1
|
Hart RG, Kota D, Li F, Zhang M, Ramallo D, Price AJ, Otterpohl KL, Smith SJ, Dunn AR, Huising MO, Liu J, Chandrasekar I. Myosin II tension sensors visualize force generation within the actin cytoskeleton in living cells. J Cell Sci 2024; 137:jcs262281. [PMID: 39369303 DOI: 10.1242/jcs.262281] [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: 05/20/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
Nonmuscle myosin II (NMII) generates cytoskeletal forces that drive cell division, embryogenesis, muscle contraction and many other cellular functions. However, at present there is no method that can directly measure the forces generated by myosins in living cells. Here, we describe a Förster resonance energy transfer (FRET)-based tension sensor that can detect myosin-associated force along the filamentous actin network. Fluorescence lifetime imaging microscopy (FLIM)-FRET measurements indicate that the forces generated by NMII isoform B (NMIIB) exhibit significant spatial and temporal heterogeneity as a function of donor lifetime and fluorophore energy exchange. These measurements provide a proxy for inferred forces that vary widely along the actin cytoskeleton. This initial report highlights the potential utility of myosin-based tension sensors in elucidating the roles of cytoskeletal contractility in a wide variety of contexts.
Collapse
Affiliation(s)
- Ryan G Hart
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA
| | - Divya Kota
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, SD 57701, USA
| | - Fangjia Li
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Mengdi Zhang
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Diego Ramallo
- Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Andrew J Price
- Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Karla L Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Steve J Smith
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, SD 57701, USA
| | - Alexander R Dunn
- Department of Chemical Biology, Stanford University, Stanford, CA 94305, USA
| | - Mark O Huising
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA
- Department of Physiology and Membrane Biology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Jing Liu
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Department of Physics and Astronomy, Purdue University, West Lafayette, IN 46907
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
2
|
Pathak A, Willis KG, Bankaitis VA, McDermott MI. Mammalian START-like phosphatidylinositol transfer proteins - Physiological perspectives and roles in cancer biology. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159529. [PMID: 38945251 DOI: 10.1016/j.bbalip.2024.159529] [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: 03/29/2024] [Revised: 06/09/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
PtdIns and its phosphorylated derivatives, the phosphoinositides, are the biochemical components of a major pathway of intracellular signaling in all eukaryotic cells. These lipids are few in terms of cohort of unique positional isomers, and are quantitatively minor species of the bulk cellular lipidome. Nevertheless, phosphoinositides regulate an impressively diverse set of biological processes. It is from that perspective that perturbations in phosphoinositide-dependent signaling pathways are increasingly being recognized as causal foundations of many human diseases - including cancer. Although phosphatidylinositol transfer proteins (PITPs) are not enzymes, these proteins are physiologically significant regulators of phosphoinositide signaling. As such, PITPs are conserved throughout the eukaryotic kingdom. Their biological importance notwithstanding, PITPs remain understudied. Herein, we review current information regarding PITP biology primarily focusing on how derangements in PITP function disrupt key signaling/developmental pathways and are associated with a growing list of pathologies in mammals.
Collapse
Affiliation(s)
- Adrija Pathak
- Department of Cell Biology and Genetics, Texas A&M Health Science Center, College Station, Texas, 77843, USA; Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843, USA
| | - Katelyn G Willis
- Department of Cell Biology and Genetics, Texas A&M Health Science Center, College Station, Texas, 77843, USA
| | - Vytas A Bankaitis
- Department of Cell Biology and Genetics, Texas A&M Health Science Center, College Station, Texas, 77843, USA; Department of Chemistry, Texas A&M University, College Station, Texas 77843 USA
| | - Mark I McDermott
- Department of Cell Biology and Genetics, Texas A&M Health Science Center, College Station, Texas, 77843, USA.
| |
Collapse
|
3
|
Consalvo KM, Rijal R, Beruvides SL, Mitchell R, Beauchemin K, Collins D, Scoggin J, Scott J, Gomer RH. PTEN and the PTEN-like phosphatase CnrN have both distinct and overlapping roles in a Dictyostelium chemorepulsion pathway. J Cell Sci 2024; 137:jcs262054. [PMID: 38940195 PMCID: PMC11317092 DOI: 10.1242/jcs.262054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
Little is known about eukaryotic chemorepulsion. The enzymes phosphatase and tensin homolog (PTEN) and CnrN dephosphorylate phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3] to phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. Dictyostelium discoideum cells require both PTEN and CnrN to induce chemorepulsion of cells away from the secreted chemorepellent protein AprA. How D. discoideum cells utilize two proteins with redundant phosphatase activities in response to AprA is unclear. Here, we show that D. discoideum cells require both PTEN and CnrN to locally inhibit Ras activation, decrease basal levels of PI(3,4,5)P3 and increase basal numbers of macropinosomes, and AprA prevents this increase. AprA requires both PTEN and CnrN to increase PI(4,5)P2 levels, decrease PI(3,4,5)P3 levels, inhibit proliferation, decrease myosin II phosphorylation and increase filopod sizes. PTEN, but not CnrN, decreases basal levels of PI(4,5)P2, and AprA requires PTEN, but not CnrN, to induce cell roundness. Together, our results suggest that CnrN and PTEN play unique roles in AprA-induced chemorepulsion.
Collapse
Affiliation(s)
- Kristen M. Consalvo
- Department of Biology, Texas A&M University, College Station, TX 77843-3474, USA
| | - Ramesh Rijal
- Department of Biology, Texas A&M University, College Station, TX 77843-3474, USA
| | - Steven L. Beruvides
- Department of Biology, Texas A&M University, College Station, TX 77843-3474, USA
| | - Ryan Mitchell
- Department of Biology, Texas A&M University, College Station, TX 77843-3474, USA
| | - Karissa Beauchemin
- Department of Biology, Texas A&M University, College Station, TX 77843-3474, USA
| | - Danni Collins
- Department of Biology, Texas A&M University, College Station, TX 77843-3474, USA
| | - Jack Scoggin
- Department of Biology, Texas A&M University, College Station, TX 77843-3474, USA
| | - Jerome Scott
- Department of Biology, Texas A&M University, College Station, TX 77843-3474, USA
| | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, TX 77843-3474, USA
| |
Collapse
|
4
|
Li K, Li T, Niu Y, Gao Y, Shi Y, He Y, Zhang X, Wang Y, Cao J, Hu X, Chen M, Shi R. Decreased NMIIA heavy chain phosphorylation at S1943 promotes mitoxantrone resistance by upregulating BCRP and N-cadherin expression in breast cancer cells. Biochem Cell Biol 2024; 102:213-225. [PMID: 38190650 DOI: 10.1139/bcb-2023-0232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024] Open
Abstract
Mitoxantrone (MX) is an effective treatment for breast cancer; however, high efflux of MX that is accomplished by breast cancer resistance protein (BCRP) leads to acquired multidrug resistance (MDR), reducing MX's therapeutic efficacy in breast cancer. Non-muscle myosin IIA (NMIIA) and its heavy phosphorylation at S1943 have been revealed to play key roles in tumor metastasis and progression, including in breast cancer; however, their molecular function in BCRP-mediated MDR in breast cancer remains unknown. In this study, we revealed that the expression of NMIIA heavy chain phosphorylation at S1943 was downregulated in BCRP-overexpressing breast cancer MCF-7/MX cells, and stable expression of NMIIA-S1943A mutant increased BCRP expression and promoted the resistance of MCF-7/MX cells to MX. Meanwhile, NMIIA S1943 phosphorylation induced by epidermal growth factor (EGF) was accompanied by the downregulation of BCRP in MCF-7/MX cells. Furthermore, stable expression of NMIIA-S1943A in MCF-7/MX cells resulted in upregulation of N-cadherin and the accumulation of β-catenin on the cell surface, which inhibited the nucleus translocation of β-catenin and Wnt/β-catenin-based proliferative signaling. EGF stimulation of MCF-7/MX cells showed the downregulation of N-cadherin and β-catenin. Our results suggest that decreased NMIIA heavy phosphorylation at S1943 increases BCRP expression and promotes MX resistance in breast cancer cells via upregulating N-cadherin expression.
Collapse
Affiliation(s)
- Kemin Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Tian Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Yanan Niu
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Yu Gao
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Yifan Shi
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Yifan He
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Xuanping Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Yan Wang
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Jing Cao
- Department of Critical Care Medicine, the First Hospital of Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Xiaoling Hu
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Min Chen
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| | - Ruizan Shi
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, People's Republic of China
| |
Collapse
|
5
|
Assadsangabi A, Evans CA, Corfe BM, Lobo AJ. Exploring Predictive Biomarkers of Relapse in Ulcerative Colitis: A Proteomics Approach. Inflamm Bowel Dis 2024; 30:808-819. [PMID: 37889841 DOI: 10.1093/ibd/izad241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Indexed: 10/29/2023]
Abstract
INTRODUCTION AND AIMS Risk stratification of subjects with a history of inflammatory bowel disease (IBD) into those likely to relapse and those who will remain quiescent continues to be a significant challenge. The aim of this study was to investigate whether certain proteomic signature profiles or biomarkers during remission are associated with future disease relapse in patients with ulcerative colitis (UC). METHODS Endoscopic rectal samples from patients with UC in clinical, endoscopic, and histological remission at index endoscopy were collected, as well as samplers from normal control individuals. The patients were stratified to early relapsers (ERs) if they developed clinical signs of UC flare within 6 months of index endoscopy or nonrelapsers (NRs) if there was no relapse after 36 months of follow-up. The pooled rectal samples from ERs, NRs, and control individuals were subjected to nano-liquid chromatography and tandem mass spectrometry as per standard iTRAQ (isobaric tags for relative and absolute quantitation) workflow methodology. Selected proteomics-yielded candidates were subjected to orthogonal validation via immunoblotting, in a biomarker discovery exercise. RESULTS Sixty-one patients were included, of whom 8 had clinical relapse within 6 months from the index endoscopy, and 43 patients had no clinical symptoms of relapse within the 36-month follow-up period. Ten patients who had clinical signs of relapse between 6 and 36 months were excluded. Seventeen control individuals were also included. Soluble proteomics analyses between ERs, NRs, and control individuals revealed a series of upregulated and downregulated proteins. Following orthogonal validation, upregulated TRX (P = .001) and IGHA1 (P = .001) were observed in ERs relative to NRs. CONCLUSIONS Several novel candidate tissue biomarkers have been identified in this study, which could discriminate patients with UC at risk of early relapse from those in long-term sustained remission. Our findings may pave the way for pre-emptive UC disease monitoring and therapeutic decision making.
Collapse
Affiliation(s)
- Arash Assadsangabi
- Molecular Gastroenterology Research Group, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
- Gastroenterology Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom
- Gastroenterology Department, Salford Royal NHS Foundation Trust, Salford, United Kingdom
- Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Caroline A Evans
- Molecular Gastroenterology Research Group, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
- Biological and Systems Engineering Group, ChELSI Institute, Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Bernard M Corfe
- Molecular Gastroenterology Research Group, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Alan J Lobo
- Molecular Gastroenterology Research Group, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
- Gastroenterology Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom
| |
Collapse
|
6
|
Consalvo KM, Rijal R, Beruvides SL, Mitchell R, Beauchemin K, Collins D, Scoggin J, Scott J, Gomer RH. PTEN and the PTEN-like phosphatase CnrN have both distinct and overlapping roles in a Dictyostelium chemorepulsion pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.23.581751. [PMID: 38464111 PMCID: PMC10925239 DOI: 10.1101/2024.02.23.581751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The directed movement of eukaryotic cells is crucial for processes such as embryogenesis and immune cell trafficking. The enzyme Phosphatase and tensin homolog (PTEN) dephosphorylates phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P 3 ] to phosphatidylinositol 4,5-bisphosphate [PI(4,5)P 2 ]. Dictyostelium discoideum cells require both PTEN and the PTEN-like phosphatase CnrN to locally inhibit Ras activation to induce biased movement of cells away from the secreted chemorepellent protein AprA. Both PTEN and CnrN decrease basal levels of PI(3,4,5)P 3 and increase basal numbers of macropinosomes, and AprA prevents this increase. AprA requires both PTEN and CnrN to increase PI(4,5)P 2 levels, decrease PI(3,4,5)P 3 levels, inhibit proliferation, decrease myosin II phosphorylation, and increase filopod sizes. AprA causes PTEN, similar to CnrN, to localize to the side of the cell towards AprA in an AprA gradient. However, PTEN and CnrN also have distinct roles in some signaling pathways. PTEN, but not CnrN, decreases basal levels of PI(4,5)P 2 , AprA requires PTEN, but not CnrN, to induce cell roundness, and CnrN and PTEN have different effects on the number of filopods and pseudopods, and the sizes of filopods. Together, our results suggest that CnrN and PTEN play unique roles in D. discoideum signaling pathways, and possibly dephosphorylate PI(3,4,5)P 3 in different membrane domains, to mediate chemorepulsion away from AprA.
Collapse
|
7
|
Niu F, Li L, Wang L, Xiao J, Xu S, Liu Y, Lin L, Yu C, Wei Z. Autoinhibition and activation of myosin VI revealed by its cryo-EM structure. Nat Commun 2024; 15:1187. [PMID: 38331992 PMCID: PMC10853514 DOI: 10.1038/s41467-024-45424-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Myosin VI is the only molecular motor that moves towards the minus end along actin filaments. Numerous cellular processes require myosin VI and tight regulations of the motor's activity. Defects in myosin VI activity are known to cause genetic diseases such as deafness and cardiomyopathy. However, the molecular mechanisms underlying the activity regulation of myosin VI remain elusive. Here, we determined the high-resolution cryo-electron microscopic structure of myosin VI in its autoinhibited state. Our structure reveals that autoinhibited myosin VI adopts a compact, monomeric conformation via extensive interactions between the head and tail domains, orchestrated by an elongated single-α-helix region resembling a "spine". This autoinhibited structure effectively blocks cargo binding sites and represses the motor's ATPase activity. Certain cargo adaptors such as GIPC can release multiple inhibitory interactions and promote motor activity, pointing to a cargo-mediated activation of the processive motor. Moreover, our structural findings allow rationalization of disease-associated mutations in myosin VI. Beyond the activity regulation mechanisms of myosin VI, our study also sheds lights on how activities of other myosin motors such as myosin VII and X might be regulated.
Collapse
Affiliation(s)
- Fengfeng Niu
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China
| | - Lingxuan Li
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Lei Wang
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jinman Xiao
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China
| | - Shun Xu
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yong Liu
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Leishu Lin
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Cong Yu
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Zhiyi Wei
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
8
|
Bourdais A, Dehapiot B, Halet G. MRCK activates mouse oocyte myosin II for spindle rotation and male pronucleus centration. J Cell Biol 2023; 222:e202211029. [PMID: 37651121 PMCID: PMC10470461 DOI: 10.1083/jcb.202211029] [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: 11/08/2022] [Revised: 06/24/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
Asymmetric meiotic divisions in oocytes rely on spindle positioning in close vicinity to the cortex. In metaphase II mouse oocytes, eccentric spindle positioning triggers cortical polarization, including the build-up of an actin cap surrounded by a ring of activated myosin II. While the role of the actin cap in promoting polar body formation is established, ring myosin II activation mechanisms and functions have remained elusive. Here, we show that ring myosin II activation requires myotonic dystrophy kinase-related Cdc42-binding kinase (MRCK), downstream of polarized Cdc42. MRCK inhibition resulted in spindle rotation defects during anaphase II, precluding polar body extrusion. Remarkably, disengagement of segregated chromatids from the anaphase spindle could rescue rotation. We further show that the MRCK/myosin II pathway is activated in the fertilization cone and is required for male pronucleus migration toward the center of the zygote. These findings provide novel insights into the mechanism of myosin II activation in oocytes and its role in orchestrating asymmetric division and pronucleus centration.
Collapse
Affiliation(s)
- Anne Bourdais
- University of Rennes, CNRS - UMR 6290, Institute of Genetics and Development of Rennes, Rennes, France
| | - Benoit Dehapiot
- University of Rennes, CNRS - UMR 6290, Institute of Genetics and Development of Rennes, Rennes, France
| | - Guillaume Halet
- University of Rennes, CNRS - UMR 6290, Institute of Genetics and Development of Rennes, Rennes, France
| |
Collapse
|
9
|
Canon L, Kikuti C, Planelles-Herrero VJ, Lin T, Mayeux F, Sirkia H, Lee YI, Heidsieck L, Velikovsky L, David A, Liu X, Moussaoui D, Forest E, Höök P, Petersen KJ, Morgan TE, Di Cicco A, Sirés-Campos J, Derivery E, Lévy D, Delevoye C, Sweeney HL, Houdusse A. How myosin VI traps its off-state, is activated and dimerizes. Nat Commun 2023; 14:6732. [PMID: 37872146 PMCID: PMC10593786 DOI: 10.1038/s41467-023-42376-2] [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: 01/05/2023] [Accepted: 10/10/2023] [Indexed: 10/25/2023] Open
Abstract
Myosin VI (Myo6) is the only minus-end directed nanomotor on actin, allowing it to uniquely contribute to numerous cellular functions. As for other nanomotors, the proper functioning of Myo6 relies on precise spatiotemporal control of motor activity via a poorly defined off-state and interactions with partners. Our structural, functional, and cellular studies reveal key features of myosin regulation and indicate that not all partners can activate Myo6. TOM1 and Dab2 cannot bind the off-state, while GIPC1 binds Myo6, releases its auto-inhibition and triggers proximal dimerization. Myo6 partners thus differentially recruit Myo6. We solved a crystal structure of the proximal dimerization domain, and show that its disruption compromises endocytosis in HeLa cells, emphasizing the importance of Myo6 dimerization. Finally, we show that the L926Q deafness mutation disrupts Myo6 auto-inhibition and indirectly impairs proximal dimerization. Our study thus demonstrates the importance of partners in the control of Myo6 auto-inhibition, localization, and activation.
Collapse
Affiliation(s)
- Louise Canon
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
| | - Carlos Kikuti
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
| | - Vicente J Planelles-Herrero
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
| | - Tianming Lin
- Department of Pharmacology & Therapeutics and the Myology Institute, University of Florida College of Medicine, PO Box 100267, Gainesville, Florida, 32610-0267, USA
| | - Franck Mayeux
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
| | - Helena Sirkia
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
| | - Young Il Lee
- Department of Pharmacology & Therapeutics and the Myology Institute, University of Florida College of Medicine, PO Box 100267, Gainesville, Florida, 32610-0267, USA
| | - Leila Heidsieck
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
| | - Léonid Velikovsky
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
| | - Amandine David
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
| | - Xiaoyan Liu
- Department of Pharmacology & Therapeutics and the Myology Institute, University of Florida College of Medicine, PO Box 100267, Gainesville, Florida, 32610-0267, USA
| | - Dihia Moussaoui
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
| | - Emma Forest
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
- École Nationale Supérieure de Chimie de Montpellier, 240 Avenue du Professeur Emile Jeanbrau, 34090, Montpellier, France
| | - Peter Höök
- Department of Pharmacology & Therapeutics and the Myology Institute, University of Florida College of Medicine, PO Box 100267, Gainesville, Florida, 32610-0267, USA
| | - Karl J Petersen
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
| | | | - Aurélie Di Cicco
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, 75005, Paris, France
| | - Julia Sirés-Campos
- Structure et Compartimentation Membranaire, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
| | | | - Daniel Lévy
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, 75005, Paris, France
| | - Cédric Delevoye
- Structure et Compartimentation Membranaire, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France
| | - H Lee Sweeney
- Department of Pharmacology & Therapeutics and the Myology Institute, University of Florida College of Medicine, PO Box 100267, Gainesville, Florida, 32610-0267, USA.
| | - Anne Houdusse
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258, Paris cedex 05, France.
| |
Collapse
|
10
|
Wang C, Ding J, Wei Q, Du S, Gong X, Chew TG. Mechanosensitive accumulation of non-muscle myosin IIB during mitosis requires its translocation activity. iScience 2023; 26:107773. [PMID: 37720093 PMCID: PMC10504539 DOI: 10.1016/j.isci.2023.107773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/02/2023] [Accepted: 08/26/2023] [Indexed: 09/19/2023] Open
Abstract
Non-muscle myosin II (NMII) is a force-generating mechanosensitive enzyme that responds to mechanical forces. NMIIs mechanoaccumulate at the cell cortex in response to mechanical forces. It is essential for cells to mechanically adapt to the physical environment, failure of which results in mitotic defects when dividing in confined environment. Much less is known about how NMII mechanoaccumulation is regulated during mitosis. We show that mitotic cells respond to compressive stress by promoting accumulation of active RhoA at the cell cortex as in interphase cells. RhoA mechanoresponse during mitosis activates and stabilizes NMIIB via ROCK signaling, leading to NMIIB mechanoaccumulation at the cell cortex. Using disease-related myosin II mutations, we found that NMIIB mechanoaccumulation requires its motor activity that translocates actin filaments, but not just its actin-binding function. Thus, the motor activity coordinates structural movement and nucleotide state changes to fine-tune actin-binding affinity optimal for NMIIs to generate and respond to forces.
Collapse
Affiliation(s)
- Chao Wang
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- The Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
| | - Jingjing Ding
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- The Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
| | - Qiaodong Wei
- Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shoukang Du
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- The Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
| | - Xiaobo Gong
- Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ting Gang Chew
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- The Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
| |
Collapse
|
11
|
Brunet T. Cell contractility in early animal evolution. Curr Biol 2023; 33:R966-R985. [PMID: 37751712 DOI: 10.1016/j.cub.2023.07.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Tissue deformation mediated by collective cell contractility is a signature characteristic of animals. In most animals, fast and reversible contractions of muscle cells mediate behavior, while slow and irreversible contractions of epithelial or mesenchymal cells play a key role in morphogenesis. Animal tissue contractility relies on the activity of the actin/myosin II complex (together referred to as 'actomyosin'), an ancient and versatile molecular machinery that performs a broad range of functions in development and physiology. This review synthesizes emerging insights from morphological and molecular studies into the evolutionary history of animal contractile tissue. The most ancient functions of actomyosin are cell crawling and cytokinesis, which are found in a wide variety of unicellular eukaryotes and in individual metazoan cells. Another contractile functional module, apical constriction, is universal in metazoans and shared with choanoflagellates, their closest known living relatives. The evolution of animal contractile tissue involved two key innovations: firstly, the ability to coordinate and integrate actomyosin assembly across multiple cells, notably to generate supracellular cables, which ensure tissue integrity but also allow coordinated morphogenesis and movements at the organism scale; and secondly, the evolution of dedicated contractile cell types for adult movement, belonging to two broad categories respectively defined by the expression of the fast (striated-type) and slow (smooth/non-muscle-type) myosin II paralogs. Both contractile cell types ancestrally resembled generic contractile epithelial or mesenchymal cells and might have played a versatile role in both behavior and morphogenesis. Modern animal contractile cells span a continuum between unspecialized contractile epithelia (which underlie behavior in modern placozoans), epithelia with supracellular actomyosin cables (found in modern sponges), epitheliomuscular tissues (with a concentration of actomyosin cables in basal processes, for example in sea anemones), and specialized muscle tissue that has lost most or all epithelial properties (as in ctenophores, jellyfish and bilaterians). Recent studies in a broad range of metazoans have begun to reveal the molecular basis of these transitions, powered by the elaboration of the contractile apparatus and the evolution of 'core regulatory complexes' of transcription factors specifying contractile cell identity.
Collapse
Affiliation(s)
- Thibaut Brunet
- Institut Pasteur, Université Paris-Cité, CNRS UMR3691, Evolutionary Cell Biology and Evolution of Morphogenesis Unit, 25-28 Rue du Docteur Roux, 75015 Paris, France.
| |
Collapse
|
12
|
Brito C, Pereira JM, Mesquita FS, Cabanes D, Sousa S. Src-Dependent NM2A Tyrosine Phosphorylation Regulates Actomyosin Remodeling. Cells 2023; 12:1871. [PMID: 37508535 PMCID: PMC10377941 DOI: 10.3390/cells12141871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/07/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Non-muscle myosin 2A (NM2A) is a key cytoskeletal enzyme that, along with actin, assembles into actomyosin filaments inside cells. NM2A is fundamental for cell adhesion and motility, playing important functions in different stages of development and during the progression of viral and bacterial infections. Phosphorylation events regulate the activity and the cellular localization of NM2A. We previously identified the tyrosine phosphorylation of residue 158 (pTyr158) in the motor domain of the NM2A heavy chain. This phosphorylation can be promoted by Listeria monocytogenes infection of epithelial cells and is dependent on Src kinase; however, its molecular role is unknown. Here, we show that the status of pTyr158 defines cytoskeletal organization, affects the assembly/disassembly of focal adhesions, and interferes with cell migration. Cells overexpressing a non-phosphorylatable NM2A variant or expressing reduced levels of Src kinase display increased stress fibers and larger focal adhesions, suggesting an altered contraction status consistent with the increased NM2A activity that we also observed. We propose NM2A pTyr158 as a novel layer of regulation of actomyosin cytoskeleton organization.
Collapse
Affiliation(s)
- Cláudia Brito
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
- MCBiology PhD Program-Instituto de Ciências Biomédicas Abel Salazar-ICBAS, University of Porto, 4050-313 Porto, Portugal
| | - Joana M Pereira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
- MCBiology PhD Program-Instituto de Ciências Biomédicas Abel Salazar-ICBAS, University of Porto, 4050-313 Porto, Portugal
| | - Francisco S Mesquita
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
| | - Didier Cabanes
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
| | - Sandra Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC, Instituto de Biologia Celular e Molecular, 4200-135 Porto, Portugal
| |
Collapse
|
13
|
Zhang W, Wu Y, J Gunst S. Membrane adhesion junctions regulate airway smooth muscle phenotype and function. Physiol Rev 2023; 103:2321-2347. [PMID: 36796098 PMCID: PMC10243546 DOI: 10.1152/physrev.00020.2022] [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: 05/31/2022] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
The local environment surrounding airway smooth muscle (ASM) cells has profound effects on the physiological and phenotypic properties of ASM tissues. ASM is continually subjected to the mechanical forces generated during breathing and to the constituents of its surrounding extracellular milieu. The smooth muscle cells within the airways continually modulate their properties to adapt to these changing environmental influences. Smooth muscle cells connect to the extracellular cell matrix (ECM) at membrane adhesion junctions that provide mechanical coupling between smooth muscle cells within the tissue. Membrane adhesion junctions also sense local environmental signals and transduce them to cytoplasmic and nuclear signaling pathways in the ASM cell. Adhesion junctions are composed of clusters of transmembrane integrin proteins that bind to ECM proteins outside the cell and to large multiprotein complexes in the submembranous cytoplasm. Physiological conditions and stimuli from the surrounding ECM are sensed by integrin proteins and transduced by submembranous adhesion complexes to signaling pathways to the cytoskeleton and nucleus. The transmission of information between the local environment of the cells and intracellular processes enables ASM cells to rapidly adapt their physiological properties to modulating influences in their extracellular environment: mechanical and physical forces that impinge on the cell, ECM constituents, local mediators, and metabolites. The structure and molecular organization of adhesion junction complexes and the actin cytoskeleton are dynamic and constantly changing in response to environmental influences. The ability of ASM to rapidly accommodate to the ever-changing conditions and fluctuating physical forces within its local environment is essential for its normal physiological function.
Collapse
Affiliation(s)
- Wenwu Zhang
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Yidi Wu
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Susan J Gunst
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| |
Collapse
|
14
|
Abstract
Under relaxing conditions, the two heads of myosin II interact with each other and with the proximal part (S2) of the myosin tail, establishing the interacting-heads motif (IHM), found in myosin molecules and thick filaments of muscle and nonmuscle cells. The IHM is normally thought of as a single, unique structure, but there are several variants. In the simplest ("canonical") IHM, occurring in most relaxed thick filaments and in heavy meromyosin, the interacting heads bend back and interact with S2, and the motif lies parallel to the filament surface. In one variant, occurring in insect indirect flight muscle, there is no S2-head interaction and the motif is perpendicular to the filament. In a second variant, found in smooth and nonmuscle single myosin molecules in their inhibited (10S) conformation, S2 is shifted ∼20 Å from the canonical form and the tail folds twice and wraps around the interacting heads. These molecule and filament IHM variants have important energetic and pathophysiological consequences. (1) The canonical motif, with S2-head interaction, correlates with the super-relaxed (SRX) state of myosin. The absence of S2-head interaction in insects may account for the lower stability of this IHM and apparent absence of SRX in indirect flight muscle, contributing to the quick initiation of flight in insects. (2) The ∼20 Å shift of S2 in 10S myosin molecules means that S2-head interactions are different from those in the canonical IHM. This variant therefore cannot be used to analyze the impact of myosin mutations on S2-head interactions that occur in filaments, as has been proposed. It can be used, instead, to analyze the structural impact of mutations in smooth and nonmuscle myosin.
Collapse
Affiliation(s)
- Raúl Padrón
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Debabrata Dutta
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Roger Craig
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA
| |
Collapse
|
15
|
Konietzny A, Wegmann S, Mikhaylova M. The endoplasmic reticulum puts a new spin on synaptic tagging. Trends Neurosci 2023; 46:32-44. [PMID: 36428191 DOI: 10.1016/j.tins.2022.10.012] [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: 08/18/2022] [Revised: 10/12/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022]
Abstract
The heterogeneity of the endoplasmic reticulum (ER) makes it a versatile platform for a broad range of homeostatic processes, ranging from calcium regulation to synthesis and trafficking of proteins and lipids. It is not surprising that neurons use this organelle to fine-tune synaptic properties and thereby provide specificity to synaptic inputs. In this review, we discuss the mechanisms that enable activity-dependent ER recruitment into dendritic spines, with a focus on molecular mechanisms that mediate transport and retention of the ER in spines. The role of calcium signaling in spine ER, synaptopodin 'tagging' of active synapses, and the formation of the spine apparatus (SA) are highlighted. Finally, we discuss the role of liquid-liquid phase separation as a possible driving force in these processes.
Collapse
Affiliation(s)
- Anja Konietzny
- AG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin, Germany; Guest Group 'Neuronal Protein Transport', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Wegmann
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Marina Mikhaylova
- AG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Berlin, Germany; Guest Group 'Neuronal Protein Transport', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
16
|
Ivanov AI, Lechuga S, Marino‐Melendez A, Naydenov NG. Unique and redundant functions of cytoplasmic actins and nonmuscle myosin II isoforms at epithelial junctions. Ann N Y Acad Sci 2022; 1515:61-74. [PMID: 35673768 PMCID: PMC9489603 DOI: 10.1111/nyas.14808] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The integrity and functions of epithelial barriers depend on the formation of adherens junctions (AJs) and tight junctions (TJs). A characteristic feature of AJs and TJs is their association with the cortical cytoskeleton composed of actin filaments and nonmuscle myosin II (NM-II) motors. Mechanical forces generated by the actomyosin cytoskeleton are essential for junctional assembly, stability, and remodeling. Epithelial cells express two different actin proteins and three NM-II isoforms, all known to be associated with AJs and TJs. Despite their structural similarity, different actin and NM-II isoforms have distinct biochemical properties, cellular distribution, and functions. The diversity of epithelial actins and myosin motors could be essential for the regulation of different steps of junctional formation, maturation, and disassembly. This review focuses on the roles of actin and NM-II isoforms in controlling the integrity and barrier properties of various epithelia. We discuss the effects of the depletion of individual actin isoforms and NM-II motors on the assembly and barrier function of AJs and TJs in model epithelial monolayers in vitro. We also describe the functional consequences of either total or tissue-specific gene knockout of different actins and NM-II motors, with a focus on the development and integrity of different epithelia in vivo.
Collapse
Affiliation(s)
- Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Armando Marino‐Melendez
- Department of Inflammation and Immunity, Lerner Research InstituteCleveland ClinicClevelandOhioUSA
| | - Nayden G. Naydenov
- Department of Inflammation and Immunity, Lerner Research InstituteCleveland ClinicClevelandOhioUSA
| |
Collapse
|
17
|
McMillan SN, Scarff CA. Cryo-electron microscopy analysis of myosin at work and at rest. Curr Opin Struct Biol 2022; 75:102391. [DOI: 10.1016/j.sbi.2022.102391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 01/01/2023]
|
18
|
Yun H, Im HJ, Choe C, Roh S. Effect of LOXL2 on metastasis through remodeling of the cell surface matrix in non-small cell lung cancer cells. Gene 2022; 830:146504. [PMID: 35483499 DOI: 10.1016/j.gene.2022.146504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 12/17/2022]
Abstract
Lung cancer is the prominent cause of cancer-associated death primarily because of distant metastatic disease. The metastatic potential of non-small cell lung cancer (NSCLC) is associated with tumor cell aggregation. However, the systemic mechanotransduction mechanism by which tumor cells dynamically aggregate and disseminate is poorly understood, especially in NSCLC. In this study, we examine whether the cell surface matrix plays an important role in metastasis. We used poly-2-hydroxyethyl methacrylate-based 3D spheroid formation methods to mimic in vivo metastatic lesions. Supra-structural analysis of human NSCLC A549 cells stained with ruthenium red for transmission electron microscopy (TEM) showed that glycocalyx surrounding the cell surface in 2D culture decreases in 3D culture. Comprehensive gene expression analysis revealed that the genes associated with cell adhesion were distinctly enriched in A549 cell spheroids. Of these, downregulation of the tumor metastatic microenvironment facilitator LOXL2, a copper-dependent enzyme catalyzing posttranslational oxidative deamination of peptidyl lysine, was of special interest. Knockdown of LOXL2 thickened the cell surface matrix in 2D culture and impaired compact aggregate formation in 3D culture. Moreover, A549 cell spheroids with endogenous overexpression of LOXL2 increased their dissemination on basement extracellular matrix Matrigel. Overall, these data imply that cell detachment-downregulated LOXL2 contributes to cell surface matrix remodeling, leading to collective dissemination of free-floating aggregates.
Collapse
Affiliation(s)
- Heesu Yun
- Cellular Reprogramming and Embryo Biotechnology Lab, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Hee-Jeong Im
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, USA; Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL, USA
| | - Chungyoul Choe
- Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University, School of Medicine, Seoul, Republic of Korea.
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology Lab, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea.
| |
Collapse
|
19
|
Nomura T, Hayakawa K, Sato N, Obinata T. Periodic Stretching of Cultured Myotubes Enhances Myofibril Assembly. Zoolog Sci 2022; 39. [DOI: 10.2108/zs220015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/22/2022] [Indexed: 11/17/2022]
Affiliation(s)
- Takahiro Nomura
- Department of Biology, Graduate School of Science and Technology, Chiba University, Yayoi-cho, Inage-ku, Chiba 263-8522, Chiba, Japan
| | - Kimihide Hayakawa
- Department of Biology, Graduate School of Science and Technology, Chiba University, Yayoi-cho, Inage-ku, Chiba 263-8522, Chiba, Japan
| | - Naruki Sato
- Department of Biology, Graduate School of Science and Technology, Chiba University, Yayoi-cho, Inage-ku, Chiba 263-8522, Chiba, Japan
| | - Takashi Obinata
- Department of Biology, Graduate School of Science and Technology, Chiba University, Yayoi-cho, Inage-ku, Chiba 263-8522, Chiba, Japan
| |
Collapse
|
20
|
Weißenbruch K, Fladung M, Grewe J, Baulesch L, Schwarz US, Bastmeyer M. Nonmuscle myosin IIA dynamically guides regulatory light chain phosphorylation and assembly of nonmuscle myosin IIB. Eur J Cell Biol 2022; 101:151213. [DOI: 10.1016/j.ejcb.2022.151213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 01/27/2023] Open
|
21
|
Erdener ŞE, Küreli G, Dalkara T. Contractile apparatus in CNS capillary pericytes. NEUROPHOTONICS 2022; 9:021904. [PMID: 35106320 PMCID: PMC8785978 DOI: 10.1117/1.nph.9.2.021904] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
Significance: Whether or not capillary pericytes contribute to blood flow regulation in the brain and retina has long been debated. This was partly caused by failure of detecting the contractile protein α -smooth muscle actin ( α -SMA) in capillary pericytes. Aim: The aim of this review is to summarize recent developments in detecting α -SMA and contractility in capillary pericytes and the relevant literature on the biology of actin filaments. Results: Evidence suggests that for visualization of the small amounts of α -SMA in downstream mid-capillary pericytes, actin depolymerization must be prevented during tissue processing. Actin filaments turnover is mainly based on de/re-polymerization rather than transcription of the monomeric form, hence, small amounts of α -SMA mRNA may evade detection by transcriptomic studies. Similarly, transgenic mice expressing fluorescent reporters under the α -SMA promoter may yield low fluorescence due to limited transcriptional activity in mid-capillary pericytes. Recent studies show that pericytes including mid-capillary ones express several actin isoforms and myosin heavy chain type 11, the partner of α -SMA in mediating contraction. Emerging evidence also suggests that actin polymerization in pericytes may have a role in regulating the tone of downstream capillaries. Conclusions: With guidance of actin biology, innovative labeling and imaging techniques can reveal the molecular machinery of contraction in pericytes.
Collapse
Affiliation(s)
- Şefik E. Erdener
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| | - Gülce Küreli
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| | - Turgay Dalkara
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| |
Collapse
|
22
|
Yanagida A, Corujo-Simon E, Revell CK, Sahu P, Stirparo GG, Aspalter IM, Winkel AK, Peters R, De Belly H, Cassani DAD, Achouri S, Blumenfeld R, Franze K, Hannezo E, Paluch EK, Nichols J, Chalut KJ. Cell surface fluctuations regulate early embryonic lineage sorting. Cell 2022; 185:777-793.e20. [PMID: 35196500 PMCID: PMC8896887 DOI: 10.1016/j.cell.2022.01.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 10/22/2021] [Accepted: 01/26/2022] [Indexed: 01/24/2023]
Abstract
In development, lineage segregation is coordinated in time and space. An important example is the mammalian inner cell mass, in which the primitive endoderm (PrE, founder of the yolk sac) physically segregates from the epiblast (EPI, founder of the fetus). While the molecular requirements have been well studied, the physical mechanisms determining spatial segregation between EPI and PrE remain elusive. Here, we investigate the mechanical basis of EPI and PrE sorting. We find that rather than the differences in static cell surface mechanical parameters as in classical sorting models, it is the differences in surface fluctuations that robustly ensure physical lineage sorting. These differential surface fluctuations systematically correlate with differential cellular fluidity, which we propose together constitute a non-equilibrium sorting mechanism for EPI and PrE lineages. By combining experiments and modeling, we identify cell surface dynamics as a key factor orchestrating the correct spatial segregation of the founder embryonic lineages.
Collapse
Affiliation(s)
- Ayaka Yanagida
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; Centre for Trophoblast Research, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Elena Corujo-Simon
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Christopher K Revell
- Cavendish Laboratory, Department of Physics, University of Cambridge, JJ Thomson Ave, Cambridge CB3 0HE, UK
| | - Preeti Sahu
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria
| | - Giuliano G Stirparo
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Irene M Aspalter
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Alex K Winkel
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Ruby Peters
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Henry De Belly
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Davide A D Cassani
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Sarra Achouri
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; Centre for Trophoblast Research, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Raphael Blumenfeld
- Gonville & Caius College, University of Cambridge, Trinity St., Cambridge CB2 1TA, UK
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Edouard Hannezo
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria
| | - Ewa K Paluch
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK.
| | - Jennifer Nichols
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; Centre for Trophoblast Research, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK.
| | - Kevin J Chalut
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK.
| |
Collapse
|
23
|
Konietzny A, Grendel J, Kadek A, Bucher M, Han Y, Hertrich N, Dekkers DHW, Demmers JAA, Grünewald K, Uetrecht C, Mikhaylova M. Caldendrin and myosin V regulate synaptic spine apparatus localization via ER stabilization in dendritic spines. EMBO J 2022; 41:e106523. [PMID: 34935159 PMCID: PMC8844991 DOI: 10.15252/embj.2020106523] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/08/2021] [Accepted: 11/19/2021] [Indexed: 11/21/2022] Open
Abstract
Excitatory synapses of principal hippocampal neurons are frequently located on dendritic spines. The dynamic strengthening or weakening of individual inputs results in structural and molecular diversity of dendritic spines. Active spines with large calcium ion (Ca2+ ) transients are frequently invaded by a single protrusion from the endoplasmic reticulum (ER), which is dynamically transported into spines via the actin-based motor myosin V. An increase in synaptic strength correlates with stable anchoring of the ER, followed by the formation of an organelle referred to as the spine apparatus. Here, we show that myosin V binds the Ca2+ sensor caldendrin, a brain-specific homolog of the well-known myosin V interactor calmodulin. While calmodulin is an essential activator of myosin V motor function, we found that caldendrin acts as an inhibitor of processive myosin V movement. In mouse and rat hippocampal neurons, caldendrin regulates spine apparatus localization to a subset of dendritic spines through a myosin V-dependent pathway. We propose that caldendrin transforms myosin into a stationary F-actin tether that enables the localization of ER tubules and formation of the spine apparatus in dendritic spines.
Collapse
Affiliation(s)
- Anja Konietzny
- RG OptobiologyInstitute of BiologyHumboldt Universität zu BerlinBerlinGermany
- Guest Group Neuronal Protein TransportCenter for Molecular NeurobiologyZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Jasper Grendel
- RG OptobiologyInstitute of BiologyHumboldt Universität zu BerlinBerlinGermany
- Guest Group Neuronal Protein TransportCenter for Molecular NeurobiologyZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Alan Kadek
- Leibniz Institute for Experimental Virology (HPI)HamburgGermany
- European XFEL GmbHSchenefeldGermany
| | - Michael Bucher
- RG OptobiologyInstitute of BiologyHumboldt Universität zu BerlinBerlinGermany
- Guest Group Neuronal Protein TransportCenter for Molecular NeurobiologyZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Yuhao Han
- RG OptobiologyInstitute of BiologyHumboldt Universität zu BerlinBerlinGermany
- Guest Group Neuronal Protein TransportCenter for Molecular NeurobiologyZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Centre for Structural Systems BiologyHamburgGermany
| | - Nathalie Hertrich
- RG OptobiologyInstitute of BiologyHumboldt Universität zu BerlinBerlinGermany
- Guest Group Neuronal Protein TransportCenter for Molecular NeurobiologyZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | | | | | - Kay Grünewald
- Leibniz Institute for Experimental Virology (HPI)HamburgGermany
- Centre for Structural Systems BiologyHamburgGermany
- Department of ChemistryUniversity of HamburgHamburgGermany
| | - Charlotte Uetrecht
- Leibniz Institute for Experimental Virology (HPI)HamburgGermany
- European XFEL GmbHSchenefeldGermany
- Centre for Structural Systems BiologyHamburgGermany
| | - Marina Mikhaylova
- RG OptobiologyInstitute of BiologyHumboldt Universität zu BerlinBerlinGermany
- Guest Group Neuronal Protein TransportCenter for Molecular NeurobiologyZMNHUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
24
|
Baker K, Geeves MA, Mulvihill DP. Acetylation stabilises calmodulin-regulated calcium signalling. FEBS Lett 2022; 596:762-771. [PMID: 35100446 PMCID: PMC9303947 DOI: 10.1002/1873-3468.14304] [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: 11/30/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/01/2022]
Abstract
Calmodulin is a conserved calcium signalling protein that regulates a wide range of cellular functions. Amino‐terminal acetylation is a ubiquitous post‐translational modification that affects the majority of human proteins, to stabilise structure, as well as regulate function and proteolytic degradation. Here, we present data on the impact of amino‐terminal acetylation upon structure and calcium signalling function of fission yeast calmodulin. We show that NatA‐dependent acetylation stabilises the helical structure of the Schizosaccharomyces pombe calmodulin, impacting its ability to associate with myosin at endocytic foci. We go on to show that this conserved modification impacts both the calcium‐binding capacity of yeast and human calmodulins. These findings have significant implications for research undertaken into this highly conserved essential protein.
Collapse
Affiliation(s)
- Karen Baker
- School of Biosciences, University of Kent, Canterbury, Kent, CT2 7NJ, UK
| | - Michael A Geeves
- School of Biosciences, University of Kent, Canterbury, Kent, CT2 7NJ, UK
| | - Daniel P Mulvihill
- School of Biosciences, University of Kent, Canterbury, Kent, CT2 7NJ, UK
| |
Collapse
|
25
|
Pepper I, Galkin VE. Actomyosin Complex. Subcell Biochem 2022; 99:421-470. [PMID: 36151385 PMCID: PMC9710302 DOI: 10.1007/978-3-031-00793-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Formation of cross-bridges between actin and myosin occurs ubiquitously in eukaryotic cells and mediates muscle contraction, intracellular cargo transport, and cytoskeletal remodeling. Myosin motors repeatedly bind to and dissociate from actin filaments in a cycle that transduces the chemical energy from ATP hydrolysis into mechanical force generation. While the general layout of surface elements within the actin-binding interface is conserved among myosin classes, sequence divergence within these motifs alters the specific contacts involved in the actomyosin interaction as well as the kinetics of mechanochemical cycle phases. Additionally, diverse lever arm structures influence the motility and force production of myosin molecules during their actin interactions. The structural differences generated by myosin's molecular evolution have fine-tuned the kinetics of its isoforms and adapted them for their individual cellular roles. In this chapter, we will characterize the structural and biochemical basis of the actin-myosin interaction and explain its relationship with myosin's cellular roles, with emphasis on the structural variation among myosin isoforms that enables their functional specialization. We will also discuss the impact of accessory proteins, such as the troponin-tropomyosin complex and myosin-binding protein C, on the formation and regulation of actomyosin cross-bridges.
Collapse
Affiliation(s)
- Ian Pepper
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Vitold E Galkin
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
26
|
Gunther LK, Cirilo JA, Desetty R, Yengo CM. Deafness mutation in the MYO3A motor domain impairs actin protrusion elongation mechanism. Mol Biol Cell 2021; 33:ar5. [PMID: 34788109 PMCID: PMC8886822 DOI: 10.1091/mbc.e21-05-0232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Class III myosins are actin-based motors proposed to transport cargo to the distal tips of stereocilia in the inner ear hair cells and/or to participate in stereocilia length regulation, which is especially important during development. Mutations in the MYO3A gene are associated with delayed onset deafness. A previous study demonstrated that L697W, a dominant deafness mutation, disrupts MYO3A ATPase and motor properties but does not impair its ability to localize to the tips of actin protrusions. In the current study, we characterized the transient kinetic mechanism of the L697W motor ATPase cycle. Our kinetic analysis demonstrates that the mutation slows the ADP release and ATP hydrolysis steps, which results in a slight reduction in the duty ratio and slows detachment kinetics. Fluorescence recovery after photobleaching (FRAP) of filopodia tip localized L697W and WT MYO3A in COS-7 cells revealed that the mutant does not alter turnover or average intensity at the actin protrusion tips. We demonstrate that the mutation slows filopodia extension velocity in COS-7 cells which correlates with its twofold slower in vitro actin gliding velocity. Overall, this work allowed us to propose a model for how the motor properties of MYO3A are crucial for facilitating actin protrusion length regulation.
Collapse
Affiliation(s)
- Laura K Gunther
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, 17033
| | - Joseph A Cirilo
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, 17033
| | - Rohini Desetty
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, 17033
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, 17033
| |
Collapse
|
27
|
Truong Quang BA, Peters R, Cassani DAD, Chugh P, Clark AG, Agnew M, Charras G, Paluch EK. Extent of myosin penetration within the actin cortex regulates cell surface mechanics. Nat Commun 2021; 12:6511. [PMID: 34764258 PMCID: PMC8586027 DOI: 10.1038/s41467-021-26611-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 10/04/2021] [Indexed: 12/11/2022] Open
Abstract
In animal cells, shape is mostly determined by the actomyosin cortex, a thin cytoskeletal network underlying the plasma membrane. Myosin motors generate tension in the cortex, and tension gradients result in cellular deformations. As such, many cell morphogenesis studies have focused on the mechanisms controlling myosin activity and recruitment to the cortex. Here, we demonstrate using super-resolution microscopy that myosin does not always overlap with actin at the cortex, but remains restricted towards the cytoplasm in cells with low cortex tension. We propose that this restricted penetration results from steric hindrance, as myosin minifilaments are considerably larger than the cortical actin meshsize. We identify myosin activity and actin network architecture as key regulators of myosin penetration into the cortex, and show that increasing myosin penetration increases cortical tension. Our study reveals that the spatial coordination of myosin and actin at the cortex regulates cell surface mechanics, and unveils an important mechanism whereby myosin size controls its action by limiting minifilament penetration into the cortical actin network. More generally, our findings suggest that protein size could regulate function in dense cytoskeletal structures. Cellular deformations are largely driven by contractile forces generated by myosin motors in the submembraneous actin cortex. Here we show that these forces are controlled not simply by cortical myosin levels, but rather by myosins spatial arrangement, specifically the extent of their overlap with cortical actin.
Collapse
Affiliation(s)
- Binh An Truong Quang
- MRC Laboratory for Molecular Cell Biology, University College London, London, WC1E 6BT, UK
| | - Ruby Peters
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
| | - Davide A D Cassani
- MRC Laboratory for Molecular Cell Biology, University College London, London, WC1E 6BT, UK
| | - Priyamvada Chugh
- MRC Laboratory for Molecular Cell Biology, University College London, London, WC1E 6BT, UK
| | - Andrew G Clark
- MRC Laboratory for Molecular Cell Biology, University College London, London, WC1E 6BT, UK.,University of Stuttgart, Institute of Cell Biology and Immunology, Allmandring 31, 70569, Stuttgart, Germany
| | - Meghan Agnew
- MRC Laboratory for Molecular Cell Biology, University College London, London, WC1E 6BT, UK
| | - Guillaume Charras
- London Centre for Nanotechnology, University College London, London, WC1H 0AH, UK.,Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Ewa K Paluch
- MRC Laboratory for Molecular Cell Biology, University College London, London, WC1E 6BT, UK. .,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK.
| |
Collapse
|
28
|
Ready DF, Chang HC. Calcium waves facilitate and coordinate the contraction of endfeet actin stress fibers in Drosophila interommatidial cells. Development 2021; 148:272616. [PMID: 34698814 DOI: 10.1242/dev.199700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 10/18/2021] [Indexed: 01/04/2023]
Abstract
Actomyosin contraction shapes the Drosophila eye's panoramic view. The convex curvature of the retinal epithelium, organized in ∼800 close-packed ommatidia, depends upon a fourfold condensation of the retinal floor mediated by contraction of actin stress fibers in the endfeet of interommatidial cells (IOCs). How these tensile forces are coordinated is not known. Here, we discover a novel phenomenon: Ca2+ waves regularly propagate across the IOC network in pupal and adult eyes. Genetic evidence demonstrates that IOC waves are independent of phototransduction, but require inositol 1,4,5-triphosphate receptor (IP3R), suggesting these waves are mediated by Ca2+ releases from ER stores. Removal of IP3R disrupts stress fibers in IOC endfeet and increases the basal retinal surface by ∼40%, linking IOC waves to facilitating stress fiber contraction and floor morphogenesis. Further, IP3R loss disrupts the organization of a collagen IV network underneath the IOC endfeet, implicating ECM and its interaction with stress fibers in eye morphogenesis. We propose that coordinated cytosolic Ca2+ increases in IOC waves promote stress fiber contractions, ensuring an organized application of the planar tensile forces that condense the retinal floor.
Collapse
Affiliation(s)
- Donald F Ready
- Department of Biological Sciences, Purdue University, 915 West State Street, West Lafayette, Indiana 47907-2054, USA
| | - Henry C Chang
- Department of Biological Sciences, Purdue University, 915 West State Street, West Lafayette, Indiana 47907-2054, USA
| |
Collapse
|
29
|
Wang Y, Zhao X, Gao M, Wan X, Guo Y, Qu Y, Chen Y, Li T, Liu H, Jiang M, Wang F, Sun X. Myosin 1f-mediated activation of microglia contributes to the photoreceptor degeneration in a mouse model of retinal detachment. Cell Death Dis 2021; 12:926. [PMID: 34628463 PMCID: PMC8502177 DOI: 10.1038/s41419-021-03983-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 11/14/2022]
Abstract
Photoreceptor death and neurodegeneration is the leading cause of irreversible vision loss. The inflammatory response of microglia plays an important role in the process of neurodegeneration. In this study, we chose retinal detachment as the model of photoreceptor degeneration. We found Myosin 1f was upregulated after retinal detachment, and it was specifically expressed in microglia. Deficiency of myosin 1f protected against photoreceptor apoptosis by inhibiting microglia activation. The elimination of microglia can abolish the protective effect of myosin 1f deficiency. After stimulation by LPS, microglia with myosin 1f deficiency showed downregulation of the MAPK and AKT pathways. Our results demonstrated that myosin 1f plays a crucial role in microglia-induced neuroinflammation after retinal injury and photoreceptor degeneration by regulating two classic inflammatory pathways and thereby decreasing the expression of inflammatory cytokines. Knockout of myosin 1f reduces the intensity of the immune response and prevents cell death of photoreceptor, suggesting that myosin 1f can be inhibited to prevent a decline in visual acuity after retinal detachment.
Collapse
Affiliation(s)
- Yimin Wang
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Xiaohuan Zhao
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Min Gao
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoling Wan
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Yinong Guo
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Yingying Qu
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Translational Medicine Center, Shanghai General Hospital, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Tong Li
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
| | - Haiyun Liu
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
| | - Mei Jiang
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Feng Wang
- Shanghai Institute of Immunology, Translational Medicine Center, Shanghai General Hospital, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiaodong Sun
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- National Clinical Research Center for Eye Disease, Shanghai, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.
| |
Collapse
|
30
|
Porro C, Pennella A, Panaro MA, Trotta T. Functional Role of Non-Muscle Myosin II in Microglia: An Updated Review. Int J Mol Sci 2021; 22:ijms22136687. [PMID: 34206505 PMCID: PMC8267657 DOI: 10.3390/ijms22136687] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 02/08/2023] Open
Abstract
Myosins are a remarkable superfamily of actin-based motor proteins that use the energy derived from ATP hydrolysis to translocate actin filaments and to produce force. Myosins are abundant in different types of tissues and involved in a large variety of cellular functions. Several classes of the myosin superfamily are expressed in the nervous system; among them, non-muscle myosin II (NM II) is expressed in both neurons and non-neuronal brain cells, such as astrocytes, oligodendrocytes, endothelial cells, and microglia. In the nervous system, NM II modulates a variety of functions, such as vesicle transport, phagocytosis, cell migration, cell adhesion and morphology, secretion, transcription, and cytokinesis, as well as playing key roles during brain development, inflammation, repair, and myelination functions. In this review, we will provide a brief overview of recent emerging roles of NM II in resting and activated microglia cells, the principal regulators of immune processes in the central nervous system (CNS) in both physiological and pathological conditions. When stimulated, microglial cells react and produce a number of mediators, such as pro-inflammatory cytokines, free radicals, and nitric oxide, that enhance inflammation and contribute to neurodegenerative diseases. Inhibition of NM II could be a new therapeutic target to treat or to prevent CNS diseases.
Collapse
Affiliation(s)
- Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
| | - Antonio Pennella
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy;
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
- Correspondence:
| |
Collapse
|
31
|
Arthur AL, Crawford A, Houdusse A, Titus MA. VASP-mediated actin dynamics activate and recruit a filopodia myosin. eLife 2021; 10:68082. [PMID: 34042588 PMCID: PMC8352590 DOI: 10.7554/elife.68082] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022] Open
Abstract
Filopodia are thin, actin-based structures that cells use to interact with their environments. Filopodia initiation requires a suite of conserved proteins but the mechanism remains poorly understood. The actin polymerase VASP and a MyTH-FERM (MF) myosin, DdMyo7 in amoeba, are essential for filopodia initiation. DdMyo7 is localized to dynamic regions of the actin-rich cortex. Analysis of VASP mutants and treatment of cells with anti-actin drugs shows that myosin recruitment and activation in Dictyostelium requires localized VASP-dependent actin polymerization. Targeting of DdMyo7 to the cortex alone is not sufficient for filopodia initiation; VASP activity is also required. The actin regulator locally produces a cortical actin network that activates myosin and together they shape the actin network to promote extension of parallel bundles of actin during filopodia formation. This work reveals how filopodia initiation requires close collaboration between an actin-binding protein, the state of the actin cytoskeleton and MF myosin activity.
Collapse
Affiliation(s)
- Ashley L Arthur
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Amy Crawford
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| | - Anne Houdusse
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, Paris, France
| | - Margaret A Titus
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, United States
| |
Collapse
|
32
|
Gunther LK, Rohde JA, Tang W, Cirilo JA, Marang CP, Scott BD, Thomas DD, Debold EP, Yengo CM. FRET and optical trapping reveal mechanisms of actin activation of the power stroke and phosphate release in myosin V. J Biol Chem 2021; 295:17383-17397. [PMID: 33453985 DOI: 10.1074/jbc.ra120.015632] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/06/2020] [Indexed: 11/06/2022] Open
Abstract
Myosins generate force and motion by precisely coordinating their mechanical and chemical cycles, but the nature and timing of this coordination remains controversial. We utilized a FRET approach to examine the kinetics of structural changes in the force-generating lever arm in myosin V. We directly compared the FRET results with single-molecule mechanical events examined by optical trapping. We introduced a mutation (S217A) in the conserved switch I region of the active site to examine how myosin couples structural changes in the actin- and nucleotide-binding regions with force generation. Specifically, S217A enhanced the maximum rate of lever arm priming (recovery stroke) while slowing ATP hydrolysis, demonstrating that it uncouples these two steps. We determined that the mutation dramatically slows both actin-induced rotation of the lever arm (power stroke) and phosphate release (≥10-fold), whereas our simulations suggest that the maximum rate of both steps is unchanged by the mutation. Time-resolved FRET revealed that the structure of the pre- and post-power stroke conformations and mole fractions of these conformations were not altered by the mutation. Optical trapping results demonstrated that S217A does not dramatically alter unitary displacements or slow the working stroke rate constant, consistent with the mutation disrupting an actin-induced conformational change prior to the power stroke. We propose that communication between the actin- and nucleotide-binding regions of myosin assures a proper actin-binding interface and active site have formed before producing a power stroke. Variability in this coupling is likely crucial for mediating motor-based functions such as muscle contraction and intracellular transport.
Collapse
Affiliation(s)
- Laura K Gunther
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania, USA
| | - John A Rohde
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Wanjian Tang
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania, USA
| | - Joseph A Cirilo
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania, USA
| | - Christopher P Marang
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, USA
| | - Brent D Scott
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Edward P Debold
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, USA
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania, USA.
| |
Collapse
|
33
|
Ramirez I, Gholkar AA, Velasquez EF, Guo X, Tofig B, Damoiseaux R, Torres JZ. The myosin regulatory light chain Myl5 localizes to mitotic spindle poles and is required for proper cell division. Cytoskeleton (Hoboken) 2021; 78:23-35. [PMID: 33641240 DOI: 10.1002/cm.21654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/18/2022]
Abstract
Myosins are ATP-dependent actin-based molecular motors critical for diverse cellular processes like intracellular trafficking, cell motility, and cell invasion. During cell division, myosin MYO10 is important for proper mitotic spindle assembly, the anchoring of the spindle to the cortex, and positioning of the spindle to the cell mid-plane. However, myosins are regulated by myosin regulatory light chains (RLCs), and whether RLCs are important for cell division has remained unexplored. Here, we have determined that the previously uncharacterized myosin RLC Myl5 associates with the mitotic spindle and is required for cell division. We show that Myl5 localizes to the leading edge and filopodia during interphase and to mitotic spindle poles and spindle microtubules during early mitosis. Importantly, depletion of Myl5 led to defects in mitotic spindle assembly, chromosome congression, and chromosome segregation and to a slower transition through mitosis. Furthermore, Myl5 bound to MYO10 in vitro and co-localized with MYO10 at the spindle poles. These results suggest that Myl5 is important for cell division and that it may be performing its function through MYO10.
Collapse
Affiliation(s)
- Ivan Ramirez
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA
| | - Ankur A Gholkar
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA
| | - Erick F Velasquez
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA
| | - Xiao Guo
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA
| | - Bobby Tofig
- California NanoSystems Institute, Los Angeles, California, USA
| | - Robert Damoiseaux
- California NanoSystems Institute, Los Angeles, California, USA.,Department of Molecular and Medical Pharmacology, Los Angeles, California, USA
| | - Jorge Z Torres
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| |
Collapse
|
34
|
Hildebrand JD, Leventry AD, Aideyman OP, Majewski JC, Haddad JA, Bisi DC, Kaufmann N. A modifier screen identifies regulators of cytoskeletal architecture as mediators of Shroom-dependent changes in tissue morphology. Biol Open 2021; 10:bio.055640. [PMID: 33504488 PMCID: PMC7875558 DOI: 10.1242/bio.055640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Regulation of cell architecture is critical in the formation of tissues during animal development. The mechanisms that control cell shape must be both dynamic and stable in order to establish and maintain the correct cellular organization. Previous work has identified Shroom family proteins as essential regulators of cell morphology during vertebrate development. Shroom proteins regulate cell architecture by directing the subcellular distribution and activation of Rho-kinase, which results in the localized activation of non-muscle myosin II. Because the Shroom-Rock-myosin II module is conserved in most animal model systems, we have utilized Drosophila melanogaster to further investigate the pathways and components that are required for Shroom to define cell shape and tissue architecture. Using a phenotype-based heterozygous F1 genetic screen for modifiers of Shroom activity, we identified several cytoskeletal and signaling protein that may cooperate with Shroom. We show that two of these proteins, Enabled and Short stop, are required for ShroomA-induced changes in tissue morphology and are apically enriched in response to Shroom expression. While the recruitment of Ena is necessary, it is not sufficient to redefine cell morphology. Additionally, this requirement for Ena appears to be context dependent, as a variant of Shroom that is apically localized, binds to Rock, but lacks the Ena binding site, is still capable of inducing changes in tissue architecture. These data point to important cellular pathways that may regulate contractility or facilitate Shroom-mediated changes in cell and tissue morphology. Summary: Using Drosophila as a model system, we identify F-actin and microtubules as important determinants of how cells and tissues respond to Shroom induced contractility.
Collapse
Affiliation(s)
- Jeffrey D Hildebrand
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Adam D Leventry
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Omoregie P Aideyman
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - John C Majewski
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - James A Haddad
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Dawn C Bisi
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Nancy Kaufmann
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
35
|
The effect of sport and physical activity on transport proteins: implications for cancer prevention and control. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021. [PMID: 33485483 DOI: 10.1016/bs.apcsb.2020.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
The present contribution briefly overviews the major biological functions of the plasma membrane and of the transport proteins (transporters), which enable the movement of different molecules and substrates (either charged or uncharged) by passive (facilitated diffusion) or active transport. In particular, transporters are overviewed at the level of the skeletal muscles, which represent a highly complex, heterogeneous, plastic and dynamic tissue and are one of the most abundant tissues in humans, accounting for up to 40% of their total weight and containing up to 50%-75% of all body proteins. Moreover, it is shown how sport and physical activity finely tune and modulate human proteome, especially in terms of structural and functional improvements concerning the density of the transport proteins. These changes are among the factors responsible for the positive outcomes of training, which involve mainly the cardiovascular and the endocrine/metabolic systems. Different kinds of training (strength and endurance) enable to achieve such improvements, even though there seems to exist a dose-relationship intensity-dependent effect, with responses after 6-8 weeks of exercise and disappearing in the chronic period (years of training). Finally, exercise-induced changes at the level of transporters can play a role in terms of cancer prevention and management. Regular physical activity and exercise can, indeed, counteract the side-effects of chemotherapy drugs, including doxorubicin and other anthracycline derivatives, which may impair the functions of cardiac and skeletal muscles, probably modulating the expression of multidrug resistance proteins.
Collapse
|
36
|
Cao Y, Jia P, Wu Z, Huang M, Chen S, Zhang J, Huang B, Lei C. A novel SNP of MYO1A gene associated with heat-tolerance in Chinese cattle. Anim Biotechnol 2020; 33:810-815. [PMID: 33146068 DOI: 10.1080/10495398.2020.1837147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
With the advent of global climate change, heat-tolerance is becoming more and more important to the sustainability of animal husbandry production systems. Previous studies have shown that MYO1A gene associated with pigmentation may be closely related to heat-tolerance in cattle. In this study, a novel missense mutation (NC_037332.1 g.56390345 A > G) was first detected in MYO1A in 891 individuals of 35 cattle breeds, which transformed the amino acid isoleucine into valine. The purpose of this study was to determine the allele frequencies distribution of this locus in Chinese indigenous cattle and to analyze the relationship between this locus and heat-tolerance. Further analysis showed that frequency of wild allele A decreased gradually from northern cattle to southern cattle, whereas frequency of mutant type allele G showed the opposite pattern, which was consistent with the distribution of various climatic conditions of China. Additionally, association analysis was carried out between genotypes and four climatic conditions (annual mean temperature (T), relative humidity (H), temperature-humidity index (THI) and average annual sunshine hours (100-cloudiness) (SR)). Analysis results showed that genotypes were significantly correlated with climatic conditions. Therefore, our results suggest that the novel SNP (rs209559414) is related to heat-tolerance trait of Chinese indigenous cattle.
Collapse
Affiliation(s)
- Yanhong Cao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.,The Animal Husbandry Research Institute of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Peng Jia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zhuyue Wu
- The Animal Husbandry Research Institute of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Mingguang Huang
- The Animal Husbandry Research Institute of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Shaomei Chen
- The Animal Husbandry Research Institute of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jicai Zhang
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Bizhi Huang
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
37
|
Niu F, Sun K, Wei W, Yu C, Wei Z. F-actin disassembly factor MICAL1 binding to Myosin Va mediates cargo unloading during cytokinesis. SCIENCE ADVANCES 2020; 6:6/45/eabb1307. [PMID: 33158857 PMCID: PMC7673715 DOI: 10.1126/sciadv.abb1307] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 09/25/2020] [Indexed: 05/08/2023]
Abstract
Motor-mediated intracellular trafficking requires motors to position cargoes at proper locations. Myosin Va (MyoVa), an actin-based motor, is a classic model for studying cargo transport. However, the molecular basis underlying cargo unloading in MyoVa-mediated transport has remained enigmatic. We have identified MICAL1, an F-actin disassembly regulator, as a binding partner of MyoVa and shown that MICAL1-MyoVa interaction is critical for localization of MyoVa at the midbody. By binding to MICAL1, MyoVa-mediated transport is terminated, resulting in vesicle unloading at the midbody for efficient cytokinesis. The MyoVa/MICAL1 complex structure reveals that MICAL1 and F-actin assembly factors, Spires, share an overlapped binding surface on MyoVa, suggesting a regulatory role of F-actin dynamics in cargo unloading. Down-regulating F-actin disassembly by a MICAL1 mutant significantly reduces MyoVa and vesicles accumulating at the midbody. Collectively, our findings demonstrate that MyoVa binds to MICAL1 at the midbody destination and triggers F-actin disassembly to unload the vesicle cargo.
Collapse
Affiliation(s)
- Fengfeng Niu
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Kang Sun
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China
| | - Wenjie Wei
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Core Research Facilities, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Cong Yu
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China
| | - Zhiyi Wei
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
38
|
Xiang X, Qiu R. Cargo-Mediated Activation of Cytoplasmic Dynein in vivo. Front Cell Dev Biol 2020; 8:598952. [PMID: 33195284 PMCID: PMC7649786 DOI: 10.3389/fcell.2020.598952] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
Abstract
Cytoplasmic dynein-1 is a minus-end-directed microtubule motor that transports a variety of cargoes including early endosomes, late endosomes and other organelles. In many cell types, dynein accumulates at the microtubule plus end, where it interacts with its cargo to be moved toward the minus end. Dynein binds to its various cargoes via the dynactin complex and specific cargo adapters. Dynactin and some of the coiled-coil-domain-containing cargo adapters not only link dynein to cargo but also activate dynein motility, which implies that dynein is activated by its cellular cargo. Structural studies indicate that a dynein dimer switches between the autoinhibited phi state and an open state; and the binding of dynactin and a cargo adapter to the dynein tails causes the dynein motor domains to have a parallel configuration, allowing dynein to walk processively along a microtubule. Recently, the dynein regulator LIS1 has been shown to be required for dynein activation in vivo, and its mechanism of action involves preventing dynein from switching back to the autoinhibited state. In this review, we will discuss our current understanding of dynein activation and point out the gaps of knowledge on the spatial regulation of dynein in live cells. In addition, we will emphasize the importance of studying a complete set of dynein regulators for a better understanding of dynein regulation in vivo.
Collapse
Affiliation(s)
- Xin Xiang
- Department of Biochemistry and Molecular Biology, The Uniformed Services University of the Health Sciences - F. Edward Hébert School of Medicine, Bethesda, MD, United States
| | | |
Collapse
|
39
|
Platenkamp A, Detmar E, Sepulveda L, Ritz A, Rogers SL, Applewhite DA. The Drosophila melanogaster Rab GAP RN-tre cross-talks with the Rho1 signaling pathway to regulate nonmuscle myosin II localization and function. Mol Biol Cell 2020; 31:2379-2397. [PMID: 32816624 PMCID: PMC7851959 DOI: 10.1091/mbc.e20-03-0181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
To identify novel regulators of nonmuscle myosin II (NMII) we performed an image-based RNA interference screen using stable Drosophila melanogaster S2 cells expressing the enhanced green fluorescent protein (EGFP)-tagged regulatory light chain (RLC) of NMII and mCherry-Actin. We identified the Rab-specific GTPase-activating protein (GAP) RN-tre as necessary for the assembly of NMII RLC into contractile actin networks. Depletion of RN-tre led to a punctate NMII phenotype, similar to what is observed following depletion of proteins in the Rho1 pathway. Depletion of RN-tre also led to a decrease in active Rho1 and a decrease in phosphomyosin-positive cells by immunostaining, while expression of constitutively active Rho or Rho-kinase (Rok) rescues the punctate phenotype. Functionally, RN-tre depletion led to an increase in actin retrograde flow rate and cellular contractility in S2 and S2R+ cells, respectively. Regulation of NMII by RN-tre is only partially dependent on its GAP activity as overexpression of constitutively active Rabs inactivated by RN-tre failed to alter NMII RLC localization, while a GAP-dead version of RN-tre partially restored phosphomyosin staining. Collectively, our results suggest that RN-tre plays an important regulatory role in NMII RLC distribution, phosphorylation, and function, likely through Rho1 signaling and putatively serving as a link between the secretion machinery and actomyosin contractility.
Collapse
Affiliation(s)
| | - Elizabeth Detmar
- Department of Biology & Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280
| | - Liz Sepulveda
- Department of Biology, Reed College, Portland, OR 97202
| | - Anna Ritz
- Department of Biology, Reed College, Portland, OR 97202
| | - Stephen L Rogers
- Department of Biology & Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280
| | | |
Collapse
|
40
|
Zhang W, Gunst SJ. S100A4 is activated by RhoA and catalyses the polymerization of non-muscle myosin, adhesion complex assembly and contraction in airway smooth muscle. J Physiol 2020; 598:4573-4590. [PMID: 32767681 DOI: 10.1113/jp280111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 06/29/2020] [Indexed: 12/27/2022] Open
Abstract
KEY POINTS S100A4 is expressed in many tissues, including smooth muscle (SM), but its physiologic function is unknown. S100A4 regulates the motility of metastatic cancer cells by binding to non-muscle (NM) myosin II. Contractile stimulation causes the polymerization of NM myosin in airway SM, which is necessary for tension development. NM myosin regulates the assembly of adhesion junction signalling complexes (adhesomes) that catalyse actin polymerization. In airway SM, ACh (acetylcholine) stimulated the binding of S100A4 to the NM myosin heavy chain, which was catalysed by RhoA GTPase via the RhoA-binding protein, rhotekin. The binding of S100A4 to NM myosin was required for NM myosin polymerization, adhesome assembly and actin polymerization. S100A4 plays a critical function in the regulation of airway SM contraction by catalysing NM myosin filament assembly. The interaction of S100A4 with NM myosin may also play an important role in the physiologic function of other tissues. ABSTRACT S100A4 binds to the heavy chain of non-muscle (NM) myosin II and can regulate the motility of crawling cells. S100A4 is widely expressed in many tissues including smooth muscle (SM), although its role in the regulation of their physiologic function is not known. We hypothesized that S100A4 contributes to the regulation of contraction in airway SM by regulating a pool of NM myosin II at the cell cortex. NM myosin II undergoes polymerization in airway SM and regulates contraction by catalysing the assembly of integrin-associated adhesome complexes that activate pathways that catalyse actin polymerization. ACh stimulated the interaction of S100A4 with NM myosin II in airway SM at the cell cortex and catalysed NM myosin filament assembly. RhoA GTPase regulated the activation of S100A4 via rhotekin, which facilitated the formation of a complex between RhoA, S100A4 and NM myosin II. The depletion of S100A4, RhoA or rhotekin from airway SM tissues using short hairpin RNA or small interfering RNA prevented NM myosin II polymerization as well as the recruitment of vinculin and paxillin to adhesome signalling complexes in response to ACh, and inhibited actin polymerization and tension development. S100A4 depletion did not affect ACh-stimulated SM myosin regulatory light chain phosphorylation. The results show that S100A4 plays a critical role in tension development in airway SM tissue by catalysing NM myosin filament assembly, and that the interaction of S100A4 with NM myosin in response to contractile stimulation is activated by RhoA GTPase. These results may be broadly relevant to the physiologic function of S100A4 in other cell and tissue types.
Collapse
Affiliation(s)
- Wenwu Zhang
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Susan J Gunst
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
41
|
Costa AR, Sousa MM. Non-Muscle Myosin II in Axonal Cell Biology: From the Growth Cone to the Axon Initial Segment. Cells 2020; 9:cells9091961. [PMID: 32858875 PMCID: PMC7563147 DOI: 10.3390/cells9091961] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
By binding to actin filaments, non-muscle myosin II (NMII) generates actomyosin networks that hold unique contractile properties. Their dynamic nature is essential for neuronal biology including the establishment of polarity, growth cone formation and motility, axon growth during development (and axon regeneration in the adult), radial and longitudinal axonal tension, and synapse formation and function. In this review, we discuss the current knowledge on the spatial distribution and function of the actomyosin cytoskeleton in different axonal compartments. We highlight some of the apparent contradictions and open questions in the field, including the role of NMII in the regulation of axon growth and regeneration, the possibility that NMII structural arrangement along the axon shaft may control both radial and longitudinal contractility, and the mechanism and functional purpose underlying NMII enrichment in the axon initial segment. With the advances in live cell imaging and super resolution microscopy, it is expected that in the near future the spatial distribution of NMII in the axon, and the mechanisms by which it participates in axonal biology will be further untangled.
Collapse
|
42
|
Conventional and Non-Conventional Roles of Non-Muscle Myosin II-Actin in Neuronal Development and Degeneration. Cells 2020; 9:cells9091926. [PMID: 32825197 PMCID: PMC7566000 DOI: 10.3390/cells9091926] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Myosins are motor proteins that use chemical energy to produce mechanical forces driving actin cytoskeletal dynamics. In the brain, the conventional non-muscle myosin II (NMII) regulates actin filament cytoskeletal assembly and contractile forces during structural remodeling of axons and dendrites, contributing to morphology, polarization, and migration of neurons during brain development. NMII isoforms also participate in neurotransmission and synaptic plasticity by driving actin cytoskeletal dynamics during synaptic vesicle release and retrieval, and formation, maturation, and remodeling of dendritic spines. NMIIs are expressed differentially in cerebral non-neuronal cells, such as microglia, astrocytes, and endothelial cells, wherein they play key functions in inflammation, myelination, and repair. Besides major efforts to understand the physiological functions and regulatory mechanisms of NMIIs in the nervous system, their contributions to brain pathologies are still largely unclear. Nonetheless, genetic mutations or deregulation of NMII and its regulatory effectors are linked to autism, schizophrenia, intellectual disability, and neurodegeneration, indicating non-conventional roles of NMIIs in cellular mechanisms underlying neurodevelopmental and neurodegenerative disorders. Here, we summarize the emerging biological roles of NMIIs in the brain, and discuss how actomyosin signaling contributes to dysfunction of neurons and glial cells in the context of neurological disorders. This knowledge is relevant for a deep understanding of NMIIs on the pathogenesis and therapeutics of neuropsychiatric and neurodegenerative diseases.
Collapse
|
43
|
Myosin XVI in the Nervous System. Cells 2020; 9:cells9081903. [PMID: 32824179 PMCID: PMC7464383 DOI: 10.3390/cells9081903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/07/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022] Open
Abstract
The myosin family is a large inventory of actin-associated motor proteins that participate in a diverse array of cellular functions. Several myosin classes are expressed in neural cells and play important roles in neural functioning. A recently discovered member of the myosin superfamily, the vertebrate-specific myosin XVI (Myo16) class is expressed predominantly in neural tissues and appears to be involved in the development and proper functioning of the nervous system. Accordingly, the alterations of MYO16 has been linked to neurological disorders. Although the role of Myo16 as a generic actin-associated motor is still enigmatic, the N-, and C-terminal extensions that flank the motor domain seem to confer unique structural features and versatile interactions to the protein. Recent biochemical and physiological examinations portray Myo16 as a signal transduction element that integrates cell signaling pathways to actin cytoskeleton reorganization. This review discusses the current knowledge of the structure-function relation of Myo16. In light of its prevalent localization, the emphasis is laid on the neural aspects.
Collapse
|
44
|
Tang VW. Collagen, stiffness, and adhesion: the evolutionary basis of vertebrate mechanobiology. Mol Biol Cell 2020; 31:1823-1834. [PMID: 32730166 PMCID: PMC7525820 DOI: 10.1091/mbc.e19-12-0709] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/11/2020] [Accepted: 05/28/2020] [Indexed: 01/09/2023] Open
Abstract
The emergence of collagen I in vertebrates resulted in a dramatic increase in the stiffness of the extracellular environment, supporting long-range force propagation and the development of low-compliant tissues necessary for the development of vertebrate traits including pressurized circulation and renal filtration. Vertebrates have also evolved integrins that can bind to collagens, resulting in the generation of higher tension and more efficient force transmission in the extracellular matrix. The stiffer environment provides an opportunity for the vertebrates to create new structures such as the stress fibers, new cell types such as endothelial cells, new developmental processes such as neural crest delamination, and new tissue organizations such as the blood-brain barrier. Molecular players found only in vertebrates allow the modification of conserved mechanisms as well as the design of novel strategies that can better serve the physiological needs of the vertebrates. These innovations collectively contribute to novel morphogenetic behaviors and unprecedented increases in the complexities of tissue mechanics and functions.
Collapse
Affiliation(s)
- Vivian W. Tang
- Department of Cell and Developmental Biology, University of Illinois, Urbana–Champaign, Urbana, IL 61801
| |
Collapse
|
45
|
Alexandrova AY, Chikina AS, Svitkina TM. Actin cytoskeleton in mesenchymal-to-amoeboid transition of cancer cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:197-256. [PMID: 33066874 DOI: 10.1016/bs.ircmb.2020.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During development of metastasis, tumor cells migrate through different tissues and encounter different extracellular matrices. An ability of cells to adapt mechanisms of their migration to these diverse environmental conditions, called migration plasticity, gives tumor cells an advantage over normal cells for long distant dissemination. Different modes of individual cell motility-mesenchymal and amoeboid-are driven by different molecular mechanisms, which largely depend on functions of the actin cytoskeleton that can be modulated in a wide range by cellular signaling mechanisms in response to environmental conditions. Various triggers can switch one motility mode to another, but regulations of these transitions are incompletely understood. However, understanding of the mechanisms driving migration plasticity is instrumental for finding anti-cancer treatment capable to stop cancer metastasis. In this review, we discuss cytoskeletal features, which allow the individually migrating cells to switch between mesenchymal and amoeboid migrating modes, called mesenchymal-to-amoeboid transition (MAT). We briefly describe main characteristics of different cell migration modes, and then discuss the triggering factors that initiate MAT with special attention to cytoskeletal features essential for migration plasticity.
Collapse
Affiliation(s)
- Antonina Y Alexandrova
- Laboratory of Mechanisms of Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia.
| | - Aleksandra S Chikina
- Cell Migration and Invasion and Spatio-Temporal Regulation of Antigen Presentation teams, UMR144/U932 Institut Curie, Paris, France
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
46
|
Zhernov I, Diez S, Braun M, Lansky Z. Intrinsically Disordered Domain of Kinesin-3 Kif14 Enables Unique Functional Diversity. Curr Biol 2020; 30:3342-3351.e5. [PMID: 32649913 DOI: 10.1016/j.cub.2020.06.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/06/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022]
Abstract
In addition to their force-generating motor domains, kinesin motor proteins feature various accessory domains enabling them to fulfill a variety of functions in the cell. Human kinesin-3, Kif14, localizes to the midbody of the mitotic spindle and is involved in the progression of cytokinesis. The specific motor properties enabling Kif14's cellular functions, however, remain unknown. Here, we show in vitro that the intrinsically disordered N-terminal domain of Kif14 enables unique functional diversity of the kinesin. Using single molecule TIRF microscopy, we found that Kif14 exists either as a diffusible monomer or as processive dimer and that the disordered domain (1) enables diffusibility of the monomeric Kif14, (2) renders the dimeric Kif14 super-processive and enables the kinesin to pass through highly crowded areas, (3) enables robust, autonomous Kif14 tracking of growing microtubule tips, independent of microtubule end-binding (EB) proteins, and (4) is sufficient to enable crosslinking of parallel microtubules and necessary to enable Kif14-driven sliding of antiparallel ones. We explain these features of Kif14 by the observed diffusible interaction of the disordered domain with the microtubule lattice and the observed increased affinity of the disordered domain for GTP-bound tubulin. We suggest that the disordered domain tethers the motor domain to the microtubule providing a diffusible foothold and a regulatory hub, tuning the kinesin's interaction with microtubules. Our findings thus exemplify pliable protein tethering as a fundamental mechanism of molecular motor regulation.
Collapse
Affiliation(s)
- Ilia Zhernov
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Prague West, Czech Republic; Faculty of Mathematics and Physics, Charles University, Ke Karlovu 3, 121 16 Prague, Czech Republic
| | - Stefan Diez
- B CUBE - Center for Molecular Bioengineering, TU Dresden, Tatzberg 41, 01307 Dresden, Germany; Cluster of Excellence Physics of Life, TU Dresden, Tatzberg 47/49, 01307 Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstr. 108, Dresden 01307, Germany
| | - Marcus Braun
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Prague West, Czech Republic.
| | - Zdenek Lansky
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Prague West, Czech Republic.
| |
Collapse
|
47
|
Non-Muscle Myosin 2A (NM2A): Structure, Regulation and Function. Cells 2020; 9:cells9071590. [PMID: 32630196 PMCID: PMC7408548 DOI: 10.3390/cells9071590] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/30/2022] Open
Abstract
Non-muscle myosin 2A (NM2A) is a motor cytoskeletal enzyme with crucial importance from the early stages of development until adulthood. Due to its capacity to convert chemical energy into force, NM2A powers the contraction of the actomyosin cytoskeleton, required for proper cell division, adhesion and migration, among other cellular functions. Although NM2A has been extensively studied, new findings revealed that a lot remains to be discovered concerning its spatiotemporal regulation in the intracellular environment. In recent years, new functions were attributed to NM2A and its activity was associated to a plethora of illnesses, including neurological disorders and infectious diseases. Here, we provide a concise overview on the current knowledge regarding the structure, the function and the regulation of NM2A. In addition, we recapitulate NM2A-associated diseases and discuss its potential as a therapeutic target.
Collapse
|
48
|
Aguilar-Cuenca R, Llorente-González C, Chapman JR, Talayero VC, Garrido-Casado M, Delgado-Arévalo C, Millán-Salanova M, Shabanowitz J, Hunt DF, Sellers JR, Heissler SM, Vicente-Manzanares M. Tyrosine Phosphorylation of the Myosin Regulatory Light Chain Controls Non-muscle Myosin II Assembly and Function in Migrating Cells. Curr Biol 2020; 30:2446-2458.e6. [PMID: 32502416 DOI: 10.1016/j.cub.2020.04.057] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 03/04/2020] [Accepted: 04/22/2020] [Indexed: 10/24/2022]
Abstract
Active non-muscle myosin II (NMII) enables migratory cell polarization and controls dynamic cellular processes, such as focal adhesion formation and turnover and cell division. Filament assembly and force generation depend on NMII activation through the phosphorylation of Ser19 of the regulatory light chain (RLC). Here, we identify amino acid Tyr (Y) 155 of the RLC as a novel regulatory site that spatially controls NMII function. We show that Y155 is phosphorylated in vitro by the Tyr kinase domain of epidermal growth factor (EGF) receptor. In cells, phosphorylation of Y155, or its phospho-mimetic mutation (Glu), prevents the interaction of RLC with the myosin heavy chain (MHCII) to form functional NMII units. Conversely, Y155 mutation to a structurally similar but non-phosphorylatable amino acid (Phe) restores the more dynamic cellular functions of NMII, such as myosin filament formation and nascent adhesion assembly, but not those requiring stable actomyosin bundles, e.g., focal adhesion elongation or migratory front-back polarization. In live cells, phospho-Y155 RLC is prominently featured in protrusions, where it prevents NMII assembly. Our data indicate that Y155 phosphorylation constitutes a novel regulatory mechanism that contributes to the compartmentalization of NMII assembly and function in live cells.
Collapse
Affiliation(s)
- Rocío Aguilar-Cuenca
- Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; Universidad Autónoma de Madrid School of Medicine, 28006 Madrid, Spain
| | - Clara Llorente-González
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| | - Jessica R Chapman
- Department of Chemistry, University of Virginia, Charlottesville, VA 22903, USA
| | - Vanessa C Talayero
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| | - Marina Garrido-Casado
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| | - Cristina Delgado-Arévalo
- Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; Universidad Autónoma de Madrid School of Medicine, 28006 Madrid, Spain
| | - María Millán-Salanova
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| | - Jeffrey Shabanowitz
- Department of Chemistry, University of Virginia, Charlottesville, VA 22903, USA
| | - Donald F Hunt
- Department of Pathology, University of Virginia, Charlottesville, VA 22903, USA; Department of Chemistry, University of Virginia, Charlottesville, VA 22903, USA
| | - James R Sellers
- Cell Biology and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sarah M Heissler
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
49
|
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: 0.5] [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
|
50
|
Martin AC. The Physical Mechanisms of Drosophila Gastrulation: Mesoderm and Endoderm Invagination. Genetics 2020; 214:543-560. [PMID: 32132154 PMCID: PMC7054018 DOI: 10.1534/genetics.119.301292] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/21/2019] [Indexed: 12/14/2022] Open
Abstract
A critical juncture in early development is the partitioning of cells that will adopt different fates into three germ layers: the ectoderm, the mesoderm, and the endoderm. This step is achieved through the internalization of specified cells from the outermost surface layer, through a process called gastrulation. In Drosophila, gastrulation is achieved through cell shape changes (i.e., apical constriction) that change tissue curvature and lead to the folding of a surface epithelium. Folding of embryonic tissue results in mesoderm and endoderm invagination, not as individual cells, but as collective tissue units. The tractability of Drosophila as a model system is best exemplified by how much we know about Drosophila gastrulation, from the signals that pattern the embryo to the molecular components that generate force, and how these components are organized to promote cell and tissue shape changes. For mesoderm invagination, graded signaling by the morphogen, Spätzle, sets up a gradient in transcriptional activity that leads to the expression of a secreted ligand (Folded gastrulation) and a transmembrane protein (T48). Together with the GPCR Mist, which is expressed in the mesoderm, and the GPCR Smog, which is expressed uniformly, these signals activate heterotrimeric G-protein and small Rho-family G-protein signaling to promote apical contractility and changes in cell and tissue shape. A notable feature of this signaling pathway is its intricate organization in both space and time. At the cellular level, signaling components and the cytoskeleton exhibit striking polarity, not only along the apical-basal cell axis, but also within the apical domain. Furthermore, gene expression controls a highly choreographed chain of events, the dynamics of which are critical for primordium invagination; it does not simply throw the cytoskeletal "on" switch. Finally, studies of Drosophila gastrulation have provided insight into how global tissue mechanics and movements are intertwined as multiple tissues simultaneously change shape. Overall, these studies have contributed to the view that cells respond to forces that propagate over great distances, demonstrating that cellular decisions, and, ultimately, tissue shape changes, proceed by integrating cues across an entire embryo.
Collapse
Affiliation(s)
- Adam C Martin
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142
| |
Collapse
|