1
|
Jin L, Qin Y, Zhao Y, Zhou X, Zeng Y. Endothelial cytoskeleton in mechanotransduction and vascular diseases. J Biomech 2025; 182:112579. [PMID: 39938443 DOI: 10.1016/j.jbiomech.2025.112579] [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: 11/27/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
The cytoskeleton is an important structural component that regulates various aspects of cell morphology, movement, and intracellular signaling. It plays a pivotal role in the cellular response to biomechanical stimuli, particularly in endothelial cells, which are critical for vascular homeostasis and the pathogenesis of cardiovascular diseases. Mechanical forces, such as shear and tension, activate intracellular signaling cascades that regulate transcription, translation, and cellular behaviors. Despite extensive research into cytoskeletal functions, the precise mechanisms by which the cytoskeleton transduces mechanical signals remain incompletely understood. This review focuses on the role of cytoskeletal components in membrane, cytoplasm, and nucleus in mechanotransduction, with an emphasis on their structure, mechanical and biological behaviors, dynamic interactions, and response to mechanical forces. The collaboration between membrane cytoskeleton, cytoplasmic cytoskeleton, and nucleoskeleton is indispensable for endothelial cells to respond to mechanical stimuli. Understanding their mechanoresponsive mechanisms is essential for advancing therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Linlu Jin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Yixue Qin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Yunran Zhao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Xintong Zhou
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China.
| |
Collapse
|
2
|
Gawor M, Lehka L, Lambert D, Toseland CP. Actin from within - how nuclear myosins and actin regulate nuclear architecture and mechanics. J Cell Sci 2025; 138:JCS263550. [PMID: 39927755 PMCID: PMC11883275 DOI: 10.1242/jcs.263550] [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: 02/11/2025] Open
Abstract
Over the past two decades, significant progress has been made in understanding mechanotransduction to the nucleus. Nevertheless, most research has focused on outside-in signalling orchestrated by external mechanical stimuli. Emerging evidence highlights the importance of intrinsic nuclear mechanisms in the mechanoresponse. The discovery of actin and associated motor proteins, such as myosins, in the nucleus, along with advances in chromatin organisation research, has raised new questions about the contribution of intranuclear architecture and mechanics. Nuclear actin and myosins are present in various compartments of the nucleus, particularly at sites of DNA processing and modification. These proteins can function as hubs and scaffolds, cross-linking distant chromatin regions and thereby impacting local and global nuclear membrane shape. Importantly, nuclear myosins are force-sensitive and nuclear actin cooperates with mechanosensors, suggesting a multi-level contribution to nuclear mechanics. The crosstalk between nuclear myosins and actin has significant implications for cell mechanical plasticity and the prevention of pathological conditions. Here, we review the recent impactful findings that highlight the roles of nuclear actin and myosins in nuclear organisation. Additionally, we discuss potential links between these proteins and emphasize the importance of using new methodologies to unravel nuclear-derived regulatory mechanisms distinct from the cytoskeleton.
Collapse
Affiliation(s)
- Marta Gawor
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Lilya Lehka
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Danielle Lambert
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2RX, UK
| | - Christopher P. Toseland
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2RX, UK
| |
Collapse
|
3
|
Miladinović A, Antiga L, Venit T, Bayona-Hernandez A, Červenka J, Labala RK, Kolář M, Castaño E, Sztacho M, Hozák P. The perinucleolar compartment and the oncogenic super-enhancers are part of the same phase-separated structure filled with phosphatidylinositol 4,5-bisphosphate and long non-coding RNA HANR. Adv Biol Regul 2025; 95:101069. [PMID: 39648081 DOI: 10.1016/j.jbior.2024.101069] [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: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 12/10/2024]
Abstract
The liquid-liquid phase separation in the cell nucleus regulates various processes such as gene regulation and transcription control, chromatin organization, and DNA repair. A plethora of proteins and RNAs contribute to the formation of biomolecular condensates and recently, several nuclear phosphoinositides were shown to be a part of these membrane-less complexes within the nucleus as well. Here we lipid-interacting RNA sequencing (LIPRNAseq) and confocal microscopy to uncover the RNA-binding capacity and localization of phosphatidylinositol 4,5 bisphosphate (PIP2). We discovered the consensus PIP2-binding AU-rich RNA motif and identified long non-coding RNA HANR (lncHANR) to colocalize with PIP2 in the proximity to the nucleolus in the perinucleolar compartment (PNC). Colocalization studies with different nuclear markers reveal that PIP2-HANR presence in the PNC correlates with oncogenic super-enhancers, and both PNC and oncogenic enhancers are part of the same structure. As lncHANR, PNC, and oncogenic super-enhancers are associated with cancer cell lines and tumors, we suggest that they can serve as interchangeable prognostic markers. Understanding of the interplay between lipid metabolism, and lncRNAs in subnuclear compartment phase separation can lead to future improvement in treatment strategies and personalized cancer management approaches.
Collapse
Affiliation(s)
- Ana Miladinović
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Ludovica Antiga
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tomáš Venit
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Andrea Bayona-Hernandez
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Unidad de Biología Integrativa, Centro de Investigación Científica de Yucatán, Calle 43, Número 130, Chuburná de Hidalgo, Mérida, Yucatán, CP 97205, Mexico
| | - Jakub Červenka
- Laboratory of Proteomics, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, Prague, Czech Republic; Laboratory of Applied Proteome Analyses, Research Center PIGMOD, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - Rajendra Kumar Labala
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Michal Kolář
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Enrique Castaño
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Unidad de Biología Integrativa, Centro de Investigación Científica de Yucatán, Calle 43, Número 130, Chuburná de Hidalgo, Mérida, Yucatán, CP 97205, Mexico
| | - Martin Sztacho
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Laboratory of Cancer Cell Architecture, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Pavel Hozák
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
4
|
Morales PN, Coons AN, Koopman AJ, Patel S, Chase PB, Parvatiyar MS, Pinto JR. Post-translational modifications of vertebrate striated muscle myosin heavy chains. Cytoskeleton (Hoboken) 2024; 81:832-842. [PMID: 38587113 PMCID: PMC11458826 DOI: 10.1002/cm.21857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/06/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024]
Abstract
Post-translational modifications (PTMs) play a crucial role in regulating the function of many sarcomeric proteins, including myosin. Myosins comprise a family of motor proteins that play fundamental roles in cell motility in general and muscle contraction in particular. A myosin molecule consists of two myosin heavy chains (MyHCs) and two pairs of myosin light chains (MLCs); two MLCs are associated with the neck region of each MyHC's N-terminal head domain, while the two MyHC C-terminal tails form a coiled-coil that polymerizes with other MyHCs to form the thick filament backbone. Myosin undergoes extensive PTMs, and dysregulation of these PTMs may lead to abnormal muscle function and contribute to the development of myopathies and cardiovascular disorders. Recent studies have uncovered the significance of PTMs in regulating MyHC function and showed how these PTMs may provide additional modulation of contractile processes. Here, we discuss MyHC PTMs that have been biochemically and/or functionally studied in mammals' and rodents' striated muscle. We have identified hotspots or specific regions in three isoforms of myosin (MYH2, MYH6, and MYH7) where the prevalence of PTMs is more frequent and could potentially play a significant role in fine-tuning the activity of these proteins.
Collapse
Affiliation(s)
- Paula Nieto Morales
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306 USA
| | - Arianna N. Coons
- Department of Biological Science, Florida State University, Tallahassee, FL 32306 USA
| | - Amelia J. Koopman
- Department of Biological Science, Florida State University, Tallahassee, FL 32306 USA
| | - Sonu Patel
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306 USA
| | - P. Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306 USA
| | - Michelle S. Parvatiyar
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306 USA
| | - Jose R. Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306 USA
| |
Collapse
|
5
|
Solomatina ES, Nishkomaeva EN, Kovaleva AV, Tvorogova AV, Potashnikova DM, Saidova AA. Parameters of Cell Death and Proliferation of Prostate Cancer Cells with Altered Expression of Myosin 1C Isoforms. DOKL BIOCHEM BIOPHYS 2024; 514:16-22. [PMID: 38189886 PMCID: PMC11021239 DOI: 10.1134/s1607672923700588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 01/09/2024]
Abstract
Myosin 1C is a monomeric myosin motor with a truncated tail domain. Such motors are referred as slow "tension sensors." Three isoforms of myosin 1C differ in short N-termed amino acid sequences, the functional differences between isoforms have not been elucidated. Myosin 1C isoform A was described as a diagnostic marker for prostate cancer, but its role in tumor transformation remains unknown. Based on data on the functions of myosin 1C, we hypothesized the potential role of myosin 1C isoforms in maintaining the tumor phenotype of prostate cancer cells. In our work, we showed that a decrease in the expression level of myosin 1C isoform C leads to an increase in the proliferative activity of prostate tumor cells.
Collapse
Affiliation(s)
- E S Solomatina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - E N Nishkomaeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - A V Kovaleva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, Moscow State University, Moscow, Russia
| | - A V Tvorogova
- Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - D M Potashnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - A A Saidova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
- Faculty of Biology, Moscow State University, Moscow, Russia.
| |
Collapse
|
6
|
Venit T, Sapkota O, Abdrabou WS, Loganathan P, Pasricha R, Mahmood SR, El Said NH, Sherif S, Thomas S, Abdelrazig S, Amin S, Bedognetti D, Idaghdour Y, Magzoub M, Percipalle P. Positive regulation of oxidative phosphorylation by nuclear myosin 1 protects cells from metabolic reprogramming and tumorigenesis in mice. Nat Commun 2023; 14:6328. [PMID: 37816864 PMCID: PMC10564744 DOI: 10.1038/s41467-023-42093-w] [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: 08/04/2022] [Accepted: 09/29/2023] [Indexed: 10/12/2023] Open
Abstract
Metabolic reprogramming is one of the hallmarks of tumorigenesis. Here, we show that nuclear myosin 1 (NM1) serves as a key regulator of cellular metabolism. NM1 directly affects mitochondrial oxidative phosphorylation (OXPHOS) by regulating mitochondrial transcription factors TFAM and PGC1α, and its deletion leads to underdeveloped mitochondria inner cristae and mitochondrial redistribution within the cell. These changes are associated with reduced OXPHOS gene expression, decreased mitochondrial DNA copy number, and deregulated mitochondrial dynamics, which lead to metabolic reprogramming of NM1 KO cells from OXPHOS to aerobic glycolysis.This, in turn, is associated with a metabolomic profile typical for cancer cells, namely increased amino acid-, fatty acid-, and sugar metabolism, and increased glucose uptake, lactate production, and intracellular acidity. NM1 KO cells form solid tumors in a mouse model, suggesting that the metabolic switch towards aerobic glycolysis provides a sufficient carcinogenic signal. We suggest that NM1 plays a role as a tumor suppressor and that NM1 depletion may contribute to the Warburg effect at the onset of tumorigenesis.
Collapse
Affiliation(s)
- Tomas Venit
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | - Oscar Sapkota
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | - Wael Said Abdrabou
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
- Center for Genomics and Systems Biology, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | - Palanikumar Loganathan
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | - Renu Pasricha
- Core Technology Platforms, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | - Syed Raza Mahmood
- Center for Genomics and Systems Biology, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | - Nadine Hosny El Said
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | - Shimaa Sherif
- Translational Medicine Department, Research Branch, Sidra Medicine, Doha, Qatar
| | - Sneha Thomas
- Core Technology Platforms, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | - Salah Abdelrazig
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | - Shady Amin
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | - Davide Bedognetti
- Translational Medicine Department, Research Branch, Sidra Medicine, Doha, Qatar
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa, Genoa, Italy
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Youssef Idaghdour
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
- Center for Genomics and Systems Biology, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | - Mazin Magzoub
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates
| | - Piergiorgio Percipalle
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates.
- Center for Genomics and Systems Biology, New York University Abu Dhabi (NYUAD), P.O. Box, 129188, Abu Dhabi, United Arab Emirates.
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91, Stockholm, Sweden.
| |
Collapse
|
7
|
Balaban C, Sztacho M, Antiga L, Miladinović A, Harata M, Hozák P. PIP2-Effector Protein MPRIP Regulates RNA Polymerase II Condensation and Transcription. Biomolecules 2023; 13:biom13030426. [PMID: 36979361 PMCID: PMC10046169 DOI: 10.3390/biom13030426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
The specific post-translational modifications of the C-terminal domain (CTD) of the Rpb1 subunit of RNA polymerase II (RNAPII) correlate with different stages of transcription. The phosphorylation of the Ser5 residues of this domain associates with the initiation condensates, which are formed through liquid-liquid phase separation (LLPS). The subsequent Tyr1 phosphorylation of the CTD peaks at the promoter-proximal region and is involved in the pause-release of RNAPII. By implementing super-resolution microscopy techniques, we previously reported that the nuclear Phosphatidylinositol 4,5-bisphosphate (PIP2) associates with the Ser5-phosphorylated-RNAPII complex and facilitates the RNAPII transcription. In this study, we identified Myosin Phosphatase Rho-Interacting Protein (MPRIP) as a novel regulator of the RNAPII transcription that recruits Tyr1-phosphorylated CTD (Tyr1P-CTD) to nuclear PIP2-containing structures. The depletion of MPRIP increases the number of the initiation condensates, indicating a defect in the transcription. We hypothesize that MPRIP regulates the condensation and transcription through affecting the association of the RNAPII complex with nuclear PIP2-rich structures. The identification of Tyr1P-CTD as an interactor of PIP2 and MPRIP further points to a regulatory role in RNAPII pause-release, where the susceptibility of the transcriptional complex to leave the initiation condensate depends on its association with nuclear PIP2-rich structures. Moreover, the N-terminal domain of MPRIP, which is responsible for the interaction with the Tyr1P-CTD, contains an F-actin binding region that offers an explanation of how nuclear F-actin formations can affect the RNAPII transcription and condensation. Overall, our findings shed light on the role of PIP2 in RNAPII transcription through identifying the F-actin binding protein MPRIP as a transcription regulator and a determinant of the condensation of RNAPII.
Collapse
Affiliation(s)
- Can Balaban
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Martin Sztacho
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
- Correspondence: (M.S.); (P.H.)
| | - Ludovica Antiga
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Ana Miladinović
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Masahiko Harata
- Laboratory of Molecular Biochemistry, Division of Life Science, Graduate School of Agricultural Science, Tohoku University, 468-1, Aramaki Aza Aoba, Aoba-ku, Sendai 980-0845, Japan
| | - Pavel Hozák
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
- Correspondence: (M.S.); (P.H.)
| |
Collapse
|
8
|
Matozo T, Kogachi L, de Alencar BC. Myosin motors on the pathway of viral infections. Cytoskeleton (Hoboken) 2022; 79:41-63. [PMID: 35842902 DOI: 10.1002/cm.21718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/25/2022] [Accepted: 07/07/2022] [Indexed: 01/30/2023]
Abstract
Molecular motors are microscopic machines that use energy from adenosine triphosphate (ATP) hydrolysis to generate movement. While kinesins and dynein are molecular motors associated with microtubule tracks, myosins bind to and move on actin filaments. Mammalian cells express several myosin motors. They power cellular processes such as endo- and exocytosis, intracellular trafficking, transcription, migration, and cytokinesis. As viruses navigate through cells, they may take advantage or be hindered by host components and machinery, including the cytoskeleton. This review delves into myosins' cell roles and compares them to their reported functions in viral infections. In most cases, the previously described myosin functions align with their reported role in viral infections, although not in all cases. This opens the possibility that knowledge obtained from studying myosins in viral infections might shed light on new physiological roles for myosins in cells. However, given the high number of myosins expressed and the variety of viruses investigated in the different studies, it is challenging to infer whether the interactions found are specific to a single virus or can be applied to other viruses with the same characteristics. We conclude that the participation of myosins in viral cycles is still a largely unexplored area, especially concerning unconventional myosins.
Collapse
Affiliation(s)
- Tais Matozo
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Leticia Kogachi
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bruna Cunha de Alencar
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
9
|
Understanding the key functions of Myosins in viral infection. Biochem Soc Trans 2022; 50:597-607. [PMID: 35212367 DOI: 10.1042/bst20211239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/10/2022] [Accepted: 02/15/2022] [Indexed: 11/17/2022]
Abstract
Myosins, a class of actin-based motor proteins existing in almost any organism, are originally considered only involved in driving muscle contraction, reshaping actin cytoskeleton, and anchoring or transporting cargoes, including protein complexes, organelles, vesicles. However, accumulating evidence reveals that myosins also play vital roles in viral infection, depending on viral species and infection stages. This review systemically summarizes the described various myosins, the performed functions, and the involved mechanisms or molecular pathways during viral infection. Meanwhile, the existing issues are also discussed. Additionally, the important technologies or agents, including siRNA, gene editing, and myosin inhibitors, would facilitate dissecting the actions and mechanisms for described and undescribed myosins, which could be adopted to prevent or control viral infection are also characterized.
Collapse
|
10
|
Horníková L, Bruštíková K, Huérfano S, Forstová J. Nuclear Cytoskeleton in Virus Infection. Int J Mol Sci 2022; 23:ijms23010578. [PMID: 35009004 PMCID: PMC8745530 DOI: 10.3390/ijms23010578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 02/04/2023] Open
Abstract
The nuclear lamina is the main component of the nuclear cytoskeleton that maintains the integrity of the nucleus. However, it represents a natural barrier for viruses replicating in the cell nucleus. The lamina blocks viruses from being trafficked to the nucleus for replication, but it also impedes the nuclear egress of the progeny of viral particles. Thus, viruses have evolved mechanisms to overcome this obstacle. Large viruses induce the assembly of multiprotein complexes that are anchored to the inner nuclear membrane. Important components of these complexes are the viral and cellular kinases phosphorylating the lamina and promoting its disaggregation, therefore allowing virus egress. Small viruses also use cellular kinases to induce lamina phosphorylation and the subsequent disruption in order to facilitate the import of viral particles during the early stages of infection or during their nuclear egress. Another component of the nuclear cytoskeleton, nuclear actin, is exploited by viruses for the intranuclear movement of their particles from the replication sites to the nuclear periphery. This study focuses on exploitation of the nuclear cytoskeleton by viruses, although this is just the beginning for many viruses, and promises to reveal the mechanisms and dynamic of physiological and pathological processes in the nucleus.
Collapse
|
11
|
Goelzer M, Goelzer J, Ferguson ML, Neu CP, Uzer G. Nuclear envelope mechanobiology: linking the nuclear structure and function. Nucleus 2021; 12:90-114. [PMID: 34455929 PMCID: PMC8432354 DOI: 10.1080/19491034.2021.1962610] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 01/10/2023] Open
Abstract
The nucleus, central to cellular activity, relies on both direct mechanical input as well as its molecular transducers to sense external stimuli and respond by regulating intra-nuclear chromatin organization that determines cell function and fate. In mesenchymal stem cells of musculoskeletal tissues, changes in nuclear structures are emerging as a key modulator of their differentiation and proliferation programs. In this review we will first introduce the structural elements of the nucleoskeleton and discuss the current literature on how nuclear structure and signaling are altered in relation to environmental and tissue level mechanical cues. We will focus on state-of-the-art techniques to apply mechanical force and methods to measure nuclear mechanics in conjunction with DNA, RNA, and protein visualization in living cells. Ultimately, combining real-time nuclear deformations and chromatin dynamics can be a powerful tool to study mechanisms of how forces affect the dynamics of genome function.
Collapse
Affiliation(s)
- Matthew Goelzer
- Materials Science and Engineering, Boise State University, Boise, ID, US
| | | | - Matthew L. Ferguson
- Biomolecular Science, Boise State University, Boise, ID, US
- Physics, Boise State University, Boise, ID, US
| | - Corey P. Neu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado, Boulder, CO, US
| | - Gunes Uzer
- Mechanical and Biomedical Engineering, Boise State University, Boise, ID, US
| |
Collapse
|
12
|
Sharif SB, Zamani N, Chadwick BP. BAZ1B the Protean Protein. Genes (Basel) 2021; 12:genes12101541. [PMID: 34680936 PMCID: PMC8536118 DOI: 10.3390/genes12101541] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 02/02/2023] Open
Abstract
The bromodomain adjacent to the zinc finger domain 1B (BAZ1B) or Williams syndrome transcription factor (WSTF) are just two of the names referring the same protein that is encoded by the WBSCR9 gene and is among the 26-28 genes that are lost from one copy of 7q11.23 in Williams syndrome (WS: OMIM 194050). Patients afflicted by this contiguous gene deletion disorder present with a range of symptoms including cardiovascular complications, developmental defects as well as a characteristic cognitive and behavioral profile. Studies in patients with atypical deletions and mouse models support BAZ1B hemizygosity as a contributing factor to some of the phenotypes. Focused analysis on BAZ1B has revealed this to be a versatile nuclear protein with a central role in chromatin remodeling through two distinct complexes as well as being involved in the replication and repair of DNA, transcriptional processes involving RNA Polymerases I, II, and III as well as possessing kinase activity. Here, we provide a comprehensive review to summarize the many aspects of BAZ1B function including its recent link to cancer.
Collapse
Affiliation(s)
- Shahin Behrouz Sharif
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA;
| | - Nina Zamani
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA;
| | - Brian P. Chadwick
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA;
- Correspondence:
| |
Collapse
|
13
|
Record J, Saeed MB, Venit T, Percipalle P, Westerberg LS. Journey to the Center of the Cell: Cytoplasmic and Nuclear Actin in Immune Cell Functions. Front Cell Dev Biol 2021; 9:682294. [PMID: 34422807 PMCID: PMC8375500 DOI: 10.3389/fcell.2021.682294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Actin cytoskeletal dynamics drive cellular shape changes, linking numerous cell functions to physiological and pathological cues. Mutations in actin regulators that are differentially expressed or enriched in immune cells cause severe human diseases known as primary immunodeficiencies underscoring the importance of efficienct actin remodeling in immune cell homeostasis. Here we discuss recent findings on how immune cells sense the mechanical properties of their environement. Moreover, while the organization and biochemical regulation of cytoplasmic actin have been extensively studied, nuclear actin reorganization is a rapidly emerging field that has only begun to be explored in immune cells. Based on the critical and multifaceted contributions of cytoplasmic actin in immune cell functionality, nuclear actin regulation is anticipated to have a large impact on our understanding of immune cell development and functionality.
Collapse
Affiliation(s)
- Julien Record
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Mezida B. Saeed
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Tomas Venit
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
| | - Piergiorgio Percipalle
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Lisa S. Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
14
|
Venit T, El Said NH, Mahmood SR, Percipalle P. A dynamic actin-dependent nucleoskeleton and cell identity. J Biochem 2021; 169:243-257. [PMID: 33351909 DOI: 10.1093/jb/mvaa133] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/27/2020] [Indexed: 12/19/2022] Open
Abstract
Actin is an essential regulator of cellular functions. In the eukaryotic cell nucleus, actin regulates chromatin as a bona fide component of chromatin remodelling complexes, it associates with nuclear RNA polymerases to regulate transcription and is involved in co-transcriptional assembly of nascent RNAs into ribonucleoprotein complexes. Actin dynamics are, therefore, emerging as a major regulatory factor affecting diverse cellular processes. Importantly, the involvement of actin dynamics in nuclear functions is redefining the concept of nucleoskeleton from a rigid scaffold to a dynamic entity that is likely linked to the three-dimensional organization of the nuclear genome. In this review, we discuss how nuclear actin, by regulating chromatin structure through phase separation may contribute to the architecture of the nuclear genome during cell differentiation and facilitate the expression of specific gene programs. We focus specifically on mitochondrial genes and how their dysregulation in the absence of actin raises important questions about the role of cytoskeletal proteins in regulating chromatin structure. The discovery of a novel pool of mitochondrial actin that serves as 'mitoskeleton' to facilitate organization of mtDNA supports a general role for actin in genome architecture and a possible function of distinct actin pools in the communication between nucleus and mitochondria.
Collapse
Affiliation(s)
- Tomas Venit
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), PO Box 129188, Abu Dhabi United Arab Emirates
| | - Nadine Hosny El Said
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), PO Box 129188, Abu Dhabi United Arab Emirates
| | - Syed Raza Mahmood
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), PO Box 129188, Abu Dhabi United Arab Emirates.,Department of Biology, New York University, 100 Washington Square East, 1009 Silver Center, New York, NY 10003, USA
| | - Piergiorgio Percipalle
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), PO Box 129188, Abu Dhabi United Arab Emirates.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Svante Arrhenius väg 20C, 114 18 Stockholm, Sweden
| |
Collapse
|
15
|
Miyamoto K, Harata M. Nucleoskeleton proteins for nuclear dynamics. J Biochem 2021; 169:237-241. [PMID: 33479767 DOI: 10.1093/jb/mvab006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
The eukaryotic nucleus shows organized structures of chromosomes, transcriptional components and their associated proteins. It has been believed that such a dense nuclear environment prevents the formation of a cytoskeleton-like network of protein filaments. However, accumulating evidence suggests that the cell nucleus also possesses structural filamentous components to support nuclear organization and compartments, which are referred to as nucleoskeleton proteins. Nucleoskeleton proteins including lamins and actin influence nuclear dynamics including transcriptional regulation, chromatin organization and DNA damage responses. Furthermore, these nucleoskeleton proteins play a pivotal role in cellular differentiation and animal development. In this commentary, we discuss how nucleoskeleton-based regulatory mechanisms orchestrate nuclear dynamics.
Collapse
Affiliation(s)
- Kei Miyamoto
- Graduate School of Biology-Oriented Science and Technology, Kindai University, 930 Nishimitani, Kinokawa-shi, Wakayama 649-6493, Japan
| | - Masahiko Harata
- Laboratory of Molecular Biology, Graduate School of Agricultural Science, Tohoku University, Aramaki Aza-Aoba 468-1, Aoba-ku, Sendai 980-0845, Japan
| |
Collapse
|
16
|
Cook AW, Toseland CP. The roles of nuclear myosin in the DNA damage response. J Biochem 2021; 169:265-271. [PMID: 33035317 DOI: 10.1093/jb/mvaa113] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023] Open
Abstract
Myosin within the nucleus has often been overlooked due to their importance in cytoplasmic processes and a lack of investigation. However, more recently, it has been shown that their nuclear roles are just as fundamental to cell function and survival with roles in transcription, DNA damage and viral replication. Myosins can act as molecular transporters and anchors that rely on their actin binding and ATPase capabilities. Their roles within the DNA damage response can varies from a transcriptional response, moving chromatin and stabilizing chromosome contacts. This review aims to highlight their key roles in the DNA damage response and how they impact nuclear organization and transcription.
Collapse
Affiliation(s)
- Alexander W Cook
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | | |
Collapse
|
17
|
The F-Actin-Binding MPRIP Forms Phase-Separated Condensates and Associates with PI(4,5)P2 and Active RNA Polymerase II in the Cell Nucleus. Cells 2021; 10:cells10040848. [PMID: 33918018 PMCID: PMC8068864 DOI: 10.3390/cells10040848] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/25/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Here, we provide evidence for the presence of Myosin phosphatase rho-interacting protein (MPRIP), an F-actin-binding protein, in the cell nucleus. The MPRIP protein binds to Phosphatidylinositol 4,5-bisphosphate (PIP2) and localizes to the nuclear speckles and nuclear lipid islets which are known to be involved in transcription. We identified MPRIP as a component of RNA Polymerase II/Nuclear Myosin 1 complex and showed that MPRIP forms phase-separated condensates which are able to bind nuclear F-actin fibers. Notably, the fibrous MPRIP preserves its liquid-like properties and reforms the spherical shaped condensates when F-actin is disassembled. Moreover, we show that the phase separation of MPRIP is driven by its long intrinsically disordered region at the C-terminus. We propose that the PIP2/MPRIP association might contribute to the regulation of RNAPII transcription via phase separation and nuclear actin polymerization.
Collapse
|
18
|
Limited Proteolysis-Coupled Mass Spectrometry Identifies Phosphatidylinositol 4,5-Bisphosphate Effectors in Human Nuclear Proteome. Cells 2021; 10:cells10010068. [PMID: 33406800 PMCID: PMC7824793 DOI: 10.3390/cells10010068] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/23/2020] [Accepted: 12/29/2020] [Indexed: 12/20/2022] Open
Abstract
Specific nuclear sub-compartments that are regions of fundamental processes such as gene expression or DNA repair, contain phosphoinositides (PIPs). PIPs thus potentially represent signals for the localization of specific proteins into different nuclear functional domains. We performed limited proteolysis followed by label-free quantitative mass spectrometry and identified nuclear protein effectors of the most abundant PIP—phosphatidylinositol 4,5-bisphosphate (PIP2). We identified 515 proteins with PIP2-binding capacity of which 191 ‘exposed’ proteins represent a direct PIP2 interactors and 324 ‘hidden’ proteins, where PIP2 binding was increased upon trypsin treatment. Gene ontology analysis revealed that ‘exposed’ proteins are involved in the gene expression as regulators of Pol II, mRNA splicing, and cell cycle. They localize mainly to non-membrane bound organelles—nuclear speckles and nucleolus and are connected to the actin nucleoskeleton. ‘Hidden’ proteins are linked to the gene expression, RNA splicing and transport, cell cycle regulation, and response to heat or viral infection. These proteins localize to the nuclear envelope, nuclear pore complex, or chromatin. Bioinformatic analysis of peptides bound in both groups revealed that PIP2-binding motifs are in general hydrophilic. Our data provide an insight into the molecular mechanism of nuclear PIP2 protein interaction and advance the methodology applicable for further studies of PIPs or other protein ligands.
Collapse
|
19
|
Giese S, Reindl T, Reinke PYA, Zattelman L, Fedorov R, Henn A, Taft MH, Manstein DJ. Mechanochemical properties of human myosin-1C are modulated by isoform-specific differences in the N-terminal extension. J Biol Chem 2020; 296:100128. [PMID: 33257319 PMCID: PMC7948490 DOI: 10.1074/jbc.ra120.015187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 11/24/2022] Open
Abstract
Myosin-1C is a single-headed, short-tailed member of the myosin class I subfamily that supports a variety of actin-based functions in the cytosol and nucleus. In vertebrates, alternative splicing of the MYO1C gene leads to the production of three isoforms, myosin-1C0, myosin-1C16, and myosin-1C35, that carry N-terminal extensions of different lengths. However, it is not clear how these extensions affect the chemomechanical coupling of human myosin-1C isoforms. Here, we report on the motor activity of the different myosin-1C isoforms measuring the unloaded velocities of constructs lacking the C-terminal lipid-binding domain on nitrocellulose-coated glass surfaces and full-length constructs on reconstituted, supported lipid bilayers. The higher yields of purified proteins obtained with constructs lacking the lipid-binding domain allowed a detailed characterization of the individual kinetic steps of human myosin-1C isoforms in their productive interaction with nucleotides and filamentous actin. Isoform-specific differences include 18-fold changes in the maximum power output per myosin-1C motor and 4-fold changes in the velocity and the resistive force at which maximum power output occurs. Our results support a model in which the isoform-specific N-terminal extensions affect chemomechanical coupling by combined steric and allosteric effects, thereby reducing both the length of the working stroke and the rate of ADP release in the absence of external loads by a factor of 2 for myosin-1C35. As the large change in maximum power output shows, the functional differences between the isoforms are further amplified by the presence of external loads.
Collapse
Affiliation(s)
- Sven Giese
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany
| | - Theresia Reindl
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany
| | - Patrick Y A Reinke
- Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
| | - Lilach Zattelman
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel; Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Roman Fedorov
- Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
| | - Arnon Henn
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel; Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Manuel H Taft
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany.
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, Fritz-Hartmann-Centre for Medical Research, Hannover Medical School, Hannover, Germany; Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
20
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
21
|
Abstract
The presence of actin in the nucleus has historically been a highly contentious issue. It is now, however, well accepted that actin has physiologically important roles in the nucleus. In this Review, we describe the evolution of our thinking about actin in the nucleus starting with evidence supporting its involvement in transcription, chromatin remodeling and intranuclear movements. We also review the growing literature on the mechanisms that regulate the import and export of actin and how post-translational modifications of actin could regulate nuclear actin. We end with an extended discussion of the role of nuclear actin in the repair of DNA double stranded breaks.
Collapse
Affiliation(s)
- Leonid Serebryannyy
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Primal de Lanerolle
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, United States.
| |
Collapse
|
22
|
Cook AW, Gough RE, Toseland CP. Nuclear myosins - roles for molecular transporters and anchors. J Cell Sci 2020; 133:133/11/jcs242420. [PMID: 32499319 DOI: 10.1242/jcs.242420] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The myosin family of molecular motors are well-characterised cytoskeletal proteins. However, myosins are also present in the nucleus, where they have been shown to have roles in transcription, DNA repair and viral infections. Despite their involvement in these fundamental cellular processes, our understanding of these functions and their regulation remains limited. Recently, research on nuclear myosins has been gathering pace, and this Review will evaluate the current state of the field. Here, we will focus on the variation in structure of nuclear myosins, their nuclear import and their roles within transcription, DNA damage, chromatin organisation and viral infections. We will also consider both the biochemical and biophysical properties and restraints that are placed on these multifunctional motors, and how they link to their cytoplasmic counterparts. By highlighting these properties and processes, we show just how integral nuclear myosins are for cellular survival.
Collapse
Affiliation(s)
- Alexander W Cook
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | - Rosemarie E Gough
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | | |
Collapse
|
23
|
Pradhan R, Nallappa MJ, Sengupta K. Lamin A/C modulates spatial organization and function of the Hsp70 gene locus via nuclear myosin I. J Cell Sci 2020; 133:jcs236265. [PMID: 31988151 DOI: 10.1242/jcs.236265] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 01/13/2020] [Indexed: 02/06/2023] Open
Abstract
The structure-function relationship of the nucleus is tightly regulated, especially during heat shock. Typically, heat shock activates molecular chaperones that prevent protein misfolding and preserve genome integrity. However, the molecular mechanisms that regulate nuclear structure-function relationships during heat shock remain unclear. Here, we show that lamin A and C (hereafter lamin A/C; both lamin A and C are encoded by LMNA) are required for heat-shock-mediated transcriptional induction of the Hsp70 gene locus (HSPA genes). Interestingly, lamin A/C regulates redistribution of nuclear myosin I (NM1) into the nucleus upon heat shock, and depletion of either lamin A/C or NM1 abrogates heat-shock-induced repositioning of Hsp70 gene locus away from the nuclear envelope. Lamins and NM1 also regulate spatial positioning of the SC35 (also known as SRSF2) speckles - important nuclear landmarks that modulates Hsp70 gene locus expression upon heat shock. This suggests an intricate crosstalk between nuclear lamins, NM1 and SC35 organization in modulating transcriptional responses of the Hsp70 gene locus during heat shock. Taken together, this study unravels a novel role for lamin A/C in the regulation of the spatial dynamics and function of the Hsp70 gene locus upon heat shock, via the nuclear motor protein NM1.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Roopali Pradhan
- Biology, Main Building, First Floor, Room B-216, Indian Institute of Science Education and Research (IISER), Pune 411008, India
| | - Muhunden Jayakrishnan Nallappa
- Biology, Main Building, First Floor, Room B-216, Indian Institute of Science Education and Research (IISER), Pune 411008, India
| | - Kundan Sengupta
- Biology, Main Building, First Floor, Room B-216, Indian Institute of Science Education and Research (IISER), Pune 411008, India
| |
Collapse
|
24
|
|
25
|
Venit T, Mahmood SR, Endara-Coll M, Percipalle P. Nuclear actin and myosin in chromatin regulation and maintenance of genome integrity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 355:67-108. [DOI: 10.1016/bs.ircmb.2020.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
|
26
|
Ranade D, Pradhan R, Jayakrishnan M, Hegde S, Sengupta K. Lamin A/C and Emerin depletion impacts chromatin organization and dynamics in the interphase nucleus. BMC Mol Cell Biol 2019; 20:11. [PMID: 31117946 PMCID: PMC6532135 DOI: 10.1186/s12860-019-0192-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 04/16/2019] [Indexed: 12/26/2022] Open
Abstract
Background Nuclear lamins are type V intermediate filament proteins that maintain nuclear structure and function. Furthermore, Emerin - an interactor of Lamin A/C, facilitates crosstalk between the cytoskeleton and the nucleus as it also interacts with actin and Nuclear Myosin 1 (NM1). Results Here we show that the depletion of Lamin A/C or Emerin, alters the localization of the nuclear motor protein - Nuclear Myosin 1 (NM1) that manifests as an increase in NM1 foci in the nucleus and are rescued to basal levels upon the combined knockdown of Lamin A/C and Emerin. Furthermore, Lamin A/C-Emerin co-depletion destabilizes cytoskeletal organization as it increases actin stress fibers. This further impinges on nuclear organization, as it enhances chromatin mobility more toward the nuclear interior in Lamin A/C-Emerin co-depleted cells. This enhanced chromatin mobility was restored to basal levels either upon inhibition of Nuclear Myosin 1 (NM1) activity or actin depolymerization. In addition, the combined loss of Lamin A/C and Emerin alters the otherwise highly conserved spatial positions of chromosome territories. Furthermore, knockdown of Lamin A/C or Lamin A/C-Emerin combined, deregulates expression levels of a candidate subset of genes. Amongst these genes, both KLK10 (Chr.19, Lamina Associated Domain (LAD+)) and MADH2 (Chr.18, LAD-) were significantly repressed, while BCL2L12 (Chr.19, LAD-) is de-repressed. These genes differentially reposition with respect to the nuclear envelope. Conclusions Taken together, these studies underscore a remarkable interplay between Lamin A/C and Emerin in modulating cytoskeletal organization of actin and NM1 that impinges on chromatin dynamics and function in the interphase nucleus. Electronic supplementary material The online version of this article (10.1186/s12860-019-0192-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Devika Ranade
- Indian Institute of Science Education and Research (IISER)-Pune, Dr. Homi Bhabha Road, Biology, Room#B-216, 1st Floor, Main Building, Pashan, Pune, Maharashtra, 411008, India
| | - Roopali Pradhan
- Indian Institute of Science Education and Research (IISER)-Pune, Dr. Homi Bhabha Road, Biology, Room#B-216, 1st Floor, Main Building, Pashan, Pune, Maharashtra, 411008, India
| | - Muhunden Jayakrishnan
- Indian Institute of Science Education and Research (IISER)-Pune, Dr. Homi Bhabha Road, Biology, Room#B-216, 1st Floor, Main Building, Pashan, Pune, Maharashtra, 411008, India
| | - Sushmitha Hegde
- Indian Institute of Science Education and Research (IISER)-Pune, Dr. Homi Bhabha Road, Biology, Room#B-216, 1st Floor, Main Building, Pashan, Pune, Maharashtra, 411008, India
| | - Kundan Sengupta
- Indian Institute of Science Education and Research (IISER)-Pune, Dr. Homi Bhabha Road, Biology, Room#B-216, 1st Floor, Main Building, Pashan, Pune, Maharashtra, 411008, India.
| |
Collapse
|
27
|
Maintenance of epigenetic landscape requires CIZ1 and is corrupted in differentiated fibroblasts in long-term culture. Nat Commun 2019; 10:460. [PMID: 30692537 PMCID: PMC6484225 DOI: 10.1038/s41467-018-08072-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 12/04/2018] [Indexed: 01/09/2023] Open
Abstract
The inactive X chromosome (Xi) serves as a model for establishment and maintenance of repressed chromatin and the function of polycomb repressive complexes (PRC1/2). Here we show that Xi transiently relocates from the nuclear periphery towards the interior during its replication, in a process dependent on CIZ1. Compromised relocation of Xi in CIZ1-null primary mouse embryonic fibroblasts is accompanied by loss of PRC-mediated H2AK119Ub1 and H3K27me3, increased solubility of PRC2 catalytic subunit EZH2, and genome-wide deregulation of polycomb-regulated genes. Xi position in S phase is also corrupted in cells adapted to long-term culture (WT or CIZ1-null), and also accompanied by specific changes in EZH2 and its targets. The data are consistent with the idea that chromatin relocation during S phase contributes to maintenance of epigenetic landscape in primary cells, and that elevated soluble EZH2 is part of an error-prone mechanism by which modifying enzyme meets template when chromatin relocation is compromised. The inactive X chromosome (Xi) is a model for establishment and maintenance of repressed chromatin and the function of polycomb repressive complexes. Here the authors show that Xi transiently relocates from the nuclear periphery during replication in a CIZ1-dependent manner, which plays a role in maintaining PRC-mediated repressed chromatin.
Collapse
|
28
|
Kelpsch DJ, Tootle TL. Nuclear Actin: From Discovery to Function. Anat Rec (Hoboken) 2018; 301:1999-2013. [PMID: 30312531 PMCID: PMC6289869 DOI: 10.1002/ar.23959] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/20/2018] [Accepted: 05/14/2018] [Indexed: 01/02/2023]
Abstract
While actin was discovered in the nucleus over 50 years ago, research lagged for decades due to strong skepticism. The revitalization of research into nuclear actin occurred after it was found that cellular stresses induce the nuclear localization and alter the structure of actin. These studies provided the first hints that actin has a nuclear function. Subsequently, it was established that the nuclear import and export of actin is highly regulated. While the structures of nuclear actin remain unclear, it can function as monomers, polymers, and even rods. Furthermore, even within a given structure, distinct pools of nuclear actin that can be differentially labeled have been identified. Numerous mechanistic studies have uncovered an array of functions for nuclear actin. It regulates the activity of RNA polymerases, as well as specific transcription factors. Actin also modulates the activity of several chromatin remodeling complexes and histone deacetylases, to ultimately impinge on transcriptional programing and DNA damage repair. Further, nuclear actin mediates chromatin movement and organization. It has roles in meiosis and mitosis, and these functions may be functionally conserved from ancient bacterial actin homologs. The structure and integrity of the nuclear envelope and sub-nuclear compartments are also regulated by nuclear actin. Furthermore, nuclear actin contributes to human diseases like cancer, neurodegeneration, and myopathies. Here, we explore the early discovery of actin in the nucleus and discuss the forms and functions of nuclear actin in both normal and disease contexts. Anat Rec, 301:1999-2013, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Daniel J. Kelpsch
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, 51 Newton Rd, 1-500 BSB, Iowa City, IA 52242
| | - Tina L. Tootle
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, 51 Newton Rd, 1-500 BSB, Iowa City, IA 52242
| |
Collapse
|
29
|
An actin-based nucleoskeleton involved in gene regulation and genome organization. Biochem Biophys Res Commun 2018; 506:378-386. [DOI: 10.1016/j.bbrc.2017.11.206] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 11/30/2017] [Indexed: 12/21/2022]
|
30
|
Calcium and Nuclear Signaling in Prostate Cancer. Int J Mol Sci 2018; 19:ijms19041237. [PMID: 29671777 PMCID: PMC5979488 DOI: 10.3390/ijms19041237] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/15/2018] [Accepted: 04/17/2018] [Indexed: 02/06/2023] Open
Abstract
Recently, there have been a number of developments in the fields of calcium and nuclear signaling that point to new avenues for a more effective diagnosis and treatment of prostate cancer. An example is the discovery of new classes of molecules involved in calcium-regulated nuclear import and nuclear calcium signaling, from the G protein-coupled receptor (GPCR) and myosin families. This review surveys the new state of the calcium and nuclear signaling fields with the aim of identifying the unifying themes that hold out promise in the context of the problems presented by prostate cancer. Genomic perturbations, kinase cascades, developmental pathways, and channels and transporters are covered, with an emphasis on nuclear transport and functions. Special attention is paid to the molecular mechanisms behind prostate cancer progression to the malignant forms and the unfavorable response to anti-androgen treatment. The survey leads to some new hypotheses that connect heretofore disparate results and may present a translational interest.
Collapse
|
31
|
Nevzorov I, Sidorenko E, Wang W, Zhao H, Vartiainen MK. Myosin-1C uses a novel phosphoinositide-dependent pathway for nuclear localization. EMBO Rep 2018; 19:290-304. [PMID: 29330316 PMCID: PMC5797967 DOI: 10.15252/embr.201744296] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 12/05/2017] [Accepted: 12/11/2017] [Indexed: 01/07/2023] Open
Abstract
Accurate control of macromolecule transport between nucleus and cytoplasm underlines several essential biological processes, including gene expression. According to the canonical model, nuclear import of soluble proteins is based on nuclear localization signals and transport factors. We challenge this view by showing that nuclear localization of the actin-dependent motor protein Myosin-1C (Myo1C) resembles the diffusion-retention mechanism utilized by inner nuclear membrane proteins. We show that Myo1C constantly shuttles in and out of the nucleus and that its nuclear localization does not require soluble factors, but is dependent on phosphoinositide binding. Nuclear import of Myo1C is preceded by its interaction with the endoplasmic reticulum, and phosphoinositide binding is specifically required for nuclear import, but not nuclear retention, of Myo1C. Our results therefore demonstrate, for the first time, that membrane association and binding to nuclear partners is sufficient to drive nuclear localization of also soluble proteins, opening new perspectives to evolution of cellular protein sorting mechanisms.
Collapse
Affiliation(s)
- Ilja Nevzorov
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ekaterina Sidorenko
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Weihuan Wang
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
- College of Life Sciences, Northwest A&F University, Yangling, Shanxi, China
| | - Hongxia Zhao
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Maria K Vartiainen
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
32
|
Sankovski E, Abroi A, Ustav M, Ustav M. Nuclear myosin 1 associates with papillomavirus E2 regulatory protein and influences viral replication. Virology 2018; 514:142-155. [PMID: 29179037 DOI: 10.1016/j.virol.2017.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 11/09/2017] [Accepted: 11/16/2017] [Indexed: 11/25/2022]
Abstract
Nuclear myosin 1c (NM1) associates with RNA polymerases and is a partner in the chromatin remodeling complex B-WICH. This complex, which also contains WSTF and SNF2h proteins, is involved in transcriptional regulation. We report herein that papillomavirus protein E2 binds to NM1 and co-precipitates with the WSTF and SNF2h proteins. Our data suggest that E2 associates with the cellular B-WICH complex through binding to NM1. E2 and NM1 associate via their N-terminal domains and this interaction is ATP dependent. The cellular multifunctional protein Brd4 and beta-actin are also present in the NM1-E2 complex. NM1 downregulation by siRNA increases the replication of the BPV1 and HPV5 genomes but does not affect HPV18 genome replication. These results suggest that the B-WICH complex may play a role in the papillomavirus life cycle through NM1 and E2 protein interaction.
Collapse
Affiliation(s)
- Eve Sankovski
- University of Tartu, Institute of Technology, Nooruse 1, 50411 Tartu, Estonia
| | - Aare Abroi
- Estonian Biocentre, Riia 23, 51010 Tartu, Estonia
| | - Mart Ustav
- University of Tartu, Institute of Technology, Nooruse 1, 50411 Tartu, Estonia; Icosagen Cell Factory OÜ, Eerika tee 1, Õssu küla, Ülenurme vald, 61713 Tartumaa, Estonia
| | - Mart Ustav
- University of Tartu, Institute of Technology, Nooruse 1, 50411 Tartu, Estonia; Icosagen Cell Factory OÜ, Eerika tee 1, Õssu küla, Ülenurme vald, 61713 Tartumaa, Estonia; Estonian Academy of Sciences, Kohtu 6, 10130 Tallinn, Estonia.
| |
Collapse
|
33
|
Majewski L, Nowak J, Sobczak M, Karatsai O, Havrylov S, Lenartowski R, Suszek M, Lenartowska M, Redowicz MJ. Myosin VI in the nucleus of neurosecretory PC12 cells: Stimulation-dependent nuclear translocation and interaction with nuclear proteins. Nucleus 2018; 9:125-141. [PMID: 29293066 PMCID: PMC5973263 DOI: 10.1080/19491034.2017.1421881] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/23/2017] [Accepted: 12/18/2017] [Indexed: 02/07/2023] Open
Abstract
Myosin VI (MVI) is a unique actin-based motor protein moving towards the minus end of actin filaments, in the opposite direction than other known myosins. Besides well described functions of MVI in endocytosis and maintenance of Golgi apparatus, there are few reports showing its involvement in transcription. We previously demonstrated that in neurosecretory PC12 cells MVI was present in the cytoplasm and nucleus, and its depletion caused substantial inhibition of cell migration and proliferation. Here, we show an increase in nuclear localization of MVI upon cell stimulation, and identification of potential nuclear localization (NLS) and nuclear export (NES) signals within MVI heavy chain. These signals seem to be functional as the MVI nuclear presence was affected by the inhibitors of nuclear import (ivermectin) and export (leptomycin B). In nuclei of stimulated cells, MVI colocalized with active RNA polymerase II, BrUTP-containing transcription sites and transcription factor SP1 as well as SC35 and PML proteins, markers of nuclear speckles and PML bodies, respectively. Mass spectrometry analysis of samples of a GST-pull-down assay with the MVI tail domain as a "bait" identified several new potential MVI binding partners. Among them are proteins involved in transcription and post-transcriptional processes. We confirmed interaction of MVI with heterogeneous nuclear ribonucleoprotein U (hnRNPU) and nucleolin, proteins involved in pre-mRNA binding and transport, and nucleolar function, respectively. Our data provide an insight into mechanisms of involvement of MVI in nuclear processes via interaction with nuclear proteins and support a notion for important role(s) for MVI in gene expression.
Collapse
Affiliation(s)
- Lukasz Majewski
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Jolanta Nowak
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Magdalena Sobczak
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Olena Karatsai
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Serhiy Havrylov
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Robert Lenartowski
- Laboratory of Isotope and Instrumental Analysis, Department of Cellular and Molecular Biology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Malgorzata Suszek
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Marta Lenartowska
- Laboratory of Developmental Biology, Department of Cellular and Molecular Biology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Maria Jolanta Redowicz
- Laboratory of Molecular Basis of Cell Motility, Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
34
|
Johnston JR, Chase PB, Pinto JR. Troponin through the looking-glass: emerging roles beyond regulation of striated muscle contraction. Oncotarget 2017; 9:1461-1482. [PMID: 29416706 PMCID: PMC5787451 DOI: 10.18632/oncotarget.22879] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/20/2017] [Indexed: 01/03/2023] Open
Abstract
Troponin is a heterotrimeric Ca2+-binding protein that has a well-established role in regulating striated muscle contraction. However, mounting evidence points to novel cellular functions of troponin, with profound implications in cancer, cardiomyopathy pathogenesis and skeletal muscle aging. Here, we highlight the non-canonical roles and aberrant expression patterns of troponin beyond the sarcomeric milieu. Utilizing bioinformatics tools and online databases, we also provide pathway, subcellular localization, and protein-protein/DNA interaction analyses that support a role for troponin in multiple subcellular compartments. This emerging knowledge challenges the conventional view of troponin as a sarcomere-specific protein exclusively involved in muscle contraction and may transform the way we think about sarcomeric proteins, particularly in the context of human disease and aging.
Collapse
Affiliation(s)
- Jamie R Johnston
- Department of Biomedical Sciences, The Florida State University College of Medicine, Tallahassee, FL, 32306-4300, USA
| | - P Bryant Chase
- Department of Biological Science, The Florida State University, Tallahassee, FL, 32306-4370, USA
| | - Jose Renato Pinto
- Department of Biomedical Sciences, The Florida State University College of Medicine, Tallahassee, FL, 32306-4300, USA
| |
Collapse
|
35
|
Zattelman L, Regev R, Ušaj M, Reinke PYA, Giese S, Samson AO, Taft MH, Manstein DJ, Henn A. N-terminal splicing extensions of the human MYO1C gene fine-tune the kinetics of the three full-length myosin IC isoforms. J Biol Chem 2017; 292:17804-17818. [PMID: 28893906 DOI: 10.1074/jbc.m117.794008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/06/2017] [Indexed: 12/28/2022] Open
Abstract
The MYO1C gene produces three alternatively spliced isoforms, differing only in their N-terminal regions (NTRs). These isoforms, which exhibit both specific and overlapping nuclear and cytoplasmic functions, have different expression levels and nuclear-cytoplasmic partitioning. To investigate the effect of NTR extensions on the enzymatic behavior of individual isoforms, we overexpressed and purified the three full-length human isoforms from suspension-adapted HEK cells. MYO1CC favored the actomyosin closed state (AMC), MYO1C16 populated the actomyosin open state (AMO) and AMC equally, and MYO1C35 favored the AMO state. Moreover, the full-length constructs isomerized before ADP release, which has not been observed previously in truncated MYO1CC constructs. Furthermore, global numerical simulation analysis predicted that MYO1C35 populated the actomyosin·ADP closed state (AMDC) 5-fold more than the actomyosin·ADP open state (AMDO) and to a greater degree than MYO1CC and MYO1C16 (4- and 2-fold, respectively). On the basis of a homology model of the 35-amino acid NTR of MYO1C35 (NTR35) docked to the X-ray structure of MYO1CC, we predicted that MYO1C35 NTR residue Arg-21 would engage in a specific interaction with post-relay helix residue Glu-469, which affects the mechanics of the myosin power stroke. In addition, we found that adding the NTR35 peptide to MYO1CC yielded a protein that transiently mimics MYO1C35 kinetic behavior. By contrast, NTR35, which harbors the R21G mutation, was unable to confer MYO1C35-like kinetic behavior. Thus, the NTRs affect the specific nucleotide-binding properties of MYO1C isoforms, adding to their kinetic diversity. We propose that this level of fine-tuning within MYO1C broadens its adaptability within cells.
Collapse
Affiliation(s)
- Lilach Zattelman
- From the Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Ronit Regev
- From the Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Marko Ušaj
- From the Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Patrick Y A Reinke
- the Institute for Biophysical Chemistry, Hannover Medical School, OE 4350, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Sven Giese
- the Institute for Biophysical Chemistry, Hannover Medical School, OE 4350, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Abraham O Samson
- the Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311520, Israel, and
| | - Manuel H Taft
- the Institute for Biophysical Chemistry, Hannover Medical School, OE 4350, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Dietmar J Manstein
- the Institute for Biophysical Chemistry, Hannover Medical School, OE 4350, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Arnon Henn
- From the Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel,
| |
Collapse
|
36
|
Gupta P, Martin R, Knölker HJ, Nihalani D, Kumar Sinha D. Myosin-1 inhibition by PClP affects membrane shape, cortical actin distribution and lipid droplet dynamics in early Zebrafish embryos. PLoS One 2017; 12:e0180301. [PMID: 28678859 PMCID: PMC5498032 DOI: 10.1371/journal.pone.0180301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/13/2017] [Indexed: 12/22/2022] Open
Abstract
Myosin-1 (Myo1) represents a mechanical link between the membrane and actin-cytoskeleton in animal cells. We have studied the effect of Myo1 inhibitor PClP in 1-8 cell Zebrafish embryos. Our results indicate a unique involvement of Myo1 in early development of Zebrafish embryos. Inhibition of Myo1 (by PClP) and Myo2 (by Blebbistatin) lead to arrest in cell division. While Myo1 isoforms appears to be important for both the formation and the maintenance of cleavage furrows, Myo2 is required only for the formation of furrows. We found that the blastodisc of the embryo, which contains a thick actin cortex (~13 μm), is loaded with cortical Myo1. Myo1 appears to be crucial for maintaining the blastodisc morphology and the actin cortex thickness. In addition to cell division and furrow formation, inhibition of Myo1 has a drastic effect on the dynamics and distribution of lipid droplets (LDs) in the blastodisc near the cleavage furrow. All these results above are effects of Myo1 inhibition exclusively; Myo2 inhibition by blebbistatin does not show such phenotypes. Therefore, our results demonstrate a potential role for Myo1 in the maintenance and formation of furrow, blastodisc morphology, cell-division and LD organization within the blastodisc during early embryogenesis.
Collapse
MESH Headings
- Actin Cytoskeleton/drug effects
- Actin Cytoskeleton/metabolism
- Actins/genetics
- Actins/metabolism
- Animals
- Blastomeres/cytology
- Blastomeres/metabolism
- Blastomeres/ultrastructure
- Blotting, Western
- Cell Division/drug effects
- Cell Division/genetics
- Cell Membrane/metabolism
- Embryo, Nonmammalian/embryology
- Embryo, Nonmammalian/metabolism
- Embryo, Nonmammalian/ultrastructure
- Gene Expression Regulation, Developmental
- Heterocyclic Compounds, 4 or More Rings/pharmacology
- Hydrocarbons, Chlorinated/pharmacology
- Lipid Droplets/metabolism
- Microscopy, Electron, Scanning
- Microscopy, Fluorescence
- Myosin Heavy Chains/antagonists & inhibitors
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Pyrroles/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
- Zebrafish/embryology
- Zebrafish/genetics
- Zebrafish/metabolism
- Zebrafish Proteins/antagonists & inhibitors
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
| | - René Martin
- Department Chemie, TU Dresden, Dresden, Germany
| | | | - Deepak Nihalani
- Dept. Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | | |
Collapse
|
37
|
Masters TA, Kendrick-Jones J, Buss F. Myosins: Domain Organisation, Motor Properties, Physiological Roles and Cellular Functions. Handb Exp Pharmacol 2017; 235:77-122. [PMID: 27757761 DOI: 10.1007/164_2016_29] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myosins are cytoskeletal motor proteins that use energy derived from ATP hydrolysis to generate force and movement along actin filaments. Humans express 38 myosin genes belonging to 12 classes that participate in a diverse range of crucial activities, including muscle contraction, intracellular trafficking, cell division, motility, actin cytoskeletal organisation and cell signalling. Myosin malfunction has been implicated a variety of disorders including deafness, hypertrophic cardiomyopathy, Usher syndrome, Griscelli syndrome and cancer. In this chapter, we will first discuss the key structural and kinetic features that are conserved across the myosin family. Thereafter, we summarise for each member in turn its unique functional and structural adaptations, cellular roles and associated pathologies. Finally, we address the broad therapeutic potential for pharmacological interventions that target myosin family members.
Collapse
Affiliation(s)
- Thomas A Masters
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK.
| | | | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| |
Collapse
|
38
|
Zhang Y, Najmi SM, Schneider DA. Transcription factors that influence RNA polymerases I and II: To what extent is mechanism of action conserved? BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1860:246-255. [PMID: 27989933 DOI: 10.1016/j.bbagrm.2016.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/07/2016] [Accepted: 10/25/2016] [Indexed: 01/05/2023]
Abstract
In eukaryotic cells, nuclear RNA synthesis is accomplished by at least three unique, multisubunit RNA polymerases. The roles of these enzymes are generally partitioned into the synthesis of the three major classes of RNA: rRNA, mRNA, and tRNA for RNA polymerases I, II, and III respectively. Consistent with their unique cellular roles, each enzyme has a complement of specialized transcription factors and enzymatic properties. However, not all transcription factors have evolved to affect only one eukaryotic RNA polymerase. In fact, many factors have been shown to influence the activities of multiple nuclear RNA polymerases. This review focuses on a subset of these factors, specifically addressing the mechanisms by which these proteins influence RNA polymerases I and II.
Collapse
Affiliation(s)
- Yinfeng Zhang
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Saman M Najmi
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - David A Schneider
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
39
|
Visuttijai K, Pettersson J, Mehrbani Azar Y, van den Bout I, Örndal C, Marcickiewicz J, Nilsson S, Hörnquist M, Olsson B, Ejeskär K, Behboudi A. Lowered Expression of Tumor Suppressor Candidate MYO1C Stimulates Cell Proliferation, Suppresses Cell Adhesion and Activates AKT. PLoS One 2016; 11:e0164063. [PMID: 27716847 PMCID: PMC5055341 DOI: 10.1371/journal.pone.0164063] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 09/18/2016] [Indexed: 12/12/2022] Open
Abstract
Myosin-1C (MYO1C) is a tumor suppressor candidate located in a region of recurrent losses distal to TP53. Myo1c can tightly and specifically bind to PIP2, the substrate of Phosphoinositide 3-kinase (PI3K), and to Rictor, suggesting a role for MYO1C in the PI3K pathway. This study was designed to examine MYO1C expression status in a panel of well-stratified endometrial carcinomas as well as to assess the biological significance of MYO1C as a tumor suppressor in vitro. We found a significant correlation between the tumor stage and lowered expression of MYO1C in endometrial carcinoma samples. In cell transfection experiments, we found a negative correlation between MYO1C expression and cell proliferation, and MYO1C silencing resulted in diminished cell migration and adhesion. Cells expressing excess of MYO1C had low basal level of phosphorylated protein kinase B (PKB, a.k.a. AKT) and cells with knocked down MYO1C expression showed a quicker phosphorylated AKT (pAKT) response in reaction to serum stimulation. Taken together the present study gives further evidence for tumor suppressor activity of MYO1C and suggests MYO1C mediates its tumor suppressor function through inhibition of PI3K pathway and its involvement in loss of contact inhibition.
Collapse
Affiliation(s)
- Kittichate Visuttijai
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
- Department of Medical and Clinical Genetics, Sahlgrenska Academy, University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Jennifer Pettersson
- Department of Medical and Clinical Genetics, Sahlgrenska Academy, University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Yashar Mehrbani Azar
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
| | - Iman van den Bout
- Department of physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, 0007, South Africa
| | - Charlotte Örndal
- Department of Pathology, Sahlgrenska University Hospital, SE-413 45, Gothenburg, Sweden
| | - Janusz Marcickiewicz
- Department of Obstetrics and Gynecology, Halland Hospital Varberg, SE- 432 37, Varberg, Sweden
| | - Staffan Nilsson
- Institute of Mathematical Statistics, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - Michael Hörnquist
- Department of Science and Technology, University of Linköping, ITN, SE-601 74, Norrköping, Sweden
| | - Björn Olsson
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
| | - Katarina Ejeskär
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
| | - Afrouz Behboudi
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
- * E-mail:
| |
Collapse
|
40
|
Venit T, Kalendová A, Petr M, Dzijak R, Pastorek L, Rohožková J, Malohlava J, Hozák P. Nuclear myosin I regulates cell membrane tension. Sci Rep 2016; 6:30864. [PMID: 27480647 PMCID: PMC4969604 DOI: 10.1038/srep30864] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 07/12/2016] [Indexed: 11/09/2022] Open
Abstract
Plasma membrane tension is an important feature that determines the cell shape and influences processes such as cell motility, spreading, endocytosis and exocytosis. Unconventional class 1 myosins are potent regulators of plasma membrane tension because they physically link the plasma membrane with adjacent cytoskeleton. We identified nuclear myosin 1 (NM1) - a putative nuclear isoform of myosin 1c (Myo1c) - as a new player in the field. Although having specific nuclear functions, NM1 localizes predominantly to the plasma membrane. Deletion of NM1 causes more than a 50% increase in the elasticity of the plasma membrane around the actin cytoskeleton as measured by atomic force microscopy. This higher elasticity of NM1 knock-out cells leads to 25% higher resistance to short-term hypotonic environment and rapid cell swelling. In contrast, overexpression of NM1 in wild type cells leads to an additional 30% reduction of their survival. We have shown that NM1 has a direct functional role in the cytoplasm as a dynamic linker between the cell membrane and the underlying cytoskeleton, regulating the degree of effective plasma membrane tension.
Collapse
Affiliation(s)
- Tomáš Venit
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic.,Faculty of Science, Charles University in Prague, Albertov 6, 128 43 Prague, Czech Republic
| | - Alžběta Kalendová
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Martin Petr
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Rastislav Dzijak
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Lukáš Pastorek
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Jana Rohožková
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Jakub Malohlava
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University in Olomouc, Hnevotinska 3, 775 15 Olomouc, Czech Republic
| | - Pavel Hozák
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| |
Collapse
|
41
|
Abstract
Myosin-I molecular motors are proposed to play various cellular roles related to membrane dynamics and trafficking. In this Cell Science at a Glance article and the accompanying poster, we review and illustrate the proposed cellular functions of metazoan myosin-I molecular motors by examining the structural, biochemical, mechanical and cell biological evidence for their proposed molecular roles. We highlight evidence for the roles of myosin-I isoforms in regulating membrane tension and actin architecture, powering plasma membrane and organelle deformation, participating in membrane trafficking, and functioning as a tension-sensitive dock or tether. Collectively, myosin-I motors have been implicated in increasingly complex cellular phenomena, yet how a single isoform accomplishes multiple types of molecular functions is still an active area of investigation. To fully understand the underlying physiology, it is now essential to piece together different approaches of biological investigation. This article will appeal to investigators who study immunology, metabolic diseases, endosomal trafficking, cell motility, cancer and kidney disease, and to those who are interested in how cellular membranes are coupled to the underlying actin cytoskeleton in a variety of different applications.
Collapse
Affiliation(s)
- Betsy B McIntosh
- Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | - E Michael Ostap
- Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| |
Collapse
|
42
|
Kulashreshtha M, Mehta IS, Kumar P, Rao BJ. Chromosome territory relocation during DNA repair requires nuclear myosin 1 recruitment to chromatin mediated by ϒ-H2AX signaling. Nucleic Acids Res 2016; 44:8272-91. [PMID: 27365048 PMCID: PMC5041470 DOI: 10.1093/nar/gkw573] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 06/03/2016] [Indexed: 11/22/2022] Open
Abstract
During DNA damage response (DDR), certain gene rich chromosome territories (CTs) relocate to newer positions within interphase nuclei and revert to their native locations following repair. Such dynamic relocation of CTs has been observed under various cellular conditions, however, the underlying mechanistic basis of the same has remained largely elusive. In this study, we aim to understand the temporal and molecular details of such crosstalk between DDR signaling and CT relocation dynamics. We demonstrate that signaling at DNA double strand breaks (DSBs) by the phosphorylated histone variant (ϒ-H2AX) is a pre-requisite for damage induced CT relocation, as cells deficient in ϒ-H2AX signaling fail to exhibit such a response. Inhibition of Rad51 or DNA Ligase IV mediated late steps of double strand break repair does not seem to abrogate CT relocation completely. Upon DNA damage, an increase in the levels of chromatin bound motor protein nuclear myosin 1 (NM1) ensues, which appears to be functionally linked to ϒ-H2AX signaling. Importantly, the motor function of NM1 is essential for its recruitment to chromatin and CT relocation following damage. Taking these observations together, we propose that early DDR sensing and signaling result in NM1 recruitment to chromosomes which in turn guides DNA damage induced CT relocation.
Collapse
Affiliation(s)
- Mugdha Kulashreshtha
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Ishita S Mehta
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India UM-DAE Centre for Excellence in Basic Sciences, Biological Sciences, Kalina Campus, Santacruz (E), Mumbai, Maharashtra 400098, India
| | - Pradeep Kumar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India UM-DAE Centre for Excellence in Basic Sciences, Biological Sciences, Kalina Campus, Santacruz (E), Mumbai, Maharashtra 400098, India
| | - Basuthkar J Rao
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| |
Collapse
|
43
|
Maly IV, Hofmann WA. Calcium-regulated import of myosin IC into the nucleus. Cytoskeleton (Hoboken) 2016; 73:341-50. [PMID: 27192697 DOI: 10.1002/cm.21310] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 05/09/2016] [Accepted: 05/16/2016] [Indexed: 01/07/2023]
Abstract
Myosin IC is a molecular motor involved in intracellular transport, cell motility, and transcription. Its mechanical properties are regulated by calcium via calmodulin binding, and its functions in the nucleus depend on import from the cytoplasm. The import has recently been shown to be mediated by the nuclear localization signal located within the calmodulin-binding domain. In the present paper, it is demonstrated that mutations in the calmodulin-binding sequence shift the intracellular distribution of myosin IC to the nucleus. The redistribution is displayed by isoform B, described originally as the "nuclear myosin," but is particularly pronounced with isoform C, the normally cytoplasmic isoform. Furthermore, experimental elevation of the intracellular calcium concentration induces a rapid import of myosin into the nucleus. The import is blocked by the importin β inhibitor importazole. These findings are consistent with a mechanism whereby calmodulin binding prevents recognition of the nuclear localization sequence by importin β, and the steric inhibition of import is released by cell signaling leading to the intracellular calcium elevation. The results establish a mechanistic connection between the calcium regulation of the motor function of myosin IC in the cytoplasm and the induction of its import into the nucleus. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ivan V Maly
- Department of Physiology and Biophysics, University at Buffalo-State University of New York, Buffalo, New York
| | - Wilma A Hofmann
- Department of Physiology and Biophysics, University at Buffalo-State University of New York, Buffalo, New York
| |
Collapse
|
44
|
Kruppa AJ, Kendrick-Jones J, Buss F. Myosins, Actin and Autophagy. Traffic 2016; 17:878-90. [PMID: 27146966 PMCID: PMC4957615 DOI: 10.1111/tra.12410] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/02/2016] [Accepted: 05/02/2016] [Indexed: 12/20/2022]
Abstract
Myosin motor proteins working together with the actin cytoskeleton drive a wide range of cellular processes. In this review, we focus on their roles in autophagy – the pathway the cell uses to ensure homeostasis by targeting pathogens, misfolded proteins and damaged organelles for degradation. The actin cytoskeleton regulated by a host of nucleating, anchoring and stabilizing proteins provides the filament network for the delivery of essential membrane vesicles from different cellular compartments to the autophagosome. Actin networks have also been implicated in structurally supporting the expanding phagophore, moving autophagosomes and enabling efficient fusion with the lysosome. Only a few myosins have so far been shown to play a role in autophagy. Non‐muscle myosin IIA functions in the early stages delivering membrane for the initial formation of the autophagosome, whereas myosin IC and myosin VI are involved in the final stages providing specific membranes for autophagosome maturation and its fusion with the lysosome.
Collapse
Affiliation(s)
- Antonina J Kruppa
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
| | - John Kendrick-Jones
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
| |
Collapse
|
45
|
Actin, actin-binding proteins, and actin-related proteins in the nucleus. Histochem Cell Biol 2016; 145:373-88. [PMID: 26847179 DOI: 10.1007/s00418-015-1400-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2015] [Indexed: 10/25/2022]
Abstract
Extensive research in the past decade has significantly broadened our view about the role actin plays in the life of the cell and added novel aspects to actin research. One of these new aspects is the discovery of the existence of nuclear actin which became evident only recently. Nuclear activities including transcriptional activation in the case of all three RNA polymerases, editing and nuclear export of mRNAs, and chromatin remodeling all depend on actin. It also became clear that there is a fine-tuned equilibrium between cytoplasmic and nuclear actin pools and that this balance is ensured by an export-import system dedicated to actin. After over half a century of research on conventional actin and its organizing partners in the cytoplasm, it was also an unexpected finding that the nucleus contains more than 30 actin-binding proteins and new classes of actin-related proteins which are not able to form filaments but had evolved nuclear-specific functions. The actin-binding and actin-related proteins in the nucleus have been linked to RNA transcription and processing, nuclear transport, and chromatin remodeling. In this paper, we attempt to provide an overview of the wide range of information that is now available about actin, actin-binding, and actin-related proteins in the nucleus.
Collapse
|
46
|
Petrini E, Baillet V, Cridge J, Hogan CJ, Guillaume C, Ke H, Brandetti E, Walker S, Koohy H, Spivakov M, Varga-Weisz P. A new phosphate-starvation response in fission yeast requires the endocytic function of myosin I. J Cell Sci 2015; 128:3707-13. [PMID: 26345368 PMCID: PMC4631780 DOI: 10.1242/jcs.171314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/28/2015] [Indexed: 12/15/2022] Open
Abstract
Endocytosis is essential for uptake of many substances into the cell, but how it links to nutritional signalling is poorly understood. Here, we show a new role for endocytosis in regulating the response to low phosphate in Schizosaccharomyces pombe. Loss of function of myosin I (Myo1), Sla2/End4 or Arp2, proteins involved in the early steps of endocytosis, led to increased proliferation in low-phosphate medium compared to controls. We show that once cells are deprived of phosphate they undergo a quiescence response that is dependent on the endocytic function of Myo1. Transcriptomic analysis revealed a wide perturbation of gene expression with induction of stress-regulated genes upon phosphate starvation in wild-type but not Δmyo1 cells. Thus, endocytosis plays a pivotal role in mediating the cellular response to nutrients, bridging the external environment and internal molecular functions of the cell. Highlighted Article: The endocytic machinery, including the type 1 myosin Myo1, is required to establish a quiescence response to low-phosphate stress in fission yeast.
Collapse
Affiliation(s)
- Edoardo Petrini
- Nuclear Dynamics, Babraham Institute, Cambridge CB22 3AT, USA
| | | | - Jake Cridge
- Nuclear Dynamics, Babraham Institute, Cambridge CB22 3AT, USA
| | | | - Cindy Guillaume
- Nuclear Dynamics, Babraham Institute, Cambridge CB22 3AT, USA
| | - Huiling Ke
- Nuclear Dynamics, Babraham Institute, Cambridge CB22 3AT, USA
| | - Elisa Brandetti
- Nuclear Dynamics, Babraham Institute, Cambridge CB22 3AT, USA
| | - Simon Walker
- Imaging Facility, Babraham Institute, Cambridge CB22 3AT, USA
| | - Hashem Koohy
- Nuclear Dynamics, Babraham Institute, Cambridge CB22 3AT, USA
| | | | | |
Collapse
|
47
|
Abstract
Myosins are molecular motors that generate force to power a wide array of motile cellular functions. Myosins have the inherent ability to change their ATPase kinetics and force-generating properties when they encounter mechanical loads; however, little is known about the structural elements in myosin responsible for force sensing. Recent structural and biophysical studies have shown that myosin-I isoforms, Myosin-Ib (Myo1b) and Myosin-Ic (Myo1c), have similar unloaded kinetics and sequences but substantially different responses to forces that resist their working strokes. Myo1b has the properties of a tension-sensing anchor, slowing its actin-detachment kinetics by two orders of magnitude with just 1 pN of resisting force, whereas Myo1c has the properties of a slow transporter, generating power without slowing under 1-pN loads that would stall Myo1b. To examine the structural elements that lead to differences in force sensing, we used single-molecule and ensemble kinetic techniques to show that the myosin-I N-terminal region (NTR) plays a critical role in tuning myosin-I mechanochemistry. We found that replacing the Myo1c NTR with the Myo1b NTR changes the identity of the primary force-sensitive transition of Myo1c, resulting in sensitivity to forces of <2 pN. Additionally, we found that the NTR plays an important role in stabilizing the post-power-stroke conformation. These results identify the NTR as an important structural element in myosin force sensing and suggest a mechanism for generating diversity of function among myosin isoforms.
Collapse
|
48
|
Almuzzaini B, Sarshad AA, Farrants AKÖ, Percipalle P. Nuclear myosin 1 contributes to a chromatin landscape compatible with RNA polymerase II transcription activation. BMC Biol 2015; 13:35. [PMID: 26044184 PMCID: PMC4486089 DOI: 10.1186/s12915-015-0147-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 06/02/2015] [Indexed: 12/11/2022] Open
Abstract
Background Nuclear myosin 1c (NM1) is emerging as a regulator of transcription and chromatin organization. Results Using chromatin immunoprecipitation and deep sequencing (ChIP-Seq) in combination with molecular analyses, we investigated the global association of NM1 with the mammalian genome. Analysis of the ChIP-Seq data demonstrates that NM1 binds across the entire mammalian genome with occupancy peaks correlating with distributions of RNA Polymerase II (Pol II) and active epigenetic marks at class II gene promoters. In mouse embryonic fibroblasts subjected to RNAi mediated NM1 gene silencing, we show that NM1 synergizes with polymerase-associated actin to maintain active Pol II at the promoter. NM1 also co-localizes with the nucleosome remodeler SNF2h at class II promoters where they assemble together with WSTF as part of the B-WICH complex. A high resolution micrococcal nuclease (MNase) assay and quantitative real time PCR shows that this mechanism is required for local chromatin remodeling. Following B-WICH assembly, NM1 mediates physical recruitment of the histone acetyl transferase PCAF and the histone methyl transferase Set1/Ash2 to maintain and preserve H3K9acetylation and H3K4trimethylation for active transcription. Conclusions We propose a novel genome-wide mechanism where myosin synergizes with Pol II-associated actin to link the polymerase machinery with permissive chromatin for transcription activation. Electronic supplementary material The online version of this article (doi:10.1186/s12915-015-0147-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bader Almuzzaini
- Department of Cell and Molecular Biology, Karolinska Institute, Box 285, SE-171 77, Stockholm, Sweden.
| | - Aishe A Sarshad
- Department of Cell and Molecular Biology, Karolinska Institute, Box 285, SE-171 77, Stockholm, Sweden. .,Present address: National Institute of Health, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD, 20892-3675, USA.
| | - Ann-Kristin Östlund Farrants
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91, Stockholm, Sweden.
| | - Piergiorgio Percipalle
- Department of Cell and Molecular Biology, Karolinska Institute, Box 285, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
49
|
Myosin VI regulates gene pairing and transcriptional pause release in T cells. Proc Natl Acad Sci U S A 2015; 112:E1587-93. [PMID: 25770220 DOI: 10.1073/pnas.1502461112] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Naive CD4 T cells differentiate into several effector lineages, which generate a stronger and more rapid response to previously encountered immunological challenges. Although effector function is a key feature of adaptive immunity, the molecular basis of this process is poorly understood. Here, we investigated the spatiotemporal regulation of cytokine gene expression in resting and restimulated effector T helper 1 (Th1) cells. We found that the Lymphotoxin (LT)/TNF alleles, which encode TNF-α, were closely juxtaposed shortly after T-cell receptor (TCR) engagement, when transcription factors are limiting. Allelic pairing required a nuclear myosin, myosin VI, which is rapidly recruited to the LT/TNF locus upon restimulation. Furthermore, transcription was paused at the TNF locus and other related genes in resting Th1 cells and released in a myosin VI-dependent manner following activation. We propose that homologous pairing and myosin VI-mediated transcriptional pause release account for the rapid and efficient expression of genes induced by an external stimulus.
Collapse
|
50
|
Class I myosins have overlapping and specialized functions in left-right asymmetric development in Drosophila. Genetics 2015; 199:1183-99. [PMID: 25659376 DOI: 10.1534/genetics.115.174698] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 02/04/2015] [Indexed: 11/18/2022] Open
Abstract
The class I myosin genes are conserved in diverse organisms, and their gene products are involved in actin dynamics, endocytosis, and signal transduction. Drosophila melanogaster has three class I myosin genes, Myosin 31DF (Myo31DF), Myosin 61F (Myo61F), and Myosin 95E (Myo95E). Myo31DF, Myo61F, and Myo95E belong to the Myosin ID, Myosin IC, and Myosin IB families, respectively. Previous loss-of-function analyses of Myo31DF and Myo61F revealed important roles in left-right (LR) asymmetric development and enterocyte maintenance, respectively. However, it was difficult to elucidate their roles in vivo, because of potential redundant activities. Here we generated class I myosin double and triple mutants to address this issue. We found that the triple mutant was viable and fertile, indicating that all three class I myosins were dispensable for survival. A loss-of-function analysis revealed further that Myo31DF and Myo61F, but not Myo95E, had redundant functions in promoting the dextral LR asymmetric development of the male genitalia. Myo61F overexpression is known to antagonize the dextral activity of Myo31DF in various Drosophila organs. Thus, the LR-reversing activity of overexpressed Myo61F may not reflect its physiological function. The endogenous activity of Myo61F in promoting dextral LR asymmetric development was observed in the male genitalia, but not the embryonic gut, another LR asymmetric organ. Thus, Myo61F and Myo31DF, but not Myo95E, play tissue-specific, redundant roles in LR asymmetric development. Our studies also revealed differential colocalization of the class I myosins with filamentous (F)-actin in the brush border of intestinal enterocytes.
Collapse
|