Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(31): 15550-15559, 2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31235578

RESUMO

The ability of glioblastoma to disperse through the brain contributes to its lethality, and blocking this behavior has been an appealing therapeutic approach. Although a number of proinvasive signaling pathways are active in glioblastoma, many are redundant, so targeting one can be overcome by activating another. However, these pathways converge on nonredundant components of the cytoskeleton, and we have shown that inhibiting one of these-the myosin II family of cytoskeletal motors-blocks glioblastoma invasion even with simultaneous activation of multiple upstream promigratory pathways. Myosin IIA and IIB are the most prevalent isoforms of myosin II in glioblastoma, and we now show that codeleting these myosins markedly impairs tumorigenesis and significantly prolongs survival in a rodent model of this disease. However, while targeting just myosin IIA also impairs tumor invasion, it surprisingly increases tumor proliferation in a manner that depends on environmental mechanics. On soft surfaces myosin IIA deletion enhances ERK1/2 activity, while on stiff surfaces it enhances the activity of NFκB, not only in glioblastoma but in triple-negative breast carcinoma and normal keratinocytes as well. We conclude myosin IIA suppresses tumorigenesis in at least two ways that are modulated by the mechanics of the tumor and its stroma. Our results also suggest that inhibiting tumor invasion can enhance tumor proliferation and that effective therapy requires targeting cellular components that drive both proliferation and invasion simultaneously.


Assuntos
Carcinogênese/metabolismo , Citoesqueleto/metabolismo , Glioblastoma/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Neoplasias/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Animais , Carcinogênese/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Citoesqueleto/genética , Citoesqueleto/patologia , Glioblastoma/genética , Glioblastoma/patologia , Camundongos , Proteínas de Neoplasias/genética , Miosina não Muscular Tipo IIA/genética
2.
J Biol Chem ; 292(8): 3099-3111, 2017 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-28053086

RESUMO

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.


Assuntos
Adesão Celular , Movimento Celular , Cadeias Pesadas de Miosina/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Animais , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Humanos , Camundongos , Cadeias Pesadas de Miosina/análise , Miosina não Muscular Tipo IIA/análise , Fosforilação
3.
J Biol Chem ; 291(12): 6083-95, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26763235

RESUMO

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.


Assuntos
Fibroblastos/fisiologia , Miosina Tipo II/fisiologia , Fibrose Pulmonar/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Movimento Celular , Polaridade Celular , Forma Celular , Matriz Extracelular/fisiologia , Feminino , Humanos , Pulmão/metabolismo , Pulmão/patologia , Camundongos Endogâmicos C57BL , Fenótipo , Fibrose Pulmonar/patologia
4.
Am J Pathol ; 186(5): 1351-60, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26988652

RESUMO

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.


Assuntos
Complexos Multiproteicos/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Neoplasias da Bexiga Urinária/patologia , Benzamidas/farmacologia , Caderinas/metabolismo , Movimento Celular/fisiologia , Dioxóis/farmacologia , Humanos , Alvo Mecanístico do Complexo 2 de Rapamicina , Invasividade Neoplásica , Fosforilação/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Transdução de Sinais/fisiologia , Proteína Smad2/metabolismo , Proteína Smad4/metabolismo , Células Tumorais Cultivadas , Regulação para Cima/fisiologia , Neoplasias da Bexiga Urinária/fisiopatologia , Vimentina/metabolismo
5.
Stem Cells ; 33(7): 2114-2125, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25827713

RESUMO

Advanced cancers display cellular heterogeneity driven by self-renewing, tumorigenic cancer stem cells (CSCs). The use of cell lines to model CSCs is challenging due to the difficulty of identifying and isolating cell populations that possess differences in self-renewal and tumor initiation. To overcome these barriers in triple-negative breast cancer (TNBC), we developed a CSC system using a green fluorescent protein (GFP) reporter for the promoter of the well-established pluripotency gene NANOG. NANOG-GFP+ cells gave rise to both GFP+ and GFP(-) cells, and GFP+ cells possessed increased levels of the embryonic stem cell transcription factors NANOG, SOX2, and OCT4 and elevated self-renewal and tumor initiation capacities. GFP+ cells also expressed mesenchymal markers and demonstrated increased invasion. Compared with the well-established CSC markers CD24(-) /CD44(+) , CD49f, and aldehyde dehydrogenase (ALDH) activity, our NANOG-GFP reporter system demonstrated increased enrichment for CSCs. To explore the utility of this system as a screening platform, we performed a flow cytometry screen that confirmed increased CSC marker expression in the GFP+ population and identified new cell surface markers elevated in TNBC CSCs, including junctional adhesion molecule-A (JAM-A). JAM-A was highly expressed in GFP+ cells and patient-derived xenograft ALDH+ CSCs compared with the GFP(-) and ALDH(-) cells, respectively. Depletion of JAM-A compromised self-renewal, whereas JAM-A overexpression induced self-renewal in GFP(-) cells. Our data indicate that we have defined and developed a robust system to monitor differences between CSCs and non-CSCs in TNBC that can be used to identify CSC-specific targets for the development of future therapeutic strategies.


Assuntos
Genes Reporter/genética , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
6.
Dev Biol ; 382(1): 136-48, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23911870

RESUMO

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.


Assuntos
Ectoderma/embriologia , Perda do Embrião/patologia , Integrases/metabolismo , Queratina-5/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Trofoblastos/metabolismo , Trofoblastos/patologia , Alelos , Animais , Animais Recém-Nascidos , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Bovinos , Linhagem da Célula , Proliferação de Células , Cruzamentos Genéticos , Ectoderma/metabolismo , Ectoderma/patologia , Perda do Embrião/metabolismo , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Desenvolvimento Embrionário , Epiderme/embriologia , Epiderme/metabolismo , Epiderme/patologia , Feminino , Deleção de Genes , Genótipo , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos Knockout , Gravidez , Saco Vitelino/metabolismo
7.
J Cell Sci ; 125(Pt 20): 4934-44, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22899719

RESUMO

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.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina , Cálcio , Movimento Celular , Dictyostelium , Proteínas de Protozoários , Cálcio/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Agregação Celular/genética , Movimento Celular/genética , Movimento Celular/fisiologia , Quimiotaxia/genética , Quimiotaxia/fisiologia , AMP Cíclico/metabolismo , Dictyostelium/genética , Dictyostelium/metabolismo , Dictyostelium/fisiologia , Humanos , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Fosforilação , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Pseudópodes/metabolismo , Deleção de Sequência
8.
Proc Natl Acad Sci U S A ; 108(44): 17991-6, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22025714

RESUMO

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.


Assuntos
Transição Epitelial-Mesenquimal , Miosina Tipo II/metabolismo , Isoformas de Proteínas/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Linhagem Celular , Camundongos , Fosforilação
9.
Eukaryot Cell ; 10(4): 604-10, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21357476

RESUMO

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.


Assuntos
Dictyostelium/metabolismo , Miosina Tipo II/metabolismo , Proteína Fosfatase 2/metabolismo , Subunidades Proteicas/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Movimento Celular/fisiologia , Citocinese/fisiologia , Dictyostelium/citologia , Dictyostelium/genética , Marcação de Genes , Miosina Tipo II/genética , Fosforilação , Proteína Fosfatase 2/genética , Subunidades Proteicas/genética , Proteínas de Protozoários/genética
10.
Traffic ; 10(12): 1773-84, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19843280

RESUMO

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.


Assuntos
Amoeba/metabolismo , Miosina Tipo II/metabolismo , Pressão Osmótica , Proteínas Quinases/metabolismo , Amoeba/enzimologia , Animais , Proteínas de Fluorescência Verde/genética , Miosina Tipo II/genética , Proteínas Quinases/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
11.
BMC Cell Biol ; 12: 52, 2011 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-22136066

RESUMO

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.


Assuntos
Cadeias Leves de Miosina/metabolismo , Miosina Tipo II/metabolismo , Serina/metabolismo , Treonina/metabolismo , Domínio Catalítico , Divisão Celular , Células Cultivadas , Células HeLa , Humanos , Cadeias Leves de Miosina/química , Miosina Tipo II/química , Fosforilação , Serina/química , Treonina/química
12.
Exp Cell Res ; 316(6): 980-91, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20132815

RESUMO

Wound healing in the skin is an important and complex process that involves 3-dimensional tissue reorganization, including matrix and chemokine-triggered cell migration, paracrine signaling, and matrix remodeling. The molecular signals and underlying mechanisms that stimulate myosin II activity during skin wound healing have not been elucidated. To begin understanding the signaling pathways involved in the activation of myosin II in this process, we have evaluated myosin II activation in migrating primary human keratinocytes in response to scratch wounding in vitro. We report here that myosin II activation and recruitment to the cytoskeleton in wounded keratinocytes are biphasic. Post-wounding, a rapid phosphorylation of myosin II regulatory light chain (RLC) occurs with resultant translocation of myosin IIA to the cell cortex, far in advance of the later polarization and cell migration. During this acute-phase of myosin II activation, pharmacological approaches reveal p38-MAP kinase and cytosolic calcium as having critical roles in the phosphorylation driving cytoskeletal assembly. Although p38-MAPK has known roles in keratinocyte migration, and known roles in leading-edge focal complex dynamics, to our knowledge this is the first report of p38-MAPK acting as an upstream activator of myosin II phosphorylation and assembly during any type of wound response.


Assuntos
Epiderme/patologia , Epiderme/fisiologia , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Cicatrização/fisiologia , Movimento Celular/fisiologia , Inibidores Enzimáticos/metabolismo , Células Epidérmicas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Cadeias Leves de Miosina/metabolismo , Fosforilação , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
J Biol Chem ; 284(40): 27377-83, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19661065

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Citocinese , Proteínas Associadas aos Microtúbulos/metabolismo , Miosina Tipo II/metabolismo , Fosfoproteínas/metabolismo , Animais , Células COS , Chlorocebus aethiops , Deleção de Genes , Antígenos HLA-D/genética , Antígenos HLA-D/metabolismo , Células HeLa , Humanos , Cadeias Leves de Miosina/deficiência , Cadeias Leves de Miosina/genética , Cadeias Leves de Miosina/metabolismo , Miosina Tipo II/genética , Fosforilação , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo
14.
Biomed Microdevices ; 12(3): 543-53, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20309736

RESUMO

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.


Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Quimiotaxia , Citometria de Fluxo/instrumentação , Técnicas Analíticas Microfluídicas/instrumentação , Microscopia/instrumentação , Miosina Tipo II/metabolismo , Técnicas de Cultura de Células/instrumentação , Linhagem Celular Tumoral , Separação Celular/instrumentação , Desenho de Equipamento , Análise de Falha de Equipamento , Análise de Injeção de Fluxo/instrumentação , Humanos
15.
Mol Biol Cell ; 16(5): 2248-62, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15728726

RESUMO

We have identified a new protein kinase in Dictyostelium discoideum that carries the same conserved class of "alpha-kinase" catalytic domain as reported previously in myosin heavy chain kinases (MHCKs) in this amoeba but that has a completely novel domain organization. The protein contains an N-terminal von Willebrand factor A (vWFA)-like motif and is therefore named VwkA. Manipulation of VwkA expression level via high copy number plasmids (VwkA++ cells) or gene disruption (vwkA null cells) results in an array of cellular defects, including impaired growth and multinucleation in suspension culture, impaired development, and alterations in myosin II abundance and assembly. Despite sequence similarity to MHCKs, the purified protein failed to phosphorylate myosin II in vitro. Autophosphorylation activity, however, was enhanced by calcium/calmodulin, and the enzyme can be precipitated from cellular lysates with calmodulin-agarose, suggesting that VwkA may directly bind calmodulin. VwkA is cytosolic in distribution but enriched on the membranes of the contractile vacuole and Golgi-like structures in the cell. We propose that VwkA likely acts indirectly to influence myosin II abundance and assembly behavior and possibly has broader roles than previously characterized alpha kinases in this organism, which all seem to be MHCKs.


Assuntos
Dictyostelium/enzimologia , Proteínas Quinases/química , Proteínas Quinases/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência de Bases , Calmodulina/metabolismo , Clonagem Molecular , DNA Complementar/genética , DNA de Protozoário/genética , Dictyostelium/genética , Dictyostelium/crescimento & desenvolvimento , Expressão Gênica , Genes de Protozoários , Complexo de Golgi/enzimologia , Dados de Sequência Molecular , Miosina Tipo II/metabolismo , Proteínas Quinases/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Vacúolos/enzimologia , Fator de von Willebrand/química , Fator de von Willebrand/genética
16.
Mol Biol Cell ; 16(9): 4256-66, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15987738

RESUMO

Myosin II filament assembly in Dictyostelium discoideum is regulated via phosphorylation of residues located in the carboxyl-terminal portion of the myosin II heavy chain (MHC) tail. A series of novel protein kinases in this system are capable of phosphorylating these residues in vitro, driving filament disassembly. Previous studies have demonstrated that at least three of these kinases (MHCK A, MHCK B, and MHCK C) display differential localization patterns in living cells. We have created a collection of single, double, and triple gene knockout cell lines for this family of kinases. Analysis of these lines reveals that three MHC kinases appear to represent the majority of cellular activity capable of driving myosin II filament disassembly, and reveals that cytokinesis defects increase with the number of kinases disrupted. Using biochemical fractionation of cytoskeletons and in vivo measurements via fluorescence recovery after photobleaching (FRAP), we find that myosin II overassembly increases incrementally in the mutants, with the MHCK A(-)/B(-)/C(-) triple mutant showing severe myosin II overassembly. These studies suggest that the full complement of MHC kinases that significantly contribute to growth phase and cytokinesis myosin II disassembly in this organism has now been identified.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Citocinese/fisiologia , Citoesqueleto/enzimologia , Dictyostelium/enzimologia , Miosina Tipo II/fisiologia , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Linhagem Celular , Dictyostelium/crescimento & desenvolvimento , Genes Reporter , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas de Protozoários , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
17.
Cancer Res ; 66(9): 4725-33, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16651425

RESUMO

Initial stages of tumor cell metastasis involve an epithelial-mesenchyme transition that involves activation of amoeboid migration and loss of cell-cell adhesion. The actomyosin cytoskeleton has fundamental but poorly understood roles in these events. Myosin II, an abundant force-producing protein, has roles in cell body translocation and retraction of the posterior of the cell during migration. Recent studies have suggested that this protein may also have roles in leading edge protrusive events. The metastasis-promoting protein metastasin-1, a regulator of myosin II assembly, colocalizes with myosin IIA at the leading edge of cancer cells, suggesting direct roles for myosin II in metastatic behavior. We have assessed the roles of specific myosin II isoforms during lamellar spreading of MDA-MB-231 breast cancer cells on extracellular matrix. We find that the two major myosin II isoforms IIA and IIB are both expressed in these cells, and both are recruited dramatically to the lamellar margin during active spreading on fibronectin. There is also a transient increase in regulatory light chain phosphorylation that correlates the recruitment of myosin IIA and myosin IIB into this spreading margin. Pharmacologic inhibition of myosin II or myosin light chain kinase dramatically reduced spreading. Depletion of myosin IIA via small interfering RNA impaired migration but enhanced lamellar spreading, whereas depletion of myosin IIB impaired not only migration but also impaired initial rates of lamellar spreading. These results indicate that both isoforms are critical for the mechanics of cell migration, with myosin IIB seeming to have a preferential role in the mechanics of lamellar protrusion.


Assuntos
Neoplasias da Mama/patologia , Movimento Celular/fisiologia , Miosina não Muscular Tipo IIA/fisiologia , Miosina não Muscular Tipo IIB/fisiologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Fibronectinas , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Quinase de Cadeia Leve de Miosina/metabolismo , Miosina não Muscular Tipo IIA/antagonistas & inibidores , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/antagonistas & inibidores , Miosina não Muscular Tipo IIB/genética , Miosina não Muscular Tipo IIB/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/genética , Transfecção , Quinases Associadas a rho
18.
Tissue Eng Part A ; 24(15-16): 1218-1227, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29397789

RESUMO

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.


Assuntos
Cartilagem/enzimologia , Condrogênese , Matriz Extracelular/metabolismo , Células-Tronco Mesenquimais/enzimologia , Transdução de Sinais , Quinases Associadas a rho/antagonistas & inibidores , Transporte Biológico Ativo , Cartilagem/citologia , Células Cultivadas , Citoesqueleto/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual , Quinases Associadas a rho/metabolismo
19.
Oncotarget ; 7(30): 47586-47592, 2016 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-27285763

RESUMO

Despite many advances in the treatment of breast cancer, it remains one of the leading causes of death among women. One hurdle for effective therapy is the treatment of the highly invasive and tumorigenic subpopulation of tumors called cancer stem cells (CSCs). CSCs, when stimulated with EGF, migrate through a physiological 3D collagen matrix at a higher velocity than non-stem cancer cells (non-SCCs). This increased invasion is due, in part, by an enhanced nuclear translocation ability of CSCs. We observed no difference between CSC and non-SCC in cellular migration rates on a 2D surface. Furthermore, during transwell migration using large diameter transwell pores, both CSC and non-SCC populations migrated with similar efficiency. However, when challenged with more restrictive transwells, CSCs were dramatically more capable of transwell migration. These results implicate nuclear translocation as a major rate limiting factor for CSC dissemination. We further show that non-muscle myosin IIB is critical for this enhanced nuclear translocation and the ability for cancer stem cells to efficiently migrate through restrictive 3D environments. These studies suggest that cytoskeletal elements upregulated in CSCs, such as myosin IIB, may be valuable targets for intervention in cancer stem cell dispersal from tumors.


Assuntos
Núcleo Celular/metabolismo , Células-Tronco Neoplásicas/patologia , Miosina não Muscular Tipo IIB/fisiologia , Transporte Ativo do Núcleo Celular , Linhagem Celular Tumoral , Movimento Celular , Humanos , Invasividade Neoplásica
20.
J Cell Biol ; 210(4): 583-94, 2015 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-26261182

RESUMO

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.


Assuntos
Movimento Celular , Núcleo Celular/fisiologia , Miosina não Muscular Tipo IIB/fisiologia , Transporte Ativo do Núcleo Celular , Animais , Fenômenos Biomecânicos , Linhagem Celular Tumoral , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA