Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 30(1): 312-23, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26381755

RESUMO

Proper bioriented attachment of microtubules and kinetochores is essential for the precise distribution of duplicated chromosomes to each daughter cell. An aberrant kinetochore-microtubule attachment results in chromosome instability, which leads to cellular transformation or apoptosis. In this article, we show that ubiquitin-associated protein 2-like (UBAP2L) is necessary for correct kinetochore-microtubule attachment. Depletion of UBAP2L inhibited chromosome alignment in metaphase and delayed progression to anaphase by activating spindle assembly checkpoint signaling. In addition, UBAP2L knockdown increased side-on attachment of kinetochores along the microtubules and suppressed stable kinetochore fiber formation. A proteomics analysis identified protein arginine methyltransferase (PRMT)1 as a direct interaction partner of UBAP2L. UBAP2L has an arginine- and glycine-rich motif called the RGG/RG or GAR motif in the N terminus. Biochemical analysis confirmed that arginine residues in the RGG/RG motif of UBAP2L were directly methylated by PRMT1. Finally, we demonstrated that the RGG/RG motif of UBAP2L is essential for the proper alignment of chromosomes in metaphase for the accurate distribution of chromosomes. Our results show a possible role for arginine methylation in UBAP2L for the progression of mitosis.


Assuntos
Proteínas de Transporte/metabolismo , Cinetocoros/metabolismo , Processamento de Proteína Pós-Traducional , Motivos de Aminoácidos , Arginina/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/genética , Células HEK293 , Células HeLa , Humanos , Metilação , Microtúbulos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteína-Arginina N-Metiltransferases/química , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
2.
Cancer Sci ; 107(9): 1315-20, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27316377

RESUMO

Nek2 (NIMA-related kinase 2) is a serine-threonine kinase and human homolog of the mitotic regulator NIMA of Aspergillus nidulan. We reported the efficiency of Nek2 siRNA in several cancer xenograft models using cholangiocarcinoma, breast cancer and colorectal cancer. Pancreatic cancer is difficult to treat due to its rapid progression and resistance to chemotherapy. Novel treatments are urgently required to improve survival in pancreatic cancer, and siRNA are a promising therapeutic option. However, finding an in vivo drug delivery system of siRNA remains a major problem for clinical application. In this study, the overexpression of Nek2 was identified in pancreatic cancer cell lines. Nek2 siRNA inhibited tumor growth in a subcutaneous xenograft mouse model of pancreatic cancer, prolonged the survival time in an intraperitoneal xenograft mouse model and efficiently prevented the progression of liver metastasis using a portal venous port-catheter system. Taken together, Nek2 is an effective therapeutic target in pancreatic cancer. An adequate delivery system is considered important in treating advanced pancreatic cancer, such as peritoneal dissemination and liver metastasis. Further investigations are required on the safety and side effects of the portal venous port-catheter system. We hope that Nek2 siRNA will be a novel therapeutic strategy for pancreatic cancer with liver metastasis and peritoneal dissemination.


Assuntos
Neoplasias Hepáticas/secundário , Quinases Relacionadas a NIMA/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Veia Porta , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Dispositivos de Acesso Vascular , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Modelos Animais de Doenças , Expressão Gênica , Técnicas de Silenciamento de Genes , Terapia Genética , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Masculino , Camundongos , Camundongos Nus , Interferência de RNA , Ratos , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Tumour Biol ; 37(4): 4531-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26503212

RESUMO

Protein arginine methylation, which is mediated by a family of protein arginine methyltransferases (PRMTs), is associated with numerous fundamental cellular processes. Accumulating studies have revealed that the expression of multiple PRMTs promotes cancer progression. In this study, we examined the role of PRMT1 in ovarian cancer cells. PRMT1 is expressed in multiple ovarian cancer cells, and the depletion of its expression suppressed colony formation, in vivo proliferation, migration, and invasion. To gain insight into PRMT1-mediated cancer progression, we searched for novel substrates of PRMT1. We found that FAM98A, whose physiological function is unknown, was arginine-methylated by PRMT1. FAM98A is expressed in numerous ovarian cancer cell lines and is important for the malignant characteristics of ovarian cancer cells. Our results indicate the possible role of the PRMT1-FAM98A pathway in cancer progression.


Assuntos
Movimento Celular , Processamento de Proteína Pós-Traducional , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas/metabolismo , Proteínas Repressoras/metabolismo , Animais , Linhagem Celular Tumoral , Células HEK293 , Humanos , Metilação , Camundongos Nus , Invasividade Neoplásica , Transplante de Neoplasias
4.
Tumour Biol ; 37(1): 763-72, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26245992

RESUMO

Ubiquitination is essential for various biological processes, such as signal transduction, intracellular trafficking, and protein degradation. Accumulating evidence has demonstrated that ubiquitination plays a crucial role in cancer development. In this report, we examine the expression and function of ubiquitin-conjugating enzyme E2S (UBE2S) in breast cancer. Immunohistochemical analysis revealed that UBE2S is highly expressed in breast cancer. The depletion of UBE2S by siRNA induced disruption of the actin cytoskeleton and focal adhesions. Interestingly, phosphorylation of FAK at Tyr397, which is important for the transduction of integrin-mediated signaling, was significantly reduced by UBE2S knockdown. We also show that UBE2S knockdown suppressed the malignant characteristics of breast cancer cells, such as migration, invasion, and anchorage-independent growth. Our results indicate that UBE2S could be a potential target for breast cancer treatment.


Assuntos
Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Enzimas de Conjugação de Ubiquitina/metabolismo , Citoesqueleto de Actina/metabolismo , Idoso , Anoikis , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Transformação Celular Neoplásica , Citoplasma/metabolismo , Feminino , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Imuno-Histoquímica , Integrinas/metabolismo , Células MCF-7 , Pessoa de Meia-Idade , Invasividade Neoplásica , Fosforilação , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Ubiquitinação
5.
Exp Cell Res ; 332(1): 78-88, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25523619

RESUMO

Invadopodia are specialized actin-based microdomains of the plasma membrane that combine adhesive properties with matrix degrading activities. Proper functioning of the bone, immune, and vascular systems depend on these organelles, and their relevance in cancer cells is linked to tumor metastasis. The elucidation of the mechanisms driving invadopodia formation is a prerequisite to understanding their role and ultimately to controlling their functions. Special AT-rich sequence-binding protein 2 (SATB2) was reported to suppress tumor cell migration and metastasis. However, the mechanism of action of SATB2 is unknown. Here, we show that SATB2 inhibits invadopodia formation in HCT116 cells and that the molecular scaffold palladin is inhibited by exogenous expression of SATB2. To confirm this association, we elucidated the function of palladin in HCT116 using a knock down strategy. Palladin knock down reduced cell migration and invasion and inhibited invadopodia formation. This phenotype was confirmed by a rescue experiment. We then demonstrated that palladin expression in SATB2-expressing cells restored invasion and invadopodia formation. Our results showed that SATB2 action is mediated by palladin inhibition and the SATB2/palladin pathway is associated with invadopodia formation in colorectal cancer cells.


Assuntos
Extensões da Superfície Celular/fisiologia , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/fisiologia , Fosfoproteínas/metabolismo , Fatores de Transcrição/fisiologia , Movimento Celular , Células HCT116 , Humanos , Transporte Proteico
6.
J Cell Sci ; 126(Pt 15): 3263-70, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23704356

