Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Biochem Biophys Res Commun ; 527(3): 716-722, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32423824

RESUMO

p27Kip1, a member of the Cip/Kip family of cyclin-dependent kinase (CDK) inhibitors, is now known as a multifunctional protein that plays crucial roles in cell architecture and migration by regulating rearrangements of the actin cytoskeleton and microtubules. The intracellular level of p27Kip1 is increased by anti-proliferative stimuli, such as mitogen deprivation and contact inhibition, which also induce formation of primary cilia, microtubule-based membranous organelles that protrude from the cell surface. However, it remains unknown whether p27Kip1 is associated with ciliogenesis. Here, we have generated p27Kip1-knockout hTERT-immortalized human retinal pigment epithelial cells, and found that ciliogenesis is almost completely disrupted in p27Kip1-knockout cells. The defect of ciliogenesis is rescued by the exogenous expression of wild-type p27Kip1 and, surprisingly, its 86-140 amino acid region, which is neither responsible for CDK inhibition nor remodeling of the actin cytoskeleton and microtubules. Moreover, transmission electron microscopy and immunofluorescence analyses reveal that p27Kip1 abrogation impairs one of the earliest events of ciliogenesis, docking of the Ehd1-associated preciliary vesicles to the distal appendages of the basal body. Our findings identify a novel CDK-independent function of p27Kip1 in primary cilia formation.


Assuntos
Cílios/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Epitélio Pigmentado da Retina/citologia , Linhagem Celular , Cílios/ultraestrutura , Inibidor de Quinase Dependente de Ciclina p27/genética , Técnicas de Inativação de Genes , Humanos , Epitélio Pigmentado da Retina/metabolismo
2.
Int J Mol Sci ; 21(17)2020 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-32825105

RESUMO

Cilia are antenna-like structures present in many vertebrate cells. These organelles detect extracellular cues, transduce signals into the cell, and play an essential role in ensuring correct cell proliferation, migration, and differentiation in a spatiotemporal manner. Not surprisingly, dysregulation of cilia can cause various diseases, including cancer and ciliopathies, which are complex disorders caused by mutations in genes regulating ciliary function. The structure and function of cilia are dynamically regulated through various mechanisms, among which E3 ubiquitin ligases and deubiquitinases play crucial roles. These enzymes regulate the degradation and stabilization of ciliary proteins through the ubiquitin-proteasome system. In this review, we briefly highlight the role of cilia in ciliopathy and cancer; describe the roles of E3 ubiquitin ligases and deubiquitinases in ciliogenesis, ciliopathy, and cancer; and highlight some of the E3 ubiquitin ligases and deubiquitinases that are potential therapeutic targets for these disorders.


Assuntos
Ciliopatias/tratamento farmacológico , Enzimas Desubiquitinantes/metabolismo , Neoplasias/tratamento farmacológico , Ubiquitina-Proteína Ligases/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Ciliopatias/metabolismo , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Neoplasias/metabolismo , Ubiquitinação/efeitos dos fármacos
3.
Proc Jpn Acad Ser B Phys Biol Sci ; 95(8): 479-493, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31611503

RESUMO

Intermediate filaments (IFs), in coordination with microfilaments and microtubules, form the structural framework of the cytoskeleton and nucleus, thereby providing mechanical support against cellular stresses and anchoring intracellular organelles in place. The assembly and disassembly of IFs are mainly regulated by the phosphorylation of IF proteins. These phosphorylation states can be tracked using antibodies raised against phosphopeptides in the target proteins. IFs exert their functions through interactions with not only structural proteins, but also non-structural proteins involved in cell signaling, such as stress responses, apoptosis, and cell proliferation. This review highlights findings related to how IFs regulate cell division through phosphorylation cascades and how trichoplein, a centriolar protein originally identified as a keratin-associated protein, regulates the cell cycle through primary cilium formation.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Filamentos Intermediários/metabolismo , Animais , Proliferação de Células , Homeostase , Humanos , Fosforilação , Processamento de Proteína Pós-Traducional
4.
Cancer Sci ; 109(9): 2632-2640, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29949679

