1
|
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
|
2
|
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
|
3
|
Maxian O, Mogilner A. Helical motors and formins synergize to compact chiral filopodial bundles: A theoretical perspective. Eur J Cell Biol 2024; 103:151383. [PMID: 38237507 DOI: 10.1016/j.ejcb.2023.151383] [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: 07/24/2023] [Revised: 12/19/2023] [Accepted: 12/30/2023] [Indexed: 01/28/2024] Open
Abstract
Chiral actin bundles have been shown to play an important role in cell dynamics, but our understanding of the molecular mechanisms which combine to generate chirality remains incomplete. To address this, we numerically simulate a crosslinked filopodial bundle under the actions of helical myosin motors and/or formins and examine the collective buckling and twisting of the actin bundle. We first show that a number of proposed mechanisms to buckle polymerizing actin bundles without motor activity fail under biologically-realistic parameters. We then demonstrate that a simplified model of myosin spinning action at the bundle base effectively "braids" the bundle, but cannot control compaction at the fiber tips. Finally, we show that formin-mediated polymerization and motor activity can act synergitically to compact filopodium bundles, as motor activity bends filaments into shapes that activate twist forces induced by formins. Stochastic fluctuations of actin polymerization rates and slower cross linking dynamics both increase buckling and decrease compaction. We discuss implications of our findings for mechanisms of cytoskeletal chirality.
Collapse
Affiliation(s)
- Ondrej Maxian
- Courant Institute, New York University, New York, NY 10012, USA; Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60615, USA; Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60615, USA
| | - Alex Mogilner
- Courant Institute, New York University, New York, NY 10012, USA; Department of Biology, New York University, New York, NY 10012, USA.
| |
Collapse
|
4
|
Kim OV, Litvinov RI, Gagne AL, French DL, Brass LF, Weisel JW. Megakaryocyte-induced contraction of plasma clots: cellular mechanisms and structural mechanobiology. Blood 2024; 143:548-560. [PMID: 37944157 PMCID: PMC11033616 DOI: 10.1182/blood.2023021545] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/17/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
ABSTRACT Nonmuscle cell contractility is an essential feature underlying diverse cellular processes such as motility, morphogenesis, division and genome replication, intracellular transport, and secretion. Blood clot contraction is a well-studied process driven by contracting platelets. Megakaryocytes (MKs), which are the precursors to platelets, can be found in bone marrow and lungs. Although they express many of the same proteins and structures found in platelets, little is known about their ability to engage with extracellular proteins such as fibrin and contract. Here, we have measured the ability of MKs to compress plasma clots. Megakaryocytes derived from human induced pluripotent stem cells (iPSCs) were suspended in human platelet-free blood plasma and stimulated with thrombin. Using real-time macroscale optical tracking, confocal microscopy, and biomechanical measurements, we found that activated iPSC-derived MKs (iMKs) caused macroscopic volumetric clot shrinkage, as well as densification and stiffening of the fibrin network via fibrin-attached plasma membrane protrusions undergoing extension-retraction cycles that cause shortening and bending of fibrin fibers. Contraction induced by iMKs involved 2 kinetic phases with distinct rates and durations. It was suppressed by inhibitors of nonmuscle myosin IIA, actin polymerization, and integrin αIIbβ3-fibrin interactions, indicating that the molecular mechanisms of iMK contractility were similar or identical to those in activated platelets. Our findings provide new insights into MK biomechanics and suggest that iMKs can be used as a model system to study platelet contractility. Physiologically, the ability of MKs to contract plasma clots may play a role in the mechanical remodeling of intravascular blood clots and thrombi.
Collapse
Affiliation(s)
- Oleg V. Kim
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute, Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA
| | - Rustem I. Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Alyssa L. Gagne
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Deborah L. French
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Lawrence F. Brass
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - John W. Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
5
|
Chen X, Arciola JM, Lee YI, Wong PHP, Yin H, Tao Q, Jin Y, Qin X, Sweeney HL, Park H. Myo10 tail is crucial for promoting long filopodia. J Biol Chem 2024; 300:105523. [PMID: 38043799 PMCID: PMC10790087 DOI: 10.1016/j.jbc.2023.105523] [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: 07/03/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023] Open
Abstract
Filopodia are slender cellular protrusions containing parallel actin bundles involved in environmental sensing and signaling, cell adhesion and migration, and growth cone guidance and extension. Myosin 10 (Myo10), an unconventional actin-based motor protein, was reported to induce filopodial initiation with its motor domain. However, the roles of the multifunctional tail domain of Myo10 in filopodial formation and elongation remain elusive. Herein, we generated several constructs of Myo10-full-length Myo10, Myo10 with a truncated tail (Myo10 HMM), and Myo10 containing four mutations to disrupt its coiled-coil domain (Myo10 CC mutant). We found that the truncation of the tail domain decreased filopodial formation and filopodial length, while four mutations in the coiled-coil domain disrupted the motion of Myo10 toward filopodial tips and the elongation of filopodia. Furthermore, we found that filopodia elongated through multiple elongation cycles, which was supported by the Myo10 tail. These findings suggest that Myo10 tail is crucial for promoting long filopodia.
Collapse
Affiliation(s)
- Xingxiang Chen
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | | | - Young Il Lee
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, USA
| | - Pak Hung Philip Wong
- Department of Physics, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Haoran Yin
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Quanqing Tao
- Department of Physics, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Yuqi Jin
- Department of Physics, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Xianan Qin
- Department of Physics, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - H Lee Sweeney
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, USA; Myology Institute, University of Florida College of Medicine, Gainesville, Florida, USA.
| | - Hyokeun Park
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China; Department of Physics, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China.
| |
Collapse
|
6
|
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
|
7
|
Daghestani MH, Alqahtani HA, AlBakheet A, Al Deery M, Awartani KA, Daghestani MH, Kaya N, Warsy A, Coskun S, Colak D. Global Transcriptional Profiling of Granulosa Cells from Polycystic Ovary Syndrome Patients: Comparative Analyses of Patients with or without History of Ovarian Hyperstimulation Syndrome Reveals Distinct Biomarkers and Pathways. J Clin Med 2022; 11:jcm11236941. [PMID: 36498516 PMCID: PMC9740016 DOI: 10.3390/jcm11236941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/09/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022] Open
Abstract
Ovarian hyperstimulation syndrome (OHSS) is often a complication of polycystic ovarian syndrome (PCOS), the most frequent disorder of the endocrine system, which affects women in their reproductive years. The etiology of OHSS is multifactorial, though the factors involved are not apparent. In an attempt to unveil the molecular basis of OHSS, we conducted transcriptome analysis of total RNA extracted from granulosa cells from PCOS patients with a history of OHSS (n = 6) and compared them to those with no history of OHSS (n = 18). We identified 59 significantly dysregulated genes (48 down-regulated, 11 up-regulated) in the PCOS with OHSS group compared to the PCOS without OHSS group (p-value < 0.01, fold change >1.5). Functional, pathway and network analyses revealed genes involved in cellular development, inflammatory and immune response, cellular growth and proliferation (including DCN, VIM, LIFR, GRN, IL33, INSR, KLF2, FOXO1, VEGF, RDX, PLCL1, PAPPA, and ZFP36), and significant alterations in the PPAR, IL6, IL10, JAK/STAT and NF-κB signaling pathways. Array findings were validated using quantitative RT-PCR. To the best of our knowledge, this is the largest cohort of Saudi PCOS cases (with or without OHSS) to date that was analyzed using a transcriptomic approach. Our data demonstrate alterations in various gene networks and pathways that may be involved in the pathophysiology of OHSS. Further studies are warranted to confirm the findings.
Collapse
Affiliation(s)
- Maha H. Daghestani
- Department of Zoology, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
- Correspondence: (M.H.D.); (D.C.)
| | - Huda A. Alqahtani
- Department of Zoology, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - AlBandary AlBakheet
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Mashael Al Deery
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Khalid A. Awartani
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Mazin H. Daghestani
- Department of Obstetrics and Gynecology, Umm-Al-Qura University, Makkah 24382, Saudi Arabia
| | - Namik Kaya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Arjumand Warsy
- Central Laboratory, Center for Women Scientific and Medical Studies, King Saud University, Riyadh 11451, Saudi Arabia
| | - Serdar Coskun
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Dilek Colak
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
- Correspondence: (M.H.D.); (D.C.)
| |
Collapse
|
8
|
Sato O, Sakai T, Choo YY, Ikebe R, Watanabe TM, Ikebe M. Mitochondria-associated myosin 19 processively transports mitochondria on actin tracks in living cells. J Biol Chem 2022; 298:101883. [PMID: 35367209 PMCID: PMC9065997 DOI: 10.1016/j.jbc.2022.101883] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 11/25/2022] Open
Abstract
Mitochondria are fundamentally important in cell function, and their malfunction can cause the development of cancer, cardiovascular disease, and neuronal disorders. Myosin 19 (Myo19) shows discrete localization with mitochondria and is thought to play an important role in mitochondrial dynamics and function; however, the function of Myo19 in mitochondrial dynamics at the cellular and molecular levels is poorly understood. Critical missing information is whether Myo19 is a processive motor that is suitable for transportation of mitochondria. Here, we show for the first time that single Myo19 molecules processively move on actin filaments and can transport mitochondria in cells. We demonstrate that Myo19 dimers having a leucine zipper processively moved on cellular actin tracks in demembraned cells with a velocity of 50 to 60 nm/s and a run length of ∼0.4 μm, similar to the movement of isolated mitochondria from Myo19 dimer-transfected cells on actin tracks, suggesting that the Myo19 dimer can transport mitochondria. Furthermore, we show single molecules of Myo19 dimers processively moved on single actin filaments with a large step size of ∼34 nm. Importantly, WT Myo19 single molecules without the leucine zipper processively move in filopodia in living cells similar to Myo19 dimers, whereas deletion of the tail domain abolished such active movement. These results suggest that Myo19 can processively move on actin filaments when two Myo19 monomers form a dimer, presumably as a result of tail-tail association. In conclusion, Myo19 molecules can directly transport mitochondria on actin tracks within living cells.
Collapse
Affiliation(s)
- Osamu Sato
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, USA
| | - Tsuyoshi Sakai
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, USA
| | - Young-Yeon Choo
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, USA
| | - Reiko Ikebe
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, USA
| | - Tomonobu M Watanabe
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, USA.
| |
Collapse
|
9
|
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: 5] [Impact Index Per Article: 1.7] [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
|
10
|
Myosin-X and talin modulate integrin activity at filopodia tips. Cell Rep 2021; 36:109716. [PMID: 34525374 PMCID: PMC8456781 DOI: 10.1016/j.celrep.2021.109716] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 07/06/2021] [Accepted: 08/24/2021] [Indexed: 11/20/2022] Open
Abstract
Filopodia assemble unique integrin-adhesion complexes to sense the extracellular matrix. However, the mechanisms of integrin regulation in filopodia are poorly defined. Here, we report that active integrins accumulate at the tip of myosin-X (MYO10)-positive filopodia, while inactive integrins are uniformly distributed. We identify talin and MYO10 as the principal integrin activators in filopodia. In addition, deletion of MYO10's FERM domain, or mutation of its β1-integrin-binding residues, reveals MYO10 as facilitating integrin activation, but not transport, in filopodia. However, MYO10's isolated FERM domain alone cannot activate integrins, potentially because of binding to both integrin tails. Finally, because a chimera construct generated by swapping MYO10-FERM by talin-FERM enables integrin activation in filopodia, our data indicate that an integrin-binding FERM domain coupled to a myosin motor is a core requirement for integrin activation in filopodia. Therefore, we propose a two-step integrin activation model in filopodia: receptor tethering by MYO10 followed by talin-mediated integrin activation.
Collapse
|
11
|
Cirilo JA, Gunther LK, Yengo CM. Functional Role of Class III Myosins in Hair Cells. Front Cell Dev Biol 2021; 9:643856. [PMID: 33718386 PMCID: PMC7947357 DOI: 10.3389/fcell.2021.643856] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/01/2021] [Indexed: 11/14/2022] Open
Abstract
Cytoskeletal motors produce force and motion using the energy from ATP hydrolysis and function in a variety of mechanical roles in cells including muscle contraction, cargo transport, and cell division. Actin-based myosin motors have been shown to play crucial roles in the development and function of the stereocilia of auditory and vestibular inner ear hair cells. Hair cells can contain hundreds of stereocilia, which rely on myosin motors to elongate, organize, and stabilize their structure. Mutations in many stereocilia-associated myosins have been shown to cause hearing loss in both humans and animal models suggesting that each myosin isoform has a specific function in these unique parallel actin bundle-based protrusions. Here we review what is known about the classes of myosins that function in the stereocilia, with a special focus on class III myosins that harbor point mutations associated with delayed onset hearing loss. Much has been learned about the role of the two class III myosin isoforms, MYO3A and MYO3B, in maintaining the precise stereocilia lengths required for normal hearing. We propose a model for how class III myosins play a key role in regulating stereocilia lengths and demonstrate how their motor and regulatory properties are particularly well suited for this function. We conclude that ongoing studies on class III myosins and other stereocilia-associated myosins are extremely important and may lead to novel therapeutic strategies for the treatment of hearing loss due to stereocilia degeneration.
Collapse
Affiliation(s)
- Joseph A Cirilo
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Laura K Gunther
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
12
|
Ljubojevic N, Henderson JM, Zurzolo C. The Ways of Actin: Why Tunneling Nanotubes Are Unique Cell Protrusions. Trends Cell Biol 2020; 31:130-142. [PMID: 33309107 DOI: 10.1016/j.tcb.2020.11.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/09/2020] [Accepted: 11/16/2020] [Indexed: 12/22/2022]
Abstract
Actin remodeling is at the heart of the response of cells to external or internal stimuli, allowing a variety of membrane protrusions to form. Fifteen years ago, tunneling nanotubes (TNTs) were identified, bringing a novel addition to the family of actin-supported cellular protrusions. Their unique property as conduits for cargo transfer between distant cells emphasizes the unique nature of TNTs among other protrusions. While TNTs in different pathological and physiological scenarios have been described, the molecular basis of how TNTs form is not well understood. In this review, we discuss the role of several actin regulators in the formation of TNTs and suggest potential players based on their comparison with other actin-based protrusions. New perspectives for discovering a distinct TNT formation pathway would enable us to target them in treating the increasing number of TNT-involved pathologies.
Collapse
Affiliation(s)
- Nina Ljubojevic
- Membrane Traffic and Pathogenesis, Institut Pasteur, UMR3691 CNRS, 75015 Paris, France; Sorbonne Université, ED394 - Physiologie, Physiopathologie et Thérapeutique, 75005 Paris, France
| | - J Michael Henderson
- Membrane Traffic and Pathogenesis, Institut Pasteur, UMR3691 CNRS, 75015 Paris, France; Laboratoire Physico-Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005 Paris, France
| | - Chiara Zurzolo
- Membrane Traffic and Pathogenesis, Institut Pasteur, UMR3691 CNRS, 75015 Paris, France.
| |
Collapse
|
13
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
14
|
|
15
|
Tunneling Nanotubes and the Eye: Intercellular Communication and Implications for Ocular Health and Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7246785. [PMID: 32352005 PMCID: PMC7171654 DOI: 10.1155/2020/7246785] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/10/2020] [Indexed: 12/31/2022]
Abstract
Cellular communication is an essential process for the development and maintenance of all tissues including the eye. Recently, a new method of cellular communication has been described, which relies on formation of tubules, called tunneling nanotubes (TNTs). These structures connect the cytoplasm of adjacent cells and allow the direct transport of cellular cargo between cells without the need for secretion into the extracellular milieu. TNTs may be an important mechanism for signaling between cells that reside long distances from each other or for cells in aqueous environments, where diffusion-based signaling is challenging. Given the wide range of cargoes transported, such as lysosomes, endosomes, mitochondria, viruses, and miRNAs, TNTs may play a role in normal homeostatic processes in the eye as well as function in ocular disease. This review will describe TNT cellular communication in ocular cell cultures and the mammalian eye in vivo, the role of TNTs in mitochondrial transport with an emphasis on mitochondrial eye diseases, and molecules involved in TNT biogenesis and their function in eyes, and finally, we will describe TNT formation in inflammation, cancer, and stem cells, focusing on pathological processes of particular interest to vision scientists.
Collapse
|
16
|
Qin X, Yoo H, Man Cheng HC, Nguyen QQ, Li J, Liu X, Prunetti L, Chen X, Liu T, Sweeney HL, Park H. Simultaneous tracking of two motor domains reveals near simultaneous steps and stutter steps of myosin 10 on actin filament bundles. Biochem Biophys Res Commun 2020; 525:S0006-291X(20)30305-3. [PMID: 32081426 PMCID: PMC7429334 DOI: 10.1016/j.bbrc.2020.02.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 11/30/2022]
Abstract
Myosin X (Myo10) has several unique design features including dimerization via an anti-parallel coiled coil and a long lever arm, which allow it to preferentially move on actin bundles. To understand the stepping behavior of single Myo10 on actin bundles, we labeled two heads of Myo10 dimers with different fluorophores. Unlike previously described for myosin V (Myo5) and VI (Myo6), which display alternating hand-over-hand stepping, Myo10 frequently took near simultaneous steps of both heads, and less frequently, 2-3 steps of one head before the other head stepped. We suggest that this behavior results from the unusual kinetic features of Myo10, in conjunction with the structural properties of the motor domain/lever arm, which will favor movement on actin bundles rather than on single filaments.
Collapse
Affiliation(s)
- Xianan Qin
- Department of Physics, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong
| | - Hanna Yoo
- Division of Life Science, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong
| | - Harry Chun Man Cheng
- Division of Life Science, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong
| | - Quang Quan Nguyen
- Department of Physics, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong
| | - Jing Li
- Department of Physics, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong
| | - Xiaoyan Liu
- Department of Pharmacology and Therapeutics and the Myology Institute, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Laurence Prunetti
- Department of Pharmacology and Therapeutics and the Myology Institute, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Xingxiang Chen
- Division of Life Science, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong
| | - Teng Liu
- Department of Physics, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong
| | - H Lee Sweeney
- Department of Pharmacology and Therapeutics and the Myology Institute, University of Florida College of Medicine, Gainesville, FL, 32610, USA.
| | - Hyokeun Park
- Department of Physics, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong; Division of Life Science, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong.
| |
Collapse
|
17
|
Sun YY, Bradley JM, Keller KE. Phenotypic and Functional Alterations in Tunneling Nanotubes Formed by Glaucomatous Trabecular Meshwork Cells. Invest Ophthalmol Vis Sci 2020; 60:4583-4595. [PMID: 31675075 PMCID: PMC6827425 DOI: 10.1167/iovs.19-28084] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Purpose Trabecular meshwork (TM) cells detect and coordinate responses to intraocular pressure (IOP) in the eye. TM cells become dysfunctional in glaucoma where IOP is often elevated. Recently, we showed that normal TM (NTM) cells communicate by forming tubular connections called tunneling nanotubes (TNTs). Here, we investigated TNTs in glaucomatous TM (GTM) cells. Methods Primary GTM and NTM cells were established from cadaver eyes. Transfer of Vybrant DiO and DiD-labeled vesicles via TNT connections was measured. Imaris software measured the number and length of cell protrusions from immunofluorescent confocal images. Live-cell imaging of the actin cytoskeleton was performed. The distribution of myosin-X, a regulator of TNTs/filopodia, was investigated in TM cells and tissue. Results GTM cells contained significantly more transferred fluorescent vesicles than NTM cells (49.6% vs. 35%). Although NTM cells had more protrusions at the cell surface than GTM cells (7.61 vs. 4.65 protrusions/cell), GTM protrusions were significantly longer (12.1 μm vs. 9.76 μm). Live-cell imaging demonstrated that the GTM actin cytoskeleton was less dynamic, and vesicle transfer between cells was significantly slower than NTM cells. Furthermore, rearrangement of the actin cortex adjacent to the TNT may influence TNT formation. Myosin-X immunostaining was punctate and disorganized in GTM cells and tissue compared to age-matched NTM controls. Conclusions Together, our data demonstrate that GTM cells have phenotypic and functional differences in their TNTs. Significantly slower vesicle transfer via TNTs in GTM cells may delay the timely propagation of cellular signals when pressures become elevated in glaucoma.
Collapse
Affiliation(s)
- Ying Ying Sun
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - John M Bradley
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Kate E Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
18
|
Abstract
For many years, major differences in morphology, motility, and mechanical characteristics have been observed between transformed cancer and normal cells. In this review, we consider these differences as linked to different states of normal and transformed cells that involve distinct mechanosensing and motility pathways. There is a strong correlation between repeated tissue healing and/or inflammation and the probability of cancer, both of which involve growth in adult tissues. Many factors are likely needed to enable growth, including the loss of rigidity sensing, but recent evidence indicates that microRNAs have important roles in causing the depletion of growth-suppressing proteins. One microRNA, miR-21, is overexpressed in many different tissues during both healing and cancer. Normal cells can become transformed by the depletion of cytoskeletal proteins that results in the loss of mechanosensing, particularly rigidity sensing. Conversely, the transformed state can be reversed by the expression of cytoskeletal proteins-without direct alteration of hormone receptor levels. In this review, we consider the different stereotypical forms of motility and mechanosensory systems. A major difference between normal and transformed cells involves a sensitivity of transformed cells to mechanical perturbations. Thus, understanding the different mechanical characteristics of transformed cells may enable new approaches to treating wound healing and cancer.
Collapse
Affiliation(s)
- Michael Sheetz
- Mechanobiology Institute and Department of Biological Sciences, National University of Singapore, Singapore 117411
- Molecular MechanoMedicine Program and Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555, USA;
| |
Collapse
|
19
|
Gudbergsson JM, Duroux M. Cripto-1 localizes to dynamic and shed filopodia associated with cellular migration in glioblastoma cells. Eur J Cell Biol 2019; 98:151044. [PMID: 31543278 DOI: 10.1016/j.ejcb.2019.151044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/22/2019] [Accepted: 07/24/2019] [Indexed: 12/17/2022] Open
Abstract
Cripto-1 is a protein participating in tissue orientation during embryogenesis but has also been implicated in a wide variety of cancers, such as colon, lung and breast cancer. Cripto-1 plays a role in the regulation of different pathways, including TGF-β/Smad and Wnt/β-catenin, which are highly associated with cell migration both during embryonal development and cancer progression. Little is known about the detailed subcellular localization of cripto-1 and how it participates in the directional movement of cells. In this study, the subcellular localization of cripto-1 in glioblastoma cells was investigated in vitro with high-resolution microscopy techniques. Cripto-1 was found to be localized to dynamic and shed filopodia and transported between cells through tunneling nanotubes. Our results connect the refined subcellular localization of cripto-1 to its functions in cellular orientation and migration.
Collapse
Affiliation(s)
- Johann Mar Gudbergsson
- Laboratory of Immunology and Cancer Biology, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Fredrik Bajers Vej 3B, 9220 Aalborg Ø, Denmark.
| | - Meg Duroux
- Laboratory of Immunology and Cancer Biology, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Fredrik Bajers Vej 3B, 9220 Aalborg Ø, Denmark.
| |
Collapse
|
20
|
Alieva NO, Efremov AK, Hu S, Oh D, Chen Z, Natarajan M, Ong HT, Jégou A, Romet-Lemonne G, Groves JT, Sheetz MP, Yan J, Bershadsky AD. Myosin IIA and formin dependent mechanosensitivity of filopodia adhesion. Nat Commun 2019; 10:3593. [PMID: 31399564 PMCID: PMC6689027 DOI: 10.1038/s41467-019-10964-w] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 06/07/2019] [Indexed: 12/21/2022] Open
Abstract
Filopodia, dynamic membrane protrusions driven by polymerization of an actin filament core, can adhere to the extracellular matrix and experience both external and cell-generated pulling forces. The role of such forces in filopodia adhesion is however insufficiently understood. Here, we study filopodia induced by overexpression of myosin X, typical for cancer cells. The lifetime of such filopodia positively correlates with the presence of myosin IIA filaments at the filopodia bases. Application of pulling forces to the filopodia tips through attached fibronectin-coated laser-trapped beads results in sustained growth of the filopodia. Pharmacological inhibition or knockdown of myosin IIA abolishes the filopodia adhesion to the beads. Formin inhibitor SMIFH2, which causes detachment of actin filaments from formin molecules, produces similar effect. Thus, centripetal force generated by myosin IIA filaments at the base of filopodium and transmitted to the tip through actin core in a formin-dependent fashion is required for filopodia adhesion.
Collapse
Affiliation(s)
- N O Alieva
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - A K Efremov
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore.,Center for BioImaging Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117557, Singapore
| | - S Hu
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - D Oh
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Z Chen
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore.,Department of Chemistry, University of California, Berkeley, CA, 94720, USA
| | - M Natarajan
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - H T Ong
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - A Jégou
- Institut Jacques Monod, CNRS, Université de Paris, 15 rue Helene Brion, F-75013, Paris, France
| | - G Romet-Lemonne
- Institut Jacques Monod, CNRS, Université de Paris, 15 rue Helene Brion, F-75013, Paris, France
| | - J T Groves
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore.,Department of Chemistry, University of California, Berkeley, CA, 94720, USA
| | - M P Sheetz
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore.,Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - J Yan
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore.,Center for BioImaging Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117557, Singapore.,Department of Physics, National University of Singapore, Singapore, 117542, Singapore
| | - A D Bershadsky
- Mechanobiology Institute, National University of Singapore, T-lab, 5A Engineering Drive 1, Singapore, 117411, Singapore. .,Weizmann Institute of Science, Herzl St 234, Rehovot, 7610001, Israel.
| |
Collapse
|
21
|
Sun YY, Yang YF, Keller KE. Myosin-X Silencing in the Trabecular Meshwork Suggests a Role for Tunneling Nanotubes in Outflow Regulation. Invest Ophthalmol Vis Sci 2019; 60:843-851. [PMID: 30807639 PMCID: PMC6390986 DOI: 10.1167/iovs.18-26055] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose The actin cytoskeleton plays a key role in outflow regulation through the trabecular meshwork (TM). Although actin stress fibers are a target of glaucoma therapies, the role of other actin cellular structures is unclear. Myosin-X (Myo10) is an actin-binding protein that is involved in tunneling nanotube (TNT) and filopodia formation. Here, we inhibited Myo10 pharmacologically or by gene silencing to investigate the role of filopodia/TNTs in the TM. Methods Short hairpin RNA interference (RNAi) silencing lentivirus targeting myosin-X (shMyo10) was generated. Human anterior segments were perfused with shMyo10 or CK-666, an Arp2/3 inhibitor. Confocal microscopy investigated the colocalization of Myo10 with matrix metalloproteinase (MMPs). Western immunoblotting investigated the protein levels of MMPs and extracellular matrix (ECM) proteins. MMP activity and phagocytosis assays were performed. Results CK-666 and shMyo10-silencing lentivirus caused a significant reduction in outflow rates in anterior segment perfusion culture, an ex vivo method to study intraocular pressure regulation. In human TM cells, Myo10 colocalized with MMP2, MMP14, and cortactin in podosome-like structures, which function as regions of focal ECM degradation. Furthermore, MMP activity, thrombospondin-1 and SPARC protein levels were significantly reduced in the media of CK-666-treated and shMyo10-silenced TM cells. However, neither Myo10 silencing or CK-666 treatment significantly affected phagocytic uptake. Conclusions Inhibiting filopodia/TNTs caused opposite effects on outflow compared with inhibiting stress fibers. Moreover, Myo10 may also play a role in focal ECM degradation in TM cells. Our results provide additional insight into the function of actin supramolecular assemblies and actin-binding proteins in outflow regulation.
Collapse
Affiliation(s)
- Ying Ying Sun
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Yong-Feng Yang
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Kate E Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
22
|
Caporizzo MA, Fishman CE, Sato O, Jamiolkowski RM, Ikebe M, Goldman YE. The Antiparallel Dimerization of Myosin X Imparts Bundle Selectivity for Processive Motility. Biophys J 2019; 114:1400-1410. [PMID: 29590597 DOI: 10.1016/j.bpj.2018.01.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 01/26/2018] [Accepted: 01/30/2018] [Indexed: 02/06/2023] Open
Abstract
Myosin X is an unconventional actin-based molecular motor involved in filopodial formation, microtubule-actin filament interaction, and cell migration. Myosin X is an important component of filopodia regulation, localizing to tips of growing filopodia by an unclear targeting mechanism. The native α-helical dimerization domain of myosin X is thought to associate with antiparallel polarity of the two amino acid chains, making myosin X the only myosin that is currently considered to form antiparallel dimers. This study aims to determine if antiparallel dimerization of myosin X imparts selectivity toward actin bundles by comparing the motility of parallel and antiparallel dimers of myosin X on single and fascin-bundled actin filaments. Antiparallel myosin X dimers exhibit selective processivity on fascin-bundled actin and are only weakly processive on single actin filaments below saturating [ATP]. Artificial forced parallel dimers of myosin X are robustly processive on both single and bundled actin, exhibiting no selectivity. To determine the relationship between gating of the reaction steps and observed differences in motility, a mathematical model was developed to correlate the parameters of motility with the biochemical and mechanical kinetics of the dimer. Results from the model, constrained by experimental data, suggest that the probability of binding forward, toward the barbed end of the actin filament, is lower in antiparallel myosin X on single actin filaments compared to fascin-actin bundles and compared to constructs of myosin X with parallel dimerization.
Collapse
Affiliation(s)
- Matthew A Caporizzo
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Claire E Fishman
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Osamu Sato
- Department of Cellular and Molecular Biology, University of Texas Science Center, Tyler, Texas
| | - Ryan M Jamiolkowski
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, University of Texas Science Center, Tyler, Texas
| | - Yale E Goldman
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
23
|
Tokuo H, Bhawan J, Coluccio LM. Myosin X is required for efficient melanoblast migration and melanoma initiation and metastasis. Sci Rep 2018; 8:10449. [PMID: 29993000 PMCID: PMC6041326 DOI: 10.1038/s41598-018-28717-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 06/15/2018] [Indexed: 12/20/2022] Open
Abstract
Myosin X (Myo10), an actin-associated molecular motor, has a clear role in filopodia induction and cell migration in vitro, but its role in vivo in mammals is not well understood. Here, we investigate the role of Myo10 in melanocyte lineage and melanoma induction. We found that Myo10 knockout (Myo10KO) mice exhibit a white spot on their belly caused by reduced melanoblast migration. Myo10KO mice crossed with available mice that conditionally express in melanocytes the BRAFV600E mutation combined with Pten silencing exhibited reduced melanoma development and metastasis, which extended medial survival time. Knockdown of Myo10 (Myo10kd) in B16F1 mouse melanoma cell lines decreased lung colonization after tail-vein injection. Myo10kd also inhibited long protrusion (LP) formation by reducing the transportation of its cargo molecule vasodilator-stimulated phosphoprotein (VASP) to the leading edge of migrating cells. These findings provide the first genetic evidence for the involvement of Myo10 not only in melanoblast migration, but also in melanoma development and metastasis.
Collapse
Affiliation(s)
- Hiroshi Tokuo
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, 02118, USA.
| | - Jag Bhawan
- Department of Dermatology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Lynne M Coluccio
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, 02118, USA
| |
Collapse
|
24
|
|
25
|
Abstract
The delivery of intracellular material within cells is crucial for maintaining normal function. Myosins transport a wide variety of cargo, ranging from vesicles to ribonuclear protein particles (RNPs), in plants, fungi, and metazoa. The properties of a given myosin transporter are adapted to move on different actin filament tracks, either on the disordered actin networks at the cell cortex or along highly organized actin bundles to distribute their cargo in a localized manner or move it across long distances in the cell. Transport is controlled by selective recruitment of the myosin to its cargo that also plays a role in activation of the motor.
Collapse
Affiliation(s)
- Margaret A Titus
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
26
|
Li YR, Yang WX. Myosins as fundamental components during tumorigenesis: diverse and indispensable. Oncotarget 2018; 7:46785-46812. [PMID: 27121062 PMCID: PMC5216836 DOI: 10.18632/oncotarget.8800] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 04/10/2016] [Indexed: 12/11/2022] Open
Abstract
Myosin is a kind of actin-based motor protein. As the crucial functions of myosin during tumorigenesis have become increasingly apparent, the profile of myosin in the field of cancer research has also been growing. Eighteen distinct classes of myosins have been discovered in the past twenty years and constitute a diverse superfamily. Various myosins share similar structures. They all convert energy from ATP hydrolysis to exert mechanical stress upon interactions with microfilaments. Ongoing research is increasingly suggesting that at least seven kinds of myosins participate in the formation and development of cancer. Myosins play essential roles in cytokinesis failure, chromosomal and centrosomal amplification, multipolar spindle formation and DNA microsatellite instability. These are all prerequisites of tumor formation. Subsequently, myosins activate various processes of tumor invasion and metastasis development including cell migration, adhesion, protrusion formation, loss of cell polarity and suppression of apoptosis. In this review, we summarize the current understanding of the roles of myosins during tumorigenesis and discuss the factors and mechanisms which may regulate myosins in tumor progression. Furthermore, we put forward a completely new concept of “chromomyosin” to demonstrate the pivotal functions of myosins during karyokinesis and how this acts to optimize the functions of the members of the myosin superfamily.
Collapse
Affiliation(s)
- Yan-Ruide Li
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
27
|
He K, Sakai T, Tsukasaki Y, Watanabe TM, Ikebe M. Myosin X is recruited to nascent focal adhesions at the leading edge and induces multi-cycle filopodial elongation. Sci Rep 2017; 7:13685. [PMID: 29057977 PMCID: PMC5651867 DOI: 10.1038/s41598-017-06147-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 05/16/2017] [Indexed: 12/16/2022] Open
Abstract
Filopodia protrude from the leading edge of cells and play important roles in cell motility. Here we report the mechanism of myosin X (encoded by Myo10)-induced multi-cycle filopodia extension. We found that actin, Arp2/3, vinculin and integrin-β first accumulated at the cell's leading edge. Myosin X was then gathered at these sites, gradually clustered by lateral movement, and subsequently initiated filopodia formation. During filopodia extension, we found the translocation of Arp2/3 and integrin-β along filopodia. Arp2/3 and integrin-β then became localized at the tip of filopodia, from where myosin X initiated the second extension of filopodia with a change in extension direction, thus producing long filopodia. Elimination of integrin-β, Arp2/3 and vinculin by siRNA significantly attenuated the myosin-X-induced long filopodia formation. We propose the following mechanism. Myosin X accumulates at nascent focal adhesions at the cell's leading edge, where myosin X promotes actin convergence to create the base of filopodia. Then myosin X moves to the filopodia tip and attracts integrin-β and Arp2/3 for further actin nucleation. The tip-located myosin X then initiates the second cycle of filopodia elongation to produce the long filopodia.
Collapse
Affiliation(s)
- Kangmin He
- Institute of Vascular Medicine, Peking University Third Hospital and Academy for Advanced Interdisciplinary Studies, Peking University, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.,Graduate School of Frontier Biosciences, Osaka University, Osaka, 5650871, Japan.,Department of Cell Biology, Harvard Medical School, and Cellular and Molecular Medicine Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Tsuyoshi Sakai
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, TX75708, USA
| | - Yoshikazu Tsukasaki
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, TX75708, USA.,Laboratory for Comprehensive Bioimaging, RIKEN Quantitative Biology Center (QBiC), Osaka, 5650874, Japan.,Department of Pharmacology, University of Illinois Chicago College of Medicine, Boston Children's Hospital, Chicago, Illinois 60612, USA
| | - Tomonobu M Watanabe
- Graduate School of Frontier Biosciences, Osaka University, Osaka, 5650871, Japan. .,Laboratory for Comprehensive Bioimaging, RIKEN Quantitative Biology Center (QBiC), Osaka, 5650874, Japan. .,World Premier International Research Center Initiative, iFReC, Osaka University, Osaka, 5650871, Japan.
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, TX75708, USA.
| |
Collapse
|
28
|
ROS induced distribution of mitochondria to filopodia by Myo19 depends on a class specific tryptophan in the motor domain. Sci Rep 2017; 7:11577. [PMID: 28912530 PMCID: PMC5599611 DOI: 10.1038/s41598-017-11002-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/17/2017] [Indexed: 01/08/2023] Open
Abstract
The role of the actin cytoskeleton in relation to mitochondria function and dynamics is only recently beginning to be recognized. Myo19 is an actin-based motor that is bound to the outer mitochondrial membrane and promotes the localization of mitochondria to filopodia in response to glucose starvation. However, how glucose starvation induces mitochondria localization to filopodia, what are the dynamics of this process and which enzymatic adaptation allows the translocation of mitochondria to filopodia are not known. Here we show that reactive oxygen species (ROS) mimic and mediate the glucose starvation induced phenotype. In addition, time-lapse fluorescent microscopy reveals that ROS-induced Myo19 motility is a highly dynamic process which is coupled to filopodia elongation and retraction. Interestingly, Myo19 motility is inhibited by back-to-consensus-mutation of a unique residue of class XIX myosins in the motor domain. Kinetic analysis of the purified mutant Myo19 motor domain reveals that the duty ratio (time spent strongly bound to actin) is highly compromised in comparison to that of the WT motor domain, indicating that Myo19 unique motor properties are necessary to propel mitochondria to filopodia tips. In summary, our study demonstrates the contribution of actin-based motility to the mitochondrial localization to filopodia by specific cellular cues.
Collapse
|
29
|
Defective Gpsm2/Gα i3 signalling disrupts stereocilia development and growth cone actin dynamics in Chudley-McCullough syndrome. Nat Commun 2017; 8:14907. [PMID: 28387217 PMCID: PMC5385604 DOI: 10.1038/ncomms14907] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 02/13/2017] [Indexed: 01/14/2023] Open
Abstract
Mutations in GPSM2 cause Chudley-McCullough syndrome (CMCS), an autosomal recessive neurological disorder characterized by early-onset sensorineural deafness and brain anomalies. Here, we show that mutation of the mouse orthologue of GPSM2 affects actin-rich stereocilia elongation in auditory and vestibular hair cells, causing deafness and balance defects. The G-protein subunit Gαi3, a well-documented partner of Gpsm2, participates in the elongation process, and its absence also causes hearing deficits. We show that Gpsm2 defines an ∼200 nm nanodomain at the tips of stereocilia and this localization requires the presence of Gαi3, myosin 15 and whirlin. Using single-molecule tracking, we report that loss of Gpsm2 leads to decreased outgrowth and a disruption of actin dynamics in neuronal growth cones. Our results elucidate the aetiology of CMCS and highlight a new molecular role for Gpsm2/Gαi3 in the regulation of actin dynamics in epithelial and neuronal tissues. Mutations in GPSM2 cause a rare disease characterized by deafness and brain abnormalities. Here the authors show that Gpsm2 forms a molecular complex with a heterotrimeric G-protein subunit, whirlin and a myosin motor to regulate actin dynamics in neurons and auditory hair cell stereocilia.
Collapse
|
30
|
Activated full-length myosin-X moves processively on filopodia with large steps toward diverse two-dimensional directions. Sci Rep 2017; 7:44237. [PMID: 28287133 PMCID: PMC5346999 DOI: 10.1038/srep44237] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 02/06/2017] [Indexed: 12/25/2022] Open
Abstract
Myosin-X, (Myo 10), is an unconventional myosin that transports the specific cargos to filopodial tips, and is associated with the mechanism underlying filopodia formation and extension. To clarify the innate motor characteristic, we studied the single molecule movement of a full-length myosin-X construct with leucine zipper at the C-terminal end of the tail (M10FullLZ) and the tail-truncated myosin-X without artificial dimerization motif (BAP-M101–979HMM). M10FullLZ localizes at the tip of filopodia like myosin-X full-length (M10Full). M10FullLZ moves on actin filaments in the presence of PI(3,4,5)P3, an activator of myosin-X. Single molecule motility analysis revealed that the step sizes of both M10FullLZ and BAP-M101–979HMM are widely distributed on single actin filaments that is consistent with electron microscopy observation. M10FullLZ moves on filopodial actin bundles of cells with a mean step size (~36 nm), similar to the step size on single actin filaments (~38 nm). Cartesian plot analysis revealed that M10FullLZ meandered on filopodial actin bundles to both x- and y- directions. These results suggest that the lever-arm of full-length myosin-X is flexible enough to processively steps on different actin filaments within the actin bundles of filopodia. This characteristic of myosin-X may facilitate actin filament convergence for filopodia production.
Collapse
|
31
|
Masters TA, Kendrick-Jones J, Buss F. Myosins: Domain Organisation, Motor Properties, Physiological Roles and Cellular Functions. Handb Exp Pharmacol 2017; 235:77-122. [PMID: 27757761 DOI: 10.1007/164_2016_29] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myosins are cytoskeletal motor proteins that use energy derived from ATP hydrolysis to generate force and movement along actin filaments. Humans express 38 myosin genes belonging to 12 classes that participate in a diverse range of crucial activities, including muscle contraction, intracellular trafficking, cell division, motility, actin cytoskeletal organisation and cell signalling. Myosin malfunction has been implicated a variety of disorders including deafness, hypertrophic cardiomyopathy, Usher syndrome, Griscelli syndrome and cancer. In this chapter, we will first discuss the key structural and kinetic features that are conserved across the myosin family. Thereafter, we summarise for each member in turn its unique functional and structural adaptations, cellular roles and associated pathologies. Finally, we address the broad therapeutic potential for pharmacological interventions that target myosin family members.
Collapse
Affiliation(s)
- Thomas A Masters
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK.
| | | | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| |
Collapse
|
32
|
Delage E, Cervantes DC, Pénard E, Schmitt C, Syan S, Disanza A, Scita G, Zurzolo C. Differential identity of Filopodia and Tunneling Nanotubes revealed by the opposite functions of actin regulatory complexes. Sci Rep 2016; 6:39632. [PMID: 28008977 PMCID: PMC5180355 DOI: 10.1038/srep39632] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 11/25/2016] [Indexed: 11/09/2022] Open
Abstract
Tunneling Nanotubes (TNTs) are actin enriched filopodia-like protrusions that play a pivotal role in long-range intercellular communication. Different pathogens use TNT-like structures as "freeways" to propagate across cells. TNTs are also implicated in cancer and neurodegenerative diseases, making them promising therapeutic targets. Understanding the mechanism of their formation, and their relation with filopodia is of fundamental importance to uncover their physiological function, particularly since filopodia, differently from TNTs, are not able to mediate transfer of cargo between distant cells. Here we studied different regulatory complexes of actin, which play a role in the formation of both these structures. We demonstrate that the filopodia-promoting CDC42/IRSp53/VASP network negatively regulates TNT formation and impairs TNT-mediated intercellular vesicle transfer. Conversely, elevation of Eps8, an actin regulatory protein that inhibits the extension of filopodia in neurons, increases TNT formation. Notably, Eps8-mediated TNT induction requires Eps8 bundling but not its capping activity. Thus, despite their structural similarities, filopodia and TNTs form through distinct molecular mechanisms. Our results further suggest that a switch in the molecular composition in common actin regulatory complexes is critical in driving the formation of either type of membrane protrusion.
Collapse
Affiliation(s)
- Elise Delage
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| | - Diégo Cordero Cervantes
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| | - Esthel Pénard
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| | - Christine Schmitt
- Ultrapole, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| | - Sylvie Syan
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| | - Andrea Disanza
- FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Giorgio Scita
- FIRC Institute of Molecular Oncology, 20139 Milan, Italy.,Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20122 Milan, Italy
| | - Chiara Zurzolo
- Unité Trafic Membranaire et Pathogenèse, Institut Pasteur, 25-28 Rue du Docteur Roux, 75724 Paris CEDEX 15, France
| |
Collapse
|
33
|
MyTH4-FERM myosins have an ancient and conserved role in filopod formation. Proc Natl Acad Sci U S A 2016; 113:E8059-E8068. [PMID: 27911821 DOI: 10.1073/pnas.1615392113] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The formation of filopodia in Metazoa and Amoebozoa requires the activity of myosin 10 (Myo10) in mammalian cells and of Dictyostelium unconventional myosin 7 (DdMyo7) in the social amoeba Dictyostelium However, the exact roles of these MyTH4-FERM myosins (myosin tail homology 4-band 4.1, ezrin, radixin, moesin; MF) in the initiation and elongation of filopodia are not well defined and may reflect conserved functions among phylogenetically diverse MF myosins. Phylogenetic analysis of MF myosin domains suggests that a single ancestral MF myosin existed with a structure similar to DdMyo7, which has two MF domains, and that subsequent duplications in the metazoan lineage produced its functional homolog Myo10. The essential functional features of the DdMyo7 myosin were identified using quantitative live-cell imaging to characterize the ability of various mutants to rescue filopod formation in myo7-null cells. The two MF domains were found to function redundantly in filopod formation with the C-terminal FERM domain regulating both the number of filopodia and their elongation velocity. DdMyo7 mutants consisting solely of the motor plus a single MyTH4 domain were found to be capable of rescuing the formation of filopodia, establishing the minimal elements necessary for the function of this myosin. Interestingly, a chimeric myosin with the Myo10 MF domain fused to the DdMyo7 motor also was capable of rescuing filopod formation in the myo7-null mutant, supporting fundamental functional conservation between these two distant myosins. Together, these findings reveal that MF myosins have an ancient and conserved role in filopod formation.
Collapse
|
34
|
Baboolal TG, Mashanov GI, Nenasheva TA, Peckham M, Molloy JE. A Combination of Diffusion and Active Translocation Localizes Myosin 10 to the Filopodial Tip. J Biol Chem 2016; 291:22373-22385. [PMID: 27566544 PMCID: PMC5077179 DOI: 10.1074/jbc.m116.730689] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 08/24/2016] [Indexed: 11/06/2022] Open
Abstract
Myosin 10 is an actin-based molecular motor that localizes to the tips of filopodia in mammalian cells. To understand how it is targeted to this distinct region of the cell, we have used total internal reflection fluorescence microscopy to study the movement of individual full-length and truncated GFP-tagged molecules. Truncation mutants lacking the motor region failed to localize to filopodial tips but still bound transiently at the plasma membrane. Deletion of the single α-helical and anti-parallel coiled-coil forming regions, which lie between the motor and pleckstrin homology domains, reduced the instantaneous velocity of intrafilopodial movement but did not affect the number of substrate adherent filopodia. Deletion of the anti-parallel coiled-coil forming region, but not the EKR-rich region of the single α-helical domain, restored intrafilopodial trafficking, suggesting this region is important in determining myosin 10 motility. We propose a model by which myosin 10 rapidly targets to the filopodial tip via a sequential reduction in dimensionality. Molecules first undergo rapid diffusion within the three-dimensional volume of the cell body. They then exhibit periods of slower two-dimensional diffusion in the plane of the plasma membrane. Finally, they move in a unidimensional, highly directed manner along the polarized actin filament bundle within the filopodium becoming confined to a single point at the tip. Here we have observed directly each phase of the trafficking process using single molecule fluorescence imaging of live cells and have quantified our observations using single particle tracking, autocorrelation analysis, and kymographs.
Collapse
Affiliation(s)
- Thomas G Baboolal
- From the Astbury Centre for Structural Molecular Biology and Institute of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT and
| | - Gregory I Mashanov
- The Francis Crick Institute, Mill Hill Laboratory, London NW7 1AA, United Kingdom
| | - Tatiana A Nenasheva
- The Francis Crick Institute, Mill Hill Laboratory, London NW7 1AA, United Kingdom
| | - Michelle Peckham
- From the Astbury Centre for Structural Molecular Biology and Institute of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT and
| | - Justin E Molloy
- The Francis Crick Institute, Mill Hill Laboratory, London NW7 1AA, United Kingdom
| |
Collapse
|
35
|
Tsai FC, Koenderink GH. Shape control of lipid bilayer membranes by confined actin bundles. SOFT MATTER 2015; 11:8834-8847. [PMID: 26395896 DOI: 10.1039/c5sm01583a] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In living cells, lipid membranes and biopolymers determine each other's conformation in a delicate force balance. Cellular polymers such as actin filaments are strongly confined by the plasma membrane in cell protrusions such as lamellipodia and filopodia. Conversely, protrusion formation is facilitated by actin-driven membrane deformation and these protrusions are maintained by dense actin networks or bundles of actin filaments. Here we investigate the mechanical interplay between actin bundles and lipid bilayer membranes by reconstituting a minimal model system based on cell-sized liposomes with encapsulated actin filaments bundled by fascin. To address the competition between the deformability of the membrane and the enclosed actin bundles, we tune the bundle stiffness (through the fascin-to-actin molar ratio) and the membrane rigidity (through protein decoration). Using confocal microscopy and quantitative image analysis, we show that actin bundles deform the liposomes into a rich set of morphologies. For liposomes having a small membrane bending rigidity, the actin bundles tend to generate finger-like membrane protrusions that resemble cellular filopodia. Stiffer bundles formed at high crosslink density stay straight in the liposome body, whereas softer bundles formed at low crosslink density are bent and kinked. When the membrane has a large bending rigidity, membrane protrusions are suppressed. In this case, membrane enclosure forces the actin bundles to organize into cortical rings, to minimize the energy cost associated with filament bending. Our results highlight the importance of taking into account mechanical interactions between the actin cytoskeleton and the membrane to understand cell shape control.
Collapse
Affiliation(s)
- Feng-Ching Tsai
- FOM Institute AMOLF, Systems Biophysics Department, Science Park 104, 1098 XG Amsterdam, The Netherlands.
| | - Gijsje Hendrika Koenderink
- FOM Institute AMOLF, Systems Biophysics Department, Science Park 104, 1098 XG Amsterdam, The Netherlands.
| |
Collapse
|
36
|
Yochelis A, Ebrahim S, Millis B, Cui R, Kachar B, Naoz M, Gov NS. Self-organization of waves and pulse trains by molecular motors in cellular protrusions. Sci Rep 2015; 5:13521. [PMID: 26335545 PMCID: PMC4558574 DOI: 10.1038/srep13521] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/29/2015] [Indexed: 11/13/2022] Open
Abstract
Actin-based cellular protrusions are an ubiquitous feature of cells, performing a variety of critical functions ranging from cell-cell communication to cell motility. The formation and maintenance of these protrusions relies on the transport of proteins via myosin motors, to the protrusion tip. While tip-directed motion leads to accumulation of motors (and their molecular cargo) at the protrusion tip, it is observed that motors also form rearward moving, periodic and isolated aggregates. The origins and mechanisms of these aggregates, and whether they are important for the recycling of motors, remain open puzzles. Motivated by novel myosin-XV experiments, a mass conserving reaction-diffusion-advection model is proposed. The model incorporates a non-linear cooperative interaction between motors, which converts them between an active and an inactive state. Specifically, the type of aggregate formed (traveling waves or pulse-trains) is linked to the kinetics of motors at the protrusion tip which is introduced by a boundary condition. These pattern selection mechanisms are found not only to qualitatively agree with empirical observations but open new vistas to the transport phenomena by molecular motors in general.
Collapse
Affiliation(s)
- A Yochelis
- Department of Solar Energy and Environmental Physics, Swiss Institute for Dryland Environmental and Energy Research, Blaustein Institutes for Desert Research (BIDR), Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Ben-Gurion 84990, Israel
| | - S Ebrahim
- Laboratory of Cell Structure and Dynamics, NIDCD, National Institutes of Health, 50 South Drive, Bethesda, MD 20892-8027, USA
| | - B Millis
- Laboratory of Cell Structure and Dynamics, NIDCD, National Institutes of Health, 50 South Drive, Bethesda, MD 20892-8027, USA
| | - R Cui
- Laboratory of Cell Structure and Dynamics, NIDCD, National Institutes of Health, 50 South Drive, Bethesda, MD 20892-8027, USA
| | - B Kachar
- Laboratory of Cell Structure and Dynamics, NIDCD, National Institutes of Health, 50 South Drive, Bethesda, MD 20892-8027, USA
| | - M Naoz
- Department of Chemical Physics, Weizmann Institute of Science, P.O.B. 26, Rehovot, Israel 76100
| | - N S Gov
- Department of Chemical Physics, Weizmann Institute of Science, P.O.B. 26, Rehovot, Israel 76100
| |
Collapse
|
37
|
Orly G, Naoz M, Gov NS. Physical model for the geometry of actin-based cellular protrusions. Biophys J 2015; 107:576-587. [PMID: 25099797 DOI: 10.1016/j.bpj.2014.05.040] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 05/09/2014] [Accepted: 05/28/2014] [Indexed: 11/17/2022] Open
Abstract
Actin-based cellular protrusions are a ubiquitous feature of cell morphology, e.g., filopodia and microvilli, serving a huge variety of functions. Despite this, there is still no comprehensive model for the mechanisms that determine the geometry of these protrusions. We present here a detailed computational model that addresses a combination of multiple biochemical and physical processes involved in the dynamic regulation of the shape of these protrusions. We specifically explore the role of actin polymerization in determining both the height and width of the protrusions. Furthermore, we show that our generalized model can explain multiple morphological features of these systems, and account for the effects of specific proteins and mutations.
Collapse
Affiliation(s)
- G Orly
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot, Israel
| | - M Naoz
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot, Israel
| | - N S Gov
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
38
|
Lai M, Guo Y, Ma J, Yu H, Zhao D, Fan W, Ju X, Sheikh MA, Malik YS, Xiong W, Guo W, Zhu X. Myosin X regulates neuronal radial migration through interacting with N-cadherin. Front Cell Neurosci 2015; 9:326. [PMID: 26347613 PMCID: PMC4539528 DOI: 10.3389/fncel.2015.00326] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 08/06/2015] [Indexed: 11/23/2022] Open
Abstract
Proper brain function depends on correct neuronal migration during development, which is known to be regulated by cytoskeletal dynamics and cell-cell adhesion. Myosin X (Myo10), an uncharacteristic member of the myosin family, is an important regulator of cytoskeleton that modulates cell motilities in many different cellular contexts. We previously reported that Myo10 was required for neuronal migration in the developing cerebral cortex, but the underlying mechanism was still largely unknown. Here, we found that knockdown of Myo10 expression disturbed the adherence of migrating neurons to radial glial fibers through abolishing surface Neuronal cadherin (N-cadherin) expression, thereby impaired neuronal migration in the developmental cortex. Next, we found Myo10 interacted with N-cadherin cellular domain through its FERM domain. Furthermore, we found knockdown of Myo10 disrupted N-cadherin subcellular distribution and led to localization of N-cadherin into Golgi apparatus and endosomal sorting vesicle. Taking together, these results reveal a novel mechanism of Myo10 interacting with N-cadherin and regulating its cell-surface expression, which is required for neuronal adhesion and migration.
Collapse
Affiliation(s)
- Mingming Lai
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China ; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Dali University Dali, China
| | - Ye Guo
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Jun Ma
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Huali Yu
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Dongdong Zhao
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Wenqiang Fan
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Xingda Ju
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Muhammad A Sheikh
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Yousra S Malik
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China
| | - Wencheng Xiong
- Department of Neurology, Georgia Regents University, Augusta GA, USA
| | - Weixiang Guo
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences Beijing, China
| | - Xiaojuan Zhu
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University Changchun, China ; State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences Beijing, China
| |
Collapse
|
39
|
Abstract
Myosin-X (Myo10) is a motor protein best known for its role in filopodia formation. New research implicates Myo10 in a number of disease states including cancer metastasis and pathogen infection. This review focuses on these developments with emphasis on the emerging roles of Myo10 in formation of cancer cell protrusions and metastasis. A number of aggressive cancers show high levels of Myo10 expression and knockdown of Myo10 has been shown to dramatically limit cancer cell motility in 2D and 3D systems. Myo10 knockdown also limits spread of intracellular pathogens marburgvirus and Shigella flexneri. Consideration is given to how these properties might arise and potential paths of future research.
Collapse
Affiliation(s)
- David S Courson
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Richard E Cheney
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
40
|
Chou AM, Sem KP, Wright GD, Sudhaharan T, Ahmed S. Dynamin1 is a novel target for IRSp53 protein and works with mammalian enabled (Mena) protein and Eps8 to regulate filopodial dynamics. J Biol Chem 2014; 289:24383-96. [PMID: 25031323 DOI: 10.1074/jbc.m114.553883] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Filopodia are dynamic actin-based structures that play roles in processes such as cell migration, wound healing, and axonal guidance. Cdc42 induces filopodial formation through IRSp53, an Inverse-Bin-Amphiphysins-Rvs (I-BAR) domain protein. Previous work from a number of laboratories has shown that IRSp53 generates filopodia by coupling membrane protrusion with actin dynamics through its Src homology 3 domain binding partners. Here, we show that dynamin1 (Dyn1), the large guanosine triphosphatase, is an interacting partner of IRSp53 through pulldown and Förster resonance energy transfer analysis, and we explore its role in filopodial formation. In neuroblastoma cells, Dyn1 localizes to filopodia, associated tip complexes, and the leading edge just behind the anti-capping protein mammalian enabled (Mena). Dyn1 knockdown reduces filopodial formation, which can be rescued by overexpressing wild-type Dyn1 but not the GTPase mutant Dyn1-K44A and the loss-of-function actin binding domain mutant Dyn1-K/E. Interestingly, dynasore, an inhibitor of Dyn GTPase, also reduced filopodial number and increased their lifetime. Using rapid time-lapse total internal reflection fluorescence microscopy, we show that Dyn1 and Mena localize to filopodia only during initiation and assembly. Dyn1 actin binding domain mutant inhibits filopodial formation, suggesting a role in actin elongation. In contrast, Eps8, an actin capping protein, is seen most strongly at filopodial tips during disassembly. Taken together, the results suggest IRSp53 partners with Dyn1, Mena, and Eps8 to regulate filopodial dynamics.
Collapse
Affiliation(s)
- Ai Mei Chou
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Kai Ping Sem
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Graham Daniel Wright
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Thankiah Sudhaharan
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Sohail Ahmed
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| |
Collapse
|
41
|
Myosin-10 produces its power-stroke in two phases and moves processively along a single actin filament under low load. Proc Natl Acad Sci U S A 2014; 111:E1833-42. [PMID: 24753602 DOI: 10.1073/pnas.1320122111] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Myosin-10 is an actin-based molecular motor that participates in essential intracellular processes such as filopodia formation/extension, phagocytosis, cell migration, and mitotic spindle maintenance. To study this motor protein's mechano-chemical properties, we used a recombinant, truncated form of myosin-10 consisting of the first 936 amino acids, followed by a GCN4 leucine zipper motif, to force dimerization. Negative-stain electron microscopy reveals that the majority of molecules are dimeric with a head-to-head contour distance of ∼50 nm. In vitro motility assays show that myosin-10 moves actin filaments smoothly with a velocity of ∼310 nm/s. Steady-state and transient kinetic analysis of the ATPase cycle shows that the ADP release rate (∼13 s(-1)) is similar to the maximum ATPase activity (∼12-14 s(-1)) and therefore contributes to rate limitation of the enzymatic cycle. Single molecule optical tweezers experiments show that under intermediate load (∼0.5 pN), myosin-10 interacts intermittently with actin and produces a power stroke of ∼17 nm, composed of an initial 15-nm and subsequent 2-nm movement. At low optical trap loads, we observed staircase-like processive movements of myosin-10 interacting with the actin filament, consisting of up to six ∼35-nm steps per binding interaction. We discuss the implications of this load-dependent processivity of myosin-10 as a filopodial transport motor.
Collapse
|
42
|
Sackmann E, Smith AS. Physics of cell adhesion: some lessons from cell-mimetic systems. SOFT MATTER 2014; 10:1644-59. [PMID: 24651316 PMCID: PMC4028615 DOI: 10.1039/c3sm51910d] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Cell adhesion is a paradigm of the ubiquitous interplay of cell signalling, modulation of material properties and biological functions of cells. It is controlled by competition of short range attractive forces, medium range repellant forces and the elastic stresses associated with local and global deformation of the composite cell envelopes. We review the basic physical rules governing the physics of cell adhesion learned by studying cell-mimetic systems and demonstrate the importance of these rules in the context of cellular systems. We review how adhesion induced micro-domains couple to the intracellular actin and microtubule networks allowing cells to generate strong forces with a minimum of attractive cell adhesion molecules (CAMs) and to manipulate other cells through filopodia over micrometer distances. The adhesion strength can be adapted to external force fluctuations within seconds by varying the density of attractive and repellant CAMs through exocytosis and endocytosis or protease-mediated dismantling of the CAM-cytoskeleton link. Adhesion domains form local end global biochemical reaction centres enabling the control of enzymes. Actin-microtubule crosstalk at adhesion foci facilitates the mechanical stabilization of polarized cell shapes. Axon growth in tissue is guided by attractive and repulsive clues controlled by antagonistic signalling pathways.
Collapse
Affiliation(s)
- Erich Sackmann
- Physics Department Technical University Munich, Germany
- Department of Physics, Ludwig-Maximillian University, Munich, Germany
| | - Ana-Sunčana Smith
- Institute for Theoretical Physics, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
- Institute Rud̷er Bošković, Zagreb, Croatia.
| |
Collapse
|
43
|
Lai M, Li L, Ju X, Yu H, Zhu X. Cloning, characterization, and promoter analysis of mouse Myo10 gene. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2013; 32:354-65. [PMID: 23742061 DOI: 10.1080/15257770.2013.790551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Myosin X (Myo10) is an unconventional myosin associated with filopodia motility. Recent studies show that in addition to full-length Myo10, brain expresses a shorter form of Myo10 that lacks a myosin motor domain named headless Myo10. Herein, we analyzed and cloned 2-kb of the 5'-upstream sequences of mouse full-length Myo10 (fMyo10) and headless Myo10 (hMyo10) to understand the transcriptional regulation of the Myo10 gene. The putative transcription factor binding sites and CpG island were analyzed by a bioinformatic approach. Luciferase reporter assays showed that the 2-kb of 5'-upstream sequences of both fMyo10 and hMyo10 had promoter activities.
Collapse
Affiliation(s)
- Mingming Lai
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | | | | | | | | |
Collapse
|
44
|
Bao J, Huck D, Gunther LK, Sellers JR, Sakamoto T. Actin structure-dependent stepping of myosin 5a and 10 during processive movement. PLoS One 2013; 8:e74936. [PMID: 24069366 PMCID: PMC3777900 DOI: 10.1371/journal.pone.0074936] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 08/07/2013] [Indexed: 11/22/2022] Open
Abstract
How myosin 10, an unconventional myosin, walks processively along actin is still controversial. Here, we used single molecule fluorescence techniques, TIRF and FIONA, to study the motility and the stepping mechanism of dimerized myosin 10 heavy-meromyosin-like fragment on both single actin filaments and two-dimensional F-actin rafts cross-linked by fascin or α-actinin. As a control, we also tracked and analyzed the stepping behavior of the well characterized processive motor myosin 5a. We have shown that myosin 10 moves processively along both single actin filaments and F-actin rafts with a step size of 31 nm. Moreover, myosin 10 moves more processively on fascin-F-actin rafts than on α-actinin-F-actin rafts, whereas myosin 5a shows no such selectivity. Finally, on fascin-F-actin rafts, myosin 10 has more frequent side steps to adjacent actin filaments than myosin 5a in the F-actin rafts. Together, these results reveal further single molecule features of myosin 10 on various actin structures, which may help to understand its cellular functions.
Collapse
Affiliation(s)
- Jianjun Bao
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan, United States of America
| | - Daniel Huck
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan, United States of America
| | - Laura K. Gunther
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan, United States of America
| | - James R. Sellers
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Takeshi Sakamoto
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan, United States of America
- Department of Physiology, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
45
|
Linch M, Sanz-Garcia M, Soriano E, Zhang Y, Riou P, Rosse C, Cameron A, Knowles P, Purkiss A, Kjaer S, McDonald NQ, Parker PJ. A cancer-associated mutation in atypical protein kinase Cι occurs in a substrate-specific recruitment motif. Sci Signal 2013; 6:ra82. [PMID: 24045153 DOI: 10.1126/scisignal.2004068] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024]
Abstract
Atypical protein kinase Cι (PKCι) has roles in cell growth, cellular polarity, and migration, and its abundance is frequently increased in cancer. We identified a protein interaction surface containing a dibasic motif (RIPR) that bound a distinct subset of PKCι substrates including lethal giant larvae 2 (LLGL2) and myosin X, but not other substrates such as Par3. Further characterization demonstrated that Arg471 in this motif was important for binding to LLGL2, whereas Arg474 was critical for interaction with myosin X, indicating that multiple complexes could be formed through this motif. A somatic mutation of the dibasic motif (R471C) was the most frequent mutation of PKCι in human cancer, and the intact dibasic motif was required for normal polarized epithelial morphogenesis in three-dimensional cysts. Thus, the R471C substitution is a change-of-function mutation acting at this substrate-specific recruitment site to selectively disrupt the polarizing activity of PKCι.
Collapse
Affiliation(s)
- Mark Linch
- 1Protein Phosphorylation Laboratory, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Bornschlögl T. How filopodia pull: what we know about the mechanics and dynamics of filopodia. Cytoskeleton (Hoboken) 2013; 70:590-603. [PMID: 23959922 DOI: 10.1002/cm.21130] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/31/2013] [Accepted: 08/01/2013] [Indexed: 01/04/2023]
Abstract
In recent years, the dynamic, hair-like cell protrusions called filopodia have attracted considerable attention. They have been found in a multitude of different cell types and are often called "sensory organelles," since they seem to sense the mechanical and chemical environment of a cell. Once formed, filopodia can exhibit complex behavior, they can grow and retract, push or pull, and transform into distinct structures. They are often found to make first adhesive contact with the extracellular matrix, pathogens or with adjacent cells, and to subsequently exert pulling forces. Much is known about the cytoskeletal players involved in filopodia formation, but only recently have we started to explore the mechanics of filopodia together with the related cytoskeletal dynamics. This review summarizes current advancements in our understanding of the mechanics and dynamics of filopodia, with a focus on the molecular mechanisms behind filopodial force exertion.
Collapse
Affiliation(s)
- Thomas Bornschlögl
- Institut Curie, Laboratoire, Physico-Chimie UMR CNRS, 168, 11 Rue Pierre et Marie Curie, 75005, Paris, France
| |
Collapse
|
47
|
Pinkoviezky I, Gov NS. Transport dynamics of molecular motors that switch between an active and inactive state. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2013; 88:022714. [PMID: 24032871 DOI: 10.1103/physreve.88.022714] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 06/11/2013] [Indexed: 06/02/2023]
Abstract
Molecular motors are involved in key transport processes in the cell. Many of these motors can switch from an active to a nonactive state, either spontaneously or depending on their interaction with other molecules. When active, the motors move processively along the filaments, while when inactive they are stationary. We treat here the simple case of spontaneously switching motors, between the active and inactive states, along an open linear track. We use our recent analogy with vehicular traffic, where we go beyond the mean-field description. We map the phase diagram of this system, and find that it clearly breaks the symmetry between the different phases, as compared to the standard total asymmetric exclusion process. We make several predictions that may be testable using molecular motors in vitro and in living cells.
Collapse
Affiliation(s)
- I Pinkoviezky
- Department of Chemical Physics, Weizmann Institute of Science, P.O. Box 26, Rehovot, Israel 76100
| | | |
Collapse
|
48
|
Gousset K, Marzo L, Commere PH, Zurzolo C. Myo10 is a key regulator of TNT formation in neuronal cells. J Cell Sci 2013; 126:4424-35. [PMID: 23886947 DOI: 10.1242/jcs.129239] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cell-to-cell communication is essential in multicellular organisms. Tunneling nanotubes (TNTs) have emerged as a new type of intercellular spreading mechanism allowing the transport of various signals, organelles and pathogens. Here, we study the role of the unconventional molecular motor myosin-X (Myo10) in the formation of functional TNTs within neuronal CAD cells. Myo10 protein expression increases the number of TNTs and the transfer of vesicles between co-cultured cells. We also show that TNT formation requires both the motor and tail domains of the protein, and identify the F2 lobe of the FERM domain within the Myo10 tail as necessary for TNT formation. Taken together, these results indicate that, in neuronal cells, TNTs can arise from a subset of Myo10-driven dorsal filopodia, independent of its binding to integrins and N-cadherins. In addition our data highlight the existence of different mechanisms for the establishment and regulation of TNTs in neuronal cells and other cell types.
Collapse
Affiliation(s)
- Karine Gousset
- Institut Pasteur, 25 Rue du Dr Roux, Unité de Traffic Membranaire et Pathogenèse, 75724 Paris Cedex 15, France
| | | | | | | |
Collapse
|
49
|
Single quantum dot tracking reveals that an individual multivalent HIV-1 Tat protein transduction domain can activate machinery for lateral transport and endocytosis. Mol Cell Biol 2013; 33:3036-49. [PMID: 23732912 DOI: 10.1128/mcb.01717-12] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The mechanisms underlying the cellular entry of the HIV-1 Tat protein transduction domain (TatP) and the molecular information necessary to improve the transduction efficiency of TatP remain unclear due to the technical limitations for direct visualization of TatP's behavior in cells. Using confocal microscopy, total internal reflection fluorescence microscopy, and four-dimensional microscopy, we developed a single-molecule tracking assay for TatP labeled with quantum dots (QDs) to examine the kinetics of TatP initially and immediately before, at the beginning of, and immediately after entry into living cells. We report that even when the number of multivalent TatP (mTatP)-QDs bound to a cell was low, each single mTatP-QD first locally induced the cell's lateral transport machinery to move the mTatP-QD toward the center of the cell body upon cross-linking of heparan sulfate proteoglycans. The centripetal and lateral movements were linked to the integrity and flow of actomyosin and microtubules. Individual mTatP underwent lipid raft-mediated temporal confinement, followed by complete immobilization, which ultimately led to endocytotic internalization. However, bivalent TatP did not sufficiently promote either cell surface movement or internalization. Together, these findings provide clues regarding the mechanisms of TatP cell entry and indicate that increasing the valence of TatP on nanoparticles allows them to behave as cargo delivery nanomachines.
Collapse
|
50
|
Gallo G. Mechanisms underlying the initiation and dynamics of neuronal filopodia: from neurite formation to synaptogenesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 301:95-156. [PMID: 23317818 DOI: 10.1016/b978-0-12-407704-1.00003-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Filopodia are finger-like cellular protrusions found throughout the metazoan kingdom and perform fundamental cellular functions during development and cell migration. Neurons exhibit a wide variety of extremely complex morphologies. In the nervous system, filopodia underlie many major morphogenetic events. Filopodia have roles spanning the initiation and guidance of neuronal processes, axons and dendrites to the formation of synaptic connections. This chapter addresses the mechanisms of the formation and dynamics of neuronal filopodia. Some of the major lessons learned from the study of neuronal filopodia are (1) there are multiple mechanisms that can regulate filopodia in a context-dependent manner, (2) that filopodia are specialized subcellular domains, (3) that filopodia exhibit dynamic membrane recycling which also controls aspects of filopodial dynamics, (4) that neuronal filopodia contain machinery for the orchestration of the actin and microtubule cytoskeleton, and (5) localized protein synthesis contributes to neuronal filopodial dynamics.
Collapse
Affiliation(s)
- Gianluca Gallo
- Shriners Hospitals Pediatric Research Center, Center for Neural Repair and Rehabilitation, Department of Anatomy and Cell Biology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|