RESUMO

Centralspindlin, which is composed of MgcRacGAP and MKLP1, is essential for central spindle formation and cytokinetic furrow ingression. MgcRacGAP utilizes its GAP domain to inactivate Rac1 and induce furrow ingression in mammalian cells. In this report, we present a novel regulatory mechanism for furrowing that is mediated by the phosphorylation of SHC SH2-domain binding protein 1 (SHCBP1), a binding partner of centralspindlin, by Aurora B (AurB). AurB phosphorylates Ser634 of SHCBP1 during mitosis. We generated a phosphorylation site mutant, S634A-SHCBP1, which was prematurely recruited to the central spindle during anaphase and inhibited furrowing. An in vitro GAP assay demonstrated that SHCBP1 can suppress the MgcRacGAP-mediated inactivation of Rac1. In addition, the inhibition of Rac1 activity rescued the furrowing defect induced by S634A-SHCBP1 expression. Thus, AurB phosphorylates SHCBP1 to prevent the premature localization of SHCBP1 to the central spindle and ensures that MgcRacGAP inactivates Rac1 to promote the ingression of the cytokinetic furrow.


Assuntos
Aurora Quinase B/metabolismo , Ciclo Celular/fisiologia , Citocinese/fisiologia , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Fuso Acromático/metabolismo , Sequência de Aminoácidos , Aurora Quinase B/genética , Ciclo Celular/genética , Citocinese/genética , Células HeLa , Humanos , Dados de Sequência Molecular , Fosforilação , Proteínas Adaptadoras da Sinalização Shc/genética , Fuso Acromático/química , Fuso Acromático/genética
7.
J Cell Sci ; 126(Pt 16): 3627-37, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23750008

RESUMO

Polo-like kinase 1 (PLK1) is a widely conserved serine/threonine kinase that regulates progression of multiple stages of mitosis. Although extensive studies about PLK1 functions during cell division have been performed, it is still not known how PLK1 regulates myosin II activation at the equatorial cortex and ingression of the cleavage furrow. In this report, we show that an actin/myosin-II-binding protein, supervillin (SVIL), is a substrate of PLK1. PLK1 phosphorylates Ser238 of SVIL, which can promote the localization of SVIL to the central spindle and association with PRC1. Expression of a PLK1 phosphorylation site mutant, S238A-SVIL, inhibited myosin II activation at the equatorial cortex and induced aberrant furrowing. SVIL has both actin- and myosin-II-binding regions in the N-terminus. Expression of ΔMyo-SVIL (deleted of the myosin-II-binding region), but not of ΔAct-SVIL (deleted of actin-binding region), reduced myosin II activation and caused defects in furrowing. Our study indicates a possible role of phosphorylated SVIL as a molecular link between the central spindle and the contractile ring to coordinate the activation of myosin II for the ingression of the cleavage furrow.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Miosina Tipo II/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fuso Acromático/metabolismo , Proteínas de Ciclo Celular/genética , Citocinese/fisiologia , Células HeLa , Humanos , Proteínas de Membrana/genética , Proteínas dos Microfilamentos/genética , Miosina Tipo II/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Transfecção , Quinase 1 Polo-Like
8.
Carcinogenesis ; 35(9): 1993-2001, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24675530

RESUMO

Pleomorphic adenoma gene like-2 (PLAGL2), a member of the PLAG gene family, is a C2H2 zinc finger transcriptional factor that is involved in cellular transformation and apoptosis. In this report, we show that PLAGL2 is associated with the organization of stress fibers and with small guanosine triphosphatase (GTPase) activity. Depletion of PLAGL2 in two different ovarian cancer cell lines, ES-2 and HEY, induced activation of RhoA, whereas activity of Rac1 was suppressed. Organization of actin stress fibers and focal adhesions was significantly promoted by PLAGL2 knockdown in a RhoA-dependent manner. Conversely, exogenous expression of PLAGL2 in MDA-MB-231 cells, a breast cancer cell line, resulted in the activation of Rac1 and the inactivation of RhoA. In addition, PLAGL2 expression induced lamellipodia formation and disruption of stress fiber formation. Finally, we show that CHN1 expression is essential for Rac1 inactivation in PLAGL2-depleted cells. Our results demonstrate a crucial role of PLAGL2 in actin dynamics and give further insight into the role of PLAGL2 in cellular transformation and apoptosis.


Assuntos
Movimento Celular , Proteínas de Ligação a DNA/fisiologia , Proteínas de Ligação a RNA/fisiologia , Fibras de Estresse/metabolismo , Fatores de Transcrição/fisiologia , Linhagem Celular Tumoral , Quimerina 1/metabolismo , Humanos , Pseudópodes/metabolismo , Pseudópodes/patologia , Fibras de Estresse/patologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
9.
Cancer Sci ; 105(12): 1526-32, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25250919

RESUMO

The striatin family of proteins, comprising STRN, STRN3 and STRN4, are multidomain-containing proteins that associate with additional proteins to form a large protein complex. We previously reported that STRN4 directly associated with protein kinases, such as MINK1, TNIK and MAP4K4, which are associated with tumor suppression or tumor progression. However, it remains unclear whether STRN4 is associated with tumor progression. In this report, we examined the role that STRN4 plays in cancer malignancy. We show that depletion of STRN4 suppresses proliferation, migration, invasion and the anchorage-independent growth of cancer cells. In addition, STRN4 knockdown increases the sensitivity of pancreatic cancer cells to gemcitabine. Finally, we show that STRN4 knockdown suppresses the proliferation and metastasis of cancer cells in mice. Our results demonstrate a possible role of STRN4 in tumor progression.


Assuntos
Proteínas de Ligação a Calmodulina/genética , Proteínas de Ligação a Calmodulina/metabolismo , Metástase Neoplásica/patologia , Neoplasias Experimentais/patologia , Proteínas do Tecido Nervoso/metabolismo , Neoplasias Pancreáticas/patologia , Animais , Anoikis , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células HCT116 , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Neoplasias Experimentais/genética , Proteínas do Tecido Nervoso/genética , Neoplasias Pancreáticas/genética , RNA Interferente Pequeno/metabolismo , Gencitabina
10.
Mol Cell Biochem ; 389(1-2): 9-16, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24337944

RESUMO

Membrane blebs are round-shaped dynamic membrane protrusions that occur under many physiological conditions. Membrane bleb production is primarily controlled by actin cytoskeletal rearrangements mediated by RhoA. Tre2-Bub2-Cdc16 (TBC) domain-containing proteins are negative regulators of the Rab family of small GTPases and contain a highly conserved TBC domain. In this report, we show that the expression of TBC1D15 is associated with the activity of RhoA and the production of membrane blebs. Depletion of TBC1D15 induced activation of RhoA and membrane blebbing, which was abolished by the addition of an inhibitor for RhoA signaling. In addition, we show that TBC1D15 is required for the accumulation of RhoA at the equatorial cortex for the ingression of the cytokinetic furrow during cytokinesis. Our results demonstrate a novel role for TBC1D15 in the regulation of RhoA during membrane blebbing and cytokinesis.


Assuntos
Proteínas Ativadoras de GTPase/genética , Inativação Gênica/fisiologia , Membranas/fisiologia , Proteína rhoA de Ligação ao GTP/genética , Linhagem Celular Tumoral , Citocinese/genética , Citocinese/fisiologia , Proteínas Ativadoras de GTPase/metabolismo , Células HeLa , Humanos , Transdução de Sinais/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo
11.
FEBS J ; 291(5): 927-944, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38009294

RESUMO

There has been a great deal of research on cell division and its mechanisms; however, its processes still have many unknowns. To find novel proteins that regulate cell division, we performed the screening using siRNAs and/or the expression plasmid of the target genes and identified leucine zipper protein 1 (LUZP1). Recent studies have shown that LUZP1 interacts with various proteins and stabilizes the actin cytoskeleton; however, the function of LUZP1 in mitosis is not known. In this study, we found that LUZP1 colocalized with the chromosomal passenger complex (CPC) at the centromere in metaphase and at the central spindle in anaphase and that these LUZP1 localizations were regulated by CPC activity and kinesin family member 20A (KIF20A). Mass spectrometry analysis identified that LUZP1 interacted with death-associated protein kinase 3 (DAPK3), one regulator of the cleavage furrow ingression in cytokinesis. In addition, we found that LUZP1 also interacted with myosin light chain 9 (MYL9), a substrate of DAPK3, and comprehensively inhibited MYL9 phosphorylation by DAPK3. In line with a known role for MYL9 in the actin-myosin contraction, LUZP1 suppression accelerated the constriction velocity at the division plane in our time-lapse analysis. Our study indicates that LUZP1 is a novel regulator for cytokinesis that regulates the constriction velocity of the contractile ring.


Assuntos
Citocinese , Zíper de Leucina , Citocinese/genética , Constrição , Citoesqueleto de Actina , Mitose
12.
J Biol Chem ; 287(30): 25019-29, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22665485

RESUMO

Cytokinesis is initiated by constriction of the cleavage furrow and terminated by abscission of the intercellular bridge that connects two separating daughter cells. The complicated processes of cytokinesis are coordinated by phosphorylation and dephosphorylation mediated by protein kinases and phosphatases. Mammalian Misshapen-like kinase 1 (MINK1) is a member of the germinal center kinases and is known to regulate cytoskeletal organization and oncogene-induced cell senescence. To search for novel regulators of cytokinesis, we performed a screen using a library of siRNAs and found that MINK1 was essential for cytokinesis. Time-lapse analysis revealed that MINK1-depleted cells were able to initiate furrowing but that abscission was disrupted. STRN4 (Zinedin) is a regulatory subunit of protein phosphatase 2A (PP2A) and was recently shown to be a component of a novel protein complex called striatin-interacting phosphatase and kinase (STRIPAK). Mass spectrometry analysis showed that MINK1 was a component of STRIPAK and that MINK1 directly interacted with STRN4. Similar to MINK1 depletion, STRN4-knockdown induced multinucleated cells and inhibited the completion of abscission. In addition, STRN4 reduced MINK1 activity in the presence of catalytic and structural subunits of PP2A. Our study identifies a novel regulatory network of protein kinases and phosphatases that regulate the completion of abscission.


Assuntos
Proteínas de Ligação a Calmodulina/metabolismo , Citocinese/fisiologia , Complexos Multienzimáticos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteína Fosfatase 2/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Ligação a Calmodulina/genética , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Complexos Multienzimáticos/genética , Proteínas do Tecido Nervoso/genética , Proteína Fosfatase 2/genética , Proteínas Serina-Treonina Quinases/genética
13.
Mol Cell Biochem ; 374(1-2): 105-11, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23129259

RESUMO

Dynamic remodeling of the actin cytoskeleton is crucial for biological processes such as cell migration and cell spreading. S100A10 is a member of the S100 protein family and is involved in intracellular trafficking and cell migration. In this study, we examined the role of S100A10 in actin cytoskeletal organization and cell spreading. Depletion of S100A10 induced disruption of stress fiber formation and delay in cell spreading. Rac1 activation during spreading was suppressed by S100A10 knockdown, and exogenous expression of active Rac1 restored the ability of cells to spread in the absence of S100A10. Our results demonstrate the crucial role of S100A10 in actin dynamics promoting cell spreading via Rac1 activation.


Assuntos
Citoesqueleto de Actina/metabolismo , Anexina A2/genética , Anexina A2/metabolismo , Proteínas S100/genética , Proteínas S100/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Células HEK293 , Células HeLa , Humanos , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno , Fibras de Estresse
14.
J Biol Chem ; 285(6): 3806-3814, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19948721

RESUMO

Nitric oxide (NO) plays a pivotal role in tumorigenesis, particularly with relation to cancer cell invasion and metastasis. NO can reversibly couple to cysteine thiols to form an S-nitrosothiol, which regulates the enzymatic activities of target proteins. c-Src is a tyrosine kinase that promotes cancer cell invasion and metastasis. Interestingly, c-Src can be activated by NO stimulation. However, mechanisms by which NO stimulates Src kinase activity have not been elucidated. We report here that NO causes S-nitrosylation of c-Src at cysteine 498 (Cys(498)) to stimulate its kinase activity. Cys(498) is conserved among Src family kinases, and Cys(506) of c-Yes, which corresponds to Cys(498) of c-Src, was also important for the NO-mediated activation of c-Yes. Estrogens may work synergistically with NO to induce the proliferation and migration of many kinds of breast cancer cells. For example, beta-estradiol induces the expression of endothelial nitric synthase and production of NO in MCF7 cells. We found that activation of c-Src in MCF7 cells by beta-estradiol stimulation was mediated by the S-nitrosylation of Cys(498). In addition, we report that disruption of E-cadherin junctions and enhancement of cell invasion by beta-estradiol stimulation was mediated by NO-dependent activation of c-Src. These results identify a novel signaling pathway that links NO and Src family kinases to cancer cell invasion and metastasis.


Assuntos
Movimento Celular , Cisteína/metabolismo , Óxido Nítrico/metabolismo , Quinases da Família src/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Caderinas/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Estradiol/farmacologia , Humanos , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutação , Doadores de Óxido Nítrico/farmacologia , Nitrosação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fyn/genética , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Proteínas Proto-Oncogênicas c-yes/genética , Proteínas Proto-Oncogênicas c-yes/metabolismo , Interferência de RNA , S-Nitroso-N-Acetilpenicilamina/farmacologia , Homologia de Sequência de Aminoácidos , Quinases da Família src/genética
17.
Eur J Haematol ; 86(3): 191-8, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21114537

RESUMO

Internal tandem duplication of FMS-like receptor tyrosine kinase 3 (FLT3/ITD) within its juxtamembrane domain is a frequent mutation in adult acute myeloid leukaemia (AML). This mutation causes constitutive activation of FLT3 and is associated with poor prognosis. The high relapse rate of FLT3/ITD-positive AML might be partly because of insufficient eradication of slow-cycling leukaemic stem cells in the bone marrow microenvironment. ß1 integrin mediates haematopoietic stem and progenitor cell homing along with their retention in the bone marrow and also inhibits haematopoietic proliferation and differentiation. Here, we demonstrate that inhibition of FLT3/ITD kinase activity by a FLT3 selective inhibitor named FI-700 decreases affinity of α4ß1 integrin to soluble VCAM-1. α4ß1 integrin deactivation by FI-700 is independent of Rap1, which is the critical regulator of integrin inside-out signalling. In addition, selective inhibition of FLT3/ITD induces Pyk2 dephosphorylation together with the inhibition of phosphatidylinositol-3-kinase (PI3K)/Akt pathway. Both wild-type and ITD-FLT3 proteins co-immunoprecipitated with ß1 integrin and Pyk2 indicating the signal crosstalk between FLT3, ß1 integrin and Pyk2. These results collectively indicated that the inhibition of FLT3 kinase might contribute not only to the induction of apoptosis, but also to the leukaemia cell detachment from the bone marrow microenvironment in the treatment of AML.


Assuntos
Quinase 2 de Adesão Focal/fisiologia , Integrina alfa4beta1/fisiologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/fisiopatologia , Mutação , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/fisiologia , Animais , Adesão Celular/genética , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Duplicação Gênica , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Camundongos , Complexos Multiproteicos , Fosforilação , Piridinas/farmacologia , Pirimidinas/farmacologia , Complexo Shelterina , Transdução de Sinais , Sequências de Repetição em Tandem , Proteínas de Ligação a Telômeros/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores
18.
Cancer Sci ; 101(5): 1163-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20345485

RESUMO

Nek2 (NIMA-related kinase 2) is involved in cell division and mitotic regulation by centrosome splitting. We previously reported that Nek2 depletion causes growth suppression and cell death in cholangiocarcinoma and breast cancer cells. In this report, we examine the effect of a combination treatment using Nek2 siRNA with the cytotoxic chemotherapeutic agent cisplatin (CDDP) on colorectal cancer. Nek2 was overexpressed in all colorectal cancer cell lines examined (HCT-15, DLD-1, Colo205, and Colo320). Nek2 short-interfering RNA (siRNA) resulted in the inhibition of cell proliferation and the induction of apoptosis in vitro. Nek2 siRNA suppressed tumor growth compared to control siRNA in a xenograft mouse model. To investigate the potential utility of Nek2 siRNA for clinical cancer therapy, we examine the effect of a combination treatment using Nek2 siRNA with CDDP on colorectal cancer. The combined administration of both Nek2 siRNA and CDDP inhibited cell proliferation and induced apoptotic cell death in vitro. Furthermore, the combined administration of both Nek2 siRNA and CDDP suppressed tumor growth compared to either the single administration of Nek2 siRNA or the combined administration of control siRNA and CDDP. Our results suggest that combination treatment using Nek2 siRNA and chemotherapeutic agents may be an effective therapeutic option for colorectal cancer.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Neoplasias Colorretais/terapia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , RNA Interferente Pequeno/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Terapia Combinada , Humanos , Quinases Relacionadas a NIMA
19.
Biochem Biophys Res Commun ; 400(2): 230-5, 2010 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-20727856

RESUMO

Gap junctional communication, which is mediated by the connexin protein family, is essential for the maintenance of normal tissue function and homeostasis. Loss of intercellular communication results in a failure to coordinately regulate cellular functions, and it can facilitate tumorigenesis. Expression of oncogenes and stimulation with cytokines has been shown to suppress intercellular communication; however, the exact mechanism by which intercellular communication is disrupted by these factors remains uncertain. In this report, we show that Akt is essential for the disruption of gap junctional communication in v-Src-transformed cells. In addition, inhibition of Akt restores gap junctional communication after it is suppressed by TNF-α signaling. Furthermore, we demonstrate that the expression of a constitutively active form of Akt1, but not of Akt2 or Akt3, is sufficient to suppress gap junctional communication. Our results clearly define Akt1 as one of the critical regulators of gap junctional communication.


Assuntos
Comunicação Celular , Junções Comunicantes/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Linhagem Celular Transformada , Junções Comunicantes/enzimologia , Camundongos , Proteína Oncogênica pp60(v-src)/metabolismo , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
20.
Exp Cell Res ; 315(8): 1384-92, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19268464

RESUMO

Both MMP-2 and MMP-9 play critical roles in tumor invasion, but their productions are differentially controlled. While the promoter region of MMP-9 has the conserved proximal AP-1 binding site, that of the MMP-2 has a noncanonical AP-1 site. To assess the role of AP-1 function, we examined the effects of dominant-negative Fos (DeltaFos), BATF and siRNA against c-Jun on MMP production in v-Crk-transformed cells which have augmented production of MMP-2 and MMP-9. Suppression of AP-1 dependent transcription by conditional expression of dominant-negative Fos (DeltaFos) and BATF substantially inhibited not only MMP-9 production but also MMP-2 production. The ChIP analysis showed the direct association of AP-1 and MMP-2 promoter region. In addition, silencing of c-Jun expression by siRNA transfection suppressed MMP-2 and MMP-9 production and in vitro invasiveness. Furthermore, the invadopodia formation of v-Crk-transformed cells could be suppressed by BATF expression or c-Jun siRNA treatment. Taken together, AP-1 appears to play a critical role in the production of MMP-2 and MMP-9 and invadopodia formation of v-Crk-transformed cells.


Assuntos
Regulação da Expressão Gênica , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Invasividade Neoplásica , Proteína Oncogênica v-crk/genética , Fator de Transcrição AP-1/metabolismo , Animais , Northern Blotting , Linhagem Celular , Linhagem Celular Transformada , Deleção de Genes , Immunoblotting , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Proteínas Oncogênicas v-fos/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA