessing stemness characteristics that are closely associated with tumor proliferation, recurrence and resistance to therapy. Recent studies have shown that different cytoskeletal components and remodeling processes have a profound impact on the behavior of CSCs. In this review, we outline the different cytoskeletal components regulating the properties of CSCs and discuss current and ongoing therapeutic strategies targeting the cytoskeleton. Given the many challenges currently faced in targeted cancer therapy, a deeper comprehension of the molecular events involved in the interaction of the cytoskeleton and CSCs will help us identify more effective therapeutic strategies to eliminate CSCs and ultimately improve patient survival.
Collapse
Affiliation(s)
- Yuqiang Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Dan Wang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Heming Ge
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Cenap Güngör
- Department of General Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Xuejun Gong
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yongheng Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
34
|
Oosterheert W, Klink BU, Belyy A, Pospich S, Raunser S. Structural basis of actin filament assembly and aging. Nature 2022; 611:374-379. [DOI: 10.1038/s41586-022-05241-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/16/2022] [Indexed: 12/12/2022]
Abstract
AbstractThe dynamic turnover of actin filaments (F-actin) controls cellular motility in eukaryotes and is coupled to changes in the F-actin nucleotide state1–3. It remains unclear how F-actin hydrolyses ATP and subsequently undergoes subtle conformational rearrangements that ultimately lead to filament depolymerization by actin-binding proteins. Here we present cryo-electron microscopy structures of F-actin in all nucleotide states, polymerized in the presence of Mg2+ or Ca2+ at approximately 2.2 Å resolution. The structures show that actin polymerization induces the relocation of water molecules in the nucleotide-binding pocket, activating one of them for the nucleophilic attack of ATP. Unexpectedly, the back door for the subsequent release of inorganic phosphate (Pi) is closed in all structures, indicating that Pi release occurs transiently. The small changes in the nucleotide-binding pocket after ATP hydrolysis and Pi release are sensed by a key amino acid, amplified and transmitted to the filament periphery. Furthermore, differences in the positions of water molecules in the nucleotide-binding pocket explain why Ca2+-actin shows slower polymerization rates than Mg2+-actin. Our work elucidates the solvent-driven rearrangements that govern actin filament assembly and aging and lays the foundation for the rational design of drugs and small molecules for imaging and therapeutic applications.
Collapse
|
35
|
Anti-PTK7 Monoclonal Antibodies Exhibit Anti-Tumor Activity at the Cellular Level and in Mouse Xenograft Models of Esophageal Squamous Cell Carcinoma. Int J Mol Sci 2022; 23:ijms232012195. [PMID: 36293051 PMCID: PMC9603586 DOI: 10.3390/ijms232012195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/24/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
PTK7 is a catalytically defective receptor protein tyrosine kinase upregulated in various cancers, including esophageal squamous cell carcinoma (ESCC). In previous studies, we observed a positive correlation between PTK7 expression levels and tumorigenicity in various ESCC cell lines and xenograft mice with ESCC KYSE-30 cells. In this study, we analyzed the effects of anti-PTK7 monoclonal antibodies (mAbs) on the tumorigenic activity in KYSE-30 cells and in mouse xenograft models. PTK7 mAb-32 and mAb-43 bind with a high affinity to the extracellular domain of PTK7. PTK7 mAbs significantly reduced three-dimensional cell proliferation, adhesion, wound healing, and migration. PTK7 mAbs also reduce chemotactic invasiveness by decreasing MMP-9 secretion. PTK7 mAbs decreased actin cytoskeleton levels in the cortical region of KYSE-30 cells. PTK7 mAbs reduced the phosphorylation of ERK, SRC, and FAK. In a mouse xenograft model of ESCC using KYSE-30 cells, PTK7 mAbs reduced tumor growth in terms of volume, weight, and the number of Ki-67-positive cells. These results demonstrated that PTK7 mAbs can inhibit the tumorigenicity of ESCC at the cellular level and in vivo by blocking the function of PTK7. Considering the anticancer activities of PTK7 mAbs, we propose that PTK7 mAbs can be used in an effective treatment strategy for PTK7-positive malignancies, such as ESCC.
Collapse
|
36
|
Alqassim SS. Functional Mimicry of Eukaryotic Actin Assembly by Pathogen Effector Proteins. Int J Mol Sci 2022; 23:ijms231911606. [PMID: 36232907 PMCID: PMC9569871 DOI: 10.3390/ijms231911606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
The actin cytoskeleton lies at the heart of many essential cellular processes. There are hundreds of proteins that cells use to control the size and shape of actin cytoskeletal networks. As such, various pathogens utilize different strategies to hijack the infected eukaryotic host actin dynamics for their benefit. These include the control of upstream signaling pathways that lead to actin assembly, control of eukaryotic actin assembly factors, encoding toxins that distort regular actin dynamics, or by encoding effectors that directly interact with and assemble actin filaments. The latter class of effectors is unique in that, quite often, they assemble actin in a straightforward manner using novel sequences, folds, and molecular mechanisms. The study of these mechanisms promises to provide major insights into the fundamental determinants of actin assembly, as well as a deeper understanding of host-pathogen interactions in general, and contribute to therapeutic development efforts targeting their respective pathogens. This review discusses mechanisms and highlights shared and unique features of actin assembly by pathogen effectors that directly bind and assemble actin, focusing on eukaryotic actin nucleator functional mimics Rickettsia Sca2 (formin mimic), Burkholderia BimA (Ena/VASP mimic), and Vibrio VopL (tandem WH2-motif mimic).
Collapse
Affiliation(s)
- Saif S Alqassim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14, Dubai Health Care City, Dubai P.O. Box 505055, United Arab Emirates
| |
Collapse
|
37
|
Bhandary M, Sales Conniff A, Miranda K, Heller LC. Acute Effects of Intratumor DNA Electrotransfer. Pharmaceutics 2022; 14:pharmaceutics14102097. [PMID: 36297532 PMCID: PMC9611921 DOI: 10.3390/pharmaceutics14102097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/14/2022] Open
Abstract
Intratumor therapeutic DNA electroporation or electrotransfer is in clinical trials in the United States and is under development in many other countries. Acute changes in endogenous gene expression in response to DNA or to pulse application may significantly modulate the therapeutic efficacy of the expressed proteins. Oligonucleotide arrays were used in this study to quantify changes in mRNA expression in B16-F10 mouse melanoma tumors four hours after DNA electrotransfer. The data were subjected to the DAVID v6.8 web server for functional annotation to reveal regulated genes and genetic pathways. Gene ontology analysis revealed several molecular functions related to cytoskeletal remodeling and inflammatory signaling. In B16-F10 cells, F-actin remodeling was confirmed by phalloidin staining in cells that received pulse application alone or in the presence of DNA. Chemokine secretion was confirmed in cells receiving DNA electrotransfer. These results indicate that pulse application alone or in the presence of DNA may modulate the therapeutic efficacy of therapeutic DNA electrotransfer.
Collapse
|
38
|
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
|
39
|
Stelling-Férez J, Gabaldón JA, Nicolás FJ. Oleanolic acid stimulation of cell migration involves a biphasic signaling mechanism. Sci Rep 2022; 12:15065. [PMID: 36064555 PMCID: PMC9445025 DOI: 10.1038/s41598-022-17553-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 07/27/2022] [Indexed: 11/09/2022] Open
Abstract
Cell migration is a critical process for wound healing, a physiological phenomenon needed for proper skin restoration after injury. Wound healing can be compromised under pathological conditions. Natural bioactive terpenoids have shown promising therapeutic properties in wound healing. Oleanolic acid (OA), a triterpenoid, enhances in vitro and in vivo cell migration. However, the underlying signaling mechanisms and pathways triggered by OA are poorly understood. We have previously shown that OA activates epidermal growth factor receptor (EGFR) and downstream effectors such as mitogen-activated protein (MAP) kinase cascade and c-Jun N-terminal kinase (JNK), leading to c-Jun transcription factor phosphorylation, all of which are involved in migration. We performed protein expression or migration front protein subcellular localization assays, which showed that OA induces c-Jun activation and its nuclear translocation, which precisely overlaps at wound-edge cells. Furthermore, c-Jun phosphorylation was independent of EGFR activation. Additionally, OA promoted actin cytoskeleton and focal adhesion (FA) dynamization. In fact, OA induced the recruitment of regulator proteins to FAs to dynamize these structures during migration. Moreover, OA changed paxillin distribution and activated focal adhesion kinase (FAK) at focal adhesions (FAs). The molecular implications of these observations are discussed.
Collapse
Affiliation(s)
- Javier Stelling-Férez
- Department of Nutrition and Food Technology, Health Sciences PhD Program, Universidad Católica de Murcia (UCAM), Campus de los Jerónimos nº135, Guadalupe, 30107, Murcia, Spain.,Regeneration, Molecular Oncology and TGF-ß, Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain
| | - José Antonio Gabaldón
- Department of Nutrition and Food Technology, Health Sciences PhD Program, Universidad Católica de Murcia (UCAM), Campus de los Jerónimos nº135, Guadalupe, 30107, Murcia, Spain
| | - Francisco José Nicolás
- Regeneration, Molecular Oncology and TGF-ß, Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain.
| |
Collapse
|
40
|
Schneider J, Jasnin M. Capturing actin assemblies in cells using in situ cryo-electron tomography. Eur J Cell Biol 2022; 101:151224. [PMID: 35500467 DOI: 10.1016/j.ejcb.2022.151224] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 11/21/2022] Open
Abstract
Actin contributes to an exceptionally wide range of cellular processes through the assembly and disassembly of highly dynamic and ordered structures. Visualizing these structures in cells can help us understand how the molecular players of the actin machinery work together to produce force-generating systems. In recent years, cryo-electron tomography (cryo-ET) has become the method of choice for structural analysis of the cell interior at the molecular scale. Here we review advances in cryo-ET workflows that have enabled this transformation, especially the automation of sample preparation procedures, data collection, and processing. We discuss new structural analyses of dynamic actin assemblies in cryo-preserved cells, which have provided mechanistic insights into actin assembly and function at the nanoscale. Finally, we highlight the latest visual proteomics studies of actin filaments and their interactors reaching sub-nanometer resolutions in cells.
Collapse
Affiliation(s)
- Jonathan Schneider
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Marion Jasnin
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany.
| |
Collapse
|
41
|
Structure of Arp2/3 complex at a branched actin filament junction resolved by single-particle cryo-electron microscopy. Proc Natl Acad Sci U S A 2022; 119:e2202723119. [PMID: 35622886 DOI: 10.1073/pnas.2202723119] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
SignificanceActin filament nucleation by Arp2/3 complex must be triggered by activators like WASP family proteins. Understanding how WASP proteins activate Arp2/3 complex has been a major challenge due to a lack of high-resolution structures of the complex in an activated state. We determined a high-resolution (∼3.9 Å) structure of the WASP-activated Arp2/3 complex at a branch junction and used biochemical, cell biological, and molecular dynamic simulations to understand the mechanism of WASP-mediated activation. This work shows in detail the contacts between the fully activated Arp2/3 complex, the nucleated daughter actin filament, and the mother actin filament and provides important insights into how conformational rearrangements in the Arp2/3 complex are stimulated during activation.
Collapse
|
42
|
Daly CA, Hall ET, Ogden SK. Regulatory mechanisms of cytoneme-based morphogen transport. Cell Mol Life Sci 2022; 79:119. [PMID: 35119540 PMCID: PMC8816744 DOI: 10.1007/s00018-022-04148-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 01/07/2023]
Abstract
During development and tissue homeostasis, cells must communicate with their neighbors to ensure coordinated responses to instructional cues. Cues such as morphogens and growth factors signal at both short and long ranges in temporal- and tissue-specific manners to guide cell fate determination, provide positional information, and to activate growth and survival responses. The precise mechanisms by which such signals traverse the extracellular environment to ensure reliable delivery to their intended cellular targets are not yet clear. One model for how this occurs suggests that specialized filopodia called cytonemes extend between signal-producing and -receiving cells to function as membrane-bound highways along which information flows. A growing body of evidence supports a crucial role for cytonemes in cell-to-cell communication. Despite this, the molecular mechanisms by which cytonemes are initiated, how they grow, and how they deliver specific signals are only starting to be revealed. Herein, we discuss recent advances toward improved understanding of cytoneme biology. We discuss similarities and differences between cytonemes and other types of cellular extensions, summarize what is known about how they originate, and discuss molecular mechanisms by which their activity may be controlled in development and tissue homeostasis. We conclude by highlighting important open questions regarding cytoneme biology, and comment on how a clear understanding of their function may provide opportunities for treating or preventing disease.
