1
|
Garone ME, Chase SE, Zhang C, Krendel M. Myosin 1e deficiency affects migration of 4T1 breast cancer cells. Cytoskeleton (Hoboken) 2024; 81:723-736. [PMID: 38140937 PMCID: PMC11193843 DOI: 10.1002/cm.21819] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/03/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023]
Abstract
Metastasis of breast cancer cells to distant tissue sites is responsible for the majority of deaths associated with breast cancer. Previously we have examined the role of class I myosin motor protein, myosin 1e (myo1e), in cancer metastasis using the Mouse Mammary Tumor Virus-Polyoma Middle T Antigen (MMTV-PyMT) mouse model. Mice deficient in myo1e formed tumors with a more differentiated phenotype relative to the wild-type mice and formed no detectable lung metastases. In the current study, we investigated how the absence of myo1e affects cell migration and invasion in vitro, using the highly invasive and migratory breast cancer cell line, 4T1. 4T1 cells deficient in myo1e exhibited an altered morphology and slower rates of migration in the wound-healing and transwell migration assays compared to the WT 4T1 cells. While integrin trafficking and Golgi reorientation did not appear to be altered upon myo1e loss, we observed lower rates of focal adhesion disassembly in myo1e-deficient cells, which could help explain the cell migration defect.
Collapse
Affiliation(s)
- Michael E. Garone
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Sharon E. Chase
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Chunling Zhang
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Mira Krendel
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
2
|
Liu PJ, Sayeeda K, Zhuang C, Krendel M. Roles of myosin 1e and the actin cytoskeleton in kidney functions and familial kidney disease. Cytoskeleton (Hoboken) 2024; 81:737-752. [PMID: 38708443 PMCID: PMC11538376 DOI: 10.1002/cm.21861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 05/07/2024]
Abstract
Mammalian kidneys are responsible for removing metabolic waste and maintaining fluid and electrolyte homeostasis via selective filtration. One of the proteins closely linked to selective renal filtration is myosin 1e (Myo1e), an actin-dependent molecular motor found in the specialized kidney epithelial cells involved in the assembly and maintenance of the renal filter. Point mutations in the gene encoding Myo1e, MYO1E, have been linked to familial kidney disease, and Myo1e knockout in mice leads to the disruption of selective filtration. In this review, we discuss the role of the actin cytoskeleton in renal filtration, the known and hypothesized functions of Myo1e, and the possible explanations for the impact of MYO1E mutations on renal function.
Collapse
Affiliation(s)
- Pei-Ju Liu
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Kazi Sayeeda
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Cindy Zhuang
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Mira Krendel
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
3
|
Li T, Song Y, Wei L, Song X, Duan R. Disulfidptosis: a novel cell death modality induced by actin cytoskeleton collapse and a promising target for cancer therapeutics. Cell Commun Signal 2024; 22:491. [PMID: 39394612 PMCID: PMC11470700 DOI: 10.1186/s12964-024-01871-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024] Open
Abstract
Disulfidptosis is a novel discovered form of programmed cell death (PCD) that diverges from apoptosis, necroptosis, ferroptosis, and cuproptosis, stemming from disulfide stress-induced cytoskeletal collapse. In cancer cells exhibiting heightened expression of the solute carrier family 7 member 11 (SLC7A11), excessive cystine importation and reduction will deplete nicotinamide adenine dinucleotide phosphate (NADPH) under glucose deprivation, followed by an increase in intracellular disulfide stress and aberrant disulfide bond formation within actin networks, ultimately culminating in cytoskeletal collapse and disulfidptosis. Disulfidptosis involves crucial physiological processes in eukaryotic cells, such as cystine and glucose uptake, NADPH metabolism, and actin dynamics. The Rac1-WRC pathway-mediated actin polymerization is also implicated in this cell death due to its contribution to disulfide bond formation. However, the precise mechanisms underlying disulfidptosis and its role in tumors are not well understood. This is probably due to the multifaceted functionalities of SLC7A11 within cells and the complexities of the downstream pathways driving disulfidptosis. This review describes the critical roles of SLC7A11 in cells and summarizes recent research advancements in the potential pathways of disulfidptosis. Moreover, the less-studied aspects of this newly discovered cell death process are highlighted to stimulate further investigations in this field.
Collapse
Affiliation(s)
- Tianyi Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Song
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Lijuan Wei
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Xiangyi Song
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Ruifeng Duan
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China.
| |
Collapse
|
4
|
Xu M, Rutkowski DM, Rebowski G, Boczkowska M, Pollard LW, Dominguez R, Vavylonis D, Ostap EM. Myosin-I synergizes with Arp2/3 complex to enhance the pushing forces of branched actin networks. SCIENCE ADVANCES 2024; 10:eado5788. [PMID: 39270022 PMCID: PMC11397503 DOI: 10.1126/sciadv.ado5788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024]
Abstract
Class I myosins (myosin-Is) colocalize with Arp2/3 complex-nucleated actin networks at sites of membrane protrusion and invagination, but the mechanisms by which myosin-I motor activity coordinates with branched actin assembly to generate force are unknown. We mimicked the interplay of these proteins using the "comet tail" bead motility assay, where branched actin networks are nucleated by the Arp2/3 complex on the surface of beads coated with myosin-I and nucleation-promoting factor. We observed that myosin-I increased bead movement efficiency by thinning actin networks without affecting growth rates. Myosin-I triggered symmetry breaking and comet tail formation in dense networks resistant to spontaneous fracturing. Even with arrested actin assembly, myosin-I alone could break the network. Computational modeling recapitulated these observations, suggesting myosin-I acts as a repulsive force shaping the network's architecture and boosting its force-generating capacity. We propose that myosin-I leverages its power stroke to amplify the forces generated by Arp2/3 complex-nucleated actin networks.
Collapse
Affiliation(s)
- Mengqi Xu
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Grzegorz Rebowski
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Malgorzata Boczkowska
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Luther W. Pollard
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roberto Dominguez
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - E. Michael Ostap
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Tsai FC, Guérin G, Pernier J, Bassereau P. Actin-membrane linkers: Insights from synthetic reconstituted systems. Eur J Cell Biol 2024; 103:151402. [PMID: 38461706 DOI: 10.1016/j.ejcb.2024.151402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/10/2024] [Accepted: 02/28/2024] [Indexed: 03/12/2024] Open
Abstract
At the cell surface, the actin cytoskeleton and the plasma membrane interact reciprocally in a variety of processes related to the remodeling of the cell surface. The actin cytoskeleton has been known to modulate membrane organization and reshape the membrane. To this end, actin-membrane linking molecules play a major role in regulating actin assembly and spatially direct the interaction between the actin cytoskeleton and the membrane. While studies in cells have provided a wealth of knowledge on the molecular composition and interactions of the actin-membrane interface, the complex molecular interactions make it challenging to elucidate the precise actions of the actin-membrane linkers at the interface. Synthetic reconstituted systems, consisting of model membranes and purified proteins, have been a powerful approach to elucidate how actin-membrane linkers direct actin assembly to drive membrane shape changes. In this review, we will focus only on several actin-membrane linkers that have been studied by using reconstitution systems. We will discuss the design principles of these reconstitution systems and how they have contributed to the understanding of the cellular functions of actin-membrane linkers. Finally, we will provide a perspective on future research directions in understanding the intricate actin-membrane interaction.
Collapse
Affiliation(s)
- Feng-Ching Tsai
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Physics of Cells and Cancer, Paris 75005, France.
| | - Gwendal Guérin
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Physics of Cells and Cancer, Paris 75005, France
| | - Julien Pernier
- Tumor Cell Dynamics Unit, Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif 94800, France
| | - Patricia Bassereau
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Physics of Cells and Cancer, Paris 75005, France.
| |
Collapse
|
6
|
Fülle JB, de Almeida RA, Lawless C, Stockdale L, Yanes B, Lane EB, Garrod DR, Ballestrem C. Proximity Mapping of Desmosomes Reveals a Striking Shift in Their Molecular Neighborhood Associated With Maturation. Mol Cell Proteomics 2024; 23:100735. [PMID: 38342409 PMCID: PMC10943070 DOI: 10.1016/j.mcpro.2024.100735] [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: 05/04/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 02/13/2024] Open
Abstract
Desmosomes are multiprotein adhesion complexes that link intermediate filaments to the plasma membrane, ensuring the mechanical integrity of cells across tissues, but how they participate in the wider signaling network to exert their full function is unclear. To investigate this, we carried out protein proximity mapping using biotinylation (BioID). The combined interactomes of the essential desmosomal proteins desmocollin 2a, plakoglobin, and plakophilin 2a (Pkp2a) in Madin-Darby canine kidney epithelial cells were mapped and their differences and commonalities characterized as desmosome matured from Ca2+ dependence to the mature, Ca2+-independent, hyper-adhesive state, which predominates in tissues. Results suggest that individual desmosomal proteins have distinct roles in connecting to cellular signaling pathways and that these roles alter substantially when cells change their adhesion state. The data provide further support for a dualistic concept of desmosomes in which the properties of Pkp2a differ from those of the other, more stable proteins. This body of data provides an invaluable resource for the analysis of desmosome function.
Collapse
Affiliation(s)
- Judith B Fülle
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | | | - Craig Lawless
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Liam Stockdale
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Bian Yanes
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - E Birgitte Lane
- Skin Research Institute of Singapore, Agency of Science Technology and Research (A∗STAR), Singapore, Singapore
| | - David R Garrod
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.
| |
Collapse
|
7
|
Xu M, Rutkowski DM, Rebowski G, Boczkowska M, Pollard LW, Dominguez R, Vavylonis D, Ostap EM. Myosin-I Synergizes with Arp2/3 Complex to Enhance Pushing Forces of Branched Actin Networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579714. [PMID: 38405741 PMCID: PMC10888859 DOI: 10.1101/2024.02.09.579714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Myosin-Is colocalize with Arp2/3 complex-nucleated actin networks at sites of membrane protrusion and invagination, but the mechanisms by which myosin-I motor activity coordinates with branched actin assembly to generate force are unknown. We mimicked the interplay of these proteins using the "comet tail" bead motility assay, where branched actin networks are nucleated by Arp2/3 complex on the surface of beads coated with myosin-I and the WCA domain of N-WASP. We observed that myosin-I increased bead movement efficiency by thinning actin networks without affecting growth rates. Remarkably, myosin-I triggered symmetry breaking and comet-tail formation in dense networks resistant to spontaneous fracturing. Even with arrested actin assembly, myosin-I alone could break the network. Computational modeling recapitulated these observations suggesting myosin-I acts as a repulsive force shaping the network's architecture and boosting its force-generating capacity. We propose that myosin-I leverages its power stroke to amplify the forces generated by Arp2/3 complex-nucleated actin networks.
Collapse
Affiliation(s)
- Mengqi Xu
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | | | - Grzegorz Rebowski
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Malgorzata Boczkowska
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Luther W Pollard
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Roberto Dominguez
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | | | - E Michael Ostap
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
8
|
Yang M, Batey JE, Dong B. Automated Five-Dimensional Single Particle Tracking by Bifocal Parallax Dark-Field Microscopy with Electronic Tunable Lens. Anal Chem 2024; 96:1-5. [PMID: 38153091 DOI: 10.1021/acs.analchem.3c04543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
We present a novel method for the precise tracking of plasmonic gold nanorods (AuNRs) in live cells, enabling a comprehensive understanding of the nanocargo's cellular dynamics. Traditional single particle tracking (SPT) struggles with accurately determining all five spatial parameters (x, y, z, ϕ, and θ) in live cells due to various challenges. Our innovation combines electronic tunable lens (ETL) technology with bifocal parallax dark-field (DF) microscopy, allowing continuous adjustment of the imaging focal plane for automatic tracking of both translational and rotational movements of AuNRs. This 5D single-particle orientation and rotational tracking (5D SPORT) method achieves remarkable precision, with 3D localization precisions of 9 (x), 10 (y), and 15 nm (z) and angular resolutions below 2°. To showcase its applicability, we investigated intracellular transport of nanocargos using transferrin-modified AuNRs as the imaging probe. Differentiated transport stages, such as active transport and pause period, were clearly unveiled from the observed dynamics in 5D. This advancement in single particle tracking holds promise for a wide range of applications in biomedical research, particularly when combined with other imaging modalities, such as light sheet fluorescence microscopy.
Collapse
Affiliation(s)
- Meek Yang
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - James Ethan Batey
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Bin Dong
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas 72701, United States
| |
Collapse
|
9
|
Pedersen RT, Snoberger A, Pyrpassopoulos S, Safer D, Drubin DG, Ostap EM. Endocytic myosin-1 is a force-insensitive, power-generating motor. J Cell Biol 2023; 222:e202303095. [PMID: 37549220 PMCID: PMC10406613 DOI: 10.1083/jcb.202303095] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/17/2023] [Accepted: 07/24/2023] [Indexed: 08/09/2023] Open
Abstract
Myosins are required for clathrin-mediated endocytosis, but their precise molecular roles in this process are not known. This is, in part, because the biophysical properties of the relevant motors have not been investigated. Myosins have diverse mechanochemical activities, ranging from powerful contractility against mechanical loads to force-sensitive anchoring. To better understand the essential molecular contribution of myosin to endocytosis, we studied the in vitro force-dependent kinetics of the Saccharomyces cerevisiae endocytic type I myosin called Myo5, a motor whose role in clathrin-mediated endocytosis has been meticulously studied in vivo. We report that Myo5 is a low-duty-ratio motor that is activated ∼10-fold by phosphorylation and that its working stroke and actin-detachment kinetics are relatively force-insensitive. Strikingly, the in vitro mechanochemistry of Myo5 is more like that of cardiac myosin than that of slow anchoring myosin-1s found on endosomal membranes. We, therefore, propose that Myo5 generates power to augment actin assembly-based forces during endocytosis in cells.
Collapse
Affiliation(s)
- Ross T.A. Pedersen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Aaron Snoberger
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Serapion Pyrpassopoulos
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Safer
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David G. Drubin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - E. Michael Ostap
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Willet AH, Chen JS, Ren L, Gould KL. Membrane binding of endocytic myosin-1s is inhibited by a class of ankyrin repeat proteins. Mol Biol Cell 2023; 34:br17. [PMID: 37531259 PMCID: PMC10559312 DOI: 10.1091/mbc.e23-06-0233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/04/2023] Open
Abstract
Myosin-1s are monomeric actin-based motors that function at membranes. Myo1 is the single myosin-1 isoform in Schizosaccharomyces pombe that works redundantly with Wsp1-Vrp1 to activate the Arp2/3 complex for endocytosis. Here, we identified Ank1 as an uncharacterized cytoplasmic Myo1 binding partner. We found that in ank1Δ cells, Myo1 dramatically redistributed from endocytic patches to decorate the entire plasma membrane and endocytosis was defective. Biochemical analysis and structural predictions suggested that the Ank1 ankyrin repeats bind the Myo1 lever arm and the Ank1 acidic tail binds the Myo1 TH1 domain to prevent TH1-dependent Myo1 membrane binding. Indeed, Ank1 overexpression precluded Myo1 membrane localization and recombinant Ank1 reduced purified Myo1 liposome binding in vitro. Based on biochemical and cell biological analyses, we propose budding yeast Ank1 and human OSTF1 are functional Ank1 orthologs and that cytoplasmic sequestration by small ankyrin repeat proteins is a conserved mechanism regulating myosin-1s in endocytosis.
Collapse
Affiliation(s)
- Alaina H. Willet
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Jun-Song Chen
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Liping Ren
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Kathleen L. Gould
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
11
|
Liu S, Liu P, Fei X, Zhu C, Hou J, Wang X, Pan Y. Analysis and validation of the potential of the MYO1E gene in pancreatic adenocarcinoma based on a bioinformatics approach. Oncol Lett 2023; 26:285. [PMID: 37274465 PMCID: PMC10236097 DOI: 10.3892/ol.2023.13871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/22/2023] [Indexed: 06/06/2023] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a common digestive cancer, and its prognosis is poor. Myosin 1E (MYO1E) is a class I myosin family member whose expression and function have not been reported in PAAD. In the present study, bioinformatics analysis was used to explore the expression levels of MYO1E in PAAD and its prognostic value, and the immunological role of MYO1E in PAAD was analyzed. The study revealed that a variety of malignancies have substantially increased MYO1E expression. Further investigation demonstrated that PAAD tissues exhibited greater levels of MYO1E mRNA and protein expression than normal tissues. High MYO1E expression is associated with poor prognosis in patients with PAAD. MYO1E expression was also associated with pathological stage in patients with PAAD. Functional enrichment analysis demonstrated that MYO1E was linked to multiple tumor-related mechanisms in PAAD. The pancreatic adenocarcinoma tumor microenvironment (TME) was analyzed and it was revealed that MYO1E expression was positively associated with tumor immune cell infiltration. In addition, MYO1E was closely associated with some tumor chemokines/receptors and immune checkpoints. In vitro experiments revealed that the suppression of MYO1E expression could inhibit pancreatic adenocarcinoma cell proliferation, invasion and migration. Through preliminary analysis, the present study evaluated the potential function of MYO1E in PAAD and its function in TME, and MYO1E may become a potential biomarker for PAAD.
Collapse
Affiliation(s)
- Songbai Liu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Peng Liu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Xiaobin Fei
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Changhao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Junyi Hou
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Xing Wang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
- Department of Hepatobiliary Surgery, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Yaozhen Pan
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
- Department of Hepatobiliary Surgery, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| |
Collapse
|
12
|
Pedersen RTA, Snoberger A, Pyrpassopoulos S, Safer D, Drubin DG, Ostap EM. Endocytic myosin-1 is a force-insensitive, power-generating motor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.21.533689. [PMID: 36993306 PMCID: PMC10055380 DOI: 10.1101/2023.03.21.533689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Myosins are required for clathrin-mediated endocytosis, but their precise molecular roles in this process are not known. This is, in part, because the biophysical properties of the relevant motors have not been investigated. Myosins have diverse mechanochemical activities, ranging from powerful contractility against mechanical loads to force-sensitive anchoring. To better understand the essential molecular contribution of myosin to endocytosis, we studied the in vitro force-dependent kinetics of the Saccharomyces cerevisiae endocytic type I myosin called Myo5, a motor whose role in clathrin-mediated endocytosis has been meticulously studied in vivo. We report that Myo5 is a low-duty-ratio motor that is activated ∼10-fold by phosphorylation, and that its working stroke and actin-detachment kinetics are relatively force-insensitive. Strikingly, the in vitro mechanochemistry of Myo5 is more like that of cardiac myosin than like that of slow anchoring myosin-1s found on endosomal membranes. We therefore propose that Myo5 generates power to augment actin assembly-based forces during endocytosis in cells. Summary Pedersen, Snoberger et al. measure the force-sensitivity of the yeast endocytic the myosin-1 called Myo5 and find that it is more likely to generate power than to serve as a force-sensitive anchor in cells. Implications for Myo5's role in clathrin-mediated endocytosis are discussed.
Collapse
Affiliation(s)
- Ross TA Pedersen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
- Present address: Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218
- Equal Contribution
| | - Aaron Snoberger
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Equal Contribution
| | - Serapion Pyrpassopoulos
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Daniel Safer
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - David G Drubin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - E Michael Ostap
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
13
|
Willet AH, Chen JS, Ren L, Gould KL. Membrane binding of endocytic myosin-1s is inhibited by a class of ankyrin repeat proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.26.538419. [PMID: 37163016 PMCID: PMC10168314 DOI: 10.1101/2023.04.26.538419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Myosin-1s are monomeric actin-based motors that function at membranes. Myo1 is the single myosin-1 isoform in Schizosaccharomyces pombe that works redundantly with Wsp1-Vrp1 to activate the Arp2/3 complex for endocytosis. Here, we identified Ank1 as an uncharacterized cytoplasmic Myo1 binding partner. We found that in ank1Δ cells, Myo1 dramatically redistributed from endocytic patches to decorate the entire plasma membrane and endocytosis was defective. Biochemical analysis and structural predictions suggested that the Ank1 ankyrin repeats bind the Myo1 lever arm and the Ank1 acidic tail binds the Myo1 TH1 domain to prevent TH1-dependent Myo1 membrane binding. Indeed, Ank1 over-expression precluded Myo1 membrane localization and recombinant Ank1 blocked purified Myo1 liposome binding in vitro. Based on biochemical and cell biology analyses, we propose budding yeast Ank1 and human OSTF1 are functional Ank1 orthologs and that cytoplasmic sequestration by small ankyrin repeat proteins is a conserved mechanism regulating myosin-1s in endocytosis. Summary Fission yeast long-tailed myosin-1 binds Ank1. Ank1 ankyrin repeats associate with the Myo1 lever arm and Ank1 acidic tail binds the Myo1 TH1 domain to inhibit Myo1 membrane binding. Ank1 orthologs exists in budding yeast (Ank1) and humans (OSTF1).
Collapse
Affiliation(s)
- Alaina H Willet
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Jun-Song Chen
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Liping Ren
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Kathleen L Gould
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
14
|
Jusue-Torres I, Tiv R, Ricarte-Filho JC, Mallisetty A, Contreras-Vargas L, Godoy-Calderon MJ, Khaddour K, Kennedy K, Valyi-Nagy K, David O, Menchaca M, Kottorou A, Koutras A, Dimitrakopoulos F, Abdelhady KM, Massad M, Rubinstein I, Feldman L, Stewart J, Shimamura T, Danilova L, Hulbert A. Myo1e overexpression in lung adenocarcinoma is associated with increased risk of mortality. Sci Rep 2023; 13:4107. [PMID: 36914720 PMCID: PMC10011530 DOI: 10.1038/s41598-023-30765-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
This study aims to perform a comprehensive genomic analysis to assess the influence of overexpression of MYO1E in non-small cell lung carcinoma (NSCLC) and whether there are differences in survival and mortality risk in NSCLC patients depending on both DNA methylation and RNA expression of MYO1E. The DNA methylation probe cg13887966 was inversely correlated with MYO1E RNA expression in both LUAD and LUSC subpopulations showing that lower MYO1E RNA expression was associated with higher MYO1E DNA methylation. Late stages of lung cancer showed significantly lower MYO1E DNA methylation and significantly higher MYO1E RNA expression for LUAD but not for LUSC. Low DNA methylation as well as high RNA expression of MYO1E are associated with a shorter median survival time and an increased risk of mortality for LUAD, but not for LUSC. This study suggests that changes in MYO1E methylation and expression in LUAD patients may have an essential role in lung cancer's pathogenesis. It shows the utility of MYO1E DNA methylation and RNA expression in predicting survival for LUAD patients. Also, given the low normal expression of MYO1E in blood cells MYO1E DNA methylation has the potential to be used as circulating tumor marker in liquid biopsies.
Collapse
Affiliation(s)
| | - Richies Tiv
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | | | - Apurva Mallisetty
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Leglys Contreras-Vargas
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | | | - Karam Khaddour
- Division of Hematology Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Kathleen Kennedy
- Division of Hematology Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Klara Valyi-Nagy
- Department of Pathology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Odile David
- Department of Pathology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Martha Menchaca
- Department of Radiology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Anastasia Kottorou
- Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, Patras, Greece
| | - Angelos Koutras
- Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, Patras, Greece
| | - Foteinos Dimitrakopoulos
- Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, Patras, Greece
| | | | - Malek Massad
- Division of Cardiothoracic Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Israel Rubinstein
- Medical and Research Services, Jesse Brown VA Medical Center, Chicago, IL, USA
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Lawrence Feldman
- Division of Hematology Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Medical and Research Services, Jesse Brown VA Medical Center, Chicago, IL, USA
| | - John Stewart
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Section of Surgical Oncology, Department of Surgery, Louisiana State University, New Orleans, LA, USA
| | - Takeshi Shimamura
- Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
- Division of Cardiothoracic Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Ludmila Danilova
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Laboratory of Systems Biology and Computational Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Alicia Hulbert
- Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
- Medical and Research Services, Jesse Brown VA Medical Center, Chicago, IL, USA.
- Department of Surgery, University of Illinois College of Medicine, 909 South Wolcott Ave, COMRB Suite 5140, Chicago, IL, 60612, USA.
| |
Collapse
|
15
|
Leonov S, Inyang O, Achkasov K, Bogdan E, Kontareva E, Chen Y, Fu Y, Osipov AN, Pustovalova M, Merkher Y. Proteomic Markers for Mechanobiological Properties of Metastatic Cancer Cells. Int J Mol Sci 2023; 24:ijms24054773. [PMID: 36902201 PMCID: PMC10003476 DOI: 10.3390/ijms24054773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/26/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
The major cause (more than 90%) of all cancer-related deaths is metastasis, thus its prediction can critically affect the survival rate. Metastases are currently predicted by lymph-node status, tumor size, histopathology and genetic testing; however, all these are not infallible, and obtaining results may require weeks. The identification of new potential prognostic factors will be an important source of risk information for the practicing oncologist, potentially leading to enhanced patient care through the proactive optimization of treatment strategies. Recently, the new mechanobiology-related techniques, independent of genetics, based on the mechanical invasiveness of cancer cells (microfluidic, gel indentation assays, migration assays etc.), demonstrated a high success rate for the detection of tumor cell metastasis propensity. However, they are still far away from clinical implementation due to complexity. Hence, the exploration of novel markers related to the mechanobiological properties of tumor cells may have a direct impact on the prognosis of metastasis. Our concise review deepens our knowledge of the factors that regulate cancer cell mechanotype and invasion, and incites further studies to develop therapeutics that target multiple mechanisms of invasion for improved clinical benefit. It may open a new clinical dimension that will improve cancer prognosis and increase the effectiveness of tumor therapies.
Collapse
Affiliation(s)
- Sergey Leonov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
- Institute of Cell Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
| | - Olumide Inyang
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| | - Konstantin Achkasov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| | - Elizaveta Bogdan
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| | - Elizaveta Kontareva
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ying Fu
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Andreyan N. Osipov
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical-Biological Agency, 123098 Moscow, Russia
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia
- Correspondence:
| | - Margarita Pustovalova
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical-Biological Agency, 123098 Moscow, Russia
| | - Yulia Merkher
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow Region, Russia
| |
Collapse
|
16
|
Banerjee P, Tan X, Russell WK, Kurie JM. Analysis of Golgi Secretory Functions in Cancer. Methods Mol Biol 2022; 2557:785-810. [PMID: 36512251 DOI: 10.1007/978-1-0716-2639-9_47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer cells utilize secretory pathways for paracrine signaling and extracellular matrix remodeling to facilitate directional cell migration, invasion, and metastasis. The Golgi apparatus is a central secretory signaling hub that is often deregulated in cancer. Here we described technologies that utilize microscopic, biochemical, and proteomic approaches to analyze Golgi secretory functions in genetically heterogeneous cancer cell lines.
Collapse
Affiliation(s)
- Priyam Banerjee
- Frits and Rita Markus Bio-Imaging Resource Center, The Rockefeller University, New York, NY, USA
| | - Xiaochao Tan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William K Russell
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Jonathan M Kurie
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
17
|
Diaz-Valencia JD, Estrada-Abreo LA, Rodríguez-Cruz L, Salgado-Aguayo AR, Patiño-López G. Class I Myosins, molecular motors involved in cell migration and cancer. Cell Adh Migr 2022; 16:1-12. [PMID: 34974807 PMCID: PMC8741282 DOI: 10.1080/19336918.2021.2020705] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 01/13/2023] Open
Abstract
Class I Myosins are a subfamily of motor proteins with ATPase activity and a characteristic structure conserved in all myosins: A N-Terminal Motor Domain, a central Neck and a C terminal Tail domain. Humans have eight genes for these myosins. Class I Myosins have different functions: regulate membrane tension, participate in endocytosis, exocytosis, intracellular trafficking and cell migration. Cell migration is influenced by many cellular components including motor proteins, like myosins. Recently has been reported that changes in myosin expression have an impact on the migration of cancer cells, the formation of infiltrates and metastasis. We propose that class I myosins might be potential markers for future diagnostic, prognostic or even as therapeutic targets in leukemia and other cancers.Abbreviations: Myo1g: Myosin 1g; ALL: Acute Lymphoblastic Leukemia, TH1: Tail Homology 1; TH2: Tail Homology 2; TH3: Tail Homology 3.
Collapse
Affiliation(s)
- Juan D. Diaz-Valencia
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
| | - Laura A. Estrada-Abreo
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
- Cell Biology and Flow Cytometry Laboratory, Metropolitan Autonomous University, México City, Mexico
| | - Leonor Rodríguez-Cruz
- Cell Biology and Flow Cytometry Laboratory, Metropolitan Autonomous University, México City, Mexico
| | - Alfonso R. Salgado-Aguayo
- Rheumatic Diseases Laboratory, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Genaro Patiño-López
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
| |
Collapse
|
18
|
Liu PJ, Gunther LK, Garone ME, Zhang C, Perez D, Bi-Karchin J, Pellenz CD, Chase SE, Presti MF, Plante EL, Martin CE, Lovric S, Yengo CM, Hildebrandt F, Krendel M. Steroid-Resistant Nephrotic Syndrome-Associated MYO1E Mutations Have Differential Effects on Myosin 1e Localization, Dynamics, and Activity. J Am Soc Nephrol 2022; 33:1989-2007. [PMID: 36316095 PMCID: PMC9678034 DOI: 10.1681/asn.2021111505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 08/22/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Myo1e is a nonmuscle motor protein enriched in podocytes. Mutations in MYO1E are associated with steroid-resistant nephrotic syndrome (SRNS). Most of the MYO1E variants identified by genomic sequencing have not been functionally characterized. Here, we set out to analyze two mutations in the Myo1e motor domain, T119I and D388H, which were selected on the basis of protein sequence conservation. METHODS EGFP-tagged human Myo1e constructs were delivered into the Myo1e-KO mouse podocyte-derived cells via adenoviral infection to analyze Myo1e protein stability, Myo1e localization, and clathrin-dependent endocytosis, which is known to involve Myo1e activity. Furthermore, truncated Myo1e constructs were expressed using the baculovirus expression system and used to measure Myo1e ATPase and motor activity in vitro. RESULTS Both mutants were expressed as full-length proteins in the Myo1e-KO cells. However, unlike wild-type (WT) Myo1e, the T119I variant was not enriched at the cell junctions or clathrin-coated vesicles (CCVs). In contrast, D388H variant localization was similar to that of WT. The rate of dissociation of the D388H variant from cell-cell junctions and CCVs was decreased, suggesting this mutation affects Myo1e interactions with binding partners. ATPase activity and ability to translocate actin filaments were drastically reduced for the D388H mutant, supporting findings from cell-based experiments. CONCLUSIONS T119I and D388H mutations are deleterious to Myo1e functions. The experimental approaches used in this study can be applied to future characterization of novel MYO1E variants associated with SRNS.
Collapse
Affiliation(s)
- Pei-Ju Liu
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York
| | - Laura K. Gunther
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Michael E. Garone
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York
| | - Chunling Zhang
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York
| | - Diana Perez
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York
| | - Jing Bi-Karchin
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York
| | - Christopher D. Pellenz
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York
| | - Sharon E. Chase
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York
| | - Maria F. Presti
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York
| | - Eric L. Plante
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York
| | - Claire E. Martin
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Svjetlana Lovric
- Divison of Nephrology, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Christopher M. Yengo
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania
| | - Friedhelm Hildebrandt
- Divison of Nephrology, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mira Krendel
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York
| |
Collapse
|
19
|
Bending over backwards: BAR proteins and the actin cytoskeleton in mammalian receptor-mediated endocytosis. Eur J Cell Biol 2022; 101:151257. [PMID: 35863103 DOI: 10.1016/j.ejcb.2022.151257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/21/2022] Open
Abstract
The role of the actin cytoskeleton during receptor-mediated endocytosis (RME) has been well characterized in yeast for many years. Only more recently has the interplay between the actin cytoskeleton and RME been extensively explored in mammalian cells. These studies have revealed the central roles of BAR proteins in RME, and have demonstrated significant roles of BAR proteins in linking the actin cytoskeleton to this cellular process. The actin cytoskeleton generates and transmits mechanical force to promote the extension of receptor-bound endocytic vesicles into the cell. Many adaptor proteins link and regulate the actin cytoskeleton at the sites of endocytosis. This review will cover key effectors, adaptors and signalling molecules that help to facilitate the invagination of the cell membrane during receptor-mediated endocytosis, including recent insights gained on the roles of BAR proteins. The final part of this review will explore associations of alterations to genes encoding BAR proteins with cancer.
Collapse
|
20
|
Pernier J, Schauer K. Does the Actin Network Architecture Leverage Myosin-I Functions? BIOLOGY 2022; 11:biology11070989. [PMID: 36101369 PMCID: PMC9311500 DOI: 10.3390/biology11070989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 11/16/2022]
Abstract
The actin cytoskeleton plays crucial roles in cell morphogenesis and functions. The main partners of cortical actin are molecular motors of the myosin superfamily. Although our understanding of myosin functions is heavily based on myosin-II and its ability to dimerize, the largest and most ancient class is represented by myosin-I. Class 1 myosins are monomeric, actin-based motors that regulate a wide spectrum of functions, and whose dysregulation mediates multiple human diseases. We highlight the current challenges in identifying the “pantograph” for myosin-I motors: we need to reveal how conformational changes of myosin-I motors lead to diverse cellular as well as multicellular phenotypes. We review several mechanisms for scaling, and focus on the (re-) emerging function of class 1 myosins to remodel the actin network architecture, a higher-order dynamic scaffold that has potential to leverage molecular myosin-I functions. Undoubtfully, understanding the molecular functions of myosin-I motors will reveal unexpected stories about its big partner, the dynamic actin cytoskeleton.
Collapse
Affiliation(s)
- Julien Pernier
- Institute for Integrative Biology of the Cell (I2BC), Centre National de la Recherche Scientifique (CNRS), Commissariat à L’Énergie Atomique et aux Énergies Alternatives (CEA), Université Paris-Saclay, 91198 Gif-sur-Yvette, France;
| | - Kristine Schauer
- Tumor Cell Dynamics Unit, Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, 94800 Villejuif, France
- Correspondence:
| |
Collapse
|
21
|
Matozo T, Kogachi L, de Alencar BC. Myosin motors on the pathway of viral infections. Cytoskeleton (Hoboken) 2022; 79:41-63. [PMID: 35842902 DOI: 10.1002/cm.21718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/25/2022] [Accepted: 07/07/2022] [Indexed: 01/30/2023]
Abstract
Molecular motors are microscopic machines that use energy from adenosine triphosphate (ATP) hydrolysis to generate movement. While kinesins and dynein are molecular motors associated with microtubule tracks, myosins bind to and move on actin filaments. Mammalian cells express several myosin motors. They power cellular processes such as endo- and exocytosis, intracellular trafficking, transcription, migration, and cytokinesis. As viruses navigate through cells, they may take advantage or be hindered by host components and machinery, including the cytoskeleton. This review delves into myosins' cell roles and compares them to their reported functions in viral infections. In most cases, the previously described myosin functions align with their reported role in viral infections, although not in all cases. This opens the possibility that knowledge obtained from studying myosins in viral infections might shed light on new physiological roles for myosins in cells. However, given the high number of myosins expressed and the variety of viruses investigated in the different studies, it is challenging to infer whether the interactions found are specific to a single virus or can be applied to other viruses with the same characteristics. We conclude that the participation of myosins in viral cycles is still a largely unexplored area, especially concerning unconventional myosins.
Collapse
Affiliation(s)
- Tais Matozo
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Leticia Kogachi
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bruna Cunha de Alencar
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
22
|
Tian X, Bunda P, Ishibe S. Podocyte Endocytosis in Regulating the Glomerular Filtration Barrier. Front Med (Lausanne) 2022; 9:801837. [PMID: 35223901 PMCID: PMC8866310 DOI: 10.3389/fmed.2022.801837] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/06/2022] [Indexed: 12/26/2022] Open
Abstract
Endocytosis is a mechanism that internalizes and recycles plasma membrane components and transmembrane receptors via vesicle formation, which is mediated by clathrin-dependent and clathrin-independent signaling pathways. Podocytes are specialized, terminally differentiated epithelial cells in the kidney, located on the outermost layer of the glomerulus. These cells play an important role in maintaining the integrity of the glomerular filtration barrier in conjunction with the adjacent basement membrane and endothelial cell layers within the glomerulus. An intact podocyte endocytic machinery appears to be necessary for maintaining podocyte function. De novo pathologic human genetic mutations and loss-of-function studies of critical podocyte endocytosis genes in genetically engineered mouse models suggest that this pathway contributes to the pathophysiology of development and progression of proteinuria in chronic kidney disease. Here, we review the mechanism of cellular endocytosis and its regulation in podocyte injury in the context of glomerular diseases. A thorough understanding of podocyte endocytosis may shed novel insights into its biological function in maintaining a functioning filter and offer potential targeted therapeutic strategies for proteinuric glomerular diseases.
Collapse
Affiliation(s)
| | | | - Shuta Ishibe
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
23
|
The EMT activator ZEB1 accelerates endosomal trafficking to establish a polarity axis in lung adenocarcinoma cells. Nat Commun 2021; 12:6354. [PMID: 34732702 PMCID: PMC8566461 DOI: 10.1038/s41467-021-26677-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a transcriptionally governed process by which cancer cells establish a front-rear polarity axis that facilitates motility and invasion. Dynamic assembly of focal adhesions and other actin-based cytoskeletal structures on the leading edge of motile cells requires precise spatial and temporal control of protein trafficking. Yet, the way in which EMT-activating transcriptional programs interface with vesicular trafficking networks that effect cell polarity change remains unclear. Here, by utilizing multiple approaches to assess vesicular transport dynamics through endocytic recycling and retrograde trafficking pathways in lung adenocarcinoma cells at distinct positions on the EMT spectrum, we find that the EMT-activating transcription factor ZEB1 accelerates endocytosis and intracellular trafficking of plasma membrane-bound proteins. ZEB1 drives turnover of the MET receptor tyrosine kinase by hastening receptor endocytosis and transport to the lysosomal compartment for degradation. ZEB1 relieves a plus-end-directed microtubule-dependent kinesin motor protein (KIF13A) and a clathrin-associated adaptor protein complex subunit (AP1S2) from microRNA-dependent silencing, thereby accelerating cargo transport through the endocytic recycling and retrograde vesicular pathways, respectively. Depletion of KIF13A or AP1S2 mitigates ZEB1-dependent focal adhesion dynamics, front-rear axis polarization, and cancer cell motility. Thus, ZEB1-dependent transcriptional networks govern vesicular trafficking dynamics to effect cell polarity change. The way in which metastatic tumour cells control endocytic vesicular trafficking networks to establish a front-rear polarity axis that facilitates motility remains unclear. Here, the authors show that the EMT activator ZEB1 influences vesicular trafficking dynamics to execute cell polarity change.
Collapse
|
24
|
Link F, Borges AR, Jones NG, Engstler M. To the Surface and Back: Exo- and Endocytic Pathways in Trypanosoma brucei. Front Cell Dev Biol 2021; 9:720521. [PMID: 34422837 PMCID: PMC8377397 DOI: 10.3389/fcell.2021.720521] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/06/2021] [Indexed: 01/10/2023] Open
Abstract
Trypanosoma brucei is one of only a few unicellular pathogens that thrives extracellularly in the vertebrate host. Consequently, the cell surface plays a critical role in both immune recognition and immune evasion. The variant surface glycoprotein (VSG) coats the entire surface of the parasite and acts as a flexible shield to protect invariant proteins against immune recognition. Antigenic variation of the VSG coat is the major virulence mechanism of trypanosomes. In addition, incessant motility of the parasite contributes to its immune evasion, as the resulting fluid flow on the cell surface drags immunocomplexes toward the flagellar pocket, where they are internalized. The flagellar pocket is the sole site of endo- and exocytosis in this organism. After internalization, VSG is rapidly recycled back to the surface, whereas host antibodies are thought to be transported to the lysosome for degradation. For this essential step to work, effective machineries for both sorting and recycling of VSGs must have evolved in trypanosomes. Our understanding of the mechanisms behind VSG recycling and VSG secretion, is by far not complete. This review provides an overview of the trypanosome secretory and endosomal pathways. Longstanding questions are pinpointed that, with the advent of novel technologies, might be answered in the near future.
Collapse
Affiliation(s)
- Fabian Link
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Alyssa R Borges
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Nicola G Jones
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Markus Engstler
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, Würzburg, Germany
| |
Collapse
|
25
|
Schäringer K, Maxeiner S, Schalla C, Rütten S, Zenke M, Sechi A. LSP1-myosin1e bimolecular complex regulates focal adhesion dynamics and cell migration. FASEB J 2021; 35:e21268. [PMID: 33470457 DOI: 10.1096/fj.202000740rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 01/22/2023]
Abstract
Several cytoskeleton-associated proteins and signaling pathways work in concert to regulate actin cytoskeleton remodeling, cell adhesion, and migration. Although the leukocyte-specific protein 1 (LSP1) has been shown to interact with the actin cytoskeleton, its function in the regulation of actin cytoskeleton dynamics is, as yet, not fully understood. We have recently demonstrated that the bimolecular complex between LSP1 and myosin1e controls actin cytoskeleton remodeling during phagocytosis. In this study, we show that LSP1 downregulation severely impairs cell migration, lamellipodia formation, and focal adhesion dynamics in macrophages. Inhibition of the interaction between LSP1 and myosin1e also impairs these processes resulting in poorly motile cells, which are characterized by few and small lamellipodia. Furthermore, cells in which LSP1-myosin1e interaction is inhibited are typically associated with inefficient focal adhesion turnover. Collectively, our findings show that the LSP1-myosin1e bimolecular complex plays a pivotal role in the regulation of actin cytoskeleton remodeling and focal adhesion dynamics required for cell migration.
Collapse
Affiliation(s)
- Katja Schäringer
- Department of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Sebastian Maxeiner
- Department of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Carmen Schalla
- Department of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Stephan Rütten
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Martin Zenke
- Department of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Antonio Sechi
- Department of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
26
|
Chakrabarti R, Lee M, Higgs HN. Multiple roles for actin in secretory and endocytic pathways. Curr Biol 2021; 31:R603-R618. [PMID: 34033793 DOI: 10.1016/j.cub.2021.03.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Actin filaments play multiple roles in the secretory pathway and in endosome dynamics in mammals, including maintenance of Golgi structure, release of membrane cargo from the trans-Golgi network (TGN), endocytosis, and endosomal sorting dynamics. In addition, TGN carrier transport and endocytosis both occur by multiple mechanisms in mammals. Actin likely plays a role in at least four mammalian endocytic pathways, five pathways for membrane release from the TGN, and three processes involving endosomes. Also, the mammalian Golgi structure is highly dynamic, and actin is likely important for these dynamics. One challenge for many of these processes is the need to deal with other membrane-associated structures, such as the cortical actin network at the plasma membrane or the matrix that surrounds the Golgi. Arp2/3 complex is a major actin assembly factor in most of the processes mentioned, but roles for formins and tandem WH2-motif-containing assembly factors are being elucidated and are anticipated to grow with further study. The specific role for actin has not been defined for most of these processes, but is likely to involve the generation of force for membrane dynamics, either by actin polymerization itself or by myosin motor activity. Defining these processes mechanistically is necessary for understanding membrane dynamics in general, as well as pathways that utilize these processes, such as autophagy.
Collapse
Affiliation(s)
- Rajarshi Chakrabarti
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Miriam Lee
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Henry N Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA.
| |
Collapse
|
27
|
Pillon M, Doublet P. Myosins, an Underestimated Player in the Infectious Cycle of Pathogenic Bacteria. Int J Mol Sci 2021; 22:ijms22020615. [PMID: 33435466 PMCID: PMC7826972 DOI: 10.3390/ijms22020615] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/15/2022] Open
Abstract
Myosins play a key role in many cellular processes such as cell migration, adhesion, intracellular trafficking and internalization processes, making them ideal targets for bacteria. Through selected examples, such as enteropathogenic E. coli (EPEC), Neisseria, Salmonella, Shigella, Listeria or Chlamydia, this review aims to illustrate how bacteria target and hijack host cell myosins in order to adhere to the cell, to enter the cell by triggering their internalization, to evade from the cytosolic autonomous cell defense, to promote the biogenesis of intracellular replicative niche, to disseminate in tissues by cell-to-cell spreading, to exit out the host cell, and also to evade from macrophage phagocytosis. It highlights the diversity and sophistication of the strategy evolved by bacteria to manipulate one of their privileged targets, the actin cytoskeleton.
Collapse
Affiliation(s)
- Margaux Pillon
- CIRI, Centre International de Recherche en Infectiologie, Legionella Pathogenesis Group, Université de Lyon, 69007 Lyon, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, 69007 Lyon, France
- Ecole Normale Supérieure de Lyon, 69007 Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, 69007 Lyon, France
- Centre National de la Recherche Scientifique, UMR5308, 69007 Lyon, France
| | - Patricia Doublet
- CIRI, Centre International de Recherche en Infectiologie, Legionella Pathogenesis Group, Université de Lyon, 69007 Lyon, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, 69007 Lyon, France
- Ecole Normale Supérieure de Lyon, 69007 Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, 69007 Lyon, France
- Centre National de la Recherche Scientifique, UMR5308, 69007 Lyon, France
- Correspondence:
| |
Collapse
|
28
|
Nakamura S, Masuyama R, Sakai K, Fukuda K, Takeda K, Tanimura S. SH3P2 suppresses osteoclast differentiation through restricting membrane localization of myosin 1E. Genes Cells 2020; 25:707-717. [PMID: 32916757 DOI: 10.1111/gtc.12806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/23/2020] [Accepted: 09/02/2020] [Indexed: 01/21/2023]
Abstract
Osteoclasts are multinucleated cells responsible for bone resorption. Src homology 3 (SH3) domain-containing protein-2 (SH3P2)/osteoclast-stimulating factor-1 regulates osteoclast differentiation, but its exact role remains elusive. Here, we show that SH3P2 suppresses osteoclast differentiation. SH3P2 knockout (KO) mice displayed decreased femoral trabecular bone mass and enhanced localization of osteoclasts on the tibial trabecular bone surface, suggesting that SH3P2 suppresses bone resorption by osteoclasts. Osteoclast differentiation based on cellular multinuclearity induced by macrophage colony-stimulating factor and receptor activator of nuclear factor-κB ligand (RANKL) was enhanced in bone marrow-derived macrophages lacking SH3P2. RANKL induced SH3P2 dephosphorylation, which increased the association of actin-dependent motor protein myosin 1E (Myo1E) with SH3P2 and thereby prevented Myo1E localization to the plasma membrane. Consistent with this, Myo1E in the membrane fraction increased in SH3P2-KO cells. Together with the attenuated osteoclast differentiation in Myo1E knocked down cells, SH3P2 may suppress osteoclast differentiation by preventing their cell-to-cell fusion depending on Myo1E membrane localization.
Collapse
Affiliation(s)
- Shota Nakamura
- Department of Cell Regulation, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Ritsuko Masuyama
- Department of Gastronomy Management, College of Gastronomy Management, Ritsumeikan University, Kusatsu, Japan
| | - Kosuke Sakai
- Department of Cell Regulation, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Karin Fukuda
- Department of Cell Regulation, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kohsuke Takeda
- Department of Cell Regulation, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Susumu Tanimura
- Department of Cell Regulation, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
29
|
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
|
30
|
A myosin-7B-dependent endocytosis pathway mediates cellular entry of α-synuclein fibrils and polycation-bearing cargos. Proc Natl Acad Sci U S A 2020; 117:10865-10875. [PMID: 32366666 DOI: 10.1073/pnas.1918617117] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Cell-to-cell transmission of misfolding-prone α-synuclein (α-Syn) has emerged as a key pathological event in Parkinson's disease. This process is initiated when α-Syn-bearing fibrils enter cells via clathrin-mediated endocytosis, but the underlying mechanisms are unclear. Using a CRISPR-mediated knockout screen, we identify SLC35B2 and myosin-7B (MYO7B) as critical endocytosis regulators for α-Syn preformed fibrils (PFFs). We show that SLC35B2, as a key regulator of heparan sulfate proteoglycan (HSPG) biosynthesis, is essential for recruiting α-Syn PFFs to the cell surface because this process is mediated by interactions between negatively charged sugar moieties of HSPGs and clustered K-T-K motifs in α-Syn PFFs. By contrast, MYO7B regulates α-Syn PFF cell entry by maintaining a plasma membrane-associated actin network that controls membrane dynamics. Without MYO7B or actin filaments, many clathrin-coated pits fail to be severed from the membrane, causing accumulation of large clathrin-containing "scars" on the cell surface. Intriguingly, the requirement for MYO7B in endocytosis is restricted to α-Syn PFFs and other polycation-bearing cargos that enter cells via HSPGs. Thus, our study not only defines regulatory factors for α-Syn PFF endocytosis, but also reveals a previously unknown endocytosis mechanism for HSPG-binding cargos in general, which requires forces generated by MYO7B and actin filaments.
Collapse
|
31
|
Shen H, Bao Y, Feng C, Fu H, Mao J. Overexpression of Myo1e promotes albumin endocytosis by mouse glomerular podocytes mediated by Dynamin. PeerJ 2020; 8:e8599. [PMID: 32211226 PMCID: PMC7083160 DOI: 10.7717/peerj.8599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 01/19/2020] [Indexed: 11/20/2022] Open
Abstract
Background As a fundamental process internalizing molecules from the plasma membrane, endocytosis plays a crucial role in podocyte biology. Our previous study has identified that overexpression of Myole may enhance podocyte endocytosis. However, its potential mechanism has been not well understand. Thus, we aimed to analyze whether albumin endocytosis by mouse glomerular podocytes is dependent on Myo1e expression. Also, we aimed to elucidate whether the underlying mechanism is mediated by Dynamin. Methods Firstly, mouse podocyte cells (MPC5) were treated with different concentrations of FITC-bovine serum albumin (BSA). The fluorescence intensity and cell viability were detected by flow cytometry and MTT assays, respectively. Afterwards, the optimal concentration of FITC-BSA was determined. Secondly, MPC5 cells were treated with Myole overexpression or knockdown. Cell morphology was observed under microscope. Immunofluorescence assay was used to determine the expression of F-actin. The protein expression of nephrin and podocin was detected by western blot. Flow cytometry was used to detect MPC5 cell apoptosis with annexin V. Finally, MPC5 cells were treated with Myole overexpression and/or Dynasore (a GTPase inhibitor of Dynamin). The fluorescence intensity was detected using flow cytometry assay. Results MPC5 endocytosis BSA was elevated with a concentration-dependent manner. MTT results showed that MPC5 cell viability was inhibited with a concentration-dependent manner. Myo1e overexpression promoted podocyte endocytic FITC-BSA, which was contrary to its knockdown. Under microscope, after inhibition of Myo1e, podocyte foot process fusion was observed. Myo1e overexpression promoted the expression of cytoskeleton F-actin and podocyte-specific molecules (nephrin and podocin) in podocyte endocytic FITC-BSA. Furthermore, we found that Myo1e promoted the apoptosis of podocytes. Dynasore attenuated the increase in endocytosis of FITC-BSA induced by Myo1e overexpression, suggesting that podocytes might mediate albumin endocytosis via Myo1e-Dynamin-Albumin. Conclusion Our findings revealed that overexpression of Myo1e promotes albumin endocytosis in mouse glomerular podocyte endocytic albumin mediated by Dynamin.
Collapse
Affiliation(s)
- Huijun Shen
- Department of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Bao
- Department of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chunyue Feng
- Department of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Haidong Fu
- Department of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianhua Mao
- Department of Nephrology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
32
|
Barger SR, James ML, Pellenz CD, Krendel M, Sirotkin V. Human myosin 1e tail but not motor domain replaces fission yeast Myo1 domains to support myosin-I function during endocytosis. Exp Cell Res 2019; 384:111625. [PMID: 31542284 DOI: 10.1016/j.yexcr.2019.111625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 09/13/2019] [Accepted: 09/15/2019] [Indexed: 10/26/2022]
Abstract
In both unicellular and multicellular organisms, long-tailed class I myosins function in clathrin-mediated endocytosis. Myosin 1e (Myo1e) in vertebrates and Myo1 in fission yeast have similar domain organization, yet whether these proteins or their individual protein domains are functionally interchangeable remains unknown. In an effort to assess functional conservation of class I myosins, we tested whether human Myo1e could replace Myo1 in fission yeast Schizosaccharomyces pombe and found that it was unable to substitute for yeast Myo1. To determine if any individual protein domain is responsible for the inability of Myo1e to function in yeast, we created human-yeast myosin-I chimeras. By functionally testing these chimeric myosins in vivo, we concluded that the Myo1e motor domain is unable to function in yeast, even when combined with the yeast Myo1 tail and a full complement of yeast regulatory light chains. Conversely, the Myo1e tail, when attached to the yeast Myo1 motor domain, supports localization to endocytic actin patches and partially rescues the endocytosis defect in myo1Δ cells. Further dissection showed that both the TH1 and TH2-SH3 domains in the human Myo1e tail are required for localization and function of chimeric myosin-I at endocytic sites. Overall, this study provides insights into the role of individual myosin-I domains, expands the utility of fission yeast as a simple model system to study the effects of disease-associated MYO1E mutations, and supports a model of co-evolution between a myosin motor and its actin track.
Collapse
Affiliation(s)
- Sarah R Barger
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Michael L James
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Christopher D Pellenz
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Mira Krendel
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Vladimir Sirotkin
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
33
|
Roles of Myosin-Mediated Membrane Trafficking in TGF-β Signaling. Int J Mol Sci 2019; 20:ijms20163913. [PMID: 31408934 PMCID: PMC6719161 DOI: 10.3390/ijms20163913] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
Recent findings have revealed the role of membrane traffic in the signaling of transforming growth factor-β (TGF-β). These findings originate from the pivotal function of TGF-β in development, cell proliferation, tumor metastasis, and many other processes essential in malignancy. Actin and unconventional myosin have crucial roles in subcellular trafficking of receptors; research has also revealed a growing number of unconventional myosins that have crucial roles in TGF-β signaling. Unconventional myosins modulate the spatial organization of endocytic trafficking and tether membranes or transport them along the actin cytoskeletons. Current models do not fully explain how membrane traffic forms a bridge between TGF-β and the downstream effectors that produce its functional responsiveness, such as cell migration. In this review, we present a brief overview of the current knowledge of the TGF-β signaling pathway and the molecular components that comprise the core pathway as follows: ligands, receptors, and Smad mediators. Second, we highlight key role(s) of myosin motor-mediated protein trafficking and membrane domain segregation in the modulation of the TGF-β signaling pathway. Finally, we review future challenges and provide future prospects in this field.
Collapse
|
34
|
Cota Teixeira S, Silva Lopes D, Santos da Silva M, Cordero da Luz FA, Cirilo Gimenes SN, Borges BC, Alves da Silva A, Alves Martins F, Alves Dos Santos M, Teixeira TL, Oliveira RA, de Melo Rodrigues Ávila V, Barbosa Silva MJ, Elias MC, Martin R, Vieira da Silva C, Knölker HJ. Pentachloropseudilin Impairs Angiogenesis by Disrupting the Actin Cytoskeleton, Integrin Trafficking and the Cell Cycle. Chembiochem 2019; 20:2390-2401. [PMID: 31026110 DOI: 10.1002/cbic.201900203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Indexed: 12/21/2022]
Abstract
Class 1 myosins (Myo1s) were the first unconventional myosins identified and humans have eight known Myo1 isoforms. The Myo1 family is involved in the regulation of gene expression, cytoskeletal rearrangements, delivery of proteins to the cell surface, cell migration and spreading. Thus, the important role of Myo1s in different biological processes is evident. In this study, we have investigated the effects of pentachloropseudilin (PClP), a reversible and allosteric potent inhibitor of Myo1s, on angiogenesis. We demonstrated that treatment of cells with PClP promoted a decrease in the number of vessels. The observed inhibition of angiogenesis is likely to be related to the inhibition of cell proliferation, migration and adhesion, as well as to alteration of the actin cytoskeleton pattern, as shown on a PClP-treated HUVEC cell line. Moreover, we also demonstrated that PClP treatment partially prevented the delivery of integrins to the plasma membrane. Finally, we showed that PClP caused DNA strand breaks, which are probably repaired during the cell cycle arrest in the G1 phase. Taken together, our results suggest that Myo1s participate directly in the angiogenesis process.
Collapse
Affiliation(s)
- Samuel Cota Teixeira
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Daiana Silva Lopes
- Multidisciplinary Institute of Health, Anísio Teixeira Campus, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista, 45029-094, BA, Brazil
| | - Marcelo Santos da Silva
- Special Laboratory of Cell Cycle (LECC), Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, 05503-900, SP, Brazil.,The Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, Sir Graeme Davies Building, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| | - Felipe Andrés Cordero da Luz
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Sarah Natalie Cirilo Gimenes
- Imunopathology Laboratory, Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, 05503-900, SP, Brazil
| | - Bruna Cristina Borges
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Aline Alves da Silva
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Flávia Alves Martins
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Marlus Alves Dos Santos
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Thaise Lara Teixeira
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Ricardo A Oliveira
- Medical School, Federal University of Uberlândia, Av. Pará, Bloco 2u, 1720 - Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Veridiana de Melo Rodrigues Ávila
- Institute of Biotechnology, Federal University of Uberlândia, Av. Pará, 1720 - Bloco 2E - Sala(s) 246 - Campus Umuarama, Uberlândia, 38405-320, MG, Brazil
| | - Marcelo José Barbosa Silva
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Maria Carolina Elias
- Special Laboratory of Cell Cycle (LECC), Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, 05503-900, SP, Brazil
| | - René Martin
- Fakultät Chemie, Technische Universität Dresden, Bergstraße 66, 01069, Dresden, Germany
| | - Claudio Vieira da Silva
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Hans-Joachim Knölker
- Fakultät Chemie, Technische Universität Dresden, Bergstraße 66, 01069, Dresden, Germany
| |
Collapse
|
35
|
Manenschijn HE, Picco A, Mund M, Rivier-Cordey AS, Ries J, Kaksonen M. Type-I myosins promote actin polymerization to drive membrane bending in endocytosis. eLife 2019; 8:44215. [PMID: 31385806 PMCID: PMC6684269 DOI: 10.7554/elife.44215] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 07/23/2019] [Indexed: 12/24/2022] Open
Abstract
Clathrin-mediated endocytosis in budding yeast requires the formation of a dynamic actin network that produces the force to invaginate the plasma membrane against the intracellular turgor pressure. The type-I myosins Myo3 and Myo5 are important for endocytic membrane reshaping, but mechanistic details of their function remain scarce. Here, we studied the function of Myo3 and Myo5 during endocytosis using quantitative live-cell imaging and genetic perturbations. We show that the type-I myosins promote, in a dose-dependent way, the growth and expansion of the actin network, which controls the speed of membrane and coat internalization. We found that this myosin-activity is independent of the actin nucleation promoting activity of myosins, and cannot be compensated for by increasing actin nucleation. Our results suggest a new mechanism for type-I myosins to produce force by promoting actin filament polymerization.
Collapse
Affiliation(s)
- Hetty E Manenschijn
- Department of Biochemistry, University of Geneva, Geneva, Switzerland.,Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Andrea Picco
- Department of Biochemistry, University of Geneva, Geneva, Switzerland.,NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Markus Mund
- Department of Biochemistry, University of Geneva, Geneva, Switzerland.,Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | | | - Jonas Ries
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Marko Kaksonen
- Department of Biochemistry, University of Geneva, Geneva, Switzerland.,NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| |
Collapse
|
36
|
Kumari A, Pineau J, Sáez PJ, Maurin M, Lankar D, San Roman M, Hennig K, Boura VF, Voituriez R, Karlsson MCI, Balland M, Lennon Dumenil AM, Pierobon P. Actomyosin-driven force patterning controls endocytosis at the immune synapse. Nat Commun 2019; 10:2870. [PMID: 31253773 PMCID: PMC6599028 DOI: 10.1038/s41467-019-10751-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/30/2019] [Indexed: 11/30/2022] Open
Abstract
An important channel of cell-to-cell communication is direct contact. The immune synapse is a paradigmatic example of such type of interaction: it forms upon engagement of antigen receptors in lymphocytes by antigen-presenting cells and allows the local exchange of molecules and information. Although mechanics has been shown to play an important role in this process, how forces organize and impact on synapse function is unknown. We find that mechanical forces are spatio-temporally patterned at the immune synapse: global pulsatile myosin II-driven tangential forces are observed at the synapse periphery while localised forces generated by invadosome-like F-actin protrusions are detected at its centre. Noticeably, we observe that these force-producing actin protrusions constitute the main site of antigen extraction and endocytosis and require myosin II contractility to form. The interplay between global and local forces dictated by the organization of the actomyosin cytoskeleton therefore controls endocytosis at the immune synapse. The immune synapse promotes cellular information exchange but the role of biophysical forces in synapse function is unclear. Here, the authors show that B cells exert two types of forces, a centripetal myosin II-driven force and a central actin protrusive force at the site of antigen extraction.
Collapse
Affiliation(s)
- Anita Kumari
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France.,Université Paris Descartes, Paris, 75006, France
| | - Judith Pineau
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France.,Université Paris Descartes, Paris, 75006, France
| | - Pablo J Sáez
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France
| | - Mathieu Maurin
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France
| | - Danielle Lankar
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France
| | - Mabel San Roman
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France
| | - Katharina Hennig
- Laboratoire Interdisciplinaire de Physique, Université Joseph Fourier (Grenoble 1), 38402, Saint, Martin d'Hères Cedex 9, France
| | - Vanessa F Boura
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Raphael Voituriez
- Laboratoire de Physique Théorique de la Matière Condensée, UMR 7600 CNRS /UPMC and Laboratoire Jean Perrin, UMR 8237 CNRS /UPMC, 4 Place Jussieu, 75255, Paris, Cedex 05, France
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Martial Balland
- Laboratoire Interdisciplinaire de Physique, Université Joseph Fourier (Grenoble 1), 38402, Saint, Martin d'Hères Cedex 9, France
| | - Ana-Maria Lennon Dumenil
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France.
| | - Paolo Pierobon
- Institut Curie, PSL Research University, INSERM U932, 26 rue d'Ulm, 75248, Paris, Cedex 05, France.
| |
Collapse
|
37
|
Girón-Pérez DA, Piedra-Quintero ZL, Santos-Argumedo L. Class I myosins: Highly versatile proteins with specific functions in the immune system. J Leukoc Biol 2019; 105:973-981. [PMID: 30821871 DOI: 10.1002/jlb.1mr0918-350rrr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/04/2019] [Accepted: 02/06/2019] [Indexed: 12/20/2022] Open
Abstract
Connections established between cytoskeleton and plasma membrane are essential in cellular processes such as cell migration, vesicular trafficking, and cytokinesis. Class I myosins are motor proteins linking the actin-cytoskeleton with membrane phospholipids. Previous studies have implicated these molecules in cell functions including endocytosis, exocytosis, release of extracellular vesicles and the regulation of cell shape and membrane elasticity. In immune cells, those proteins also are involved in the formation and maintenance of immunological synapse-related signaling. Thus, these proteins are master regulators of actin cytoskeleton dynamics in different scenarios. Although the localization of class I myosins has been described in vertebrates, their functions, regulation, and mechanical properties are not very well understood. In this review, we focused on and summarized the current understanding of class I myosins in vertebrates with particular emphasis in leukocytes.
Collapse
Affiliation(s)
- Daniel Alberto Girón-Pérez
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | - Zayda Lizbeth Piedra-Quintero
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | - Leopoldo Santos-Argumedo
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
38
|
Sakai R, Fukuda R, Unida S, Aki M, Ono Y, Endo A, Kusumi S, Koga D, Fukushima T, Komada M, Okiyoneda T. The integral function of the endocytic recycling compartment is regulated by RFFL-mediated ubiquitylation of Rab11 effectors. J Cell Sci 2019; 132:jcs.228007. [PMID: 30659120 DOI: 10.1242/jcs.228007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 01/03/2019] [Indexed: 12/11/2022] Open
Abstract
Endocytic trafficking is regulated by ubiquitylation (also known as ubiquitination) of cargoes and endocytic machineries. The role of ubiquitylation in lysosomal delivery has been well documented, but its role in the recycling pathway is largely unknown. Here, we report that the ubiquitin (Ub) ligase RFFL regulates ubiquitylation of endocytic recycling regulators. An RFFL dominant-negative (DN) mutant induced clustering of endocytic recycling compartments (ERCs) and delayed endocytic cargo recycling without affecting lysosomal traffic. A BioID RFFL interactome analysis revealed that RFFL interacts with the Rab11 effectors EHD1, MICALL1 and class I Rab11-FIPs. The RFFL DN mutant strongly captured these Rab11 effectors and inhibited their ubiquitylation. The prolonged interaction of RFFL with Rab11 effectors was sufficient to induce the clustered ERC phenotype and to delay cargo recycling. RFFL directly ubiquitylates these Rab11 effectors in vitro, but RFFL knockout (KO) only reduced the ubiquitylation of Rab11-FIP1. RFFL KO had a minimal effect on the ubiquitylation of EHD1, MICALL1, and Rab11-FIP2, and failed to delay transferrin recycling. These results suggest that multiple Ub ligases including RFFL regulate the ubiquitylation of Rab11 effectors, determining the integral function of the ERC.
Collapse
Affiliation(s)
- Ryohei Sakai
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Ryosuke Fukuda
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Shin Unida
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Misaki Aki
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Yuji Ono
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Akinori Endo
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Satoshi Kusumi
- Division of Morphological Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Daisuke Koga
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical University, Asahikawa 078-8510, Hokkaido, Japan
| | - Toshiaki Fukushima
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Masayuki Komada
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Tsukasa Okiyoneda
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Hyogo 669-1337, Japan
| |
Collapse
|
39
|
Pedersen RTA, Drubin DG. Type I myosins anchor actin assembly to the plasma membrane during clathrin-mediated endocytosis. J Cell Biol 2019; 218:1138-1147. [PMID: 30659101 PMCID: PMC6446854 DOI: 10.1083/jcb.201810005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/14/2018] [Accepted: 01/04/2019] [Indexed: 12/26/2022] Open
Abstract
Actin assembly and type I myosins are both required for clathrin-mediated endocytosis. Here Pedersen and Drubin show that type I myosins anchor actin assembly factors to the plasma membrane at sites of clathrin-mediated endocytosis, facilitating force generation by actin assembly. The actin cytoskeleton generates forces on membranes for a wide range of cellular and subcellular morphogenic events, from cell migration to cytokinesis and membrane trafficking. For each of these processes, filamentous actin (F-actin) interacts with membranes and exerts force through its assembly, its associated myosin motors, or both. These two modes of force generation are well studied in isolation, but how they are coordinated in cells is mysterious. During clathrin-mediated endocytosis, F-actin assembly initiated by the Arp2/3 complex and several proteins that compose the WASP/myosin complex generates the force necessary to deform the plasma membrane into a pit. Here we present evidence that type I myosin is the key membrane anchor for endocytic actin assembly factors in budding yeast. By mooring actin assembly factors to the plasma membrane, this myosin organizes endocytic actin networks and couples actin-generated forces to the plasma membrane to drive invagination and scission. Through this unexpected mechanism, myosin facilitates force generation independent of its motor activity.
Collapse
Affiliation(s)
- Ross T A Pedersen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
| | - David G Drubin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
| |
Collapse
|
40
|
Zhang Y, Cao F, Zhou Y, Feng Z, Sit B, Krendel M, Yu CH. Tail domains of myosin-1e regulate phosphatidylinositol signaling and F-actin polymerization at the ventral layer of podosomes. Mol Biol Cell 2019; 30:622-635. [PMID: 30601698 PMCID: PMC6589698 DOI: 10.1091/mbc.e18-06-0398] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
During podosome formation, distinct phosphatidylinositol 3,4,5-trisphosphate lipid (PI(3,4,5)P3) production and F-actin polymerization take place at integrin-mediated adhesions. Membrane-associated actin regulation factors, such as myosin-1, serve as key molecules to link phosphatidylinositol signals to podosome assembly. Here, we report that long-tailed myosin-1e (Myo1e) is enriched at the ventral layer of the podosome core in a PI(3,4,5)P3-dependent manner. The combination of TH1 and TH2 (TH12) of Myo1e tail domains contains the essential motif for PI(3,4,5)P3-dependent membrane association and ventral localization at the podosome. TH12 KR2A (K772A and R782A) becomes dissociated from the plasma membrane. While F-actin polymerizations are initialized from the ventral layer of the podosome, TH12 precedes the recruitment of N-WASP and Arp2/3 in the initial phase of podosome formation. Overexpression of TH12, not TH12 KR2A, impedes PI(3,4,5)P3 signaling, restrains F-actin polymerization, and inhibits podosome formation. TH12 also suppresses gelatin degradation and migration speed of invadopodia-forming A375 melanoma cells. Thus, TH12 domain of Myo1e serves as a regulatory component to connect phosphatidylinositol signaling to F-actin polymerization at the podosome.
Collapse
Affiliation(s)
- Yage Zhang
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Fakun Cao
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Yuhuan Zhou
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Zhen Feng
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Brian Sit
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong.,Randall Division of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London WC2R 2LS, United Kingdom
| | - Mira Krendel
- SUNY Upstate Medical University, Syracuse, NY 13210
| | - Cheng-Han Yu
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| |
Collapse
|
41
|
Schöneberg J, Dambournet D, Liu TL, Forster R, Hockemeyer D, Betzig E, Drubin DG. 4D cell biology: big data image analytics and lattice light-sheet imaging reveal dynamics of clathrin-mediated endocytosis in stem cell-derived intestinal organoids. Mol Biol Cell 2018; 29:2959-2968. [PMID: 30188768 PMCID: PMC6329908 DOI: 10.1091/mbc.e18-06-0375] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
New methods in stem cell 3D organoid tissue culture, advanced imaging, and big data image analytics now allow tissue-scale 4D cell biology, but currently available analytical pipelines are inadequate for handing and analyzing the resulting gigabytes and terabytes of high-content imaging data. We expressed fluorescent protein fusions of clathrin and dynamin2 at endogenous levels in genome-edited human embryonic stem cells, which were differentiated into hESC-derived intestinal epithelial organoids. Lattice light-sheet imaging with adaptive optics (AO-LLSM) allowed us to image large volumes of these organoids (70 × 60 × 40 µm xyz) at 5.7 s/frame. We developed an open-source data analysis package termed pyLattice to process the resulting large (∼60 Gb) movie data sets and to track clathrin-mediated endocytosis (CME) events. CME tracks could be recorded from ∼35 cells at a time, resulting in ∼4000 processed tracks per movie. On the basis of their localization in the organoid, we classified CME tracks into apical, lateral, and basal events and found that CME dynamics is similar for all three classes, despite reported differences in membrane tension. pyLattice coupled with AO-LLSM makes possible quantitative high temporal and spatial resolution analysis of subcellular events within tissues.
Collapse
Affiliation(s)
| | - Daphné Dambournet
- Department of Molecular and Cell Biology, Berkeley, Berkeley, CA 94720
| | - Tsung-Li Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Ryan Forster
- Department of Molecular and Cell Biology, Berkeley, Berkeley, CA 94720
| | - Dirk Hockemeyer
- Department of Molecular and Cell Biology, Berkeley, Berkeley, CA 94720
| | - Eric Betzig
- Department of Molecular and Cell Biology, Berkeley, Berkeley, CA 94720.,Department of Physics, University of California, Berkeley, Berkeley, CA 94720.,Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| | - David G Drubin
- Department of Molecular and Cell Biology, Berkeley, Berkeley, CA 94720
| |
Collapse
|
42
|
Dambournet D, Sochacki KA, Cheng AT, Akamatsu M, Taraska JW, Hockemeyer D, Drubin DG. Genome-edited human stem cells expressing fluorescently labeled endocytic markers allow quantitative analysis of clathrin-mediated endocytosis during differentiation. J Cell Biol 2018; 217:3301-3311. [PMID: 29980624 PMCID: PMC6123002 DOI: 10.1083/jcb.201710084] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/05/2017] [Accepted: 06/04/2018] [Indexed: 12/11/2022] Open
Abstract
We developed a general approach for investigation of how cellular processes become adapted for specific cell types during differentiation. Previous studies reported substantial differences in the morphology and dynamics of clathrin-mediated endocytosis (CME) sites. However, associating specific CME properties with distinct differentiated cell types and determining how these properties are developmentally specified during differentiation have been elusive. Using genome-edited human embryonic stem cells, and isogenic fibroblasts and neuronal progenitor cells derived from them, we established by live-cell imaging and platinum replica transmission electron microscopy that CME site dynamics and ultrastructure on the plasma membrane are precisely reprogrammed during differentiation. Expression levels for the endocytic adaptor protein AP2μ2 were found to underlie dramatic changes in CME dynamics and structure. Additionally, CME dependency on actin assembly and phosphoinositide-3 kinase activity are distinct for each cell type. Collectively, our results demonstrate that key CME properties are reprogrammed during differentiation at least in part through AP2μ2 expression regulation.
Collapse
Affiliation(s)
- Daphné Dambournet
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
| | - Kem A Sochacki
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | - Matthew Akamatsu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
| | - Justin W Taraska
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Dirk Hockemeyer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
| | - David G Drubin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
| |
Collapse
|
43
|
Rottner K, Faix J, Bogdan S, Linder S, Kerkhoff E. Actin assembly mechanisms at a glance. J Cell Sci 2018; 130:3427-3435. [PMID: 29032357 DOI: 10.1242/jcs.206433] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The actin cytoskeleton and associated motor proteins provide the driving forces for establishing the astonishing morphological diversity and dynamics of mammalian cells. Aside from functions in protruding and contracting cell membranes for motility, differentiation or cell division, the actin cytoskeleton provides forces to shape and move intracellular membranes of organelles and vesicles. To establish the many different actin assembly functions required in time and space, actin nucleators are targeted to specific subcellular compartments, thereby restricting the generation of specific actin filament structures to those sites. Recent research has revealed that targeting and activation of actin filament nucleators, elongators and myosin motors are tightly coordinated by conserved protein complexes to orchestrate force generation. In this Cell Science at a Glance article and the accompanying poster, we summarize and discuss the current knowledge on the corresponding protein complexes and their modes of action in actin nucleation, elongation and force generation.
Collapse
Affiliation(s)
- Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Sven Bogdan
- Institute for Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps-University of Marburg, 35032 Marburg, Germany
| | - Stefan Linder
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany
| | - Eugen Kerkhoff
- Department of Neurology, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
44
|
Ripoll L, Heiligenstein X, Hurbain I, Domingues L, Figon F, Petersen KJ, Dennis MK, Houdusse A, Marks MS, Raposo G, Delevoye C. Myosin VI and branched actin filaments mediate membrane constriction and fission of melanosomal tubule carriers. J Cell Biol 2018; 217:2709-2726. [PMID: 29875258 PMCID: PMC6080934 DOI: 10.1083/jcb.201709055] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 03/16/2018] [Accepted: 05/09/2018] [Indexed: 01/19/2023] Open
Abstract
Vesicular and tubular transport intermediates regulate organellar cargo dynamics. Transport carrier release involves local and profound membrane remodeling before fission. Pinching the neck of a budding tubule or vesicle requires mechanical forces, likely exerted by the action of molecular motors on the cytoskeleton. Here, we show that myosin VI, together with branched actin filaments, constricts the membrane of tubular carriers that are then released from melanosomes, the pigment containing lysosome-related organelles of melanocytes. By combining superresolution fluorescence microscopy, correlative light and electron microscopy, and biochemical analyses, we find that myosin VI motor activity mediates severing by constricting the neck of the tubule at specific melanosomal subdomains. Pinching of the tubules involves the cooperation of the myosin adaptor optineurin and the activity of actin nucleation machineries, including the WASH and Arp2/3 complexes. The fission and release of these tubules allows for the export of components from melanosomes, such as the SNARE VAMP7, and promotes melanosome maturation and transfer to keratinocytes. Our data reveal a new myosin VI- and actin-dependent membrane fission mechanism required for organelle function.
Collapse
Affiliation(s)
- Léa Ripoll
- Structure and Membrane Compartments, Institut Curie, Paris Sciences & Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France
| | - Xavier Heiligenstein
- Structure and Membrane Compartments, Institut Curie, Paris Sciences & Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France
| | - Ilse Hurbain
- Structure and Membrane Compartments, Institut Curie, Paris Sciences & Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France.,Cell and Tissue Imaging Facility, Institut Curie, Paris Sciences & Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France
| | - Lia Domingues
- Structure and Membrane Compartments, Institut Curie, Paris Sciences & Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France
| | - Florent Figon
- Structure and Membrane Compartments, Institut Curie, Paris Sciences & Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France.,Master BioSciences, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Karl J Petersen
- Structural Motility, Institut Curie, Paris Sciences & Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France
| | - Megan K Dennis
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA.,Departments of Pathology and Laboratory Medicine and Physiology, University of Pennsylvania, Philadelphia, PA
| | - Anne Houdusse
- Structural Motility, Institut Curie, Paris Sciences & Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France
| | - Michael S Marks
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA.,Departments of Pathology and Laboratory Medicine and Physiology, University of Pennsylvania, Philadelphia, PA
| | - Graça Raposo
- Structure and Membrane Compartments, Institut Curie, Paris Sciences & Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France.,Cell and Tissue Imaging Facility, Institut Curie, Paris Sciences & Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France
| | - Cédric Delevoye
- Structure and Membrane Compartments, Institut Curie, Paris Sciences & Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France .,Cell and Tissue Imaging Facility, Institut Curie, Paris Sciences & Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France
| |
Collapse
|
45
|
Yoshida A, Sakai N, Uekusa Y, Imaoka Y, Itagaki Y, Suzuki Y, Yoshimura SH. Morphological changes of plasma membrane and protein assembly during clathrin-mediated endocytosis. PLoS Biol 2018; 16:e2004786. [PMID: 29723197 PMCID: PMC5953504 DOI: 10.1371/journal.pbio.2004786] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 05/15/2018] [Accepted: 03/29/2018] [Indexed: 12/21/2022] Open
Abstract
Clathrin-mediated endocytosis (CME) proceeds through a series of morphological changes of the plasma membrane induced by a number of protein components. Although the spatiotemporal assembly of these proteins has been elucidated by fluorescence-based techniques, the protein-induced morphological changes of the plasma membrane have not been fully clarified in living cells. Here, we visualize membrane morphology together with protein localizations during CME by utilizing high-speed atomic force microscopy (HS-AFM) combined with a confocal laser scanning unit. The plasma membrane starts to invaginate approximately 30 s after clathrin starts to assemble, and the aperture diameter increases as clathrin accumulates. Actin rapidly accumulates around the pit and induces a small membrane swelling, which, within 30 s, rapidly covers the pit irreversibly. Inhibition of actin turnover abolishes the swelling and induces a reversible open–close motion of the pit, indicating that actin dynamics are necessary for efficient and irreversible pit closure at the end of CME. Cells communicate with their environments via the plasma membrane and various membrane proteins. Clathrin-mediated endocytosis (CME) plays a central role in such communication and proceeds with a series of multiprotein assembly, deformation of the plasma membrane, and production of a membrane vesicle that delivers extracellular signaling molecules into the cytoplasm. In this study, we utilized our home-built correlative imaging system comprising high-speed atomic force microscopy (HS-AFM) and confocal fluorescence microscopy to simultaneously image morphological changes of the plasma membrane and protein localization during CME in a living cell. The results revealed a tight correlation between the size of the pit and the amount of clathrin assembled. Actin dynamics play multiple roles in the assembly, maturation, and closing phases of the process, and affects membrane morphology, suggesting a close relationship between endocytosis and dynamic events at the cell cortex. Knock down of dynamin also affected the closing motion of the pit and showed functional correlation with actin.
Collapse
Affiliation(s)
- Aiko Yoshida
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | | | | | - Yuka Imaoka
- R&D Group, Olympus Corporation, Hachioji, Japan
| | | | - Yuki Suzuki
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | | |
Collapse
|
46
|
Capping protein-controlled actin polymerization shapes lipid membranes. Nat Commun 2018; 9:1630. [PMID: 29691404 PMCID: PMC5915599 DOI: 10.1038/s41467-018-03918-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 03/20/2018] [Indexed: 11/08/2022] Open
Abstract
Arp2/3 complex-mediated actin assembly at cell membranes drives the formation of protrusions or endocytic vesicles. To identify the mechanism by which different membrane deformations can be achieved, we reconstitute the basic membrane deformation modes of inward and outward bending in a confined geometry by encapsulating a minimal set of cytoskeletal proteins into giant unilamellar vesicles. Formation of membrane protrusions is favoured at low capping protein (CP) concentrations, whereas the formation of negatively bent domains is promoted at high CP concentrations. Addition of non-muscle myosin II results in full fission events in the vesicle system. The different deformation modes are rationalized by simulations of the underlying transient nature of the reaction kinetics. The relevance of the regulatory mechanism is supported by CP overexpression in mouse melanoma B16-F1 cells and therefore demonstrates the importance of the quantitative understanding of microscopic kinetic balances to address the diverse functionality of the cytoskeleton. Cell membrane protrusions and invaginations are both driven by actin assembly but the mechanism leading to different membrane shapes is unknown. Using a minimal system and modelling the authors reconstitute the deformation modes and identify capping protein as a regulator of both deformation types.
Collapse
|
47
|
O'Loughlin T, Masters TA, Buss F. The MYO6 interactome reveals adaptor complexes coordinating early endosome and cytoskeletal dynamics. EMBO Rep 2018; 19:e44884. [PMID: 29467281 PMCID: PMC5891429 DOI: 10.15252/embr.201744884] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 01/17/2018] [Accepted: 01/30/2018] [Indexed: 12/15/2022] Open
Abstract
The intracellular functions of myosin motors requires a number of adaptor molecules, which control cargo attachment, but also fine-tune motor activity in time and space. These motor-adaptor-cargo interactions are often weak, transient or highly regulated. To overcome these problems, we use a proximity labelling-based proteomics strategy to map the interactome of the unique minus end-directed actin motor MYO6. Detailed biochemical and functional analysis identified several distinct MYO6-adaptor modules including two complexes containing RhoGEFs: the LIFT (LARG-Induced F-actin for Tethering) complex that controls endosome positioning and motility through RHO-driven actin polymerisation; and the DISP (DOCK7-Induced Septin disPlacement) complex, a novel regulator of the septin cytoskeleton. These complexes emphasise the role of MYO6 in coordinating endosome dynamics and cytoskeletal architecture. This study provides the first in vivo interactome of a myosin motor protein and highlights the power of this approach in uncovering dynamic and functionally diverse myosin motor complexes.
Collapse
Affiliation(s)
- Thomas O'Loughlin
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Cambridge, UK
| | - Thomas A Masters
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Cambridge, UK
| | - Folma Buss
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Cambridge, UK
| |
Collapse
|
48
|
Lyraki R, Lokaj M, Soares DC, Little A, Vermeren M, Marsh JA, Wittinghofer A, Hurd T. Characterization of a novel RP2-OSTF1 interaction and its implication for actin remodelling. J Cell Sci 2018; 131:jcs.211748. [PMID: 29361551 DOI: 10.1242/jcs.211748] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/21/2017] [Indexed: 11/20/2022] Open
Abstract
Retinitis pigmentosa 2 (RP2) is the causative gene for a form of X-linked retinal degeneration. RP2 was previously shown to have GTPase-activating protein (GAP) activity towards the small GTPase ARL3 via its N-terminus, but the function of the C-terminus remains elusive. Here, we report a novel interaction between RP2 and osteoclast-stimulating factor 1 (OSTF1), an intracellular protein that indirectly enhances osteoclast formation and activity and is a negative regulator of cell motility. Moreover, this interaction is abolished by a human pathogenic mutation in RP2. We utilized a structure-based approach to pinpoint the binding interface to a strictly conserved cluster of residues on the surface of RP2 that spans both the C- and N-terminal domains of the protein, and which is structurally distinct from the ARL3-binding site. In addition, we show that RP2 is a positive regulator of cell motility in vitro, recruiting OSTF1 to the cell membrane and preventing its interaction with the migration regulator Myo1E.
Collapse
Affiliation(s)
- Rodanthi Lyraki
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Mandy Lokaj
- Structural Biology Group, Max-Planck Institut für Molekulare Physiologie, Abteilung Strukturelle Biologie, Otto-Hahn-Str. 11, 44227 Dortmund, Germany
| | - Dinesh C Soares
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Abigail Little
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Matthieu Vermeren
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.,MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Joseph A Marsh
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Alfred Wittinghofer
- Structural Biology Group, Max-Planck Institut für Molekulare Physiologie, Abteilung Strukturelle Biologie, Otto-Hahn-Str. 11, 44227 Dortmund, Germany
| | - Toby Hurd
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| |
Collapse
|
49
|
|
50
|
Makaraci P, Kim K. trans-Golgi network-bound cargo traffic. Eur J Cell Biol 2018; 97:137-149. [PMID: 29398202 DOI: 10.1016/j.ejcb.2018.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/15/2017] [Accepted: 01/16/2018] [Indexed: 12/19/2022] Open
Abstract
Cargo following the retrograde trafficking are sorted at endosomes to be targeted the trans-Golgi network (TGN), a central receiving organelle. Though molecular requirements and their interaction networks have been somewhat established, the complete understanding of the intricate nature of their action mechanisms in every step of the retrograde traffic pathway remains unachieved. This review focuses on elucidating known functions of key regulators, including scission factors at the endosome and tethering/fusion mediators at the receiving dock, TGN, as well as a diverse range of cargo.
Collapse
Affiliation(s)
- Pelin Makaraci
- Department of Biology, Missouri State University, 901 S National Ave., Springfield, MO 65807, USA
| | - Kyoungtae Kim
- Department of Biology, Missouri State University, 901 S National Ave., Springfield, MO 65807, USA.
| |
Collapse
|