1
|
Wang Y, Shi P, Liu G, Chen W, Wang YJ, Hu Y, Yang A, Wei T, Chen YC, Liang L, Liu Z, Liu YJ, Wu C. Espin enhances confined cell migration by promoting filopodia formation and contributes to cancer metastasis. EMBO Rep 2025; 26:2574-2596. [PMID: 40185977 DOI: 10.1038/s44319-025-00437-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 02/22/2025] [Accepted: 03/07/2025] [Indexed: 04/07/2025] Open
Abstract
Genes regulating the finger-like cellular protrusions-filopodia have long been implicated in cancer metastasis. However, depleting the flat lamellipodia but retaining filopodia drastically hampers cell migration on spread surface, obscuring the role of filopodia in cell motility. It has been noticed recently that cells under confinement may employ distinct migratory machineries. However, the regulating factors have mainly been focused on cell blebbing, nuclear deformation and cell rear contractility, without much emphasis on cell protrusions and even less on filopodia. Here, by micropore-based screening, we identified espin as an active regulator for confined migration and that its overexpression was associated with metastasis. In comparison to fascin, espin showed stronger actin bundling in vitro and induced shorter and thicker filopodia in cells. Combining the imaging-compatible microchannels and DNA-based tension probes, we uncovered that espin overexpression induced excessive filopodia at the leading edge and along the sides, exerting force for confined migration. Our results demonstrate an important role for filopodia and the regulating protein-espin in confined cell migration and shed new light on cytoskeletal mechanisms underlying metastasis.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
- International Cancer Institute, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Peng Shi
- Cancer Institute, Suzhou Medical College, Soochow University, 215000, Suzhou, Jiangsu, China.
| | - Geyao Liu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
- International Cancer Institute, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Wei Chen
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei Province, China
| | - Ya-Jun Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, China
| | - Yiping Hu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
- International Cancer Institute, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Ao Yang
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Tonghua Wei
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
- International Cancer Institute, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Yu-Chen Chen
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, China
| | - Ling Liang
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Zheng Liu
- The Institute for Advanced Studies, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, Hubei Province, China.
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, China.
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China.
- International Cancer Institute, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, 100191, Beijing, China.
| |
Collapse
|
2
|
McAtee C, Patel M, Hoshino D, Sung BH, von Lersner A, Shi M, Hong NH, Young A, Krystofiak E, Zijlstra A, Weaver AM. Secreted exosomes induce filopodia formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.20.604139. [PMID: 40161676 PMCID: PMC11952364 DOI: 10.1101/2024.07.20.604139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Filopodia are dynamic adhesive cytoskeletal structures that are critical for directional sensing, polarization, cell-cell adhesion, and migration of diverse cell types. Filopodia are also critical for neuronal synapse formation. While dynamic rearrangement of the actin cytoskeleton is known to be critical for filopodia biogenesis, little is known about the upstream extracellular signals. Here, we identify secreted exosomes as potent regulators of filopodia formation. Inhibition of exosome secretion inhibited the formation and stabilization of filopodia in both cancer cells and neurons and inhibited subsequent synapse formation by neurons. Rescue experiments with purified small and large extracellular vesicles (EVs) identified exosome-enriched small EVs (SEVs) as having potent filopodia-inducing activity. Proteomic analyses of cancer cell-derived SEVs identified the TGF-β family coreceptor endoglin as a key SEV-enriched cargo that regulates filopodia. Cancer cell endoglin levels also affected filopodia-dependent behaviors, including metastasis of cancer cells in chick embryos and 3D migration in collagen gels. As neurons do not express endoglin, we performed a second proteomics experiment to identify SEV cargoes regulated by endoglin that might promote filopodia in both cell types. We discovered a single SEV cargo that was altered in endoglin-KD cancer SEVs, the transmembrane protein Thrombospondin Type 1 Domain Containing 7A (THSD7A). We further found that both cancer cell and neuronal SEVs carry THSD7A and that add-back of purified THSD7A is sufficient to rescue filopodia defects of both endoglin-KD cancer cells and exosome-inhibited neurons. We also find that THSD7A induces filopodia formation through activation of the Rho GTPase, Cdc42. These findings suggest a new model for filopodia formation, triggered by exosomes carrying THSD7A.
Collapse
Affiliation(s)
- Caitlin McAtee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, USA
- Center for Extracellular Vesicle Research, Vanderbilt University School of Medicine, Nashville, USA
| | - Mikin Patel
- Department of Biological Sciences, Vanderbilt University, Nashville, USA
| | | | - Bong Hwan Sung
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, USA
- Center for Extracellular Vesicle Research, Vanderbilt University School of Medicine, Nashville, USA
| | - Ariana von Lersner
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, USA
- Center for Extracellular Vesicle Research, Vanderbilt University School of Medicine, Nashville, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, USA
| | - Mingjian Shi
- Department of Biological Sciences, Vanderbilt University, Nashville, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, USA
| | - Nan Hyung Hong
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, USA
| | - Anna Young
- Department of Biological Sciences, Vanderbilt University, Nashville, USA
| | - Evan Krystofiak
- Cell Imaging Shared Resource EM Facility, Vanderbilt University, Nashville, Tennessee, USA
| | - Andries Zijlstra
- Center for Extracellular Vesicle Research, Vanderbilt University School of Medicine, Nashville, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, USA
| | - Alissa M. Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, USA
- Center for Extracellular Vesicle Research, Vanderbilt University School of Medicine, Nashville, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, USA
| |
Collapse
|
3
|
Eddington C, Titus MA. The filopodial myosin DdMyo7 is a slow, calcium-regulated motor. J Biol Chem 2025; 301:108371. [PMID: 40043952 DOI: 10.1016/j.jbc.2025.108371] [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: 11/27/2023] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 05/08/2025] Open
Abstract
MyTH4-FERM (MF) myosins are a family of molecular motors with critical roles in the formation and organization of thin membrane protrusions supported by parallel bundles of actin - filopodia, microvilli, and stereocilia. The amoeboid MF myosin DdMyo7 is essential for filopodia formation but its mechanism of action is unknown. The motor properties of a forced-dimer of the DdMyo7 motor were characterized using an in vitro motility assay to address this question. The DdMyo7 motor associates with two different light chains, the Dictyostelium calmodulins CalA and CalB, whose binding is shown to be sensitive to the presence of calcium. Total internal reflection fluorescence motility assays of the dimerized DdMyo7 motor reveal that it is a slow, processive motor that moves along actin at ∼ 40 nm/sec, and the activity of the motor is significantly reduced in the presence of Ca2+. The speed of DdMyo7 is similar to that of other Myo7 family members such as human Myo7A and fly DmMyo7A, but is at least 10-fold slower than the mammalian filopodial MF myosin, Myo10. The results show that evolutionarily distant native filopodial myosins can promote filopodia elongation using motors with distinct properties, revealing diverse mechanisms of myosin-based filopodia formation.
Collapse
Affiliation(s)
- Casey Eddington
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, USA; Graduate Program in Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Margaret A Titus
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, USA; Graduate Program in Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
4
|
Hawley J, Lea R, Biga V, Papalopulu N, Manning C. Cellular signalling protrusions enable dynamic distant contacts in spinal cord neurogenesis. Biol Open 2025; 14:bio061765. [PMID: 39835478 PMCID: PMC11789279 DOI: 10.1242/bio.061765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
In the developing mouse ventral spinal cord, HES5, a transcription factor downstream of Notch signalling, is expressed as evenly spaced clusters of high HES5-expressing neural progenitor cells along the dorsoventral axis. While Notch signalling requires direct membrane contact for its activation, we have previously shown mathematically that contact needs to extend beyond neighbouring cells for the HES5 pattern to emerge. However, the presence of cellular structures that could enable such long-distance signalling was unclear. Here, we report that cellular protrusions are present all along the apicobasal axis of individual neural progenitor cells. Through live imaging, we show that these protrusions dynamically extend and retract reaching lengths of up to ∼20 µm, enough to extend membrane contact beyond adjacent cells. The Notch ligand DLL1 was found to colocalise with protrusions, further supporting the idea that Notch signalling can be transduced at a distance. The effect of protrusions on the HES5 pattern was tested by reducing the density of protrusions using the CDC42 inhibitor ML141, leading to a tendency to decrease the distance between high HES5 cell clusters. However, this tendency was not significant and leaves an open question about their role in the fine-grained organisation of neurogenesis.
Collapse
Affiliation(s)
- Joshua Hawley
- Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Robert Lea
- Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Veronica Biga
- Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Nancy Papalopulu
- Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Cerys Manning
- Faculty of Biology Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
5
|
Shangguan J, Rock RS. Hundreds of myosin 10s are pushed to the tips of filopodia and could cause traffic jams on actin. eLife 2024; 12:RP90603. [PMID: 39480891 PMCID: PMC11527427 DOI: 10.7554/elife.90603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Abstract
Myosin 10 (Myo10) is a motor protein known for its role in filopodia formation. Although Myo10-driven filopodial dynamics have been characterized, there is no information about the absolute number of Myo10 molecules during the filopodial lifecycle. To better understand molecular stoichiometries and packing restraints in filopodia, we measured Myo10 abundance in these structures. We combined SDS-PAGE densitometry with epifluorescence microscopy to quantitate HaloTag-labeled Myo10 in U2OS cells. About 6% of total intracellular Myo10 localizes to filopodia, where it enriches at opposite cellular ends. Hundreds of Myo10s are in a typical filopodium, and their distribution across filopodia is log-normal. Some filopodial tips even contain more Myo10 than accessible binding sites on the actin filament bundle. Live-cell movies reveal a dense cluster of over a hundred Myo10 molecules that initiates filopodial elongation. Hundreds of Myo10 molecules continue to accumulate during filopodial growth, but accumulation ceases when retraction begins. Rates of filopodial elongation, second-phase elongation, and retraction are inversely related to Myo10 quantities. Our estimates of Myo10 molecules in filopodia provide insight into the physics of packing Myo10, its cargo, and other filopodia-associated proteins in narrow membrane compartments. Our protocol provides a framework for future work analyzing Myo10 abundance and distribution upon perturbation.
Collapse
Affiliation(s)
- Julia Shangguan
- Department of Biochemistry and Molecular Biology, University of ChicagoChicagoUnited States
| | - Ronald S Rock
- Department of Biochemistry and Molecular Biology, The Institute for Biophysical Dynamics, University of ChicagoChicagoUnited States
| |
Collapse
|
6
|
Li T, Song Y, Wei L, Song X, Duan R. Disulfidptosis: a novel cell death modality induced by actin cytoskeleton collapse and a promising target for cancer therapeutics. Cell Commun Signal 2024; 22:491. [PMID: 39394612 PMCID: PMC11470700 DOI: 10.1186/s12964-024-01871-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024] Open
Abstract
Disulfidptosis is a novel discovered form of programmed cell death (PCD) that diverges from apoptosis, necroptosis, ferroptosis, and cuproptosis, stemming from disulfide stress-induced cytoskeletal collapse. In cancer cells exhibiting heightened expression of the solute carrier family 7 member 11 (SLC7A11), excessive cystine importation and reduction will deplete nicotinamide adenine dinucleotide phosphate (NADPH) under glucose deprivation, followed by an increase in intracellular disulfide stress and aberrant disulfide bond formation within actin networks, ultimately culminating in cytoskeletal collapse and disulfidptosis. Disulfidptosis involves crucial physiological processes in eukaryotic cells, such as cystine and glucose uptake, NADPH metabolism, and actin dynamics. The Rac1-WRC pathway-mediated actin polymerization is also implicated in this cell death due to its contribution to disulfide bond formation. However, the precise mechanisms underlying disulfidptosis and its role in tumors are not well understood. This is probably due to the multifaceted functionalities of SLC7A11 within cells and the complexities of the downstream pathways driving disulfidptosis. This review describes the critical roles of SLC7A11 in cells and summarizes recent research advancements in the potential pathways of disulfidptosis. Moreover, the less-studied aspects of this newly discovered cell death process are highlighted to stimulate further investigations in this field.
Collapse
Affiliation(s)
- Tianyi Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Song
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Lijuan Wei
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Xiangyi Song
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Ruifeng Duan
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China.
| |
Collapse
|
7
|
Lambert C, Karger M, Steffen A, Tang Y, Döring H, Stradal TEB, Lappalainen P, Faix J, Bieling P, Rottner K. Differential interference with actin-binding protein function by acute Cytochalasin B. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.611976. [PMID: 39372773 PMCID: PMC11451763 DOI: 10.1101/2024.09.11.611976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Dynamic actin filament remodeling is crucial for a plethora of fundamental cell biological processes, ranging from cell division and migration to cell communication, intracellular trafficking or tissue development. Cytochalasin B and -D are fungal secondary metabolites frequently used for interference with such processes. Although generally assumed to block actin filament polymerization at their rapidly growing barbed ends and compete with regulators at these sites, our molecular understanding of their precise effects in dynamic actin structures is scarce. Here we combine live cell imaging and analysis of fluorescent actin-binding protein dynamics with acute treatment of lamellipodia in migrating cells with cytochalasin B. Our results show that in spite of an abrupt halt of lamellipodium protrusion, cytochalasin B affects various actin filament barbed end-binding proteins in a differential fashion. Cytochalasin B enhances instead of diminishes the accumulation of prominent barbed end-binding factors such as Ena/VASP family proteins and heterodimeric capping protein (CP) in the lamellipodium. Similar results were obtained with cytochalasin D. All these effects are highly specific, as cytochalasin-induced VASP accumulation requires the presence of CP, but not vice versa , and coincides with abrogation of both actin and VASP turnover. Cytochalasin B can also increase apparent barbed end interactions with the actin-binding β-tentacle of CP and partially mimic its Arp2/3 complex-promoting activity in the lamellipodium. In conclusion, our results reveal a new spectrum of cytochalasin activities on barbed end-binding factors, with important implications for the interpretation of their effects on dynamic actin structures.
Collapse
|
8
|
Nozumi M, Sato Y, Nishiyama-Usuda M, Igarashi M. Identification of z-axis filopodia in growth cones using super-resolution microscopy. J Neurochem 2024; 168:2974-2988. [PMID: 38946488 DOI: 10.1111/jnc.16162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/02/2024] [Accepted: 06/10/2024] [Indexed: 07/02/2024]
Abstract
A growth cone is a highly motile tip of an extending axon that is crucial for neural network formation. Three-dimensional-structured illumination microscopy, a type of super-resolution light microscopy with a resolution that overcomes the optical diffraction limitation (ca. 200 nm) of conventional light microscopy, is well suited for studying the molecular dynamics of intracellular events. Using this technique, we discovered a novel type of filopodia distributed along the z-axis ("z-filopodia") within the growth cone. Z-filopodia were typically oriented in the direction of axon growth, not attached to the substratum, protruded spontaneously without microtubule invasion, and had a lifetime that was considerably shorter than that of conventional filopodia. Z-filopodia formation and dynamics were regulated by actin-regulatory proteins, such as vasodilator-stimulated phosphoprotein, fascin, and cofilin. Chromophore-assisted laser inactivation of cofilin induced the rapid turnover of z-filopodia. An axon guidance receptor, neuropilin-1, was concentrated in z-filopodia and was transported together with them, whereas its ligand, semaphorin-3A, was selectively bound to them. Membrane domains associated with z-filopodia were also specialized and resembled those of lipid rafts, and their behaviors were closely related to those of neuropilin-1. The results suggest that z-filopodia have unique turnover properties, and unlike xy-filopodia, do not function as force-generating structures for axon extension.
Collapse
Affiliation(s)
- Motohiro Nozumi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine, and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Yuta Sato
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine, and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Miyako Nishiyama-Usuda
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine, and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine, and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
9
|
Shangguan J, Rock RS. Hundreds of myosin 10s are pushed to the tips of filopodia and could cause traffic jams on actin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.26.546598. [PMID: 37425746 PMCID: PMC10327019 DOI: 10.1101/2023.06.26.546598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Myosin 10 (Myo10) is a motor protein known for its role in filopodia formation. Although Myo10-driven filopodial dynamics have been characterized, there is no information about the absolute number of Myo10 molecules during the filopodial lifecycle. To better understand molecular stoichiometries and packing restraints in filopodia, we measured Myo10 abundance in these structures. We combined SDS-PAGE densitometry with epifluorescence microscopy to quantitate HaloTag-labeled Myo10 in U2OS cells. About 6% of total intracellular Myo10 localizes to filopodia, where it enriches at opposite cellular ends. Hundreds of Myo10s are in a typical filopodium, and their distribution across filopodia is log-normal. Some filopodial tips even contain more Myo10 than accessible binding sites on the actin filament bundle. Live-cell movies reveal a dense cluster of over a hundred Myo10 molecules that initiates filopodial elongation. Hundreds of Myo10 molecules continue to accumulate during filopodial growth, but accumulation ceases when retraction begins. Rates of filopodial elongation, second-phase elongation, and retraction are inversely related to Myo10 quantities. Our estimates of Myo10 molecules in filopodia provide insight into the physics of packing Myo10, its cargo, and other filopodia-associated proteins in narrow membrane compartments. Our protocol provides a framework for future work analyzing Myo10 abundance and distribution upon perturbation.
Collapse
Affiliation(s)
- Julia Shangguan
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois, USA
| | - Ronald S Rock
- Department of Biochemistry and Molecular Biology, The Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
10
|
Jawahar A, Vermeil J, Heuvingh J, du Roure O, Piel M. The third dimension of the actin cortex. Curr Opin Cell Biol 2024; 89:102381. [PMID: 38905917 DOI: 10.1016/j.ceb.2024.102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/15/2024] [Accepted: 05/24/2024] [Indexed: 06/23/2024]
Abstract
The actin cortex, commonly described as a thin 2-dimensional layer of actin filaments beneath the plasma membrane, is beginning to be recognized as part of a more dynamic and three-dimensional composite material. In this review, we focus on the elements that contribute to the three-dimensional architecture of the actin cortex. We also argue that actin-rich structures such as filopodia and stress fibers can be viewed as specialized integral parts of the 3D actin cortex. This broadens our definition of the cortex, shifting from its simplified characterization as a thin, two-dimensional layer of actin filaments.
Collapse
Affiliation(s)
- Anumita Jawahar
- Physique et Mécanique des Milieux Hétérogènes, ESPCI Paris, PSL University, CNRS, Université Paris Cité, Sorbonne Université, Paris, France; Institut Curie and Institut Pierre Gilles de Gennes, PSL University, CNRS, Paris, France.
| | - Joseph Vermeil
- Physique et Mécanique des Milieux Hétérogènes, ESPCI Paris, PSL University, CNRS, Université Paris Cité, Sorbonne Université, Paris, France; 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, Université Paris Cité, Sorbonne Université, Paris, France
| | - Olivia du Roure
- Physique et Mécanique des Milieux Hétérogènes, ESPCI Paris, PSL University, CNRS, Université Paris Cité, Sorbonne Université, Paris, France
| | - Matthieu Piel
- Institut Curie and Institut Pierre Gilles de Gennes, PSL University, CNRS, Paris, France
| |
Collapse
|
11
|
Iorio R, Petricca S, Mattei V, Delle Monache S. Horizontal mitochondrial transfer as a novel bioenergetic tool for mesenchymal stromal/stem cells: molecular mechanisms and therapeutic potential in a variety of diseases. J Transl Med 2024; 22:491. [PMID: 38790026 PMCID: PMC11127344 DOI: 10.1186/s12967-024-05047-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/29/2024] [Indexed: 05/26/2024] Open
Abstract
Intercellular mitochondrial transfer (MT) is a newly discovered form of cell-to-cell signalling involving the active incorporation of healthy mitochondria into stressed/injured recipient cells, contributing to the restoration of bioenergetic profile and cell viability, reduction of inflammatory processes and normalisation of calcium dynamics. Recent evidence has shown that MT can occur through multiple cellular structures and mechanisms: tunneling nanotubes (TNTs), via gap junctions (GJs), mediated by extracellular vesicles (EVs) and other mechanisms (cell fusion, mitochondrial extrusion and migrasome-mediated mitocytosis) and in different contexts, such as under physiological (tissue homeostasis and stemness maintenance) and pathological conditions (hypoxia, inflammation and cancer). As Mesenchimal Stromal/ Stem Cells (MSC)-mediated MT has emerged as a critical regulatory and restorative mechanism for cell and tissue regeneration and damage repair in recent years, its potential in stem cell therapy has received increasing attention. In particular, the potential therapeutic role of MSCs has been reported in several articles, suggesting that MSCs can enhance tissue repair after injury via MT and membrane vesicle release. For these reasons, in this review, we will discuss the different mechanisms of MSCs-mediated MT and therapeutic effects on different diseases such as neuronal, ischaemic, vascular and pulmonary diseases. Therefore, understanding the molecular and cellular mechanisms of MT and demonstrating its efficacy could be an important milestone that lays the foundation for future clinical trials.
Collapse
Affiliation(s)
- Roberto Iorio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100, L'Aquila, Italy
| | - Sabrina Petricca
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100, L'Aquila, Italy
| | - Vincenzo Mattei
- Dipartimento di Scienze della Vita, Della Salute e delle Professioni Sanitarie, Link Campus University, Via del Casale di San Pio V 44, 00165, Rome, Italy.
| | - Simona Delle Monache
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100, L'Aquila, Italy.
| |
Collapse
|
12
|
Zhou C, Huang M, Wang S, Chu S, Zhang Z, Chen N. Tunneling nanotubes: The transport highway for astrocyte-neuron communication in the central nervous system. Brain Res Bull 2024; 209:110921. [PMID: 38447659 DOI: 10.1016/j.brainresbull.2024.110921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/08/2024]
Abstract
Tunneling nanotubes (TNTs) have emerged as pivotal structures for intercellular communication, enabling the transfer of cellular components across distant cells. Their involvement in neurological disorders has attracted considerable scientific interest. This review delineates the functions of TNTs within the central nervous system, examining their role in the transmission of bioenergetic substrates, and signaling molecules, and their multifaceted impact on both physiological and pathological processes, with an emphasis on neurodegenerative diseases. The review highlights the selectivity and specificity of TNTs as dedicated pathways for intercellular cargo delivery, particularly under stress conditions that provoke increased TNT formation. The potential of TNTs as therapeutic targets is explored in depth. We pay particular attention to the interactions between astrocytes and neurons mediated by TNTs, which are fundamental to brain architecture and function. Dysfunctions in these interactions are implicated in the spread of protein aggregates and mitochondrial anomalies, contributing to the pathogenesis of neurodegenerative diseases. The review culminates with a synthesis of the current understanding of TNT biology and identifies research gaps, advocating for intensified exploration into TNTs as a promising therapeutic frontier.
Collapse
Affiliation(s)
- Cuixiang Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Min Huang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shasha Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Naihong Chen
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
13
|
Ebrahimi AM, Gawlik W, Wojciechowski AM, Rajfur Z. Cell-particles interaction - selective uptake and transport of microdiamonds. Commun Biol 2024; 7:318. [PMID: 38480800 PMCID: PMC10937934 DOI: 10.1038/s42003-024-05974-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 02/26/2024] [Indexed: 03/17/2024] Open
Abstract
Diamond particles have recently emerged as novel agents in cellular studies because of their superb biocompatibility. Their unique characteristics, including small size and the presence of fluorescent color centers, stimulate many important applications. However, the mechanism of interaction between cells and diamond particles-uptake, transport, and final localization within cells-is not yet fully understood. Herein, we show a novel, to the best of our knowledge, cell behavior wherein cells actively target and uptake diamond particles rather than latex beads from their surroundings, followed by their active transport within cells. Furthermore, we demonstrate that myosin-X is involved in cell-particle interaction, while myosin-II does not participate in particle uptake and transport. These results can have important implications for drug delivery and improve sensing methods that use diamond particles.
Collapse
Affiliation(s)
- Armin M Ebrahimi
- Marian Smoluchowski Institute of Physics, Jagiellonian University, 30-348, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348, Kraków, Poland
| | - Wojciech Gawlik
- Marian Smoluchowski Institute of Physics, Jagiellonian University, 30-348, Kraków, Poland
| | - Adam M Wojciechowski
- Marian Smoluchowski Institute of Physics, Jagiellonian University, 30-348, Kraków, Poland.
| | - Zenon Rajfur
- Marian Smoluchowski Institute of Physics, Jagiellonian University, 30-348, Kraków, Poland.
- Jagiellonian Center of Biomedical Imaging, Jagiellonian University, 30-348, Kraków, Poland.
| |
Collapse
|
14
|
Eddington C, Schwartz JK, Titus MA. filoVision - using deep learning and tip markers to automate filopodia analysis. J Cell Sci 2024; 137:jcs261274. [PMID: 38264939 PMCID: PMC10941656 DOI: 10.1242/jcs.261274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024] Open
Abstract
Filopodia are slender, actin-filled membrane projections used by various cell types for environment exploration. Analyzing filopodia often involves visualizing them using actin, filopodia tip or membrane markers. Due to the diversity of cell types that extend filopodia, from amoeboid to mammalian, it can be challenging for some to find a reliable filopodia analysis workflow suited for their cell type and preferred visualization method. The lack of an automated workflow capable of analyzing amoeboid filopodia with only a filopodia tip label prompted the development of filoVision. filoVision is an adaptable deep learning platform featuring the tools filoTips and filoSkeleton. filoTips labels filopodia tips and the cytosol using a single tip marker, allowing information extraction without actin or membrane markers. In contrast, filoSkeleton combines tip marker signals with actin labeling for a more comprehensive analysis of filopodia shafts in addition to tip protein analysis. The ZeroCostDL4Mic deep learning framework facilitates accessibility and customization for different datasets and cell types, making filoVision a flexible tool for automated analysis of tip-marked filopodia across various cell types and user data.
Collapse
Affiliation(s)
- Casey Eddington
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
- Graduate Program in Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jessica K. Schwartz
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Margaret A. Titus
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
- Graduate Program in Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
15
|
Yim YI, Pedrosa A, Wu X, Chinthalapudi K, Cheney RE, Hammer JA. Mechanisms underlying Myosin 10's contribution to the maintenance of mitotic spindle bipolarity. Mol Biol Cell 2024; 35:ar14. [PMID: 38019611 PMCID: PMC10881153 DOI: 10.1091/mbc.e23-07-0282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Myosin 10 (Myo10) couples microtubules and integrin-based adhesions to movement along actin filaments via its microtubule-binding MyTH4 domain and integrin-binding FERM domain, respectively. Here we show that Myo10-depleted HeLa cells and mouse embryo fibroblasts (MEFs) both exhibit a pronounced increase in the frequency of multipolar spindles. Staining of unsynchronized metaphase cells showed that the primary driver of spindle multipolarity in Myo10-depleted MEFs and in Myo10-depleted HeLa cells lacking supernumerary centrosomes is pericentriolar material (PCM) fragmentation, which creates y-tubulin-positive acentriolar foci that serve as extra spindle poles. For HeLa cells possessing supernumerary centrosomes, Myo10 depletion further accentuates spindle multipolarity by impairing the clustering of the extra spindle poles. Complementation experiments show that Myo10 must interact with both microtubules and integrins to promote PCM/pole integrity. Conversely, Myo10 only needs interact with integrins to promote supernumerary centrosome clustering. Importantly, images of metaphase Halo-Myo10 knockin cells show that the myosin localizes exclusively to the spindle and the tips of adhesive retraction fibers. We conclude that Myo10 promotes PCM/pole integrity in part by interacting with spindle microtubules, and that it promotes supernumerary centrosome clustering by supporting retraction fiber-based cell adhesion, which likely serves to anchor the microtubule-based forces driving pole focusing.
Collapse
Affiliation(s)
- Yang-In Yim
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Antonio Pedrosa
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xufeng Wu
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
16
|
Davoudi P, Do DN, Colombo S, Rathgeber B, Sargolzaei M, Plastow G, Wang Z, Hu G, Valipour S, Miar Y. Genome-wide association studies for economically important traits in mink using copy number variation. Sci Rep 2024; 14:24. [PMID: 38167844 PMCID: PMC10762091 DOI: 10.1038/s41598-023-50497-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Copy number variations (CNVs) are structural variants consisting of duplications and deletions of DNA segments, which are known to play important roles in the genetics of complex traits in livestock species. However, CNV-based genome-wide association studies (GWAS) have remained unexplored in American mink. Therefore, the purpose of the current study was to investigate the association between CNVs and complex traits in American mink. A CNV-based GWAS was performed with the ParseCNV2 software program using deregressed estimated breeding values of 27 traits as pseudophenotypes, categorized into traits of growth and feed efficiency, reproduction, pelt quality, and Aleutian disease tests. The study identified a total of 10,137 CNVs (6968 duplications and 3169 deletions) using the Affymetrix Mink 70K single nucleotide polymorphism (SNP) array in 2986 American mink. The association analyses identified 250 CNV regions (CNVRs) associated with at least one of the studied traits. These CNVRs overlapped with a total of 320 potential candidate genes, and among them, several genes have been known to be related to the traits such as ARID1B, APPL1, TOX, and GPC5 (growth and feed efficiency traits); GRM1, RNASE10, WNT3, WNT3A, and WNT9B (reproduction traits); MYO10, and LIMS1 (pelt quality traits); and IFNGR2, APEX1, UBE3A, and STX11 (Aleutian disease tests). Overall, the results of the study provide potential candidate genes that may regulate economically important traits and therefore may be used as genetic markers in mink genomic breeding programs.
Collapse
Affiliation(s)
- Pourya Davoudi
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Duy Ngoc Do
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Stefanie Colombo
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Bruce Rathgeber
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Mehdi Sargolzaei
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
- Select Sires Inc., Plain City, OH, USA
| | - Graham Plastow
- Livestock Gentec, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Zhiquan Wang
- Livestock Gentec, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Guoyu Hu
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Shafagh Valipour
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Younes Miar
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada.
| |
Collapse
|
17
|
Panda SK, Torsiello M, Rehman A, Desiderio V, Del Vecchio V. Mitochondrial Transfer Between Mesenchymal Stem Cells and Cancer Cells. Methods Mol Biol 2024; 2835:39-48. [PMID: 39105904 DOI: 10.1007/978-1-0716-3995-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Mitochondrial transfer (MT) is a biological process that allows a donor cell to horizontally share its own mitochondria with a recipient cell. Mitochondria are highly dynamic membrane-bound sub-cellular organelles prominently involved in the regulation of the cell energy balance, calcium homeostasis, and apoptotic machinery activation. They physiologically undergo fusion and fission processes in response to the cell requirement, with a continuous morphological re-arrangement. This structural and functional plasticity is at the basis of the MT, described in tissue regeneration, cardiac and neurological diseases, as well as in cancer. Here, the MT has been observed in the tumor micro-environment (TME) from the adipose-derived stem cells (ASCs) to the cancer cells, eventually reverting the lack of the mitochondria respiration function, or enhancing their motility and drug resistance. In this chapter, we outline some key protocols for evaluating this exciting phenomenon of MT. These methodological and technical approaches are very important, considering all the limitations that scientists constantly face, especially in this field of the research.
Collapse
Affiliation(s)
- S Kumar Panda
- Department of Experimental Medicine, Histology and Embryology Section, University of Campania "L. Vanvitelli", Naples, Italy
| | - M Torsiello
- Department of Experimental Medicine, Histology and Embryology Section, University of Campania "L. Vanvitelli", Naples, Italy
| | - A Rehman
- Department of Experimental Medicine, Histology and Embryology Section, University of Campania "L. Vanvitelli", Naples, Italy
| | - Vincenzo Desiderio
- Department of Experimental Medicine, Histology and Embryology Section, University of Campania "L. Vanvitelli", Naples, Italy
| | - V Del Vecchio
- Department of Experimental Medicine, Histology and Embryology Section, University of Campania "L. Vanvitelli", Naples, Italy.
| |
Collapse
|
18
|
Cirilo JA, Liao X, Perrin BJ, Yengo CM. The dynamics of actin protrusions can be controlled by tip-localized myosin motors. J Biol Chem 2024; 300:105516. [PMID: 38042485 PMCID: PMC10801316 DOI: 10.1016/j.jbc.2023.105516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023] Open
Abstract
Class III myosins localize to inner ear hair cell stereocilia and are thought to be crucial for stereocilia length regulation. Mutations within the motor domain of MYO3A that disrupt its intrinsic motor properties have been associated with non-syndromic hearing loss, suggesting that the motor properties of MYO3A are critical for its function within stereocilia. In this study, we investigated the impact of a MYO3A hearing loss mutation, H442N, using both in vitro motor assays and cell biological studies. Our results demonstrate the mutation causes a dramatic increase in intrinsic motor properties, actin-activated ATPase and in vitro actin gliding velocity, as well as an increase in actin protrusion extension velocity. We propose that both "gain of function" and "loss of function" mutations in MYO3A can impair stereocilia length regulation, which is crucial for stereocilia formation during development and normal hearing. Furthermore, we generated chimeric MYO3A constructs that replace the MYO3A motor and neck domain with the motor and neck domain of other myosins. We found that duty ratio, fraction of ATPase cycle myosin is strongly bound to actin, is a critical motor property that dictates the ability to tip localize within filopodia. In addition, in vitro actin gliding velocities correlated extremely well with filopodial extension velocities over a wide range of gliding and extension velocities. Taken together, our data suggest a model in which tip-localized myosin motors exert force that slides the membrane tip-ward, which can combat membrane tension and enhance the actin polymerization rate that ultimately drives protrusion elongation.
Collapse
Affiliation(s)
- Joseph A Cirilo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Xiayi Liao
- Department of Biology, Indiana University - Purdue University, Indianapolis, Indiana, USA
| | - Benjamin J Perrin
- Department of Biology, Indiana University - Purdue University, Indianapolis, Indiana, USA
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
| |
Collapse
|
19
|
Suarez C, Winkelman JD, Harker AJ, Ye HJ, McCall PM, Morganthaler AN, Gardel ML, Kovar DR. Reconstitution of the transition from a lamellipodia- to filopodia-like actin network with purified proteins. Eur J Cell Biol 2023; 102:151367. [PMID: 37890285 DOI: 10.1016/j.ejcb.2023.151367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
How cells utilize complex mixtures of actin binding proteins to assemble and maintain functionally diverse actin filament networks with distinct architectures and dynamics within a common cytoplasm is a longstanding question in cell biology. A compelling example of complex and specialized actin structures in cells are filopodia which sense extracellular chemical and mechanical signals to help steer motile cells. Filopodia have distinct actin architecture, composed of long, parallel actin filaments bundled by fascin, which form finger-like membrane protrusions. Elongation of the parallel actin filaments in filopodia can be mediated by two processive actin filament elongation factors, formin and Ena/VASP, which localize to the tips of filopodia. There remains debate as to how the architecture of filopodia are generated, with one hypothesis proposing that filopodia are generated from the lamellipodia, which consists of densely packed, branched actin filaments nucleated by Arp2/3 complex and kept short by capping protein. It remains unclear if different actin filament elongation factors are necessary and sufficient to facilitate the emergence of filopodia with diverse characteristics from a highly dense network of short-branched capped filaments. To address this question, we combined bead motility and micropatterning biomimetic assays with multi-color Total Internal Reflection Fluorescence microscopy imaging, to successfully reconstitute the formation of filopodia-like networks (FLN) from densely-branched lamellipodia-like networks (LLN) with eight purified proteins (actin, profilin, Arp2/3 complex, Wasp pWA, fascin, capping protein, VASP and formin mDia2). Saturating capping protein concentrations inhibit FLN assembly, but the addition of either formin or Ena/VASP differentially rescues the formation of FLN from LLN. Specifically, we found that formin/mDia2-generated FLNs are relatively long and lack capping protein, whereas VASP-generated FLNs are comparatively short and contain capping protein, indicating that the actin elongation factor can affect the architecture and composition of FLN emerging from LLN. Our biomimetic reconstitution systems reveal that formin or VASP are necessary and sufficient to induce the transition from a LLN to a FLN, and establish robust in vitro platforms to investigate FLN assembly mechanisms.
Collapse
Affiliation(s)
- Cristian Suarez
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA.
| | - Jonathan D Winkelman
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Alyssa J Harker
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Hannah J Ye
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Patrick M McCall
- Department of Physics, The University of Chicago, Chicago, IL 60637, USA; James Franck Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Alisha N Morganthaler
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Margaret L Gardel
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Department of Physics, The University of Chicago, Chicago, IL 60637, USA; James Franck Institute, The University of Chicago, Chicago, IL 60637, USA; Pritzker School for Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
20
|
Guin S, Halder SC, Ghosh SR, Jana AD. Electric field-driven up-and-down motion of the flexible tail of Al 13+ cluster system-a nano-scale flipper. J Mol Model 2023; 29:383. [PMID: 37987928 DOI: 10.1007/s00894-023-05781-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 11/03/2023] [Indexed: 11/22/2023]
Abstract
CONTEXT Dynamic metal nanoclusters have become a hot area of research in the field of nanoscience and nanotechnology due to their potential applications in micro devices. One such dynamic cluster is a quasi-planar ground state (GS) Al13+ cluster which exhibits an electric field driven up and down flipping motion of the flexible tail which oscillates with respect to the mean plane. A Car-Parrinello molecular dynamics (CPMD) simulation has been carried out to understand the nature of dynamics of the cluster. CPMD simulation study reveals that the flexible tail region of the Al13+ isomeric system (two ground states M1, M2 and a transition state TS connecting them) can be engaged in a systematic up down flipping motion by the application of a transverse electric field. A saw tooth electric field of amplitude 5.19 V/nm is sufficient to induce the up-and-down flipping oscillation of the cluster, which has an average oscillation frequency of around 20 THz. AIM, NICS and AdNDP analyses also have been carried out to understand the fluxional nature of the cluster from the electronic structural perspective. Electronic structural analysis of selected optimized intermediate states in the presence of transverse electric field has also been analyzed to correlate the electronic structure with the dynamic nature of the cluster. METHODS Single-point energies of all intermediate states between two minima of Al13+ clusters connected through a transition state cluster. Optimized geometries of Al13+ clusters in the presence of electric field of different strengths have been carried out by using the Gaussian 03 package. 6-311 + G(d) basis set and B3LYP hybrid density functional have been utilized for these studies. To establish the flipping motion, Car-Parrinello molecular dynamics (CPMD) has been performed using the cp.x module of the Quantum ESPRESSO 6.3.0 program package using the Perdew-Burke-Ernzerhof (PBE) functional, plane-wave basis set and ultrasoft pseudopotentials. ORTEP-3 and POV ray-3.7 software packages have been used for visualization and graphics generation. Atoms in molecule (AIM), Adaptive Natural Density Partitioning (AdNDP) analysis have been carried out using Multiwfn 3.7 program package.
Collapse
Affiliation(s)
- Surajit Guin
- Department of Physics, Behala College, Parnasree, Kolkata, 700060, West Bengal, India
| | - Sasthi Charan Halder
- Department of Physics, Behala College, Parnasree, Kolkata, 700060, West Bengal, India
| | - Sourav Ranjan Ghosh
- Department of Physics, Behala College, Parnasree, Kolkata, 700060, West Bengal, India
- Department of Physics, Heritage Institute of Technology, Kolkata, 700107, India
| | - Atish Dipankar Jana
- Department of Physics, Behala College, Parnasree, Kolkata, 700060, West Bengal, India.
- Institute of Astronomy Space and Earth Science, AJ 316, Salt Lake, Kolkata, 700091, India.
| |
Collapse
|
21
|
Nishita M, Kamizaki K, Hoshi K, Aruga K, Nishikaku I, Shibuya H, Matsumoto K, Minami Y. Rho family small GTPase Rif regulates Wnt5a-Ror1-Dvl2 signaling and promotes lung adenocarcinoma progression. J Biol Chem 2023; 299:105248. [PMID: 37703992 PMCID: PMC10570955 DOI: 10.1016/j.jbc.2023.105248] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023] Open
Abstract
Rho in filopodia (Rif), a member of the Rho family of small GTPases, induces filopodia formation primarily on the dorsal surface of cells; however, its function remains largely unclear. Here, we show that Rif interacts with Ror1, a receptor for Wnt5a that can also induce dorsal filopodia. Our immunohistochemical analysis revealed a high frequency of coexpression of Ror1 and Rif in lung adenocarcinoma. Lung adenocarcinoma cells cultured on Matrigel established front-rear polarity with massive filopodia on their front surfaces, where Ror1 and Rif were accumulated. Suppression of Ror1 or Rif expression inhibited cell proliferation, survival, and invasion, accompanied by the loss of filopodia and cell polarity in vitro, and prevented tumor growth in vivo. Furthermore, we found that Rif was required to activate Wnt5a-Ror1 signaling at the cell surface leading to phosphorylation of the Wnt signaling pathway hub protein Dvl2, which was further promoted by culturing the cells on Matrigel. Our findings reveal a novel function of Rif in mediating Wnt5a-Ror1-Dvl2 signaling, which is associated with the formation of polarized filopodia on 3D matrices in lung adenocarcinoma cells.
Collapse
Affiliation(s)
- Michiru Nishita
- Department of Biochemistry, Fukushima Medical University School of Medicine, Fukushima, Japan.
| | - Koki Kamizaki
- Division of Cell Physiology, Department of Physiology and Cell Biology, Kobe University, Graduate School of Medicine, Kobe, Japan
| | - Kyoka Hoshi
- Department of Biochemistry, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kana Aruga
- Division of Cell Physiology, Department of Physiology and Cell Biology, Kobe University, Graduate School of Medicine, Kobe, Japan
| | - Ikumi Nishikaku
- Division of Cell Physiology, Department of Physiology and Cell Biology, Kobe University, Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Shibuya
- Department of Molecular Cell Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma, Kanazawa, Japan; WPI-Nano Life Science Institute, Kanazawa University, Kakuma, Kanazawa, Japan
| | - Yasuhiro Minami
- Division of Cell Physiology, Department of Physiology and Cell Biology, Kobe University, Graduate School of Medicine, Kobe, Japan.
| |
Collapse
|
22
|
Li W, Chung WL, Kozlov MM, Medalia O, Geiger B, Bershadsky AD. Chiral growth of adherent filopodia. Biophys J 2023; 122:3704-3721. [PMID: 37301982 PMCID: PMC10541518 DOI: 10.1016/j.bpj.2023.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/03/2023] [Accepted: 06/06/2023] [Indexed: 06/12/2023] Open
Abstract
Adherent filopodia are elongated finger-like membrane protrusions, extending from the edges of diverse cell types and participating in cell adhesion, spreading, migration, and environmental sensing. The formation and elongation of filopodia are driven by the polymerization of parallel actin filaments, comprising the filopodia cytoskeletal core. Here, we report that adherent filopodia, formed during the spreading of cultured cells on galectin-8-coated substrates, tend to change the direction of their extension in a chiral fashion, acquiring a left-bent shape. Cryoelectron tomography examination indicated that turning of the filopodia tip to the left is accompanied by the displacement of the actin core bundle to the right of the filopodia midline. Reduction of the adhesion to galectin-8 by treatment with thiodigalactoside abolished this filopodia chirality. By modulating the expression of a variety of actin-associated filopodia proteins, we identified myosin-X and formin DAAM1 as major filopodia chirality promoting factors. Formin mDia1, actin filament elongation factor VASP, and actin filament cross-linker fascin were also shown to be involved. Thus, the simple actin cytoskeleton of filopodia, together with a small number of associated proteins are sufficient to drive a complex navigation process, manifested by the development of left-right asymmetry in these cellular protrusions.
Collapse
Affiliation(s)
- Wenhong Li
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Wen-Lu Chung
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Michael M Kozlov
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Benjamin Geiger
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| | - Alexander D Bershadsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel; Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
23
|
Kapustin A, Tsakali SS, Whitehead M, Chennell G, Wu MY, Molenaar C, Kutikhin A, Bogdanov L, Sinitsky M, Rubina K, Clayton A, Verweij FJ, Pegtel DM, Zingaro S, Lobov A, Zainullina B, Owen D, Parsons M, Cheney RE, Warren D, Humphries MJ, Iskratsch T, Holt M, Shanahan CM. Extracellular vesicles stimulate smooth muscle cell migration by presenting collagen VI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.17.551257. [PMID: 37645762 PMCID: PMC10462164 DOI: 10.1101/2023.08.17.551257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The extracellular matrix (ECM) supports blood vessel architecture and functionality and undergoes active remodelling during vascular repair and atherogenesis. Vascular smooth muscle cells (VSMCs) are essential for vessel repair and, via their secretome, are able to invade from the vessel media into the intima to mediate ECM remodelling. Accumulation of fibronectin (FN) is a hallmark of early vascular repair and atherosclerosis and here we show that FN stimulates VSMCs to secrete small extracellular vesicles (sEVs) by activating the β1 integrin/FAK/Src pathway as well as Arp2/3-dependent branching of the actin cytoskeleton. Spatially, sEV were secreted via filopodia-like cellular protrusions at the leading edge of migrating cells. We found that sEVs are trapped by the ECM in vitro and colocalise with FN in symptomatic atherosclerotic plaques in vivo. Functionally, ECM-trapped sEVs induced the formation of focal adhesions (FA) with enhanced pulling forces at the cellular periphery. Proteomic and GO pathway analysis revealed that VSMC-derived sEVs display a cell adhesion signature and are specifically enriched with collagen VI. In vitro assays identified collagen VI as playing the key role in cell adhesion and invasion. Taken together our data suggests that the accumulation of FN is a key early event in vessel repair acting to promote secretion of collage VI enriched sEVs by VSMCs. These sEVs stimulate migration and invasion by triggering peripheral focal adhesion formation and actomyosin contraction to exert sufficient traction forces to enable VSMC movement within the complex vascular ECM network.
Collapse
Affiliation(s)
- Alexander Kapustin
- School of Cardiovascular and Metabolic Medicine & Sciences, James Black Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK, Tel. 020 7848 5221, FAX 020 7848 5193
| | - Sofia Serena Tsakali
- School of Cardiovascular and Metabolic Medicine & Sciences, James Black Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK, Tel. 020 7848 5221, FAX 020 7848 5193
| | - Meredith Whitehead
- School of Cardiovascular and Metabolic Medicine & Sciences, James Black Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK, Tel. 020 7848 5221, FAX 020 7848 5193
| | - George Chennell
- Wohl Cellular Imaging Centre, King’s College London, 5 Cutcombe Road, London, SE5 9NU
| | - Meng-Ying Wu
- School of Cardiovascular and Metabolic Medicine & Sciences, James Black Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK, Tel. 020 7848 5221, FAX 020 7848 5193
| | - Chris Molenaar
- School of Cardiovascular and Metabolic Medicine & Sciences, James Black Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK, Tel. 020 7848 5221, FAX 020 7848 5193
| | - Anton Kutikhin
- Laboratory for Molecular, Translational and Digital Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo, 650002, Russian Federation
| | - Leo Bogdanov
- Laboratory for Molecular, Translational and Digital Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo, 650002, Russian Federation
| | - Maxim Sinitsky
- Laboratory for Molecular, Translational and Digital Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo, 650002, Russian Federation
| | - Kseniya Rubina
- Laboratory of Morphogenesis and Tissue Reparation, Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky av. 27-1, Moscow, 119991, Russia, tel/fax +74959329904
| | - Aled Clayton
- Tissue Microenvironment Research Group, Division of Cancer & Genetics, School of Medicine, Cardiff University, Tenovus Building, Cardiff, UK, CF14 2XN
| | - Frederik J Verweij
- Division of Cell Biology, Neurobiology & Biophysics, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Dirk Michiel Pegtel
- Amsterdam UMC, Location Vrije Universiteit Amsterdam, Department of Pathology, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Simona Zingaro
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL UK
| | - Arseniy Lobov
- Laboratory of Regenerative Biomedicine, Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretskiy Prospekt, 194064, St. Petersburg, Russia
| | - Bozhana Zainullina
- Centre for Molecular and Cell Technologies, Research Park, St. Petersburg State University, 7/9 Universitetskaya Embankment, 199034, St. Petersburg, Russia
| | - Dylan Owen
- Institute of Immunology and Immunotherapy, School of Mathematics and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, B15 2TT, UK
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL UK
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Derek Warren
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK, NR4 7TJ
| | - Martin James Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Thomas Iskratsch
- School of Engineering and Materials Science, Faculty of Science and Engineering, Queen Mary University of London, Engineering Building, Mile End Road, E1 4NS
| | - Mark Holt
- Amsterdam UMC, Location Vrije Universiteit Amsterdam, Department of Pathology, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Catherine M Shanahan
- School of Cardiovascular and Metabolic Medicine & Sciences, James Black Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK, Tel. 020 7848 5221, FAX 020 7848 5193
| |
Collapse
|
24
|
Ostrowska-Podhorodecka Z, Ali A, Norouzi M, Ding I, Abbasi S, Arora PD, Wong THF, Magalhaes M, McCulloch CA. Vimentin-mediated myosin 10 aggregation at tips of cell extensions drives MT1-MMP-dependent collagen degradation in colorectal cancer. FASEB J 2023; 37:e23097. [PMID: 37440280 DOI: 10.1096/fj.202300672r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/09/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023]
Abstract
Colorectal cancer (CRC) is a high prevalence adenocarcinoma with progressive increases in metastasis-related mortality, but the mechanisms governing the extracellular matrix (ECM) degradation important for metastasis in CRC are not well-defined. We investigated a functional relationship between vimentin (Vim) and myosin 10 (Myo10), and whether this relationship is associated with cancer progression. We tested the hypothesis that Vim regulates the aggregation of Myo10 at the tips of cell extensions, which increases membrane-type 1 matrix metalloproteinase (MT1-MMP)-associated local collagen proteolysis and ECM degradation. Analysis of CRC samples revealed colocalization of Vim with Myo10 and MT1-MMP in cell extensions adjacent to sites of collagen degradation, suggesting an association with local cell invasion. We analyzed cultured CRC cells and fibroblasts and found that Vim accelerates aggregation of Myo10 at cell tips, which increases the cell extension rate. Vim stabilizes the interaction of Myo10 with MT1-MMP, which in turn increases collagenolysis. Vim depletion reduced the aggregation of Myo10 at the cell extension tips and MT1-MMP-dependent collagenolysis. We propose that Vim interacts with Myo10, which in turn associates with MT1-MMP to facilitate the transport of these molecules to the termini of cell extensions and there enhance cancer invasion of soft connective tissues.
Collapse
Affiliation(s)
| | - Aiman Ali
- Oral Pathology and Oral Medicine, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Masoud Norouzi
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Isabel Ding
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Sevil Abbasi
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Pamma D Arora
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Timothy H F Wong
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Marco Magalhaes
- Oral Pathology and Oral Medicine, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
- Cancer Invasion and Metastasis Laboratory, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
- Dental and Maxillofacial Sciences Department, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | | |
Collapse
|
25
|
Yim YI, Pedrosa A, Wu X, Chinthalapudi K, Cheney RE, Hammer JA. Myosin 10 uses its MyTH4 and FERM domains differentially to support two aspects of spindle pole biology required for mitotic spindle bipolarity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545002. [PMID: 37398378 PMCID: PMC10312724 DOI: 10.1101/2023.06.15.545002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Myosin 10 (Myo10) has the ability to link actin filaments to integrin-based adhesions and to microtubules by virtue of its integrin-binding FERM domain and microtubule-binding MyTH4 domain, respectively. Here we used Myo10 knockout cells to define Myo10's contribution to the maintenance of spindle bipolarity, and complementation to quantitate the relative contributions of its MyTH4 and FERM domains. Myo10 knockout HeLa cells and mouse embryo fibroblasts (MEFs) both exhibit a pronounced increase in the frequency of multipolar spindles. Staining of unsynchronized metaphase cells showed that the primary driver of spindle multipolarity in knockout MEFs and knockout HeLa cells lacking supernumerary centrosomes is pericentriolar material (PCM) fragmentation, which creates γ-tubulin-positive acentriolar foci that serve as additional spindle poles. For HeLa cells possessing supernumerary centrosomes, Myo10 depletion further accentuates spindle multipolarity by impairing the clustering of the extra spindle poles. Complementation experiments show that Myo10 must interact with both integrins and microtubules to promote PCM/pole integrity. Conversely, Myo10's ability to promote the clustering of supernumerary centrosomes only requires that it interact with integrins. Importantly, images of Halo-Myo10 knock-in cells show that the myosin localizes exclusively within adhesive retraction fibers during mitosis. Based on these and other results, we conclude that Myo10 promotes PCM/pole integrity at a distance, and that it facilitates supernumerary centrosome clustering by promoting retraction fiber-based cell adhesion, which likely provides an anchor for the microtubule-based forces driving pole focusing.
Collapse
Affiliation(s)
- Yang-In Yim
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Antonio Pedrosa
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Xufeng Wu
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
26
|
Crozet F, Letort G, Bulteau R, Da Silva C, Eichmuller A, Tortorelli AF, Blévinal J, Belle M, Dumont J, Piolot T, Dauphin A, Coulpier F, Chédotal A, Maître JL, Verlhac MH, Clarke HJ, Terret ME. Filopodia-like protrusions of adjacent somatic cells shape the developmental potential of oocytes. Life Sci Alliance 2023; 6:e202301963. [PMID: 36944420 PMCID: PMC10029974 DOI: 10.26508/lsa.202301963] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 03/23/2023] Open
Abstract
The oocyte must grow and mature before fertilization, thanks to a close dialogue with the somatic cells that surround it. Part of this communication is through filopodia-like protrusions, called transzonal projections (TZPs), sent by the somatic cells to the oocyte membrane. To investigate the contribution of TZPs to oocyte quality, we impaired their structure by generating a full knockout mouse of the TZP structural component myosin-X (MYO10). Using spinning disk and super-resolution microscopy combined with a machine-learning approach to phenotype oocyte morphology, we show that the lack of Myo10 decreases TZP density during oocyte growth. Reduction in TZPs does not prevent oocyte growth but impairs oocyte-matrix integrity. Importantly, we reveal by transcriptomic analysis that gene expression is altered in TZP-deprived oocytes and that oocyte maturation and subsequent early embryonic development are partially affected, effectively reducing mouse fertility. We propose that TZPs play a role in the structural integrity of the germline-somatic complex, which is essential for regulating gene expression in the oocyte and thus its developmental potential.
Collapse
Affiliation(s)
- Flora Crozet
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Université Paris Cité, Paris, France
| | - Gaëlle Letort
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Université Paris Cité, Paris, France
| | - Rose Bulteau
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Christelle Da Silva
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Adrien Eichmuller
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
- Institut Curie, PSL Research University, Sorbonne Université, CNRS UMR 3215, INSERM U934, Paris, France
| | - Anna Francesca Tortorelli
- Institut Curie, PSL Research University, Sorbonne Université, CNRS UMR 3215, INSERM U934, Paris, France
| | | | - Morgane Belle
- Institut de la Vision, UMRS968/UMR7210/UM80, Paris, France
| | - Julien Dumont
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Tristan Piolot
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Aurélien Dauphin
- Institut Curie, PSL Research University, Sorbonne Université, CNRS UMR 3215, INSERM U934, Paris, France
| | - Fanny Coulpier
- Genomics Core Facility, Institut de Biologie de l'ENS, Département de biologie, Ecole normale supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Alain Chédotal
- Institut de la Vision, UMRS968/UMR7210/UM80, Paris, France
| | - Jean-Léon Maître
- Institut Curie, PSL Research University, Sorbonne Université, CNRS UMR 3215, INSERM U934, Paris, France
| | - Marie-Hélène Verlhac
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada
| | - Marie-Emilie Terret
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| |
Collapse
|
27
|
Hein JI, Scholz J, Körber S, Kaufmann T, Faix J. Unleashed Actin Assembly in Capping Protein-Deficient B16-F1 Cells Enables Identification of Multiple Factors Contributing to Filopodium Formation. Cells 2023; 12:cells12060890. [PMID: 36980231 PMCID: PMC10047565 DOI: 10.3390/cells12060890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
Background: Filopodia are dynamic, finger-like actin-filament bundles that overcome membrane tension by forces generated through actin polymerization at their tips to allow extension of these structures a few microns beyond the cell periphery. Actin assembly of these protrusions is regulated by accessory proteins including heterodimeric capping protein (CP) or Ena/VASP actin polymerases to either terminate or promote filament growth. Accordingly, the depletion of CP in B16-F1 melanoma cells was previously shown to cause an explosive formation of filopodia. In Ena/VASP-deficient cells, CP depletion appeared to result in ruffling instead of inducing filopodia, implying that Ena/VASP proteins are absolutely essential for filopodia formation. However, this hypothesis was not yet experimentally confirmed. Methods: Here, we used B16-F1 cells and CRISPR/Cas9 technology to eliminate CP either alone or in combination with Ena/VASP or other factors residing at filopodia tips, followed by quantifications of filopodia length and number. Results: Unexpectedly, we find massive formations of filopodia even in the absence of CP and Ena/VASP proteins. Notably, combined inactivation of Ena/VASP, unconventional myosin-X and the formin FMNL3 was required to markedly impair filopodia formation in CP-deficient cells. Conclusions: Taken together, our results reveal that, besides Ena/VASP proteins, numerous other factors contribute to filopodia formation.
Collapse
Affiliation(s)
| | | | | | | | - Jan Faix
- Correspondence: ; Tel.: +49-511-532-2928
| |
Collapse
|
28
|
Zhang K, Huang M, Li A, Wen J, Yan L, Li Y, Guo L, Senthil KS, Zhou Y, Chen G, Liu Y, Zhang X, Yao X, Qin D, Su H. DIAPH3 condensates formed by liquid-liquid phase separation act as a regulatory hub for stress-induced actin cytoskeleton remodeling. Cell Rep 2023; 42:111986. [PMID: 36640348 DOI: 10.1016/j.celrep.2022.111986] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/24/2022] [Accepted: 12/23/2022] [Indexed: 01/11/2023] Open
Abstract
Membraneless condensates, such as stress granules (SGs) and processing bodies (P-bodies), have attracted wide attention due to their unique feature of rapid response to stress without first requiring nuclear feedback. In this study, we identify diaphanous-related formin 3 (DIAPH3), an actin nucleator, as a scaffold protein to initiate liquid-liquid phase separation (LLPS) and form abundant cytosolic phase-separated DIAPH3 granules (D-granules) in mammalian cells such as HeLa, HEK293, and fibroblasts under various stress conditions. Neither mRNAs nor known stress-associated condensate markers, such as G3BP1, G3BP2, and TIA1 for SGs and DCP1A for P-bodies, are detected in D-granules. Using overexpression and knockout of DIAPH3, pharmacological interventions, and optogenetics, we further demonstrate that stress-induced D-granules spatially sequester DIAPH3 within the condensation to inhibit the assembly of actin filaments in filopodia. This study reveals that D-granules formed by LLPS act as a regulatory hub for actin cytoskeletal remodeling in response to stress.