Collapse
Affiliation(s)
- Christina A Daly
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl. MS340, Memphis, TN, 38105, USA
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, MS 1500, Memphis, TN, 38105, USA
| | - Eric T Hall
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl. MS340, Memphis, TN, 38105, USA
| | - Stacey K Ogden
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl. MS340, Memphis, TN, 38105, USA.
| |
Collapse
|
43
|
Illescas M, Peñas A, Arenas J, Martín MA, Ugalde C. Regulation of Mitochondrial Function by the Actin Cytoskeleton. Front Cell Dev Biol 2022; 9:795838. [PMID: 34993202 PMCID: PMC8725978 DOI: 10.3389/fcell.2021.795838] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/03/2021] [Indexed: 12/13/2022] Open
Abstract
The regulatory role of actin cytoskeleton on mitochondrial function is a growing research field, but the underlying molecular mechanisms remain poorly understood. Specific actin-binding proteins (ABPs), such as Gelsolin, have also been shown to participate in the pathophysiology of mitochondrial OXPHOS disorders through yet to be defined mechanisms. In this mini-review, we will summarize the experimental evidence supporting the fundamental roles of actin cytoskeleton and ABPs on mitochondrial trafficking, dynamics, biogenesis, metabolism and apoptosis, with a particular focus on Gelsolin involvement in mitochondrial disorders. The functional interplay between the actin cytoskeleton, ABPs and mitochondrial membranes for the regulation of cellular homeostasis thus emerges as a new exciting field for future research and therapeutic approaches.
Collapse
Affiliation(s)
- María Illescas
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Ana Peñas
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Joaquín Arenas
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Miguel A Martín
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Cristina Ugalde
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| |
Collapse
|
44
|
Koronfel M, Kounatidis I, Mwangangi DM, Vyas N, Okolo C, Jadhav A, Fish T, Chotchuang P, Schulte A, Robinson RC, Harkiolaki M. Correlative cryo-imaging of the cellular universe with soft X-rays and laser light used to track F-actin structures in mammalian cells. Acta Crystallogr D Struct Biol 2021; 77:1479-1485. [PMID: 34866605 PMCID: PMC8647181 DOI: 10.1107/s2059798321010329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/06/2021] [Indexed: 11/16/2022] Open
Abstract
Imaging of actin filaments is crucial due to the integral role that they play in many cellular functions such as intracellular transport, membrane remodelling and cell motility. Visualizing actin filaments has so far relied on fluorescence microscopy and electron microscopy/tomography. The former lacks the capacity to capture the overall local ultrastructure, while the latter requires rigorous sample preparation that can lead to potential artefacts, and only delivers relatively small volumes of imaging data at the thinnest areas of a cell. In this work, a correlative approach utilizing in situ super-resolution fluorescence imaging and cryo X-ray tomography was used to image bundles of actin filaments deep inside cells under near-native conditions. In this case, fluorescence 3D imaging localized the actin bundles within the intracellular space, while X-ray tomograms of the same areas provided detailed views of the local ultrastructure. Using this new approach, actin trails connecting vesicles in the perinuclear area and hotspots of actin presence within and around multivesicular bodies were observed. The characteristic prevalence of filamentous actin in cytoplasmic extensions was also documented.
Collapse
Affiliation(s)
- Mohamed Koronfel
- Beamline B24, Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, United Kingdom
| | - Ilias Kounatidis
- Beamline B24, Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, United Kingdom
| | - Dennis M. Mwangangi
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore 138673, Singapore
| | - Nina Vyas
- Beamline B24, Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, United Kingdom
| | - Chidinma Okolo
- Beamline B24, Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, United Kingdom
| | - Archana Jadhav
- Beamline B24, Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, United Kingdom
| | - Tom Fish
- Beamline B24, Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, United Kingdom
| | - Phatcharin Chotchuang
- Research Institute for Interdisciplinary Science (RIIS), Okayama University, Okayama 700-8530, Japan
| | - Albert Schulte
- Research Institute for Interdisciplinary Science (RIIS), Okayama University, Okayama 700-8530, Japan
| | - Robert C. Robinson
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore 138673, Singapore
- Research Institute for Interdisciplinary Science (RIIS), Okayama University, Okayama 700-8530, Japan
- School of Biomolecular Science and Engineering (BSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Maria Harkiolaki
- Beamline B24, Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, United Kingdom
| |
Collapse
|
45
|
Pospich S, Sweeney HL, Houdusse A, Raunser S. High-resolution structures of the actomyosin-V complex in three nucleotide states provide insights into the force generation mechanism. eLife 2021; 10:e73724. [PMID: 34812732 PMCID: PMC8735999 DOI: 10.7554/elife.73724] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
The molecular motor myosin undergoes a series of major structural transitions during its force-producing motor cycle. The underlying mechanism and its coupling to ATP hydrolysis and actin binding are only partially understood, mostly due to sparse structural data on actin-bound states of myosin. Here, we report 26 high-resolution cryo-EM structures of the actomyosin-V complex in the strong-ADP, rigor, and a previously unseen post-rigor transition state that binds the ATP analog AppNHp. The structures reveal a high flexibility of myosin in each state and provide valuable insights into the structural transitions of myosin-V upon ADP release and binding of AppNHp, as well as the actomyosin interface. In addition, they show how myosin is able to specifically alter the structure of F-actin.
Collapse
Affiliation(s)
- Sabrina Pospich
- Department of Structural Biochemistry, Max Planck Institute of Molecular PhysiologyDortmundGermany
| | - H Lee Sweeney
- Department of Pharmacology and Therapeutics and the Myology Institute, University of FloridaGainesvilleUnited States
| | - Anne Houdusse
- Structural Motility, Institut Curie, Centre National de la Recherche ScientifiqueParisFrance
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular PhysiologyDortmundGermany
| |
Collapse
|
46
|
Nasufović V, Küllmer F, Bößneck J, Dahse H, Görls H, Bellstedt P, Stallforth P, Arndt H. Total Synthesis and Bioactivity Mapping of Geodiamolide H. Chemistry 2021; 27:11633-11642. [PMID: 34032329 PMCID: PMC8453818 DOI: 10.1002/chem.202100989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Indexed: 01/24/2023]
Abstract
The first total synthesis of the actin-stabilizing marine natural product geodiamolide H was achieved. Solid-phase based peptide assembly paired with scalable stereoselective syntheses of polyketide building blocks and an optimized esterification set the stage for investigating the key ring-closing metathesis. Geodiamolide H and synthetic analogues were characterized for their toxicity and for antiproliferative effects in cellulo, by characterising actin polymerization induction in vitro, and by docking on the F-actin target and property computation in silico, for a better understanding of structure-activity relationships (SAR). A non-natural analogue of geodiamolide H was discovered to be most potent in the series, suggesting significant potential for tool compound design.
Collapse
Affiliation(s)
- Veselin Nasufović
- Institut für Organische Chemie und Makromolekulare ChemieFriedrich-Schiller-Universität (FSU)Humboldtstr. 1007743JenaGermany
| | - Florian Küllmer
- Institut für Organische Chemie und Makromolekulare ChemieFriedrich-Schiller-Universität (FSU)Humboldtstr. 1007743JenaGermany
| | - Johanna Bößneck
- Institut für Organische Chemie und Makromolekulare ChemieFriedrich-Schiller-Universität (FSU)Humboldtstr. 1007743JenaGermany
| | - Hans‐Martin Dahse
- Abteilungen Infektionsbiologie und PaläobiotechnologieLeibniz-Institut für Naturstoffforschung – Hans-Knöll-InstitutBeutenbergstr. 11a07745JenaGermany
| | - Helmar Görls
- Institut für Anorganische und Analytische ChemieFriedrich-Schiller-Universität (FSU)Humboldtstr. 807743JenaGermany
| | - Peter Bellstedt
- NMR-PlattformFriedrich-Schiller-Universität (FSU)Humboldtstr. 1007743JenaGermany
| | - Pierre Stallforth
- Abteilungen Infektionsbiologie und PaläobiotechnologieLeibniz-Institut für Naturstoffforschung – Hans-Knöll-InstitutBeutenbergstr. 11a07745JenaGermany
| | - Hans‐Dieter Arndt
- Institut für Organische Chemie und Makromolekulare ChemieFriedrich-Schiller-Universität (FSU)Humboldtstr. 1007743JenaGermany
| |
Collapse
|
47
|
Rabaglino MB, O’Doherty A, Bojsen-Møller Secher J, Lonergan P, Hyttel P, Fair T, Kadarmideen HN. Application of multi-omics data integration and machine learning approaches to identify epigenetic and transcriptomic differences between in vitro and in vivo produced bovine embryos. PLoS One 2021; 16:e0252096. [PMID: 34029343 PMCID: PMC8143403 DOI: 10.1371/journal.pone.0252096] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/09/2021] [Indexed: 01/16/2023] Open
Abstract
Pregnancy rates for in vitro produced (IVP) embryos are usually lower than for embryos produced in vivo after ovarian superovulation (MOET). This is potentially due to alterations in their trophectoderm (TE), the outermost layer in physical contact with the maternal endometrium. The main objective was to apply a multi-omics data integration approach to identify both temporally differentially expressed and differentially methylated genes (DEG and DMG), between IVP and MOET embryos, that could impact TE function. To start, four and five published transcriptomic and epigenomic datasets, respectively, were processed for data integration. Second, DEG from day 7 to days 13 and 16 and DMG from day 7 to day 17 were determined in the TE from IVP vs. MOET embryos. Third, genes that were both DE and DM were subjected to hierarchical clustering and functional enrichment analysis. Finally, findings were validated through a machine learning approach with two additional datasets from day 15 embryos. There were 1535 DEG and 6360 DMG, with 490 overlapped genes, whose expression profiles at days 13 and 16 resulted in three main clusters. Cluster 1 (188) and Cluster 2 (191) genes were down-regulated at day 13 or day 16, respectively, while Cluster 3 genes (111) were up-regulated at both days, in IVP embryos compared to MOET embryos. The top enriched terms were the KEGG pathway "focal adhesion" in Cluster 1 (FDR = 0.003), and the cellular component: "extracellular exosome" in Cluster 2 (FDR<0.0001), also enriched in Cluster 1 (FDR = 0.04). According to the machine learning approach, genes in Cluster 1 showed a similar expression pattern between IVP and less developed (short) MOET conceptuses; and between MOET and DKK1-treated (advanced) IVP conceptuses. In conclusion, these results suggest that early conceptuses derived from IVP embryos exhibit epigenomic and transcriptomic changes that later affect its elongation and focal adhesion, impairing post-transfer survival.
Collapse
Affiliation(s)
- Maria B. Rabaglino
- Quantitative Genetics, Bioinformatics and Computational Biology Group, Department of Applied Mathematics and Computer Science, Technical University of Denmark, Lyngby, Denmark
| | - Alan O’Doherty
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Jan Bojsen-Møller Secher
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Patrick Lonergan
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Poul Hyttel
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Trudee Fair
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Haja N. Kadarmideen
- Quantitative Genetics, Bioinformatics and Computational Biology Group, Department of Applied Mathematics and Computer Science, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
48
|
Nishimura Y, Shi S, Zhang F, Liu R, Takagi Y, Bershadsky AD, Viasnoff V, Sellers JR. The formin inhibitor SMIFH2 inhibits members of the myosin superfamily. J Cell Sci 2021; 134:237818. [PMID: 33589498 PMCID: PMC8121067 DOI: 10.1242/jcs.253708] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 02/03/2021] [Indexed: 12/31/2022] Open
Abstract
The small molecular inhibitor of formin FH2 domains, SMIFH2, is widely used in cell biological studies. It inhibits formin-driven actin polymerization in vitro, but not polymerization of pure actin. It is active against several types of formin from different species. Here, we found that SMIFH2 inhibits retrograde flow of myosin 2 filaments and contraction of stress fibers. We further checked the effect of SMIFH2 on non-muscle myosin 2A and skeletal muscle myosin 2 in vitro, and found that SMIFH2 inhibits activity of myosin ATPase and the ability to translocate actin filaments in the gliding actin in vitro motility assay. Inhibition of non-muscle myosin 2A in vitro required a higher concentration of SMIFH2 compared with that needed to inhibit retrograde flow and stress fiber contraction in cells. We also found that SMIFH2 inhibits several other non-muscle myosin types, including bovine myosin 10, Drosophila myosin 7a and Drosophila myosin 5, more efficiently than it inhibits formins. These off-target inhibitions demand additional careful analysis in each case when solely SMIFH2 is used to probe formin functions. This article has an associated First Person interview with Yukako Nishimura, joint first author of the paper.
Collapse
Affiliation(s)
- Yukako Nishimura
- Mechanobiology Institute (MBI), National University of Singapore, Singapore 117411, Singapore
| | - Shidong Shi
- Mechanobiology Institute (MBI), National University of Singapore, Singapore 117411, Singapore
| | - Fang Zhang
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rong Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yasuharu Takagi
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander D Bershadsky
- Mechanobiology Institute (MBI), National University of Singapore, Singapore 117411, Singapore.,Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Virgile Viasnoff
- Mechanobiology Institute (MBI), National University of Singapore, Singapore 117411, Singapore.,CNRS UMI 3639 BMC, Singapore 117411, Singapore.,Department of Biological Sciences, National university of Singapore, Singapore 117558, Singapore
| | - James R Sellers
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
49
|
Pospich S, Küllmer F, Nasufović V, Funk J, Belyy A, Bieling P, Arndt H, Raunser S. Cryo‐EM Resolves Molecular Recognition Of An Optojasp Photoswitch Bound To Actin Filaments In Both Switch States. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202013193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Sabrina Pospich
- Department of Structural Biochemistry Max Planck Institute of Molecular Physiology Otto-Hahn-Str. 11 44227 Dortmund Germany
| | - Florian Küllmer
- Institute of Organic Chemistry and Macromolecular Chemistry Friedrich-Schiller-University Humboldtstr. 10 07743 Jena Germany
| | - Veselin Nasufović
- Institute of Organic Chemistry and Macromolecular Chemistry Friedrich-Schiller-University Humboldtstr. 10 07743 Jena Germany
| | - Johanna Funk
- Department of Systemic Cell Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Str. 11 44227 Dortmund Germany
| | - Alexander Belyy
- Department of Structural Biochemistry Max Planck Institute of Molecular Physiology Otto-Hahn-Str. 11 44227 Dortmund Germany
| | - Peter Bieling
- Department of Systemic Cell Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Str. 11 44227 Dortmund Germany
| | - Hans‐Dieter Arndt
- Institute of Organic Chemistry and Macromolecular Chemistry Friedrich-Schiller-University Humboldtstr. 10 07743 Jena Germany
| | - Stefan Raunser
- Department of Structural Biochemistry Max Planck Institute of Molecular Physiology Otto-Hahn-Str. 11 44227 Dortmund Germany
| |
Collapse
|
50
|
The multiple roles of actin-binding proteins at invadopodia. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021. [PMID: 33962752 DOI: 10.1016/bs.ircmb.2021.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Invadopodia are actin-rich membrane protrusions that facilitate cancer cell dissemination by focusing on proteolytic activity and clearing paths for migration through physical barriers, such as basement membranes, dense extracellular matrices, and endothelial cell junctions. Invadopodium formation and activity require spatially and temporally regulated changes in actin filament organization and dynamics. About three decades of research have led to a remarkable understanding of how these changes are orchestrated by sequential recruitment and coordinated activity of different sets of actin-binding proteins. In this chapter, we provide an update on the roles of the actin cytoskeleton during the main stages of invadopodium development with a particular focus on actin polymerization machineries and production of pushing forces driving extracellular matrix remodeling.
Collapse
|