Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mikrochim Acta ; 191(6): 340, 2024 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-38787447

RESUMO

A new sandwich-type electrochemical biosensing platform was developed by gold @polyphthalenediamine nanohybrids (AuNP@PoPD) as the sensing platform and phosphorus doped reduced graphene oxide-hemin-palladium nanoparticles (PrGO-Hemin-PdNP) as the signal amplifier for phosphatidylinositol proteoglycan 3 (GPC3). AuNP@PoPD, co-electrodeposited into the screen printed electrode with high conductivity and stability, is dedicated to assembling the primary GPC3 aptamer (GPC3Apt). The second GPC3Apt immobilized on the high conductivity and large surface area of PrGO-Hemin-PdNP was utilized as an electrochemical signal reporter by hemin oxidation (PrGO-Hemin-PdNP-GPC3Apt). In the range 0.001-10.0 ng/mL, the hemin oxidation current signal of the electrochemical aptasensor increased log-linearly with the concentration of GPC3, the lowest detection limit was 0.13 pg/mL, and the sensitivity was 2.073 µA/µM/cm2. The aptasensor exhibited good sensing performance in a human serum sample with the relative error of 4.31-8.07%. The sandwich sensor showed good selectivity and stability for detection GPC3 in human serum samples, providing a new efficient and sensitive method for detecting HCC markers.


Assuntos
Aptâmeros de Nucleotídeos , Técnicas Biossensoriais , Técnicas Eletroquímicas , Glipicanas , Ouro , Grafite , Hemina , Limite de Detecção , Nanopartículas Metálicas , Paládio , Glipicanas/sangue , Humanos , Técnicas Eletroquímicas/métodos , Técnicas Eletroquímicas/instrumentação , Aptâmeros de Nucleotídeos/química , Hemina/química , Grafite/química , Paládio/química , Ouro/química , Técnicas Biossensoriais/métodos , Nanopartículas Metálicas/química , Eletrodos
2.
MedComm (2020) ; 5(5): e548, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38645664

RESUMO

Identifying new targets for overcoming radioresistance is crucial for improving the efficacy of lung cancer radiotherapy, given that tumor cell resistance is a leading cause of treatment failure. Recent research has spotlighted the significance of Musashi2 (MSI2) in cancer biology. In this study, we first demonstrated that MSI2 plays a key function in regulating the radiosensitivity of lung cancer. The expression of MSI2 is negatively correlated with overall survival in cancer patients, and the knockdown of MSI2 inhibits tumorigenesis and increases radiosensitivity of lung cancer cells. Cellular radiosensitivity, which is closely linked to DNA damage, is influenced by MSI2 interaction with ataxia telangiectasia mutated and Rad3-related kinase (ATR) and checkpoint kinase 1 (CHK1) post-irradiation; moreover, knockdown of MSI2 inhibits the ATR-mediated DNA damage response pathway. RNA-binding motif protein 17 (RBM17), which is implicated in DNA damage repair, exhibits increased interaction with MSI2 post-irradiation. We found that knockdown of RBM17 disrupted the interaction between MSI2 and ATR post-irradiation and increased the radiosensitivity of lung cancer cells. Furthermore, we revealed the potential mechanism of MSI2 recruitment into the nucleus with the assistance of RBM17 to activate ATR to promote radioresistance. This study provides novel insights into the potential application of MSI2 as a new target in lung cancer radiotherapy.

3.
Cancer Lett ; 575: 216404, 2023 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-37739210

RESUMO

Elevated expression and genetic aberration of IRTKS, also named as BAIAP2L1, have been observed in many tumors, especially in tumor progression. however, the molecular and cellular mechanisms involved in the IRTKS-enhanced tumor progression are obscure. Here we show that higher IRTKS level specifically increases histone H3 lysine 9 trimethylation (H3K9me3) by promoting accumulation of the histone methyltransferase SETDB1. Furthermore, we reveal that IRTKS recruits the deubiquitinase OTUD4 to remove Lys48-linked polyubiquitination at K182/K1050 sites of SETDB1, thus blocking SETDB1 degradation via the ubiquitin-proteasome pathway. Interestingly, the enhanced IRTKS-OTUD4-SETDB1-H3K9me3 axis leads to a general decrease in chromatin accessibility, which inhibits transcription of CDH1 encoding E-cadherin, a key molecule essential for maintaining epithelial cell phenotype, and therefore results in epithelial-mesenchymal transition (EMT) and malignant cell metastasis. Clinically, the elevated IRTKS levels in tumor specimens correlate with SETDB1 levels, but negatively associate with survival time. Our data reveal a novel mechanism for the IRTKS-enhanced tumor progression, where IRTKS cooperates with OTUD4 to enhance SETDB1-mediated H3K9 trimethylation that promotes tumor metastasis via suppressing E-cadherin expression. This study also provides a potential approach to reduce the activity and stability of the known therapeutic target SETDB1 possibly through regulating IRTKS or deubiquitinase OTUD4.

4.
J Clin Biochem Nutr ; 72(2): 139-146, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36936869

RESUMO

M2-type polarization of tumor associated-macrophage (TAM) is involved in the malignancy of gastrointestinal stromal tumor (GIST) progression. ETS variant 1 (ETV1) has been previously validated to regulate GIST pathogenesis. Our study intended to explore the role and mechanism of ETV1 in mediating the M2-polarization of TAM in GIST progression. First, we analyzed the correlation between ETV1 expression and M2-polarization in GIST tissues. IL-4 was used to treat THP-1-derived TAM cells and IL-4-stimulated TAM were co-cultured with GIST-T1 cells to mimic the GIST microenvironment. A loss-of-function assay was performed to explore the role of ETV1. Results showed that ETV1 elevation was positively correlated with M2-polarization. IL-4-induced TAM promoted ETV1 expression, silencing ETV1 inhibited proliferation, invasion and KIT activation in IL-4-treated GIST cells, while cell apoptosis was enhanced. Besides, co-culture of ETV1-silenced GIST cells significantly depressed M2-polarization in TAM, presented as decreased levels of CD206, Agr-1 and cytokines, as well as the proportion of CD206-positive TAM. PDE3A was positively correlated with ETV1 and M2-polarization. Overexpressing PDE3A reversed the inhibitory effects of ETV1 silencing. Generally, ETV1 inhibition depressed M2-polarization of TAM in GIST and its promotion on pathological aggravation via down-regulating PDE3A. This evidence may provide a new target for GIST regulation.

5.
Pathol Res Pract ; 237: 154020, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35868058

RESUMO

Lysine acetyltransferase 7 (KAT7) was upregulated in gastric cancer (GC) patient tissues, and associated with poor prognosis and metastasis. However, its specific role in GC remains unclear. This study aimed to annotate the role of KAT7 in GC cells. The results showed that the overexpression of KAT7 promoted cell growth, migration, and invasion, while KAT7 inhibition has the opposite effect. Besides, KAT7 participated in cell cycle phase distribution and epithelial-mesenchymal transition (EMT) process of GC cells. In addition, KAT7 promoted the transcription and nuclear translocation of Yes-associated protein 1 (YAP1) in MKN45 cells. Silence of YAP1 partly reversed the promoting effect of KAT7 on GC cells progression. In summary, this study indicates that KAT7 promoted GC cells progression through promoting YAP1 activation, contributes to understand the specific role of KAT7 in GC.


Assuntos
Lisina Acetiltransferases , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patologia , Proteínas de Sinalização YAP , Movimento Celular , Regulação Neoplásica da Expressão Gênica/genética , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Proliferação de Células , Lisina Acetiltransferases/metabolismo , Histona Acetiltransferases
6.
Materials (Basel) ; 13(17)2020 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-32887240

RESUMO

Secondary aluminum dross (SAD) is regarded as a solid waste of aluminum recycling process that creates serious environmental and health concerns. However, SAD can also be used as a good source of aluminum, so that utilizing the SAD for the production of valuable products is a promising approach of recycling such waste. In the present work, a novel eco-friendly three-step process was proposed for the synthesis of cryolite (Na3AlF6) from the SAD, and it consisted of (1) water-washing pretreatment of SAD, (2) extraction of Al component via pyro-hydrometallurgy, including low-temperature alkaline smelting, water leaching and purification of leachate in sequence, (3) precipitation of cryolite from the purified NaAlO2 solution using the carbonation method. By analysis of the parameter optimization for each procedure, it was found that the maximum hydrolysis efficiency of aluminum nitride (AlN) in the SAD was around 68.3% accompanied with an extraction efficiency of Al reaching 91.5%. On this basis, the cryolite of high quality was synthesized under the following optimal carbonation conditions: reaction temperature of 75 °C, NaAlO2 concentration of 0.11 mol/L, F/(6Al) molar ratio of 1.10, and 99.99% CO2 gas pressure, and flow rate of 0.2 MPa and 0.5 L/min respectively. The formation of Na3AlF6 phase can be detected by XRD. The morphological feature observed by SEM revealed that the as-synthesized cryolite had a polyhedral shape (~1 µm size) with obvious agglomeration. The chemical composition and ignition loss of the as-synthesized cryolite complied well with the requirements of the Chinese national standard (GB/T 4291-2017).