Collapse
Affiliation(s)
- Ke Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Miaodan Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Ang Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jing Wen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yunhao Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Liman Guo
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Kumaran Satyanarayanan Senthil
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yangyang Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Guobing Chen
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
| | - Yong Liu
- Laboratory of Neuroscience in Health and Disease Institute, Guangzhou First People's Hospital School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaofei Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaoli Yao
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| |
Collapse
|
29
|
Pokrant T, Hein JI, Körber S, Disanza A, Pich A, Scita G, Rottner K, Faix J. Ena/VASP clustering at microspike tips involves lamellipodin but not I-BAR proteins, and absolutely requires unconventional myosin-X. Proc Natl Acad Sci U S A 2023; 120:e2217437120. [PMID: 36598940 PMCID: PMC9926217 DOI: 10.1073/pnas.2217437120] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/14/2022] [Indexed: 01/05/2023] Open
Abstract
Sheet-like membrane protrusions at the leading edge, termed lamellipodia, drive 2D-cell migration using active actin polymerization. Microspikes comprise actin-filament bundles embedded within lamellipodia, but the molecular mechanisms driving their formation and their potential functional relevance have remained elusive. Microspike formation requires the specific activity of clustered Ena/VASP proteins at their tips to enable processive actin assembly in the presence of capping protein, but the factors and mechanisms mediating Ena/VASP clustering are poorly understood. Systematic analyses of B16-F1 melanoma mutants lacking potential candidate proteins revealed that neither inverse BAR-domain proteins, nor lamellipodin or Abi is essential for clustering, although they differentially contribute to lamellipodial VASP accumulation. In contrast, unconventional myosin-X (MyoX) identified here as proximal to VASP was obligatory for Ena/VASP clustering and microspike formation. Interestingly, and despite the invariable distribution of other relevant marker proteins, the width of lamellipodia in MyoX-KO mutants was significantly reduced as compared with B16-F1 control, suggesting that microspikes contribute to lamellipodium stability. Consistently, MyoX removal caused marked defects in protrusion and random 2D-cell migration. Strikingly, Ena/VASP-deficiency also uncoupled MyoX cluster dynamics from actin assembly in lamellipodia, establishing their tight functional association in microspike formation.
Collapse
Affiliation(s)
- Thomas Pokrant
- Institute for Biophysical Chemistry, Hannover Medical School, 30625Hannover, Germany
| | - Jens Ingo Hein
- Institute for Biophysical Chemistry, Hannover Medical School, 30625Hannover, Germany
| | - Sarah Körber
- Institute for Biophysical Chemistry, Hannover Medical School, 30625Hannover, Germany
| | - Andrea Disanza
- IFOM ETS (Istituto Fondazione di Oncologia Molecolare ETS), - The AIRC (Italian Association for Cancer Research) Institute of Molecular Oncology, 20139Milan, Italy
| | - Andreas Pich
- Research Core Unit Proteomics, Hannover Medical School, 30625Hannover, Germany
| | - Giorgio Scita
- IFOM ETS (Istituto Fondazione di Oncologia Molecolare ETS), - The AIRC (Italian Association for Cancer Research) Institute of Molecular Oncology, 20139Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, 20139 Milan, Italy
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106Braunschweig, Germany
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, 38124Braunschweig, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, 30625Hannover, Germany
| |
Collapse
|
30
|
Pokrant T, Hein JI, Körber S, Disanza A, Pich A, Scita G, Rottner K, Faix J. Ena/VASP clustering at microspike tips involves lamellipodin but not I-BAR proteins, and absolutely requires unconventional myosin-X. Proc Natl Acad Sci U S A 2023. [PMID: 36598940 DOI: 10.1101/2022.05.12.491613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
Sheet-like membrane protrusions at the leading edge, termed lamellipodia, drive 2D-cell migration using active actin polymerization. Microspikes comprise actin-filament bundles embedded within lamellipodia, but the molecular mechanisms driving their formation and their potential functional relevance have remained elusive. Microspike formation requires the specific activity of clustered Ena/VASP proteins at their tips to enable processive actin assembly in the presence of capping protein, but the factors and mechanisms mediating Ena/VASP clustering are poorly understood. Systematic analyses of B16-F1 melanoma mutants lacking potential candidate proteins revealed that neither inverse BAR-domain proteins, nor lamellipodin or Abi is essential for clustering, although they differentially contribute to lamellipodial VASP accumulation. In contrast, unconventional myosin-X (MyoX) identified here as proximal to VASP was obligatory for Ena/VASP clustering and microspike formation. Interestingly, and despite the invariable distribution of other relevant marker proteins, the width of lamellipodia in MyoX-KO mutants was significantly reduced as compared with B16-F1 control, suggesting that microspikes contribute to lamellipodium stability. Consistently, MyoX removal caused marked defects in protrusion and random 2D-cell migration. Strikingly, Ena/VASP-deficiency also uncoupled MyoX cluster dynamics from actin assembly in lamellipodia, establishing their tight functional association in microspike formation.
Collapse
Affiliation(s)
- Thomas Pokrant
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Jens Ingo Hein
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Sarah Körber
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Andrea Disanza
- IFOM ETS (Istituto Fondazione di Oncologia Molecolare ETS), - The AIRC (Italian Association for Cancer Research) Institute of Molecular Oncology, 20139 Milan, Italy
| | - Andreas Pich
- Research Core Unit Proteomics, Hannover Medical School, 30625 Hannover, Germany
| | - Giorgio Scita
- IFOM ETS (Istituto Fondazione di Oncologia Molecolare ETS), - The AIRC (Italian Association for Cancer Research) Institute of Molecular Oncology, 20139 Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, 20139 Milan, Italy
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany
- Molecular Cell Biology Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
31
|
Fitz GN, Weck ML, Bodnya C, Perkins OL, Tyska MJ. Protrusion growth driven by myosin-generated force. Dev Cell 2023; 58:18-33.e6. [PMID: 36626869 PMCID: PMC9940483 DOI: 10.1016/j.devcel.2022.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/10/2022] [Accepted: 11/29/2022] [Indexed: 01/11/2023]
Abstract
Actin-based protrusions extend from the surface of all eukaryotic cells, where they support diverse activities essential for life. Models of protrusion growth hypothesize that actin filament assembly exerts force for pushing the plasma membrane outward. However, membrane-associated myosin motors are also abundant in protrusions, although their potential for contributing, growth-promoting force remains unexplored. Using an inducible system that docks myosin motor domains to membrane-binding modules with temporal control, we found that application of myosin-generated force to the membrane is sufficient for driving robust protrusion elongation in human, mouse, and pig cell culture models. Protrusion growth scaled with motor accumulation, required barbed-end-directed force, and was independent of cargo delivery or recruitment of canonical elongation factors. Application of growth-promoting force was also supported by structurally distinct myosin motors and membrane-binding modules. Thus, myosin-generated force can drive protrusion growth, and this mechanism is likely active in diverse biological contexts.
Collapse
Affiliation(s)
- Gillian N Fitz
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Meredith L Weck
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Caroline Bodnya
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Olivia L Perkins
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
32
|
Tu Y, Pal K, Austin J, Wang X. Filopodial adhesive force in discrete nodes revealed by integrin molecular tension imaging. Curr Biol 2022; 32:4386-4396.e3. [PMID: 36084647 PMCID: PMC9613586 DOI: 10.1016/j.cub.2022.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/30/2022] [Accepted: 08/15/2022] [Indexed: 10/14/2022]
Abstract
Filopodia are narrow cell extensions involved in various physiological processes. Integrins mediate filopodia adhesion and likely transmit adhesive force to regulate filopodia formation and functions, but the force is extremely weak to study and remains poorly understood. Using integrative tension sensor (ITS), we imaged filopodia adhesive force at the single molecular tension level and investigated the force dynamics and sources. Results show that filopodia integrin tension (FIT) is generated in discrete foci (force nodes) along single filopodia with a spacing of ∼1 μm. Inhibitions of actin polymerization or myosin II activity markedly reduced FIT signals in force nodes at filopodia tips and at filopodia bases, respectively, suggesting differential force sources of FIT in the distal force nodes and proximal ones in filopodia. Using two ITS constructs with different force thresholds for activation, we showed that the molecular force level of FIT is greater at filopodia bases than that at filopodia tips. We also tested the role of vinculin and myosin X in the FIT transmission. With vinculin knockout in cells, filopodia and associated force nodes were still formed normally, suggesting that vinculin is dispensable for the formation of filopodia and force nodes. However, vinculin is indeed required for the transmission of strong FIT (capable of rupturing DNA in a shear conformation), as the strong FIT vanished in filopodia with vinculin knockout. Co-imaging of FIT and myosin X shows no apparent co-localization, demonstrating that myosin X is not directly responsible for generating FIT, despite its prominent role in filopodium elongation.
Collapse
Affiliation(s)
- Ying Tu
- Department of Physics and Astronomy, Iowa State University, Ames, IA 50011, USA
| | - Kaushik Pal
- Department of Physics and Astronomy, Iowa State University, Ames, IA 50011, USA
| | - Jacob Austin
- Department of Physics and Astronomy, Iowa State University, Ames, IA 50011, USA
| | - Xuefeng Wang
- Department of Physics and Astronomy, Iowa State University, Ames, IA 50011, USA; Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
33
|
Selective binding and transport of protocadherin 15 isoforms by stereocilia unconventional myosins in a heterologous expression system. Sci Rep 2022; 12:13764. [PMID: 35962067 PMCID: PMC9374675 DOI: 10.1038/s41598-022-17757-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/30/2022] [Indexed: 11/09/2022] Open
Abstract
During hair cell development, the mechanoelectrical transduction (MET) apparatus is assembled at the stereocilia tips, where it coexists with the stereocilia actin regulatory machinery. While the myosin-based tipward transport of actin regulatory proteins is well studied, isoform complexity and built-in redundancies in the MET apparatus have limited our understanding of how MET components are transported. We used a heterologous expression system to elucidate the myosin selective transport of isoforms of protocadherin 15 (PCDH15), the protein that mechanically gates the MET apparatus. We show that MYO7A selectively transports the CD3 isoform while MYO3A and MYO3B transports the CD2 isoform. Furthermore, MYO15A showed an insignificant role in the transport of PCDH15, and none of the myosins tested transport PCDH15-CD1. Our data suggest an important role for MYO3A, MYO3B, and MYO7A in the MET apparatus formation and highlight the intricate nature of MET and actin regulation during development and functional maturation of the stereocilia bundle.
Collapse
|
34
|
Ou H, Wang L, Xi Z, Shen H, Jiang Y, Zhou F, Liu Y, Zhou Y. MYO10 contributes to the malignant phenotypes of colorectal cancer via RACK1 by activating integrin/Src/FAK signaling. Cancer Sci 2022; 113:3838-3851. [PMID: 35912545 DOI: 10.1111/cas.15519] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 11/28/2022] Open
Abstract
Liver metastases still remain a major cause of colorectal cancer (CRC) patient death. MYO10 is upregulated in several tumor types, however, its significance and the underlying mechanism in CRC is not entirely clear. Here we found that MYO10 was highly expressed in CRC tumor tissues, especially in liver metastasis tissues. MYO10 knockout reduced CRC cell proliferation, invasion, and migration in vitro, and CRC metastasis in vivo. We identified RACK1 by LC-MS/MS and demonstrated that MYO10 interacts with and stabilizes RACK1. Mechanistically, MYO10 promotes CRC cell progression and metastasis via ubiquitination-mediated RACK1 degradation and integrin/Src/FAK signaling activation. Therefore, the MYO10/RACK1/integrin/Src/FAK axis may play an important role in CRC progression and metastasis.
Collapse
Affiliation(s)
- Haibin Ou
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Lili Wang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Ziyao Xi
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Hui Shen
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yaofei Jiang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Fuxiang Zhou
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yu Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yunfeng Zhou
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
35
|
Matozo T, Kogachi L, de Alencar BC. Myosin motors on the pathway of viral infections. Cytoskeleton (Hoboken) 2022; 79:41-63. [PMID: 35842902 DOI: 10.1002/cm.21718] [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: 04/27/2022] [Revised: 06/25/2022] [Accepted: 07/07/2022] [Indexed: 01/30/2023]
Abstract
Molecular motors are microscopic machines that use energy from adenosine triphosphate (ATP) hydrolysis to generate movement. While kinesins and dynein are molecular motors associated with microtubule tracks, myosins bind to and move on actin filaments. Mammalian cells express several myosin motors. They power cellular processes such as endo- and exocytosis, intracellular trafficking, transcription, migration, and cytokinesis. As viruses navigate through cells, they may take advantage or be hindered by host components and machinery, including the cytoskeleton. This review delves into myosins' cell roles and compares them to their reported functions in viral infections. In most cases, the previously described myosin functions align with their reported role in viral infections, although not in all cases. This opens the possibility that knowledge obtained from studying myosins in viral infections might shed light on new physiological roles for myosins in cells. However, given the high number of myosins expressed and the variety of viruses investigated in the different studies, it is challenging to infer whether the interactions found are specific to a single virus or can be applied to other viruses with the same characteristics. We conclude that the participation of myosins in viral cycles is still a largely unexplored area, especially concerning unconventional myosins.
Collapse
Affiliation(s)
- Tais Matozo
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Leticia Kogachi
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bruna Cunha de Alencar
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
36
|
Hensel A, Stahl P, Moews L, König L, Patwardhan R, Höing A, Schulze N, Nalbant P, Stauber RH, Knauer SK. The Taspase1/Myosin1f-axis regulates filopodia dynamics. iScience 2022; 25:104355. [PMID: 35601920 PMCID: PMC9121324 DOI: 10.1016/j.isci.2022.104355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/04/2022] [Accepted: 04/28/2022] [Indexed: 11/15/2022] Open
Abstract
The unique threonine protease Tasp1 impacts not only ordered development and cell proliferation but also pathologies. However, its substrates and the underlying molecular mechanisms remain poorly understood. We demonstrate that the unconventional Myo1f is a Tasp1 substrate and unravel the physiological relevance of this proteolysis. We classify Myo1f as a nucleo-cytoplasmic shuttle protein, allowing its unhindered processing by nuclear Tasp1 and an association with chromatin. Moreover, we show that Myo1f induces filopodia resulting in increased cellular adhesion and migration. Importantly, filopodia formation was antagonized by Tasp1-mediated proteolysis, supported by an inverse correlation between Myo1f concentration and Tasp1 expression level. The Tasp1/Myo1f-axis might be relevant in human hematopoiesis as reduced Tasp1 expression coincided with increased Myo1f concentrations and filopodia in macrophages compared to monocytes and vice versa. In sum, we discovered Tasp1-mediated proteolysis of Myo1f as a mechanism to fine-tune filopodia formation, inter alia relevant for cells of the immune system. Myosin1f is a nucleo-cytoplasmic shuttle protein temporarily located in the nucleus Myosin1f induces filopodia resulting in increased cellular adhesion and migration The protease Taspase1 cleaves Myosin1f, thereby impairing its function Taspase1 and Myosin1f inversely correlate in immune cell differentiation
Collapse
Affiliation(s)
- Astrid Hensel
- Department of Molecular Biology II, Center of Medical Biotechnology (ZMB), University Duisburg-Essen, 45141 Essen, Germany
- Corresponding author
| | - Paul Stahl
- Department of Molecular Biology II, Center of Medical Biotechnology (ZMB), University Duisburg-Essen, 45141 Essen, Germany
| | - Lisa Moews
- Department of Molecular Biology II, Center of Medical Biotechnology (ZMB), University Duisburg-Essen, 45141 Essen, Germany
| | - Lena König
- Department of Molecular Biology II, Center of Medical Biotechnology (ZMB), University Duisburg-Essen, 45141 Essen, Germany
| | - Rutuja Patwardhan
- Department of Molecular Cell Biology, Center of Medical Biotechnology (ZMB), University Duisburg-Essen, 45141 Essen, Germany
| | - Alexander Höing
- Department of Molecular Biology II, Center of Medical Biotechnology (ZMB), University Duisburg-Essen, 45141 Essen, Germany
| | - Nina Schulze
- Imaging Center Campus Essen (ICCE), Center of Medical Biotechnology (ZMB), University Duisburg-Essen, 45141 Essen, Germany
| | - Perihan Nalbant
- Department of Molecular Cell Biology, Center of Medical Biotechnology (ZMB), University Duisburg-Essen, 45141 Essen, Germany
| | - Roland H. Stauber
- Department of Molecular and Cellular Oncology/ENT, University Mainz Medical Center, 55131 Mainz, Germany
| | - Shirley K. Knauer
- Department of Molecular Biology II, Center of Medical Biotechnology (ZMB), University Duisburg-Essen, 45141 Essen, Germany
- Corresponding author
| |
Collapse
|
37
|
Yang YF, Sun YY, Peters DM, Keller KE. The Effects of Mechanical Stretch on Integrins and Filopodial-Associated Proteins in Normal and Glaucomatous Trabecular Meshwork Cells. Front Cell Dev Biol 2022; 10:886706. [PMID: 35573666 PMCID: PMC9100841 DOI: 10.3389/fcell.2022.886706] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 01/29/2023] Open
Abstract
The trabecular meshwork (TM) is the tissue responsible for regulating aqueous humor fluid egress from the anterior eye. If drainage is impaired, intraocular pressure (IOP) becomes elevated, which is a primary risk factor for primary open angle glaucoma. TM cells sense elevated IOP via changes in their biomechanical environment. Filopodia cellular protrusions and integrin transmembrane proteins may play roles in detecting IOP elevation, yet this has not been studied in detail in the TM. Here, we investigate integrins and filopodial proteins, such as myosin-X (Myo10), in response to mechanical stretch, an in vitro technique that produces mechanical alterations mimicking elevated IOP. Pull-down assays showed Myo10 binding to α5 but not the β1 subunit, αvβ3, and αvβ5 integrins. Several of these integrins colocalized in nascent adhesions in the filopodial tip and shaft. Using conformation-specific antibodies, we found that β1 integrin, but not α5 or αvβ3 integrins, were activated following 1-h mechanical stretch. Cadherin -11 (CDH11), a cell adhesion molecule, did not bind to Myo10, but was associated with filopodia. Interestingly, CDH11 was downregulated on the TM cell surface following 1-h mechanical stretch. In glaucoma cells, CDH11 protein levels were increased. Finally, mechanical stretch caused a small, yet significant increase in Myo10 protein levels in glaucoma cells, but did not affect cellular communication of fluorescent vesicles via filopodia-like tunneling nanotubes. Together, these data suggest that TM cell adhesion proteins, β1 integrin and CDH11, have relatively rapid responses to mechanical stretch, which suggests a central role in sensing changes in IOP elevation in situ.
Collapse
Affiliation(s)
- Yong-Feng Yang
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Ying Ying Sun
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Donna M. Peters
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Kate E. Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States,Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, United States,*Correspondence: Kate E. Keller,
| |
Collapse
|
38
|
Application of piconewton forces to individual filopodia reveals mechanosensory role of L-type Ca 2+ channels. Biomaterials 2022; 284:121477. [PMID: 35395455 DOI: 10.1016/j.biomaterials.2022.121477] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/18/2022] [Indexed: 11/02/2022]
Abstract
Filopodia are ubiquitous membrane projections that play crucial role in guiding cell migration on rigid substrates and through extracellular matrix by utilizing yet unknown mechanosensing molecular pathways. As recent studies show that Ca2+ channels localized to filopodia play an important role in regulation of their formation and since some Ca2+ channels are known to be mechanosensitive, force-dependent activity of filopodial Ca2+ channels might be linked to filopodia's mechanosensing function. We tested this hypothesis by monitoring changes in the intra-filopodial Ca2+ level in response to application of stretching force to individual filopodia of several cell types using optical tweezers. Results show that stretching forces of tens of pN strongly promote Ca2+ influx into filopodia, causing persistent Ca2+ oscillations that last for minutes even after the force is released. Several known mechanosensitive Ca2+ channels, such as Piezo 1, Piezo 2 and TRPV4, were found to be dispensable for the observed force-dependent Ca2+ influx, while L-type Ca2+ channels appear to be a key player in the discovered phenomenon. As previous studies have shown that intra-filopodial transient Ca2+ signals play an important role in guidance of cell migration, our results suggest that the force-dependent activation of L-type Ca2+ channels may contribute to this process. Overall, our study reveals an intricate interplay between mechanical forces and Ca2+ signaling in filopodia, providing novel mechanistic insights for the force-dependent filopodia functions in guidance of cell migration.
Collapse
|
39
|
Understanding the key functions of Myosins in viral infection. Biochem Soc Trans 2022; 50:597-607. [PMID: 35212367 DOI: 10.1042/bst20211239] [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: 11/02/2021] [Revised: 02/10/2022] [Accepted: 02/15/2022] [Indexed: 11/17/2022]
Abstract
Myosins, a class of actin-based motor proteins existing in almost any organism, are originally considered only involved in driving muscle contraction, reshaping actin cytoskeleton, and anchoring or transporting cargoes, including protein complexes, organelles, vesicles. However, accumulating evidence reveals that myosins also play vital roles in viral infection, depending on viral species and infection stages. This review systemically summarizes the described various myosins, the performed functions, and the involved mechanisms or molecular pathways during viral infection. Meanwhile, the existing issues are also discussed. Additionally, the important technologies or agents, including siRNA, gene editing, and myosin inhibitors, would facilitate dissecting the actions and mechanisms for described and undescribed myosins, which could be adopted to prevent or control viral infection are also characterized.
Collapse
|
40
|
Alberico HC, Woods DC. Role of Granulosa Cells in the Aging Ovarian Landscape: A Focus on Mitochondrial and Metabolic Function. Front Physiol 2022; 12:800739. [PMID: 35153812 PMCID: PMC8829508 DOI: 10.3389/fphys.2021.800739] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/29/2021] [Indexed: 01/11/2023] Open
Abstract
Mitochondria are at the intersection of aging and fertility, with research efforts centered largely on the role that these specialized organelles play in the relatively rapid decline in oocyte quality that occurs as females approach reproductive senescence. In addition to various roles in oocyte maturation, fertilization, and embryogenesis, mitochondria are critical to granulosa cell function. Herein, we provide a review of the literature pertaining to the role of mitochondria in granulosa cell function, with emphasis on how mitochondrial aging in granulosa cells may impact reproduction in female mammals.
Collapse
|
41
|
Nozaki R, Kasamatsu A, Moss J, Uzawa K. Lysyl hydroxylase 2 deficiency promotes filopodia formation and fibroblast migration. Biochem Biophys Res Commun 2022; 587:146-152. [PMID: 34875533 PMCID: PMC10020999 DOI: 10.1016/j.bbrc.2021.11.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 11/19/2022]
Abstract
Lysyl hydroxylase 2 (LH2) regulates intermolecular cross-linking of collagen molecules. Accumulation of LH2-modified collagen, which is highly stable and resistant to collagenase cleavage, is one cause of fibrosis. We previously demonstrated that conventional LH2 knockout mice showed embryonic lethality. Here we established LH2 conditional knockout mice using a tamoxifen-inducible Cre system. Morphological analysis of LH2-deficient fibroblasts by microscopy showed a dramatic increase in the number of filopodia, the finger-like cell surface projections that enable cell movement. The tips and leading edges of these filopodia exhibited up-regulated expression of Myosin-X (Myo10), a regulator of filopodial integrity. Wound healing assays demonstrated that migration of LH2-deficient cells was significantly faster than that of control cells. Gene expression profiling data also supported this phenotype. Together these findings indicate that LH2 deficiency may prevent fibrosis through decreased accumulation of LH2-cross-linked collagen, and that fibroblasts with faster migration contribute to enhanced wound healing activity. In conclusion, our cellular models provide evidence that LH2 deficiency plays a critical role in cell migration mediated through filopodia formation. Understanding the precise role of this phenotype in LH2-deficient cells may be helpful to define the pathogenesis of fibrosis. As such, detailed analyses of fibrosis and wound healing using LH2-deficient mouse models are needed.
Collapse
Affiliation(s)
- Ryunosuke Nozaki
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Kasamatsu
- Department of Dentistry and Oral-Maxillofacial Surgery, Chiba University Hospital, Chiba, Japan.
| | - Joel Moss
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Katsuhiro Uzawa
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba, Japan; Department of Dentistry and Oral-Maxillofacial Surgery, Chiba University Hospital, Chiba, Japan.
| |
Collapse
|
42
|
Vyshnava SS, Kanderi DK, Dowlathabad MR. Confocal laser scanning microscopy study of intercellular events in filopodia using 3-mercaptopropoinc acid capped CdSe/ZnS quantum dots. Micron 2022; 153:103200. [PMID: 34973488 DOI: 10.1016/j.micron.2021.103200] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 11/18/2022]
Abstract
Physico-chemical mobility of cells in three dimensions is dependent on the development of filipodia, which is the fundamental instinct for survival and other cellular functions in live cells. Specifically, our present research paper describes the synthesis of 3-Mercaptopropoinc acid (MPA) capped CdSe/ZnS quantum dots (QDs), which are biocompatible and utilized for cellular bioimaging applications. Using the pancreatic cell lines BXCP3 cells, we successfully demonstrated the applicability of MPA-capped QDs for intercellular filopodia imaging. Employing these QDs, we examined the dynamics of filopodia formation in real-time along the Z-axis by using confocal laser microscopy.
Collapse
Affiliation(s)
| | - Dileep Kumar Kanderi
- Department of Microbiology, Sri Krishnadevaraya University, Anantapuram, A.P, India.
| | | |
Collapse
|
43
|
Gradilla AC, Guerrero I. Hedgehog on track: Long-distant signal transport and transfer through direct cell-to-cell contact. Curr Top Dev Biol 2022; 150:1-24. [DOI: 10.1016/bs.ctdb.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
44
|
Routledge D, Rogers S, Ono Y, Brunt L, Meniel V, Tornillo G, Ashktorab H, Phesse TJ, Scholpp S. The scaffolding protein flot2 promotes cytoneme-based transport of wnt3 in gastric cancer. eLife 2022; 11:77376. [PMID: 36040316 PMCID: PMC9457691 DOI: 10.7554/elife.77376] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
The Wnt/β-catenin signalling pathway regulates multiple cellular processes during development and many diseases, including cell proliferation, migration, and differentiation. Despite their hydrophobic nature, Wnt proteins exert their function over long distances to induce paracrine signalling. Recent studies have identified several factors involved in Wnt secretion; however, our understanding of how Wnt ligands are transported between cells to interact with their cognate receptors is still debated. Here, we demonstrate that gastric cancer cells utilise cytonemes to transport Wnt3 intercellularly to promote proliferation and cell survival. Furthermore, we identify the membrane-bound scaffolding protein Flotillin-2 (Flot2), frequently overexpressed in gastric cancer, as a modulator of these cytonemes. Together with the Wnt co-receptor and cytoneme initiator Ror2, Flot2 determines the number and length of Wnt3 cytonemes in gastric cancer. Finally, we show that Flotillins are also necessary for Wnt8a cytonemes during zebrafish embryogenesis, suggesting a conserved mechanism for Flotillin-mediated Wnt transport on cytonemes in development and disease.
Collapse
Affiliation(s)
- Daniel Routledge
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of ExeterExeterUnited Kingdom
| | - Sally Rogers
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of ExeterExeterUnited Kingdom
| | - Yosuke Ono
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of ExeterExeterUnited Kingdom
| | - Lucy Brunt
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of ExeterExeterUnited Kingdom
| | - Valerie Meniel
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff UniversityCardiffUnited Kingdom
| | - Giusy Tornillo
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff UniversityCardiffUnited Kingdom
| | - Hassan Ashktorab
- Department of Medicine, Howard UniversityWashingtonUnited States
| | - Toby J Phesse
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff UniversityCardiffUnited Kingdom,The Peter Doherty Institute for Infection and Immunity, The University of MelbourneMelbourneAustralia
| | - Steffen Scholpp
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of ExeterExeterUnited Kingdom
| |
Collapse
|
45
|
A large-scale nanoscopy and biochemistry analysis of postsynaptic dendritic spines. Nat Neurosci 2021; 24:1151-1162. [PMID: 34168338 DOI: 10.1038/s41593-021-00874-w] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 05/13/2021] [Indexed: 02/05/2023]
Abstract
Dendritic spines, the postsynaptic compartments of excitatory neurotransmission, have different shapes classified from 'stubby' to 'mushroom-like'. Whereas mushroom spines are essential for adult brain function, stubby spines disappear during brain maturation. It is still unclear whether and how they differ in protein composition. To address this, we combined electron microscopy and quantitative biochemistry with super-resolution microscopy to annotate more than 47,000 spines for more than 100 synaptic targets. Surprisingly, mushroom and stubby spines have similar average protein copy numbers and topologies. However, an analysis of the correlation of each protein to the postsynaptic density mass, used as a marker of synaptic strength, showed substantially more significant results for the mushroom spines. Secretion and trafficking proteins correlated particularly poorly to the strength of stubby spines. This suggests that stubby spines are less likely to adequately respond to dynamic changes in synaptic transmission than mushroom spines, which possibly explains their loss during brain maturation.
Collapse
|
46
|
Nair S, Rocha‐Ferreira E, Fleiss B, Nijboer CH, Gressens P, Mallard C, Hagberg H. Neuroprotection offered by mesenchymal stem cells in perinatal brain injury: Role of mitochondria, inflammation, and reactive oxygen species. J Neurochem 2021; 158:59-73. [PMID: 33314066 PMCID: PMC8359360 DOI: 10.1111/jnc.15267] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022]
Abstract
Preclinical studies have shown that mesenchymal stem cells have a positive effect in perinatal brain injury models. The mechanisms that cause these neurotherapeutic effects are not entirely intelligible. Mitochondrial damage, inflammation, and reactive oxygen species are considered to be critically involved in the development of injury. Mesenchymal stem cells have immunomodulatory action and exert mitoprotective effects which attenuate production of reactive oxygen species and promote restoration of tissue function and metabolism after perinatal insults. This review summarizes the present state, the underlying causes, challenges and possibilities for effective clinical translation of mesenchymal stem cell therapy.
Collapse
Affiliation(s)
- Syam Nair
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Eridan Rocha‐Ferreira
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Bobbi Fleiss
- School of Health and Biomedical SciencesRMIT UniversityBundooraVictoriaAustralia
- Université de Paris, NeuroDiderotParisFrance
| | - Cora H Nijboer
- Department for Developmental Origins of DiseaseUniversity Medical Center Utrecht Brain Center and Wilhelmina Children’s Hospital, Utrecht UniversityUtrechtNetherlands
| | | | - Carina Mallard
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Neuroscience and PhysiologySahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Henrik Hagberg
- Centre of Perinatal Medicine and Health, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
47
|
Zhang S, Saunders T. Mechanical processes underlying precise and robust cell matching. Semin Cell Dev Biol 2021; 120:75-84. [PMID: 34130903 DOI: 10.1016/j.semcdb.2021.06.003] [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: 03/05/2021] [Revised: 05/27/2021] [Accepted: 06/04/2021] [Indexed: 11/26/2022]
Abstract
During the development of complicated multicellular organisms, the robust formation of specific cell-cell connections (cell matching) is required for the generation of precise tissue structures. Mismatches or misconnections can lead to various diseases. Diverse mechanical cues, including differential adhesion and temporally varying cell contractility, are involved in regulating the process of cell-cell recognition and contact formation. Cells often start the process of cell matching through contact via filopodia protrusions, mediated by specific adhesion interactions at the cell surface. These adhesion interactions give rise to differential mechanical signals that can be further perceived by the cells. In conjunction with contractions generated by the actomyosin networks within the cells, this differentially coded adhesion information can be translated to reposition and sort cells. Here, we review the role of these different cell matching components and suggest how these mechanical factors cooperate with each other to facilitate specificity in cell-cell contact formation.
Collapse
Affiliation(s)
- Shaobo Zhang
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Timothy Saunders
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, Singapore; Warwick Medical School, University of Warwick, Coventry, United Kingdom.
| |
Collapse
|
48
|
Arthur AL, Crawford A, Houdusse A, Titus MA. VASP-mediated actin dynamics activate and recruit a filopodia myosin. eLife 2021; 10:68082. [PMID: 34042588 PMCID: PMC8352590 DOI: 10.7554/elife.68082] [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: 03/04/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022] Open
Abstract
Filopodia are thin, actin-based structures that cells use to interact with their environments. Filopodia initiation requires a suite of conserved proteins but the mechanism remains poorly understood. The actin polymerase VASP and a MyTH-FERM (MF) myosin, DdMyo7 in amoeba, are essential for filopodia initiation. DdMyo7 is localized to dynamic regions of the actin-rich cortex. Analysis of VASP mutants and treatment of cells with anti-actin drugs shows that myosin recruitment and activation in Dictyostelium requires localized VASP-dependent actin polymerization. Targeting of DdMyo7 to the cortex alone is not sufficient for filopodia initiation; VASP activity is also required. The actin regulator locally produces a cortical actin network that activates myosin and together they shape the actin network to promote extension of parallel bundles of actin during filopodia formation. This work reveals how filopodia initiation requires close collaboration between an actin-binding protein, the state of the actin cytoskeleton and MF myosin activity.
Collapse
Affiliation(s)
- Ashley L Arthur
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Amy Crawford
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Anne Houdusse
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, Paris, France
| | - Margaret A Titus
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| |
Collapse
|
49
|
Abstract
Filopodia, microvilli and stereocilia represent an important group of plasma membrane protrusions. These specialized projections are supported by parallel bundles of actin filaments and have critical roles in sensing the external environment, increasing cell surface area, and acting as mechanosensors. While actin-associated proteins are essential for actin-filament elongation and bundling in these protrusions, myosin motors have a surprising role in the formation and extension of filopodia and stereocilia and in the organization of microvilli. Actin regulators and specific myosins collaborate in controlling the length of these structures. Myosins can transport cargoes along the length of these protrusions, and, in the case of stereocilia and microvilli, interactions with adaptors and cargoes can also serve to anchor adhesion receptors to the actin-rich core via functionally conserved motor-adaptor complexes. This review highlights recent progress in understanding the diverse roles myosins play in filopodia, microvilli and stereocilia.
Collapse
Affiliation(s)
- Anne Houdusse
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, 75005 Paris, France.
| | - Margaret A Titus
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
50
|
Cordero Cervantes D, Zurzolo C. Peering into tunneling nanotubes-The path forward. EMBO J 2021; 40:e105789. [PMID: 33646572 PMCID: PMC8047439 DOI: 10.15252/embj.2020105789] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/21/2020] [Accepted: 01/15/2021] [Indexed: 12/19/2022] Open
Abstract
The identification of Tunneling Nanotubes (TNTs) and TNT-like structures signified a critical turning point in the field of cell-cell communication. With hypothesized roles in development and disease progression, TNTs' ability to transport biological cargo between distant cells has elevated these structures to a unique and privileged position among other mechanisms of intercellular communication. However, the field faces numerous challenges-some of the most pressing issues being the demonstration of TNTs in vivo and understanding how they form and function. Another stumbling block is represented by the vast disparity in structures classified as TNTs. In order to address this ambiguity, we propose a clear nomenclature and provide a comprehensive overview of the existing knowledge concerning TNTs. We also discuss their structure, formation-related pathways, biological function, as well as their proposed role in disease. Furthermore, we pinpoint gaps and dichotomies found across the field and highlight unexplored research avenues. Lastly, we review the methods employed to date and suggest the application of new technologies to better understand these elusive biological structures.
Collapse
Affiliation(s)
| | - Chiara Zurzolo
- Institut PasteurMembrane Traffic and PathogenesisParisFrance
| |
Collapse
|