Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Exp Cell Res ; 386(1): 111707, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31693874

ABSTRACT

Cytokinesis of animal cells requires contraction of a contractile ring, composed of actin filaments and myosin II filaments. Phosphorylation of myosin II regulatory light chain (MRLC) promotes contraction of the actomyosin ring by activating myosin II motor activity. Both Rho-associated coiled-coil kinase (Rho kinase/ROCK) and Zipper-interacting protein kinase (ZIP kinase/ZIPK) have been reported to phosphorylate MRLC at the contractile ring. However, it remains unclear whether these kinases function independently of each other. Here, we clarified that ROCK colocalizes and forms a complex with ZIPK at telophase. As ROCK is reported to phosphorylate and activate ZIPK in vitro, we hypothesized that ZIPK phosphorylated by ROCK contributes to control cytokinesis. To address this, we expressed EGFP-ZIPK wild type (WT), a non-phosphorylatable mutant (T265A) or a phosphorylation-mimicking mutant (T265D) in HeLa cells and treated these cells with a ROCK inhibitor. Decrease in phosphorylated MRLC and a delay of furrow ingression by the ROCK inhibitor were rescued by the expression of EGFP-ZIPK-T265D, but not EGFP-ZIPK-WT or -T265A. This suggests that ROCK regulates MRLC phosphorylation followed by furrow ingression, through ZIPK phosphorylation.


Subject(s)
Cytokinesis , Death-Associated Protein Kinases/metabolism , rho-Associated Kinases/metabolism , Death-Associated Protein Kinases/genetics , HeLa Cells , Humans , Loss of Function Mutation , Myosin Light Chains/metabolism , Phosphorylation
2.
Biochem Biophys Res Commun ; 533(4): 1095-1101, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33032825

ABSTRACT

Death-associated protein kinase 3 (DAPK3), a member of the DAPK family, contributes to cytokinesis by phosphorylating myosin II regulatory light chain (MRLC). Missense mutations in DAPK3, T112M, D161N, and P216S, were observed in the lung, colon, and cervical cancers, respectively, but the effects of these mutations on cytokinesis remain unclear. Here, we show that cells expressing EGFP-DAPK3-T112M, -D161N, or -P216S exhibited reduced rates of cytokinesis, with an increased ratio of multinucleated cells. In addition, these cells exhibited reduced levels of phosphorylated MRLC at the contractile ring. Collectively, our data demonstrates that cancer-associated DAPK3 mutations impair cytokinesis by reducing phosphorylated MRLC.


Subject(s)
Cytokinesis/genetics , Death-Associated Protein Kinases/genetics , Myosin Light Chains/metabolism , Death-Associated Protein Kinases/metabolism , HeLa Cells , Humans , Mutation, Missense , Phosphorylation
3.
Biochem Biophys Res Commun ; 459(4): 686-91, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25769953

ABSTRACT

Zipper-interacting protein kinase (ZIPK) is known to regulate several functions such as apoptosis, smooth muscle contraction, and cell migration. While exogenously expressed GFP-ZIPK localizes to the cleavage furrow, role of ZIPK in cytokinesis is obscure. Here, we show that ZIPK is a major MRLC kinase during mitosis. Moreover, ZIPK siRNA-mediated knockdown causes delay of cytokinesis. The delay in cytokinesis of ZIPK-knockdown cells was rescued by the exogenous diphosphorylation-mimicking MRLC mutant. Taken together, these findings suggest that ZIPK plays a role in the progression and completion of cytokinesis through MRLC phosphorylation.


Subject(s)
Cell Division , Death-Associated Protein Kinases/metabolism , Myosin Type II/metabolism , Cell Line , Death-Associated Protein Kinases/genetics , Humans , Phosphorylation , RNA, Small Interfering/genetics
4.
Exp Cell Res ; 318(8): 915-24, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22374324

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.


Subject(s)
Actins/metabolism , Myosin Light Chains/metabolism , Myosin Type II/metabolism , Actin Cytoskeleton/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cytokinesis/physiology , HeLa Cells , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Myosin Type II/antagonists & inhibitors , Phosphorylation , Thiazolidines/pharmacology
5.
Biochem Biophys Res Commun ; 417(2): 686-91, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22166199

ABSTRACT

Myosin II is activated by the monophosphorylation of its regulatory light chain (MRLC) at Ser19 (1P-MRLC). Its ATPase activity is further enhanced by MRLC diphosphorylation at Thr18/Ser19 (2P-MRLC). As these phosphorylated MRLCs are colocalized with their heavy chains at the contractile ring in dividing cells, we believe that the phosphorylated MRLC acts as a subunit of the activated myosin II during cytokinesis. However, the distinct role(s) of 1P- and 2P-MRLC during cytokinesis has not been elucidated. In this study, a monoclonal antibody (4F12) specific for 2P-MRLC was raised and used to examine the roles of 2P-MRLC in cultured mammalian cells. Our confocal microscopic observations using 4F12 revealed that 2P-MRLC localized to the contractile ring, and, unexpectedly, to the midzone also. Interestingly, 2P-MRLC did not colocalize with 1P-MRLC, myosin II heavy chain, and F-actin at the midzone. These results suggest that 2P-MRLC has a role different from that of 1P-MRLC at the midzone, and is not a subunit of myosin II.


Subject(s)
Cytokinesis , Myosin Light Chains/metabolism , Antibodies, Monoclonal , HeLa Cells , Humans , Myosin Light Chains/genetics , Phosphorylation
6.
Biochem J ; 435(3): 569-76, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21231914

ABSTRACT

Myosin II ATPase activity is enhanced by the phosphorylation of MRLC (myosin II regulatory light chain) in non-muscle cells. It is well known that pMRLC (phosphorylated MRLC) co-localizes with F-actin (filamentous actin) in the CR (contractile ring) of dividing cells. Recently, we reported that HeLa cells expressing non-phosphorylatable MRLC show a delay in the speed of furrow ingression, suggesting that pMRLC plays an important role in the control of furrow ingression. However, it is still unclear how pMRLC regulates myosin II and F-actin at the CR to control furrow ingression during cytokinesis. In the present study, to clarify the roles of pMRLC, we measured the turnover of myosin II and actin at the CR in dividing HeLa cells expressing fluorescent-tagged MRLCs and actin by FRAP (fluorescence recovery after photobleaching). A myosin II inhibitor, blebbistatin, caused an enhancement of the turnover of MRLC and actin at the CR, which induced a delay in furrow ingression. Furthermore, only non-phosphorylatable MRLC and a Rho-kinase inhibitor, Y-27632, accelerated the turnover of MRLC and actin at the CR. Interestingly, the effect of Y-27632 was cancelled in the cell expressing phosphomimic MRLCs. Taken together, these results reveal that pMRLC reduces the turnover of myosin II and also actin at the CR. In conclusion, we show that the enhancement of myosin II and actin turnover at the CR induced slower furrowing in dividing HeLa cells.


Subject(s)
Actins/metabolism , Mitosis/physiology , Myosin Type II/metabolism , Actins/genetics , Amides , Cell Membrane/physiology , Gene Expression Regulation/physiology , HeLa Cells , Heterocyclic Compounds, 4 or More Rings , Humans , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Myosin Type II/genetics , Protein Transport , Pyridines
7.
Cytoskeleton (Hoboken) ; 79(9-11): 94-104, 2022 09.
Article in English | MEDLINE | ID: mdl-36053962

ABSTRACT

Microtubule stability and dynamics regulations are essential for vital cellular processes, such as microtubule-dependent axonal transport. Dynamin is involved in many membrane fission events, such as clathrin-mediated endocytosis. The ubiquitously expressed dynamin-2 has been reported to regulate microtubule stability. However, the underlying molecular mechanisms remain unclear. This study aimed to investigate the roles of intrinsic properties of dynamin-2 on microtubule regulation by rescue experiments. A heterozygous DNM2 mutation in HeLa cells was generated, and an increase in the level of stabilized microtubules in these heterozygous cells was observed. The expression of wild-type dynamin-2 in heterozygous cells reduced stabilized microtubules. Conversely, the expression of self-assembly-defective mutants of dynamin-2 in the heterozygous cells failed to decrease stabilized microtubules. This indicated that the self-assembling ability of dynamin-2 is necessary for regulating microtubule stability. Moreover, the heterozygous cells expressing the GTPase-defective dynamin-2 mutant, K44A, reduced microtubule stabilization, similar to the cells expressing wild-type dynamin-2, suggesting that GTPase activity of dynamin-2 is not essential for regulating microtubule stability. These results showed that the mechanism of microtubule regulation by dynamin-2 is diverse from that of endocytosis.


Subject(s)
Dynamins , Endocytosis , Microtubules , Humans , Dynamins/genetics , Endocytosis/physiology , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , HeLa Cells , Microtubules/metabolism
8.
Genes Cells ; 14(5): 555-68, 2009 May.
Article in English | MEDLINE | ID: mdl-19371382

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.


Subject(s)
Cytokinesis/physiology , Myosin Light Chains/metabolism , Myosin Type II/metabolism , Actins/drug effects , Actins/metabolism , Amides/pharmacology , Cytokinesis/drug effects , HeLa Cells , Humans , Myosin Light Chains/drug effects , Myosin Type II/drug effects , Phosphorylation , Pyridines/pharmacology
9.
Exp Cell Res ; 315(7): 1336-45, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19331814

ABSTRACT

Microtubule reorganization is necessary for many cellular functions such as cell migration, cell polarity and cell division. Dynamin was originally identified as a microtubule-binding protein. Previous limited digestion experiment revealed that C-terminal 100-amino acids proline rich domain (PRD) of dynamin is responsible for microtubule binding in vitro. However, as obvious localization of dynamin along microtubules is only observed at the spindle midzone during mitosis but not in interphase cells, it remains unclear how dynamin interacts with microtubules in vivo. Here, we report that GFP-dynamin-2-(1-786), a truncated mutant lacking a C-terminal portion of the PRD, localized along microtubules in interphase HeLa cells. GFP-dynamin-2-wild type (WT) and GFP-dynamin-2-(1-745), a construct that was further truncated to remove the entire PRD, localized in discrete punctate structures but not along microtubules. These data suggest that the N-terminal (residues 746-786) but not the entire PRD is necessary for the interaction of dynamin-2 with microtubules in the cell and that the C-terminus of PRD (787-870) negatively regulate this interaction. Microtubules in cells expressing GFP-dynamin-2-(1-786) were stabilized against exposure to cold. These results provide a first evidence for a regulated interaction of dynamin-2 with microtubules in cultured mammalian cells.


Subject(s)
Dynamin II/metabolism , Microtubules/metabolism , Animals , Cell Cycle/physiology , Dynamin II/genetics , HeLa Cells , Humans , Nocodazole/metabolism , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tubulin Modulators/metabolism
10.
Sci Rep ; 9(1): 12729, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31484968

ABSTRACT

Cytokinesis is initiated by the formation and ingression of the cleavage furrow. Phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] accumulation followed by RhoA translocation to the cleavage furrow are prerequisites for cytokinesis progression. Here, we investigated whether phospholipase C (PLC)-related catalytically inactive protein (PRIP), a metabolic modulator of PI(4,5)P2, regulates PI(4,5)P2-mediated cytokinesis. We found that PRIP localised to the cleavage furrow during cytokinesis. Moreover, HeLa cells with silenced PRIP displayed abnormal cytokinesis. Importantly, PI(4,5)P2 accumulation at the cleavage furrow, as well as the localisation of RhoA and phospho-myosin II regulatory light chain to the cleavage furrow, were reduced in PRIP-silenced cells. The overexpression of oculocerebrorenal syndrome of Lowe-1 (OCRL1), a phosphatidylinositol-5-phosphatase, in cells decreased PI(4,5)P2 levels during early cytokinesis and resulted in cytokinesis abnormalities. However, these abnormal cytokinesis phenotypes were ameliorated by the co-expression of PRIP but not by co-expression of a PI(4,5)P2-unbound PRIP mutant. Collectively, our results indicate that PRIP is a component at the cleavage furrow that maintains PI(4,5)P2 metabolism and regulates RhoA-dependent progression of cytokinesis. Thus, we propose that PRIP regulates phosphoinositide metabolism correctively and mediates normal cytokinesis progression.


Subject(s)
Cell Membrane/metabolism , Cytokinesis , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphoinositide Phospholipase C/metabolism , Cell Membrane/genetics , HeLa Cells , Humans , Phosphoinositide Phospholipase C/genetics , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism
12.
Cytoskeleton (Hoboken) ; 72(12): 609-20, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26663899

ABSTRACT

Myosin II regulatory light chain (MRLC) is canonically known as a subunit of conventional myosin (myosin II), which tunes cytoplasmic contractility in cells. Recent studies have also revealed the noncanonical functions of MRLC, such as engagement with other proteins including unconventional myosins. Three MRLC isoforms (MRLC1, MRLC2, and MRLC3) are known in humans. The characteristics of MRLC2 are well known, but those of MRLC1 and MRLC3 are unclear; therefore, the properties of the three MRLC isoforms were investigated. The MRLCs were all phosphorylated at Thr18/Ser19, which is required for myosin II stimulation. MRLC mRNAs were expressed at the same level throughout the cell cycle in HeLa cells. The MRLCs colocalized with each other and their turnover rate was similar to that of myosin II heavy chain. Depletion of all the MRLCs perturbed cell spreading. The overproduction of MRLC2 or MRLC3, but not MRLC1, could effectively compensate for this defect, suggesting that MRLC2 and MRLC3 play dominant roles in cell spreading. Finally, computer simulations of the three-dimensional protein structures indicated that the location of the N-terminus of MRLC1 differs from that of MRLC2 or MRLC3, depending on its sequence. Thus, these MRLC isoforms have overlapping but distinct functions have been proposed.


Subject(s)
Myosin Light Chains/metabolism , Protein Isoforms/metabolism , HeLa Cells , Humans
13.
PLoS One ; 8(8): e70965, 2013.
Article in English | MEDLINE | ID: mdl-23951055

ABSTRACT

Non-muscle myosin II is stimulated by monophosphorylation of its regulatory light chain (MRLC) at Ser19 (1P-MRLC). MRLC diphosphorylation at Thr18/Ser19 (2P-MRLC) further enhances the ATPase activity of myosin II. Phosphorylated MRLCs localize to the contractile ring and regulate cytokinesis as subunits of activated myosin II. Recently, we reported that 2P-MRLC, but not 1P-MRLC, localizes to the midzone independently of myosin II heavy chain during cytokinesis in cultured mammalian cells. However, the mechanism underlying the distinct localization of 1P- and 2P-MRLC during cytokinesis is unknown. Here, we showed that depletion of the Rho signaling proteins MKLP1, MgcRacGAP, or ECT2 inhibited the localization of 1P-MRLC to the contractile ring but not the localization of 2P-MRLC to the midzone. In contrast, depleting or inhibiting a midzone-localizing kinase, Aurora B, perturbed the localization of 2P-MRLC to the midzone but not the localization of 1P-MRLC to the contractile ring. We did not observe any change in the localization of phosphorylated MRLC in myosin light-chain kinase (MLCK)-inhibited cells. Furrow regression was observed in Aurora B- and 2P-MRLC-inhibited cells but not in 1P-MRLC-perturbed dividing cells. Furthermore, Aurora B bound to 2P-MRLC in vitro and in vivo. These results suggest that Aurora B, but not Rho/MLCK signaling, is essential for the localization of 2P-MRLC to the midzone in dividing HeLa cells.


Subject(s)
Aurora Kinase B/metabolism , Cytokinesis , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/metabolism , Signal Transduction , rho-Associated Kinases/metabolism , Aurora Kinase B/genetics , Blotting, Western , Fluorescent Antibody Technique , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , HeLa Cells , Humans , Microscopy, Confocal , Myosin-Light-Chain Kinase/genetics , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , rho-Associated Kinases/genetics
14.
J Biochem ; 148(5): 533-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20889493

ABSTRACT

The large GTPase dynamin is strongly accumulated in the constricted area including midzonal microtubules of dividing cells. The proline-rich domain (PRD) of dynamin has been considered as a microtubule-binding domain. However, it remains unclear how PRD controls dynamin-microtubule interaction in mitotic cells. Here, we found that the microtubule-binding activity of PRD is low in dynamin-2. One of the mitosis-specific kinase activities to PRD in HeLa cells was identified as cyclin B-Cdc2 kinase. The kinase phosphorylated PRD at Ser(764) and/or Thr(766) and reduced the microtubule-binding activity of PRD. These results suggest that phosphorylation of PRD by cyclin B-Cdc2 kinase plays an important role to control dynamin-2-microtubule interaction in mitotic HeLa cells.


Subject(s)
Dynamin II/metabolism , Microtubules/metabolism , Proline-Rich Protein Domains/physiology , CDC2 Protein Kinase/metabolism , Dynamin II/chemistry , HeLa Cells , Humans , Mitosis/physiology , Phosphorylation
15.
Genes Cells ; 12(1): 49-61, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17212654

ABSTRACT

Dvl, an important component of the Wnt signalling pathway, is thought to be involved in synaptogenesis. In this study, we investigated whether Dvl regulates neurotransmitter release. Knockdown of Dvl in PC12 cells suppressed K(+)-induced dopamine release, and this phenotype was restored by expression of Dvl-1. We identified synaptotagmin (Syt) I, which is involved in neurotransmitter release, as a Dvl-binding protein. Dvl directly bound to the C2B domain of Syt I. Dvl colocalized with Syt I at the tip of neurites of differentiated PC12 cells and of neurons in the rat dorsal root ganglion. Dvl and Syt I was located in large dense-core vesicles, which contain dopamine. In addition, endocytosis of vesicles containing Syt I was suppressed in Dvl knockdown PC12 cells. Dvl inhibited the binding of Syt I to the complex consisting of syntaxin-1A and SNAP-25. Furthermore, micro2-adaptin of AP-2, which is known to play a role in endocytosis, formed a complex with Dvl and Syt I. Taken together, these results suggest that Dvl is involved in endo- and exocytotic processes through the binding to Syt I.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Endocytosis/physiology , Exocytosis/physiology , Phosphoproteins/metabolism , Synaptotagmin I/metabolism , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding Sites , Cell Differentiation , Dishevelled Proteins , Dopamine/metabolism , Humans , PC12 Cells , Phosphoproteins/analysis , Phosphoproteins/genetics , Protein Structure, Tertiary , Rats , Secretory Vesicles/metabolism , Synaptotagmin I/analysis , Synaptotagmin I/genetics
16.
Arch Biochem Biophys ; 456(2): 224-31, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17094935

ABSTRACT

Adhesive cells show complex mechanical interactions with the substrate, however the exact mechanism of such interactions, termed traction forces, is still unclear. To address this question we have measured traction forces of fibroblasts treated with agents that affect the myosin II-dependent contractile mechanism. Using the potent myosin II inhibitor blebbistatin, we demonstrate that traction forces are strongly dependent on a functional myosin II heavy chain. Since myosin II is regulated by both the myosin light chain kinase (MLCK) and, directly or indirectly, the Rho-associated kinase (ROCK), we examined the effects of inhibitors against these kinases. Interestingly, inhibition of the myosin light chain kinase had no detectable effect, while inhibition of the Rho-dependent kinase caused strong inhibition of traction forces. Our results indicate that ROCK and MLCK play non-redundant roles in regulating myosin II functions, and that a subset of myosin II, regulated by the Rho small GTPase, may be responsible for the regulation of traction forces in migrating fibroblasts.


Subject(s)
Cell Adhesion/physiology , Intracellular Signaling Peptides and Proteins/physiology , Mechanotransduction, Cellular/physiology , Myosin Light Chains/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Cell Adhesion/drug effects , Homeostasis/physiology , Mechanotransduction, Cellular/drug effects , Mice , NIH 3T3 Cells , Phosphorylation , Stress, Mechanical , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL