Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Med (Berl) ; 98(5): 761-773, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32318747

RESUMO

Tight junction protein 1 (TJP1) is a membrane-associated cytosolic protein important for cell-cell communication in intercellular barriers in epithelial and non-epithelial cells. Here, we explored the functional involvement of TJP1 in non-epithelial tumors such as soft tissue sarcoma, especially in leiomyosarcoma (LMS). TJP1 expression in soft tissue sarcoma was analyzed in normal and tumor tissues as well as from public datasets such as the TCGA provisional dataset, in which TJP1 expression was compared with other subtypes such as undifferentiated sarcomas, and myxofibrosarcomas. SK-LMS-1 cell lines with reduced TJP1 expression showed attenuated anchorage-independent colony formation as well as reduced intercellular aggregation on non-coated culture plates compared with control as well as parental SK-LMS-1 cells. Transcriptome profiling following TJP1 knockdown in SK-LMS-1 cells suggested the involvement of several signaling pathways, including NF-κB pathway and growth factor receptor signaling. In addition, TJP1 downregulation induced enhanced response against anti-cancer agents, doxorubicin and gefitinib. Taken together, these results suggest that TJP1 contributes to sarcoma genesis and might be useful therapeutic target. KEY MESSAGES: • TJP1 expression at RNA level higher in tumor than in normal tissues of sarcoma. • Targeting TJP1 attenuates cell-cell aggregation and anchorage-independent growth. • Targeting TJP1 is beneficial in anti-cancer therapy in LMS.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/genética , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Leiomiossarcoma/genética , Proteína da Zônula de Oclusão-1/genética , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Proliferação de Células , Edição de Genes , Perfilação da Expressão Gênica , Inativação Gênica , Humanos , Janus Quinases/metabolismo , Leiomiossarcoma/tratamento farmacológico , Leiomiossarcoma/metabolismo , Leiomiossarcoma/patologia , NF-kappa B/metabolismo , RNA Interferente Pequeno/genética , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Transcriptoma , Proteína da Zônula de Oclusão-1/antagonistas & inibidores
2.
J Clin Med ; 8(8)2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31390831

RESUMO

Corneal dystrophies (CDs) are a diverse group of inherited disorders with a heterogeneous genetic background. Here, we report the identification of a novel ZNF143 heterozygous missense mutation in three individuals of the same family with clinical and pathological features that are consistent with endothelial CD. Ophthalmologic examination revealed diffuse corneal clouding and edema with decreased endothelial cell density. Pathological findings showed increased corneal thickness due to edema of basal epithelial cells and stroma, and abnormal metaplastic endothelium with stratified epithelium-like changes. Patients' metaplastic corneal endothelial cells expressed predominantly cytokerain 7, cytokeratin 19, and E-cadherin. Although Sanger sequencing did not detect any mutation associated with endothelial CDs, whole exome sequencing identified the ZNF143 c.937G>C p.(Asp313His) mutation as a candidate gene for our patients' endothelial CD. In-vitro functional studies demonstrated that mutant ZNF143 promoted the mesenchymal-to-epithelial transition; it upregulated the expression of genes associated with epithelialization in human corneal endothelial cells. Additionally, proinflammatory cytokine responsive genes were significantly enriched after mutant ZNF143 transfection, which may contribute to the severe phenotype of the three patients. These findings link a mutation in ZNF143 with endothelial CD for the first time.

3.
Cells ; 8(4)2019 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-30935019

RESUMO

Autophagy is a cellular process that disrupts and uses unnecessary or malfunctioning components for cellular homeostasis. Evidence has shown a role for autophagy in tumor cell survival, but the molecular determinants that define sensitivity against autophagic regulation in cancers are not clear. Importantly, we found that breast cancer cells with low expression levels of a zinc-finger protein, ZNF143 (MCF7 sh-ZNF143), showed better survival than control cells (MCF7 sh-Control) under starvation, which was compromised with chloroquine, an autophagy inhibitor. In addition, there were more autophagic vesicles in MCF7 sh-ZNF143 cells than in MCF7 sh-Control cells, and proteins related with the autophagic process, such as Beclin1, p62, and ATGs, were altered in cells with less ZNF143. ZNF143 knockdown affected the stability of p53, which showed a dependence on MG132, a proteasome inhibitor. Data from proteome profiling in breast cancer cells with less ZNF143 suggest a role of NAD(P)H quinone dehydrogenase 1(NQO1) for p53 stability. Taken together, we showed that a subset of breast cancer cells with low expression of ZNF143 might exhibit better survival via an autophagic process by regulating the p53⁻Beclin1 axis, corroborating the necessity of blocking autophagy for the best therapy.


Assuntos
Proteína Beclina-1/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , NAD(P)H Desidrogenase (Quinona)/metabolismo , Transdução de Sinais , Estresse Fisiológico , Transativadores/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Apoptose , Autofagia , Neoplasias da Mama/ultraestrutura , Linhagem Celular Tumoral , Sobrevivência Celular , Intervalo Livre de Doença , Feminino , Humanos , Estabilidade Proteica , Vacúolos/metabolismo , Vacúolos/ultraestrutura
4.
J Cell Mol Med ; 23(6): 4043-4053, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30933430

RESUMO

Several studies have shown that expression of zinc-finger protein 143 (ZNF143) is closely related to tumour progression including colon cancer. However, it remains unclear how ZNF143 expression is related to tumour progression within the tumour microenvironment. Here, we investigated whether ZNF143 expression affects the tumour microenvironment and tumour progression by screening molecules secreted by colon cancer cells stably expressing short-hairpin RNAs against ZNF143 or control RNAs. We observed that secretion of interleukin (IL)-8 was increased when ZNF143 expression was reduced in two colon cancer cell lines. The mRNA and protein levels of IL-8 were increased in cells following ZNF143 knockdown, and this effect was reversed when ZNF143 expression was restored. The Janus tyrosine kinase/signal transducer and activator of transcription (JAK/STAT) and extracellular signal-regulated kinase pathways were also shown to contribute to IL-8 expression in ZNF143-knockdown cells. The expression levels of ZNF143 and IL-8 were inversely correlated with three-dimensionally grown spheroids and colon cancer tissues. THP-1 cells were differentiated when cells were incubated with condition media from colon cancer cell with less ZNF143, drastically. Loss of ZNF143 may contribute to the development of colon cancer by regulating intracellular and intercellular signalling for cell plasticity and the tumour microenvironment respectively.


Assuntos
Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Interleucina-8/genética , Transativadores/genética , Diferenciação Celular/genética , Linhagem Celular Tumoral , Progressão da Doença , MAP Quinases Reguladas por Sinal Extracelular/genética , Células HCT116 , Células HT29 , Humanos , Transcrição Gênica/genética , Microambiente Tumoral/genética
5.
BMB Rep ; 50(12): 621-627, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29065970

RESUMO

We previously reported the involvement of zinc-finger protein 143 (ZNF143) on cancer cell motility in colon cancer cells. Here, ZNF143 was further characterized in breast cancer. Immunohistochemistry was used to determine the expression of ZNF143 in normal tissues and in tissues from metastatic breast cancer at various stages. Notably, ZNF143 was selectively expressed in duct and gland epithelium of normal breast tissues, which decreased when the tissue became malignant. To determine the molecular mechanism how ZNF143 affects breast cancer progression, it was knocked down by infecting benign breast cancer cells with shorthairpin (sh) RNA-lentiviral particles against ZNF143 (MCF7 sh-ZNF143). MCF7 sh-ZNF143 cells showed different cell-cell contacts and actin filament (F-actin) structures when compared with MCF7 sh-Control cells. In migration and invasion assays, ZNF143 knockdown induced increased cellular motility in breast carcinoma cells. This was reduced by the recovery of ZNF143 expression. Taken together, these results suggest that ZNF143 expression contributes to breast cancer progression. [BMB Reports 2017; 50(12): 621-627].


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Movimento Celular , Transativadores/metabolismo , Movimento Celular/genética , Feminino , Humanos , Células MCF-7 , Reação em Cadeia da Polimerase em Tempo Real , Transativadores/genética
6.
Eur J Pharmacol ; 769: 257-65, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26607468

RESUMO

The proto-oncogene c-Myc has been implicated in a variety of cellular processes, such as proliferation, differentiation and apoptosis. Several c-Myc targets have been studied; however, selective regulation of c-Myc is not easy in cancer cells. Herein, we attempt to identify chemical compounds that induce cell death in c-Myc-overexpressing cells (STF-cMyc and STF-Control) by conducting MTS assays on approximately 4000 chemical compounds. One compound, C604, induced cell death in STF-cMyc cells but not STF-Control cells. Apoptotic proteins, including caspase-3 and poly(ADP-ribose) polymerase (PARP), were cleaved in C604-treated STF-cMyc cells. In addition, SW620, HCT116 and NCI-H23 cells, which exhibit higher basal levels of c-Myc, underwent apoptotic cell death in response to C604, suggesting a role for C604 as an inducer of apoptosis in cancer cells with c-Myc amplification. C604 induced cell cycle arrest at the G2/M phase in cells, which was not affected by apoptotic inhibitors. Interestingly, C604 induced accumulation of c-Myc and Cdc25A proteins. In summary, a chemical compound was identified that may induce cell death in cancer cells with c-Myc amplification specifically through an apoptotic pathway.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/genética , Pirimidinas/farmacologia , Tiazóis/farmacologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Expressão Gênica , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , Proto-Oncogene Mas
7.
BMB Rep ; 48(2): 115-20, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25027596

RESUMO

Tight junction protein 1 (TJP1), a component of tight junction, has been reported to play a role in protein networks as an adaptor protein, and TJP1 expression is altered during tumor development. Here, we found that TJP1 expression was increased at the RNA and protein levels in TGF-ß-stimulated lung cancer cells, A549. SB431542, a type-I TGF-ß receptor inhibitor, as well as SB203580, a p38 kinase inhibitor, significantly abrogated the effect of TGF-ß on TJP1 expression. Diphenyleneiodonium, an NADPH oxidase inhibitor, also attenuated TJP1 expression in response to TGF-ß in lung cancer cells. When TJP1 expression was reduced by shRNA lentiviral particles in A549 cells (A549-sh TJP1), wound healing was much lower than in cells infected with control viral particles. Taken together, these data suggest that TGF-ß enhances TJP1 expression, which may play a role beyond structural support in tight junctions during cancer development.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Proteína da Zônula de Oclusão-1/metabolismo , Benzamidas/farmacologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Dioxóis/farmacologia , Humanos , Imidazóis/farmacologia , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/metabolismo , Oniocompostos/farmacologia , Piridinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteína da Zônula de Oclusão-1/antagonistas & inibidores , Proteína da Zônula de Oclusão-1/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Mol Carcinog ; 53 Suppl 1: E161-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24009065

RESUMO

To investigate the role of zinc-finger protein 143 in cancer cells, we stably introduced ZNF143 expression knockdown by infecting colon cancer cells with short hairpin (sh) RNA-lentiviral particles against ZNF143 (HCT116 sh-ZNF143). Compared to sh-control cells, HCT116 sh-ZNF143 cells showed faster wound healing, increased migration through Transwell chambers, and increased invasion through Matrigel in Transwell chambers. ZNF143 knockdown increased transcriptional expression of ZEB1. Additionally, ZNF143 regulated E-cadherin transcriptional expression. Small interfering-RNA-mediated silencing of ZEB1 expression affected motility in HCT116 sh-ZNF143 cells. These data suggest that ZNF143 is involved in cellular motility through a ZEB1-E-cadherin-linked pathway in colon cancer cells.


Assuntos
Caderinas/metabolismo , Movimento Celular , Neoplasias do Colo/patologia , Proteínas de Homeodomínio/metabolismo , Transativadores/fisiologia , Fatores de Transcrição/metabolismo , Apoptose , Western Blotting , Caderinas/genética , Adesão Celular , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Citometria de Fluxo , Proteínas de Homeodomínio/genética , Humanos , Técnicas Imunoenzimáticas , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Cicatrização , Homeobox 1 de Ligação a E-box em Dedo de Zinco
9.
Mol Cells ; 32(5): 415-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21909943

RESUMO

Previously, we reported that the expression of zinc-finger protein 143 (ZNF143) was induced by insulin-like growth factor-1 (IGF-1) via reactive oxygen species (ROS)- and phosphatidylinositide-3-kinase (PI3-kinase)-linked pathways in colon cancer cells. Here, we investigated whether GAIP-interacting protein, C-terminus (GIPC), a binding partner of IGF-1R, is involved in ZNF143 expression through IGF-1 and IGF-1R signaling in colon cancer cells. The knockdown of GIPC in colon cancer cells reduced ZNF143 expression in response to IGF-1. IGF-1 signaling through its receptor, leading to the phosphorylation and activation of the PI3-kinase-Akt pathway and mitogenactivated protein kinases (MAPKs) was unaffected by the knockdown of GIPC, indicating the independence of the GIPC-linked pathway from PI3-kinase- and MAPK-linked signaling in IGF-1-induced ZNF143 expression. In accordance with previous results in breast cancer cells (Choi et al., 2010), the knockdown of GIPC reduced ROS production in response to IGF-1 in colon cancer cells. Furthermore, the knockdown of GIPC reduced the expression of Rad51, which is regulated by ZNF143, in response to IGF-1 in colon cancer cells. Taken together, these data suggest that GIPC is involved in IGF-1 signaling leading to ZNF143 expression through the regulation of ROS production, which may play a role for colon cancer tumorigenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias do Colo/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Transativadores/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Células HCT116 , Humanos , MAP Quinase Quinase 4/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Rad51 Recombinase/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Exp Mol Med ; 42(10): 696-702, 2010 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-20733350

RESUMO

Expression of zinc-finger protein 143 (ZNF143), a human homolog of the Xenopus transcriptional activator protein Staf, is induced by various DNA-damaging agents including etoposide, doxorubicin, and gamma-irradiation. ZNF143 binds to cisplatin-modified DNA, and its levels are increased in cancer cells that are resistant to anticancer drugs, including cisplatin, suggesting that it plays a role in carcinogenesis and cancer cell survival. However, the mechanism of ZNF143 induction in cancer cells remains unclear. Both insulin-like growth factor-1 (IGF-1) and its receptor (IGF-1R) have been reported to be overexpressed in cancer cells and to be related to anticancer drug resistance, but the identity of the relevant signaling mediators is still being investigated. In the present study, we observed that IGF-1 was able to induce ZNF143 expression in HCT116 human colon cancer cells and that wortmannin, an inhibitor of phosphatidylinositide 3- kinase (PI3-kinase), inhibited this induction, as did diphenyleneiodonium (DPI), an NADPH oxidase inhibitor, and monodansylcardavarine (MDC), a receptor internalization inhibitor. Treatment with MDC decreased the IGF-1-stimulated generation of reactive oxygen species. Taken together, these data suggest that IGF-1 induces ZNF143 expression in cancer cells via PI3-kinase and reactive oxygen species generation during receptor internalization.


Assuntos
Neoplasias do Colo/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transativadores/biossíntese , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Neoplasias do Colo/enzimologia , Neoplasias do Colo/genética , Células HCT116 , Humanos , Transdução de Sinais , Transativadores/genética
11.
Cancer Lett ; 294(2): 254-63, 2010 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-20206441

RESUMO

Insulin-like growth factor-1 (IGF-1)/IGF-1 receptor signaling participates in a variety of cellular processes, including cell survival, growth, and proliferation. Increased expression of IGF-1R and activation of its downstream signaling components have been implicated in human cancers. Although a regulatory role for IGF-1R has been established, the relationship between IGF-1R and its binding partner, GAIP-interacting protein C-terminus (GIPC), in terms of promoting cell proliferation, remains unclear. We found that siRNA-mediated silencing of GIPC expression decreased IGF-1-mediated IGF-1R phosphorylation and cellular proliferation in breast cancer models. IGF-1-mediated cellular proliferation was also inhibited by N-acetylcysteine, which implicates reactive oxygen species generation. siRNA-mediated silencing of GIPC expression also decreased IGF-1-mediated reactive oxygen species generation. Taken together, these data suggest that GIPC contributes to IGF-1-induced cancer cell proliferation via the regulation of reactive oxygen species production.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptor IGF Tipo 1/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Receptor IGF Tipo 1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...