Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Tissue Eng Part A ; 24(15-16): 1218-1227, 2018 08.
Article in English | MEDLINE | ID: mdl-29397789

ABSTRACT

Human mesenchymal stem cell (hMSC)-based chondrogenesis is a key process used to develop tissue engineered cartilage constructs from stem cells, but the resulting constructs have inferior biochemical and biomechanical properties compared to native articular cartilage. Transforming growth factor ß containing medium is commonly applied to cell layers of hMSCs, which aggregate upon centrifugation to form 3-D constructs. The aggregation process leads to a high cell density condition, which can cause nutrient limitations during long-term culture and, subsequently, inferior quality of tissue engineered constructs. Our objective is to modulate the aggregation process by targeting RhoA/ROCK signaling pathway, the chief modulator of actomyosin contractility, to enhance the end quality of the engineered constructs. Through ROCK inhibition, repression of cytoskeletal tension in chondrogenic hMSCs was achieved along with less dense aggregates with enhanced transport properties. ROCK inhibition also led to significantly increased cartilaginous extracellular matrix accumulation. These findings can be used to create an improved microenvironment for hMSC-derived tissue engineered cartilage culture. We expect that these findings will ultimately lead to improved cartilaginous tissue development from hMSCs.


Subject(s)
Cartilage/enzymology , Chondrogenesis , Extracellular Matrix/metabolism , Mesenchymal Stem Cells/enzymology , Signal Transduction , rho-Associated Kinases/antagonists & inhibitors , Biological Transport, Active , Cartilage/cytology , Cells, Cultured , Cytoskeleton/metabolism , Humans , Mesenchymal Stem Cells/cytology , Tissue Engineering , rho-Associated Kinases/metabolism
2.
J Biol Chem ; 292(8): 3099-3111, 2017 02 24.
Article in English | MEDLINE | ID: mdl-28053086

ABSTRACT

Non-muscle myosin II (NMII) is a conserved force-producing cytoskeletal enzyme with important but poorly understood roles in cell migration. To investigate myosin heavy chain (MHC) phosphorylation roles in 3D migration, we expressed GFP-tagged NMIIA wild-type or mutant constructs in cells depleted of endogenous NMIIA protein. We find that individual mutation or double mutation of Ser-1916 or Ser-1943 to alanine potently blocks recruitment of GFP-NM-IIA filaments to leading edge protrusions in 2D, and this in turn blocks maturation of anterior focal adhesions. When placed in 3D collagen gels, cells expressing wild-type GFP MHC-IIA behave like parental cells, displaying robust and active formation and retraction of protrusions. However, cells depleted of NMIIA or cells expressing the mutant GFP MHC-IIA display severe defects in invasion and in stabilizing protrusions in 3D. These studies reveal an NMIIA-specific role in 3D invasion that requires competence for NMIIA phosphorylation at Ser-1916 and Ser-1943. In sum, these results demonstrate a critical and previously unrecognized role for NMIIA phosphorylation in 3D invasion.


Subject(s)
Cell Adhesion , Cell Movement , Myosin Heavy Chains/metabolism , Nonmuscle Myosin Type IIA/metabolism , Animals , COS Cells , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Humans , Mice , Myosin Heavy Chains/analysis , Nonmuscle Myosin Type IIA/analysis , Phosphorylation
3.
Am J Pathol ; 186(5): 1351-60, 2016 05.
Article in English | MEDLINE | ID: mdl-26988652

ABSTRACT

Our prior work identified the mammalian target of rapamycin complex 2 (mTORC2) as a key regulator of bladder cancer cell migration and invasion, although upstream growth factor mediators of this pathway in bladder cancer have not been well delineated. We tested whether transforming growth factor (TGF)-ß, which can function as a promotility factor in bladder cancer cells, could regulate mTORC2-dependent bladder cancer cell motility and invasion. In human bladder cancers, the highest levels of phosphorylated SMAD2, a TGF-ß signaling intermediate, were present in high-grade invasive bladder cancers and associated with more frequent recurrence and decreased disease-specific survival. Increased expression of TGF-ß isoforms, receptors, and signaling components was detected in invasive high-grade bladder cancer cells that expressed Vimentin and lacked E-cadherin. Application of TGF-ß induced phosphorylation of the Ser473 residue of AKT, a selective target of mTORC2, in a SMAD2- and SMAD4-independent manner and increased bladder cancer cell migration in a modified scratch wound assay and invasion through Matrigel. Inhibition of TGF-ß receptor I using SB431542 ablated TGF-ß-induced migration and invasion. A similar effect was seen when Rictor, a key mTORC2 component, was selectively silenced. Our results suggest that TGF-ß can induce bladder cancer cell invasion via mTORC2 signaling, which may be applicable in most bladder cancers.


Subject(s)
Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Transforming Growth Factor beta/physiology , Urinary Bladder Neoplasms/pathology , Benzamides/pharmacology , Cadherins/metabolism , Cell Movement/physiology , Dioxoles/pharmacology , Humans , Mechanistic Target of Rapamycin Complex 2 , Neoplasm Invasiveness , Phosphorylation/physiology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction/physiology , Smad2 Protein/metabolism , Smad4 Protein/metabolism , Tumor Cells, Cultured , Up-Regulation/physiology , Urinary Bladder Neoplasms/physiopathology , Vimentin/metabolism
4.
J Biol Chem ; 291(12): 6083-95, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26763235

ABSTRACT

Pro-fibrotic mesenchymal cells are known to be the key effector cells of fibroproliferative disease, but the specific matrix signals and the induced cellular responses that drive the fibrogenic phenotype remain to be elucidated. The key mediators of the fibroblast fibrogenic phenotype were characterized using a novel assay system that measures fibroblast behavior in response to actual normal and fibrotic lung tissue. Using this system, we demonstrate that normal lung promotes fibroblast motility and polarization, while fibrotic lung immobilizes the fibroblast and promotes myofibroblast differentiation. These context-specific phenotypes are surprisingly both mediated by myosin II. The role of myosin II is supported by the observation of an increase in myosin phosphorylation and a change in intracellular distribution in fibroblasts on fibrotic lung, as compared with normal lung. Moreover, loss of myosin II activity has opposing effects on protrusive activity in fibroblasts on normal and fibrotic lung. Loss of myosin II also selectively inhibits myofibroblast differentiation in fibroblasts on fibrotic lung. Importantly, these findings are recapitulated by varying the matrix stiffness of polyacrylamide gels in the range of normal and fibrotic lung tissue. Comparison of the effects of myosin inhibition on lung tissue with that of polyacrylamide gels suggests that matrix fiber organization drives the fibroblast phenotype under conditions of normal/soft lung, while matrix stiffness drives the phenotype under conditions of fibrotic/stiff lung. This work defines novel roles for myosin II as a key regulatory effector molecule of the pro-fibrotic phenotype, in response to biophysical properties of the matrix.


Subject(s)
Fibroblasts/physiology , Myosin Type II/physiology , Pulmonary Fibrosis/metabolism , Animals , Cell Differentiation , Cell Line , Cell Movement , Cell Polarity , Cell Shape , Extracellular Matrix/physiology , Female , Humans , Lung/metabolism , Lung/pathology , Mice, Inbred C57BL , Phenotype , Pulmonary Fibrosis/pathology
5.
J Cell Biol ; 210(4): 583-94, 2015 Aug 17.
Article in English | MEDLINE | ID: mdl-26261182

ABSTRACT

Non-muscle myosin II (NMII) is reported to play multiple roles during cell migration and invasion. However, the exact biophysical roles of different NMII isoforms during these processes remain poorly understood. We analyzed the contributions of NMIIA and NMIIB in three-dimensional (3D) migration and in generating the forces required for efficient invasion by mammary gland carcinoma cells. Using traction force microscopy and microfluidic invasion devices, we demonstrated that NMIIA is critical for generating force during active protrusion, and NMIIB plays a major role in applying force on the nucleus to facilitate nuclear translocation through tight spaces. We further demonstrate that the nuclear membrane protein nesprin-2 is a possible linker coupling NMIIB-based force generation to nuclear translocation. Together, these data reveal a central biophysical role for NMIIB in nuclear translocation during 3D invasive migration, a result with relevance not only to cancer metastasis but for 3D migration in other settings such as embryonic cell migration and wound healing.


Subject(s)
Cell Movement , Cell Nucleus/physiology , Nonmuscle Myosin Type IIB/physiology , Active Transport, Cell Nucleus , Animals , Biomechanical Phenomena , Cell Line, Tumor , Mice , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism
6.
Curr Biol ; 25(2): 175-186, 2015 Jan 19.
Article in English | MEDLINE | ID: mdl-25544611

ABSTRACT

BACKGROUND: Cell migration requires coordinated formation of focal adhesions (FAs) and assembly and contraction of the actin cytoskeleton. Nonmuscle myosin II (MII) is a critical mediator of contractility and FA dynamics in cell migration. Signaling downstream of the small GTPase Rac1 also regulates FA and actin dynamics, but its role in regulation of MII during migration is less clear. RESULTS: We found that Rac1 promotes association of MIIA with FA. Live-cell imaging showed that, whereas most MIIA at the leading edge assembled into dorsal contractile arcs, a substantial subset assembled in or was captured within maturing FA, and this behavior was promoted by active Rac1. Protein kinase C (PKC) activation was necessary and sufficient for integrin- and Rac1-dependent phosphorylation of MIIA heavy chain (HC) on serine1916 (S1916) and recruitment to FA. S1916 phosphorylation of MIIA HC and localization in FA was enhanced during cell spreading and ECM stiffness mechanosensing, suggesting upregulation of this pathway during physiological Rac1 activation. Phosphomimic and nonphosphorylatable MIIA HC mutants demonstrated that S1916 phosphorylation was necessary and sufficient for the capture and assembly of MIIA minifilaments in FA. S1916 phosphorylation was also sufficient to promote the rapid assembly of FAs to enhance cell migration and for the modulation of traction force, spreading, and migration by ECM stiffness. CONCLUSIONS: Our study reveals for the first time that Rac1 and integrin activation regulates MIIA HC phosphorylation through a PKC-dependent mechanism that promotes MIIA association with FAs and acts as a critical modulator of cell migration and mechanosensing.


Subject(s)
Cell Movement , Focal Adhesions/metabolism , Molecular Motor Proteins/genetics , Myosin Heavy Chains/genetics , Signal Transduction , rac1 GTP-Binding Protein/genetics , Cell Line , Humans , Mechanotransduction, Cellular/physiology , Molecular Motor Proteins/metabolism , Myosin Heavy Chains/metabolism , Phosphorylation , rac1 GTP-Binding Protein/metabolism
7.
PLoS One ; 8(11): e81081, 2013.
Article in English | MEDLINE | ID: mdl-24312263

ABSTRACT

Bladder cancer is the fourth most common cause of cancer in males in the United States. Invasive behavior is a major determinant of prognosis. In this study, we identified mammalian target of rapamycin complex 2 (mTORC2) as a central regulator of bladder cancer cell migration and invasion. mTORC2 activity was assessed by the extent of phosphorylation of Ser473 in AKT and determined to be approximately 5-fold higher in specimens of invasive human bladder cancer as opposed to non-invasive human bladder cancer. The immortalized malignant bladder cell lines, UMUC-3, J82 and T24 demonstrated higher baseline mTORC2 activity relative to the benign bladder papilloma-derived cell line RT4 and the normal urothelial cell line HU1. The malignant bladder cancer cells also demonstrated increased migration in transwell and denudation assays, increased invasion of matrigel, and increased capacity to invade human bladder specimens. Gene silencing of rictor, a critical component of mTORC2, substantially inhibited bladder cancer cell migration and invasion. This was accompanied by a significant decrease in Rac1 activation and paxillin phosphorylation. These studies identify mTORC2 as a major target for neutralizing bladder cancer invasion.


Subject(s)
Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Cell Movement , Disease Progression , Humans , Mechanistic Target of Rapamycin Complex 2 , Neoplasm Invasiveness , Signal Transduction , rac1 GTP-Binding Protein/metabolism
8.
Dev Biol ; 382(1): 136-48, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23911870

ABSTRACT

In studies initially focused on roles of nonmuscle myosin IIA (NMIIA) in the developing mouse epidermis, we have discovered that a previously described cytokeratin 5 (K5)-Cre gene construct is expressed in early embryo development. Mice carrying floxed alleles of the nonmuscle myosin II heavy chain gene (NMHC IIA(flox/flox)) were crossed with the K5-Cre line. The progeny of newborn pups did not show a Mendelian genotype distribution, suggesting embryonic lethality. Analysis of post-implantation conceptuses from embryonic day (E)9.5 to E13.5 revealed poorly developed embryos and defective placentas, with significantly reduced labyrinth surface area and blood vessel vascularization. These results suggested the novel possibility that the bovine K5 promoter-driven Cre-recombinase was active early in trophoblast-lineage cells that give rise to the placenta. To test this possibility, K5-Cre transgenic mice were crossed with the mT/mG reporter mouse in which activation of GFP expression indicates Cre transgene expression. We observed activation of K5-Cre-driven GFP expression in the ectoplacental cone, in the extraembryonic ectoderm, and in trophoblast giant cells in the E6.5 embryo. In addition, we observed GFP expression at E11.5 to E13.5 in both the labyrinth of the placenta and the yolk sac. NMIIA expression was detected in these same cell types in normal embryos, as well as in E13.5 yolk sac and labyrinth. These findings taken together suggest that NMHC IIA may play critical roles in the early trophoblast-derived ectoplacental cone and extraembryonic ectoderm, as well as in the yolk sac and labyrinth tissues that form later. Our findings are consistent with phenotypes of constitutive NMIIA knockout mice made earlier, that displayed labyrinth and yolk sac-specific defects, but our findings extend those observations by suggesting possible NMIIA roles in trophoblast lineages as well. These results furthermore demonstrate that K5-Cre gene constructs, previously reported to be activated starting at approximately E12.5 in the forming epidermis, may be widely useful as drivers for activation of cre/lox based gene excision in early embryo extraembronic trophoblast tissues as well.


Subject(s)
Ectoderm/embryology , Embryo Loss/pathology , Integrases/metabolism , Keratin-5/metabolism , Nonmuscle Myosin Type IIA/metabolism , Trophoblasts/metabolism , Trophoblasts/pathology , Alleles , Animals , Animals, Newborn , Blood Vessels/embryology , Blood Vessels/metabolism , Cattle , Cell Lineage , Cell Proliferation , Crosses, Genetic , Ectoderm/metabolism , Ectoderm/pathology , Embryo Loss/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Embryonic Development , Epidermis/embryology , Epidermis/metabolism , Epidermis/pathology , Female , Gene Deletion , Genotype , Ki-67 Antigen/metabolism , Male , Mice , Mice, Knockout , Pregnancy , Yolk Sac/metabolism
9.
Sci Signal ; 6(284): ra60, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23861542

ABSTRACT

Tumor necrosis factor-α (TNF-α) elicits its biological activities through activation of TNF receptor 1 (TNFR1, also known as p55) and TNFR2 (also known as p75). The activities of both receptors are required for the TNF-α-induced proinflammatory response. The adaptor protein TNFR-associated factor 2 (TRAF2) is critical for either p55- or p75-mediated activation of nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling, as well as for target gene expression. We identified nonmuscle myosin II (myosin) as a binding partner of p75. TNF-α-dependent signaling by p75 and induction of target gene expression persisted substantially longer in cells deficient in myosin regulatory light chain (MRLC; a component of myosin) than in cells replete in myosin. In resting endothelial cells, myosin was bound constitutively to the intracellular region of p75, a region that overlaps with the TRAF2-binding domain, and TNF-α caused the rapid dissociation of myosin from p75. At early time points after exposure to TNF-α, p75 activated Rho-associated kinase 1 (ROCK1). Inhibition of ROCK1 activity blocked TNF-α-dependent phosphorylation of MRLC and the dissociation of myosin from p75. ROCK1-dependent release of myosin was necessary for the TNF-α-dependent recruitment of TRAF2 to p75 and for p75-specific activation of NF-κB and MAPK signaling. Thus, our findings have revealed a previously uncharacterized, noncanonical regulatory function of myosin in cytokine signaling.


Subject(s)
Cytosol/metabolism , Gene Expression Regulation/physiology , Human Umbilical Vein Endothelial Cells/metabolism , MAP Kinase Signaling System/physiology , Myosin Type II/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Humans , Myosin Type II/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/genetics , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
10.
J Cell Sci ; 125(Pt 20): 4934-44, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22899719

ABSTRACT

Behavioral analyses of the deletion mutants of the four known myosin II heavy chain (Mhc) kinases of Dictyostelium discoideum revealed that all play a minor role in the efficiency of basic cell motility, but none play a role in chemotaxis in a spatial gradient of cAMP generated in vitro. However, the two kinases MhckA and MhckC were essential for chemotaxis in a spatial gradient of Ca(2+), shear-induced directed movement, and reorientation in the front of waves of cAMP during natural aggregation. The phenotypes of the mutants mhckA(-) and mhckC(-) were highly similar to that of the Ca(2+) channel/receptor mutant iplA(-) and the myosin II phosphorylation mutant 3XALA, which produces constitutively unphosphorylated myosin II. These results demonstrate that IplA, MhckA and MhckC play a selective role in chemotaxis in a spatial gradient of Ca(2+), but not cAMP, and suggest that Ca(2+) chemotaxis plays a role in the orientation of cells in the front of cAMP waves during natural aggregation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases , Calcium , Cell Movement , Dictyostelium , Protozoan Proteins , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Aggregation/genetics , Cell Movement/genetics , Cell Movement/physiology , Chemotaxis/genetics , Chemotaxis/physiology , Cyclic AMP/metabolism , Dictyostelium/genetics , Dictyostelium/metabolism , Dictyostelium/physiology , Humans , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Phosphorylation , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Pseudopodia/metabolism , Sequence Deletion
11.
BMC Cell Biol ; 12: 52, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-22136066

ABSTRACT

BACKGROUND: Phosphorylation of non-muscle myosin II regulatory light chain (RLC) at Thr18/Ser19 is well established as a key regulatory event that controls myosin II assembly and activation, both in vitro and in living cells. RLC can also be phosphorylated at Ser1/Ser2/Thr9 by protein kinase C (PKC). Biophysical studies show that phosphorylation at these sites leads to an increase in the Km of myosin light chain kinase (MLCK) for RLC, thereby indirectly inhibiting myosin II activity. Despite unequivocal evidence that PKC phosphorylation at Ser1/Ser2/Thr9 can regulate myosin II function in vitro, there is little evidence that this mechanism regulates myosin II function in live cells. RESULTS: The purpose of these studies was to investigate the role of Ser1/Ser2/Thr9 phosphorylation in live cells. To do this we utilized phospho-specific antibodies and created GFP-tagged RLC reporters with phosphomimetic aspartic acid substitutions or unphosphorylatable alanine substitutions at the putative inhibitory sites or the previously characterized activation sites. Cell lines stably expressing the RLC-GFP constructs were assayed for myosin recruitment during cell division, the ability to complete cell division, and myosin assembly levels under resting or spreading conditions. Our data shows that manipulation of the activation sites (Thr18/Ser19) significantly alters myosin II function in a number of these assays while manipulation of the putative inhibitory sites (Ser1/Ser2/Thr9) does not. CONCLUSIONS: These studies suggest that inhibitory phosphorylation of RLC is not a substantial regulatory mechanism, although we cannot rule out its role in other cellular processes or perhaps other types of cells or tissues in vivo.


Subject(s)
Myosin Light Chains/metabolism , Myosin Type II/metabolism , Serine/metabolism , Threonine/metabolism , Catalytic Domain , Cell Division , Cells, Cultured , HeLa Cells , Humans , Myosin Light Chains/chemistry , Myosin Type II/chemistry , Phosphorylation , Serine/chemistry , Threonine/chemistry
12.
Proc Natl Acad Sci U S A ; 108(44): 17991-6, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22025714

ABSTRACT

Despite functional significance of nonmuscle myosin II in cell migration and invasion, its role in epithelial-mesenchymal transition (EMT) or TGF-ß signaling is unknown. Analysis of normal mammary gland expression revealed that myosin IIC is expressed in luminal cells, whereas myosin IIB expression is up-regulated in myoepithelial cells that have more mesenchymal characteristics. Furthermore, TGF-ß induction of EMT in nontransformed murine mammary gland epithelial cells results in an isoform switch from myosin IIC to myosin IIB and increased phosphorylation of myosin heavy chain (MHC) IIA on target sites known to regulate filament dynamics (S1916, S1943). These expression and phosphorylation changes are downstream of heterogeneous nuclear ribonucleoprotein-E1 (E1), an effector of TGF-ß signaling. E1 knockdown drives cells into a migratory, invasive mesenchymal state and concomitantly up-regulates MHC IIB expression and MHC IIA phosphorylation. Abrogation of myosin IIB expression in the E1 knockdown cells has no effect on 2D migration but significantly reduced transmigration and macrophage-stimulated collagen invasion. These studies indicate that transition between myosin IIC/myosin IIB expression is a critical feature of EMT that contributes to increases in invasive behavior.


Subject(s)
Epithelial-Mesenchymal Transition , Myosin Type II/metabolism , Protein Isoforms/metabolism , Transforming Growth Factor beta/physiology , Animals , Cell Line , Mice , Phosphorylation
13.
Mol Biol Cell ; 22(13): 2270-81, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21562226

ABSTRACT

Abnormalities in the huntingtin protein (Htt) are associated with Huntington's disease. Despite its importance, the function of Htt is largely unknown. We show that Htt is required for normal chemotaxis and cytokinesis in Dictyostelium discoideum. Cells lacking Htt showed slower migration toward the chemoattractant cAMP and contained lower levels of cortical myosin II, which is likely due to defects in dephosphorylation of myosin II mediated by protein phosphatase 2A (PP2A). htt(-) cells also failed to maintain myosin II in the cortex of the cleavage furrow, generating unseparated daughter cells connected through a thin cytoplasmic bridge. Furthermore, similar to Dictyostelium htt(-) cells, siRNA-mediated knockdown of human HTT also decreased the PP2A activity in HeLa cells. Our data indicate that Htt regulates the phosphorylation status of myosin II during chemotaxis and cytokinesis through PP2A.


Subject(s)
Chemotaxis/physiology , Cytokinesis/physiology , Dictyostelium/cytology , Dictyostelium/metabolism , Microtubule-Associated Proteins/metabolism , Myosin Type II/metabolism , Cell Division/genetics , Cell Division/radiation effects , Cell Movement/genetics , Cell Movement/physiology , Cells, Cultured , Chemotaxis/genetics , Cyclic AMP/metabolism , Dictyostelium/genetics , HeLa Cells , Humans , Microtubule-Associated Proteins/genetics , Myosin Type II/genetics , Phosphorylation , Protein Phosphatase 2/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
14.
Eukaryot Cell ; 10(4): 604-10, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21357476

ABSTRACT

In Dictyostelium discoideum, myosin II resides predominantly in a soluble pool as the result of phosphorylation of the myosin heavy chain (MHC), and dephosphorylation of the MHC is required for myosin II filament assembly, recruitment to the cytoskeleton, and force production. Protein phosphatase type 2A (PP2A) was identified in earlier studies in Dictyostelium as a key biochemical activity that can drive MHC dephosphorylation. We report here gene targeting and cell biological studies addressing the roles of candidate PP2A B regulatory subunits (phr2aBα and phr2aBß) in myosin II assembly control in vivo. Dictyostelium phr2aBα- and phr2aBß-null cells show delayed development, reduction in the assembly of myosin II in cytoskeletal ghost assays, and defects in cytokinesis when grown in suspension compared to parental cell lines. These results demonstrate that the PP2A B subunits phr2aBα and phr2aBß contribute to myosin II assembly control in vivo, with phr2aBα having the predominant role facilitating MHC dephosphorylation to facilitate filament assembly.


Subject(s)
Dictyostelium/metabolism , Myosin Type II/metabolism , Protein Phosphatase 2/metabolism , Protein Subunits/metabolism , Protozoan Proteins/metabolism , Animals , Cell Movement/physiology , Cytokinesis/physiology , Dictyostelium/cytology , Dictyostelium/genetics , Gene Targeting , Myosin Type II/genetics , Phosphorylation , Protein Phosphatase 2/genetics , Protein Subunits/genetics , Protozoan Proteins/genetics
15.
J Mol Biol ; 407(5): 673-86, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21316371

ABSTRACT

Myosin II motor proteins play important roles in cell migration. Although myosin II filament assembly plays a key role in the stabilization of focal contacts at the leading edge of migrating cells, the mechanisms and signaling pathways regulating the localized assembly of lamellipodial myosin II filaments are poorly understood. We performed a proteomic analysis of myosin heavy chain (MHC) phosphorylation sites in MDA-MB 231 breast cancer cells to identify MHC phosphorylation sites that are activated during integrin engagement and lamellar extension on fibronectin. Fibronectin-activated MHC phosphorylation was identified on novel and previously recognized consensus sites for phosphorylation by protein kinase C and casein kinase II (CK-II). S1943, a CK-II consensus site, was highly phosphorylated in response to matrix engagement, and phosphoantibody staining revealed phosphorylation on myosin II assembled into leading-edge lamellae. Surprisingly, neither pharmacological reduction nor small inhibitory RNA reduction in CK-II activity reduced this stimulated S1943 phosphorylation. Our data demonstrate that S1943 phosphorylation is upregulated during lamellar protrusion, and that CK-II does not appear to be the kinase responsible for this matrix-induced phosphorylation event.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/physiology , Myosin Type II/metabolism , Proteome/analysis , Animals , COS Cells , Casein Kinase II/chemistry , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Female , Fibronectins/metabolism , HeLa Cells , Humans , Models, Molecular , Myosin Heavy Chains/chemistry , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosin Type II/chemistry , Myosin Type II/genetics , Phosphorylation , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pseudopodia/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
16.
Biomed Microdevices ; 12(3): 543-53, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20309736

ABSTRACT

To study the roles of nonmuscle myosin II (NM-II) during invasive cell migration, microfluidic migration chambers have been designed and fabricated using photo- and soft-lithography microfabrication techniques. The chamber consists of two channels separated by a vertical barrier with multiple bays of pores with widths varying from 6 microm to 16 microm, and lengths varying from 25 microm to 50 microm. The cells are plated in the channel on one side of the barrier while a chemoattractant is flowed through the channel on the other side of the barrier. In these chambers, cells can be observed with transmitted light or fluorescence optics while they chemotax through various sized pores that impose differential mechanical resistance to transmigration. As an initial test of this device, we compared breast-cancer cell chemotactic transmigration through different pore sizes with and without inhibition of NM-II. Two distinct rates were observed as cells attempted to pull their nucleus through the smaller pores, and the faster nuclear transit mode was critically dependent on NM-II motor activity. The ability to monitor cells as they chemotax through pores of different dimensions within a single experimental system provides novel information on how pore size affects cell morphology and migration rate, providing a dramatic improvement of imaging potential relative to other in vitro transmigration systems such as Boyden chambers.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Chemotaxis , Flow Cytometry/instrumentation , Microfluidic Analytical Techniques/instrumentation , Microscopy/instrumentation , Myosin Type II/metabolism , Cell Culture Techniques/instrumentation , Cell Line, Tumor , Cell Separation/instrumentation , Equipment Design , Equipment Failure Analysis , Flow Injection Analysis/instrumentation , Humans
17.
Traffic ; 10(12): 1773-84, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19843280

ABSTRACT

The contractile vacuole (CV) is a dynamic organelle that enables Dictyostelium amoeba and other protist to maintain osmotic homeostasis by expelling excess water. In the present study, we have uncovered a mechanism that coordinates the mechanics of the CV with myosin II, regulated by VwkA, an unconventional protein kinase that is conserved in an array of protozoa. Green fluorescent protein (GFP)-VwkA fusion proteins localize persistently to the CV during both filling and expulsion phases of water. In vwkA null cells, the established CV marker dajumin still localizes to the CV, but these structures are large, spherical and severely impaired for discharge. Furthermore, myosin II cortical localization and assembly are abnormal in vwkA null cells. Parallel analysis of wild-type cells treated with myosin II inhibitors or of myosin II null cells also results in enlarged CVs with impaired dynamics. We suggest that the myosin II cortical cytoskeleton, regulated by VwkA, serves a critical conserved role in the periodic contractions of the CV, as part of the osmotic protective mechanism of protozoa.


Subject(s)
Amoeba/metabolism , Myosin Type II/metabolism , Osmotic Pressure , Protein Kinases/metabolism , Amoeba/enzymology , Animals , Green Fluorescent Proteins/genetics , Myosin Type II/genetics , Protein Kinases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
18.
J Biol Chem ; 284(40): 27377-83, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19661065

ABSTRACT

During cell division, the mechanisms by which myosin II is recruited to the contractile ring are not fully understood. Much recent work has focused on a model in which spatially restricted de novo filament assembly occurs at the cell equator via localized myosin II regulatory light chain (RLC) phosphorylation, stimulated by the RhoA-activating centralspindlin complex. Here, we show that a recombinant myosin IIA protein that assembles constitutively and is incapable of binding RLC still displays strong localization to the furrow in mammalian cells. Furthermore, this RLC-deficient myosin II efficiently drives cytokinesis, demonstrating that centralspindlin-based RLC phosphorylation is not necessary for myosin II localization during furrowing. Myosin II truncation analysis further reveals two distinct myosin II tail properties that contribute to furrow localization: a central tail domain mediating cortical furrow binding to heterologous binding partners and a carboxyl-terminal region mediating co-assembly with existing furrow myosin IIA or IIB filaments.


Subject(s)
Cell Cycle Proteins/metabolism , Cytokinesis , Microtubule-Associated Proteins/metabolism , Myosin Type II/metabolism , Phosphoproteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Gene Deletion , HLA-D Antigens/genetics , HLA-D Antigens/metabolism , HeLa Cells , Humans , Myosin Light Chains/deficiency , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Myosin Type II/genetics , Phosphorylation , Protein Transport , Recombinant Fusion Proteins/metabolism
19.
Mol Biol Cell ; 20(1): 338-47, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18971378

ABSTRACT

To better understand the mechanism controlling nonmuscle myosin II (NM-II) assembly in mammalian cells, mutant NM-IIA constructs were created to allow tests in live cells of two widely studied models for filament assembly control. A GFP-NM-IIA construct lacking the RLC binding domain (DeltaIQ2) destabilizes the 10S sequestered monomer state and results in a severe defect in recycling monomers during spreading, and from the posterior to the leading edge during polarized migration. A GFP-NM-IIA construct lacking the nonhelical tailpiece (Deltatailpiece) is competent for leading edge assembly, but overassembles, suggesting defects in disassembly from lamellae subsequent to initial recruitment. The Deltatailpiece phenotype was recapitulated by a GFP-NM-IIA construct carrying a mutation in a mapped tailpiece phosphorylation site (S1943A), validating the importance of the tailpiece and tailpiece phosphorylation in normal lamellar myosin II assembly control. These results demonstrate that both the 6S/10S conformational change and the tailpiece contribute to the localization and assembly of myosin II in mammalian cells. This work furthermore offers cellular insights that help explain platelet and leukocyte defects associated with R1933-stop alleles of patients afflicted with human MYH9-related disorder.


Subject(s)
Cell Movement/physiology , Myosin Type II/metabolism , Recombinant Fusion Proteins/metabolism , Actins/metabolism , Amino Acid Sequence , Animals , Cytoskeleton/metabolism , Fluorescence Recovery After Photobleaching , HeLa Cells , Humans , Models, Molecular , Mutagenesis , Myosin Type II/chemistry , Myosin Type II/genetics , Protein Conformation , Recombinant Fusion Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...