1
|
Mangione MC, Gould KL. Molecular form and function of the cytokinetic ring. J Cell Sci 2019; 132:132/12/jcs226928. [PMID: 31209062 DOI: 10.1242/jcs.226928] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Animal cells, amoebas and yeast divide using a force-generating, actin- and myosin-based contractile ring or 'cytokinetic ring' (CR). Despite intensive research, questions remain about the spatial organization of CR components, the mechanism by which the CR generates force, and how other cellular processes are coordinated with the CR for successful membrane ingression and ultimate cell separation. This Review highlights new findings about the spatial relationship of the CR to the plasma membrane and the arrangement of molecules within the CR from studies using advanced microscopy techniques, as well as mechanistic information obtained from in vitro approaches. We also consider advances in understanding coordinated cellular processes that impact the architecture and function of the CR.
Collapse
Affiliation(s)
- MariaSanta C Mangione
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Kathleen L Gould
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| |
Collapse
|
2
|
Emergent mechanics of actomyosin drive punctuated contractions and shape network morphology in the cell cortex. PLoS Comput Biol 2018; 14:e1006344. [PMID: 30222728 PMCID: PMC6171965 DOI: 10.1371/journal.pcbi.1006344] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/04/2018] [Accepted: 07/05/2018] [Indexed: 11/24/2022] Open
Abstract
Filamentous actin (F-actin) and non-muscle myosin II motors drive cell motility and cell shape changes that guide large scale tissue movements during embryonic morphogenesis. To gain a better understanding of the role of actomyosin in vivo, we have developed a two-dimensional (2D) computational model to study emergent phenomena of dynamic unbranched actomyosin arrays in the cell cortex. These phenomena include actomyosin punctuated contractions, or "actin asters" that form within quiescent F-actin networks. Punctuated contractions involve both formation of high intensity aster-like structures and disassembly of those same structures. Our 2D model allows us to explore the kinematics of filament polarity sorting, segregation of motors, and morphology of F-actin arrays that emerge as the model structure and biophysical properties are varied. Our model demonstrates the complex, emergent feedback between filament reorganization and motor transport that generate as well as disassemble actin asters. Since intracellular actomyosin dynamics are thought to be controlled by localization of scaffold proteins that bind F-actin or their myosin motors we also apply our 2D model to recapitulate in vitro studies that have revealed complex patterns of actomyosin that assemble from patterning filaments and motor complexes with microcontact printing. Although we use a minimal representation of filament, motor, and cross-linker biophysics, our model establishes a framework for investigating the role of other actin binding proteins, how they might alter actomyosin dynamics, and makes predictions that can be tested experimentally within live cells as well as within in vitro models. Recent genetic and mechanical studies of embryonic development reveal a critical role for intracellular scaffolds in generating the shape of the embryo and constructing internal organs. In this paper we developed computer simulations of these scaffolds, composed of filamentous actin (F-actin), a rod-like protein polymer, and mini-thick filaments, composed of non-muscle myosin II, forming a two headed spring-like complex of motor proteins that can walk on, and remodel F-actin networks. Using simulations of these dynamic interactions, we can carry out virtual experiments where we change the physics and chemistry of F-actin polymers, their associated myosin motors, and cross-linkers and observe the changes in scaffolds that emerge. For example, by modulating the motor stiffness of the myosin motors in our model we can observe the formation or loss of large aster-like structures. Such fine-grained control over the physical properties of motors or filaments within simulations are not currently possible with biological experiments, even where mutant proteins or small molecule inhibitors can be targeted to specific sites on filaments or motors. Our approach reflects a growing adoption of simulation methods to investigate microscopic features that shape actomyosin arrays and the mesoscale effects of molecular scale processes. We expect predictions from these models will drive more refined experimental approaches to expose the many roles of actomyosin in development.
Collapse
|
3
|
Spira F, Cuylen-Haering S, Mehta S, Samwer M, Reversat A, Verma A, Oldenbourg R, Sixt M, Gerlich DW. Cytokinesis in vertebrate cells initiates by contraction of an equatorial actomyosin network composed of randomly oriented filaments. eLife 2017; 6. [PMID: 29106370 PMCID: PMC5673306 DOI: 10.7554/elife.30867] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 10/28/2017] [Indexed: 12/21/2022] Open
Abstract
The actomyosin ring generates force to ingress the cytokinetic cleavage furrow in animal cells, yet its filament organization and the mechanism of contractility is not well understood. We quantified actin filament order in human cells using fluorescence polarization microscopy and found that cleavage furrow ingression initiates by contraction of an equatorial actin network with randomly oriented filaments. The network subsequently gradually reoriented actin filaments along the cell equator. This strictly depended on myosin II activity, suggesting local network reorganization by mechanical forces. Cortical laser microsurgery revealed that during cytokinesis progression, mechanical tension increased substantially along the direction of the cell equator, while the network contracted laterally along the pole-to-pole axis without a detectable increase in tension. Our data suggest that an asymmetric increase in cortical tension promotes filament reorientation along the cytokinetic cleavage furrow, which might have implications for diverse other biological processes involving actomyosin rings.
Collapse
Affiliation(s)
- Felix Spira
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Sara Cuylen-Haering
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Shalin Mehta
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, United States
| | - Matthias Samwer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Anne Reversat
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Amitabh Verma
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, United States
| | - Rudolf Oldenbourg
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, United States
| | - Michael Sixt
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Daniel W Gerlich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| |
Collapse
|
4
|
Dorn JF, Zhang L, Phi TT, Lacroix B, Maddox PS, Liu J, Maddox AS. A theoretical model of cytokinesis implicates feedback between membrane curvature and cytoskeletal organization in asymmetric cytokinetic furrowing. Mol Biol Cell 2016; 27:1286-99. [PMID: 26912796 PMCID: PMC4831882 DOI: 10.1091/mbc.e15-06-0374] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 02/16/2016] [Indexed: 11/11/2022] Open
Abstract
Furrow ingression is asymmetric in cytokinesis in the Caenorhabditis elegans zygote. A combination of quantitative high-resolution live-cell microscopy and theoretical modeling revealed a mechanistic basis for asymmetry: feedback among membrane curvature, cytoskeletal alignment, and contractility. The model also suggests that asymmetry promotes energy efficiency. During cytokinesis, the cell undergoes a dramatic shape change as it divides into two daughter cells. Cell shape changes in cytokinesis are driven by a cortical ring rich in actin filaments and nonmuscle myosin II. The ring closes via actomyosin contraction coupled with actin depolymerization. Of interest, ring closure and hence the furrow ingression are nonconcentric (asymmetric) within the division plane across Metazoa. This nonconcentricity can occur and persist even without preexisting asymmetric cues, such as spindle placement or cellular adhesions. Cell-autonomous asymmetry is not explained by current models. We combined quantitative high-resolution live-cell microscopy with theoretical modeling to explore the mechanistic basis for asymmetric cytokinesis in the Caenorhabditis elegans zygote, with the goal of uncovering basic principles of ring closure. Our theoretical model suggests that feedback among membrane curvature, cytoskeletal alignment, and contractility is responsible for asymmetric cytokinetic furrowing. It also accurately predicts experimental perturbations of conserved ring proteins. The model further suggests that curvature-mediated filament alignment speeds up furrow closure while promoting energy efficiency. Collectively our work underscores the importance of membrane–cytoskeletal anchoring and suggests conserved molecular mechanisms for this activity.
Collapse
Affiliation(s)
- Jonas F Dorn
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Li Zhang
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Tan-Trao Phi
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | | | - Paul S Maddox
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Jian Liu
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20814
| | - Amy Shaub Maddox
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
5
|
Insight into actin organization and function in cytokinesis from analysis of fission yeast mutants. Genetics 2013; 194:435-46. [PMID: 23589458 DOI: 10.1534/genetics.113.149716] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Actin is a key cytoskeletal protein with multiple roles in cellular processes such as polarized growth, cytokinesis, endocytosis, and cell migration. Actin is present in all eukaryotes as highly dynamic filamentous structures, such as linear cables and branched filaments. Detailed investigation of the molecular role of actin in various processes has been hampered due to the multifunctionality of the protein and the lack of alleles defective in specific processes. The actin cytoskeleton of the fission yeast, Schizosaccharomyces pombe, has been extensively characterized and contains structures analogous to those in other cell types. In this study, primarily with the view to uncover actin function in cytokinesis, we generated a large bank of fission yeast actin mutants that affect the organization of distinct actin structures and/or discrete physiological functions of actin. Our screen identified 17 mutants with specific defects in cytokinesis. Some of these cytokinesis mutants helped in dissecting the function of specific actin structures during ring assembly. Further genetic analysis of some of these actin mutants revealed multiple genetic interactions with mutants previously known to affect the actomyosin ring assembly. We also characterize a mutant allele of actin that is suppressed upon overexpression of Cdc8p-tropomyosin, underscoring the utility of this mutant bank. Another 22 mutant alleles, defective in polarized growth and/or other functions of actin obtained from this screen, are also described in this article. This mutant bank should be a valuable resource to study the physiological and biochemical functions of actin.
Collapse
|
6
|
Biphasic assembly of the contractile apparatus during the first two cell division cycles in zebrafish embryos. ZYGOTE 2013; 22:218-28. [DOI: 10.1017/s0967199413000051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SummaryThe large and optically clear embryos of the zebrafish provide an excellent model system in which to study the dynamic assembly of the essential contractile band components, actin and myosin, via double fluorescent labelling in combination with confocal microscopy. We report the rapid appearance (i.e. within <2 min) of a restricted arc of F-actin patches along the prospective furrow plane in a central, apical region of the blastodisc cortex. These patches then fused with each other end-to-end forming multiple actin cables, which were subsequently bundled together forming an F-actin band. During this initial assembly phase, the F-actin-based structure did not elongate laterally, but was still restricted to an arc extending ~15° either side of the blastodisc apex. This initial assembly phase was then followed by an extension phase, where additional F-actin patches were added to each end of the original arc, thus extending it out to the edges of the blastodisc. The dynamics of phosphorylated myosin light chain 2 (MLC2) recruitment to this F-actin scaffold also reflect the two-phase nature of the contractile apparatus assembly. MLC2 was not associated with the initial F-actin arc, but MLC2 clusters were recruited and assembled into the extending ends of the band. We propose that the MLC2-free central region of the contractile apparatus acts to position and then extend the cleavage furrow in the correct plane, while the actomyosin ends alone generate the force required for furrow ingression. This biphasic assembly strategy may be required to successfully divide the early cells of large embryos.
Collapse
|
7
|
Huang J, Huang Y, Yu H, Subramanian D, Padmanabhan A, Thadani R, Tao Y, Tang X, Wedlich-Soldner R, Balasubramanian MK. Nonmedially assembled F-actin cables incorporate into the actomyosin ring in fission yeast. ACTA ACUST UNITED AC 2013. [PMID: 23185032 PMCID: PMC3514790 DOI: 10.1083/jcb.201209044] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In many eukaryotes, cytokinesis requires the assembly and constriction of an actomyosin-based contractile ring. Despite the central role of this ring in cytokinesis, the mechanism of F-actin assembly and accumulation in the ring is not fully understood. In this paper, we investigate the mechanism of F-actin assembly during cytokinesis in Schizosaccharomyces pombe using lifeact as a probe to monitor actin dynamics. Previous work has shown that F-actin in the actomyosin ring is assembled de novo at the division site. Surprisingly, we find that a significant fraction of F-actin in the ring was recruited from formin-Cdc12p nucleated long actin cables that were generated at multiple nonmedial locations and incorporated into the ring by a combination of myosin II and myosin V activities. Our results, together with findings in animal cells, suggest that de novo F-actin assembly at the division site and directed transport of F-actin cables assembled elsewhere can contribute to ring assembly.
Collapse
Affiliation(s)
- Junqi Huang
- Cell Division Laboratory, Temasek Life Sciences Laboratory, Singapore 117604
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Roubinet C, Tran PT, Piel M. Common mechanisms regulating cell cortex properties during cell division and cell migration. Cytoskeleton (Hoboken) 2012; 69:957-72. [PMID: 23125194 DOI: 10.1002/cm.21086] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Revised: 09/28/2012] [Accepted: 10/02/2012] [Indexed: 12/14/2022]
Abstract
Single cell morphogenesis results from a balance of forces involving internal pressure (also called turgor pressure in plants and fungi) and the plastic and dynamic outer shell of the cell. Dominated by the cell wall in plants and fungi, mechanical properties of the outer shell of animal cells arise from the cell cortex, which is mostly composed of the plasma membrane (and membrane proteins) and the underlying meshwork of actin filaments and myosin motors (and associated proteins). In this review, following Bray and White [1988; Science 239:883-889], we draw a parallel between the regulation of the cell cortex during cell division and cell migration in animal cells. Starting from the similarities in shape changes and underlying mechanical properties, we further propose that the analogy between cell division and cell migration might run deeper, down to the basic molecular mechanisms driving cell cortex remodeling. We focus our attention on how an heterogeneous and dynamic cortex can be generated to allow cell shape changes while preserving cell integrity.
Collapse
Affiliation(s)
- Chantal Roubinet
- Université de Toulouse, UPS, Centre de Biologie du Développement, Bâtiment 4R3, 118 route de Narbonne, F-31062 Toulouse, France.
| | | | | |
Collapse
|
9
|
Jayadev R, Kuk CY, Low SH, Murata-Hori M. Calcium sensitivity of α-actinin is required for equatorial actin assembly during cytokinesis. Cell Cycle 2012; 11:1929-37. [PMID: 22544326 DOI: 10.4161/cc.20277] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The actin cross-linking protein, α-actinin, plays a crucial role in mediating furrow ingression during cytokinesis. However, the mechanism by which its dynamics are regulated during this process is poorly understood. Here we have investigated the role of calcium sensitivity of α-actinin in the regulation of its dynamics by generating a functional calcium-insensitive mutant (EFM). GFP-tagged EFM (EFM-GFP) localized to the equatorial regions during cell division. However, the maximal equatorial accumulation of EFM-GFP was significantly smaller in comparison to α-actinin-GFP when it was expressed in normal cells and cells depleted of endogenous α-actinin. No apparent defects in cytokinesis were observed in these cells. However, F-actin levels at the equator were significantly reduced in cells expressing EFM-GFP as compared with α-actinin-GFP at furrow initiation but were recovered during furrow ingression. These results suggest that calcium sensitivity of α-actinin is required for its equatorial accumulation that is crucial for the initial equatorial actin assembly but is dispensable for cytokinesis. Equatorial RhoA localization was not affected by EFM-GFP overexpression, suggesting that equatorial actin assembly is predominantly driven by the RhoA-dependent mechanism. Our observations shed new light on the role and regulation of the accumulation of pre-existing actin filaments in equatorial actin assembly during cytokinesis.
Collapse
Affiliation(s)
- Ranjay Jayadev
- Cell Biology Program, Temasek Life Sciences Laboratory; Singapore
| | | | | | | |
Collapse
|
10
|
Phosphorylation of myosin II regulatory light chain controls its accumulation, not that of actin, at the contractile ring in HeLa cells. Exp Cell Res 2012; 318:915-24. [PMID: 22374324 DOI: 10.1016/j.yexcr.2012.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 02/02/2012] [Accepted: 02/12/2012] [Indexed: 11/24/2022]
Abstract
During cytokinesis in eukaryotic cells, an actomyosin-based contractile ring (CR) is assembled along the equator of the cell. Myosin II ATPase activity is stimulated by the phosphorylation of the myosin II regulatory light chain (MRLC) in vitro, and phosphorylated MRLC localizes at the CR in various types of cells. Previous studies have determined that phosphorylated MRLC plays an important role in CR furrowing. However, the role of phosphorylated MRLC in CR assembly remains unknown. Here, we have used confocal microscopy to observe dividing HeLa cells expressing fluorescent protein-tagged MRLC mutants and actin during CR assembly near the cortex. Di-phosphomimic MRLC accumulated at the cell equator earlier than non-phosphorylatable MRLC and actin. Interestingly, perturbation of myosin II activity by non-phosphorylatable MRLC expression or treatment with blebbistatin, a myosin II inhibitor, did not alter the time of actin accumulation at the cell equator. Furthermore, inhibition of actin polymerization by treatment with latrunculin A had no effect on MRLC accumulation at the cell equator. Taken together, these data suggest that phosphorylated MRLC temporally controls its own accumulation, but not that of actin, in cultured mammalian cells.
Collapse
|
11
|
Abstract
Over the past half century, the Xenopus laevis embryo has become a popular model system for studying vertebrate early development at molecular, cellular, and multicellular levels. The year-round availability of easily fertilized eggs, the embryo's large size and rapid development, and the hardiness of both adults and offspring against a wide range of laboratory conditions provide unmatched advantages for a variety of approaches, particularly "cutting and pasting" experiments, to explore embryogenesis. There is, however, a common perception that the Xenopus embryo is intractable for microscope work, due to its store of large, refractile yolk platelets and abundant cortical pigmentation. This chapter presents easily adapted protocols to surmount, and in some cases take advantage of, these optical properties to facilitate live-cell microscopic analysis of commonly used experimental manipulations of early Xenopus embryos.
Collapse
Affiliation(s)
- Michael V Danilchik
- Department of Integrative Biosciences, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
12
|
Chankitisakul V, Tharasanit T, Tasripoo K, Techakumphu M. Chronological Reorganization of Microtubules, Actin Microfilaments, and Chromatin during the First Cell Cycle in Swamp Buffalo (Bubalus bubalis) Embryos. Vet Med Int 2010; 2010:382989. [PMID: 21234419 PMCID: PMC3014712 DOI: 10.4061/2010/382989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 11/03/2010] [Indexed: 11/20/2022] Open
Abstract
This paper aimed to study the dynamics of early embryonic development, in terms of redistribution of cytoskeleton (microtubules, actin microfilaments) and chromatin configurations during the first cell cycle in swamp buffalo embryos. Oocytes were matured and fertilized in vitro, and they were fixed at various time points after IVF. At 6 h after IVF, 44.4% matured oocytes were penetrated by spermatozoa. Partial ZP digestion, however, did not improve fertilization rate compared to control (P > .05). At 12 h after IVF, the fertilized oocytes progressed to the second meiotic division and formed the female pronucleus simultaneously with the paternal chromatin continued to decondense. A sperm aster was observed radiating from the base of the decondensing sperm head. At 18 h after IVF, most presumptive zygotes had reached the pronuclear stage. The sperm aster was concurrently enlarged to assist the migration and apposition of pronuclei. Cell cleavage was facilitated by microfilaments and firstly observed by 30 h after IVF. In conclusion, the cytoskeleton actively involves with the process of fertilization and cleavage in swamp buffalo oocytes. The centrosomal material is paternally inherited. Fertilization failure is predominantly caused by poor sperm penetration. However, partial digestion of ZP did not improve fertilization rate.
Collapse
Affiliation(s)
- Vibuntita Chankitisakul
- Department of Obstetrics, Gynaecology and Reproduction, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | | | | | | |
Collapse
|
13
|
Impact of marine drugs on cytoskeleton-mediated reproductive events. Mar Drugs 2010; 8:881-915. [PMID: 20479959 PMCID: PMC2866467 DOI: 10.3390/md8040881] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 03/02/2010] [Accepted: 03/23/2010] [Indexed: 12/30/2022] Open
Abstract
Marine organisms represent an important source of novel bioactive compounds, often showing unique modes of action. Such drugs may be useful tools to study complex processes such as reproduction; which is characterized by many crucial steps that start at gamete maturation and activation and virtually end at the first developmental stages. During these processes cytoskeletal elements such as microfilaments and microtubules play a key-role. In this review we describe: (i) the involvement of such structures in both cellular and in vitro processes; (ii) the toxins that target the cytoskeletal elements and dynamics; (iii) the main steps of reproduction and the marine drugs that interfere with these cytoskeleton-mediated processes. We show that marine drugs, acting on microfilaments and microtubules, exert a wide range of impacts on reproductive events including sperm maturation and motility, oocyte maturation, fertilization, and early embryo development.
Collapse
|
14
|
Coffman VC, Nile AH, Lee IJ, Liu H, Wu JQ. Roles of formin nodes and myosin motor activity in Mid1p-dependent contractile-ring assembly during fission yeast cytokinesis. Mol Biol Cell 2010; 20:5195-210. [PMID: 19864459 DOI: 10.1091/mbc.e09-05-0428] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Two prevailing models have emerged to explain the mechanism of contractile-ring assembly during cytokinesis in the fission yeast Schizosaccharomyces pombe: the spot/leading cable model and the search, capture, pull, and release (SCPR) model. We tested some of the basic assumptions of the two models. Monte Carlo simulations of the SCPR model require that the formin Cdc12p is present in >30 nodes from which actin filaments are nucleated and captured by myosin-II in neighboring nodes. The force produced by myosin motors pulls the nodes together to form a compact contractile ring. Live microscopy of cells expressing Cdc12p fluorescent fusion proteins shows for the first time that Cdc12p localizes to a broad band of 30-50 dynamic nodes, where actin filaments are nucleated in random directions. The proposed progenitor spot, essential for the spot/leading cable model, usually disappears without nucleating actin filaments. alpha-Actinin ain1 deletion cells form a normal contractile ring through nodes in the absence of the spot. Myosin motor activity is required to condense the nodes into a contractile ring, based on slower or absent node condensation in myo2-E1 and UCS rng3-65 mutants. Taken together, these data provide strong support for the SCPR model of contractile-ring formation in cytokinesis.
Collapse
Affiliation(s)
- Valerie C Coffman
- Department of Molecular Genetics, Graduate Program of Molecular, Cellular, and Developmental Biology, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
15
|
Abstract
Cleavage furrows (CFs) have been isolated from dividing sea urchin eggs and the protein constituents have been analyzed by two-dimensional gel electrophoresis (Fujimoto & Mabuchi, J. Biochem. 122, 518-524, 1997). Two proteins of 51 and 32 kDa, respectively, have been found to be enriched in the CF preparation. Here, we show that these proteins are identical to the protein elongation factor 1alpha (EF-1alpha) and 1beta (EF-1beta), respectively. Furthermore, the CF 51-kDa protein is identical to the 51-kDa protein which had been isolated as a component of the microtubule organizing granules of mitotic sea urchin eggs. The 51-kDa protein bundles F-actin in vitro. This activity is suppressed by Ca(2+)/calmodulin or GTPgammaS. The 32-kDa protein binds EF-1alpha both in vitro and in cell extract, and is shown to suppress the F-actin-bundling activity of the 51-kDa protein. Microinjection of a monoclonal antibody against the 51-kDa protein or that of His-tagged 32-kDa protein into dividing sea urchin eggs at the onset of cleavage leads to failure of cytokinesis. These results strongly suggest that EF-1alpha is involved in maintenance of the structure of the contractile ring and EF-1beta regulates the F-actin-bundling activity of EF-1alpha.
Collapse
Affiliation(s)
- Hirotaka Fujimoto
- Department of Cell Biology, National Institute for Basic Biology, Okazaki, Aichi 444-8585, Japan
| | | |
Collapse
|
16
|
Asano S, Hamao K, Hosoya H. Direct evidence for roles of phosphorylated regulatory light chain of myosin II in furrow ingression during cytokinesis in HeLa cells. Genes Cells 2009; 14:555-68. [PMID: 19371382 DOI: 10.1111/j.1365-2443.2009.01288.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Phosphorylation of myosin II is thought to play an important role in cytokinesis. Although it is well known that phosphorylated regulatory light chain of myosin II (P-MRLC) localizes along the contractile ring, it is not clear how P-MRLC controls myosin II and F-actin in furrow ingression during cytokinesis. To elucidate roles of P-MRLC in furrow ingression, HeLa cells transfected with EGFP-tagged wild-type or each MRLC mutant were observed using a live-imaging microscope. Time-lapse observation revealed that a delay of furrow ingression was observed in the nonphosphorylatable form of MRLC (AA-MRLC)-expressing cell but not in the wild-type or phospho-mimic MRLC-expressing cell. Among each form of MRLC-expressing cell, the total amount of P-MRLC including phospho-mimic MRLCs was smallest in the cell expressing AA-MRLC. However, the amount of F-actin and myosin II at the contractile ring in the AA-MRLC-expressing cell was the same as that in the normal cell. Interestingly, delay of furrow ingression by a Rho-kinase inhibitor, Y27632, was rescued by phospho-mimic MRLCs. These results suggest that the P-MRLC is essential for the progress of furrow ingression but not the retainment of F-actin and myosin II in the contractile ring of dividing HeLa cells.
Collapse
Affiliation(s)
- Satoshi Asano
- Department of Biological Science, Graduate School of Science, Hiroshima University, Higashi-Hiroshima 739-8526, Japan
| | | | | |
Collapse
|
17
|
Alsop GB, Chen W, Foss M, Tseng KF, Zhang D. Redistribution of actin during assembly and reassembly of the contractile ring in grasshopper spermatocytes. PLoS One 2009; 4:e4892. [PMID: 19287500 PMCID: PMC2654139 DOI: 10.1371/journal.pone.0004892] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Accepted: 02/17/2009] [Indexed: 11/19/2022] Open
Abstract
Cytokinesis in animal cells requires the assembly of an actomyosin contractile ring to cleave the cell. The ring is highly dynamic; it assembles and disassembles during each cell cleavage, resulting in the recurrent redistribution of actin. To investigate this process in grasshopper spermatocytes, we mechanically manipulated the spindle to induce actin redistribution into ectopic contractile rings, around reassembled lateral spindles. To enhance visualization of actin, we folded the spindle at its equator to convert the remnants of the partially assembled ring into a concentrated source of actin. Filaments from the disintegrating ring aligned along reorganizing spindle microtubules, suggesting that their incorporation into the new ring was mediated by microtubules. We tracked incorporation by speckling actin filaments with Qdots and/or labeling them with Alexa 488-phalloidin. The pattern of movement implied that actin was transported along spindle microtubules, before entering the ring. By double-labeling dividing cells, we imaged actin filaments moving along microtubules near the contractile ring. Together, our findings indicate that in one mechanism of actin redistribution, actin filaments are transported along spindle microtubule tracks in a plus-end–directed fashion. After reaching the spindle midzone, the filaments could be transported laterally to the ring. Notably, actin filaments undergo a dramatic trajectory change as they enter the ring, implying the existence of a pulling force. Two other mechanisms of actin redistribution, cortical flow and de novo assembly, are also present in grasshopper, suggesting that actin converges at the nascent contractile ring from diffuse sources within the cytoplasm and cortex, mediated by spindle microtubules.
Collapse
Affiliation(s)
- G. Bradley Alsop
- Department of Zoology, Oregon State University, Corvallis, Oregon, United States of America
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, United States of America
| | - Wei Chen
- Department of Zoology, Oregon State University, Corvallis, Oregon, United States of America
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, United States of America
| | - Margit Foss
- Department of Zoology, Oregon State University, Corvallis, Oregon, United States of America
| | - Kuo-Fu Tseng
- Department of Zoology, Oregon State University, Corvallis, Oregon, United States of America
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, United States of America
| | - Dahong Zhang
- Department of Zoology, Oregon State University, Corvallis, Oregon, United States of America
- Center for Genome Research and Biocomputing (CGRB), Oregon State University, Corvallis, Oregon, United States of America
- * E-mail:
| |
Collapse
|
18
|
Yumura S, Ueda M, Sako Y, Kitanishi-Yumura T, Yanagida T. Multiple Mechanisms for Accumulation of Myosin II Filaments at the Equator During Cytokinesis. Traffic 2008; 9:2089-99. [DOI: 10.1111/j.1600-0854.2008.00837.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
19
|
Thoms JAI, Loch HM, Bamburg JR, Gunning PW, Weinberger RP. A tropomyosin 1 induced defect in cytokinesis can be rescued by elevated expression of cofilin. ACTA ACUST UNITED AC 2008; 65:979-90. [DOI: 10.1002/cm.20320] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
20
|
Danilchik MV, Brown EE. Membrane dynamics of cleavage furrow closure in Xenopus laevis. Dev Dyn 2008; 237:565-79. [PMID: 18224710 DOI: 10.1002/dvdy.21442] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Epithelial membrane polarity develops early in Xenopus development, with membrane inserted along the earliest cleavage furrows by means of localized exocytosis. The added surface constitutes a new basolateral domain important for early morphogenesis. This basolateral surface becomes isolated from the outside by furrow closure, a zippering of adjacent apical-basolateral margins. Time-lapse microscopy of membrane-labeled embryos revealed two distinct kinds of protrusive activity in furrow closure. Early in furrowing, protrusive activity was associated with purse-string contractility along the apical-basolateral margins. Later in furrow progression, a basolateral protrusive zone developed entirely within the new membrane domain, with long motile filopodia extending in contractile bands from the exposed surfaces. Filopodia interacting with opposing cell surfaces across the cleavage furrow appeared to mediate blastomere-blastomere adhesion, contact spreading and lamellipodial protrusion. Interference with these dynamic activities prevented furrow closure, indicating a basic role for both marginal and basolateral protrusive activities in early embryogenesis.
Collapse
Affiliation(s)
- Michael V Danilchik
- Department of Integrative Biosciences, Oregon Health and Science University, Portland, Oregon 97239-3097, USA.
| | | |
Collapse
|
21
|
Li WM, Webb SE, Chan CM, Miller AL. Multiple roles of the furrow deepening Ca2+ transient during cytokinesis in zebrafish embryos. Dev Biol 2008; 316:228-48. [PMID: 18313658 DOI: 10.1016/j.ydbio.2008.01.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2007] [Revised: 01/11/2008] [Accepted: 01/11/2008] [Indexed: 01/16/2023]
Abstract
The generation of a required series of localized Ca(2+) transients during cytokinesis in zebrafish embryos suggests that Ca(2+) plays a necessary role in regulating this process. Here, we report that cortical actin remodeling, characterized by the reorganization of the contractile band and the formation during furrow deepening of pericleavage F-actin enrichments (PAEs), requires a localized increase in intracellular Ca(2+), which is released from IP(3)-sensitive stores. We demonstrate that VAMP-2 vesicle fusion at the deepening furrow also requires Ca(2+) released via IP(3) receptors, as well as the presence of PAEs and the action of calpains. Finally, by expressing a dominant-negative form of the kinesin-like protein, kif23, we demonstrate that its recruitment to the furrow region is required for VAMP-2 vesicle transport; and via FRAP analysis, that kif23 localization is also Ca(2+)-dependent. Collectively, our data demonstrate that a localized increase in intracellular Ca(2+) is involved in regulating several key events during furrow deepening and subsequent apposition.
Collapse
Affiliation(s)
- Wai Ming Li
- Department of Biology, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | | | | | | |
Collapse
|
22
|
Chávez-Munguía B, Talamás-Rohana P, Ríos A, González-Lázaro M, Martínez-Palomo A. Entamoeba histolytica: Fibrilar aggregates in dividing trophozoites. Exp Parasitol 2008; 118:280-4. [PMID: 17870071 DOI: 10.1016/j.exppara.2007.08.001] [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: 04/19/2007] [Revised: 08/02/2007] [Accepted: 08/03/2007] [Indexed: 11/22/2022]
Abstract
Entamoeba histolytica trophozoite cytokinesis is dependent upon cytoskeletal elements such as filamentous actin and myosin. Here we present confocal and transmission electron microscopy studies of this process. A sequence in the formation of the contractile ring was shown with rhodamine-phalloidine staining. Ultrastructural analysis revealed the presence of fibrilar aggregates in the cytoplasm of dividing trophozoites. Among them two filaments of different diameter were identified. These aggregates presented repeating assemblies of thin and thick filaments that in cross section revealed a muscle-like appearance. Our results suggest that these aggregates constitute the contractile ring responsible for the separation of daughter cells.
Collapse
Affiliation(s)
- Bibiana Chávez-Munguía
- Department of Experimental Pathology, Center for Research and Advanced Studies, Av. IPN 2508, Zacatenco 07360, Mexico City, Mexico.
| | | | | | | | | |
Collapse
|
23
|
Kuhn TB, Bamburg JR. Tropomyosin and ADF/cofilin as collaborators and competitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 644:232-49. [PMID: 19209826 DOI: 10.1007/978-0-387-85766-4_18] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dynamics of actin filaments is pivotal to many fundamental cellular processes such as Dcytokinesis, motility, morphology, vesicle and organelle transport, gene transcription and senescence. In vivo kinetics of actin filament dynamics is far from the equilibrium in vitro and these profound differences are attributed to large number of regulatory proteins. In particular, proteins of the ADF/cofilin family greatly increase actin filament dynamics by severing filaments and enhancing depolymerization of ADP-actin monomers from their pointed ends. Cofilin binds cooperatively to a minor conformer of F-actin in which the subunits are slightly under rotated along the filament helical axis. At high stoichiometry of cofilin to actin subunits, cofilin actually stabilizes actin filaments. Many isoforms oftropomyosin appear to compete with ADF/cofilin proteins for binding to actin filaments. Tropomyosin isoforms studied to date prefer binding to the "untwisted" conformer of F-actin and through their protection and stabilization of F-actin, recruit myosin II and assemble different actin superstructures from the cofilin-actin filaments. However, some tropomyosin isoforms may synergize with ADF/cofilin to enhance filament dynamics, suggesting that the different isoforms of tropomyosins, many of which show developmental or tissue specific expression profiles, play major roles in the assembly and turnover of actin superstructures. Different actin superstructures can overlap both spatially and temporally within a cell, but can be differentiated from each other based upon their kinetic and kinematic properties. Furthermore, local regulation of ADF/cofilin activity through signal transduction pathways could be one mechanism to alter the dynamic balance in F-actin-binding of certain tropomyosin isoforms in subcellular domains.
Collapse
Affiliation(s)
- Thomas B Kuhn
- Department of Chemistry, University of Alaska Fairbanks, Fairbanks, Alaska, USA
| | | |
Collapse
|
24
|
Kamasaki T, Osumi M, Mabuchi I. Three-dimensional arrangement of F-actin in the contractile ring of fission yeast. ACTA ACUST UNITED AC 2007; 178:765-71. [PMID: 17724118 PMCID: PMC2064542 DOI: 10.1083/jcb.200612018] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The contractile ring, which is required for cytokinesis in animal and yeast cells, consists mainly of actin filaments. Here, we investigate the directionality of the filaments in fission yeast using myosin S1 decoration and electron microscopy. The contractile ring is composed of around 1,000 to 2,000 filaments each around 0.6 mum in length. During the early stages of cytokinesis, the ring consists of two semicircular populations of parallel filaments of opposite directionality. At later stages, before contraction, the ring filaments show mixed directionality. We consider that the ring is initially assembled from a single site in the division plane and that filaments subsequently rearrange before contraction initiates.
Collapse
Affiliation(s)
- Tomoko Kamasaki
- Division of Biology, School of Arts and Sciences, University of Tokyo, Tokyo 153-8902, Japan
| | | | | |
Collapse
|
25
|
Zhou M, Wang YL. Distinct pathways for the early recruitment of myosin II and actin to the cytokinetic furrow. Mol Biol Cell 2007; 19:318-26. [PMID: 17959823 DOI: 10.1091/mbc.e07-08-0783] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Equatorial organization of myosin II and actin has been recognized as a universal event in cytokinesis of animal cells. Current models for the formation of equatorial cortex favor either directional cortical transport toward the equator or localized de novo assembly. However, this process has never been analyzed directly in dividing mammalian cells at a high resolution. Here we applied total internal reflection fluorescence microscope (TIRF-M), coupled with spatial temporal image correlation spectroscopy (STICS) and a new analytical approach termed temporal differential microscopy (TDM), to image the dynamics of myosin II and actin during the assembly of equatorial cortex. Our results indicated distinct and at least partially independent mechanisms for the early equatorial recruitment of myosin and actin filaments. Cortical myosin showed no detectable directional flow during early cytokinesis. In addition to equatorial assembly, we showed that localized inhibition of disassembly contributed to the formation of the equatorial myosin band. In contrast to myosin, actin filaments underwent a striking flux toward the equator. Myosin motor activity was required for the actin flux, but not for actin concentration in the furrow, suggesting that there was a flux-independent, de novo mechanism for actin recruitment along the equator. Our results indicate that cytokinesis involves signals that regulate both assembly and disassembly activities and argue against mechanisms that are coupled to global cortical movements.
Collapse
Affiliation(s)
- Mian Zhou
- University of Massachusetts Medical School, Department of Physiology, Worcester, MA 01605, USA
| | | |
Collapse
|
26
|
Urven LE, Yabe T, Pelegri F. A role for non-muscle myosin II function in furrow maturation in the early zebrafish embryo. J Cell Sci 2007; 119:4342-52. [PMID: 17038547 DOI: 10.1242/jcs.03197] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Cytokinesis in early zebrafish embryos involves coordinated changes in the f-actin- and microtubule-based cytoskeleton, and the recruitment of adhesion junction components to the furrow. We show that exposure to inhibitors of non-muscle myosin II function does not affect furrow ingression during the early cleavage cycles but interferes with the recruitment of pericleavage f-actin and cortical beta-catenin aggregates to the furrow, as well as the remodeling of the furrow microtubule array. This remodeling is in turn required for the distal aggregation of the zebrafish germ plasm. Embryos with reduced myosin activity also exhibit at late stages of cytokinesis a stabilized contractile ring apparatus that appears as a ladder-like pattern of short f-actin cables, supporting a role for myosin function in the disassembly of the contractile ring after furrow formation. Our studies support a role for myosin function in furrow maturation that is independent of furrow ingression and which is essential for the recruitment of furrow components and the remodeling of the cytoskeleton during cytokinesis.
Collapse
Affiliation(s)
- Lance E Urven
- Laboratory of Genetics, University of Wisconsin - Madison, WI 53706, USA
| | | | | |
Collapse
|
27
|
Danilchik MV, Brown EE, Riegert K. Intrinsic chiral properties of the Xenopus egg cortex: an early indicator of left-right asymmetry? Development 2006; 133:4517-26. [PMID: 17050623 DOI: 10.1242/dev.02642] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Vertebrate embryos define an anatomic plane of bilateral symmetry by establishing rudimentary anteroposterior and dorsoventral (DV) axes. A left-right (LR) axis also emerges, presaging eventual morphological asymmetries of the heart and other viscera. In the radially symmetric egg of Xenopus laevis, the earliest steps in DV axis determination are driven by microtubule-dependent localization of maternal components toward the prospective dorsal side. LR axis determination is linked in time to this DV-determining process, but the earliest steps are unclear. Significantly, no cytoskeletal polarization has been identified in early embryos capable of lateral displacement of maternal components. Cleaving Xenopus embryos and parthenogenetically activated eggs treated with 2,3-butanedione monoxime (BDM) undergo a dramatic large-scale torsion, with the cortex of the animal hemisphere shearing in an exclusively counterclockwise direction past the vegetal cortex. Long actin fibers develop in a shear zone paralleling the equator. Drug experiments indicate that the actin is not organized by microtubules, and depends on the reorganization of preexisting f-actin fibers rather than new actin polymerization. The invariant chirality of this drug response suggests a maternally inherited, microfilament-dependent organization within the egg cortex that could play an early role in LR axis determination during the first cell cycle. Consistent with this hypothesis, brief disruption of cortical actin during the first cell cycle randomizes the LR orientation of tadpole heart and gut.
Collapse
Affiliation(s)
- Michael V Danilchik
- Department of Integrative Biosciences, Oregon Health and Science University, Portland, OR 97239-3097, USA.
| | | | | |
Collapse
|
28
|
Miyauchi K, Yamamoto Y, Kosaka T, Hosoya H. Myosin II activity is not essential for recruitment of myosin II to the furrow in dividing HeLa cells. Biochem Biophys Res Commun 2006; 350:543-8. [PMID: 17022938 DOI: 10.1016/j.bbrc.2006.09.065] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Accepted: 09/15/2006] [Indexed: 11/21/2022]
Abstract
To elucidate whether phosphorylation of myosin II regulatory light chain (MRLC) is essential for myosin II recruitment to the furrow during cytokinesis, HeLa cells transfected with three types of GFP-tagged recombinant MRLCs, wild-type MRLC, non-phosphorylated form of MRLC, and phosphorylated form of MRLC, were examined. Living cell-imaging showed that both phosphorylated and non-phosphorylated form of MRLCs were recruited to the equator at the same time after anaphase onset, suggesting that phosphorylation of MRLC is not responsible for recruitment of myosin II to the equator. Moreover, the treatment with an inhibitor of myosin II activity, blebbistatin, induced no effect on recruitment of those three recombinant MRLCs. During cytokinesis, phosphorylated but not non-phosphorylated form of MRLC was retained in the equator. These results suggest that phosphorylation of MRLC is essential for retainment of myosin II in the furrow but not for initial recruitment of myosin II to the furrow in dividing HeLa cells.
Collapse
Affiliation(s)
- Kenji Miyauchi
- Department of Biological Science, Graduate School of Science, Hiroshima University, Higashi-Hiroshima 739-8526, Japan
| | | | | | | |
Collapse
|
29
|
Sun QY, Schatten H. Regulation of dynamic events by microfilaments during oocyte maturation and fertilization. Reproduction 2006; 131:193-205. [PMID: 16452714 DOI: 10.1530/rep.1.00847] [Citation(s) in RCA: 200] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Actin filaments (microfilaments) regulate various dynamic events during oocyte meiotic maturation and fertilization. In most species, microfilaments are not required for germinal vesicle breakdown and meiotic spindle formation, but they mediate peripheral nucleus (chromosome) migration, cortical spindle anchorage, homologous chromosome separation, cortex development/maintenance, polarity establishment, and first polar body emission during oocyte maturation. Peripheral cortical granule migration is controlled by microfilaments, while mitochondria movement is mediated by microtubules. During fertilization, microfilaments are involved in sperm incorporation, spindle rotation (mouse), cortical granule exocytosis, second polar body emission and cleavage ring formation, but are not required for pronuclear apposition (except for the mouse). Many of the events are driven by the dynamic interactions between myosin and actin filaments whose polymerization is regulated by RhoA, Cdc42, Arp2/3 and other signaling molecules. Studies have also shown that oocyte cortex organization and polarity formation mediated by actin filaments are regulated by mitogen-activated protein kinase, myosin light-chain kinase, protein kinase C and its substrate p-MARKS as well as PAR proteins. The completion of several dynamic events, including homologous chromosome separation, spindle anchorage, spindle rotation, vesicle organelle transport and pronuclear apposition (mouse), requires interactions between microfilaments and microtubules, but determination of how the two systems of the cytoskeleton precisely cross-link, and which proteins link microfilaments to microtubules to perform functions in eggs, requires further studies. Finally, the meaning of microfilament-mediated oocyte polarity versus embryo polarity and embryo development in different species (Drosophila, Xenopus and mouse) is discussed.
Collapse
Affiliation(s)
- Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, China
| | | |
Collapse
|
30
|
Abstract
Formins constitute a diverse protein family present in all eukaryotes examined. They are defined by the presence of a formin homology 2 (FH2) domain, which possesses intrinsic and conserved functions regulating cytoskeletal dynamics. Over the past few years, formins have become recognized as potent nucleators of linear actin filaments that control a large variety of cellular and morphogenetic functions. Here, we review the molecular principles of formin-induced cytoskeletal rearrangements and their consequences for a growing number of biological processes.
Collapse
Affiliation(s)
- Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, 30623 Hannover, Germany.
| | | |
Collapse
|
31
|
Abstract
The mechanism underlying cytokinesis, the final step in cell division, remains one of the major unsolved questions in basic cell biology. Thanks to advances in functional genomics and proteomics, we are now able to assemble a "parts list" of proteins involved in cytokinesis. In this review, we discuss how to relate this parts list to biological mechanism. For easier analysis, we split cytokinesis into discrete steps: cleavage plane specification, rearrangement of microtubule structures, contractile ring assembly, ring ingression, and completion. We report on the advances that have been made to understand these steps and how they can be integrated into a global understanding of cytokinesis. We also discuss the extent to which classic questions have been answered and identify major outstanding questions.
Collapse
Affiliation(s)
- Ulrike S Eggert
- Dana-Farber Cancer Institute and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
32
|
Tanaka K, Okubo Y, Abe H. Involvement of slingshot in the Rho-mediated dephosphorylation of ADF/cofilin during Xenopus cleavage. Zoolog Sci 2006; 22:971-84. [PMID: 16219977 DOI: 10.2108/zsj.22.971] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
ADF/cofilin is a key regulator for actin dynamics during cytokinesis. Its activity is suppressed by phosphorylation and reactivated by dephosphorylation. Little is known, however, about regulatory mechanisms of ADF/cofilin function during formation of contractile ring actin filaments. Using Xenopus cycling extracts, we found that ADF/cofilin was dephosphorylated at prophase and telophase. In addition, constitutively active Rho GTPase induced dephosphorylation of ADF/cofilin in the egg extracts. This dephosphorylation was inhibited by Na(3)VO (4) but not by other conventional phosphatase-inhibitors. We cloned a Xenopus homologue of Slingshot phosphatase (XSSH), originally identified in Drosophila and human as an ADF/cofilin phosphatase, and raised antibody specific for the catalytic domain of XSSH. This inhibitory antibody significantly suppressed the Rho-induced dephosphorylation of ADF/cofilin in extracts, suggesting that the dephosphorylation at telophase is dependent on XSSH. XSSH bound to actin filaments with a dissociation constant of 0.4 microM, and the ADF/cofilin phosphatase activity was increased in the presence of F-actin. When latrunculin A, a G-actin-sequestering drug, was added to extracts, both Rho-induced actin polymerization and dephosphorylation of ADF/cofilin were markedly inhibited. Jasplakinolide, an actin-stabilizing drug, alone induced actin polymerization in the extracts and lead to dephosphorylation of ADF/cofilin. These results suggest that Rho-induced dephosphorylation of ADF/cofilin is dependent on the XSSH activation that is caused by increase in the amount of F-actin induced by Rho signaling. XSSH colocalized with both actin filaments and ADF/cofilin in the actin patches formed on the surface of the early cleavage furrow. Injection of inhibitory antibody blocked cleavage of blastomeres. Thus, XSSH may reorganize actin filaments through dephosphorylation and reactivation of ADF/cofilin at early stage of contractile ring formation.
Collapse
|
33
|
Darenfed H, Mandato CA. Wound-induced contractile ring: a model for cytokinesis. Biochem Cell Biol 2006; 83:711-20. [PMID: 16333322 DOI: 10.1139/o05-164] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The actomyosin-based contractile ring is required for several biological processes, such as wound healing and cytokinesis of animal cells. Despite progress in defining the roles of this structure in both wound closure and cell division, we still do not fully understand how an actomyosin ring is spatially and temporally assembled, nor do we understand the molecular mechanism of its contraction. Recent results have demonstrated that microtubule-dependent local assembly of F-actin and myosin-II is present in wound closure and is similar to that in cytokinesis in animal cells. Furthermore, signalling factors such as small Rho GTPases have been shown to be involved in the regulation of actin dynamics during both processes. In this review we address recent findings in an attempt to better understand the dynamics of actomyosin contractile rings during wound healing as compared with the final step of animal cell division.
Collapse
Affiliation(s)
- Hassina Darenfed
- Department of Anatomy and Cell Biology, Mc Gill University, Montreal, QC, Canada
| | | |
Collapse
|
34
|
Nakano K, Mabuchi I. Actin-depolymerizing protein Adf1 is required for formation and maintenance of the contractile ring during cytokinesis in fission yeast. Mol Biol Cell 2006; 17:1933-45. [PMID: 16467379 PMCID: PMC1415287 DOI: 10.1091/mbc.e05-09-0900] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The role of the actin-depolymerizing factor (ADF)/cofilin-family protein Adf1 in cytokinesis of fission yeast cells was studied. Adf1 was required for accumulation of actin at the division site by depolymerizing actin at the cell ends, assembly of the contractile ring through severing actin filaments, and maintenance of the contractile ring once formed. Genetic and cytological analyses suggested that it collaborates with profilin and capping protein in the mitotic reorganization of the actin cytoskeleton. Furthermore, it was unexpectedly found that Adf1 and myosin-II also collaborate in assembling the contractile ring. Tropomyosin was shown to antagonize the function of Adf1 in the contractile ring. We propose that formation and maintenance of the contractile ring are achieved by a balanced collaboration of these proteins.
Collapse
Affiliation(s)
- Kentaro Nakano
- Division of Biology, Department of Life Sciences, Graduate Program in Interdisciplinary Sciences, School of Arts and Sciences, University of Tokyo, Tokyo 153-8902, Japan.
| | | |
Collapse
|
35
|
Ziyyat A, Rubinstein E, Monier-Gavelle F, Barraud V, Kulski O, Prenant M, Boucheix C, Bomsel M, Wolf JP. CD9 controls the formation of clusters that contain tetraspanins and the integrin α6β1, which are involved in human and mouse gamete fusion. J Cell Sci 2006; 119:416-24. [PMID: 16418227 DOI: 10.1242/jcs.02730] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The process of gamete fusion has been largely studied in the mouse and has revealed the crucial role of the tetraspanin CD9. By contrast, human gamete fusion remains largely unknown. We now show that an anti-α6 integrin mAb (GoH3) strongly inhibited human sperm-egg fusion in human zona-free eggs. Furthermore, a mAb directed against CD151, a tetraspanin known to associate with α6β1, partially inhibited sperm-egg fusion. By contrast, the addition of an anti-CD9 mAb to zona free eggs had no effect. The integrin α6β1, CD151 and CD9 tetraspanins were evenly distributed on human zona-intact oocytes. On zona-free eggs, the integrin α6β1 and tetraspanin CD151 patched and co-localized but the tetraspanin CD9 remained unchanged. CD9 mAb prevented α6β1 integrin clustering and gamete fusion when added prior to, but not after, zona removal. Antibody-mediated aggregation of integrin α6β1 yielded patches that were bigger and more heterogeneous in mouse oocytes lacking CD9. Moreover, a strong labelling of α6β1 could be observed at the sperm entry point. Altogether, these data show that CD9 controls the redistribution of some membrane proteins including the α6β1 integrin into clusters that may be necessary for gamete fusion.
Collapse
Affiliation(s)
- Ahmed Ziyyat
- Université Paris 13, Laboratoire de Biologie de la Reproduction, UPRES 3410, UFR SMBH, Bobigny, France
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Dean SO, Rogers SL, Stuurman N, Vale RD, Spudich JA. Distinct pathways control recruitment and maintenance of myosin II at the cleavage furrow during cytokinesis. Proc Natl Acad Sci U S A 2005; 102:13473-8. [PMID: 16174742 PMCID: PMC1200093 DOI: 10.1073/pnas.0506810102] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The correct localization of myosin II to the equatorial cortex is crucial for proper cell division. Here, we examine a collection of genes that cause defects in cytokinesis and reveal with live cell imaging two distinct phases of myosin II localization. Three genes in the rho1 signaling pathway, pebble (a Rho guanidine nucleotide exchange factor), rho1, and rho kinase, are required for the initial recruitment of myosin II to the equatorial cortex. This initial localization mechanism does not require F-actin or the two components of the centralspindlin complex, the mitotic kinesin pavarotti/MKLP1 and racGAP50c/CYK-4. However, F-actin, the centralspindlin complex, formin (diaphanous), and profilin (chickadee) are required to stably maintain myosin II at the furrow. In the absence of these latter genes, myosin II delocalizes from the equatorial cortex and undergoes highly dynamic appearances and disappearances around the entire cell cortex, sometimes associated with abnormal contractions or blebbing. Our findings support a model in which a rho kinase-dependent event, possibly myosin II regulatory light chain phosphorylation, is required for the initial recruitment to the furrow, whereas the assembly of parallel, unbranched actin filaments, generated by formin-mediated actin nucleation, is required for maintaining myosin II exclusively at the equatorial cortex.
Collapse
Affiliation(s)
- Sara O Dean
- Department of Biochemistry, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
37
|
Murthy K, Wadsworth P. Myosin-II-Dependent Localization and Dynamics of F-Actin during Cytokinesis. Curr Biol 2005; 15:724-31. [PMID: 15854904 DOI: 10.1016/j.cub.2005.02.055] [Citation(s) in RCA: 221] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2005] [Revised: 02/11/2005] [Accepted: 02/14/2005] [Indexed: 11/24/2022]
Abstract
BACKGROUND The assembly of an F-actin- and myosin-II-containing contractile ring (CR) is required for cytokinesis in eukaryotic cells. Interactions between myosin II and actin in the ring are believed to generate the force that constricts the cell into two daughters. The mechanism(s) that contribute to the spatially and temporally regulated assembly and disassembly of the CR at the cell equator are poorly understood. RESULTS We generated an LLCPK1 epithelial cell line that stably expresses GFP-actin. Live confocal imaging showed accumulation of GFP-actin in the equatorial cortex from late anaphase through cytokinesis. Fluorescence recovery after photobleaching (FRAP) experiments showed that actin in the CR is highly dynamic (t(1/2) = 26 s). In some cells, movement of GFP-actin toward the equatorial region was observed and contributed to FRAP. Blocking actin dynamic turnover with jasplakinolide demonstrates that dynamic actin is required for CR formation and cytokinesis. To test the role of myosin II in actin turnover and transport during CR formation, we inhibited myosin light-chain kinase with ML7 and myosin II ATPase activity with blebbistatin. Inhibition of myosin light-chain phosphorylation resulted in clearance of GFP-actin from the equatorial region, a reduction in myosin II in the furrow, and inhibition of cytokinesis. Treatment with blebbistatin did not block CR formation but reduced FRAP of GFP-actin and prevented completion of cytokinesis. CONCLUSIONS These results demonstrate that the majority of actin in the CR is highly dynamic and establish novel roles for myosin II in the retention and dynamic turnover of actin in the CR.
Collapse
Affiliation(s)
- Kausalya Murthy
- Department of Biology and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, 01003, USA
| | | |
Collapse
|
38
|
Motegi F, Mishra M, Balasubramanian MK, Mabuchi I. Myosin-II reorganization during mitosis is controlled temporally by its dephosphorylation and spatially by Mid1 in fission yeast. ACTA ACUST UNITED AC 2004; 165:685-95. [PMID: 15184401 PMCID: PMC2172373 DOI: 10.1083/jcb.200402097] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Cytokinesis in many eukaryotes requires an actomyosin contractile ring. Here, we show that in fission yeast the myosin-II heavy chain Myo2 initially accumulates at the division site via its COOH-terminal 134 amino acids independently of F-actin. The COOH-terminal region can access to the division site at early G2, whereas intact Myo2 does so at early mitosis. Ser1444 in the Myo2 COOH-terminal region is a phosphorylation site that is dephosphorylated during early mitosis. Myo2 S1444A prematurely accumulates at the future division site and promotes formation of an F-actin ring even during interphase. The accumulation of Myo2 requires the anillin homologue Mid1 that functions in proper ring placement. Myo2 interacts with Mid1 in cell lysates, and this interaction is inhibited by an S1444D mutation in Myo2. Our results suggest that dephosphorylation of Myo2 liberates the COOH-terminal region from an intramolecular inhibition. Subsequently, dephosphorylated Myo2 is anchored by Mid1 at the medial cortex and promotes the ring assembly in cooperation with F-actin.
Collapse
Affiliation(s)
- Fumio Motegi
- Division of Biology, Department of Life Sciences, Graduate School of Arts and Science, University of Tokyo, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | | | | | | |
Collapse
|
39
|
Kishimoto Y, Koshida S, Furutani-Seiki M, Kondoh H. Zebrafish maternal-effect mutations causing cytokinesis defect without affecting mitosis or equatorial vasa deposition. Mech Dev 2004; 121:79-89. [PMID: 14706702 DOI: 10.1016/j.mod.2003.10.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Maternal-effect genes play essential roles in early embryogenesis particularly before activation of the zygotic genes. A genetic screen for mutations affecting such maternal-effect genes was carried out employing an F3 screen strategy, identifying six recessive mutations out of 60 mutagenized genomes. Three of the mutations (acytokinesis mutations: ackkt5, ackkt62 and ackkt119) caused absence of cell cleavage in the embryos derived from homozygous females regardless of the paternal genotype, without affecting nuclear divisions. These embryos are defective in generating contractile rings, ackkt62 mutation abolishing reactions to organize cortical F-actin, while other mutations causing abortive contractile ring-like structures at ectopic sites. Defect of contractile ring formation in the affected embryos leads to the absence of microtubule arrays at the prospective cleavage plane. Thus, these mutations reveal the sequence of events associated with cytokinesis, in particular, the cortical actin dynamics. It is remarkable that in all acytokinetic embryos, daughter nuclei after mitosis are arranged in spatially normal positions, and maternal vasa mRNAs accumulate in the prospective planes of the first and second cell cleavages in the total absence of cytokinesis. This indicates that the basic cell architectures of early embryos are largely established by the autonomous activities of the mitotic apparatus, without much dependence on the cell cleavage machinery.
Collapse
Affiliation(s)
- Yasuyuki Kishimoto
- Kondoh Differentiation Signaling Project (ERATO), Japan Science and Technology Corporation, 14 Yoshida-Kawaramachi, Sakyouku, Kyoto 606-8305, Japan.
| | | | | | | |
Collapse
|
40
|
Abstract
Despite a century of research into the nature of animal cell division, a molecular explanation for the positioning of the actomyosin contractile ring has remained elusive. The discovery of a novel interaction between regulators of Rho family small GTPases has revealed a link between the mitotic microtubules and the contractile ring during the later stages of mitosis. The properties of the interacting Rho regulators suggest a molecular model for the positioning and initiation of contractile ring furrowing in animal cells. In this 'double ring' model, centralspindlin complexes, localized by the action of their kinesin-like protein component, position and activate a cortical equatorial ring of Rho GTPase exchange factors. The resulting ring of activated Rho would then trigger a cascade of events leading to formation and constriction of the contractile ring.
Collapse
Affiliation(s)
- Robert Saint
- Centre for the Molecular Genetics of Development, Research School of Biological Sciences, Australian National University, Canberra ACT 0200, Australia.
| | | |
Collapse
|
41
|
Nishimura Y, Mabuchi I. An IQGAP-like protein is involved in actin assembly together with Cdc42 in the sea urchin egg. ACTA ACUST UNITED AC 2004; 56:207-18. [PMID: 14584023 DOI: 10.1002/cm.10146] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We isolated a gene homologous to human cdc42 (ucdc42) from a sea urchin cDNA library. The GTPgammaS-bound UCdc42 induced actin assembly in sea urchin egg extract. Proteins that are involved in this actin assembly system were searched using UCdc42-bound agarose beads. A 180-kDa protein (p180), which showed a homology to human IQGAPs, bound to the GTPgammaS-UCdc42 beads. Immunodepletion of p180 from the sea urchin egg extract abolished this actin assembly on the UCdc42 beads. Immunofluorescent localization of p180 was similar to that of the actin cytoskeleton in the egg cortex and it was concentrated in the cleavage furrow during cytokinesis. A possible role of p180 in actin assembly is discussed.
Collapse
Affiliation(s)
- Yukako Nishimura
- Division of Biology, Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Komaba, Meguro-ku, Tokyo
| | | |
Collapse
|
42
|
Abstract
Neurons from the Xenopus spinal cord are highly versatile and easily manipulated, making them an ideal model system to answer questions regarding the cellular and molecular basis of early neural development and function. Xenopus has been a productive model system in studies ranging from axon growth and guidance to synaptic plasticity. Exogenous molecules, such as proteins, fluorescent tracers, and nucleic acids, can be injected into early blastomeres to load tracers in all neurons or into late blastomeres to target specific classes of neurons based on established lineage maps. Xenopus spinal neurons also provide an excellent culture system, as neurons extend processes on a variety of substrata and develop at room temperature in minimal salt solutions. Live fluorescent neurons can be imaged for hours with fluorescence microscopy at room temperature in static cultures without neurotrophic support or serum. This highly reduced culture system minimizes variables that can confound interpretation of results. Cultures can be prepared at various stages of development as dissociated neurons or as spinal cord explants. Both excitatory and inhibitory neurons develop in culture, and synaptic contacts among neurons and between neurons and nonneuronal targets form naturally. The simple anatomy and rapid rostral-to-caudal development of the Xenopus spinal cord also make this an excellent in vivo model system to analyze axon guidance by identifiable classes of neurons. This chapter focuses on techniques that exploit both in vitro and in vivo qualities of this system.
Collapse
Affiliation(s)
- Timothy M Gómez
- Department of Anatomy, Cell and Molecular Biology Training Program, University of Wisconsin Medical School, Madison, Wisconsin 53706, USA
| | | | | | | |
Collapse
|
43
|
Abstract
Dynamic actin filaments contribute to cell migration, organelle movements, memory, and gene regulation. These dynamic processes are often regulated by extracellular and?or cell cycle signals. Regulation targets, not actin itself, but the factors that determine it's dynamic properties. Thus, filament nucleation, rate and duration of elongation, and depolymerization are each controlled with regard to time and?or space. Two mechanisms exist for nucleating filaments de novo, the Arp23 complex and the formins; multiple pathways regulate each. A new filament elongates rapidly but transiently before its barbed end is capped. Rapid capping allows the cell to maintain fine temporal and spatial control over F-actin distribution. Modulation of capping protein activity and its access to barbed ends is emerging as a site of local regulation. Finally, to maintain a steady state filaments must depolymerize. Depolymerization can limit the rate of new filament nucleation and elongation. The activity of ADF?cofilin, which facilitates depolymerization, is also regulated by multiple inputs. This chapter describes (1) mechanism and regulation of new filament formation, (2) mechanism of enhancing elongation at barbed ends, (3) capping proteins and their regulators, and (4) recycling of actin monomers from filamentous actin (F-actin) back to globular actin (G-actin).
Collapse
Affiliation(s)
- Sally H Zigmond
- Biology Department, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
44
|
Uyeda TQP, Nagasaki A, Yumura S. Multiple Parallelisms in Animal Cytokinesis. INTERNATIONAL REVIEW OF CYTOLOGY 2004; 240:377-432. [PMID: 15548417 DOI: 10.1016/s0074-7696(04)40004-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The process of cytokinesis in animal cells is usually presented as a relatively simple picture: A cleavage plane is first positioned in the equatorial region by the astral microtubules of the anaphase mitotic apparatus, and a contractile ring made up of parallel filaments of actin and myosin II is formed and encircles the cortex at the division site. Active sliding between the two filament systems constricts the perimeter of the cortex, leading to separation of two daughter cells. However, recent studies in both animal cells and lower eukaryotic model organisms have demonstrated that cytokinesis is actually far more complex. It is now obvious that the three key processes of cytokinesis, cleavage plane determination, equatorial furrowing, and scission, are driven by different mechanisms in different types of cells. In some cases, moreover, multiple pathways appear to have redundant functions in a single cell type. In this review, we present a novel hypothesis that incorporates recent observations on the activities of mitotic microtubules and the biochemistry of Rho-type GTPase proteins and postulates that two different sets of microtubules are responsible for the two known mechanisms of cleavage plane determination and also for two distinct mechanisms of equatorial furrowing.
Collapse
Affiliation(s)
- Taro Q P Uyeda
- Gene Function Research Center, National Institute for Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | | | | |
Collapse
|
45
|
Ono S. Regulation of Actin Filament Dynamics by Actin Depolymerizing Factor/Cofilin and Actin-Interacting Protein 1: New Blades for Twisted Filaments. Biochemistry 2003; 42:13363-70. [PMID: 14621980 DOI: 10.1021/bi034600x] [Citation(s) in RCA: 159] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Actin depolymerizing factor (ADF)/cofilin enhances turnover of actin filaments by severing and depolymerizing filaments. A number of proteins functionally interact with ADF/cofilin to modulate the dynamics of actin filaments. Actin-interacting protein 1 (AIP1) has emerged as a conserved WD-repeat protein that specifically enhances ADF/cofilin-induced actin dynamics. Interaction of AIP1 with actin was originally characterized by a yeast two-hybrid system. However, biochemical studies revealed its unique activity on ADF/cofilin-bound actin filaments. AIP1 alone has negligible effects on actin filament dynamics, whereas in the presence of ADF/cofilin, AIP1 enhances filament fragmentation by capping ends of severed filaments. Studies in model organisms demonstrated that AIP1 genetically interacts with ADF/cofilin and participates in several actin-dependent cellular events. The crystal structure of AIP1 revealed its unique structure with two seven-bladed beta-propeller domains. Thus, AIP1 is a new class of actin regulatory proteins that selectively enhances ADF/cofilin-dependent actin filament dynamics.
Collapse
Affiliation(s)
- Shoichiro Ono
- Department of Pathology, Emory University, Atlanta, Georgia 30322, USA.
| |
Collapse
|
46
|
Silverman-Gavrila RV, Forer A. Myosin localization during meiosis I of crane-fly spermatocytes gives indications about its role in division. CELL MOTILITY AND THE CYTOSKELETON 2003; 55:97-113. [PMID: 12740871 DOI: 10.1002/cm.10112] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We showed previously that in crane-fly spermatocytes myosin is required for tubulin flux [Silverman-Gavrila and Forer, 2000a: J Cell Sci 113:597-609], and for normal anaphase chromosome movement and contractile ring contraction [Silverman-Gavrila and Forer, 2001: Cell Motil Cytoskeleton 50:180-197]. Neither the identity nor the distribution of myosin(s) were known. In the present work, we used immunofluorescence and confocal microscopy to study myosin during meiosis-I of crane-fly spermatocytes compared to tubulin, actin, and skeletor, a spindle matrix protein, in order to further understand how myosin might function during cell division. Antibodies to myosin II regulatory light chain and myosin II heavy chain gave similar staining patterns, both dependent on stage: myosin is associated with nuclei, asters, centrosomes, chromosomes, spindle microtubules, midbody microtubules, and contractile rings. Myosin and actin colocalization along kinetochore fibers from prometaphase to anaphase are consistent with suggestions that acto-myosin forces in these stages propel kinetochore fibres poleward and trigger tubulin flux in kinetochore fibres, contributing in this way to poleward chromosome movement. Myosin and actin colocalization at the cell equator in cytokinesis, similar to studies in other cells [e.g., Fujiwara and Pollard, 1978: J Cell Biol 77:182-195], supports a role of actin-myosin interactions in contractile ring function. Myosin and skeletor colocalization in prometaphase spindles is consistent with a role of these proteins in spindle formation. After microtubules or actin were disrupted, myosin remained in spindles and contractile rings, suggesting that the presence of myosin in these structures does not require the continued presence of microtubules or actin. BDM (2,3 butanedione, 2 monoxime) treatment that inhibits chromosome movement and cytokinesis also altered myosin distributions in anaphase spindles and contractile rings, consistent with the physiological effects, suggesting also that myosin needs to be active in order to be properly distributed.
Collapse
|
47
|
Abstract
Eukaryotic cells use actin polymerization to change shape, move, and internalize extracellular materials by phagocytosis and endocytosis, and to form contractile structures. In addition, several pathogens have evolved to use host cell actin assembly for attachment, internalization, and cell-to-cell spread. Although cells possess multiple mechanisms for initiating actin polymerization, attention in the past five years has focused on the regulation of actin nucleation-the formation of new actin filaments from actin monomers. The Arp2/3 complex and the multiple nucleation-promoting factors (NPFs) that regulate its activity comprise the only known cellular actin-nucleating factors and may represent a universal machine, conserved across eukaryotic phyla, that nucleates new actin filaments for various cellular structures with numerous functions. This review focuses on our current understanding of the mechanism of actin nucleation by the Arp2/3 complex and NPFs and how these factors work with other cytoskeletal proteins to generate structurally and functionally diverse actin arrays in cells.
Collapse
Affiliation(s)
- Matthew D Welch
- Department of Molecular and Cell Biology, 301 LSA, University of California, Berkeley 94720-3200, USA.
| | | |
Collapse
|
48
|
Ono K, Parast M, Alberico C, Benian GM, Ono S. Specific requirement for two ADF/cofilin isoforms in distinct actin-dependent processes in Caenorhabditis elegans. J Cell Sci 2003; 116:2073-85. [PMID: 12679387 DOI: 10.1242/jcs.00421] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Actin depolymerizing factor (ADF)/cofilin is an essential enhancer of actin turnover. Multicellular organisms express multiple ADF/cofilin isoforms in different patterns of tissue distribution. However, the functional significance of different ADF/cofilin isoforms is not understood. The Caenorhabditis elegans unc-60 gene generates two ADF/cofilins, UNC-60A and UNC-60B, by alternative splicing. These two ADF/cofilin proteins have different effects on actin dynamics in vitro, but their functional difference in vivo remains unclear. Here, we demonstrate that the two isoforms are expressed in different tissues and are required for distinct morphogenetic processes. UNC-60A was ubiquitously expressed in most embryonic cells and enriched in adult gonads, intestine and oocytes. In contrast, UNC-60B was specifically expressed in the body wall muscle, vulva and spermatheca. RNA interference of UNC-60A caused embryonic lethality with variable defects in cytokinesis and developmental patterning. In severely affected embryos, a cleavage furrow was formed and progressed but reversed before completion of the cleavage. Also, in some affected embryos, positioning of the blastomeres became abnormal, which resulted in embryonic arrest. In contrast, an unc-60B-null mutant was homozygous viable, underwent normal early embryogenesis and caused disorganization of actin filaments specifically in body wall muscle. These results suggest that the ADF/cofilin isoforms play distinct roles in specific aspects of actin reorganization in vivo.
Collapse
Affiliation(s)
- Kanako Ono
- Department of Pathology, Emory University, Whitehead IO5N, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
49
|
|
50
|
Abstract
Cytokinesis in many eukaryotes requires a contractile ring of actin and myosin that cleaves the cell in two. Little is known about how actin filaments and other components assemble into this ring structure and generate force. Here we show that the contractile ring in the fission yeast Schizosaccharomyces pombe is an active site of actin assembly. This actin polymerization activity requires Arp3, the formin Cdc12, profilin and WASP, but not myosin II or IQGAP proteins. Both newly polymerized actin filaments and pre-existing actin cables can contribute to the initial assembly of the ring. Once formed, the ring remains a dynamic structure in which actin and other ring components continuously assemble and disassemble from the ring every minute. The rate of actin polymerization can influence the rate of cleavage. Thus, actin polymerization driven by the Arp2/3 complex and formins is a central process in cytokinesis. Our studies show that cytokinesis is a more dynamic process than previously thought and provide a perspective on the mechanism of cell division.
Collapse
Affiliation(s)
- Robert J Pelham
- Department of Microbiology, Columbia University College of Physicians and Surgeons, 701 West 168th Street, New York, New York 10032, USA
| | | |
Collapse
|