RESUMO

Tetraploidy, a condition in which a cell has four homologous sets of chromosomes, is often seen as a natural physiological condition but is also frequently seen in pathophysiological conditions such as cancer. Tetraploidy facilitates chromosomal instability (CIN), which is an elevated level of chromosomal loss and gain that can cause production of a wide variety of aneuploid cells that carry structural and numerical aberrations of chromosomes. The resultant genomic heterogeneity supposedly expedites karyotypic evolution that confers oncogenic potential in spite of the reduced cellular fitness caused by aneuploidy. Recent studies suggest that tetraploidy might also be associated with aging; mice with mutations in an intermediate filament protein have revealed that these tetraploidy-prone mice exhibit tissue disorders associated with aging. Cellular senescence and its accompanying senescence-associated secretory phenotype have now emerged as critical factors that link tetraploidy and tetraploidy-induced CIN with cancer, and possibly with aging. Here, we review recent findings about how tetraploidy is related to cancer and possibly to aging, and discuss underlying mechanisms of the relationship, as well as how we can exploit the properties of cells exhibiting tetraploidy-induced CIN to control these pathological conditions.


Assuntos
Envelhecimento/genética , Senescência Celular/genética , Instabilidade Cromossômica/genética , Neoplasias/genética , Tetraploidia , Animais , Humanos , Camundongos
5.
Biochem Biophys Res Commun ; 498(3): 544-550, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29518391

RESUMO

We previously reported that vimentin, GFAP, and desmin (type III intermediate filament [IF] proteins) are mitotically phosphorylated by CDK1, Aurora-B, and Rho-kinase. This phosphorylation is critical for efficient separation of these IFs and completion of cytokinesis. Keratin 5 (K5) and K14 form a heterodimer, which constitutes IF network in basal layer cells of stratified squamous epithelia. Here, we report that the solubility of K5/K14 increased in mitosis. The in vitro assays revealed that three mitotic kinases phosphorylate K5 more than K14. We then identified Thr23/Thr144, Ser30, and Thr159 on murine K5 as major phosphorylation sites for CDK1, Aurora-B, and Rho-kinase, respectively. Using site- and phosphorylation-state-specific antibodies, we demonstrated that K5-Thr23 was phosphorylated in entire cytoplasm from prometaphase to metaphase, whereas K5-Ser30 phosphorylation occurred specifically at the cleavage furrow from anaphase to telophase. Efficient K5/K14-IF separation was impaired by K5 mutations at the sites phosphorylated by these mitotic kinases. K5-Thr23 phosphorylation was widely detected in dividing K5-positive cells of murine individuals. These results suggested that mitotic reorganization of K5/K14-IF network is governed largely through K5 phosphorylation by CDK1, Aurora-B, and Rho-kinase.


Assuntos
Aurora Quinase B/metabolismo , Proteína Quinase CDC2/metabolismo , Filamentos Intermediários/metabolismo , Queratina-14/metabolismo , Queratina-15/metabolismo , Quinases Associadas a rho/metabolismo , Animais , Linhagem Celular , Células HeLa , Humanos , Camundongos Endogâmicos C57BL , Mitose , Fosforilação
6.
Biochem Biophys Res Commun ; 478(3): 1323-9, 2016 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-27565725

RESUMO

Desmin is a type III intermediate filament (IF) component protein expressed specifically in muscular cells. Desmin is phosphorylated by Aurora-B and Rho-kinase specifically at the cleavage furrow from anaphase to telophase. The disturbance of this phosphorylation results in the formation of unusual long bridge-like IF structures (IF-bridge) between two post-mitotic (daughter) cells. Here, we report that desmin also serves as an excellent substrate for the other type of mitotic kinase, Cdk1. Desmin phosphorylation by Cdk1 loses its ability to form IFs in vitro. We have identified Ser6, Ser27, and Ser31 on murine desmin as phosphorylation sites for Cdk1. Using a site- and phosphorylation-state-specific antibody for Ser31 on desmin, we have demonstrated that Cdk1 phosphorylates desmin in entire cytoplasm from prometaphase to metaphase. Desmin mutations at Cdk1 sites exhibit IF-bridge phenotype, the frequency of which is significantly increased by the addition of Aurora-B and Rho-kinase site mutations to Cdk1 site mutations. In addition, Cdk1-induced desmin phosphorylation is detected in mitotic muscular cells of murine embryonic/newborn muscles and human rhabdomyosarcoma specimens. Therefore, Cdk1-induced desmin phosphorylation is required for efficient separation of desmin-IFs and generally detected in muscular mitotic cells in vivo.


Assuntos
Proteína Quinase CDC2/metabolismo , Desmina/metabolismo , Filamentos Intermediários/metabolismo , Mitose , Músculo Esquelético/embriologia , Músculo Esquelético/metabolismo , Rabdomiossarcoma/metabolismo , Animais , Animais Recém-Nascidos , Humanos , Camundongos , Proteínas Mutantes/metabolismo , Fosforilação , Fosfosserina/metabolismo , Rabdomiossarcoma/patologia
7.
Cell Struct Funct ; 40(1): 43-50, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25748360

RESUMO

Checkpoint kinase 1 (Chk1) is a conserved protein kinase central to the cell-cycle checkpoint during DNA damage response (DDR). Until recently, ATR, a protein kinase activated in response to DNA damage or stalled replication, has been considered as the sole regulator of Chk1. Recent progress, however, has led to the identification of additional protein kinases involved in Chk1 phosphorylation, affecting the subcellular localization and binding partners of Chk1. In fact, spatio-temporal regulation of Chk1 is of critical importance not only in the DDR but also in normal cell-cycle progression. In due course, many potent inhibitors targeted to Chk1 have been developed as anticancer agents and some of these inhibitors are currently in clinical trials. In this review, we summarize the current knowledge of Chk1 regulation by phosphorylation.


Assuntos
Proteínas Quinases/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Quinase 1 do Ponto de Checagem , Quinases Ciclina-Dependentes/metabolismo , Humanos , Fosforilação , Proteínas Quinases/química , Serina/metabolismo
8.
EMBO J ; 29(16): 2802-12, 2010 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-20639859

RESUMO

14-3-3 proteins control various cellular processes, including cell cycle progression and DNA damage checkpoint. At the DNA damage checkpoint, some subtypes of 14-3-3 (beta and zeta isoforms in mammalian cells and Rad24 in fission yeast) bind to Ser345-phosphorylated Chk1 and promote its nuclear retention. Here, we report that 14-3-3gamma forms a complex with Chk1 phosphorylated at Ser296, but not at ATR sites (Ser317 and Ser345). Ser296 phosphorylation is catalysed by Chk1 itself after Chk1 phosphorylation by ATR, and then ATR sites are rapidly dephosphorylated on Ser296-phosphorylated Chk1. Although Ser345 phosphorylation is observed at nuclear DNA damage foci, it occurs more diffusely in the nucleus. The replacement of endogenous Chk1 with Chk1 mutated at Ser296 to Ala induces premature mitotic entry after ultraviolet irradiation, suggesting the importance of Ser296 phosphorylation in the DNA damage response. Although Ser296 phosphorylation induces the only marginal change in Chk1 catalytic activity, 14-3-3gamma mediates the interaction between Chk1 and Cdc25A. This ternary complex formation has an essential function in Cdc25A phosphorylation and degradation to block premature mitotic entry after DNA damage.


Assuntos
Proteínas 14-3-3/metabolismo , Dano ao DNA , Mitose , Proteínas Quinases/metabolismo , Fosfatases cdc25/metabolismo , Ciclo Celular , Núcleo Celular/metabolismo , Quinase 1 do Ponto de Checagem , Células HeLa , Humanos , Fosforilação , Ligação Proteica , Serina/metabolismo , Proteínas Contendo Repetições de beta-Transducina/metabolismo
9.
J Cell Sci ; 124(Pt 13): 2113-9, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21628425

RESUMO

Chk1 inhibits the premature activation of the cyclin-B1-Cdk1. However, it remains controversial whether Chk1 inhibits Cdk1 in the centrosome or in the nucleus before the G2-M transition. In this study, we examined the specificity of the mouse monoclonal anti-Chk1 antibody DCS-310, with which the centrosome was stained. Conditional Chk1 knockout in mouse embryonic fibroblasts reduced nuclear but not centrosomal staining with DCS-310. In Chk1(+/myc) human colon adenocarcinoma (DLD-1) cells, Chk1 was detected in the nucleus but not in the centrosome using an anti-Myc antibody. Through the combination of protein array and RNAi technologies, we identified Ccdc-151 as a protein that crossreacted with DCS-310 on the centrosome. Mitotic entry was delayed by expression of the Chk1 mutant that localized in the nucleus, although forced immobilization of Chk1 to the centrosome had little impact on the timing of mitotic entry. These results suggest that nuclear but not centrosomal Chk1 contributes to correct timing of mitotic entry.


Assuntos
Núcleo Celular/metabolismo , Mitose , Proteínas Quinases/biossíntese , Animais , Proteína Quinase CDC2/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Centrossomo/metabolismo , Quinase 1 do Ponto de Checagem , Ciclina B1/genética , Ciclina B1/metabolismo , Humanos , Camundongos , Camundongos Knockout , Mutação , Proteínas Quinases/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Interferência de RNA , RNA Interferente Pequeno
10.
Adv Sci (Weinh) ; : e2202632, 2022 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-36373718

RESUMO

Following injury, skeletal muscle regenerates but fatty tissue accumulation is seen in aged muscle or muscular dystrophies. Fibro/adipogenic progenitors (FAPs) are key players in these events; however, the effect of primary cilia on FAPs remains unclear. Here, it is reported that genetic ablation of trichoplein (TCHP), a ciliary regulator, induces ciliary elongation on FAPs after injury, which promotes muscle regeneration while inhibiting adipogenesis. The defective adipogenic differentiation of FAPs is attributed to dysfunction of cilia-dependent lipid raft dynamics, which is critical for insulin/Akt signaling. It is also found that interleukin (IL) 13 is substantially produced by intramuscular FAPs, which are upregulated by ciliary elongation and contribute to regeneration. Mechanistically, upon injury, long cilia excessively activate the IL33/ST2/JNK axis to enhance IL13 production, facilitating myoblast proliferation and M2 macrophage polarization. The results indicate that FAPs organize the regenerative responses to skeletal muscle injury via cilia-mediated insulin/Akt and ST2/JNK signaling pathways.

