Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 14(6): 373, 2023 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-37355626

RESUMO

Phosphodiesterase 4D interacting protein (PDE4DIP) is a centrosome/Golgi protein associated with cyclic nucleotide phosphodiesterases. PDE4DIP is commonly mutated in human cancers, and its alteration in mice leads to a predisposition to intestinal cancer. However, the biological function of PDE4DIP in human cancer remains obscure. Here, we report for the first time the oncogenic role of PDE4DIP in colorectal cancer (CRC) growth and adaptive MEK inhibitor (MEKi) resistance. We show that the expression of PDE4DIP is upregulated in CRC tissues and associated with the clinical characteristics and poor prognosis of CRC patients. Knockdown of PDE4DIP impairs the growth of KRAS-mutant CRC cells by inhibiting the core RAS signaling pathway. PDE4DIP plays an essential role in the full activation of oncogenic RAS/ERK signaling by suppressing the expression of the RAS GTPase-activating protein (RasGAP) neurofibromin (NF1). Mechanistically, PDE4DIP promotes the recruitment of PLCγ/PKCε to the Golgi apparatus, leading to constitutive activation of PKCε, which triggers the degradation of NF1. Upregulation of PDE4DIP results in adaptive MEKi resistance in KRAS-mutant CRC by reactivating the RAS/ERK pathway. Our work reveals a novel functional link between PDE4DIP and NF1/RAS signal transduction and suggests that targeting PDE4DIP is a promising therapeutic strategy for KRAS-mutant CRC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Neoplasias Colorretais , Proteínas do Citoesqueleto , Neurofibromina 1 , Humanos , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Resistencia a Medicamentos Antineoplásicos/genética , Mutação , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo
2.
Aging (Albany NY) ; 14(9): 3801-3812, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35504024

RESUMO

BACKGROUND: Current therapeutic strategies on patients with lymphomas remains limited. Previously we found the suppressive effect of Ophiorrhiza pumila (OPE) on hepatocarcinoma. In present study, the effect of OPE on lymphoma in vitro and in vivo were investigated. METHODS: CCK-8 assay was applied to detect the effect of OPE on cell proliferation. Flow cytometry was used to analyze the effect of OPE on cell cycle distribution and apoptosis. Xenograft mouse model was conducted to determine the anti-tumor activity of OPE. TNUEL assay was performed to detect the apoptosis in tumor tissues. Western blot and immuno-histochemistry were used to determine protein expression. RESULTS: In vitro tests indicate that OPE suppressed A20 cell proliferation in a dose- and time-dependent manner. OPE treatment induced cell cycle arrest at S phase and elevated apoptosis in A20 cells. OPE displayed a significant inhibition in tumor growth in a mouse xenograft model. OPE promoted apoptosis of tumor cell in the mouse model Cleaved caspase 3 expression and Bax/Bcl2 ratio were also enhanced. In addition, OPE suppressed A20 cell viability partially by reducing phosphorylation of EGFR. CONCLUSIONS: Our data showed that OPE suppressed the proliferation of lymphoma cells and promoted apoptosis in vitro and in vivo, which might be partially mediated by inactivating EGFR signaling.


Assuntos
Neoplasias Hepáticas , Linfoma , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Receptores ErbB , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Linfoma/tratamento farmacológico , Camundongos , Camundongos Nus , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Nat Prod Res ; 36(11): 2800-2805, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34018446

RESUMO

Two new hasubanan alkaloids, stephalonester A (1) and stephalonester B (2), together with four known compounds, stephalonine E (3), longanone (4), cephatonine (5), and prostephabyssine (6) were isolated from the whole plant of Stephania longa. Their structures were determined by HR-ESI-MS, 1 D and 2 D NMR, ECD calculations, as well as by comparison with literature values. All compounds were evaluated for their anti-inflammatory activity in vitro. Compounds 4, 5, and 6 exhibited significantly inhibitory effects on TNF-α and IL-6 production with IC50 values range from 6.54 to 30.44 µM.


Assuntos
Alcaloides , Stephania , Alcaloides/química , Alcaloides/farmacologia , Anti-Inflamatórios/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis , Estrutura Molecular , Stephania/química
4.
Oncogene ; 40(45): 6369-6380, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34588619

RESUMO

The therapeutic efficacy of 5-fluorouracil (5-FU) is often reduced by the development of drug resistance. We observed significant upregulation of lipocalin 2 (LCN2) expression in a newly established 5-FU-resistant colorectal cancer (CRC) cell line. In this study, we demonstrated that 5-FU-treated CRC cells developed resistance through LCN2 upregulation caused by LCN2 promoter demethylation and that feedback between LCN2 and NF-κB further amplified LCN2 expression. High LCN2 expression was associated with poor prognosis in CRC patients. LCN2 attenuated the cytotoxicity of 5-FU by activating the SRC/AKT/ERK-mediated antiapoptotic program. Mechanistically, the LCN2-integrin ß3 interaction enhanced integrin ß3 stability, thus recruiting SRC to the cytomembrane for autoactivation, leading to downstream AKT/ERK cascade activation. Targeting LCN2 or SRC compromised the growth of CRC cells with LCN2-induced 5-FU resistance. Our findings demonstrate a novel mechanism of acquired resistance to 5-FU, suggesting that LCN2 can be used as a biomarker and/or therapeutic target for advanced CRC.


Assuntos
Neoplasias Colorretais/patologia , Metilação de DNA , Resistencia a Medicamentos Antineoplásicos , Fluoruracila/farmacologia , Integrina beta3/metabolismo , Lipocalina-2/genética , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Metilação de DNA/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HT29 , Humanos , Integrina beta3/química , Lipocalina-2/metabolismo , Masculino , Camundongos , NF-kappa B/metabolismo , Transplante de Neoplasias , Prognóstico , Regiões Promotoras Genéticas/efeitos dos fármacos , Estabilidade Proteica , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima , Quinases da Família src/metabolismo
5.
Aging (Albany NY) ; 13(8): 11833-11859, 2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33885377

RESUMO

Transcriptome differences between Hodgkin's lymphoma (HL), diffuse large B-cell lymphoma (DLBCL), and mantle cell lymphoma (MCL), which are all derived from B cell, remained unclear. This study aimed to construct lymphoma-specific diagnostic models by screening lymphoma marker genes. Transcriptome data of HL, DLBCL, and MCL were obtained from public databases. Lymphoma marker genes were screened by comparing cases and controls as well as the intergroup differences among lymphomas. A total of 9 HL marker genes, 7 DLBCL marker genes, and 4 MCL marker genes were screened in this study. Most HL marker genes were upregulated, whereas DLBCL and MCL marker genes were downregulated compared to controls. The optimal HL-specific diagnostic model contains one marker gene (MYH2) with an AUC of 0.901. The optimal DLBCL-specific diagnostic model contains 7 marker genes (LIPF, CCDC144B, PRO2964, PHF1, SFTPA2, NTS, and HP) with an AUC of 0.951. The optimal MCL-specific diagnostic model contains 3 marker genes (IGLV3-19, IGKV4-1, and PRB3) with an AUC of 0.843. The present study reveals the transcriptome data-based differences between HL, DLBCL, and MCL, when combined with other clinical markers, may help the clinical diagnosis and prognosis.


Assuntos
Biomarcadores Tumorais/genética , Doença de Hodgkin/diagnóstico , Linfoma Difuso de Grandes Células B/diagnóstico , Linfoma de Célula do Manto/diagnóstico , Modelos Genéticos , Estudos de Casos e Controles , Conjuntos de Dados como Assunto , Diagnóstico Diferencial , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Doença de Hodgkin/genética , Doença de Hodgkin/mortalidade , Humanos , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/mortalidade , Linfoma de Célula do Manto/genética , Linfoma de Célula do Manto/mortalidade , Estadiamento de Neoplasias , Prognóstico , Intervalo Livre de Progressão , Transcriptoma/genética
6.
Cell Biol Int ; 45(5): 948-956, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33377561

RESUMO

Metastasis is a major cause of breast cancer death. MPP7 is a cell polarity controller highly linked to cell migration; however, the function of MPP7 in breast cancer remains unknown. In this study, we reported that MPP7 expression was upregulated in breast cancer tissues and high MPP7 expression predicted poor survival in patients with breast cancer. Ectopic expression of MPP7 markedly enhanced the migration and invasion in breast cancer cells. In contrast, depletion of MPP7 resulted in impaired cell mobility and metastasis. Moreover, we demonstrated that MPP7 exerted its promotional effect via modulation of EMT and activation of the EGFR/AKT cascade. Our study reveals an oncogenic role of MPP7 in breast cancer and suggests that MPP7 may serve as a potential target for exploring novel therapeutic strategies against breast cancer metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Transição Epitelial-Mesenquimal , Receptores ErbB/metabolismo , Feminino , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Proteínas de Membrana/fisiologia , Invasividade Neoplásica/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Transcriptoma/genética
7.
Cell Death Dis ; 11(10): 822, 2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-33009373

RESUMO

Hepatocellular carcinoma (HCC) is a major leading cause of cancer-related death worldwide. Alpha fetoprotein (AFP) is reactivated in a majority of hepatocellular carcinoma (HCC) and associated with poor patient outcomes. Although increasing evidence has shown that AFP can regulate HCC cell growth, the precise functions of AFP in hepatocarcinogenesis and the associated underlying mechanism remain incompletely understood. In this study, we demostrated that depleting AFP significantly suppressed diethylnitrosamine (DEN)-induced liver tumor progression in an AFP gene-deficient mouse model. Similarly, knocking down AFP expression inhibited human HCC cell proliferation and tumor growth by inducing apoptosis. AFP expression level was inversely associated with the apoptotic rate in mouse and human HCC specimens. Investigation of potential cross-talk between AFP and apoptotic signaling revealed that AFP exerted its growth-promoting effect by suppressing the Fas/FADD-mediated extrinsic apoptotic pathway. Mechanistically, AFP bound to the RNA-binding protein HuR, increasing the accumulation of HuR in the cytoplasm and subsequent inhibition of Fas mRNA translation. In addition, we found that inhibiting AFP enhanced the cytotoxicity of therapeutics to AFP-positive HCC cells by activating HuR-mediated Fas/FADD apoptotic signaling. Conclusion: Our study defined the pro-oncogenic role of AFP in HCC progression and uncovered a novel antiapoptotic mechanism connecting AFP to HuR-mediated Fas translation. Our findings suggest that AFP is involved in the pathogenesis and chemosensitivity of HCC and that blockade of AFP may be a promising strategy to treat advanced HCC.


Assuntos
Apoptose/genética , Carcinoma Hepatocelular/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , alfa-Fetoproteínas/genética , Animais , Carcinogênese/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proteína de Domínio de Morte Associada a Fas/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Camundongos , alfa-Fetoproteínas/metabolismo
8.
Chin Med ; 15: 11, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32021647

RESUMO

BACKGROUND: Ophiorrhiza pumila, belonging to the genus Ophiorrhiza (Rubiaceae), is distributed throughout tropical and subtropical Asia. In this study, we evaluated for the first time the anti-proliferation and anti-migration effects of ethanol extract of O. pumila (OPE) on HepG2 and SMMC-7721 cells, and explored the related mechanism. METHODS: OPE was prepared by percolation with 95% ethanol and its main compounds were analyzed by HPLC-MS2. The anti-proliferation effect of OPE was evaluated by the CCK-8 assay and colony formation assay. Cell cycle distribution, apoptosis, and reactive oxygen species (ROS) level were detected by flow cytometry. Migration and invasion abilities were detected by Transwell migration/invasion assays. The expression of correlated proteins was determined using western blotting. RESULTS: A total of 5 tentative compounds were identified from OPE, including pumiloside, deoxypumiloside, camptothecin, aknadinine, and ß-stigmasterol. OPE displayed strong cytostatic effects on HepG2 and SMMC-7721 cells. OPE induced G2/M phase cell cycle arrest, increased apoptosis, and augmented ROS production in these cell lines. In addition, OPE possessed a significant inhibition on cell migration and invasion by reduction of MMP-9 and MMP-2 expression. Moreover, OPE significantly suppressed the phosphorylation of p65. CONCLUSIONS: Our data showed that OPE suppresses liver cancer cell proliferation and migration, which is possibly involved with the inhibition of the NF-κB pathway.

9.
Anticancer Drugs ; 31(8): 819-827, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32097137

RESUMO

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death worldwide. Phytochemicals are important candidates for developing anticancer agents. Ziyuglycoside II is a major active compound of Sanguisorba officinalis, which exhibits antiproliferation activity in several cancers; however, its action in HCC remains unknown. In this study, we investigated the antitumor activity of ziyuglycoside II against HCC and explored the potential mechanisms. We found that ziyuglycoside II exerts significant inhibitory effects on the viability and clonogenic activity of HCC cells. The proliferation repression mediated by ziyuglycoside II was mainly due to increased apoptosis and reactive oxygen species accumulation, as well as a G0/G1 phase cell-cycle arrest. Additionally, ziyuglycoside II markedly impaired HCC cell migration and invasion, two important steps during metastasis, and these suppressive effects may be attributed to the downregulation of matrix metalloproteinases MMP2 and MMP9 expression. Moreover, ziyuglycoside II blocked the epidermal growth factor receptor/nuclear factor kappa-B (EGFR/NF-kB) signaling, which may contribute to its anticancer activity. Taken together, our findings reveal antiproliferative and antimetastasis activities of ziyuglycoside II in HCC cells, implying that ziyuglycoside II might be a promising candidate for the development of novel anti-HCC drugs.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Saponinas/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/secundário , Ciclo Celular , Movimento Celular , Proliferação de Células , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Invasividade Neoplásica , Espécies Reativas de Oxigênio , Células Tumorais Cultivadas
10.
Front Oncol ; 9: 864, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31572672

RESUMO

Animal models play crucial roles in the development of anticancer therapeutics. The ability to quickly assess the localized primary hepatocellular carcinoma (HCC) status in a non-invasive manner would significantly improve the effectiveness of anti-HCC therapeutic studies. However, to date, animal models with this advantage are extremely scarce. In this study, we developed a novel animal model for the fast assessment of drug efficacy against primary HCC in vivo. HCC was induced in immunocompetent hepatocarcinogenesis reporter (HCR) mice by diethylnitrosamine (DEN) injection and confirmed by histopathological staining. Using the bioluminescence imaging (BLI) technique, HCC progression was longitudinally visualized and monitored in a non-invasive way. Tests of two clinical drugs showed that both sorafenib and oxaliplatin significantly inhibited the BLI signal in mouse liver in a dose-dependent manner. The in vivo intensity of BLI signals was highly consistent with the final tumor burden status in mouse liver after drug treatment. The inhibitory effect of anti-HCC drugs was accurately evaluated through in vivo BLI intensity detection. Our study successfully established a bioluminescence mouse model for non-invasive real-time monitoring of HCC therapy, and this HCR mouse model would be a useful tool for potential anti-HCC drug screening and new therapeutic strategy development.

11.
Cell Biol Int ; 43(8): 863-874, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31033093

RESUMO

We had previously identified that the co-expression of transmembrane CXCL16 (TM-CXCL16) and its receptor CXCR6 is an independent risk factor for poor survival in patients with diffuse large B-cell lymphoma (DLBCL). However, the impact of the soluble form of CXCL16 (sCXCL16) on the pathogenesis of DLBCL remains unknown. In the present study, the synergistic effect of sCXCL16 and tumor necrosis factor α (TNF-α) on apoptosis in DLBCL cell lines (OCI-LY8 and OCI-LY10) was investigated in vitro. sCXCL16 reinforced TNF-α-mediated inhibition of DLBCL cell proliferation, as determined by the cell counting kit-8 assay. The results of annexin V staining showed that sCXCL16 enhanced TNF-α-induced apoptosis in OCI-LY8 and OCI-LY10 cells through a death receptor-caspase signaling pathway. The results of gene microarray suggested a significant upregulation of differentially expressed genes in the TNF signaling pathway. sCXCL16 increased the concentration of extracellular TNF-α by binding to CXCR6 to activate the nuclear factor-κB (NF-κB) signaling pathway. TNF-α also induced the secretion of sCXCL16 by increasing the expression of ADAM10, which is known to cleave TM-CXCL16 to yield sCXCL16. Moreover, bioinformatics analysis revealed that elevated TNF-α and ADAM10 expression levels in tumor tissues predicted better survival in patients with DLBCL. Thus, our study suggests that sCXCL16 enhances TNF-α-induced apoptosis of DLBCL cells, which may involve a positive feedback loop consisting of TNF-α, ADAM10, sCXCL16, and members of the NF-κB pathway. sCXCL16 and TNF-α may be used as prognostic markers in the clinic, and their combinational use is a promising approach in the context of DLBCL therapy.


Assuntos
Apoptose , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL16/fisiologia , Linfoma Difuso de Grandes Células B/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Linhagem Celular Tumoral , Humanos , Proteínas de Membrana/metabolismo , NF-kappa B/metabolismo , Receptores CXCR6/metabolismo , Transdução de Sinais
12.
Vaccine ; 36(2): 248-255, 2018 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-29199042

RESUMO

Trichinellosis is a foodborne disease that remains a public health hazard and an economic problem in food safety. Vaccines against the parasite can be an effective way to control this disease; however, commercial vaccines against Trichinella infection are not yet available. Trichinella cathepsin B proteins appear to be promising targets for vaccine development. Here, we reported for the first time the characterization of a novel cDNA that encodes Trichinella spiralis (T. spiralis) cathepsin B-like protease 2 gene (TsCPB2). The recombinant mature TsCPB2 protein was successfully expressed in E. coli system and purified with Ni-affinity chromatography. TsCPB2 expression was detected at all the developmental stages of T. spiralis and it was expressed as an excretory-secretory protein of T. spiralis muscle larvae. Immunization with TsCPB2 antigen induced a combination of humoral and cellular immune responses, which manifested as a mixed Th1/Th2 response, as well as remarkably elevated IgE level. Moreover, vaccination of mice with TsCPB2 that were subsequently challenged with T. spiralis larvae resulted in a 52.3% (P < .001) reduction in worm burden and a 51.2% (P < .001) reduction in muscle larval burden. Our results suggest that TsCPB2 induces protective immunity in Trichinella-infected mice and might be a novel vaccine candidate against trichinellosis.


Assuntos
Antígenos de Helmintos/imunologia , Catepsina B/imunologia , Proteínas Recombinantes/imunologia , Trichinella spiralis/imunologia , Triquinelose/prevenção & controle , Animais , Antígenos de Helmintos/administração & dosagem , Catepsina B/administração & dosagem , Modelos Animais de Doenças , Feminino , Imunidade Celular , Imunidade Humoral , Camundongos Endogâmicos BALB C , Músculos/parasitologia , Carga Parasitária , Proteínas Recombinantes/administração & dosagem , Triquinelose/imunologia
13.
Cancer Lett ; 402: 153-165, 2017 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-28600110

RESUMO

R-spondins play critical roles in development, stem cell survival, and tumorigenicity by modulating Wnt/ß-catenin signaling; however, the role of R-spondins in noncanonical Wnt signaling regulation remains largely unknown. We demonstrate here that R-spondin 2 (RSPO2) has an inhibitory effect on colorectal cancer (CRC) cell migration, invasion, and metastasis. Reduced RSPO2 expression was associated with tumor metastasis and poor survival in CRC patients. The metastasis-suppressive activity of RSPO2 was independent of the Wnt/ß-catenin signaling pathway but dependent on the Fzd7-mediated noncanonical Wnt signaling pathway. The physical interaction of RSPO2 and Fzd7 increased the degradation of cell surface Fzd7 via ZNRF3-mediated ubiquitination, which led to the suppression of the downstream PKC/ERK signaling cascade. In late-stage metastatic cancer, Wnt5a promoted CRC cell migration by preventing degradation of Fzd7, and RSPO2 antagonized Wnt5a-driven noncanonical Wnt signaling activation and tumor cell migration by blocking the binding of Wnt5a to the Fzd7 receptor. Our study reveals a novel RSPO2/Wnt5a-competing noncanonical Wnt signaling mechanism that regulates cellular migration and invasion, and our data suggest that secreted RSPO2 protein could serve as a potential therapy for Wnt5a/Fzd7-driven aggressive CRC tumors.


Assuntos
Movimento Celular , Neoplasias Colorretais/metabolismo , Receptores Frizzled/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Via de Sinalização Wnt , Proteína Wnt-5a/metabolismo , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Animais , Ligação Competitiva , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Receptores Frizzled/genética , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Masculino , Metaloproteinase 7 da Matriz/genética , Metaloproteinase 7 da Matriz/metabolismo , Camundongos Nus , Invasividade Neoplásica , Ligação Proteica , Proteína Quinase C/metabolismo , Estabilidade Proteica , Proteólise , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-ets , Interferência de RNA , Fatores de Tempo , Transfecção , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
14.
Mol Cell Proteomics ; 16(3): 469-484, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28087594

RESUMO

Toxoplasma kinase ROP18 is a key molecule responsible for the virulence of Toxoplasma gondii; however, the mechanisms by which ROP18 exerts parasite virulence via interaction with host proteins remain limited to a small number of identified substrates. To identify a broader array of ROP18 substrates, we successfully purified bioactive mature ROP18 and used it to probe a human proteome array. Sixty eight new putative host targets were identified. Functional annotation analysis suggested that these proteins have a variety of functions, including metabolic process, kinase activity and phosphorylation, cell growth, apoptosis and cell death, and immunity, indicating a pleiotropic role of ROP18 kinase. Among these proteins, four candidates, p53, p38, UBE2N, and Smad1, were further validated. We demonstrated that ROP18 targets p53, p38, UBE2N, and Smad1 for degradation. Importantly, we demonstrated that ROP18 phosphorylates Smad1 Ser-187 to trigger its proteasome-dependent degradation. Further functional characterization of the substrates of ROP18 may enhance understanding of the pathogenesis of Toxoplasma infection and provide new therapeutic targets. Similar strategies could be used to identify novel host targets for other microbial kinases functioning at the pathogen-host interface.


Assuntos
Análise Serial de Proteínas/métodos , Proteínas Serina-Treonina Quinases/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Linhagem Celular , Células HEK293 , Células HeLa , Humanos , Anotação de Sequência Molecular , Fosforilação , Mapas de Interação de Proteínas , Proteínas de Protozoários , Proteína Smad1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo
15.
Cancer Lett ; 390: 48-57, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28093285

RESUMO

Ataxia-telangiectasia mutated (ATM) protein kinase is a major guardian of genomic stability, and its well-established function in cancer is tumor suppression. Here, we report an oncogenic role of ATM. Using two isogenic sets of human colon cancer cell lines that differed only in their ATM status, we demonstrated that ATM deficiency significantly inhibits cancer cell proliferation, migration, and invasion. The tumor-suppressive function of ATM depletion is not modulated by the compensatory activation of ATR, but it is associated with B56γ2-mediated Chk1/p53/CD44 signaling pathways. Under normal growth conditions, the depletion of ATM prevents B56γ2 ubiquitination and degradation, which activates PP2A-mediated Chk1/p53/p21 signaling pathways, leading to senescence and cell cycle arrest. CD44 was validated as a novel ATM target based on its ability to rescue cell migration and invasion defects in ATM-depleted cells. The activation of p53 induced by ATM depletion suppresses CD44 transcription, thus resulting in epithelial-mesenchymal transition (EMT) and cell migration suppression. Our study suggests that ATM has tumorigenic potential in post-formed colon neoplasia, and it supports ATM as an appealing target for improving cancer therapy.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas Sanguíneas/metabolismo , Quinase 1 do Ponto de Checagem/metabolismo , Neoplasias do Colo/fisiopatologia , Receptores de Hialuronatos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/genética , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/genética , Imunofluorescência , Deleção de Genes , Humanos , Modelos Biológicos , Transdução de Sinais
16.
Lasers Med Sci ; 31(5): 929-36, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27059226

RESUMO

The facial flat wart (verruca plana) is one of the most common reasons for dermatology and primary care visits. Although there are many therapeutic modalities, no single therapy has been proven to be completely curative. Case reports and uncontrolled studies suggested that photodynamic therapy (PDT) with topical 5-aminolevulinic acid (ALA) can effectively treat recalcitrant facial flat warts. A total of 12 patients with recalcitrant facial flat warts were enrolled in the study. ALA gel (10 %) was applied topically to lesions and incubated for 3 h. The lesions were irradiated by an LED light of 630 ± 10 nm at dose levels of 60-100 mW/cm. Clinical assessment was conducted before and after every treatment for up to 24 weeks. Among the ten patients completing three sessions of ALA-PDT, five had complete lesions clearance, and the other five patients were significantly improved. At the 24-week follow-up, the average effective rate was 88.8 %, with no recurrences. No significant side effects were reported. A low-dose topical ALA-PDT regimen using 10 % ALA, 3 h incubation, and a red light source for three treatment sessions are suggested as the optimal scheme for the treatment of recalcitrant flat warts on the face in Chinese patients. Superior efficacy is found in elevated or active period lesions with mild side effects.


Assuntos
Ácido Aminolevulínico/uso terapêutico , Face , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/uso terapêutico , Verrugas/tratamento farmacológico , Adulto , Povo Asiático , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva
17.
World J Gastroenterol ; 21(36): 10375-84, 2015 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-26420964

RESUMO

AIM: To investigate the effects of Recql5 deficiency on liver injury induced by lipopolysaccharide/D-galactosamine (LPS/D-Gal). METHODS: Liver injury was induced in wild type (WT) or Recql5-deficient mice using LPS/D-Gal, and assessed by histological, serum transaminases, and mortality analyses. Hepatocellular apoptosis was quantified by transferase dUTP nick end labeling assay and Western blot analysis of cleaved caspase-3. Liver inflammatory chemokine and cytochrome P450 expression was analyzed by quantitative reverse transcription-PCR. Neutrophil infiltration was evaluated by myeloperoxidase activity. Expression and phosphorylation of ERK, JNK, p65, and H2A.X was determined by Western blot. Oxidative stress was evaluated by measuring malondialdehyde production and nitric oxide synthase, superoxide dismutase, glutathione peroxidase, catalase, and glutathione reductase activity. RESULTS: Following LPS/D-Gal exposure, Recql5-deficient mice exhibited enhanced liver injury, as evidenced by more severe hepatic hemorrhage, higher serum aspartate transaminase and alanine transaminase levels, and lower survival rate. As compared to WT mice, Recql5-deficient mice showed an increased number of apoptotic hepatocytes and higher cleaved caspase-3 levels. Recql5-deficient mice exhibited increased DNA damage, as evidenced by increased γ-H2A.X levels. Inflammatory cytokine levels, neutrophil infiltration, and ERK phosphorylation were also significantly increased in the knockout mice. Additionally, Recql5-deficient mice exhibited increased malondialdehyde production and elevated inducible nitric oxide synthase, superoxide dismutase, glutathione peroxidase, catalase, and glutathione reductase activity, indicative of enhanced oxidative stress. Moreover, CYP450 expression was significantly downregulated in Recql5-deficient mice after LPS/D-Gal treatment. CONCLUSION: Recql5 protects the liver against LPS/D-Gal-induced injury through suppression of hepatocyte apoptosis and oxidative stress and modulation of CYP450 expression.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Galactosamina , Hepatócitos/enzimologia , Lipopolissacarídeos , Fígado/enzimologia , RecQ Helicases/metabolismo , Animais , Apoptose , Caspase 3/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/patologia , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Citocinas/genética , Citocinas/metabolismo , Dano ao DNA , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hepatócitos/patologia , Mediadores da Inflamação/metabolismo , Fígado/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos , Estresse Oxidativo , Fosforilação , RecQ Helicases/deficiência , RecQ Helicases/genética , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
18.
Nat Commun ; 5: 3149, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24476626

RESUMO

R-spondins are a family of secreted Wnt agonists. One of the family members, R-spondin 2 (RSPO2), has an important role in embryonic development, bone formation and myogenic differentiation; however, its role in human cancers remains largely unknown. Here we show that RSPO2 expression is downregulated in human colorectal cancers (CRCs) due to promoter hypermethylation, and that the RSPO2 reduction correlates with tumour differentiation, size and metastasis. Overexpression of RSPO2 suppresses CRC cell proliferation and tumorigenicity, whereas the depletion of RSPO2 enhances tumour cell growth. RSPO2 has an inhibitory effect on Wnt/ß-catenin signaling in the CRC cells that show suppressed cell proliferation. In human CRC cells, the RSPO2-induced inhibition of Wnt signaling depends on leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5); RSPO2 interacts with LGR5 to stabilize the membrane-associated zinc and ring finger 3 (ZNRF3). Our data suggest that RSPO2 functions as a tumour suppressor in human CRCs, and these data reveal a RSPO2-induced, LGR5-dependent Wnt signaling-negative feedback loop that exerts a net growth-suppressive effect on CRC cells.


Assuntos
Neoplasias Colorretais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/patologia , Regulação para Baixo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
19.
PLoS One ; 9(1): e84874, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24416302

RESUMO

Although class IX myosins are single-headed, they demonstrate characteristics of processive movement along actin filaments. Double-headed myosins that move processively along actin filaments achieve this by successive binding of the two heads in a hand-over-hand mechanism. This mechanism, obviously, cannot operate in single-headed myosins. However, it has been proposed that a long class IX specific insertion in the myosin head domain at loop2 acts as an F-actin tether, allowing for single-headed processive movement. Here, we tested this proposal directly by analysing the movement of deletion constructs of the class IX myosin from Caenorhabditis elegans (Myo IX). Deletion of the large basic loop2 insertion led to a loss of processive behaviour, while deletion of the N-terminal head extension, a second unique domain of class IX myosins, did not influence the motility of Myo IX. The processive behaviour of Myo IX is also abolished with increasing salt concentrations. These observations directly demonstrate that the insertion located in loop2 acts as an electrostatic actin tether during movement of Myo IX along the actin track.


Assuntos
Actinas/metabolismo , Movimento , Mutagênese Insercional , Miosinas/química , Miosinas/metabolismo , Eletricidade Estática , Citoesqueleto de Actina/metabolismo , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Miosinas/genética , Concentração Osmolar , Estrutura Terciária de Proteína , Pontos Quânticos
20.
World J Gastroenterol ; 19(40): 6814-24, 2013 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-24187456

RESUMO

AIM: To investigate the effects of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) deficiency on the cytotoxicity of chemotherapeutic agents toward colorectal cancer cells. METHODS: PTEN-deficient colorectal cancer (CRC) cells were generated by human somatic cell gene targeting using the adeno-associated virus system. The cytotoxic effects of compounds including curcumin, 5-fluorouracil (5-FU), dihydroartemisinin (DHA), irinotecan (CPT-11) and oxaliplatin (OXA) on cancer cells were determined using the MTT assay. Enhanced cytotoxicity of curcumin in PTEN-deficient CRC cells was observed, and this was confirmed using clonogenic assays. Apoptosis and cell cycle progression were analyzed by flow cytometry. Levels of apoptosis and cell cycle-related proteins were examined by Western blotting. RESULTS: We developed an isogenic set of CRC cell lines that differed only in their PTEN status. Using this set of cell lines, we found that disruption of the PTEN gene had no effect on the sensitivity of CRC cells to 5-FU, CPT-11, DHA, or OXA, whereas PTEN disruption increased the sensitivity of CRC cells to curcumin. Loss of PTEN did not alter the curcumin-induced apoptosis in CRC cells. However, PTEN deficiency led to an altered pattern of curcumin-mediated cell cycle arrest. In HCT116 PTEN (+/+) cells, curcumin caused a G2/M phase arrest, whereas it caused a G0/G1 phase arrest in HCT116 PTEN (-/-) cells. Levels of cell cycle-related proteins were consistent with these respective patterns of cell cycle arrest. CONCLUSION: Curcumin shows enhanced cytotoxicity toward PTEN-deficient cancer cells, suggesting that it might be a potential chemotherapeutic agent for cancers harboring PTEN mutations.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias Colorretais/enzimologia , Curcumina/farmacologia , PTEN Fosfo-Hidrolase/deficiência , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Relação Dose-Resposta a Droga , Regulação para Baixo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Células HCT116 , Humanos , PTEN Fosfo-Hidrolase/genética , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...