Myosin II dynamics inDictyostelium: Determinants for filament assembly and translocation to the cell cortex during chemoattractant responses

2002 ◽  
Vol 53 (3) ◽  
pp. 177-188 ◽  
Author(s):  
Stephanie Levi ◽  
Mark V. Polyakov ◽  
Thomas T. Egelhoff
2020 ◽  
Vol 219 (8) ◽  
Author(s):  
Bernardo Chapa-y-Lazo ◽  
Motonari Hamanaka ◽  
Alexander Wray ◽  
Mohan K. Balasubramanian ◽  
Masanori Mishima

Nearly six decades ago, Lewis Wolpert proposed the relaxation of the polar cell cortex by the radial arrays of astral microtubules as a mechanism for cleavage furrow induction. While this mechanism has remained controversial, recent work has provided evidence for polar relaxation by astral microtubules, although its molecular mechanisms remain elusive. Here, using C. elegans embryos, we show that polar relaxation is achieved through dynein-mediated removal of myosin II from the polar cortexes. Mutants that position centrosomes closer to the polar cortex accelerated furrow induction, whereas suppression of dynein activity delayed furrowing. We show that dynein-mediated removal of myosin II from the polar cortexes triggers a bidirectional cortical flow toward the cell equator, which induces the assembly of the actomyosin contractile ring. These results provide a molecular mechanism for the aster-dependent polar relaxation, which works in parallel with equatorial stimulation to promote robust cytokinesis.


BIOPHYSICS ◽  
2006 ◽  
Vol 51 (5) ◽  
pp. 764-770
Author(s):  
D. V. Serebryanaya ◽  
O. V. Shcherbakova ◽  
T. V. Dudnakova ◽  
V. P. Shirinsky ◽  
A. V. Vorotnikov

2000 ◽  
Vol 150 (4) ◽  
pp. 823-838 ◽  
Author(s):  
Douglas N. Robinson ◽  
James A. Spudich

We have developed a system for performing interaction genetics in Dictyostelium discoideum that uses a cDNA library complementation/multicopy suppression strategy. Chemically mutagenized cells were screened for cytokinesis-deficient mutants and one mutant was subjected to library complementation. Isolates of four different genes were recovered as modifiers of this strain's cytokinesis defect. These include the cleavage furrow protein cortexillin I, a novel protein we named dynacortin, an ezrin-radixin-moesin-family protein, and coronin. The cortexillin I locus and transcript were found to be disrupted in the strain, identifying it as the affected gene. Dynacortin is localized partly to the cell cortex and becomes enriched in protrusive regions, a localization pattern that is similar to coronin and partly dependent on RacE. During cytokinesis, dynacortin is found in the cortex and is somewhat enriched at the poles. Furthermore, it appears to be reduced in the cleavage furrow. The genetic interactions and the cellular distributions of the proteins suggest a hypothesis for cytokinesis in which the contraction of the medial ring is a function of spatially restricted cortexillin I and myosin II and globally distributed dynacortin, coronin, and RacE.


2020 ◽  
Author(s):  
Nazlı Ezgi Özkan-Küçük ◽  
Mohammad Haroon Qureshi ◽  
Berfu Nur Yiğit ◽  
Altuğ Kamacıoğlu ◽  
Nima Bavili ◽  
...  

ABSTRACTSuccessful cell division requires dramatic reorganization of the cell cortex in coordination with actomyosin cytoskeleton organization, membrane trafficking and cell adhesion. Although the contractile actomyosin ring is considered as hallmark of cytokinesis, in some cell types cell adhesion systems have been shown to drive cytokinesis independently from actomyosin function. We previously reported that Protocadherin 7 (PCDH7) localizes to the mitotic cortex which is required for building up the full mitotic rounding pressure. Here, we show that PCDH7 localizes to the mitotic cell cortex and to the cleavage furrow by a palmitoylation-dependent mechanism. At the cleavage furrow, PCDH7 facilitates the activation of myosin II and successful cytokinesis. Strikingly, PCDH7 promotes cytokinesis even when the myosin II contractility and integrin mediated adhesion are blocked. This work describes a palmitoylation-dependent cortical reorganization which promotes cytokinesis under different conditions.


1999 ◽  
Vol 147 (5) ◽  
pp. 1039-1048 ◽  
Author(s):  
Wenchuan Liang ◽  
Hans M. Warrick ◽  
James A. Spudich

Myosin II thick filament assembly in Dictyostelium is regulated by phosphorylation at three threonines in the tail region of the molecule. Converting these three threonines to aspartates (3×Asp myosin II), which mimics the phosphorylated state, inhibits filament assembly in vitro, and 3×Asp myosin II fails to rescue myosin II–null phenotypes. Here we report a suppressor screen of Dictyostelium myosin II–null cells containing 3×Asp myosin II, which reveals a 21-kD region in the tail that is critical for the phosphorylation control. These data, combined with new structural evidence from electron microscopy and sequence analyses, provide evidence that thick filament assembly control involves the folding of myosin II into a bent monomer, which is unable to incorporate into thick filaments. The data are consistent with a structural model for the bent monomer in which two specific regions of the tail interact to form an antiparallel tetrameric coiled–coil structure.


2020 ◽  
Vol 127 (Suppl_1) ◽  
Author(s):  
Kuldeep Singh ◽  
Anne B Kim ◽  
Kathleen G Morgan

Non-muscle myosin II plays a role in many fundamental cellular processes including cell adhesion, migration, and cytokinesis. However, its role in vascular function is not well understood. Here, we investigated the function of non-muscle myosin II in the biomechanical properties of mouse proximal aorta. We found that blebbistatin, a specific inhibitor of non-muscle myosin II decreases agonist-induced aortic stress and stiffness in a dose-dependent manner. We also specifically demonstrate, in freshly isolated contractile aortic smooth muscle cells, using deconvolution microscopy that the NM myosin IIA isoform co-localizes with contractile filaments in the core of the cell as well as in the non-muscle cell cortex. However, the NM myosin IIB isoform is only colocalized with contractile filaments, and is excluded from the cell cortex. Furthermore, both the siRNA knockdown of NMIIA and NMIIB isoforms in a differentiated smooth muscle cell line A7r5 and blebbistatin-mediated inhibition of NM myosin II suppresses agonist-activated increases in phosphorylation of FAK Y925 and paxillin Y118. Thus, in the present study, we show, for the first time, that NM myosin II regulates aortic stiffness and that this regulation is mediated at least in part through the tension-dependent phosphorylation of focal adhesion proteins FAK and paxillin.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1595-1595
Author(s):  
Jae-Won Shin ◽  
Kyle R Spinler ◽  
Joe Swift ◽  
Dennis E Discher

Abstract Abstract 1595 Hematopoietic stem cells (HSCs) are maintained in a quiescent state in a bone marrow niche, and become activated to give rise to different types of blood cells. The hierarchical design of hematopoiesis suggests that cell division-cycle machinery of each subpopulation could be differentially regulated. Our laboratory and others have previously demonstrated that non-muscle myosin-II is one of the major cytoskeletal proteins essential for cell fate decisions via adhesion to matrix and cytokinesis, both of which influence hematopoiesis. While myosin-II is essential for embryonic development, very little is known about its roles in regulating different stages of adult hematopoiesis. To test which stages of human hematopoiesis require myosin-II, bone marrow-derived CD34+ cells were cultured in serum-free medium with different combinations of cytokines followed by pharmacological inhibition of myosin over several cell cycles with blebbistatin. Multi-color flow cytometry analysis reveals that myosin inhibition leads to selective elimination of progenitor subpopulations via apoptotic cell death, while long-term HSC (LT-HSC) subpopulations remain viable. Transplantation of CD34+-derived cells treated with blebbistatin into immuno-deficient xenograft mice (NOD/Shi-scid/IL-2Rnull; NSG) shows normal LT-HSC engraftment comparable to untreated cells, producing both myeloid and lymphoid lineages. Colony-forming assays confirm dose-dependent reduction of multipotent, granulocyte-macrophage and erythroid progenitors by myosin inhibition. Under culture conditions that promote differentiation of megakaryocytic (MK) lineages, myosin inhibition produces a 3–10-fold increase of polyploid MKs in suspension cultures, whereas inhibition of other pathways that also affect contractility, including the Rho-kinase pathway, have lesser effects. Myosin-II inhibition also softens the cell cortex and enhances fragmentation upon aspiration into a micropipette. Such shear flow-induced fragmentation of distensible MK processes is known to be key to how MKs in the marrow shed platelets (plts) into permeating capillaries. To test whether myosin inhibition increases plt generation in vivo, human CD34+-derived cells treated with blebbistatin were transplanted intra-tibially in NSG mice. Transplantation of untreated cells produced detectable circulating human CD41+ plts as early as 8hrs with a peak at 48∼72hrs, followed by a complete decline at 2 wks. Blebbistatin-treated cells produce plts at a more sustained level with a slower rate of decline after a peak. Overall, myosin inhibition enhances the generation of circulating human plts per CD41+ cell transplanted: untreated cells yield 229 ± 114 plts/day whereas blebbistatin treatment yields 804 ± 256 plts/day. Plts from both treated and untreated human cells show activated adhesion on collagen assessed ex vivo. Together these data highlight a dual role for myosin-II in adult hematopoiesis in that: 1. Myosin-II is required for survival of hematopoietic myeloid progenitors 2. Myosin-II inhibition accelerates maturation of MKs and plt generation, while maintaining survival of LT-HSCs. Disclosures: No relevant conflicts of interest to declare.


2009 ◽  
Vol 20 (1) ◽  
pp. 200-208 ◽  
Author(s):  
Guangwei Du ◽  
Michael A. Frohman

When cells cease migrating through the vasculature, adhere to extracellular matrix, and begin to spread, they exhibit rapid changes in contraction and relaxation at peripheral regions newly contacting the underlying substrata. We describe here a requirement in this process for myosin II disassembly at the cell cortex via the action of myosin phosphatase (MP), which in turn is regulated by a plasma membrane signaling lipid. Cells in suspension exhibit high levels of activity of the signaling enzyme phospholipase D2 (PLD2), elevating production of the lipid second messenger phosphatidic acid (PA) at the plasma membrane, which in turn recruits MP and stores it there in a presumed inactive state. On cell attachment, down-regulation of PLD2 activity decreases PA production, leading to MP release, myosin dephosphorylation, and actomyosin disassembly. This novel model for recruitment and restraint of MP provides a means to effect a rapid cytoskeletal reorganization at the cell cortex upon demand.


Sign in / Sign up

Export Citation Format

Share Document