151
|
Melo PN, Souza da Silveira M, Mendes Pinto I, Relvas JB. Morphofunctional programming of microglia requires distinct roles of type II myosins. Glia 2021; 69:2717-2738. [PMID: 34329508 DOI: 10.1002/glia.24067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 11/05/2022]
Abstract
The ramified morphology of microglia and the dynamics of their membrane protrusions are essential for their functions in central nervous system development, homeostasis, and disease. Although their ability to change and control shape critically depends on the actin and actomyosin cytoskeleton, the underlying regulatory mechanisms remain largely unknown. In this study, we systematically analyzed the actomyosin cytoskeleton and regulators downstream of the small GTPase RhoA in the control of microglia shape and function. Our results reveal that (i) Myh9 controls cortical tension levels and affects microglia protrusion formation, (ii) cofilin-mediated maintenance of actin turnover regulates microglia protrusion extension, and (iii) Myh10 influences microglia inflammatory activation. Overall we uncover molecular pathways that regulate microglia morphology and identify type-II myosins as important regulators of microglia biology with differential roles in the control of cell shape (Myh9) and functions (Myh10).
Collapse
Affiliation(s)
- Pedro Neves Melo
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal.,Graduate Programme in Areas of Basic and Applied Biology (GABBA), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Mariana Souza da Silveira
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal.,Instituto de Biofísica Carlos Chagas Filho (IBCCF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Inês Mendes Pinto
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal.,Life Sciences, International Iberian Nanotechnology Laboratory (INL), Braga, Portugal
| | - João Bettencourt Relvas
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal.,Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
152
|
Saito R, Satoh H, Aoba K, Hirasawa H, Miwa N. Dicalcin suppresses in vitro trophoblast attachment in human cell lines. Biochem Biophys Res Commun 2021; 570:206-213. [PMID: 34311201 DOI: 10.1016/j.bbrc.2021.07.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 01/02/2023]
Abstract
Implantation is a highly organized process that involves an interaction between a competent blastocyst and a receptive uterus. Despite significant research efforts, the molecular mechanisms governing this complex process remain elusive. Here, we investigated the effect of dicalcin, an S100-like Ca2+-binding protein, on the attachment of choriocarcinoma cells (BeWo cells) onto a monolayer of endometrial carcinoma cells (Ishikawa cells). Extracellularly administered dicalcin bound to both BeWo and Ishikawa cells. Pretreatment of BeWo spheroids with dicalcin reduced the attachment ratio of the spheroids onto the monolayer, whereas that of Ishikawa cells showed no apparent change. We identified the partial amino acid sequence of human dicalcin that exhibited maximum suppression for BeWo spheroid attachment. Transmission electron microscopy analysis revealed that the dicalcin-derived peptide caused a dilation of the intercellular junction between BeWo and ISK cells. Peptide treatment of BeWo spheroids downregulated the expression of integrinαvβ3 in BeWo cells, and induced alterations in their phalloidin-staining pattern, as measured by the length of each F-actin fiber and the thickness of the cortical stress fiber. Thus, dicalcin affects reorganization of the intracellular actin meshwork and subsequently the intensity of attachment, functioning as a novel suppressor of implantation.
Collapse
Affiliation(s)
- Ryohei Saito
- Department of Obsetrics and Gynecology, Graduate School of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama, Iruma-gun, Saitama, 350-0495, Japan
| | - Hiromasa Satoh
- Department of Physiology, Saitama Medical University, 38 Moro-hongo, Moroyama, Iruma-gun, Saitama, 350-0495, Japan
| | - Kayo Aoba
- Department of Physiology, Saitama Medical University, 38 Moro-hongo, Moroyama, Iruma-gun, Saitama, 350-0495, Japan
| | - Hajime Hirasawa
- Department of Physiology, Saitama Medical University, 38 Moro-hongo, Moroyama, Iruma-gun, Saitama, 350-0495, Japan
| | - Naofumi Miwa
- Department of Physiology, Saitama Medical University, 38 Moro-hongo, Moroyama, Iruma-gun, Saitama, 350-0495, Japan.
| |
Collapse
|
153
|
KRAP tethers IP 3 receptors to actin and licenses them to evoke cytosolic Ca 2+ signals. Nat Commun 2021; 12:4514. [PMID: 34301929 PMCID: PMC8302619 DOI: 10.1038/s41467-021-24739-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/01/2021] [Indexed: 02/06/2023] Open
Abstract
Regulation of IP3 receptors (IP3Rs) by IP3 and Ca2+ allows regenerative Ca2+ signals, the smallest being Ca2+ puffs, which arise from coordinated openings of a few clustered IP3Rs. Cells express thousands of mostly mobile IP3Rs, yet Ca2+ puffs occur at a few immobile IP3R clusters. By imaging cells with endogenous IP3Rs tagged with EGFP, we show that KRas-induced actin-interacting protein (KRAP) tethers IP3Rs to actin beneath the plasma membrane. Loss of KRAP abolishes Ca2+ puffs and the global increases in cytosolic Ca2+ concentration evoked by more intense stimulation. Over-expressing KRAP immobilizes additional IP3R clusters and results in more Ca2+ puffs and larger global Ca2+ signals. Endogenous KRAP determines which IP3Rs will respond: it tethers IP3R clusters to actin alongside sites where store-operated Ca2+ entry occurs, licenses IP3Rs to evoke Ca2+ puffs and global cytosolic Ca2+ signals, implicates the actin cytoskeleton in IP3R regulation and may allow local activation of Ca2+ entry.
Collapse
|
154
|
Flormann DAD, Schu M, Terriac E, Thalla D, Kainka L, Koch M, Gad AKB, Lautenschläger F. A novel universal algorithm for filament network tracing and cytoskeleton analysis. FASEB J 2021; 35:e21582. [PMID: 33835502 DOI: 10.1096/fj.202100048r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/06/2021] [Accepted: 03/25/2021] [Indexed: 01/01/2023]
Abstract
The rapid development of advanced microscopy techniques over recent decades has significantly increased the quality of imaging and our understanding of subcellular structures, such as the organization of the filaments of the cytoskeleton using fluorescence and electron microscopy. However, these recent improvements in imaging techniques have not been matched by similar development of techniques for computational analysis of the images of filament networks that can now be obtained. Hence, for a wide range of applications, reliable computational analysis of such two-dimensional methods remains challenging. Here, we present a new algorithm for tracing of filament networks. This software can extract many important parameters from grayscale images of filament networks, including the mesh hole size, and filament length and connectivity (also known as Coordination Number). In addition, the method allows sub-networks to be distinguished in two-dimensional images using intensity thresholding. We show that the algorithm can be used to analyze images of cytoskeleton networks obtained using different advanced microscopy methods. We have thus developed a new improved method for computational analysis of two-dimensional images of filamentous networks that has wide applications for existing imaging techniques. The algorithm is available as open-source software.
Collapse
Affiliation(s)
- Daniel A D Flormann
- Department of Physics, Saarland University, Saarbruecken, Germany.,INM - Leibniz Institute for New Materials, Saarbruecken, Germany
| | - Moritz Schu
- Department of Physics, Saarland University, Saarbruecken, Germany
| | - Emmanuel Terriac
- INM - Leibniz Institute for New Materials, Saarbruecken, Germany
| | - Divyendu Thalla
- Department of Physics, Saarland University, Saarbruecken, Germany.,INM - Leibniz Institute for New Materials, Saarbruecken, Germany
| | - Lucina Kainka
- Department of Physics, Saarland University, Saarbruecken, Germany.,INM - Leibniz Institute for New Materials, Saarbruecken, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, Saarbruecken, Germany
| | - Annica K B Gad
- Department of Oncology and Metabolism, The Medical School, Weston Park Cancer Centre, Sheffield, UK.,Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
| | - Franziska Lautenschläger
- Department of Physics, Saarland University, Saarbruecken, Germany.,INM - Leibniz Institute for New Materials, Saarbruecken, Germany
| |
Collapse
|
155
|
Kimura K, Motegi F. Fluid flow dynamics in cellular patterning. Semin Cell Dev Biol 2021; 120:3-9. [PMID: 34274213 DOI: 10.1016/j.semcdb.2021.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/24/2021] [Accepted: 07/01/2021] [Indexed: 10/20/2022]
Abstract
The development of complex forms of multicellular organisms depends on the spatial arrangement of cellular architecture and functions. The interior design of the cell is patterned by spatially biased distributions of molecules and biochemical reactions in the cytoplasm and/or on the plasma membrane. In recent years, a dynamic change in the cytoplasmic fluid flow has emerged as a key physical process of driving long-range transport of molecules to particular destinations within the cell. Here, recent experimental advances in the understanding of the generation of the various types of cytoplasmic flows and contributions to intracellular patterning are reviewed with a particular focus on feedback mechanisms between the mechanical properties of fluid flow and biochemical signaling during animal cell polarization.
Collapse
Affiliation(s)
- Kenji Kimura
- School of Science and Technology, Kwansei Gakuin University, Japan.
| | - Fumio Motegi
- Instiute for Genetic Medicine, Hokkaido University, Japan; Temasek Lifesciences Laboratory, Singapore; Mechanobiology Institute, National University of Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore.
| |
Collapse
|
156
|
Schu M, Terriac E, Koch M, Paschke S, Lautenschläger F, Flormann DAD. Scanning electron microscopy preparation of the cellular actin cortex: A quantitative comparison between critical point drying and hexamethyldisilazane drying. PLoS One 2021; 16:e0254165. [PMID: 34234360 PMCID: PMC8263306 DOI: 10.1371/journal.pone.0254165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 06/21/2021] [Indexed: 11/18/2022] Open
Abstract
The cellular cortex is an approximately 200-nm-thick actin network that lies just beneath the cell membrane. It is responsible for the mechanical properties of cells, and as such, it is involved in many cellular processes, including cell migration and cellular interactions with the environment. To develop a clear view of this dense structure, high-resolution imaging is essential. As one such technique, electron microscopy, involves complex sample preparation procedures. The final drying of these samples has significant influence on potential artifacts, like cell shrinkage and the formation of artifactual holes in the actin cortex. In this study, we compared the three most used final sample drying procedures: critical-point drying (CPD), CPD with lens tissue (CPD-LT), and hexamethyldisilazane drying. We show that both hexamethyldisilazane and CPD-LT lead to fewer artifactual mesh holes within the actin cortex than CPD. Moreover, CPD-LT leads to significant reduction in cell height compared to hexamethyldisilazane and CPD. We conclude that the final drying procedure should be chosen according to the reduction in cell height, and so CPD-LT, or according to the spatial separation of the single layers of the actin cortex, and so hexamethyldisilazane.
Collapse
Affiliation(s)
- Moritz Schu
- Leibniz Institute for New Materials (INM), Saarland University, Saarbrücken, Saarland, Germany
- Center for Biophysics, Saarland University, Saarbrücken, Saarland, Germany
| | - Emmanuel Terriac
- Leibniz Institute for New Materials (INM), Saarland University, Saarbrücken, Saarland, Germany
| | - Marcus Koch
- Leibniz Institute for New Materials (INM), Saarland University, Saarbrücken, Saarland, Germany
| | - Stephan Paschke
- Department of General and Visceral Surgery, University Hospital Ulm, Ulm, Baden-Württemberg, Germany
| | - Franziska Lautenschläger
- Leibniz Institute for New Materials (INM), Saarland University, Saarbrücken, Saarland, Germany
- Center for Biophysics, Saarland University, Saarbrücken, Saarland, Germany
| | - Daniel A. D. Flormann
- Leibniz Institute for New Materials (INM), Saarland University, Saarbrücken, Saarland, Germany
- Center for Biophysics, Saarland University, Saarbrücken, Saarland, Germany
- * E-mail:
| |
Collapse
|
157
|
Zhang DF, Yang ZC, Chen JQ, Jin XX, Qiu YD, Chen XJ, Shi HY, Liu ZG, Wang MS, Liang G, Zheng XH. Piperlongumine inhibits migration and proliferation of castration-resistant prostate cancer cells via triggering persistent DNA damage. BMC Complement Med Ther 2021; 21:195. [PMID: 34229670 PMCID: PMC8261967 DOI: 10.1186/s12906-021-03369-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 06/28/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Metastatic castration-resistant prostate cancer (CRPC) is the leading cause of death among men diagnosed with prostate cancer. Piperlongumine (PL) is a novel potential anticancer agent that has been demonstrated to exhibit anticancer efficacy against prostate cancer cells. However, the effects of PL on DNA damage and repair against CRPC have remained unclear. The aim of this study was to further explore the anticancer activity and mechanisms of action of PL against CRPC in terms of DNA damage and repair processes. METHODS The effect of PL on CRPC was evaluated by MTT assay, long-term cell proliferation, reactive oxygen species assay, western blot assay, flow cytometry assay (annexin V/PI staining), β-gal staining assay and DAPI staining assay. The capacity of PL to inhibit the invasion and migration of CRPC cells was assessed by scratch-wound assay, cell adhesion assay, transwell assay and immunofluorescence (IF) assay. The effect of PL on DNA damage and repair was determined via IF assay and comet assay. RESULTS The results showed that PL exhibited stronger anticancer activity against CRPC compared to that of taxol, cisplatin (DDP), doxorubicin (Dox), or 5-Fluorouracil (5-FU), with fewer side effects in normal cells. Importantly, PL treatment significantly decreased cell adhesion to the extracellular matrix and inhibited the migration of CRPC cells through affecting the expression and distribution of focal adhesion kinase (FAK), leading to concentration-dependent inhibition of CRPC cell proliferation and concomitantly increased cell death. Moreover, PL treatment triggered persistent DNA damage and provoked strong DNA damage responses in CRPC cells. CONCLUSION Collectively, our findings demonstrate that PL potently inhibited proliferation, migration, and invasion of CRPC cells and that these potent anticancer effects were potentially achieved via triggering persistent DNA damage in CRPC cells.
Collapse
Affiliation(s)
- Ding-Fang Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Zhi-Chun Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
- The Fifth Affiliated Hospital of Wenzhou Medical University, Affiliated Lishui Hospital of Zhejiang University, The Central Hospital of Zhejiang Lishui, Lishui, 323000, Zhejiang, People's Republic of China
| | - Jian-Qiang Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Xiang-Xiang Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Yin-da Qiu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Xiao-Jing Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Hong-Yi Shi
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Zhi-Guo Liu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Min-Shan Wang
- The Affiliated Xiangshan Hospital, Wenzhou Medical University, Ningbo, 315000, Zhejiang, China
- Hospital of Chinese Medicine of Haishu District, Ningbo, 315000, Zhejiang, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China.
| | - Xiao-Hui Zheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China.
| |
Collapse
|
158
|
Liao M, Liang X, Howard J. The narrowing of dendrite branches across nodes follows a well-defined scaling law. Proc Natl Acad Sci U S A 2021; 118:e2022395118. [PMID: 34215693 PMCID: PMC8271565 DOI: 10.1073/pnas.2022395118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The systematic variation of diameters in branched networks has tantalized biologists since the discovery of da Vinci's rule for trees. Da Vinci's rule can be formulated as a power law with exponent two: The square of the mother branch's diameter is equal to the sum of the squares of those of the daughters. Power laws, with different exponents, have been proposed for branching in circulatory systems (Murray's law with exponent 3) and in neurons (Rall's law with exponent 3/2). The laws have been derived theoretically, based on optimality arguments, but, for the most part, have not been tested rigorously. Using superresolution methods to measure the diameters of dendrites in highly branched Drosophila class IV sensory neurons, we have found that these types of power laws do not hold. In their place, we have discovered a different diameter-scaling law: The cross-sectional area is proportional to the number of dendrite tips supported by the branch plus a constant, corresponding to a minimum diameter of the terminal dendrites. The area proportionality accords with a requirement for microtubules to transport materials and nutrients for dendrite tip growth. The minimum diameter may be set by the force, on the order of a few piconewtons, required to bend membrane into the highly curved surfaces of terminal dendrites. Because the observed scaling differs from Rall's law, we propose that cell biological constraints, such as intracellular transport and protrusive forces generated by the cytoskeleton, are important in determining the branched morphology of these cells.
Collapse
Affiliation(s)
- Maijia Liao
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520
| | - Xin Liang
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520;
| |
Collapse
|
159
|
Inman A, Smutny M. Feeling the force: Multiscale force sensing and transduction at the cell-cell interface. Semin Cell Dev Biol 2021; 120:53-65. [PMID: 34238674 DOI: 10.1016/j.semcdb.2021.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 12/13/2022]
Abstract
A universal principle of all living cells is the ability to sense and respond to mechanical stimuli which is essential for many biological processes. Recent efforts have identified critical mechanosensitive molecules and response pathways involved in mechanotransduction during development and tissue homeostasis. Tissue-wide force transmission and local force sensing need to be spatiotemporally coordinated to precisely regulate essential processes during development such as tissue morphogenesis, patterning, cell migration and organogenesis. Understanding how cells identify and interpret extrinsic forces and integrate a specific response on cell and tissue level remains a major challenge. In this review we consider important cellular and physical factors in control of cell-cell mechanotransduction and discuss their significance for cell and developmental processes. We further highlight mechanosensitive macromolecules that are known to respond to external forces and present examples of how force responses can be integrated into cell and developmental programs.
Collapse
Affiliation(s)
- Angus Inman
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV47AL, UK
| | - Michael Smutny
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV47AL, UK.
| |
Collapse
|
160
|
Laplaud V, Levernier N, Pineau J, Roman MS, Barbier L, Sáez PJ, Lennon-Duménil AM, Vargas P, Kruse K, du Roure O, Piel M, Heuvingh J. Pinching the cortex of live cells reveals thickness instabilities caused by myosin II motors. SCIENCE ADVANCES 2021; 7:eabe3640. [PMID: 34215576 PMCID: PMC11057708 DOI: 10.1126/sciadv.abe3640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 05/20/2021] [Indexed: 06/13/2023]
Abstract
The cell cortex is a contractile actin meshwork, which determines cell shape and is essential for cell mechanics, migration, and division. Because its thickness is below optical resolution, there is a tendency to consider the cortex as a thin uniform two-dimensional layer. Using two mutually attracted magnetic beads, one inside the cell and the other in the extracellular medium, we pinch the cortex of dendritic cells and provide an accurate and time-resolved measure of its thickness. Our observations draw a new picture of the cell cortex as a highly dynamic layer, harboring large fluctuations in its third dimension because of actomyosin contractility. We propose that the cortex dynamics might be responsible for the fast shape-changing capacity of highly contractile cells that use amoeboid-like migration.
Collapse
Affiliation(s)
- Valentin Laplaud
- Physique et Mécanique des Milieux Hétérogènes, ESPCI Paris, PSL University, CNRS, Univ Paris, Sorbonne Université, Paris, France
- Institut Curie and Institut Pierre Gilles de Gennes, PSL University, CNRS, Paris, France
| | - Nicolas Levernier
- Department of Theoretical Physics, University of Geneva, Geneva, Switzerland
| | - Judith Pineau
- Institut Curie, INSERM U932, PSL University, Paris, France
| | | | - Lucie Barbier
- Institut Curie and Institut Pierre Gilles de Gennes, PSL University, CNRS, Paris, France
| | - Pablo J Sáez
- Institut Curie and Institut Pierre Gilles de Gennes, PSL University, CNRS, Paris, France
| | | | - Pablo Vargas
- Institut Curie and Institut Pierre Gilles de Gennes, PSL University, CNRS, Paris, France
| | - Karsten Kruse
- Departments of Biochemistry and Theoretical Physics and NCCR for Chemical Biology, University of Geneva, Geneva 1211, Switzerland
| | - Olivia du Roure
- Physique et Mécanique des Milieux Hétérogènes, ESPCI Paris, PSL University, CNRS, Univ Paris, Sorbonne Université, Paris, France.
| | - Matthieu Piel
- Institut Curie and Institut Pierre Gilles de Gennes, PSL University, CNRS, Paris, France.
| | - Julien Heuvingh
- Physique et Mécanique des Milieux Hétérogènes, ESPCI Paris, PSL University, CNRS, Univ Paris, Sorbonne Université, Paris, France.
| |
Collapse
|
161
|
Pan X, Zhou Y, Hotulainen P, Meunier FA, Wang T. The axonal radial contractility: Structural basis underlying a new form of neural plasticity. Bioessays 2021; 43:e2100033. [PMID: 34145916 DOI: 10.1002/bies.202100033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 12/25/2022]
Abstract
Axons are the longest cellular structure reaching over a meter in the case of human motor axons. They have a relatively small diameter and contain several cytoskeletal elements that mediate both material and information exchange within neurons. Recently, a novel type of axonal plasticity, termed axonal radial contractility, has been unveiled. It is represented by dynamic and transient diameter changes of the axon shaft to accommodate the passages of large organelles. Mechanisms underpinning this plasticity are not fully understood. Here, we first summarised recent evidence of the functional relevance for axon radial contractility, then discussed the underlying structural basis, reviewing nanoscopic evidence of the subtle changes. Two models are proposed to explain how actomyosin rings are organised. Possible roles of non-muscle myosin II (NM-II) in axon degeneration are discussed. Finally, we discuss the concept of periodic functional nanodomains, which could sense extracellular cues and coordinate the axonal responses. Also see the video abstract here: https://youtu.be/ojCnrJ8RCRc.
Collapse
Affiliation(s)
- Xiaorong Pan
- Center for Brain Science, School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Yimin Zhou
- Center for Brain Science, School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Pirta Hotulainen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Tong Wang
- Center for Brain Science, School of Life Science and Technology, Shanghaitech University, Shanghai, China
| |
Collapse
|
162
|
Wohl I, Sherman E. Spectral Analysis of ATP-Dependent Mechanical Vibrations in T Cells. Front Cell Dev Biol 2021; 9:590655. [PMID: 34178972 PMCID: PMC8222795 DOI: 10.3389/fcell.2021.590655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 04/27/2021] [Indexed: 11/13/2022] Open
Abstract
Mechanical vibrations affect multiple cell properties, including its diffusivity, entropy, internal content organization, and thus-function. Here, we used Differential Interference Contrast (DIC), confocal, and Total Internal Reflection Fluorescence (TIRF) microscopies to study mechanical vibrations in live (Jurkat) T cells. Vibrations were measured via the motion of intracellular particles and plasma membrane. These vibrations depend on adenosine triphosphate (ATP) consumption and on Myosin II activity. We then used spectral analysis of these vibrations to distinguish the effects of thermal agitation, ATP-dependent mechanical work and cytoskeletal visco-elasticity. Parameters of spectral analyses could be related to mean square displacement (MSD) analyses with specific advantages in characterizing intracellular mechanical work. We identified two spectral ranges where mechanical work dominated vibrations of intracellular components: 0-3 Hz for intracellular particles and the plasma-membrane, and 100-150 Hz for the plasma-membrane. The 0-3 Hz vibrations of the cell membrane that we measured in an experimental model of immune synapse (IS) are expected to affect the IS formation and function in effector cells. It may also facilitate immunological escape of extensively vibrating malignant cells.
Collapse
Affiliation(s)
| | - Eilon Sherman
- Racah Institute of Physics, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
163
|
Abstract
EPH/EPHRIN signaling is crucial to the segregation of cell populations during the morphogenesis of many tissues. In this issue, Kindberg et al. (2021. J. Cell Biol.https://doi.org/10.1083/jcb.202005216) show that EPH activation can drive both heterotypic cell repulsion and homotypic aggregation by triggering increased cortical tension.
Collapse
|
164
|
Bayir E, Sendemir A. Role of Intermediate Filaments in Blood-Brain Barrier in Health and Disease. Cells 2021; 10:cells10061400. [PMID: 34198868 PMCID: PMC8226756 DOI: 10.3390/cells10061400] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
The blood–brain barrier (BBB) is a highly selective cellular monolayer unique to the microvasculature of the central nervous system (CNS), and it mediates the communication of the CNS with the rest of the body by regulating the passage of molecules into the CNS microenvironment. Limitation of passage of substances through the BBB is mainly due to tight junctions (TJ) and adherens junctions (AJ) between brain microvascular endothelial cells. The importance of actin filaments and microtubules in establishing and maintaining TJs and AJs has been indicated; however, recent studies have shown that intermediate filaments are also important in the formation and function of cell–cell junctions. The most common intermediate filament protein in endothelial cells is vimentin. Vimentin plays a role in blood–brain barrier permeability in both cell–cell and cell–matrix interactions by affecting the actin and microtubule reorganization and by binding directly to VE-cadherin or integrin proteins. The BBB permeability increases due to the formation of stress fibers and the disruption of VE–cadherin interactions between two neighboring cells in various diseases, disrupting the fiber network of intermediate filament vimentin in different ways. Intermediate filaments may be long ignored key targets in regulation of BBB permeability in health and disease.
Collapse
Affiliation(s)
- Ece Bayir
- Ege University Central Research Test and Analysis Laboratory Application and Research Center (EGE-MATAL), Ege University, 35100 Izmir, Turkey;
| | - Aylin Sendemir
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Izmir, Turkey
- Department of Biomedical Technologies, Graduate School of Natural and Applied Science, Ege University, 35100 Izmir, Turkey
- Correspondence: ; Tel.: +90-232-3114817
| |
Collapse
|
165
|
Simao M, Régnier F, Taheraly S, Fraisse A, Tacine R, Fraudeau M, Benabid A, Feuillet V, Lambert M, Delon J, Randriamampita C. cAMP Bursts Control T Cell Directionality by Actomyosin Cytoskeleton Remodeling. Front Cell Dev Biol 2021; 9:633099. [PMID: 34095108 PMCID: PMC8173256 DOI: 10.3389/fcell.2021.633099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/22/2021] [Indexed: 01/23/2023] Open
Abstract
T lymphocyte migration is an essential step to mounting an efficient immune response. The rapid and random motility of these cells which favors their sentinel role is conditioned by chemokines as well as by the physical environment. Morphological changes, underlaid by dynamic actin cytoskeleton remodeling, are observed throughout migration but especially when the cell modifies its trajectory. However, the signaling cascade regulating the directional changes remains largely unknown. Using dynamic cell imaging, we investigated in this paper the signaling pathways involved in T cell directionality. We monitored cyclic adenosine 3′-5′ monosphosphate (cAMP) variation concomitantly with actomyosin distribution upon T lymphocyte migration and highlighted the fact that spontaneous bursts in cAMP starting from the leading edge, are sufficient to promote actomyosin redistribution triggering trajectory modification. Although cAMP is commonly considered as an immunosuppressive factor, our results suggest that, when transient, it rather favors the exploratory behavior of T cells.
Collapse
Affiliation(s)
- Morgane Simao
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Fabienne Régnier
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Sarah Taheraly
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Achille Fraisse
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France.,Master de Biologie, École Normale Supérieure de Lyon, Université Claude Bernard Lyon I, Université de Lyon, Lyon, France
| | - Rachida Tacine
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Marie Fraudeau
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Adam Benabid
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Vincent Feuillet
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Mireille Lambert
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Jérôme Delon
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | | |
Collapse
|
166
|
Schoenwaelder N, Salewski I, Engel N, Krause M, Schneider B, Müller M, Riess C, Lemcke H, Skorska A, Grosse-Thie C, Junghanss C, Maletzki C. The Individual Effects of Cyclin-Dependent Kinase Inhibitors on Head and Neck Cancer Cells-A Systematic Analysis. Cancers (Basel) 2021; 13:cancers13102396. [PMID: 34063457 PMCID: PMC8157193 DOI: 10.3390/cancers13102396] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/07/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022] Open
Abstract
Cyclin-dependent kinase inhibitors (CDKi´s) display cytotoxic activity against different malignancies, including head and neck squamous cell carcinomas (HNSCC). By coordinating the DNA damage response, these substances may be combined with cytostatics to enhance cytotoxicity. Here, we investigated the influence of different CDKi´s (palbociclib, dinaciclib, THZ1) on two HNSCC cell lines in monotherapy and combination therapy with clinically-approved drugs (5-FU, Cisplatin, cetuximab). Apoptosis/necrosis, cell cycle, invasiveness, senescence, radiation-induced γ-H2AX DNA double-strand breaks, and effects on the actin filament were studied. Furthermore, the potential to increase tumor immunogenicity was assessed by analyzing Calreticulin translocation and immune relevant surface markers. Finally, an in vivo mouse model was used to analyze the effect of dinaciclib and Cisplatin combination therapy. Dinaciclib, palbociclib, and THZ1 displayed anti-neoplastic activity after low-dose treatment, while the two latter substances slightly enhanced radiosensitivity. Dinaciclib decelerated wound healing, decreased invasiveness, and induced MHC-I, accompanied by high amounts of surface-bound Calreticulin. Numbers of early and late apoptotic cells increased initially (24 h), while necrosis dominated afterward. Antitumoral effects of the selective CDKi palbociclib were weaker, but combinations with 5-FU potentiated effects of the monotherapy. Additionally, CDKi and CDKi/chemotherapy combinations induced MHC I, indicative of enhanced immunogenicity. The in vivo studies revealed a cell line-specific response with best tumor growth control in the combination approach. Global acting CDKi's should be further investigated as targeting agents for HNSCC, either individually or in combination with selected drugs. The ability of dinaciclib to increase the immunogenicity of tumor cells renders this substance a particularly interesting candidate for immune-based oncological treatment regimens.
Collapse
Affiliation(s)
- Nina Schoenwaelder
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
- Correspondence: ; Tel.: +49-381-494-5764
| | - Inken Salewski
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
| | - Nadja Engel
- Department of Oral and Maxillofacial Surgery, Facial Plastic Surgery, University Medical Center Rostock, 18057 Rostock, Germany;
| | - Mareike Krause
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
| | - Björn Schneider
- Institute of Pathology, University Medical Center Rostock, Strempelstr.14, 18057 Rostock, Germany;
| | - Michael Müller
- Core Facility for Cell Sorting & Cell Analysis, Laboratory for Clinical Immunology, University Medical Center Rostock, 18057 Rostock, Germany;
| | - Christin Riess
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
- University Children’s Hospital, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Heiko Lemcke
- Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), University Medical Center Rostock, 18057 Rostock, Germany; (H.L.); (A.S.)
- Department of Cardiology, University Medical Center Rostock, 18059 Rostock, Germany
- Department Life, Light & Matter, Faculty of Interdisciplinary Research, University Rostock, 18059 Rostock, Germany
| | - Anna Skorska
- Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), University Medical Center Rostock, 18057 Rostock, Germany; (H.L.); (A.S.)
- Department of Cardiology, University Medical Center Rostock, 18059 Rostock, Germany
- Department Life, Light & Matter, Faculty of Interdisciplinary Research, University Rostock, 18059 Rostock, Germany
| | - Christina Grosse-Thie
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
| | - Christian Junghanss
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
| | - Claudia Maletzki
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
| |
Collapse
|
167
|
Chakrabarti R, Lee M, Higgs HN. Multiple roles for actin in secretory and endocytic pathways. Curr Biol 2021; 31:R603-R618. [PMID: 34033793 DOI: 10.1016/j.cub.2021.03.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Actin filaments play multiple roles in the secretory pathway and in endosome dynamics in mammals, including maintenance of Golgi structure, release of membrane cargo from the trans-Golgi network (TGN), endocytosis, and endosomal sorting dynamics. In addition, TGN carrier transport and endocytosis both occur by multiple mechanisms in mammals. Actin likely plays a role in at least four mammalian endocytic pathways, five pathways for membrane release from the TGN, and three processes involving endosomes. Also, the mammalian Golgi structure is highly dynamic, and actin is likely important for these dynamics. One challenge for many of these processes is the need to deal with other membrane-associated structures, such as the cortical actin network at the plasma membrane or the matrix that surrounds the Golgi. Arp2/3 complex is a major actin assembly factor in most of the processes mentioned, but roles for formins and tandem WH2-motif-containing assembly factors are being elucidated and are anticipated to grow with further study. The specific role for actin has not been defined for most of these processes, but is likely to involve the generation of force for membrane dynamics, either by actin polymerization itself or by myosin motor activity. Defining these processes mechanistically is necessary for understanding membrane dynamics in general, as well as pathways that utilize these processes, such as autophagy.
Collapse
Affiliation(s)
- Rajarshi Chakrabarti
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Miriam Lee
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Henry N Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA.
| |
Collapse
|
168
|
Mylvaganam S, Freeman SA, Grinstein S. The cytoskeleton in phagocytosis and macropinocytosis. Curr Biol 2021; 31:R619-R632. [PMID: 34033794 DOI: 10.1016/j.cub.2021.01.036] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cells of the innate immune system, notably macrophages, neutrophils and dendritic cells, perform essential antimicrobial and homeostatic functions. These functions rely on the dynamic surveillance of the environment supported by the formation of elaborate membrane protrusions. Such protrusions - pseudopodia, lamellipodia and filopodia - facilitate the sampling of the surrounding fluid by macropinocytosis, as well as the engulfment of particulates by phagocytosis. Both processes entail extreme plasma membrane deformations that require the coordinated rearrangement of cytoskeletal polymers, which exert protrusive force and drive membrane coalescence and scission. The resulting vacuolar compartments undergo pronounced remodeling and ultimate resolution by mechanisms that also involve the cytoskeleton. Here, we describe the regulation and functions of cytoskeletal assembly and remodeling during macropinocytosis and phagocytosis.
Collapse
Affiliation(s)
- Sivakami Mylvaganam
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Spencer A Freeman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Sergio Grinstein
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
169
|
Hyperthermia induced disruption of mechanical balance leads to G1 arrest and senescence in cells. Biochem J 2021; 478:179-196. [PMID: 33346336 DOI: 10.1042/bcj20200705] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022]
Abstract
Human body temperature limits below 40°C during heat stroke or fever. The implications of prolonged exposure to the physiologically relevant temperature (40°C) on cellular mechanobiology is poorly understood. Here, we have examined the effects of heat stress (40°C for 72 h incubation) in human lung adenocarcinoma (A549), mouse melanoma (B16F10), and non-cancerous mouse origin adipose tissue cells (L929). Hyperthermia increased the level of ROS, γ-H2AX and HSP70 and decreased mitochondrial membrane potential in the cells. Heat stress impaired cell division, caused G1 arrest, induced cellular senescence, and apoptosis in all the tested cell lines. The cells incubated at 40°C for 72 h displayed a significant decrease in the f-actin level and cellular traction as compared with cells incubated at 37°C. Also, the cells showed a larger focal adhesion area and stronger adhesion at 40°C than at 37°C. The mitotic cells at 40°C were unable to round up properly and displayed retracting actin stress fibers. Hyperthermia down-regulated HDAC6, increased the acetylation level of microtubules, and perturbed the chromosome alignment in the mitotic cells at 40°C. Overexpression of HDAC6 rescued the cells from the G1 arrest and reduced the delay in cell rounding at 40°C suggesting a crucial role of HDAC6 in hyperthermia mediated responses. This study elucidates the significant role of cellular traction, focal adhesions, and cytoskeletal networks in mitotic cell rounding and chromosomal misalignment. It also highlights the significance of HDAC6 in heat-evoked senile cellular responses.
Collapse
|
170
|
Semaphorin3F Drives Dendritic Spine Pruning Through Rho-GTPase Signaling. Mol Neurobiol 2021; 58:3817-3834. [PMID: 33856648 DOI: 10.1007/s12035-021-02373-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 03/24/2021] [Indexed: 12/18/2022]
Abstract
Dendritic spines of cortical pyramidal neurons are initially overproduced then remodeled substantially in the adolescent brain to achieve appropriate excitatory balance in mature circuits. Here we investigated the molecular mechanism of developmental spine pruning by Semaphorin 3F (Sema3F) and its holoreceptor complex, which consists of immunoglobulin-class adhesion molecule NrCAM, Neuropilin-2 (Npn2), and PlexinA3 (PlexA3) signaling subunits. Structure-function studies of the NrCAM-Npn2 interface showed that NrCAM stabilizes binding between Npn2 and PlexA3 necessary for Sema3F-induced spine pruning. Using a mouse neuronal culture system, we identified a dual signaling pathway for Sema3F-induced pruning, which involves activation of Tiam1-Rac1-PAK1-3 -LIMK1/2-Cofilin1 and RhoA-ROCK1/2-Myosin II in dendritic spines. Inhibitors of actin remodeling impaired spine collapse in the cortical neurons. Elucidation of these pathways expands our understanding of critical events that sculpt neuronal networks and may provide insight into how interruptions to these pathways could lead to spine dysgenesis in diseases such as autism, bipolar disorder, and schizophrenia.
Collapse
|
171
|
Sun B, Qu R, Fan T, Yang Y, Jiang X, Khan AU, Zhou Z, Zhang J, Wei K, Ouyang J, Dai J. Actin polymerization state regulates osteogenic differentiation in human adipose-derived stem cells. Cell Mol Biol Lett 2021; 26:15. [PMID: 33858321 PMCID: PMC8048231 DOI: 10.1186/s11658-021-00259-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/03/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Actin is an essential cellular protein that assembles into microfilaments and regulates numerous processes such as cell migration, maintenance of cell shape, and material transport. METHODS In this study, we explored the effect of actin polymerization state on the osteogenic differentiation of human adipose-derived stem cells (hASCs). The hASCs were treated for 7 days with different concentrations (0, 1, 5, 10, 20, and 50 nM) of jasplakinolide (JAS), a reagent that directly polymerizes F-actin. The effects of the actin polymerization state on cell proliferation, apoptosis, migration, and the maturity of focal adhesion-related proteins were assessed. In addition, western blotting and alizarin red staining assays were performed to assess osteogenic differentiation. RESULTS Cell proliferation and migration in the JAS (0, 1, 5, 10, and 20 nM) groups were higher than in the control group and the JAS (50 nM) group. The FAK, vinculin, paxillin, and talin protein expression levels were highest in the JAS (20 nM) group, while zyxin expression was highest in the JAS (50 nM) group. Western blotting showed that osteogenic differentiation in the JAS (0, 1, 5, 10, 20, and 50 nM) group was enhanced compared with that in the control group, and was strongest in the JAS (50 nM) group. CONCLUSIONS In summary, our data suggest that the actin polymerization state may promote the osteogenic differentiation of hASCs by regulating the protein expression of focal adhesion-associated proteins in a concentration-dependent manner. Our findings provide valuable information for exploring the mechanism of osteogenic differentiation in hASCs.
Collapse
Affiliation(s)
- Bing Sun
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Tingyu Fan
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Yuchao Yang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Xin Jiang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Asmat Ullah Khan
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Zhitao Zhou
- Central Laboratory, Southern Medical University, Guangzhou, China
| | - Jingliao Zhang
- Department of Foot and Ankle Surgery, Henan Luoyang Orthopedic Hospital, Zhengzhou, China
| | - Kuanhai Wei
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China.
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China.
| |
Collapse
|
172
|
Frey F, Idema T. More than just a barrier: using physical models to couple membrane shape to cell function. SOFT MATTER 2021; 17:3533-3549. [PMID: 33503097 DOI: 10.1039/d0sm01758b] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The correct execution of many cellular processes, such as division and motility, requires the cell to adopt a specific shape. Physically, these shapes are determined by the interplay of the plasma membrane and internal cellular driving factors. While the plasma membrane defines the boundary of the cell, processes inside the cell can result in the generation of forces that deform the membrane. These processes include protein binding, the assembly of protein superstructures, and the growth and contraction of cytoskeletal networks. Due to the complexity of the cell, relating observed membrane deformations back to internal processes is a challenging problem. Here, we review cell shape changes in endocytosis, cell adhesion, cell migration and cell division and discuss how by modeling membrane deformations we can investigate the inner working principles of the cell.
Collapse
Affiliation(s)
- Felix Frey
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, The Netherlands.
| | | |
Collapse
|
173
|
Nakamura M, Verboon JM, Prentiss CL, Parkhurst SM. The kinesin-like protein Pavarotti functions noncanonically to regulate actin dynamics. J Cell Biol 2021; 219:151940. [PMID: 32673395 PMCID: PMC7480107 DOI: 10.1083/jcb.201912117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/07/2020] [Accepted: 06/09/2020] [Indexed: 01/03/2023] Open
Abstract
Pavarotti, the Drosophila MKLP1 orthologue, is a kinesin-like protein that works with Tumbleweed (MgcRacGAP) as the centralspindlin complex. This complex is essential for cytokinesis, where it helps to organize the contractile actomyosin ring at the equator of dividing cells by activating the RhoGEF Pebble. Actomyosin rings also function as the driving force during cell wound repair. We previously showed that Tumbleweed and Pebble are required for the cell wound repair process. Here, we show that Pavarotti also functions during wound repair and confirm that while Pavarotti, Tumbleweed, and Pebble are all used during this cellular repair, each has a unique localization pattern and knockdown phenotype, demonstrating centralspindlin-independent functions. Surprisingly, we find that the classically microtubule-associated Pavarotti binds directly to actin in vitro and in vivo and has a noncanonical role directly regulating actin dynamics. Finally, we demonstrate that this actin regulation by Pavarotti is not specific to cellular wound repair but is also used in normal development.
Collapse
Affiliation(s)
- Mitsutoshi Nakamura
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jeffrey M Verboon
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Clara L Prentiss
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Susan M Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
174
|
Qi Y, Liu J, Chao J, Greer PA, Li S. PTEN dephosphorylates Abi1 to promote epithelial morphogenesis. J Cell Biol 2021; 219:151941. [PMID: 32673396 PMCID: PMC7480098 DOI: 10.1083/jcb.201910041] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 04/08/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
The tumor suppressor PTEN is essential for early development. Its lipid phosphatase activity converts PIP3 to PIP2 and antagonizes the PI3K–Akt pathway. In this study, we demonstrate that PTEN’s protein phosphatase activity is required for epiblast epithelial differentiation and polarization. This is accomplished by reconstitution of PTEN-null embryoid bodies with PTEN mutants that lack only PTEN’s lipid phosphatase activity or both PTEN’s lipid and protein phosphatase activities. Phosphotyrosine antibody immunoprecipitation and mass spectrometry were used to identify Abi1, a core component of the WASP-family verprolin homologous protein (WAVE) regulatory complex (WRC), as a new PTEN substrate. We demonstrate that PTEN dephosphorylation of Abi1 at Y213 and S216 results in Abi1 degradation through the calpain pathway. This leads to down-regulation of the WRC and reorganization of the actin cytoskeleton. The latter is critical to the transformation of nonpolar pluripotent stem cells into the polarized epiblast epithelium. Our findings establish a link between PTEN and WAVE-Arp2/3–regulated actin cytoskeletal dynamics in epithelial morphogenesis.
Collapse
Affiliation(s)
- Yanmei Qi
- Department of Surgery, Rutgers University Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Jie Liu
- Department of Surgery, Rutgers University Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Joshua Chao
- Department of Surgery, Rutgers University Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Peter A Greer
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Shaohua Li
- Department of Surgery, Rutgers University Robert Wood Johnson Medical School, New Brunswick, NJ
| |
Collapse
|
175
|
Lipid Rafts Interaction of the ARID3A Transcription Factor with EZRIN and G-Actin Regulates B-Cell Receptor Signaling. Diseases 2021; 9:diseases9010022. [PMID: 33804610 PMCID: PMC8005928 DOI: 10.3390/diseases9010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/12/2021] [Accepted: 03/18/2021] [Indexed: 11/17/2022] Open
Abstract
Several diseases originate via dysregulation of the actin cytoskeleton. The ARID3A/Bright transcription factor has also been implicated in malignancies, primarily those derived from hematopoietic lineages. Previously, we demonstrated that ARID3A shuttles between the nucleus and the plasma membrane, where it localizes within lipid rafts. There it interacts with components of the B-cell receptor (BCR) to reduce its ability to transmit downstream signaling. We demonstrate here that a direct component of ARID3A-regulated BCR signal strength is cortical actin. ARID3A interacts with actin exclusively within lipid rafts via the actin-binding protein EZRIN, which confines unstimulated BCRs within lipid rafts. BCR ligation discharges the ARID3A-EZRIN complex from lipid rafts, allowing the BCR to initiate downstream signaling events. The ARID3A-EZRIN interaction occurs almost exclusively within unpolymerized G-actin, where EZRIN interacts with the multifunctional ARID3A REKLES domain. These observations provide a mechanism by which a transcription factor directly regulates BCR signaling via linkage to the actin cytoskeleton with consequences for B-cell-related neoplasia.
Collapse
|
176
|
Dantas M, Lima JT, Ferreira JG. Nucleus-Cytoskeleton Crosstalk During Mitotic Entry. Front Cell Dev Biol 2021; 9:649899. [PMID: 33816500 PMCID: PMC8014196 DOI: 10.3389/fcell.2021.649899] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/26/2021] [Indexed: 12/30/2022] Open
Abstract
In preparation for mitosis, cells undergo extensive reorganization of the cytoskeleton and nucleus, so that chromosomes can be efficiently segregated into two daughter cells. Coordination of these cytoskeletal and nuclear events occurs through biochemical regulatory pathways, orchestrated by Cyclin-CDK activity. However, recent studies provide evidence that physical forces are also involved in the early steps of spindle assembly. Here, we will review how the crosstalk of physical forces and biochemical signals coordinates nuclear and cytoplasmic events during the G2-M transition, to ensure efficient spindle assembly and faithful chromosome segregation.
Collapse
Affiliation(s)
- Margarida Dantas
- Instituto de Investigação e Inovação em Saúde - i3S, University of Porto, Porto, Portugal.,BiotechHealth Ph.D. Programme, University of Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Joana T Lima
- Instituto de Investigação e Inovação em Saúde - i3S, University of Porto, Porto, Portugal.,Departamento de Biomedicina, Faculdade de Medicina, University of Porto, Porto, Portugal
| | - Jorge G Ferreira
- Instituto de Investigação e Inovação em Saúde - i3S, University of Porto, Porto, Portugal.,Departamento de Biomedicina, Faculdade de Medicina, University of Porto, Porto, Portugal
| |
Collapse
|
177
|
Nunes V, Ferreira JG. From the cytoskeleton to the nucleus: An integrated view on early spindle assembly. Semin Cell Dev Biol 2021; 117:42-51. [PMID: 33726956 DOI: 10.1016/j.semcdb.2021.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/01/2022]
Abstract
Accurate chromosome segregation requires a complete restructuring of cellular organization. Microtubules remodel to assemble a mitotic spindle and the actin cytoskeleton rearranges to form a stiff actomyosin cortex. These cytoplasmic events must be spatially and temporally coordinated with mitotic chromosome condensation and nuclear envelope permeabilization, in order to ensure mitotic timing and fidelity. Here, we discuss the main cytoskeletal and nuclear events that occur during mitotic entry in proliferating animal cells, focusing on their coordinated contribution for early mitotic spindle assembly. We will also explore recent progress in understanding their regulatory biochemical and mechanical pathways.
Collapse
Affiliation(s)
- Vanessa Nunes
- Instituto de Investigação e Inovação em Saúde - i3S, University of Porto, Porto, Portugal; BiotechHealth PhD Programe, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Jorge G Ferreira
- Instituto de Investigação e Inovação em Saúde - i3S, University of Porto, Porto, Portugal; Departamento de Biomedicina, Faculdade de Medicina, University of Porto, Porto, Portugal.
| |
Collapse
|
178
|
Lee H, Bonin K, Guthold M. Human mammary epithelial cells in a mature, stratified epithelial layer flatten and stiffen compared to single and confluent cells. Biochim Biophys Acta Gen Subj 2021; 1865:129891. [PMID: 33689830 DOI: 10.1016/j.bbagen.2021.129891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/07/2021] [Accepted: 03/03/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND The epithelium forms a protective barrier against external biological, chemical and physical insults. So far, AFM-based, micro-mechanical measurements have only been performed on single cells and confluent cells, but not yet on cells in mature layers. METHODS Using a combination of atomic force, fluorescence and confocal microscopy, we determined the changes in stiffness, morphology and actin distribution of human mammary epithelial cells (HMECs) as they transition from single cells to confluency to a mature layer. RESULTS Single HMECs have a tall, round (planoconvex) morphology, have actin stress fibers at the base, have diffuse cortical actin, and have a stiffness of 1 kPa. Confluent HMECs start to become flatter, basal actin stress fibers start to disappear, and actin accumulates laterally where cells abut. Overall stiffness is still 1 kPa with two-fold higher stiffness in the abutting regions. As HMECs mature and form multilayered structures, cells on apical surfaces become flatter (apically more level), wider, and seven times stiffer (mean, 7 kPa) than single and confluent cells. The main drivers of these changes are actin filaments, as cells show strong actin accumulation in the regions where cells adjoin, and in the apical regions. CONCLUSIONS HMECs stiffen, flatten and redistribute actin upon transiting from single cells to mature, confluent layers. GENERAL SIGNIFICANCE Our findings advance the understanding of breast ductal morphogenesis and mechanical homeostasis.
Collapse
Affiliation(s)
- Hyunsu Lee
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Keith Bonin
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Martin Guthold
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA.
| |
Collapse
|
179
|
Vignaud T, Copos C, Leterrier C, Toro-Nahuelpan M, Tseng Q, Mahamid J, Blanchoin L, Mogilner A, Théry M, Kurzawa L. Stress fibres are embedded in a contractile cortical network. NATURE MATERIALS 2021; 20:410-420. [PMID: 33077951 PMCID: PMC7610471 DOI: 10.1038/s41563-020-00825-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 09/14/2020] [Indexed: 05/06/2023]
Abstract
Contractile actomyosin networks are responsible for the production of intracellular forces. There is increasing evidence that bundles of actin filaments form interconnected and interconvertible structures with the rest of the network. In this study, we explored the mechanical impact of these interconnections on the production and distribution of traction forces throughout the cell. By using a combination of hydrogel micropatterning, traction force microscopy and laser photoablation, we measured the relaxation of traction forces in response to local photoablations. Our experimental results and modelling of the mechanical response of the network revealed that bundles were fully embedded along their entire length in a continuous and contractile network of cortical filaments. Moreover, the propagation of the contraction of these bundles throughout the entire cell was dependent on this embedding. In addition, these bundles appeared to originate from the alignment and coalescence of thin and unattached cortical actin filaments from the surrounding mesh.
Collapse
Affiliation(s)
- Timothée Vignaud
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Végétale, Grenoble-Alpes University/CEA/CNRS/INRA, Grenoble, France
- CytoMorpho Lab, Hôpital Saint Louis, Institut Universitaire d'Hématologie, Université Paris Diderot/CEA/INSERM, Paris, France
- Clinique de Chirurgie Digestive et Endocrinienne, Hôtel Dieu, Nantes, France
| | - Calina Copos
- Courant Institute and Department of Biology, New York University, New York, NY, USA
| | - Christophe Leterrier
- NeuroCyto, Institute of NeuroPhysiopathology (INP), CNRS, Aix Marseille Université, Marseille, France
| | - Mauricio Toro-Nahuelpan
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Qingzong Tseng
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Végétale, Grenoble-Alpes University/CEA/CNRS/INRA, Grenoble, France
- CytoMorpho Lab, Hôpital Saint Louis, Institut Universitaire d'Hématologie, Université Paris Diderot/CEA/INSERM, Paris, France
| | - Julia Mahamid
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Laurent Blanchoin
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Végétale, Grenoble-Alpes University/CEA/CNRS/INRA, Grenoble, France
- CytoMorpho Lab, Hôpital Saint Louis, Institut Universitaire d'Hématologie, Université Paris Diderot/CEA/INSERM, Paris, France
| | - Alex Mogilner
- Courant Institute and Department of Biology, New York University, New York, NY, USA.
| | - Manuel Théry
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Végétale, Grenoble-Alpes University/CEA/CNRS/INRA, Grenoble, France.
- CytoMorpho Lab, Hôpital Saint Louis, Institut Universitaire d'Hématologie, Université Paris Diderot/CEA/INSERM, Paris, France.
| | - Laetitia Kurzawa
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Végétale, Grenoble-Alpes University/CEA/CNRS/INRA, Grenoble, France.
- CytoMorpho Lab, Hôpital Saint Louis, Institut Universitaire d'Hématologie, Université Paris Diderot/CEA/INSERM, Paris, France.
| |
Collapse
|
180
|
Mechanochemical control of epidermal stem cell divisions by B-plexins. Nat Commun 2021; 12:1308. [PMID: 33637728 PMCID: PMC7910479 DOI: 10.1038/s41467-021-21513-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 01/28/2021] [Indexed: 01/31/2023] Open
Abstract
The precise spatiotemporal control of cell proliferation is key to the morphogenesis of epithelial tissues. Epithelial cell divisions lead to tissue crowding and local changes in force distribution, which in turn suppress the rate of cell divisions. However, the molecular mechanisms underlying this mechanical feedback are largely unclear. Here, we identify a critical requirement of B-plexin transmembrane receptors in the response to crowding-induced mechanical forces during embryonic skin development. Epidermal stem cells lacking B-plexins fail to sense mechanical compression, resulting in disinhibition of the transcriptional coactivator YAP, hyperproliferation, and tissue overgrowth. Mechanistically, we show that B-plexins mediate mechanoresponses to crowding through stabilization of adhesive cell junctions and lowering of cortical stiffness. Finally, we provide evidence that the B-plexin-dependent mechanochemical feedback is also pathophysiologically relevant to limit tumor growth in basal cell carcinoma, the most common type of skin cancer. Our data define a central role of B-plexins in mechanosensation to couple cell density and cell division in development and disease.
Collapse
|
181
|
Smoothelin-like 2 Inhibits Coronin-1B to Stabilize the Apical Actin Cortex during Epithelial Morphogenesis. Curr Biol 2021; 31:696-706.e9. [PMID: 33275893 DOI: 10.1016/j.cub.2020.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 09/24/2020] [Accepted: 11/03/2020] [Indexed: 11/22/2022]
Abstract
The actin cortex is involved in many biological processes and needs to be significantly remodeled during cell differentiation. Developing epithelial cells construct a dense apical actin cortex to carry out their barrier and exchange functions. The apical cortex assembles in response to three-dimensional (3D) extracellular cues, but the regulation of this process during epithelial morphogenesis remains unknown. Here, we describe the function of Smoothelin-like 2 (SMTNL2), a member of the smooth-muscle-related Smoothelin protein family, in apical cortex maturation. SMTNL2 is induced during development in multiple epithelial tissues and localizes to the apical and junctional actin cortex in intestinal and kidney epithelial cells. SMTNL2 deficiency leads to membrane herniations in the apical domain of epithelial cells, indicative of cortex abnormalities. We find that SMTNL2 binds to actin filaments and is required to slow down the turnover of apical actin. We also characterize the SMTNL2 proximal interactome and find that SMTNL2 executes its functions partly through inhibition of coronin-1B. Although coronin-1B-mediated actin dynamics are required for early morphogenesis, its sustained activity is detrimental for the mature apical shape. SMTNL2 binds to coronin-1B through its N-terminal coiled-coil region and negates its function to stabilize the apical cortex. In sum, our results unveil a mechanism for regulating actin dynamics during epithelial morphogenesis, providing critical insights on the developmental control of the cellular cortex.
Collapse
|
182
|
Lv Z, de-Carvalho J, Telley IA, Großhans J. Cytoskeletal mechanics and dynamics in the Drosophila syncytial embryo. J Cell Sci 2021; 134:134/4/jcs246496. [PMID: 33597155 DOI: 10.1242/jcs.246496] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cell and tissue functions rely on the genetic programmes and cascades of biochemical signals. It has become evident during the past decade that the physical properties of soft material that govern the mechanics of cells and tissues play an important role in cellular function and morphology. The biophysical properties of cells and tissues are determined by the cytoskeleton, consisting of dynamic networks of F-actin and microtubules, molecular motors, crosslinkers and other associated proteins, among other factors such as cell-cell interactions. The Drosophila syncytial embryo represents a simple pseudo-tissue, with its nuclei orderly embedded in a structured cytoskeletal matrix at the embryonic cortex with no physical separation by cellular membranes. Here, we review the stereotypic dynamics and regulation of the cytoskeleton in Drosophila syncytial embryos and how cytoskeletal dynamics underlies biophysical properties and the emergence of collective features. We highlight the specific features and processes of syncytial embryos and discuss the applicability of biophysical approaches.
Collapse
Affiliation(s)
- Zhiyi Lv
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Jorge de-Carvalho
- Instituto Gulbenkian de Ciência, Fundação Calouste Gulbenkian, 2780-156 Oeiras, Portugal
| | - Ivo A Telley
- Instituto Gulbenkian de Ciência, Fundação Calouste Gulbenkian, 2780-156 Oeiras, Portugal
| | - Jörg Großhans
- Fachbereich Biologie, Philipps-Universität Marburg, 35043 Marburg, Germany
| |
Collapse
|
183
|
Phenotypic Plasticity of Cancer Cells Based on Remodeling of the Actin Cytoskeleton and Adhesive Structures. Int J Mol Sci 2021; 22:ijms22041821. [PMID: 33673054 PMCID: PMC7918886 DOI: 10.3390/ijms22041821] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 02/08/2023] Open
Abstract
There is ample evidence that, instead of a binary switch, epithelial-mesenchymal transition (EMT) in cancer results in a flexible array of phenotypes, each one uniquely suited to a stage in the invasion-metastasis cascade. The phenotypic plasticity of epithelium-derived cancer cells gives them an edge in surviving and thriving in alien environments. This review describes in detail the actin cytoskeleton and E-cadherin-based adherens junction rearrangements that cancer cells need to implement in order to achieve the advantageous epithelial/mesenchymal phenotype and plasticity of migratory phenotypes that can arise from partial EMT.
Collapse
|
184
|
Kiel C, Matallanas D, Kolch W. The Ins and Outs of RAS Effector Complexes. Biomolecules 2021; 11:236. [PMID: 33562401 PMCID: PMC7915224 DOI: 10.3390/biom11020236] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 01/31/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022] Open
Abstract
RAS oncogenes are among the most commonly mutated proteins in human cancers. They regulate a wide range of effector pathways that control cell proliferation, survival, differentiation, migration and metabolic status. Including aberrations in these pathways, RAS-dependent signaling is altered in more than half of human cancers. Targeting mutant RAS proteins and their downstream oncogenic signaling pathways has been elusive. However, recent results comprising detailed molecular studies, large scale omics studies and computational modeling have painted a new and more comprehensive portrait of RAS signaling that helps us to understand the intricacies of RAS, how its physiological and pathophysiological functions are regulated, and how we can target them. Here, we review these efforts particularly trying to relate the detailed mechanistic studies with global functional studies. We highlight the importance of computational modeling and data integration to derive an actionable understanding of RAS signaling that will allow us to design new mechanism-based therapies for RAS mutated cancers.
Collapse
Affiliation(s)
- Christina Kiel
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin 4, Ireland; (C.K.); (D.M.)
- UCD Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - David Matallanas
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin 4, Ireland; (C.K.); (D.M.)
| | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin 4, Ireland; (C.K.); (D.M.)
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
185
|
Hodgkinson T, Tsimbouri PM, Llopis-Hernandez V, Campsie P, Scurr D, Childs PG, Phillips D, Donnelly S, Wells JA, O'Brien FJ, Salmeron-Sanchez M, Burgess K, Alexander M, Vassalli M, Oreffo ROC, Reid S, France DJ, Dalby MJ. The use of nanovibration to discover specific and potent bioactive metabolites that stimulate osteogenic differentiation in mesenchymal stem cells. SCIENCE ADVANCES 2021; 7:7/9/eabb7921. [PMID: 33637520 PMCID: PMC7909882 DOI: 10.1126/sciadv.abb7921] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 01/12/2021] [Indexed: 05/02/2023]
Abstract
Bioactive metabolites have wide-ranging biological activities and are a potential source of future research and therapeutic tools. Here, we use nanovibrational stimulation to induce osteogenic differentiation of mesenchymal stem cells, in the absence of off-target, nonosteogenic differentiation. We show that this differentiation method, which does not rely on the addition of exogenous growth factors to culture media, provides an artifact-free approach to identifying bioactive metabolites that specifically and potently induce osteogenesis. We first identify a highly specific metabolite, cholesterol sulfate, an endogenous steroid. Next, a screen of other small molecules with a similar steroid scaffold identified fludrocortisone acetate with both specific and highly potent osteogenic-inducing activity. Further, we implicate cytoskeletal contractility as a measure of osteogenic potency and cell stiffness as a measure of specificity. These findings demonstrate that physical principles can be used to identify bioactive metabolites and then enable optimization of metabolite potency can be optimized by examining structure-function relationships.
Collapse
Affiliation(s)
- Tom Hodgkinson
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin D2, Ireland
| | - P Monica Tsimbouri
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Virginia Llopis-Hernandez
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Paul Campsie
- SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow G1 1QE, UK
| | - David Scurr
- School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, UK
| | - Peter G Childs
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8LT, UK
| | - David Phillips
- School of Chemistry, College of Science and Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Sam Donnelly
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Julia A Wells
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin D2, Ireland
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8LT, UK
| | - Karl Burgess
- Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, University of Glasgow, Switchback Rd., Bearsden, Glasgow G61 1BD, UK
| | - Morgan Alexander
- School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, UK
| | - Massimo Vassalli
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8LT, UK
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Stuart Reid
- SUPA Department of Biomedical Engineering, University of Strathclyde, Glasgow G1 1QE, UK
| | - David J France
- School of Chemistry, College of Science and Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
186
|
Lehtimäki JI, Rajakylä EK, Tojkander S, Lappalainen P. Generation of stress fibers through myosin-driven reorganization of the actin cortex. eLife 2021; 10:60710. [PMID: 33506761 PMCID: PMC7877910 DOI: 10.7554/elife.60710] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 01/27/2021] [Indexed: 12/26/2022] Open
Abstract
Contractile actomyosin bundles, stress fibers, govern key cellular processes including migration, adhesion, and mechanosensing. Stress fibers are thus critical for developmental morphogenesis. The most prominent actomyosin bundles, ventral stress fibers, are generated through coalescence of pre-existing stress fiber precursors. However, whether stress fibers can assemble through other mechanisms has remained elusive. We report that stress fibers can also form without requirement of pre-existing actomyosin bundles. These structures, which we named cortical stress fibers, are embedded in the cell cortex and assemble preferentially underneath the nucleus. In this process, non-muscle myosin II pulses orchestrate the reorganization of cortical actin meshwork into regular bundles, which promote reinforcement of nascent focal adhesions, and subsequent stabilization of the cortical stress fibers. These results identify a new mechanism by which stress fibers can be generated de novo from the actin cortex and establish role for stochastic myosin pulses in the assembly of functional actomyosin bundles.
Collapse
Affiliation(s)
- Jaakko I Lehtimäki
- HiLIFE Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Eeva Kaisa Rajakylä
- Section of Pathology, Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Sari Tojkander
- Section of Pathology, Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Pekka Lappalainen
- HiLIFE Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
187
|
The Actomyosin Cortex of Cells: A Thin Film of Active Matter. J Indian Inst Sci 2021. [DOI: 10.1007/s41745-020-00220-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
188
|
Gan WJ, Motegi F. Mechanochemical Control of Symmetry Breaking in the Caenorhabditis elegans Zygote. Front Cell Dev Biol 2021; 8:619869. [PMID: 33537308 PMCID: PMC7848089 DOI: 10.3389/fcell.2020.619869] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Cell polarity is the asymmetric organization of cellular components along defined axes. A key requirement for polarization is the ability of the cell to break symmetry and achieve a spatially biased organization. Despite different triggering cues in various systems, symmetry breaking (SB) usually relies on mechanochemical modulation of the actin cytoskeleton, which allows for advected movement and reorganization of cellular components. Here, the mechanisms underlying SB in Caenorhabditis elegans zygote, one of the most popular models to study cell polarity, are reviewed. A zygote initiates SB through the centrosome, which modulates mechanics of the cell cortex to establish advective flow of cortical proteins including the actin cytoskeleton and partitioning defective (PAR) proteins. The chemical signaling underlying centrosomal control of the Aurora A kinase–mediated cascade to convert the organization of the contractile actomyosin network from an apolar to polar state is also discussed.
Collapse
Affiliation(s)
- Wan Jun Gan
- Temasek Life-Sciences Laboratory, Singapore, Singapore
| | - Fumio Motegi
- Temasek Life-Sciences Laboratory, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore.,Mechanobiology Institute, National University of Singapore, Singapore, Singapore.,Institute of Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
189
|
Brunet T, Albert M, Roman W, Coyle MC, Spitzer DC, King N. A flagellate-to-amoeboid switch in the closest living relatives of animals. eLife 2021; 10:e61037. [PMID: 33448265 PMCID: PMC7895527 DOI: 10.7554/elife.61037] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 01/14/2021] [Indexed: 12/19/2022] Open
Abstract
Amoeboid cell types are fundamental to animal biology and broadly distributed across animal diversity, but their evolutionary origin is unclear. The closest living relatives of animals, the choanoflagellates, display a polarized cell architecture (with an apical flagellum encircled by microvilli) that resembles that of epithelial cells and suggests homology, but this architecture differs strikingly from the deformable phenotype of animal amoeboid cells, which instead evoke more distantly related eukaryotes, such as diverse amoebae. Here, we show that choanoflagellates subjected to confinement become amoeboid by retracting their flagella and activating myosin-based motility. This switch allows escape from confinement and is conserved across choanoflagellate diversity. The conservation of the amoeboid cell phenotype across animals and choanoflagellates, together with the conserved role of myosin, is consistent with homology of amoeboid motility in both lineages. We hypothesize that the differentiation between animal epithelial and crawling cells might have evolved from a stress-induced switch between flagellate and amoeboid forms in their single-celled ancestors.
Collapse
Affiliation(s)
- Thibaut Brunet
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Marvin Albert
- Department of Molecular Life Sciences, University of ZürichZurichSwitzerland
| | - William Roman
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBERNEDBarcelonaSpain
| | - Maxwell C Coyle
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Danielle C Spitzer
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Nicole King
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
190
|
Lenne PF, Rupprecht JF, Viasnoff V. Cell Junction Mechanics beyond the Bounds of Adhesion and Tension. Dev Cell 2021; 56:202-212. [PMID: 33453154 DOI: 10.1016/j.devcel.2020.12.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/06/2020] [Accepted: 12/21/2020] [Indexed: 12/22/2022]
Abstract
Cell-cell junctions, in particular adherens junctions, are major determinants of tissue mechanics during morphogenesis and homeostasis. In attempts to link junctional mechanics to tissue mechanics, many have utilized explicitly or implicitly equilibrium approaches based on adhesion energy, surface energy, and contractility to determine the mechanical equilibrium at junctions. However, it is increasingly clear that they have significant limitations, such as that it remains challenging to link the dynamics of the molecular components to the resulting physical properties of the junction, to its remodeling ability, and to its adhesion strength. In this perspective, we discuss recent attempts to consider the aspect of energy dissipation at junctions to draw contact points with soft matter physics where energy loss plays a critical role in adhesion theories. We set the grounds for a theoretical framework of the junction mechanics that bridges the dynamics at the molecular scale to the mechanics at the tissue scale.
Collapse
Affiliation(s)
- Pierre-François Lenne
- Aix Marseille Université, CNRS, IBDM, Turing Centre for Living Systems, 13288 Marseille, France.
| | - Jean-François Rupprecht
- Aix Marseille Université, CNRS, CPT, Turing Centre for Living Systems, 13288 Marseille, France.
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; CNRS Biomechanics of Cell Contacts, Singapore 117411, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117411, Singapore.
| |
Collapse
|
191
|
Schneider F, Colin-York H, Fritzsche M. Quantitative Bio-Imaging Tools to Dissect the Interplay of Membrane and Cytoskeletal Actin Dynamics in Immune Cells. Front Immunol 2021; 11:612542. [PMID: 33505401 PMCID: PMC7829180 DOI: 10.3389/fimmu.2020.612542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022] Open
Abstract
Cellular function is reliant on the dynamic interplay between the plasma membrane and the actin cytoskeleton. This critical relationship is of particular importance in immune cells, where both the cytoskeleton and the plasma membrane work in concert to organize and potentiate immune signaling events. Despite their importance, there remains a critical gap in understanding how these respective dynamics are coupled, and how this coupling in turn may influence immune cell function from the bottom up. In this review, we highlight recent optical technologies that could provide strategies to investigate the simultaneous dynamics of both the cytoskeleton and membrane as well as their interplay, focusing on current and future applications in immune cells. We provide a guide of the spatio-temporal scale of each technique as well as highlighting novel probes and labels that have the potential to provide insights into membrane and cytoskeletal dynamics. The quantitative biophysical tools presented here provide a new and exciting route to uncover the relationship between plasma membrane and cytoskeletal dynamics that underlies immune cell function.
Collapse
Affiliation(s)
- Falk Schneider
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Huw Colin-York
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Kennedy Institute for Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Marco Fritzsche
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Kennedy Institute for Rheumatology, University of Oxford, Oxford, United Kingdom
- Rosalind Franklin Institute, Harwell Campus, Didcot, United Kingdom
| |
Collapse
|
192
|
Brill-Karniely Y. Mechanical Measurements of Cells Using AFM: 3D or 2D Physics? Front Bioeng Biotechnol 2020; 8:605153. [PMID: 33330437 PMCID: PMC7731794 DOI: 10.3389/fbioe.2020.605153] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/08/2020] [Indexed: 12/28/2022] Open
Affiliation(s)
- Yifat Brill-Karniely
- Faculty of Medicine, The School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
193
|
Welf ES, Miles CE, Huh J, Sapoznik E, Chi J, Driscoll MK, Isogai T, Noh J, Weems AD, Pohlkamp T, Dean K, Fiolka R, Mogilner A, Danuser G. Actin-Membrane Release Initiates Cell Protrusions. Dev Cell 2020; 55:723-736.e8. [PMID: 33308479 DOI: 10.1016/j.devcel.2020.11.024] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/08/2020] [Accepted: 11/20/2020] [Indexed: 01/04/2023]
Abstract
Despite the well-established role of actin polymerization as a driving mechanism for cell protrusion, upregulated actin polymerization alone does not initiate protrusions. Using a combination of theoretical modeling and quantitative live-cell imaging experiments, we show that local depletion of actin-membrane links is needed for protrusion initiation. Specifically, we show that the actin-membrane linker ezrin is depleted prior to protrusion onset and that perturbation of ezrin's affinity for actin modulates protrusion frequency and efficiency. We also show how actin-membrane release works in concert with actin polymerization, leading to a comprehensive model for actin-driven shape changes. Actin-membrane release plays a similar role in protrusions driven by intracellular pressure. Thus, our findings suggest that protrusion initiation might be governed by a universal regulatory mechanism, whereas the mechanism of force generation determines the shape and expansion properties of the protrusion.
Collapse
Affiliation(s)
- Erik S Welf
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Christopher E Miles
- Courant Institute of Mathematical Sciences, New York University, New York, NY 10012, USA; Department of Biology, New York University, New York, NY 10012, USA
| | - Jaewon Huh
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Etai Sapoznik
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph Chi
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Meghan K Driscoll
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tadamoto Isogai
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jungsik Noh
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andrew D Weems
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Theresa Pohlkamp
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kevin Dean
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Reto Fiolka
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences, New York University, New York, NY 10012, USA; Department of Biology, New York University, New York, NY 10012, USA.
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
194
|
Strauss RE, Gourdie RG. Cx43 and the Actin Cytoskeleton: Novel Roles and Implications for Cell-Cell Junction-Based Barrier Function Regulation. Biomolecules 2020; 10:E1656. [PMID: 33321985 PMCID: PMC7764618 DOI: 10.3390/biom10121656] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Barrier function is a vital homeostatic mechanism employed by epithelial and endothelial tissue. Diseases across a wide range of tissue types involve dynamic changes in transcellular junctional complexes and the actin cytoskeleton in the regulation of substance exchange across tissue compartments. In this review, we focus on the contribution of the gap junction protein, Cx43, to the biophysical and biochemical regulation of barrier function. First, we introduce the structure and canonical channel-dependent functions of Cx43. Second, we define barrier function and examine the key molecular structures fundamental to its regulation. Third, we survey the literature on the channel-dependent roles of connexins in barrier function, with an emphasis on the role of Cx43 and the actin cytoskeleton. Lastly, we discuss findings on the channel-independent roles of Cx43 in its associations with the actin cytoskeleton and focal adhesion structures highlighted by PI3K signaling, in the potential modulation of cellular barriers. Mounting evidence of crosstalk between connexins, the cytoskeleton, focal adhesion complexes, and junctional structures has led to a growing appreciation of how barrier-modulating mechanisms may work together to effect solute and cellular flux across tissue boundaries. This new understanding could translate into improved therapeutic outcomes in the treatment of barrier-associated diseases.
Collapse
Affiliation(s)
- Randy E. Strauss
- Virginia Tech, Translational Biology Medicine and Health (TBMH) Program, Roanoke, VA 24016, USA
| | - Robert G. Gourdie
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| |
Collapse
|
195
|
van Haastert PJM. Unified control of amoeboid pseudopod extension in multiple organisms by branched F-actin in the front and parallel F-actin/myosin in the cortex. PLoS One 2020; 15:e0243442. [PMID: 33296414 PMCID: PMC7725310 DOI: 10.1371/journal.pone.0243442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023] Open
Abstract
The trajectory of moving eukaryotic cells depends on the kinetics and direction of extending pseudopods. The direction of pseudopods has been well studied to unravel mechanisms for chemotaxis, wound healing and inflammation. However, the kinetics of pseudopod extension-when and why do pseudopods start and stop- is equally important, but is largely unknown. Here the START and STOP of about 4000 pseudopods was determined in four different species, at four conditions and in nine mutants (fast amoeboids Dictyostelium and neutrophils, slow mesenchymal stem cells, and fungus B.d. chytrid with pseudopod and a flagellum). The START of a first pseudopod is a random event with a probability that is species-specific (23%/s for neutrophils). In all species and conditions, the START of a second pseudopod is strongly inhibited by the extending first pseudopod, which depends on parallel filamentous actin/myosin in the cell cortex. Pseudopods extend at a constant rate by polymerization of branched F-actin at the pseudopod tip, which requires the Scar complex. The STOP of pseudopod extension is induced by multiple inhibitory processes that evolve during pseudopod extension and mainly depend on the increasing size of the pseudopod. Surprisingly, no differences in pseudopod kinetics are detectable between polarized, unpolarized or chemotactic cells, and also not between different species except for small differences in numerical values. This suggests that the analysis has uncovered the fundament of cell movement with distinct roles for stimulatory branched F-actin in the protrusion and inhibitory parallel F-actin in the contractile cortex.
Collapse
|
196
|
Casani S, Casanova J, Llimargas M. Unravelling the distinct contribution of cell shape changes and cell intercalation to tissue morphogenesis: the case of the Drosophila trachea. Open Biol 2020; 10:200329. [PMID: 33234070 PMCID: PMC7729023 DOI: 10.1098/rsob.200329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Intercalation allows cells to exchange positions in a spatially oriented manner in an array of diverse processes, spanning convergent extension in embryonic gastrulation to the formation of tubular organs. However, given the co-occurrence of cell intercalation and changes in cell shape, it is sometimes difficult to ascertain their respective contribution to morphogenesis. A well-established model to analyse intercalation, particularly in tubular organs, is the Drosophila tracheal system. There, fibroblast growth factor (FGF) signalling at the tip of the dorsal branches generates a ‘pulling’ force believed to promote cell elongation and cell intercalation, which account for the final branch extension. Here, we used a variety of experimental conditions to study the contribution of cell elongation and cell intercalation to morphogenesis and analysed their mutual requirements. We provide evidence that cell intercalation does not require cell elongation and vice versa. We also show that the two cell behaviours are controlled by independent but simultaneous mechanisms, and that cell elongation is sufficient to account for full extension of the dorsal branch, while cell intercalation has a specific role in setting the diameter of this structure. Thus, rather than viewing changes in cell shape and cell intercalation as just redundant events that add robustness to a given morphogenetic process, we find that they can also act by contributing to different features of tissue architecture.
Collapse
Affiliation(s)
- Sandra Casani
- Institut de Biologia Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Baldiri Reixac, 10-12, 08028 Barcelona, Catalonia, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain
| | - Jordi Casanova
- Institut de Biologia Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Baldiri Reixac, 10-12, 08028 Barcelona, Catalonia, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain
| | - Marta Llimargas
- Institut de Biologia Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Baldiri Reixac, 10-12, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
197
|
Gensbittel V, Kräter M, Harlepp S, Busnelli I, Guck J, Goetz JG. Mechanical Adaptability of Tumor Cells in Metastasis. Dev Cell 2020; 56:164-179. [PMID: 33238151 DOI: 10.1016/j.devcel.2020.10.011] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/18/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022]
Abstract
The most dangerous aspect of cancer lies in metastatic progression. Tumor cells will successfully form life-threatening metastases when they undergo sequential steps along a journey from the primary tumor to distant organs. From a biomechanics standpoint, growth, invasion, intravasation, circulation, arrest/adhesion, and extravasation of tumor cells demand particular cell-mechanical properties in order to survive and complete the metastatic cascade. With metastatic cells usually being softer than their non-malignant counterparts, high deformability for both the cell and its nucleus is thought to offer a significant advantage for metastatic potential. However, it is still unclear whether there is a finely tuned but fixed mechanical state that accommodates all mechanical features required for survival throughout the cascade or whether tumor cells need to dynamically refine their properties and intracellular components at each new step encountered. Here, we review the various mechanical requirements successful cancer cells might need to fulfill along their journey and speculate on the possibility that they dynamically adapt their properties accordingly. The mechanical signature of a successful cancer cell might actually be its ability to adapt to the successive microenvironmental constraints along the different steps of the journey.
Collapse
Affiliation(s)
- Valentin Gensbittel
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Sébastien Harlepp
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Ignacio Busnelli
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany.
| | - Jacky G Goetz
- INSERM UMR_S1109, Tumor Biomechanics, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
| |
Collapse
|
198
|
Skouloudaki K, Papadopoulos DK, Hurd TW. The Molecular Network of YAP/Yorkie at the Cell Cortex and their Role in Ocular Morphogenesis. Int J Mol Sci 2020; 21:ijms21228804. [PMID: 33233821 PMCID: PMC7699867 DOI: 10.3390/ijms21228804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 12/11/2022] Open
Abstract
During development, the precise control of tissue morphogenesis requires changes in the cell number, size, shape, position, and gene expression, which are driven by both chemical and mechanical cues from the surrounding microenvironment. Such physical and architectural features inform cells about their proliferative and migratory capacity, enabling the formation and maintenance of complex tissue architecture. In polarised epithelia, the apical cell cortex, a thin actomyosin network that lies directly underneath the apical plasma membrane, functions as a platform to facilitate signal transmission between the external environment and downstream signalling pathways. One such signalling pathway culminates in the regulation of YES-associated protein (YAP) and TAZ transcriptional co-activators and their sole Drosophila homolog, Yorkie, to drive proliferation and differentiation. Recent studies have demonstrated that YAP/Yorkie exhibit a distinct function at the apical cell cortex. Here, we review recent efforts to understand the mechanisms that regulate YAP/Yki at the apical cell cortex of epithelial cells and how normal and disturbed YAP-actomyosin networks are involved in eye development and disease.
Collapse
|
199
|
Van der Meeren L, Verduijn J, Krysko DV, Skirtach AG. AFM Analysis Enables Differentiation between Apoptosis, Necroptosis, and Ferroptosis in Murine Cancer Cells. iScience 2020; 23:101816. [PMID: 33299979 PMCID: PMC7702191 DOI: 10.1016/j.isci.2020.101816] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/13/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Regulated cell death (RCD) has a fundamental role in development, pathology, and tissue homeostasis. In order to understand the RCD mechanisms, it is essential to follow these processes in real time. Here, atomic force microscopy (AFM) is applied to morphologically and mechanically characterize four RCD modalities (intrinsic and extrinsic apoptosis, necroptosis, and ferroptosis) in murine tumor cell lines. The nano-topographical analysis revealed a distinct surface morphology in case of necroptosis, ∼ 200 nm membrane disruptions are observed. Using mechanical measurements, it is possible to detect the early onset of RCD. Combined elasticity and microrheology analysis allowed for a clear distinction between apoptotic and regulated necrotic cell death. Finally, immunofluorescence analysis of the cytoskeleton structure during the RCD processes confirm the measured mechanical changes. The results of this study not only demonstrate the possibility of early real-time cell death detection but also reveal important differences in the cytoskeletal dynamics between multiple RCD modalities. AFM is a label-free method to distinguish apoptosis, necroptosis, and ferroptosis Nanotopography and subtle morphologic changes are distinct for each RCD Mechanobiology elasticity analysis reveals changes occurring at early stages of RCD Microrheology data agree with mechanobiology Young's modulus analysis
Collapse
Affiliation(s)
- Louis Van der Meeren
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium.,Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Joost Verduijn
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium.,Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Dmitri V Krysko
- Cancer Research Institute Ghent, 9000 Ghent, Belgium.,Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium.,Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russian Federation
| | - André G Skirtach
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium.,Cancer Research Institute Ghent, 9000 Ghent, Belgium
| |
Collapse
|
200
|
The Arf-GEF Steppke promotes F-actin accumulation, cell protrusions and tissue sealing during Drosophila dorsal closure. PLoS One 2020; 15:e0239357. [PMID: 33186390 PMCID: PMC7665897 DOI: 10.1371/journal.pone.0239357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/02/2020] [Indexed: 01/05/2023] Open
Abstract
Cytohesin Arf-GEFs promote actin polymerization and protrusions of cultured cells, whereas the Drosophila cytohesin, Steppke, antagonizes actomyosin networks in several developmental contexts. To reconcile these findings, we analyzed epidermal leading edge actin networks during Drosophila embryo dorsal closure. Here, Steppke is required for F-actin of the actomyosin cable and for actin-based protrusions. steppke mutant defects in the leading edge actin networks are associated with improper sealing of the dorsal midline, but are distinguishable from effects of myosin mis-regulation. Steppke localizes to leading edge cell-cell junctions with accumulations of the F-actin regulator Enabled emanating from either side. Enabled requires Steppke for full leading edge recruitment, and genetic interaction shows the proteins cooperate for dorsal closure. Inversely, Steppke over-expression induces ectopic, actin-rich, lamellar cell protrusions, an effect dependent on the Arf-GEF activity and PH domain of Steppke, but independent of Steppke recruitment to myosin-rich AJs via its coiled-coil domain. Thus, Steppke promotes actin polymerization and cell protrusions, effects that occur in conjunction with Steppke's previously reported regulation of myosin contractility during dorsal closure.
Collapse
|