7.
Cell Mol Biol Lett ; 18(2): 297-314, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23666597

RESUMO

PTPN4 is a widely expressed non-receptor protein tyrosine phosphatase. Although its overexpression inhibits cell growth, the proteins with which it interacts to regulate cell growth are unknown. In this study, we identified CrkI as a PTPN4-interacting protein using a yeast two-hybrid, and confirmed this interaction using in vitro GST pull-down and co-immunoprecipitation and co-localization assays. We further determined the interactional regions as the SH3 domain of CrkI and the proline-rich region between amino acids 462 and 468 of PTPN4. Notably, overexpression of PTPN4 inhibits CrkI-mediated proliferation and wound healing of HEK293T cells, while knockdown of PTPN4 by siRNA in Hep3B cells enhances CrkI-mediated cell growth and motility. Moreover, our data show that ectopic expression of PTPN4 reduces the phosphorylation level of CrkI in HEK293T cells. These findings suggest that PTPN4 negatively regulates cell proliferation and motility through dephosphorylation of CrkI.


Assuntos
Proteína Tirosina Fosfatase não Receptora Tipo 4/metabolismo , Proteínas Proto-Oncogênicas c-crk/metabolismo , Sequência de Aminoácidos , Movimento Celular , Proliferação de Células , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Humanos , Dados de Sequência Molecular , Fosforilação , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 4/química , Interferência de RNA , Reprodutibilidade dos Testes
8.
Nature ; 482(7386): 542-6, 2012 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-22327296

RESUMO

Menin is a tumour suppressor protein whose loss or inactivation causes multiple endocrine neoplasia 1 (MEN1), a hereditary autosomal dominant tumour syndrome that is characterized by tumorigenesis in multiple endocrine organs. Menin interacts with many proteins and is involved in a variety of cellular processes. Menin binds the JUN family transcription factor JUND and inhibits its transcriptional activity. Several MEN1 missense mutations disrupt the menin-JUND interaction, suggesting a correlation between the tumour-suppressor function of menin and its suppression of JUND-activated transcription. Menin also interacts with mixed lineage leukaemia protein 1 (MLL1), a histone H3 lysine 4 methyltransferase, and functions as an oncogenic cofactor to upregulate gene transcription and promote MLL1-fusion-protein-induced leukaemogenesis. A recent report on the tethering of MLL1 to chromatin binding factor lens epithelium-derived growth factor (LEDGF) by menin indicates that menin is a molecular adaptor coordinating the functions of multiple proteins. Despite its importance, how menin interacts with many distinct partners and regulates their functions remains poorly understood. Here we present the crystal structures of human menin in its free form and in complexes with MLL1 or with JUND, or with an MLL1-LEDGF heterodimer. These structures show that menin contains a deep pocket that binds short peptides of MLL1 or JUND in the same manner, but that it can have opposite effects on transcription. The menin-JUND interaction blocks JUN N-terminal kinase (JNK)-mediated JUND phosphorylation and suppresses JUND-induced transcription. In contrast, menin promotes gene transcription by binding the transcription activator MLL1 through the peptide pocket while still interacting with the chromatin-anchoring protein LEDGF at a distinct surface formed by both menin and MLL1.


Assuntos
Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Transcrição Gênica , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cromatina/metabolismo , Cristalografia por Raios X , Fibroblastos , Células HEK293 , Histona-Lisina N-Metiltransferase , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Proteína de Leucina Linfoide-Mieloide/química , Fosforilação , Ligação Proteica , Multimerização Proteica , Proteínas Proto-Oncogênicas c-jun/química , Relação Estrutura-Atividade
9.
Mol Cell Biochem ; 354(1-2): 11-20, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21461611

RESUMO

The epigenetic dysregulation of tumor suppressor genes plays an important role in many cancers, including hepatocellular carcinoma (HCC). In this study, we identified a new gene, family with sequence similarity 43, member B (FAM43B), based on a previous genome-wide approach. FAM43B was significantly downregulated in 60% (24/40) HCC specimens as compared to non-HCC livers. Enforced FAM43B overexpression could suppress cell growth and colony formation in vitro, and induce cell cycle delay, whereas FAM43B knockdown enhanced cell growth. The expression level of FAM43B was found related to the methylation level of FAM43B promoter in HCC cell lines and HCC specimens. The collective data suggest that the expression of FAM43B was regulated by methylation and the epigenetic silencing of FAM43B could contribute to HCC tumorigenesis by regulating cell proliferation.


Assuntos
Carcinoma Hepatocelular/genética , Proliferação de Células , Metilação de DNA , Epigênese Genética , Genes Supressores de Tumor , Neoplasias Hepáticas/genética , Carcinoma Hepatocelular/metabolismo , Ciclo Celular/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/metabolismo , Especificidade de Órgãos , Filogenia , Regiões Promotoras Genéticas , Interferência de RNA
10.
Mol Genet Genomics ; 283(5): 485-92, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20358383

RESUMO

Proliferation of liver cells can be observed in hepatocarcinogenesis, at different stages of liver development, and during liver regeneration after an injury. Does it imply that they share similar molecular mechanisms? Here, the transcriptional profiles of hepatocellular carcinoma (HCC), liver development, and liver regeneration were systematically compared as a preliminary attempt to answer this question. From the comparison, we found that advanced HCC mimics early development in terms of deprived normal liver functions and activated cellular proliferation, but advanced HCC and early development differ in expressions of cancer-related genes and their transcriptional controls. HCC and liver regeneration demonstrate different expression patterns as a whole, but regeneration is similar to dysplasia (pre-stage of HCC) in terms of their proximity to the normal state. In summary, of these three important processes, the carcinogenic progress carries the highest variance in expression; HCC pre-stage shares some resemblance with liver regeneration; and advanced HCC stage displays similarity with early development.


Assuntos
Carcinoma Hepatocelular/genética , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Regeneração Hepática/genética , Fígado/embriologia , Fígado/metabolismo , Animais , Carcinoma Hepatocelular/patologia , Análise por Conglomerados , Perfilação da Expressão Gênica , Fígado/patologia , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Estadiamento de Neoplasias , Análise de Componente Principal , Fatores de Tempo
11.
J Clin Invest ; 120(1): 223-41, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20038795

RESUMO

The epigenetic silencing of tumor suppressor genes is a crucial event during carcinogenesis and metastasis. Here, in a human genome-wide survey, we identified scavenger receptor class A, member 5 (SCARA5) as a candidate tumor suppressor gene located on chromosome 8p. We found that SCARA5 expression was frequently downregulated as a result of promoter hypermethylation and allelic imbalance and was associated with vascular invasion in human hepatocellular carcinoma (HCC). Furthermore, SCARA5 knockdown via RNAi markedly enhanced HCC cell growth in vitro, colony formation in soft agar, and invasiveness, tumorigenicity, and lung metastasis in vivo. By contrast, SCARA5 overexpression suppressed these malignant behaviors. Interestingly, SCARA5 was found to physically associate with focal adhesion kinase (FAK) and inhibit the tyrosine phosphorylation cascade of the FAK-Src-Cas signaling pathway. Conversely, silencing SCARA5 stimulated the signaling pathway via increased phosphorylation of certain tyrosine residues of FAK, Src, and p130Cas; it was also associated with activation of MMP9, a tumor metastasis-associated enzyme. Taken together, these data suggest that the plasma membrane protein SCARA5 can contribute to HCC tumorigenesis and metastasis via activation of the FAK signaling pathway.


Assuntos
Carcinoma Hepatocelular/etiologia , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Neoplasias Hepáticas/etiologia , Receptores Depuradores Classe A/fisiologia , Transdução de Sinais/fisiologia , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Cromossomos Humanos Par 8 , Metilação de DNA , Inativação Gênica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Perda de Heterozigosidade , Camundongos , Camundongos SCID , Invasividade Neoplásica , Regiões Promotoras Genéticas , Receptores Depuradores Classe A/antagonistas & inibidores , Receptores Depuradores Classe A/genética
12.
Biosci Rep ; 30(1): 1-10, 2009 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-19193193

RESUMO

In the present study we describe a novel secreted protein, named C12ORF39 (chromosome 12 open-reading framework 39), which contains a typical amidation/proteolytic processing signal (Gly-Arg-Arg motif). Interestingly, C12ORF39 protein is not hydrolysed, but is a full-length protein without signal peptides. Western blotting indicated that c-Myc-tagged C12ORF39 is secreted into culture medium in transfected HeLa cells. Quantitative RT-PCR (reverse transcription-PCR) analysis revealed that c12orf39 is mainly expressed in placenta and brain. Immunohistochemistry on formalin-fixed paraffin-embedded human term placenta using a rabbit antibody against human C12ORF39 demonstrated that the protein was localized extracellularly, surrounding the trophoblastic cells. In addition, C12ORF39 secretion could be blocked by brefeldin A, suggesting that the secretion of C12ORF39 is dependent on the Golgi apparatus. Furthermore, laser-scanning confocal microscopy also confirmed that the C12ORF39 protein co-localized with the Golgi apparatus. Taken together, although C12ORF39 is not a secreted small peptide, it can also be secreted to play a role in the biological functions of the placenta.


Assuntos
Células Epiteliais/metabolismo , Complexo de Golgi/metabolismo , Hormônios Peptídicos/metabolismo , Placenta/metabolismo , Proteínas/metabolismo , Transdução de Sinais/fisiologia , Amidas/metabolismo , Animais , Brefeldina A/farmacologia , Células COS , Chlorocebus aethiops , Cromossomos Humanos Par 12/genética , Células Epiteliais/ultraestrutura , Espaço Extracelular/metabolismo , Feminino , Complexo de Golgi/ultraestrutura , Células HeLa , Humanos , Microscopia Confocal , Fases de Leitura Aberta/genética , Hormônios Peptídicos/química , Hormônios Peptídicos/genética , Placenta/ultraestrutura , Gravidez , Inibidores da Síntese de Proteínas/farmacologia , Proteínas/química , Proteínas/genética , Trofoblastos/metabolismo , Trofoblastos/ultraestrutura
13.
FEBS Lett ; 582(21-22): 3185-92, 2008 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-18708057

RESUMO

We describe a novel secreted protein, named hOLFML1 (human olfactomedin-like protein 1), with an olfectamine domain in its C-terminus, mainly expressed in the small intestine, liver, lung and heart. Immunohistochemical staining on human small intestine indicated that the protein localizes preferentially in the intestinal villi. Interestingly, ectopic hOLFML1 promoted proliferation of HeLa cells and increased the percentage of cells in S phase. In contrast, knock down of hOLFML1 protein expression by siRNA inhibited cell proliferation and delayed the entry of cells into S phase. Our data also revealed that hOLFML1 is N-glycosylated and its secretion is triggered by serum. Taken together, these findings suggest that hOLFML1 may play a significant role in the regulation of cell proliferation in vitro.


Assuntos
Proliferação de Células , Proteínas da Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Sequência Conservada , Proteínas da Matriz Extracelular/classificação , Proteínas da Matriz Extracelular/genética , Glicoproteínas/classificação , Glicoproteínas/genética , Células HeLa , Humanos , Intestino Delgado/metabolismo , Filogenia , RNA Interferente Pequeno/genética , Distribuição Tecidual
14.
Mol Cell Biochem ; 304(1-2): 297-304, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17541814

RESUMO

HCCA2 (hepatocellular carcinoma-associated gene 2) was initially identified as a HCC (hepatocellular carcinoma)-specific protein and subsequently, a long splice variant of HCCA2 was identified as a co-activator of transcription factor YY1 (Yin Yang 1). To investigate the role of HCCA2 in HCC genesis and progression, we screened a human fetal liver cDNA library and identified a novel HCCA2-interacting protein, MAD2L2 (MAD2 mitotic arrest deficient-like 2 (yeast)). The interaction between HCCA2 and MAD2L2 was confirmed by in vitro and in vivo binding assays and the interaction domain was mapped to the N-terminus of HCCA2 by sequential deletion. HCCA2 and MAD2L2 also colocalized in the nucleus of Hela cells. Furthermore, overexpression of HCCA2 led to cell cycle arrest at G0/G1 phase and therefore inhibited cell proliferation. Our research suggests that HCCA2 may play a novel role in cell cycle regulation.


Assuntos
Proteínas Nucleares/metabolismo , Proteínas/metabolismo , Fatores de Transcrição/metabolismo , Ciclo Celular/genética , Proteínas de Ciclo Celular , Proliferação de Células , Células Cultivadas , Células HeLa , Humanos , Proteínas Mad2 , Proteínas Nucleares/genética , Ligação Proteica , Mapeamento de Interação de Proteínas , Fatores de Transcrição/genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA