1
|
Radhakrishnan R, Dronamraju VR, Leung M, Gruesen A, Solanki AK, Walterhouse S, Roehrich H, Song G, da Costa Monsanto R, Cureoglu S, Martin R, Kondkar AA, van Kuijk FJ, Montezuma SR, Knöelker HJ, Hufnagel RB, Lobo GP. The role of motor proteins in photoreceptor protein transport and visual function. Ophthalmic Genet 2022; 43:285-300. [PMID: 35470760 DOI: 10.1080/13816810.2022.2062391] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Rods and cones are photoreceptor neurons in the retina that are required for visual sensation in vertebrates, wherein the perception of vision is initiated when these neurons respond to photons in the light stimuli. The photoreceptor cell is structurally studied as outer segments (OS) and inner segments (IS) where proper protein sorting, localization, and compartmentalization are critical for phototransduction, visual function, and survival. In human retinal diseases, improper protein transport to the OS or mislocalization of proteins to the IS and other cellular compartments could lead to impaired visual responses and photoreceptor cell degeneration that ultimately cause loss of visual function. RESULTS Therefore, studying and identifying mechanisms involved in facilitating and maintaining proper protein transport in photoreceptor cells would help our understanding of pathologies involving retinal cell degeneration in inherited retinal dystrophies, age-related macular degeneration, and Usher Syndrome. CONCLUSIONS Our mini-review will discuss mechanisms of protein transport within photoreceptors and introduce a novel role for an unconventional motor protein, MYO1C, in actin-based motor transport of the visual chromophore Rhodopsin to the OS, in support of phototransduction and visual function.
Collapse
Affiliation(s)
- Rakesh Radhakrishnan
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Venkateshwara R Dronamraju
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Matthias Leung
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrew Gruesen
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ashish K Solanki
- Department of Medicine, Drug Discovery Building, Medical University of South Carolina, South Carolina, USA
| | - Stephen Walterhouse
- Department of Medicine, Drug Discovery Building, Medical University of South Carolina, South Carolina, USA
| | - Heidi Roehrich
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Grace Song
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rafael da Costa Monsanto
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sebahattin Cureoglu
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - René Martin
- Faculty of Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Altaf A Kondkar
- Department of Ophthalmology.,Glaucoma Research Chair in Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Frederik J van Kuijk
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sandra R Montezuma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Robert B Hufnagel
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Glenn P Lobo
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, Drug Discovery Building, Medical University of South Carolina, South Carolina, USA.,Department of Ophthalmology, Medical University of South Carolina, South Carolina, USA
| |
Collapse
|
2
|
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
|
3
|
Abstract
This book, a collection of chapters written by some of the leading researchers in the field of molecular motors, highlights the current understanding of the structure, molecular mechanism, and cellular roles of members of the myosin superfamily. Here, I briefly review the discovery of the first myosin motor, skeletal muscle myosin-II, and preview the contents of subsequent chapters.
Collapse
Affiliation(s)
- Lynne M Coluccio
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
4
|
|
5
|
Ko YS, Bae JA, Kim KY, Kim SJ, Sun EG, Lee KH, Kim N, Kang H, Seo YW, Kim H, Chung IJ, Kim KK. MYO1D binds with kinase domain of the EGFR family to anchor them to plasma membrane before their activation and contributes carcinogenesis. Oncogene 2019; 38:7416-7432. [DOI: 10.1038/s41388-019-0954-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 07/26/2019] [Accepted: 08/02/2019] [Indexed: 12/13/2022]
|
6
|
Arif E, Solanki AK, Srivastava P, Rahman B, Tash BR, Holzman LB, Janech MG, Martin R, Knölker HJ, Fitzgibbon WR, Deng P, Budisavljevic MN, Syn WK, Wang C, Lipschutz JH, Kwon SH, Nihalani D. The motor protein Myo1c regulates transforming growth factor-β-signaling and fibrosis in podocytes. Kidney Int 2019; 96:139-158. [PMID: 31097328 DOI: 10.1016/j.kint.2019.02.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/07/2019] [Accepted: 02/14/2019] [Indexed: 01/19/2023]
Abstract
Transforming growth factor-β (TGF-β) is known to play a critical role in the pathogenesis of many progressive podocyte diseases. However, the molecular mechanisms regulating TGF-β signaling in podocytes remain unclear. Using a podocyte-specific myosin (Myo)1c knockout, we demonstrate whether Myo1c is critical for TGF-β-signaling in podocyte disease pathogenesis. Specifically, podocyte-specific Myo1c knockout mice were resistant to fibrotic injury induced by Adriamycin or nephrotoxic serum. Further, loss of Myo1c also protected from injury in the TGF-β-dependent unilateral ureteral obstruction mouse model of renal interstitial fibrosis. Mechanistic analyses showed that loss of Myo1c significantly blunted TGF-β signaling through downregulation of canonical and non-canonical TGF-β pathways. Interestingly, nuclear rather than the cytoplasmic Myo1c was found to play a central role in controlling TGF-β signaling through transcriptional regulation. Differential expression analysis of nuclear Myo1c-associated gene promoters showed that nuclear Myo1c targeted the TGF-β responsive gene growth differentiation factor (GDF)-15 and directly bound to the GDF-15 promoter. Importantly, GDF15 was found to be involved in podocyte pathogenesis, where GDF15 was upregulated in glomeruli of patients with focal segmental glomerulosclerosis. Thus, Myo1c-mediated regulation of TGF-β-responsive genes is central to the pathogenesis of podocyte injury. Hence, inhibiting this process may have clinical application in treating podocytopathies.
Collapse
Affiliation(s)
- Ehtesham Arif
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ashish K Solanki
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Pankaj Srivastava
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Bushra Rahman
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Brian R Tash
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lawrence B Holzman
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael G Janech
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA; College of Charleston, Charleston, South Carolina, USA
| | - René Martin
- Department of Chemistry, TU Dresden, Dresden, Germany
| | | | - Wayne R Fitzgibbon
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Peifeng Deng
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Milos N Budisavljevic
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Wing-Kin Syn
- Department of Gastroenterology & Hepatology, Medical University of South Carolina, Charleston, South Carolina, USA; Section of Gastroenterology, Ralph H Johnson VA Medical Center, Charleston, South Carolina, USA; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, (UPV/EHU), Vizcaya, Spain
| | - Cindy Wang
- Department of Gastroenterology & Hepatology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Joshua H Lipschutz
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sang-Ho Kwon
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, USA
| | - Deepak Nihalani
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA.
| |
Collapse
|
7
|
Myosin isoform expressed in metastatic prostate cancer stimulates cell invasion. Sci Rep 2017; 7:8476. [PMID: 28814772 PMCID: PMC5559518 DOI: 10.1038/s41598-017-09158-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/21/2017] [Indexed: 12/15/2022] Open
Abstract
During metastasis, tumor cells migrate out of their original tissue to invade other organs. Secretion of exosomes and metalloproteases is essential for extracellular matrix remodeling, enabling migration through tissue barriers. Metastatic prostate cancer is differentiated by expression of the rare isoform A of the molecular motor myosin IC, however the function of this isoform remained unknown. Here we show that it contributes causatively to the invasive motility of prostate cancer cells. We found that the isoform associates with metalloprotease-containing exosomes and stimulates their secretion. While the data show that myosin IC is involved in prostate cancer cell migration, migration outside extracellular matrix in vitro proves little affected specifically by isoform A. Nevertheless, this isoform stimulates invasion through extracellular matrix, pointing to a critical role in secretion. Both the secretion and invasion depend on the integrity of the motor and lipid-binding domains of the protein. Our results demonstrate how myosin IC isoform A is likely to function in metastasis, driving secretion of exosomes that enable invasion of prostate cancer cells across extracellular matrix barriers. The new data identify a molecule suitable for a mechanistically grounded development into a marker and target for prognosis, detection, and treatment of invasive prostate cancer.
Collapse
|
8
|
How myosin organization of the actin cytoskeleton contributes to the cancer phenotype. Biochem Soc Trans 2017; 44:1026-34. [PMID: 27528748 DOI: 10.1042/bst20160034] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Indexed: 12/29/2022]
Abstract
The human genome contains 39 genes that encode myosin heavy chains, classified on the basis of their sequence similarity into 12 classes. Most cells express at least 12 different genes, from at least 8 different classes, which are typically composed of several class 1 genes, at least one class 2 gene and classes 5, 6, 9, 10, 18 and 19. Although the different myosin isoforms all have specific and non-overlapping roles in the cell, in combination they all contribute to the organization of the actin cytoskeleton, and the shape and phenotype of the cell. Over (or under) expression of these different myosin isoforms can have strong effects on actin organization, cell shape and contribute to the cancer phenotype as discussed in this review.
Collapse
|
9
|
Chemomechanical regulation of myosin Ic cross-bridges: Deducing the elastic properties of an ensemble from single-molecule mechanisms. PLoS Comput Biol 2017; 13:e1005566. [PMID: 28549064 PMCID: PMC5470724 DOI: 10.1371/journal.pcbi.1005566] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 06/14/2017] [Accepted: 05/11/2017] [Indexed: 12/15/2022] Open
Abstract
Myosin Ic is thought to be the principal constituent of the motor that adjusts mechanical responsiveness during adaptation to prolonged stimuli by hair cells, the sensory receptors of the inner ear. In this context myosin molecules operate neither as filaments, as occurs in muscles, nor as single or few molecules, as characterizes intracellular transport. Instead, myosin Ic molecules occur in a complex cluster in which they may exhibit cooperative properties. To better understand the motor’s remarkable function, we introduce a theoretical description of myosin Ic’s chemomechanical cycle based on experimental data from recent single-molecule studies. The cycle consists of distinct chemical states that the myosin molecule stochastically occupies. We explicitly calculate the probabilities of the occupancy of these states and show their dependence on the external force, the availability of actin, and the nucleotide concentrations as required by thermodynamic constraints. This analysis highlights that the strong binding of myosin Ic to actin is dominated by the ADP state for small external forces and by the ATP state for large forces. Our approach shows how specific parameter values of the chemomechanical cycle for myosin Ic result in behaviors distinct from those of other members of the myosin family. Integrating this single-molecule cycle into a simplified ensemble description, we predict that the average number of bound myosin heads is regulated by the external force and nucleotide concentrations. The elastic properties of such an ensemble are determined by the average number of myosin cross-bridges. Changing the binding probabilities and myosin’s stiffness under a constant force results in a mechanical relaxation which is large enough to account for fast adaptation in hair cells. Myosin molecules are biological nanomachines that transduce chemical energy into mechanical work and thus produce directed motion in living cells. These molecules proceed through cyclic reactions in which they change their conformational states upon the binding and release of nucleotides while attaching to and detaching from filaments. The myosin family consists of many distinct members with diverse functions such as muscle contraction, cargo transport, cell migration, and sensory adaptation. How these functions emerge from the biophysical properties of the individual molecules is an open question. We present an approach that integrates recent findings from single-molecule experiments into a thermodynamically consistent description of myosin Ic and demonstrate how the specific parameter values of the cycle result in a distinct function. The free variables of our description are the chemical input and external force, both of which are experimentally accessible and define the cellular environment in which these proteins function. We use this description to predict the elastic properties of an ensemble of molecules and discuss the implications for myosin Ic’s function in the inner ear as a tension regulator mediating adaptation, a hallmark of biological sensory systems. In this situation myosin molecules cooperate in an intermediate regime, neither as a large ensemble as in muscle nor as a single or a few molecules as in intracellular transport.
Collapse
|
10
|
Das JK, Das P, Ray KK, Choudhury PP, Jana SS. Mathematical Characterization of Protein Sequences Using Patterns as Chemical Group Combinations of Amino Acids. PLoS One 2016; 11:e0167651. [PMID: 27930687 PMCID: PMC5145171 DOI: 10.1371/journal.pone.0167651] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/17/2016] [Indexed: 01/08/2023] Open
Abstract
Comparison of amino acid sequence similarity is the fundamental concept behind the protein phylogenetic tree formation. By virtue of this method, we can explain the evolutionary relationships, but further explanations are not possible unless sequences are studied through the chemical nature of individual amino acids. Here we develop a new methodology to characterize the protein sequences on the basis of the chemical nature of the amino acids. We design various algorithms for studying the variation of chemical group transitions and various chemical group combinations as patterns in the protein sequences. The amino acid sequence of conventional myosin II head domain of 14 family members are taken to illustrate this new approach. We find two blocks of maximum length 6 aa as 'FPKATD' and 'Y/FTNEKL' without repeating the same chemical nature and one block of maximum length 20 aa with the repetition of chemical nature which are common among all 14 members. We also check commonality with another motor protein sub-family kinesin, KIF1A. Based on our analysis we find a common block of length 8 aa both in myosin II and KIF1A. This motif is located in the neck linker region which could be responsible for the generation of mechanical force, enabling us to find the unique blocks which remain chemically conserved across the family. We also validate our methodology with different protein families such as MYOI, Myosin light chain kinase (MLCK) and Rho-associated protein kinase (ROCK), Na+/K+-ATPase and Ca2+-ATPase. Altogether, our studies provide a new methodology for investigating the conserved amino acids' pattern in different proteins.
Collapse
Affiliation(s)
- Jayanta Kumar Das
- Applied Statistics Unit, Indian Statistical Institute, 203 B.T Road, Kolkata-700108, West Bengal, India
| | - Provas Das
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Kolkata-700032, West Bengal, India
| | - Korak Kumar Ray
- Department of Chemistry, Indian Institute of Technology-Bombay, IIT Bombay, Powai, Mumbai-400076, Maharashtra, India
| | - Pabitra Pal Choudhury
- Applied Statistics Unit, Indian Statistical Institute, 203 B.T Road, Kolkata-700108, West Bengal, India
| | - Siddhartha Sankar Jana
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Kolkata-700032, West Bengal, India
| |
Collapse
|
11
|
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
|
12
|
Makowska KA, Hughes RE, White KJ, Wells CM, Peckham M. Specific Myosins Control Actin Organization, Cell Morphology, and Migration in Prostate Cancer Cells. Cell Rep 2015; 13:2118-25. [PMID: 26670045 PMCID: PMC4688110 DOI: 10.1016/j.celrep.2015.11.012] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/27/2015] [Accepted: 11/02/2015] [Indexed: 11/16/2022] Open
Abstract
We investigated the myosin expression profile in prostate cancer cell lines and found that Myo1b, Myo9b, Myo10, and Myo18a were expressed at higher levels in cells with high metastatic potential. Moreover, Myo1b and Myo10 were expressed at higher levels in metastatic tumors. Using an siRNA-based approach, we found that knockdown of each myosin resulted in distinct phenotypes. Myo10 knockdown ablated filopodia and decreased 2D migration speed. Myo18a knockdown increased circumferential non-muscle myosin 2A-associated actin filament arrays in the lamella and reduced directional persistence of 2D migration. Myo9b knockdown increased stress fiber formation, decreased 2D migration speed, and increased directional persistence. Conversely, Myo1b knockdown increased numbers of stress fibers but did not affect 2D migration. In all cases, the cell spread area was increased and 3D migration potential was decreased. Therefore, myosins not only act as molecular motors but also directly influence actin organization and cell morphology, which can contribute to the metastatic phenotype. Myo1b, Myo9b, Myo10, and Myo18a are highly expressed in metastatic prostate cancer Knockdown of individual myosins distinctly affects the cytoskeleton and cell migration Myosins act in concert to directly influence actin organization and cell migration Misregulation of myosin expression may drive the metastatic phenotype
Collapse
Affiliation(s)
- Katarzyna A Makowska
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Ruth E Hughes
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Kathryn J White
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Claire M Wells
- Division of Cancer Studies, King's College London, London SE1 1UL, UK.
| | - Michelle Peckham
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
13
|
Li YR, Yang WX. Myosin superfamily: The multi-functional and irreplaceable factors in spermatogenesis and testicular tumors. Gene 2015; 576:195-207. [PMID: 26478466 DOI: 10.1016/j.gene.2015.10.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 09/21/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022]
Abstract
Spermatogenesis is a fundamental process in sexual development and reproduction, in which the diploid spermatogonia transform into haploid mature spermatozoa. This process is under the regulation of multiple factors and pathway. Myosin has been implicated in various aspects during spermatogenesis. Myosins constitute a diverse superfamily of actin-based molecular motors that translocate along microfilament in an ATP-dependent manner, and six kinds of myosins have been proved that function during spermatogenesis. In mitosis and meiosis, myosins play an important role in spindle assembly and positioning, karyokinesis and cytokinesis. During spermiogenesis, myosins participate in acrosomal formation, nuclear morphogenesis, mitochondrial translocation and spermatid individualization. In this review, we summarize current understanding of the functions of myosin in spermatogenesis and some reproductive system diseases such as testicular tumors and prostate cancer, and discuss the roles of possible upstream molecules which regulate myosin in these processes.
Collapse
Affiliation(s)
- Yan-Ruide Li
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
14
|
Ihnatovych I, Sielski NL, Hofmann WA. Selective expression of myosin IC Isoform A in mouse and human cell lines and mouse prostate cancer tissues. PLoS One 2014; 9:e108609. [PMID: 25259793 PMCID: PMC4178219 DOI: 10.1371/journal.pone.0108609] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/01/2014] [Indexed: 12/15/2022] Open
Abstract
Myosin IC is a single headed member of the myosin superfamily. We recently identified a novel isoform and showed that the MYOIC gene in mammalian cells encodes three isoforms (isoforms A, B, and C). Furthermore, we demonstrated that myosin IC isoform A but not isoform B exhibits a tissue specific expression pattern. In this study, we extended our analysis of myosin IC isoform expression patterns by analyzing the protein and mRNA expression in various mammalian cell lines and in various prostate specimens and tumor tissues from the transgenic mouse prostate (TRAMP) model by immunoblotting, qRT-PCR, and by indirect immunohistochemical staining of paraffin embedded prostate specimen. Analysis of a panel of mammalian cell lines showed an increased mRNA and protein expression of specifically myosin IC isoform A in a panel of human and mouse prostate cancer cell lines but not in non-cancer prostate or other (non-prostate-) cancer cell lines. Furthermore, we demonstrate that myosin IC isoform A expression is significantly increased in TRAMP mouse prostate samples with prostatic intraepithelial neoplasia (PIN) lesions and in distant site metastases in lung and liver when compared to matched normal tissues. Our observations demonstrate specific changes in the expression of myosin IC isoform A that are concurrent with the occurrence of prostate cancer in the TRAMP mouse prostate cancer model that closely mimics clinical prostate cancer. These data suggest that elevated levels of myosin IC isoform A may be a potential marker for the detection of prostate cancer.
Collapse
Affiliation(s)
- Ivanna Ihnatovych
- Department of Physiology and Biophysics, University at Buffalo-State University of New York, Buffalo, New York, United States of America
| | - Neil L. Sielski
- Department of Physiology and Biophysics, University at Buffalo-State University of New York, Buffalo, New York, United States of America
| | - Wilma A. Hofmann
- Department of Physiology and Biophysics, University at Buffalo-State University of New York, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
15
|
Sielski NL, Ihnatovych I, Hagen JJ, Hofmann WA. Tissue specific expression of myosin IC isoforms. BMC Cell Biol 2014; 15:8. [PMID: 24618359 PMCID: PMC3984714 DOI: 10.1186/1471-2121-15-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 03/03/2014] [Indexed: 11/13/2022] Open
Abstract
Background Myosin IC is a single headed member of the myosin superfamily that localizes to the cytoplasm and the nucleus and is implicated in a variety of processes in both compartments. We recently identified a novel isoform of myosin IC and showed that the MYOIC gene in mammalian cells encodes three isoforms (isoforms A, B, and C) that differ only in the addition of short isoform-specific N-terminal peptides. The expression pattern of the isoforms and the mechanisms of expression regulation remain unknown. Results To determine the expression patterns of myosin IC isoforms, we performed a comprehensive expression analysis of the two myosin IC isoforms (isoform A and B) that contain isoform-specific sequences. By immunoblotting with isoform-specific antibodies and by qRT-PCR with isoform-specific primer we demonstrate that myosin IC isoforms A and B have distinct expression patterns in mouse tissues. Specifically, we show that myosin IC isoform A is expressed in a tissue specific pattern, while myosin IC isoform B is ubiquitously expressed at comparable levels in mouse tissues. Conclusions The differences in the expression profile of the myosin IC isoforms indicate a tissue-specific MYOIC gene regulation and further suggest that the myosin IC isoforms, despite their high sequence homology, might have tissue-specific and isoform-specific functions.
Collapse
Affiliation(s)
| | | | | | - Wilma A Hofmann
- Department of Physiology and Biophysics, University at Buffalo-State University of New York, 3435 Main Street, Buffalo, NY 14214, USA.
| |
Collapse
|
16
|
Sarshad AA, Percipalle P. New Insight into Role of Myosin Motors for Activation of RNA Polymerases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 311:183-230. [DOI: 10.1016/b978-0-12-800179-0.00004-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
17
|
Arif E, Kumari B, Wagner MC, Zhou W, Holzman LB, Nihalani D. Myo1c is an unconventional myosin required for zebrafish glomerular development. Kidney Int 2013; 84:1154-65. [PMID: 23715127 PMCID: PMC3844053 DOI: 10.1038/ki.2013.201] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 03/14/2013] [Accepted: 03/21/2013] [Indexed: 12/17/2022]
Abstract
The targeting and organization of podocyte slit diaphragm proteins nephrin and neph1 is critical for development and maintenance of a functional glomerular filtration barrier. Myo1c is a non-muscle myosin motor protein that interacts directly with nephrin and neph1 and mediates their intracellular transport to the podocyte intercellular junction. Here we investigated the necessity of Myo1c in podocyte development using zebrafish as a model system. Immunofluorescence microscopy and in situ RNA hybridization analysis of zebrafish embryos showed that Myo1c is widely expressed in various tissues including the zebrafish glomerulus. Knockdown of the Myo1c gene in zebrafish using antisense morpholino derivatives resulted in an abnormal developmental phenotype that included pericardial edema and dilated renal tubules. Ultra-structural analysis of the glomerulus in Myo1c depleted zebrafish showed abnormal podocyte morphology and absence of the slit diaphragm. Consistent with these observations, the glomerular filter permeability appeared altered in zebrafish in which Myo1c expression was attenuated. The specificity of Myo1c knockdown was confirmed by a rescue experiment in which co-injection of Myo1c morpholino derivatives with orthologous Myo1c mRNA prepared from mouse cDNA lessened phenotypic abnormalities including edema in Myo1c morphants. Thus, our results demonstrate that Myo1c is necessary for podocyte morphogenesis.
Collapse
Affiliation(s)
- Ehtesham Arif
- Renal Electrolyte and Hypertension Division, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
18
|
Venit T, Dzijak R, Kalendová A, Kahle M, Rohožková J, Schmidt V, Rülicke T, Rathkolb B, Hans W, Bohla A, Eickelberg O, Stoeger T, Wolf E, Yildirim AÖ, Gailus-Durner V, Fuchs H, de Angelis MH, Hozák P. Mouse nuclear myosin I knock-out shows interchangeability and redundancy of myosin isoforms in the cell nucleus. PLoS One 2013; 8:e61406. [PMID: 23593477 PMCID: PMC3623870 DOI: 10.1371/journal.pone.0061406] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/09/2013] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Nuclear myosin I (NM1) is a nuclear isoform of the well-known "cytoplasmic" Myosin 1c protein (Myo1c). Located on the 11(th) chromosome in mice, NM1 results from an alternative start of transcription of the Myo1c gene adding an extra 16 amino acids at the N-terminus. Previous studies revealed its roles in RNA Polymerase I and RNA Polymerase II transcription, chromatin remodeling, and chromosomal movements. Its nuclear localization signal is localized in the middle of the molecule and therefore directs both Myosin 1c isoforms to the nucleus. METHODOLOGY/PRINCIPAL FINDINGS In order to trace specific functions of the NM1 isoform, we generated mice lacking the NM1 start codon without affecting the cytoplasmic Myo1c protein. Mutant mice were analyzed in a comprehensive phenotypic screen in cooperation with the German Mouse Clinic. Strikingly, no obvious phenotype related to previously described functions has been observed. However, we found minor changes in bone mineral density and the number and size of red blood cells in knock-out mice, which are most probably not related to previously described functions of NM1 in the nucleus. In Myo1c/NM1 depleted U2OS cells, the level of Pol I transcription was restored by overexpression of shRNA-resistant mouse Myo1c. Moreover, we found Myo1c interacting with Pol II. The ratio between Myo1c and NM1 proteins were similar in the nucleus and deletion of NM1 did not cause any compensatory overexpression of Myo1c protein. CONCLUSION/SIGNIFICANCE We observed that Myo1c can replace NM1 in its nuclear functions. Amount of both proteins is nearly equal and NM1 knock-out does not cause any compensatory overexpression of Myo1c. We therefore suggest that both isoforms can substitute each other in nuclear processes.
Collapse
Affiliation(s)
- Tomáš Venit
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, ASCR, v.v.i., Prague, Czech Republic
- Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Rastislav Dzijak
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, ASCR, v.v.i., Prague, Czech Republic
| | - Alžběta Kalendová
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, ASCR, v.v.i., Prague, Czech Republic
| | - Michal Kahle
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, ASCR, v.v.i., Prague, Czech Republic
| | - Jana Rohožková
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, ASCR, v.v.i., Prague, Czech Republic
| | - Volker Schmidt
- Institute of Laboratory Animal Science and Biomodels Austria, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Rülicke
- Institute of Laboratory Animal Science and Biomodels Austria, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg/Munich, Germany
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Wolfgang Hans
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg/Munich, Germany
| | - Alexander Bohla
- German Mouse Clinic, Comprehensive Pneumology Center and Institute of Lung Biology and Disease, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg/Munich, Germany
| | - Oliver Eickelberg
- German Mouse Clinic, Comprehensive Pneumology Center and Institute of Lung Biology and Disease, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg/Munich, Germany
| | - Tobias Stoeger
- German Mouse Clinic, Comprehensive Pneumology Center and Institute of Lung Biology and Disease, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg/Munich, Germany
| | - Eckhard Wolf
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ali Önder Yildirim
- German Mouse Clinic, Comprehensive Pneumology Center and Institute of Lung Biology and Disease, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg/Munich, Germany
| | - Valérie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg/Munich, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg/Munich, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg/Munich, Germany
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
- Member of German Center for Diabetes Research, Neuherberg/Munich, Germany
| | - Pavel Hozák
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, ASCR, v.v.i., Prague, Czech Republic
- * E-mail:
| |
Collapse
|
19
|
Schwab RS, Ihnatovych I, Yunus SZSA, Domaradzki T, Hofmann WA. Identification of signals that facilitate isoform specific nucleolar localization of myosin IC. Exp Cell Res 2013; 319:1111-23. [PMID: 23438938 DOI: 10.1016/j.yexcr.2013.02.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Revised: 02/13/2013] [Accepted: 02/14/2013] [Indexed: 01/29/2023]
Abstract
Myosin IC is a single headed member of the myosin superfamily that localizes to the cytoplasm and the nucleus, where it is involved in transcription by RNA polymerases I and II, intranuclear transport, and nuclear export. In mammalian cells, three isoforms of myosin IC are expressed that differ only in the addition of short isoform-specific N-terminal peptides. Despite the high sequence homology, the isoforms show differences in cellular distribution, in localization to nuclear substructures, and in their interaction with nuclear proteins through yet unknown mechanisms. In this study, we used EGFP-fusion constructs that express truncated or mutated versions of myosin IC isoforms to detect regions that are involved in isoform-specific localization. We identified two nucleolar localization signals (NoLS). One NoLS is located in the myosin IC isoform B specific N-terminal peptide, the second NoLS is located upstream of the neck region within the head domain. We demonstrate that both NoLS are functional and necessary for nucleolar localization of specifically myosin IC isoform B. Our data provide a first mechanistic explanation for the observed functional differences between the myosin IC isoforms and are an important step toward our understanding of the underlying mechanisms that regulate the various and distinct functions of myosin IC isoforms.
Collapse
Affiliation(s)
- Ryan S Schwab
- Department of Physiology and Biophysics, University at Buffalo-State University of New York, Buffalo, NY, USA
| | | | | | | | | |
Collapse
|
20
|
Ihnatovych I, Migocka-Patrzalek M, Dukh M, Hofmann WA. Identification and characterization of a novel myosin Ic isoform that localizes to the nucleus. Cytoskeleton (Hoboken) 2012; 69:555-65. [PMID: 22736583 DOI: 10.1002/cm.21040] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 05/07/2012] [Accepted: 05/09/2012] [Indexed: 11/05/2022]
Abstract
In vertebrates, two myosin Ic isoforms that localize to the cytoplasm and to the nucleus have been characterized. The isoform that predominantly localizes to the nucleus is called nuclear myosin I (NMI). NMI has been identified as a key factor involved in nuclear processes such as transcription by RNA polymerases I and II and intranuclear transport processes. We report here the identification of a previously uncharacterized third MYOIC gene product that is called isoform A. Similar to NMI, this isoform contains a unique N-terminal peptide sequence, localizes to the nucleus and colocalizes with RNA polymerase II. However, unlike NMI, upon exposure to inhibitors of RNA polymerase II transcription the newly identified isoform translocates to nuclear speckles. Furthermore, in contrast to NMI, this new isoform is absent from nucleoli and does not colocalize with RNA polymerase I. Our results suggest an unexpected diversity among nuclear myosin Ic isoforms in respect to their intranuclear localization and interaction with nuclear binding partners that could provide new insights into the regulation of myosin-dependent nuclear processes.
Collapse
Affiliation(s)
- Ivanna Ihnatovych
- Department of Physiology and Biophysics, University at Buffalo-State University of New York, Buffalo, New York 14214, USA
| | | | | | | |
Collapse
|
21
|
Cheng J, Grassart A, Drubin DG. Myosin 1E coordinates actin assembly and cargo trafficking during clathrin-mediated endocytosis. Mol Biol Cell 2012; 23:2891-904. [PMID: 22675027 PMCID: PMC3408416 DOI: 10.1091/mbc.e11-04-0383] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
An actin-dependent role is shown for Myo1E in the trafficking of newly internalized cargo to early endosomes during CME. The results establish for mammalian cells, similar to budding yeast, interdependence in the recruitment of type I myosins, WIP/WIRE, and N-WASP to endocytic sites to assemble F-actin as endocytic vesicles are being formed. Myosin 1E (Myo1E) is recruited to sites of clathrin-mediated endocytosis coincident with a burst of actin assembly. The recruitment dynamics and lifetime of Myo1E are similar to those of tagged actin polymerization regulatory proteins. Like inhibition of actin assembly, depletion of Myo1E causes reduced transferrin endocytosis and a significant delay in transferrin trafficking to perinuclear compartments, demonstrating an integral role for Myo1E in these actin-mediated steps. Mistargeting of GFP-Myo1E or its src-homology 3 domain to mitochondria results in appearance of WIP, WIRE, N-WASP, and actin filaments at the mitochondria, providing evidence for Myo1E's role in actin assembly regulation. These results suggest for mammalian cells, similar to budding yeast, interdependence in the recruitment of type I myosins, WIP/WIRE, and N-WASP to endocytic sites for Arp2/3 complex activation to assemble F-actin as endocytic vesicles are being formed.
Collapse
Affiliation(s)
- Jackie Cheng
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | | |
Collapse
|
22
|
Abstract
Myosin-X (Myo10) is an unconventional myosin with MyTH4-FERM domains that is best known for its striking localization to the tips of filopodia and its ability to induce filopodia. Although the head domain of Myo10 enables it to function as an actin-based motor, its tail contains binding sites for several molecules with central roles in cell biology, including phosphatidylinositol (3,4,5)-trisphosphate, microtubules and integrins. Myo10 also undergoes fascinating long-range movements within filopodia, which appear to represent a newly recognized system of transport. Myo10 is also unusual in that it is a myosin with important roles in the spindle, a microtubule-based structure. Exciting new studies have begun to reveal the structure and single-molecule properties of this intriguing myosin, as well as its mechanisms of regulation and induction of filopodia. At the cellular and organismal level, growing evidence demonstrates that Myo10 has crucial functions in numerous processes ranging from invadopodia formation to cell migration.
Collapse
Affiliation(s)
- Michael L Kerber
- Department of Cell and Molecular Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7545, USA
| | | |
Collapse
|
23
|
DeGiorgis JA, Cavaliere KR, Burbach JPH. Identification of molecular motors in the Woods Hole squid, Loligo pealei: an expressed sequence tag approach. Cytoskeleton (Hoboken) 2011; 68:566-77. [PMID: 21913340 DOI: 10.1002/cm.20531] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 08/26/2011] [Indexed: 12/31/2022]
Abstract
The squid giant axon and synapse are unique systems for studying neuronal function. While a few nucleotide and amino acid sequences have been obtained from squid, large scale genetic and proteomic information is lacking. We have been particularly interested in motors present in axons and their roles in transport processes. Here, to obtain genetic data and to identify motors expressed in squid, we initiated an expressed sequence tag project by single-pass sequencing mRNAs isolated from the stellate ganglia of the Woods Hole Squid, Loligo pealei. A total of 22,689 high quality expressed sequence tag (EST) sequences were obtained and subjected to basic local alignment search tool analysis. Seventy six percent of these sequences matched genes in the National Center for Bioinformatics databases. By CAP3 analysis this library contained 2459 contigs and 7568 singletons. Mining for motors successfully identified six kinesins, six myosins, a single dynein heavy chain, as well as components of the dynactin complex, and motor light chains and accessory proteins. This initiative demonstrates that EST projects represent an effective approach to obtain sequences of interest.
Collapse
Affiliation(s)
- Joseph A DeGiorgis
- Department of Biology, Providence College, Providence, Rhode Island, USA.
| | | | | |
Collapse
|
24
|
Maravillas-Montero JL, Gillespie PG, Patiño-López G, Shaw S, Santos-Argumedo L. Myosin 1c participates in B cell cytoskeleton rearrangements, is recruited to the immunologic synapse, and contributes to antigen presentation. THE JOURNAL OF IMMUNOLOGY 2011; 187:3053-63. [PMID: 21841128 DOI: 10.4049/jimmunol.1004018] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Myosin 1c (Myo1c) is a member of the unconventional class I myosins of vertebrates, which directly link the plasma membrane with the microfilament cortical web. Although this molecular motor has been implicated in cell functions such as cytoskeleton organization, cell motility, nuclear transcription, and endocytosis, its role in hematopoietic cells is largely unknown. In this study, we show that Myo1c is abundantly expressed in murine B lymphocytes and is preferentially located at the plasma membrane, especially in peripheral processes such as microvilli. We observed that this motor concentrates at the growing membrane protrusions generated during B cell spreading and that it is actively recruited to the immune synapse. Interestingly, Myo1c was detected in lipid rafts of B cells and showed strong colocalization with MHC-II, particularly after cross-linking of these molecules. By transfection of a dominant negative form of Myo1c or specific siRNA, we also detected alterations in the spreading and Ag-presenting ability of these cells. The data suggest that Myo1c is involved in the cytoskeleton dynamics and membrane protein anchoring or sorting in B lymphocytes.
Collapse
Affiliation(s)
- José L Maravillas-Montero
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City CP 07360, México
| | | | | | | | | |
Collapse
|
25
|
Motor protein Myo1c is a podocyte protein that facilitates the transport of slit diaphragm protein Neph1 to the podocyte membrane. Mol Cell Biol 2011; 31:2134-50. [PMID: 21402783 DOI: 10.1128/mcb.05051-11] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The podocyte proteins Neph1 and nephrin organize a signaling complex at the podocyte cell membrane that forms the structural framework for a functional glomerular filtration barrier. Mechanisms regulating the movement of these proteins to and from the membrane are currently unknown. This study identifies a novel interaction between Neph1 and the motor protein Myo1c, where Myo1c plays an active role in targeting Neph1 to the podocyte cell membrane. Using in vivo and in vitro experiments, we provide data supporting a direct interaction between Neph1 and Myo1c which is dynamic and actin dependent. Unlike wild-type Myo1c, the membrane localization of Neph1 was significantly reduced in podocytes expressing dominant negative Myo1c. In addition, Neph1 failed to localize at the podocyte cell membrane and cell junctions in Myo1c-depleted podocytes. We further demonstrate that similarly to Neph1, Myo1c also binds nephrin and reduces its localization at the podocyte cell membrane. A functional analysis of Myo1c knockdown cells showed defects in cell migration, as determined by a wound assay. In addition, the ability to form tight junctions was impaired in Myo1c knockdown cells, as determined by transepithelial electric resistance (TER) and bovine serum albumin (BSA) permeability assays. These results identify a novel Myo1c-dependent molecular mechanism that mediates the dynamic organization of Neph1 and nephrin at the slit diaphragm and is critical for podocyte function.
Collapse
|
26
|
Toyoda T, An D, Witczak CA, Koh HJ, Hirshman MF, Fujii N, Goodyear LJ. Myo1c regulates glucose uptake in mouse skeletal muscle. J Biol Chem 2010; 286:4133-40. [PMID: 21127070 DOI: 10.1074/jbc.m110.174938] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Contraction and insulin promote glucose uptake in skeletal muscle through GLUT4 translocation to cell surface membranes. Although the signaling mechanisms leading to GLUT4 translocation have been extensively studied in muscle, the cellular transport machinery is poorly understood. Myo1c is an actin-based motor protein implicated in GLUT4 translocation in adipocytes; however, the expression profile and role of Myo1c in skeletal muscle have not been investigated. Myo1c protein abundance was higher in more oxidative skeletal muscles and heart. Voluntary wheel exercise (4 weeks, 8.2 ± 0.8 km/day), which increased the oxidative profile of the triceps muscle, significantly increased Myo1c protein levels by ∼2-fold versus sedentary controls. In contrast, high fat feeding (9 weeks, 60% fat) significantly reduced Myo1c by 17% in tibialis anterior muscle. To study Myo1c regulation of glucose uptake, we expressed wild-type Myo1c or Myo1c mutated at the ATPase catalytic site (K111A-Myo1c) in mouse tibialis anterior muscles in vivo and assessed glucose uptake in vivo in the basal state, in response to 15 min of in situ contraction, and 15 min following maximal insulin injection (16.6 units/kg of body weight). Expression of wild-type Myo1c or K111A-Myo1c had no effect on basal glucose uptake. However, expression of wild-type Myo1c significantly increased contraction- and insulin-stimulated glucose uptake, whereas expression of K111A-Myo1c decreased both contraction-stimulated and insulin-stimulated glucose uptake. Neither wild-type nor K111A-Myo1c expression altered GLUT4 expression, and neither affected contraction- or insulin-stimulated signaling proteins. Myo1c is a novel mediator of both insulin-stimulated and contraction-stimulated glucose uptake in skeletal muscle.
Collapse
Affiliation(s)
- Taro Toyoda
- Section on Integrative Physiology and Metabolism, the Joslin Diabetes Center, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Komaba S, Coluccio LM. Localization of myosin 1b to actin protrusions requires phosphoinositide binding. J Biol Chem 2010; 285:27686-93. [PMID: 20610386 DOI: 10.1074/jbc.m109.087270] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Myosin 1b (Myo1b), a class I myosin, is a widely expressed, single-headed, actin-associated molecular motor. Transient kinetic and single-molecule studies indicate that it is kinetically slow and responds to tension. Localization and subcellular fractionation studies indicate that Myo1b associates with the plasma membrane and certain subcellular organelles such as endosomes and lysosomes. Whether Myo1b directly associates with membranes is unknown. We demonstrate here that full-length rat Myo1b binds specifically and with high affinity to phosphatidylinositol 4,5-bisphosphate (PIP(2)) and phosphatidylinositol 3,4,5-triphosphate (PIP(3)), two phosphoinositides that play important roles in cell signaling. Binding is not Ca(2+)-dependent and does not involve the calmodulin-binding IQ region in the neck domain of Myo1b. Furthermore, the binding site is contained entirely within the C-terminal tail region, which contains a putative pleckstrin homology domain. Single mutations in the putative pleckstrin homology domain abolish binding of the tail domain of Myo1b to PIP(2) and PIP(3) in vitro. These same mutations alter the distribution of Myc-tagged Myo1b at membrane protrusions in HeLa cells where PIP(2) localizes. In addition, we found that motor activity is required for Myo1b localization in filopodia. These results suggest that binding of Myo1b to phosphoinositides plays an important role in vivo by regulating localization to actin-enriched membrane projections.
Collapse
Affiliation(s)
- Shigeru Komaba
- Boston Biomedical Research Institute, Watertown, Massachusetts 02472, USA
| | | |
Collapse
|
28
|
Mehta IS, Amira M, Harvey AJ, Bridger JM. Rapid chromosome territory relocation by nuclear motor activity in response to serum removal in primary human fibroblasts. Genome Biol 2010; 11:R5. [PMID: 20070886 PMCID: PMC2847717 DOI: 10.1186/gb-2010-11-1-r5] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Revised: 11/23/2009] [Accepted: 01/13/2010] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Radial chromosome positioning in interphase nuclei is nonrandom and can alter according to developmental, differentiation, proliferation, or disease status. However, it is not yet clear when and how chromosome repositioning is elicited. RESULTS By investigating the positioning of all human chromosomes in primary fibroblasts that have left the proliferative cell cycle, we have demonstrated that in cells made quiescent by reversible growth arrest, chromosome positioning is altered considerably. We found that with the removal of serum from the culture medium, chromosome repositioning took less than 15 minutes, required energy and was inhibited by drugs affecting the polymerization of myosin and actin. We also observed that when cells became quiescent, the nuclear distribution of nuclear myosin 1 beta was dramatically different from that in proliferating cells. If we suppressed the expression of nuclear myosin 1 beta by using RNA-interference procedures, the movement of chromosomes after 15 minutes in low serum was inhibited. When high serum was restored to the serum-starved cultures, chromosome repositioning was evident only after 24 to 36 hours, and this coincided with a return to a proliferating distribution of nuclear myosin 1 beta. CONCLUSIONS These findings demonstrate that genome organization in interphase nuclei is altered considerably when cells leave the proliferative cell cycle and that repositioning of chromosomes relies on efficient functioning of an active nuclear motor complex that contains nuclear myosin 1 beta.
Collapse
Affiliation(s)
- Ishita S Mehta
- Centre for Cell and Chromosome Biology, Division of Biosciences, School of Health Sciences and Social Care, Brunel University, Kingston Lane, Uxbridge, UB8 3PH, UK.
| | | | | | | |
Collapse
|
29
|
Hofmann WA, Richards TA, de Lanerolle P. Ancient animal ancestry for nuclear myosin. J Cell Sci 2009; 122:636-43. [PMID: 19225126 DOI: 10.1242/jcs.030205] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The identification of nuclear myosin I (NMI) has raised the possibility that myosin might have had an early functional role in the eukaryotic nucleus. To investigate this possibility, we examined the molecular evolution of the vertebrate myosin-I proteins. We found that myosin I has undergone at least five duplication events in the common ancestor of the vertebrates (vertebrate-specific duplications), leading to nine myosin-I vertebrate gene families, followed by two additional myosin-I duplication events in the lineage leading to modern fish. This expansion suggests a large-scale adaptive radiation in myosin-I function in an early phase of vertebrate evolution. The branching order of the evolutionary tree suggests that the functional role of NMI predates this expansion. More specifically, in the tunicate Ciona intestinalis, we found a myosin-I protein that localizes to the nucleus, but that branches on phylogenetic trees before the duplication that led to vertebrate myosin IC and myosin IH. This relationship suggests that the common ancestor of these three proteins encoded a nuclear isoform and that the localization of myosin I to the nucleus predates the origin of the vertebrates. Thus, a functional role for NMI appears to have been present at an early stage of animal evolution prior to the rise of both myosin IC and the vertebrates, as NMI was present in the last common ancestor of vertebrates and tunicates.
Collapse
Affiliation(s)
- Wilma A Hofmann
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | |
Collapse
|
30
|
Okumura T, Utsuno H, Kuroda J, Gittenberger E, Asami T, Matsuno K. The development and evolution of left-right asymmetry in invertebrates: lessons from Drosophila and snails. Dev Dyn 2009; 237:3497-515. [PMID: 19035360 DOI: 10.1002/dvdy.21788] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The unique nature of body handedness, which is distinct from the anteroposterior and dorsoventral polarities, has been attracting growing interest in diverse biological disciplines. Recent research progress on the left-right asymmetry of animal development has focused new attention on the mechanisms underlying the development and evolution of invertebrate handedness. This exploratory review of currently available information illuminates the prospective value of Drosophila and pulmonate snails for innovative new research aimed at elucidating these mechanisms. For example, findings in Drosophila and snails suggest that an actin filament-dependent mechanism may be evolutionarily conserved in protostomes. The polarity conservation of primary asymmetry across most metazoan phyla, which visceral handedness represents, indicates developmental constraint and purifying selection as possible but unexplored mechanisms. Comparative studies using Drosophila and snails, which have the great advantages of using genetic and evolutionary approaches, will accelerate our understanding of the mechanisms governing the conservation and diversity of animal handedness.
Collapse
Affiliation(s)
- Takashi Okumura
- Department of Biological Science and Technology, Tokyo University of Science, Noda, Japan
| | | | | | | | | | | |
Collapse
|
31
|
Hozumi S, Maeda R, Taniguchi-Kanai M, Okumura T, Taniguchi K, Kawakatsu Y, Nakazawa N, Hatori R, Matsuno K. Head region of unconventional myosin I family members is responsible for the organ-specificity of their roles in left-right polarity in Drosophila. Dev Dyn 2009; 237:3528-37. [PMID: 18521948 DOI: 10.1002/dvdy.21583] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In Drosophila, Myosin31DF (Myo31DF), encoding a Myosin ID protein, has crucial roles in left-right (LR) asymmetric development. Loss of Myo31DF function leads to laterality inversion for many organs, including the embryonic gut. Here, we found that Myo31DF was required before LR asymmetric morphogenesis in the hindgut, suggesting it functions in LR patterning instead of directly in hindgut morphological changes. Myosin61F (Myo61F) encodes another Myosin I, and Myo31DF or Myo61F overexpression reverses the laterality of different organs. Myo31DF and Myo61F have domains conserved in Myosin proteins, particularly in the proteins' head regions. We studied the roles of these domains in LR patterning using overexpression analysis. The Actin-binding and ATP-binding domains were essential for both proteins, but the IQ domains, binding sites for Myosin light chains, were required only by Myo31DF. Our results also suggest that the organ specificities of the Myo31DF and Myo61F activities depended on their head regions.
Collapse
Affiliation(s)
- Shunya Hozumi
- Department of Biological Science and Technology, Tokyo University of Science, Noda, Chiba, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Odronitz F, Kollmar M. Drawing the tree of eukaryotic life based on the analysis of 2,269 manually annotated myosins from 328 species. Genome Biol 2008; 8:R196. [PMID: 17877792 PMCID: PMC2375034 DOI: 10.1186/gb-2007-8-9-r196] [Citation(s) in RCA: 289] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2007] [Revised: 09/17/2007] [Accepted: 09/18/2007] [Indexed: 01/03/2023] Open
Abstract
The tree of eukaryotic life was reconstructed based on the analysis of 2,269 myosin motor domains from 328 organisms, confirming some accepted relationships of major taxa and resolving disputed and preliminary classifications. Background The evolutionary history of organisms is expressed in phylogenetic trees. The most widely used phylogenetic trees describing the evolution of all organisms have been constructed based on single-gene phylogenies that, however, often produce conflicting results. Incongruence between phylogenetic trees can result from the violation of the orthology assumption and stochastic and systematic errors. Results Here, we have reconstructed the tree of eukaryotic life based on the analysis of 2,269 myosin motor domains from 328 organisms. All sequences were manually annotated and verified, and were grouped into 35 myosin classes, of which 16 have not been proposed previously. The resultant phylogenetic tree confirms some accepted relationships of major taxa and resolves disputed and preliminary classifications. We place the Viridiplantae after the separation of Euglenozoa, Alveolata, and Stramenopiles, we suggest a monophyletic origin of Entamoebidae, Acanthamoebidae, and Dictyosteliida, and provide evidence for the asynchronous evolution of the Mammalia and Fungi. Conclusion Our analysis of the myosins allowed combining phylogenetic information derived from class-specific trees with the information of myosin class evolution and distribution. This approach is expected to result in superior accuracy compared to single-gene or phylogenomic analyses because the orthology problem is resolved and a strong determinant not depending on any technical uncertainties is incorporated, the class distribution. Combining our analysis of the myosins with high quality analyses of other protein families, for example, that of the kinesins, could help in resolving still questionable dependencies at the origin of eukaryotic life.
Collapse
Affiliation(s)
- Florian Odronitz
- Department of NMR-based Structural Biology, Max-Planck-Institute for Biophysical Chemistry, Am Fassberg, 37077 Goettingen, Germany
| | - Martin Kollmar
- Department of NMR-based Structural Biology, Max-Planck-Institute for Biophysical Chemistry, Am Fassberg, 37077 Goettingen, Germany
| |
Collapse
|
33
|
Tsiavaliaris G, Fujita-Becker S, Dürrwang U, Diensthuber RP, Geeves MA, Manstein DJ. Mechanism, regulation, and functional properties of Dictyostelium myosin-1B. J Biol Chem 2007; 283:4520-7. [PMID: 18089562 DOI: 10.1074/jbc.m708113200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Myosin-1B is one of three long tailed class-1 myosins containing an ATP-insensitive actin-binding site in the tail region that are produced in Dictyostelium discoideum. Myosin-1B localizes to actin-rich structures at the leading edge of migrating cells where it has been implicated in the formation and retraction of membrane projections, the recycling of plasma membrane components, and intracellular particle transport. Here, we have used a combination of molecular engineering approaches to describe the kinetic and motile properties of the myosin-1B motor and its regulation by TEDS site phosphorylation. Our results show that myosin-1B is a low duty ratio motor and displays the fastest nucleotide binding kinetics of any of the Dictyostelium class-1 myosins studied so far. Different from Dictyostelium myosin-1D and myosin-1E, dephosphorylated myosin-1B is not inactivated but moves actin filaments efficiently, albeit at an up to 8-fold slower velocity in the in vitro motility assay. A further difference is that myosin-1B lacks the ability to switch between rapid movement and bearing tension upon physiological changes of free Mg2+ ions. In this respect, its motor properties appear to be more closely related to Dictyostelium myosin-2 and rabbit skeletal muscle myosin.
Collapse
Affiliation(s)
- Georgios Tsiavaliaris
- Institute for Biophysical Chemistry, OE 4350, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30623 Hannover, Germany.
| | | | | | | | | | | |
Collapse
|
34
|
Schietroma C, Yu HY, Wagner MC, Umbach JA, Bement WM, Gundersen CB. A role for myosin 1e in cortical granule exocytosis in Xenopus oocytes. J Biol Chem 2007; 282:29504-13. [PMID: 17702742 PMCID: PMC2820112 DOI: 10.1074/jbc.m705825200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Xenopus oocytes undergo dynamic structural changes during maturation and fertilization. Among these, cortical granule exocytosis and compensatory endocytosis provide effective models to study membrane trafficking. This study documents an important role for myosin 1e in cortical granule exocytosis. Myosin 1e is expressed at the earliest stage that cortical granule exocytosis can be detected in oocytes. Prior to exocytosis, myosin 1e relocates to the surface of cortical granules. Overexpression of myosin 1e augments the kinetics of cortical granule exocytosis, whereas tail-derived fragments of myosin 1e inhibit this secretory event (but not constitutive exocytosis). Finally, intracellular injection of myosin 1e antibody inhibits cortical granule exocytosis. Further experiments identified cysteine string proteins as interacting partners for myosin 1e. As constituents of the membrane of cortical granules, cysteine string proteins are also essential for cortical granule exocytosis. Future investigation of the link between myosin 1e and cysteine string proteins should help to clarify basic mechanisms of regulated exocytosis.
Collapse
Affiliation(s)
- Cataldo Schietroma
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, UCLA, Los Angeles, California 90095
| | - Hoi-Ying Yu
- Department of Zoology, University of Wisconsin, Madison, Wisconsin 53706
| | - Mark C. Wagner
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Joy A. Umbach
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, UCLA, Los Angeles, California 90095
| | - William M. Bement
- Department of Zoology, University of Wisconsin, Madison, Wisconsin 53706
| | - Cameron B. Gundersen
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, UCLA, Los Angeles, California 90095
| |
Collapse
|
35
|
Hegan PS, Mermall V, Tilney LG, Mooseker MS. Roles for Drosophila melanogaster myosin IB in maintenance of enterocyte brush-border structure and resistance to the bacterial pathogen Pseudomonas entomophila. Mol Biol Cell 2007; 18:4625-36. [PMID: 17855510 PMCID: PMC2043548 DOI: 10.1091/mbc.e07-02-0191] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Drosophila myosin IB (Myo1B) is one of two class I myosins in the Drosophila genome. In the larval and adult midgut enterocyte, Myo1B is present within the microvillus (MV) of the apical brush border (BB) where it forms lateral tethers between the MV membrane and underlying actin filament core. Expression of green fluorescent protein-Myo1B tail domain in the larval gut showed that the tail domain is sufficient for localization of Myo1B to the BB. A Myo1B deletion mutation exhibited normal larval gut physiology with respect to food uptake, clearance, and pH regulation. However, there is a threefold increase in terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive enterocyte nuclei in the Myo1B mutant. Ultrastructural analysis of mutant midgut revealed many perturbations in the BB, including membrane tethering defects, MV vesiculation, and membrane shedding. The apical localization of both singed (fascin) and Dmoesin is impaired. BBs isolated from mutant and control midgut revealed that the loss of Myo1B causes the BB membrane and underlying cytoskeleton to become destabilized. Myo1B mutant larvae also exhibit enhanced sensitivity to oral infection by the bacterial pathogen Pseudomonas entomophila, and severe cytoskeletal defects are observed in the BB of proximal midgut epithelial cells soon after infection. Resistance to P. entomophila infection is restored in Myo1B mutant larvae expressing a Myo1B transgene. These results indicate that Myo1B may play a role in the local midgut response pathway of the Imd innate immune response to Gram-negative bacterial infection.
Collapse
Affiliation(s)
- Peter S Hegan
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06520, USA.
| | | | | | | |
Collapse
|
36
|
Spéder P, Noselli S. Left-right asymmetry: class I myosins show the direction. Curr Opin Cell Biol 2006; 19:82-7. [PMID: 17174542 DOI: 10.1016/j.ceb.2006.12.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Accepted: 12/07/2006] [Indexed: 02/03/2023]
Abstract
Myosins are actin-based molecular motors that are found in almost all eukaryotes. Phylogenetic analysis allows the discrimination of 37 different types of myosins, most with unknown functions. Recent work in Drosophila has revealed a crucial role for type ID unconventional myosin in left-right asymmetry. Mutations in Myosin ID completely reverse the left-right axis (situs inversus), a phenotype that is dependent on an intact actin cytoskeleton. How this myosin might orient the left-right axis has began to be elucidated by showing that it interacts directly with beta-catenin, suggesting that myosin ID interacts with the adherens junction to control the direction of organ looping. This is the first demonstration of a role of a myosin in body patterning.
Collapse
Affiliation(s)
- Pauline Spéder
- Institute of Signalling, Developmental Biology & Cancer, CNRS - UMR 6543, University of Nice Sophia-Antipolis, Parc Valrose, 06108 Nice cedex 2, France
| | | |
Collapse
|
37
|
Hofmann WA, Vargas GM, Ramchandran R, Stojiljkovic L, Goodrich JA, de Lanerolle P. Nuclear myosin I is necessary for the formation of the first phosphodiester bond during transcription initiation by RNA polymerase II. J Cell Biochem 2006; 99:1001-9. [PMID: 16960872 DOI: 10.1002/jcb.21035] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The nuclear isoform of myosin, Nuclear Myosin I (NMI) is involved in transcription by RNA polymerase I. Previous experiments showing that antibodies to NMI inhibit transcription by RNA polymerase II using HeLa cell nuclear extract (NE) suggested that NMI might be a general transcription factor for RNA polymerases. In this study we used a minimal in vitro transcription system to investigate the involvement of NMI in transcription by RNA polymerase II in detail. We demonstrate that NMI co-purifies with RNA polymerase II and that NMI is necessary for basal transcription by RNA polymerase II because antibodies to NMI inhibit transcription while adding NMI stimulates transcription. Further investigation revealed that NMI is specifically involved in transcription initiation. Finally, by employing an abortive transcription initiation assay, we demonstrate that NMI is crucial for the formation of the first phosphodiester bond during transcription initiation.
Collapse
Affiliation(s)
- Wilma A Hofmann
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
38
|
Sumoza-Toledo A, Gillespie PG, Romero-Ramirez H, Ferreira-Ishikawa HC, Larson RE, Santos-Argumedo L. Differential localization of unconventional myosin I and nonmuscle myosin II during B cell spreading. Exp Cell Res 2006; 312:3312-22. [PMID: 16919270 DOI: 10.1016/j.yexcr.2006.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Revised: 07/01/2006] [Accepted: 07/07/2006] [Indexed: 02/06/2023]
Abstract
Cross-linking of CD44 in vitro promotes chemokinesis and actin-based dendrite formation in T and B cells. However, the mechanisms by which the adhesion molecule CD44 induces cytoskeleton activation in lymphocytes are still poorly understood. In this study, we have investigated whether myosin isoforms are involved in CD44-dependent dendrite formation in activated B cells. Pharmacological inhibition of myosin with 2,3-butanedione monoxime strongly affected spreading and dendrite formation, suggesting that these cellular motors may participate in these phenomena. Furthermore, immunofluorescence analysis showed differences in subcellular localization of class I and class II myosin during B cell spreading. In response to CD44 cross-linking, myosin-1c was polarized to lamellipodia, where F-actin was high. In contrast, the distribution of cytosplasmic nonmuscle class II myosin was not altered. Expressions of myosin-1c and II were also demonstrated in B cells by Western blot. Although the inhibition of PLCgamma, PI3K and MEK-1 activation affected the spreading and dendrite formation in activated B cells, only PLCgamma and MEK-1 inhibition correlated with absence of myosin-1c polarization. Additionally, myosin-1c polarization was observed upon cross-linking of other surface molecules, suggesting a common mechanism for B cell spreading. This work shows that class I and class II myosin are expressed in B cells, are differentially distributed, and may participate in the morphological changes of these cells.
Collapse
Affiliation(s)
- Adriana Sumoza-Toledo
- Department of Molecular Biomedicine, Centro de Investigación y Estudios Avanzados, Av. IPN #2508. Col. Zacatenco. CP 07360, México, D.F., México
| | | | | | | | | | | |
Collapse
|
39
|
Hofmann WA, Johnson T, Klapczynski M, Fan JL, de Lanerolle P. From transcription to transport: emerging roles for nuclear myosin IThis paper is one of a selection of papers published in this Special Issue, entitled 27th International West Coast Chromatin and Chromosome Conference, and has undergone the Journal's usual peer review process. Biochem Cell Biol 2006; 84:418-26. [PMID: 16936815 DOI: 10.1139/o06-069] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Myosins are a superfamily of actin-activated ATPases that, in the cytoplasm, work together with actin as molecular motors. The presence of actin in the nucleus has been known for many years. The demonstration of a nuclear isoform of a myosin, nuclear myosin I (NMI), stimulated a great deal of interest in possible intranuclear motor functions of an acto–NMI complex. NMI has been shown to be involved in transcription by RNA polymerases I and II. In both cases, NMI interacts with the respective polymerase and is critically involved in the basic process of transcription. A recent study on intranuclear long-range chromosome movement has now demonstrated a role for NMI in the translocation of chromosome regions as well. Moreover, this movement is based on an active and directed process that is facilitated by an acto–NMI complex, establishing for the first time a functional role for a motor complex consisting of actin and a myosin in the nucleus.
Collapse
Affiliation(s)
- Wilma A Hofmann
- Department of Physiology and Biophysics, University of IL at Chicago, 835 S. Wolcott, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
40
|
Dürrwang U, Fujita-Becker S, Erent M, Kull FJ, Tsiavaliaris G, Geeves MA, Manstein DJ. Dictyostelium myosin-IE is a fast molecular motor involved in phagocytosis. J Cell Sci 2006; 119:550-8. [PMID: 16443752 DOI: 10.1242/jcs.02774] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Class I myosins are single-headed motor proteins, implicated in various motile processes including organelle translocation, ion-channel gating, and cytoskeleton reorganization. Here we describe the cellular localization of myosin-IE and its role in the phagocytic uptake of solid particles and cells. A complete analysis of the kinetic and motor properties of Dictyostelium discoideum myosin-IE was achieved by the use of motor domain constructs with artificial lever arms. Class I myosins belonging to subclass IC like myosin-IE are thought to be tuned for tension maintenance or stress sensing. In contrast to this prediction, our results show myosin-IE to be a fast motor. Myosin-IE motor activity is regulated by myosin heavy chain phosphorylation, which increases the coupling efficiency between the actin and nucleotide binding sites tenfold and the motile activity more than fivefold. Changes in the level of free Mg(2+) ions, which are within the physiological range, are shown to modulate the motor activity of myosin-IE by inhibiting the release of adenosine diphosphate.
Collapse
Affiliation(s)
- Ulrike Dürrwang
- Abteilung Biophysik, Max-Planck Institut für medizinische Forschung, Jahnstr. 29, 69120 Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
41
|
Hozumi S, Maeda R, Taniguchi K, Kanai M, Shirakabe S, Sasamura T, Spéder P, Noselli S, Aigaki T, Murakami R, Matsuno K. An unconventional myosin in Drosophila reverses the default handedness in visceral organs. Nature 2006; 440:798-802. [PMID: 16598258 DOI: 10.1038/nature04625] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Accepted: 02/02/2006] [Indexed: 11/08/2022]
Abstract
The internal organs of animals often have left-right asymmetry. Although the formation of the anterior-posterior and dorsal-ventral axes in Drosophila is well understood, left-right asymmetry has not been extensively studied. Here we find that the handedness of the embryonic gut and the adult gut and testes is reversed (not randomized) in viable and fertile homozygous Myo31DF mutants. Myo31DF encodes an unconventional myosin, Drosophila MyoIA (also referred to as MyoID in mammals; refs 3, 4), and is the first actin-based motor protein to be implicated in left-right patterning. We find that Myo31DF is required in the hindgut epithelium for normal embryonic handedness. Disruption of actin filaments in the hindgut epithelium randomizes the handedness of the embryonic gut, suggesting that Myo31DF function requires the actin cytoskeleton. Consistent with this, we find that Myo31DF colocalizes with the cytoskeleton. Overexpression of Myo61F, another myosin I (ref. 4), reverses the handedness of the embryonic gut, and its knockdown also causes a left-right patterning defect. These two unconventional myosin I proteins may have antagonistic functions in left-right patterning. We suggest that the actin cytoskeleton and myosin I proteins may be crucial for generating left-right asymmetry in invertebrates.
Collapse
Affiliation(s)
- Shunya Hozumi
- Department of Biological Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
de Lanerolle P, Johnson T, Hofmann WA. Actin and myosin I in the nucleus: what next? Nat Struct Mol Biol 2005; 12:742-6. [PMID: 16142228 DOI: 10.1038/nsmb983] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2005] [Accepted: 08/04/2005] [Indexed: 11/09/2022]
Abstract
Several recent publications have demonstrated the importance of nuclear actin and nuclear myosin I in transcription. Here we review these publications and their implications. In addition, we discuss some important issues that should be addressed to gain a more comprehensive understanding of how these traditionally 'cytoplasmic' proteins are involved in transcription. We propose highly speculative models and mechanisms solely to stimulate thought and experimentation in this area.
Collapse
Affiliation(s)
- Primal de Lanerolle
- the Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois 60612, USA.
| | | | | |
Collapse
|
43
|
Lund LM, Machado VM, McQuarrie IG. Axonal isoforms of myosin-I. Biochem Biophys Res Commun 2005; 330:857-64. [PMID: 15809075 DOI: 10.1016/j.bbrc.2005.02.187] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2005] [Indexed: 10/25/2022]
Abstract
We have examined spinal motor neurons in Sprague-Dawley rats to further characterize a mechanoenzyme, myosin-Igamma (myr4), which is found in high concentration during axon tract formation in neonates. We raised an antibody to myr4 and made riboprobes for in situ hybridization. Myr4 mRNA was abundant in spinal cord motor neurons (particularly during axon regrowth). Nerves undergoing Wallerian degeneration (from a crush 7 days earlier) showed anti-myr4 labeling of the axolemma and SER--after microtubules, neurofilaments, and F-actin had already been degraded--which is consistent with a described lipid-binding domain in the tail region of myosin-Is. Newly synthesized myr4 was carried in axons by the slow component (SC) of axonal transport at 1-8 mm/day, whereas, none was carried by the fast component (FC). We conclude that SC delivers myr4 to the cytoplasmic surfaces of stationary axonal membranes (SER and axolemma). This positioning would anchor the tail domain of myr4 and leave the catalytic head domain free to interact with F-actin.
Collapse
Affiliation(s)
- Linda M Lund
- Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, USA.
| | | | | |
Collapse
|
44
|
Batters C, Wallace MI, Coluccio LM, Molloy JE. A model of stereocilia adaptation based on single molecule mechanical studies of myosin I. Philos Trans R Soc Lond B Biol Sci 2005; 359:1895-905. [PMID: 15647165 PMCID: PMC1693475 DOI: 10.1098/rstb.2004.1559] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have used an optical tweezers-based apparatus to perform single molecule mechanical experiments using the unconventional myosins, Myo1b and Myo1c. The single-headed nature and slow ATPase kinetics of these myosins make them ideal for detailed studies of the molecular mechanism of force generation by acto-myosin. Myo1c exhibits several features that have not been seen using fast skeletal muscle myosin II. (i) The working stroke occurs in two, distinct phases, producing an initial 3 nm and then a further 1.5 nm of movement. (ii) Two types of binding interaction were observed: short-lived ATP-independent binding events that produced no movement and longer-lived, ATP-dependent events that produced a full working stroke. The stiffness of both types of interaction was similar. (iii) In a new type of experiment, using feedback to apply controlled displacements to a single acto-myosin cross-bridge, we found abrupt changes in force during attachment of the acto-Myo1b cross-bridge, a result that is consistent with the classical 'T2' behaviour of single muscle fibres. Given that these myosins might exhibit the classical T2 behaviour, we propose a new model to explain the slow phase of sensory adaptation of the hair cells of the inner ear.
Collapse
Affiliation(s)
- Christopher Batters
- Division of Physical Biochemistry, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | | | | | | |
Collapse
|
45
|
Abstract
Axonal transport in neurons has been shown to be microtubule dependent, driven by the molecular motor proteins kinesin and dynein. However, organelles undergoing fast transport can often pause or rapidly change directions without apparent dissociation from their transport tracks. Cytoskeletal polymers such as neurofilaments and microtubules have also been shown to make infrequent but rapid movements in axons indicating that their transport is likely to involve molecular motors. In addition, neurons have multiple compartments that are devoid of microtubules where transport of organelles is still seen to occur. These areas are rich in other cytoskeletal polymers such as actin filaments. Transported organelles have been shown to associate with multiple motor proteins including myosins. This suggests that nonmicrotubule-based transport may be myosin driven. In this review we will focus our attention on myosin motors known to be present in neurons and evaluate the evidence that they contribute to transport or other functions in the different compartments of the neuron.
Collapse
Affiliation(s)
- Paul C Bridgman
- Department of Anatomy and Neurobiology, Washington University School of Medicine, Box 8108, 660 S. Euclid Avenue, St. Louis, Missouri 63110, USA.
| |
Collapse
|
46
|
Abstract
Development of the nervous system requires remarkable changes in cell structure that are dependent upon the cytoskeleton. The importance of specific components of the neuronal cytoskeleton, such as microtubules and neurofilaments, to neuronal function and development has been well established. Recently, increasing focus has been put on understanding the functional role of the actin cytoskeleton in neurons. Important modulators of the actin cytoskeleton are the large family of myosins, many of which (classes I, II, III, V, VI, VII, IX, and XV; Fig. 1) are expressed in developing neurons or sensory cells. Myosins are force-producing proteins that have been implicated in a wide variety of cellular functions in the developing nervous system, including neuronal migration, process outgrowth, and growth cone motility, as well as other aspects of morphogenesis, axonal transport, and synaptic and sensory functions. We review the roles that neuronal myosins play in these functions with particular focus on the first three events listed above, as well as sensory function.
Collapse
Affiliation(s)
- Michael E Brown
- Department of Anatomy and Neurobiology, Washington University School of Medicine, Box 8108, 660 S. Euclid Avenue, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
47
|
Bose A, Guilherme A, Robida SI, Nicoloro SMC, Zhou QL, Jiang ZY, Pomerleau DP, Czech MP. Glucose transporter recycling in response to insulin is facilitated by myosin Myo1c. Nature 2002; 420:821-4. [PMID: 12490950 DOI: 10.1038/nature01246] [Citation(s) in RCA: 207] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2002] [Accepted: 10/10/2002] [Indexed: 01/18/2023]
Abstract
Insulin stimulates glucose uptake in muscle and adipocytes by signalling the translocation of GLUT4 glucose transporters from intracellular membranes to the cell surface. The translocation of GLUT4 may involve signalling pathways that are both independent of and dependent on phosphatidylinositol-3-OH kinase (PI(3)K). This translocation also requires the actin cytoskeleton, and the rapid movement of GLUT4 along linear tracks may be mediated by molecular motors. Here we report that the unconventional myosin Myo1c is present in GLUT4-containing vesicles purified from 3T3-L1 adipocytes. Myo1c, which contains a motor domain, three IQ motifs and a carboxy-terminal cargo domain, is highly expressed in primary and cultured adipocytes. Insulin enhances the localization of Myo1c with GLUT4 in cortical tubulovesicular structures associated with actin filaments, and this colocalization is insensitive to wortmannin. Insulin-stimulated translocation of GLUT4 to the adipocyte plasma membrane is augmented by the expression of wild-type Myo1c and inhibited by a dominant-negative cargo domain of Myo1c. A decrease in the expression of endogenous Myo1c mediated by small interfering RNAs inhibits insulin-stimulated uptake of 2-deoxyglucose. Thus, myosin Myo1c functions in a PI(3)K-independent insulin signalling pathway that controls the movement of intracellular GLUT4-containing vesicles to the plasma membrane.
Collapse
Affiliation(s)
- Avirup Bose
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Gillespie PG, Cyr JL. Calmodulin binding to recombinant myosin-1c and myosin-1c IQ peptides. BMC BIOCHEMISTRY 2002; 3:31. [PMID: 12453307 PMCID: PMC139967 DOI: 10.1186/1471-2091-3-31] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2002] [Accepted: 11/26/2002] [Indexed: 11/10/2022]
Abstract
BACKGROUND Bullfrog myosin-1c contains three previously recognized calmodulin-binding IQ domains (IQ1, IQ2, and IQ3) in its neck region; we identified a fourth IQ domain (IQ4), located immediately adjacent to IQ3. How calmodulin binds to these IQ domains is the subject of this report. RESULTS In the presence of EGTA, calmodulin bound to synthetic peptides corresponding to IQ1, IQ2, and IQ3 with Kd values of 2-4 microM at normal ionic strength; the interaction with an IQ4 peptide was much weaker. Ca2+ substantially weakened the calmodulin-peptide affinity for all of the IQ peptides except IQ3. To reveal how calmodulin bound to the linearly arranged IQ domains of the myosin-1c neck, we used hydrodynamic measurements to determine the stoichiometry of complexes of calmodulin and myosin-1c. Purified myosin-1c and T701-Myo1c (a myosin-1c fragment with all four IQ domains and the C-terminal tail) each bound 2-3 calmodulin molecules. At a physiologically relevant temperature (25 degrees C) and under low-Ca2+ conditions, T701-Myo1c bound two calmodulins in the absence and three calmodulins in the presence of 5 microM free calmodulin. Ca2+ dissociated nearly all calmodulins from T701-Myo1c at 25 degrees C; one calmodulin was retained if 5 microM free calmodulin was present. CONCLUSIONS We inferred from these data that at 25 degrees C and normal cellular concentrations of calmodulin, calmodulin is bound to IQ1, IQ2, and IQ3 of myosin-1c when Ca2+ is low. The calmodulin bound to one of these IQ domains, probably IQ2, is only weakly associated. Upon Ca2+ elevation, all calmodulin except that bound to IQ3 should dissociate.
Collapse
Affiliation(s)
- Peter G Gillespie
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland OR 97239, USA
| | - Janet L Cyr
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland OR 97239, USA
- Present address: Department of Otolaryngology & Sensory Neuroscience Research Center, West Virginia University School of Medicine, Morgantown WV 26506, USA
| |
Collapse
|
49
|
Diefenbach TJ, Latham VM, Yimlamai D, Liu CA, Herman IM, Jay DG. Myosin 1c and myosin IIB serve opposing roles in lamellipodial dynamics of the neuronal growth cone. J Cell Biol 2002; 158:1207-17. [PMID: 12356865 PMCID: PMC2173244 DOI: 10.1083/jcb.200202028] [Citation(s) in RCA: 100] [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] [Indexed: 11/22/2022] Open
Abstract
The myosin family of motor proteins is implicated in mediating actin-based growth cone motility, but the roles of many myosins remain unclear. We previously implicated myosin 1c (M1c; formerly myosin I beta) in the retention of lamellipodia (Wang et al., 1996). Here we address the role of myosin II (MII) in chick dorsal root ganglion neuronal growth cone motility and the contribution of M1c and MII to retrograde F-actin flow using chromophore-assisted laser inactivation (CALI). CALI of MII reduced neurite outgrowth and growth cone area by 25%, suggesting a role for MII in lamellipodial expansion. Micro-CALI of MII caused a rapid reduction in local lamellipodial protrusion in growth cones with no effects on filopodial dynamics. This is opposite to micro-CALI of M1c, which caused an increase in lamellipodial protrusion. We used fiduciary beads (Forscher et al., 1992) to observe retrograde F-actin flow during the acute loss of M1c or MII. Micro-CALI of M1c reduced retrograde bead flow by 76%, whereas micro-CALI of MII or the MIIB isoform did not. Thus, M1c and MIIB serve opposite and nonredundant roles in regulating lamellipodial dynamics, and M1c activity is specifically required for retrograde F-actin flow.
Collapse
Affiliation(s)
- Thomas J Diefenbach
- Department of Physiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|
50
|
Holt JR, Gillespie SKH, Provance DW, Shah K, Shokat KM, Corey DP, Mercer JA, Gillespie PG. A chemical-genetic strategy implicates myosin-1c in adaptation by hair cells. Cell 2002; 108:371-81. [PMID: 11853671 DOI: 10.1016/s0092-8674(02)00629-3] [Citation(s) in RCA: 245] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Myosin-1c (also known as myosin-Ibeta) has been proposed to mediate the slow component of adaptation by hair cells, the sensory cells of the inner ear. To test this hypothesis, we mutated tyrosine-61 of myosin-1c to glycine, conferring susceptibility to inhibition by N(6)-modified ADP analogs. We expressed the mutant myosin-1c in utricular hair cells of transgenic mice, delivered an ADP analog through a whole-cell recording pipette, and found that the analog rapidly blocked adaptation to positive and negative deflections in transgenic cells but not in wild-type cells. The speed and specificity of inhibition suggests that myosin-1c participates in adaptation in hair cells.
Collapse
Affiliation(s)
- Jeffrey R Holt
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|