11.
J Biol Chem ; 285(10): 7657-69, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20048155

RESUMO

Gliosis is a biological process that occurs during injury repair in the central nervous system and is characterized by the overexpression of the intermediate filaments (IFs) glial fibrillary acidic protein (GFAP) and vimentin. A common thread in many retinal diseases is reactive Müller cell gliosis, an untreatable condition that leads to tissue scarring and even blindness. Here, we demonstrate that the vimentin-targeting small molecule withaferin A (WFA) is a novel chemical probe of GFAP. Using molecular modeling studies that build on the x-ray crystal structure of tetrameric vimentin rod 2B domain we reveal that the WFA binding site is conserved in the corresponding domain of tetrameric GFAP. Consequently, we demonstrate that WFA covalently binds soluble recombinant tetrameric human GFAP at cysteine 294. In cultured primary astrocytes, WFA binds to and down-regulates soluble vimentin and GFAP expression to cause cell cycle G(0)/G(1) arrest. Exploiting a chemical injury model that overexpresses vimentin and GFAP in retinal Müller glia, we demonstrate that systemic delivery of WFA down-regulates soluble vimentin and GFAP expression in mouse retinas. This pharmacological knockdown of soluble IFs results in the impairment of GFAP filament assembly and inhibition of cell proliferative response in Müller glia. We further show that a more severe GFAP filament assembly deficit manifests in vimentin-deficient mice, which is partly rescued by WFA. These findings illustrate WFA as a chemical probe of type III IFs and illuminate this class of withanolide as a potential treatment for diverse gliosis-dependent central nervous system traumatic injury conditions and diseases, and for orphan IF-dependent pathologies.


Assuntos
Ergosterol/análogos & derivados , Proteína Glial Fibrilar Ácida/metabolismo , Gliose , Retina , Degeneração Retiniana , Vimentina/metabolismo , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Ciclina D3/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Ergosterol/química , Ergosterol/metabolismo , Ergosterol/farmacologia , Proteína Glial Fibrilar Ácida/genética , Gliose/metabolismo , Gliose/patologia , Humanos , Camundongos , Camundongos Knockout , Modelos Moleculares , Estrutura Secundária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Retina/efeitos dos fármacos , Retina/metabolismo , Retina/patologia , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Vimentina/química , Vimentina/genética , Vitanolídeos
12.
Trends Cell Biol ; 31(12): 954-964, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34420822

RESUMO

Primary cilia are solitary, microtubule-based structures emanating from the surface of most vertebrate cells. Although it is understood that ciliary assembly and disassembly both depend upon and impact cell cycle progression, critical mechanistic details of these links remain unresolved. Accumulating evidence shows that the signaling pathways downstream of receptor tyrosine kinases and lysophosphatidic acid receptors control the dynamics of primary cilia. It has also become clear that primary cilia not only serve as signaling hubs but also regulate the composition of the surrounding membrane, which is likely to affect the response to growth factors. Here, we overview recent advances in understanding the interplay between primary cilia and the cell cycle, with a focus on growth factor signaling pathways.


Assuntos
Cílios , Transdução de Sinais , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Divisão Celular , Cílios/metabolismo , Humanos , Transdução de Sinais/fisiologia
13.
Cell Rep ; 34(10): 108817, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33691104

RESUMO

Primary cilia play a pivotal role in signal transduction and development and are known to serve as signaling hubs. Recent studies have shown that primary cilium dysfunction influences adipogenesis, but the mechanisms are unclear. Here, we show that mesenchymal progenitors C3H10T1/2 depleted of trichoplein, a key regulator of cilium formation, have significantly longer cilia than control cells and fail to differentiate into adipocytes. Mechanistically, the elongated cilia prevent caveolin-1- and/or GM3-positive lipid rafts from being assembled around the ciliary base where insulin receptor proteins accumulate, thereby inhibiting the insulin-Akt signaling. We further generate trichoplein knockout mice, in which adipogenic progenitors display elongated cilia and impair the lipid raft dynamics. The knockout mice on an extended high-fat diet exhibit reduced body fat and smaller adipocytes than wild-type (WT) mice. Overall, our results suggest a role for primary cilia in regulating adipogenic signal transduction via control of the lipid raft dynamics around cilia.


Assuntos
Caveolina 1/metabolismo , Cílios/metabolismo , Microdomínios da Membrana/metabolismo , Adipogenia/efeitos dos fármacos , Animais , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/genética , Aurora Quinase A/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Metabolismo Energético , Insulina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Obesidade/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais
14.
J Biol Chem ; 284(49): 34223-30, 2009 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-19837665

RESUMO

Chk1, one of the critical transducers in DNA damage/replication checkpoints, prevents entry into mitosis through inhibition of Cdk1 activity. However, it has remained unclear how this inhibition is cancelled at the G(2)/M transition. We reported recently that Chk1 is phosphorylated at Ser(286) and Ser(301) by Cdk1 during mitosis. Here, we show that mitotic Chk1 phosphorylation is accompanied by Chk1 translocation from the nucleus to the cytoplasm in prophase. This translocation advanced in accordance with prophase progression and was regulated by Crm-1-dependent nuclear export. Exogenous Chk1 mutated at Ser(286) and Ser(301) to Ala (S286A/S301A) was observed mainly in the nuclei of prophase cells, although such nuclear accumulation was hardly observed in wild-type Chk1. Induction of S286A/S301A resulted in the delay of mitotic entry. Biochemical analyses using immunoprecipitated cyclin B(1)-Cdk1 complexes revealed S286A/S301A expression to block the adequate activation of Cdk1. In support of this, S286A/S301A expression retained Wee1 at higher levels and Cdk1-induced phosphorylation of cyclin B(1) and vimentin at lower levels. A kinase-dead version of S286A/S301A also localized predominantly in the nucleus but lost the ability to delay mitotic entry. These results indicate that Chk1 phosphorylation by Cdk1 participates in cytoplasmic sequestration of Chk1 activity, which releases Cdk1 inhibition in the nucleus and promotes mitotic entry.


Assuntos
Proteína Quinase CDC2/metabolismo , Núcleo Celular/metabolismo , Regulação da Expressão Gênica , Proteínas Quinases/metabolismo , Transporte Ativo do Núcleo Celular , Ciclo Celular , Divisão Celular , Quinase 1 do Ponto de Checagem , Ciclina B1/metabolismo , Citoplasma/metabolismo , Fase G2 , Células HeLa , Humanos , Mitose , Fosforilação , Vimentina/metabolismo
15.
Biochim Biophys Acta ; 1790(10): 1345-52, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19619611

RESUMO

BACKGROUND: The Src-family non-receptor-type tyrosine kinase Lyn, which is often associated with chemotherapeutic resistance in cancer, localizes not only to the plasma membrane but also Golgi membranes. Recently, we showed that Lyn, which is synthesized in the cytosol, is transported from the Golgi to the plasma membrane along the secretory pathway. However, it is still unclear how Golgi targeting of newly synthesized Lyn is regulated. METHODS: Subcellular localization of Lyn and its mutants was determined by confocal microscopy. RESULTS: We show that the kinase domain, but not the SH3 and SH2 domains, of Lyn is required for the targeting of Lyn to the Golgi, whereas the N-terminal lipids of the Lyn SH4 domain are not sufficient for its Golgi targeting. Although intact Lyn, which colocalizes with caveolin-positive Golgi membranes, can traffic toward the plasma membrane, kinase domain-deleted Lyn is immobilized on caveolin-negative Golgi membranes. GENERAL SIGNIFICANCE: Besides the SH4 domain, the Lyn kinase domain is important for targeting of newly synthesized Lyn to the Golgi, especially caveolin-positive transport membranes. Our results provide a novel role of the Lyn catalytic domain in the Golgi targeting of newly synthesized Lyn in a manner independent of its kinase activity.


Assuntos
Caveolinas/metabolismo , Complexo de Golgi/metabolismo , Membranas Intracelulares/metabolismo , Quinases da Família src/metabolismo , Animais , Sítios de Ligação/genética , Células COS , Domínio Catalítico/genética , Membrana Celular/metabolismo , Chlorocebus aethiops , Cicloeximida/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Microscopia Confocal , Mutação , Inibidores da Síntese de Proteínas/farmacologia , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Domínios de Homologia de src/genética , Quinases da Família src/genética
16.
Exp Cell Res ; 315(7): 1117-41, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19245808

RESUMO

Src-family kinases (SFKs), which participate in various signaling events, are found at not only the plasma membrane but also several subcellular compartments, including the nucleus. Nuclear structural changes are frequently observed during transcription, cell differentiation, senescence, tumorigenesis, and cell cycle. However, little is known about signal transduction in the alteration of chromatin texture. Here, we develop a pixel imaging method for quantitatively evaluating chromatin structural changes. Growth factor stimulation increases euchromatic hypocondensation and concomitant heterochromatic hypercondensation in G(1) phase, and the levels reach a plateau by 30 min, sustain for at least 5 h and return to the basal levels after 24 h. Serum-activated SFKs in the nucleus were more frequently detected in the euchromatin areas than the heterochromatin areas. Nuclear expression of kinase-active SFKs, but not unrelated Syk kinase, drastically increases both euchromatinization and heterochromatinization in a manner dependent on the levels of nuclear tyrosine phosphorylation. However, growth factor stimulation does not induce chromatin structural changes in SYF cells lacking SFKs, and reintroduction of one SFK member into SYF cells can, albeit insufficiently, induce chromatin structural changes. These results suggest that nuclear tyrosine phosphorylation by SFKs plays an important role in chromatin structural changes upon growth factor stimulation.


Assuntos
Eucromatina/química , Eucromatina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Microscopia de Fluorescência/métodos , Conformação de Ácido Nucleico , Quinases da Família src/metabolismo , Androstadienos/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Células COS , Ciclo Celular/fisiologia , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Chlorocebus aethiops , Eucromatina/genética , Células HeLa , Histonas/metabolismo , Humanos , Inibidores de Proteínas Quinases/metabolismo , Proteína Fosfatase 2/metabolismo , Transdução de Sinais/fisiologia , Vanadatos/metabolismo , Wortmanina , Quinases da Família src/genética
17.
J Cell Biol ; 165(5): 641-52, 2004 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-15173188

RESUMO

Src-family kinases, known to participate in signaling pathways of a variety of surface receptors, are localized to the cytoplasmic side of the plasma membrane through lipid modification. We show here that Lyn, a member of the Src-family kinases, is biosynthetically transported to the plasma membrane via the Golgi pool of caveolin along the secretory pathway. The trafficking of Lyn from the Golgi apparatus to the plasma membrane is inhibited by deletion of the kinase domain or Csk-induced "closed conformation" but not by kinase inactivation. Four residues (Asp346 and Glu353 on alphaE helix, and Asp498 and Asp499 on alphaI helix) present in the C-lobe of the kinase domain, which can be exposed to the molecular surface through an "open conformation," are identified as being involved in export of Lyn from the Golgi apparatus toward the plasma membrane but not targeting to the Golgi apparatus. Thus, the kinase domain of Lyn plays a role in Lyn trafficking besides catalysis of substrate phosphorylation.


Assuntos
Cavéolas/enzimologia , Caveolinas/metabolismo , Membrana Celular/metabolismo , Complexo de Golgi/metabolismo , Quinases da Família src/biossíntese , Sequência de Aminoácidos/fisiologia , Animais , Anticorpos/farmacologia , Ácido Aspártico/metabolismo , Células COS , Cavéolas/ultraestrutura , Caveolina 1 , Membrana Celular/enzimologia , Membrana Celular/ultraestrutura , Ácido Glutâmico , Complexo de Golgi/enzimologia , Complexo de Golgi/ultraestrutura , Células HeLa , Humanos , Dados de Sequência Molecular , Mutação/genética , Estrutura Secundária de Proteína/fisiologia , Estrutura Terciária de Proteína/genética , Transporte Proteico/fisiologia , Quinases da Família src/antagonistas & inibidores
18.
Exp Cell Res ; 314(10): 2040-54, 2008 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-18457834

RESUMO

Protein-tyrosine phosphorylation is regulated by protein-tyrosine kinases and protein-tyrosine phosphatases (PTPs). Src-family tyrosine kinases (SFKs) participate in the regulation of the actin cytoskeleton. Actin filaments can be accumulated in a cap at the dorsal cell surface, which is called the cortical actin cap. Here, we show that SFKs play an important role in formation of the cortical actin cap. HeLa cells normally exhibit the cortical actin cap, one of the major sites of tyrosine phosphorylation. The cortical actin cap is disrupted by SFK inhibitors or overexpression of the Lyn SH3 domain. Csk-knockout cells form the cortical actin cap when the level of tyrosine phosphorylation is increased by Na(3)VO(4), a PTP inhibitor, and the formation of the cortical actin cap is inhibited by SFK inactivation with re-introduction of Csk. SYF cells lacking SFKs minimally exhibit the cortical actin cap even in the presence of Na(3)VO(4), and transfection with Lyn restores the cortical actin cap in the presence of Na(3)VO(4). Disruption of the cortical actin cap by dominant-negative Cdc42 causes loss of tyrosine phosphorylation at the cell top. These results suggest that SFK(s) is involved in formation of the cortical actin cap, which may serve as a platform of tyrosine phosphorylation signaling.


Assuntos
Actinas/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Quinases da Família src/metabolismo , Animais , Proteína Tirosina Quinase CSK , Linhagem Celular , Membrana Celular/ultraestrutura , Citoesqueleto/ultraestrutura , Humanos , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Pirimidinas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia , Vanadatos/metabolismo , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética
19.
Exp Cell Res ; 314(18): 3392-404, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18817770

RESUMO

Src-family kinases, cytoplasmic enzymes that participate in various signaling events, are found at not only the plasma membrane but also subcellular compartments, such as the nucleus, the Golgi apparatus and late endosomes/lysosomes. Lyn, a member of the Src-family kinases, is known to play a role in DNA damage response and cell cycle control in the nucleus. However, it is still unclear how the localization of Lyn to the nucleus is regulated. Here, we investigated the mechanism of the distribution of Lyn between the cytoplasm and the nucleus in epitheloid HeLa cells and hematopoietic THP-1 cells. Lyn was definitely detected in purified nuclei by immunofluorescence and immunoblotting analyses. Nuclear accumulation of Lyn was enhanced upon treatment of cells with leptomycin B (LMB), an inhibitor of Crm1-mediated nuclear export. Moreover, Lyn mutants lacking the sites for lipid modification were highly accumulated in the nucleus upon LMB treatment. Intriguingly, inhibition of the kinase activity of Lyn by SU6656, Csk overexpression, or point mutation in the ATP-binding site induced an increase in nuclear Lyn levels. These results suggest that Lyn being imported into and rapidly exported from the nucleus preferentially accumulates in the nucleus by inhibition of the kinase activity and lipid modification.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Núcleo Celular/metabolismo , Quinases da Família src/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Sequência de Aminoácidos , Linhagem Celular Tumoral , Núcleo Celular/química , Núcleo Celular/enzimologia , Citoplasma/enzimologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ácidos Graxos Insaturados/farmacologia , Imunofluorescência , Células HeLa , Humanos , Immunoblotting , Dados de Sequência Molecular , Mutação , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética
20.
Adv Sci (Weinh) ; 6(1): 1801138, 2019 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-30643718

RESUMO

Primary cilia detect extracellular cues and transduce these signals into cells to regulate proliferation, migration, and differentiation. Here, the function of primary cilia as signaling hubs of growth factors and morphogens is in focus. First, the molecular mechanisms regulating the assembly and disassembly of primary cilia are described. Then, the role of primary cilia in mediating growth factor and morphogen signaling to maintain human health and the potential mechanisms by which defects in these pathways contribute to human diseases, such as ciliopathy, obesity, and cancer are described. Furthermore, a novel signaling pathway by which certain growth factors stimulate cell proliferation through suppression of ciliogenesis is also described, suggesting novel therapeutic targets in cancer.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA