Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell Mol Life Sci ; 79(10): 537, 2022 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-36183290

RESUMO

Contact-based pericellular interactions play important roles in cancer progression via juxtacrine signaling pathways. The present study revealed that hypoxia-inducible factor-1α (HIF-1α), induced even in non-hypoxic conditions by cell-to-cell contact, was a critical cue responsible for the malignant characteristics of glioblastoma multiforme (GBM) cells through Notch1 signaling. Densely cultured GBM cells showed enhanced viability and resistance to temozolomide (TMZ) compared to GBM cells at a low density. Ablating Notch1 signaling by a γ-secretase inhibitor or siRNA transfection resensitized resistant GBM cells to TMZ treatment and decreased their viability under dense culture conditions. The expression of HIF-1α was significantly elevated in highly dense GBM cells even under non-hypoxic conditions. Atypical HIF-1α expression was associated with the Notch1 signaling pathway in both GBM and glioblastoma stem cells (GSC). Proteasomal degradation of HIF-1α was prevented by binding with Notch1 intracellular domain (NICD), which translocated to the nuclei of GBM cells. Silencing Notch1 signaling using a doxycycline-inducible Notch1 RNA-interfering system or treatment with chetomin, a HIF pathway inhibitor, retarded tumor development with a significant anti-cancer effect in a murine U251-xenograft model. Using GBM patient tissue microarray analysis, a significant increase in HIF-1α expression was identified in the group with Notch1 expression compared to the group without Notch1 expression among those with positive HIF-1α expression. Collectively, these findings highlight the critical role of cell-to-cell contact-dependent signaling in GBM progression. They provide a rationale for targeting HIF-1α signaling even in a non-hypoxic microenvironment.


Assuntos
Glioblastoma , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Linhagem Celular Tumoral , Doxiciclina , Glioblastoma/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , RNA Interferente Pequeno/genética , Receptor Notch1/genética , Transdução de Sinais , Temozolomida , Microambiente Tumoral
2.
Biochem Biophys Res Commun ; 599: 100-105, 2022 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-35180468

RESUMO

BACKGROUND: Luffa cylindrica stem sap (LuCS) has been ethnopharmacologically used as a cosmetic ingredients to improve the facial condition in Asians, but there is no scientific proof about the advantages of LuCS as a supplement for skin elasticity inducer. PURPOSE: Presently, we have validated the beneficial effect of LuCS in human preadipocyte and fibroblast. METHODS: In vitro activities of LuCS on expression of cellular elastin and collagen type I were validated using Western blot analysis in human fibroblasts. Effect of LuCS on preadipocyte development was performed using MDI medium containing isobutyl-methylxanthine, dexamethasone, and insulin and then evaluated using oil red O staining. RESULTS: Treatment of LuCS stimulated the expression of cellular elastin and type I procollagen in human skin fibroblasts. Exposure to LuCS induced lipid accumulation of preadipocytes via activation of CEBP/α signaling pathway in preadipocytes. Expression of collagen I, elastin, or CEBP/α mRNA was decreased by age. 3-bromo-3-methylisoxazol-5-amine enhanced the synthesis of cellular lipid in preadipocytes. CONCLUSIONS: Collectively, these results suggest the rationale of LuCS treatment in enhancing the skin condition.


Assuntos
Adipócitos/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Luffa/química , Extratos Vegetais/farmacologia , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Elastina/genética , Elastina/metabolismo , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Extratos Vegetais/química , Pró-Colágeno/genética , Pró-Colágeno/metabolismo
3.
Biochem Biophys Res Commun ; 580: 41-47, 2021 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-34619551

RESUMO

Luffa cylindrica stem sap (LuCS) has been traditionally used as a facial cosmetic supplement to enhance the skin condition of Asians. However, LuCS has yet to be described and there is no solid scientific evidence regarding the use of LuCS as an anti-wrinkle agent. In the present study, we have evaluated the functional effect of LuCS and its underlying mechanisms based on scientific evidence. Treatment with LuCS stimulated the growth and migration of human skin fibroblasts. LuCS treatment activated EGFR signaling via the enhanced expression of EGFR and down-regulation of PPARγ in human skin fibroblasts. Exposure to LuCS induced the synthesis of cellular type I procollagen and elastin in consort with the down-regulation of various proteinases including MMP-1, -2 and -9 in human skin fibroblasts. LuCS treatment also reversed the skin damage induced by UV-A irradiation in human skin fibroblasts. 3-bromo-3-methylisoxazol-5-amine was identified as the functional component using UPLC-MS-MS analysis and increased production of cellular type I procollagen. Collectively, these results suggest the efficacy of LuCS supplementation in improving the skin condition via anti-wrinkle effect.


Assuntos
Fibroblastos/efeitos dos fármacos , Luffa , Extratos Vegetais/farmacologia , Substâncias Protetoras/farmacologia , Envelhecimento da Pele/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Fibroblastos/citologia , Fibroblastos/efeitos da radiação , Humanos , Luffa/química , Extratos Vegetais/química , Caules de Planta/química , Substâncias Protetoras/química
4.
Tumour Biol ; 37(9): 12193-12202, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27230679

RESUMO

The prognosis of pancreatic cancer has not improved despite considerable and continuous effort. Dual-specificity phosphatase 28 (DUSP28) is highly expressed in human pancreatic cancers and exerts critical effects. However, knowledge of its function in pancreatic cancers is extremely limited. Here, we demonstrate the peculiar role of DUSP28 in pancreatic cancers. Analysis using the Gene Expression Omnibus public microarray database indicated higher DUSP28, MUC1, MUC4, MUC5B, MUC16 and MUC20 messenger RNA (mRNA) levels in pancreatic cancers compared with normal pancreas tissues. DUSP28 expression in human pancreatic cancer correlated positively with those of MUC1, MUC4, MUC5B, MUC16 and MUC20. In contrast, there were no significant correlations between DUSP28 and mucins in normal pancreas tissues. Decreased DUSP28 expression resulted in down-regulation of MUC5B and MUC16 at both the mRNA and protein levels; furthermore, transfection with small interfering RNA (siRNA) for MUC5B and MUC16 inhibited the migration and survival of AsPC-1 cells. In addition, transfection of siRNA for MUC5B and MUC16 resulted in a significant decrease in phosphorylation of FAK and ERK1/2 compared with transfection with scrambled-siRNA. These results collectively indicate unique links between DUSP28 and MUC5B/MUC16 and their roles in pancreatic cancer; moreover, they strongly support a rationale for targeting DUSP28 to inhibit development of malignant pancreatic cancer.


Assuntos
Antígeno Ca-125/genética , Movimento Celular/genética , Fosfatases de Especificidade Dupla/genética , Regulação Neoplásica da Expressão Gênica , Mucina-5B/genética , Neoplasias Pancreáticas/genética , Western Blotting , Antígeno Ca-125/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Fosfatases de Especificidade Dupla/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Perfilação da Expressão Gênica/métodos , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mucina-5B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Fosforilação , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
J Neurooncol ; 128(1): 29-34, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26951556

RESUMO

Glioblastoma multiforme (GBM) is the most common and aggressive type of primary brain tumor with grave prognosis. Despite the growing understanding of the complex signaling networks responsible for the initiation and progression of GBM, many experimental therapies have fallen short of their treatment goals. In the present study, we investigated the novel molecular mechanisms responsible for synergistic action of temozolomide (TMZ) and anti-VEGF therapy in GBM cells. We tested the combined effects of TMZ and VEGF blockade in four human GBM cell lines: TMZ-sensitive U251-MG and U373-MG cells, and TMZ-resistant CRT-MG and LN215-MG cells, which correlated with MGMT promoter methylation status. Treatment of TMZ along with a sublethal dosage range of SU1498, a chemical inhibitor of the VEGF receptor signaling, induced significant cell death in both TMZ-sensitive and TMZ-resistant GBM cells without changing the status of the MGMT promoter methylation. Treatment with TMZ specifically reduced the expression of NRP-1, a coreceptor of VEGF but not those of VEGF-R1 and VEGF-R2. We further confirmed the key role of NRP-1 by showing that the reduction of NRP-1 by siRNA also increased the SU1498-induced cytotoxicity of LN215-MG. These results collectively indicate that combined treatment of TMZ can sensitize GBM cells to blockade of autocrine VEGF signaling through specific down-regulation of NRP-1, which provide a rationale for further evaluation and a potential clinical trial of combinatorial therapy of TMZ and SU1498 or other VEGF inhibitors for intractable brain tumors.


Assuntos
Antineoplásicos/farmacologia , Cinamatos/farmacologia , Dacarbazina/análogos & derivados , Glioblastoma/tratamento farmacológico , Neuropilina-1/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Metilação de DNA , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Dacarbazina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Neuropilina-1/genética , Neuropilina-1/metabolismo , Regiões Promotoras Genéticas , RNA Interferente Pequeno , Temozolomida , Proteínas Supressoras de Tumor/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
Tumour Biol ; 36(12): 9385-93, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26109002

RESUMO

Pancreatic cancer is one of the most dangerous cancers and is associated with a grave prognosis. Despite increased knowledge of the complex signaling networks responsible for progression of pancreatic cancer, many challenging therapies have fallen short of expectations. In this study, we examined the anti-migratory effect of quercetin 3-O-glucoside in epidermal growth factor-induced cell migration by inhibiting EGF receptor (EGFR) signaling in several human pancreatic cancer cell lines. Treatment with quercetin, quercetin 3-O-glucoside, and quercetin 7-O-glucoside differentially suppressed epidermal growth factor-induced migration activity of human pancreatic cancer cells. In particular, quercetin 3-O-glucoside strongly inhibited the infiltration activity of pancreatic cancer cells in a dose-dependent manner. Furthermore, quercetin 3-O-glucoside exerted the anti-migratory effect even at a relatively low dose compared with other forms of quercetin. The anti-tumor effects of quercetin 3-O-glucoside were mediated by selectively inhibiting the EGFR-mediated FAK, AKT, MEK1/2, and ERK1/2 signaling pathway. Combinatorial treatment with quercetin 3-O-glucoside plus gemcitabine showed the synergistic anti-migratory effect on epidermal growth factor-induced cell migration in human pancreatic cancer cell lines. These results suggest that quercetin 3-O-glucoside has potential for anti-metastatic therapy in human pancreatic cancer.


Assuntos
Fator de Crescimento Epidérmico/genética , Receptores ErbB/genética , Flavonoides/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Glucosídeos/administração & dosagem , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Fosforilação , Quercetina/administração & dosagem , Quercetina/análogos & derivados
7.
Biochem Biophys Res Commun ; 443(4): 1218-25, 2014 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-24388986

RESUMO

Gr-1(+)CD11b(+) cells can suppress innate and adaptive immunity, and the functional immunosuppressive characteristics of these cells can be modulated by the tumor microenvironment. Since Gr-1(+)CD11(+) cells are also involved in tumor-associated angiogenesis, we hypothesized that the angiogenic nature of Gr-1(+)CD11b(+) cells could be regulated by the tumor milieu. To address this hypothesis, we imitated a tumor microenvironment by exposing Gr-1(+)CD11b(+) cells isolated from spleen of 4T1 mammary carcinoma-bearing mice to tumor-conditioned medium. Supernatants from tumor-conditioned Gr-1(+)CD11b(+) cells significantly induced capillary-like tube formation and migration of human umbilical vein endothelial cells (HUVECs) compared to naive Gr-1(+)CD11b(+) cells. Incubation of Gr-1(+)CD11b(+) cells with tumor-conditioned medium induced production of pro-angiogenic chemokines CCL2 and CXCL16. Pretreatment with an anti-CCL2 antibody, but not an anti-CXCL16 antibody, suppressed the angiogenic effects of tumor-conditioned Gr-1(+)CD11b(+) cells on HUVECs. Simultaneous neutralization of CCL2 and CXCL16 significantly inhibited tube formation and migration of HUVECs compared to the sole neutralization against CCL2. Supernatants from tumor-conditioned Gr-1(+)CD11b(+) cells induced phosphorylation of ERK1/2 in HUVECs, and inhibition of the ERK pathway blocked angiogenic effects. ERK pathway activity was partially abrogated by neutralization of CCL2 and more suppressed by simultaneous neutralization of CCL2 and CXCL16. These results collectively indicate that CCL2 and CXCL16 chemokines produced by tumor-conditioned Gr-1(+)CD11b(+) myeloid cells synergistically induce angiogenesis in vitro by stimulating the ERK1/2 signaling pathway. Thus, regulation of Gr-1(+)CD11b(+) cells in the tumor microenvironment may contribute to angiogenesis through the secretion of pro-angiogenic chemokines.


Assuntos
Quimiocina CCL2/metabolismo , Quimiocina CXCL6/metabolismo , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/imunologia , Células Mieloides/imunologia , Neovascularização Patológica/imunologia , Animais , Antígeno CD11b/metabolismo , Linhagem Celular Tumoral , Quimiocina CCL2/antagonistas & inibidores , Quimiocina CXCL16 , Quimiocina CXCL6/antagonistas & inibidores , Meios de Cultivo Condicionados , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Tolerância Imunológica , Sistema de Sinalização das MAP Quinases , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/classificação , Células Mieloides/metabolismo , Neovascularização Patológica/metabolismo , Testes de Neutralização , Receptores de Quimiocinas/metabolismo , Microambiente Tumoral/imunologia
8.
BMB Rep ; 57(7): 324-329, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38649146

RESUMO

The aim of the present study is to provide a rational background for silencing the V-set and transmembrane domain containing 2 like (VSTM2L) in consort with recognising soluble VSTM2L against cholangiocarcinoma. A therapeutic target against cholangiocarcinoma was selected using iterative patient partitioning (IPP) calculation, and it was verified by in vitro and in silico analyses. VSTM2L was selected as a potential therapeutic target against cholangiocarcinoma. Silencing the VSTM2L expression significantly attenuated the viability and survival of cholangiocarcinoma cells through blockade of the intracellular signalling pathway. In silico analysis showed that VSTM2L affected the positive regulation of cell growth in cholangiocarcinoma. Liptak's z value revealed that the expression of VSTM2L worsened the prognosis of cholangiocarcinoma patients. In addition, soluble VSTM2L was significantly detected in the whole blood of cholangiocarcinoma patients compared with that of healthy donors. Our report reveals that VSTM2L might be the potential therapeutic target and a soluble prognostic biomarker against cholangiocarcinoma. [BMB Reports 2024; 57(7): 324-329].


Assuntos
Neoplasias dos Ductos Biliares , Biomarcadores Tumorais , Colangiocarcinoma , Colangiocarcinoma/metabolismo , Colangiocarcinoma/diagnóstico , Humanos , Biomarcadores Tumorais/metabolismo , Prognóstico , Neoplasias dos Ductos Biliares/metabolismo , Neoplasias dos Ductos Biliares/patologia , Neoplasias dos Ductos Biliares/diagnóstico , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Transdução de Sinais , Masculino , Proteínas de Membrana/metabolismo
9.
J Med Food ; 26(8): 595-604, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37594560

RESUMO

Acorn (Quercus acutissima CARR.) has been used in traditional food and medicinal ethnopharmacology in Asia, and it has shown multifarious functions such as antidementia, antiobesity, and antiasthma functions. However, there is limited scientific evidence about the efficacy of acorn for ameliorating skin problems. Treatment with ethanol-extracted acorns (EeA's) ablated the expression of inducible nitric oxide synthase (iNOS), cyclooxygenase 2 (COX2), monocyte chemoattractant protein-1 (MCP-1), and interleukin (IL)-8 stimulated by tumor necrosis factor (TNF)-α in human adult low calcium high temperature (HaCaT) cells under sublethal dosages. In addition, treatment with EeA dose dependently inhibited the ex vivo hyper keratin formation induced by TNF-α in HaCaT cells in conjunction with the blockade of cytokeratin-1 (CK-1) and cytokeratin-5 (CK-5) expression. Moreover, EeA treatment stimulated the expression of hyaluronic acid (HA) expression in human fibroblasts in a dose-dependent manner. Linoleamide was identified as the functional component of EeA using preparative high-performance liquid chromatography and ultra high performance liquid chromatography-mass spectrometry-mass spectrometry analysis, and the anti-inflammatory features and enhanced HA expression were verified. Collectively, these results suggest the efficacy of EeA supplementation in improving skin problems via anti-inflammation and upregulating HA production.


Assuntos
Ácido Hialurônico , Quercus , Adulto , Humanos , Queratinócitos , Células HaCaT , Etanol
10.
BMB Rep ; 56(11): 594-599, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37357538

RESUMO

A number of therapeutic drugs have been developed from functional chemicals found in plants. Knowledge of plants used for medicinal purposes has historically been transmitted by word of mouth or through literature. The aim of the present study is to provide a systemic platform for the development of lead compounds against breast cancer based on a traditional medical text. To verify our systematic approach, integrating processes consisted of text mining of traditional medical texts, 3-D virtual docking screening, and in vitro and in vivo experimental validations were demonstrated. Our text analysis system identified rutin as a specific phytochemical traditionally used for cancer treatment. 3-D virtual screening predicted that rutin could block EGFR signaling. Thus, we validated significant anticancer effects of rutin against breast cancer cells through blockade of EGFR signaling pathway in vitro. We also demonstrated in vivo anti-cancer effects of rutin using the breast cancer recurrence in vivo models. In summary, our innovative approach might be proper for discovering new phytochemical lead compounds designing for blockade of malignant neoplasm including breast cancer. [BMB Reports 2023; 56(11): 594-599].


Assuntos
Neoplasias da Mama , Plantas Medicinais , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Plantas Medicinais/química , Compostos Fitoquímicos , Transdução de Sinais , Receptores ErbB
11.
Cell Oncol (Dordr) ; 45(2): 275-291, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35357655

RESUMO

BACKGROUND: Cholangiocarcinoma is a devastating cancer with a poor prognosis. Previous reports have presented conflicting results on the role of transforming growth factor-ß-induced protein (TGFBI) in malignant cancers. Currently, our understanding of the role of TGFBI in cholangiocarcinoma is ambiguous. The aim of the present study was to investigate the role of TGFBI in human cholangiocarcinoma. METHODS: Iterative patient partitioning (IPP) scoring and consecutive elimination methods were used to select prognostic biomarkers. mRNA and protein expression levels were determined using Gene Expression Omnibus (GEO), Western blot and ELISA analyses. Biological activities of selected biomarkers were examined using both in vitro and in vivo assays. Prognostic values were assessed using Kaplan-Meier and Liptak's z score analyses. RESULTS: TGFBI was selected as a candidate cholangiocarcinoma biomarker. GEO database analysis revealed significantly higher TGFBI mRNA expression levels in cholangiocarcinoma tissues compared to matched normal tissues. TGFBI protein was specifically detected in a soluble form in vitro and in vivo. TGFBI silencing evoked significant anti-cancer effects in vitro. Soluble TGFBI treatment aggravated the malignancy of cholangiocarcinoma cells both in vitro and in vivo through activation of the integrin beta-1 (ITGB1) dependent PPARγ signalling pathway. High TGFBI expression was associated with a poor prognosis in patients with cholangiocarcinoma. CONCLUSIONS: Our data suggest that TGFBI may serve as a promising prognostic biomarker and therapeutic target for cholangiocarcinoma.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Neoplasias dos Ductos Biliares/genética , Ductos Biliares Intra-Hepáticos/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Colangiocarcinoma/genética , Proteínas da Matriz Extracelular , Humanos , Integrina beta1 , PPAR gama , RNA Mensageiro/genética , Fator de Crescimento Transformador beta/metabolismo
12.
Int J Oncol ; 61(4)2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36043525

RESUMO

Despite the high expression of neuropilin­1 (NRP­1) in human glioblastoma (GB), the understanding of its function as a co­receptor of vascular endothelial growth factor receptors (VEGFRs) in angiogenesis is currently limited. Therefore, the aim of the present study was to elucidate the non­classical function of NRP­1 expression in human GB. Expression patterns of NRP­1 and VEGF­A were determined by sandwich ELISA, western blot analysis, or immunohistochemistry. Differential dependency of GB cells following ablation of VEGF­A signaling was validated in vitro and in vivo. Cellular mechanism responsible for distinct response to VEGF­A signaling was evaluated by western blotting and immunoprecipitation analysis. Prognostic implications were assessed using IHC analysis. GB cells exhibited differing sensitivity to silencing of vascular endothelial growth factor (VEGF)­A signaling, which resulted in a distinct expression pattern of wild­type or chondroitin­sulfated NRP­1. VEGF­A­sensitive GB exhibited the physical interaction between wild­type NRP­1 and FMS related receptor tyrosine kinase 1 (Flt­1) whereas VEGF­A­resistant GB exhibited chondroitin­sulfated NRP­1 without interaction with Flt­1. Eliminating the chondroitin sulfate modification in NRP­1 led to re­sensitization to VEGF­A signaling, and chondroitin sulfate modification was found to be associated with an adverse prognosis in patients with GB. The present study identified the distinct functions of NRP­1 in VEGF­A signaling in accordance with its unique expression type and interaction with Flt­1. The present research is expected to provide a strong basis for targeting VEGF­A signaling in patients with GB, with variable responses.


Assuntos
Glioblastoma , Neuropilina-1 , Fator A de Crescimento do Endotélio Vascular , Sulfatos de Condroitina , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Neuropilina-1/genética , Neuropilina-1/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Anticancer Res ; 42(8): 3789-3798, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35896263

RESUMO

BACKGROUND/AIM: Cholangiocarcinoma remains one of the most dangerous types of cancer. Eriodictyol is a well-known flavonoid having effective bioactivity against various malignant tumor types. However, the anticancer effect of eriodictyol against cholangiocarcinoma remains ambiguous. Thus, the aim of the present study was to investigate the effects of eriodictyol on human cholangiocarcinoma. MATERIALS AND METHODS: The biological effects of eriodictyol were validated by viability assay, colony formation and western blot analysis. The significance of heme oxygenase 1 (HMOX1) expression in cholangio-carcinoma was demonstrated using bioinformatics analysis and knockdown of HMOX1 by transfection with short interfering (si)-RNA. RESULTS: Eriodictyol highly reduced the in vitro viability of SNU-308, SNU-478, SNU-1079, and SNU-1196 cholangiocarcinoma cells compared with that of 293T cells, in a dose-dependent manner. The anticancer effect of eriodictyol was achieved by caspase-3-mediated apoptosis. In particular, eriodictyol increased HMOX1 expression, which resulted in attenuation of cholangiocarcinoma cell proliferation. In contrast, ablating HMOX1 expression by si-RNA transfection against HMOX1 made cholangiocarcinoma cells insensitive to the antiproliferative effect of eriodictyol treatment. CONCLUSION: These results collectively indicate that eriodictyol acts as an anticancer agent via regulation of HMOX1 expression against human cholangiocarcinoma.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Neoplasias dos Ductos Biliares/tratamento farmacológico , Neoplasias dos Ductos Biliares/genética , Ductos Biliares Intra-Hepáticos/metabolismo , Linhagem Celular Tumoral , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/genética , Flavanonas , Heme Oxigenase-1/genética , Humanos , RNA Interferente Pequeno/genética
14.
Microvasc Res ; 80(3): 303-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20594998

RESUMO

Patrinia villosa, a Chinese medicinal herb, is known for its anti-inflammatory effects. In the present study, we tested the pro-angiogenic efficacy of an aqueous extract of Patrinia villosa (PVE) in vitro and in vivo. Treatment with PVE significantly enhanced cell proliferation, migration, and the capillary-like structure forming activity of cultured human umbilical vein endothelial cells (HUVECs). Western blot analysis demonstrated that PVE treatment induced a time-dependent phosphorylation of FAK and Akt in HUVECs. Preincubation with a FAK inhibitor, SC203950, abolished PVE-induced proliferation of HUVECs, indicating a role for FAK in PVE-induced angiogenesis. The proangiogenic activity of PVE was confirmed by an ex vivo mouse aortic ring assay and an in vivo murine hindlimb ischemia model. Further analysis using fractions of PVE partitioned by n-hexane, EtOAc, n-BuOH, and water residue revealed that the EtOAc fraction contains the bioactive components responsible for PVE-induced migration, endothelial cord formation, FAK phosphorylation, and aortic ring sprouting. Our results provide a rationale for the use of PVE in the treatment of peripheral vascular insufficiency; they indicate the need to identify the novel pro-angiogenic chemicals in the fractions of PVE.


Assuntos
Indutores da Angiogênese/farmacologia , Células Endoteliais/efeitos dos fármacos , Ativadores de Enzimas/farmacologia , Quinase 1 de Adesão Focal/metabolismo , Isquemia/tratamento farmacológico , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica/efeitos dos fármacos , Patrinia , Extratos Vegetais/farmacologia , Animais , Western Blotting , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fracionamento Químico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/enzimologia , Ativação Enzimática , Quinase 1 de Adesão Focal/antagonistas & inibidores , Membro Posterior , Humanos , Isquemia/enzimologia , Isquemia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Imagem de Perfusão , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Técnicas de Cultura de Tecidos
15.
Cell Oncol (Dordr) ; 43(5): 821-834, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32483746

RESUMO

PURPOSE: Pancreatic cancer is one of the most aggressive cancers. Preclinical and clinical data indicate that Notch 1 ligand jagged1 (JAG1) plays a pro-oncogenic role in several malignant cancers. As yet, however, the role of JAG1 in pancreatic cancer is poorly understood. The objective of the present study was to investigate JAG1 as a therapeutic target in human pancreatic cancer. METHODS: Expression levels of Notch signaling molecules were assessed using GEO datasets and Western blot analysis, respectively. Anti-tumor effects following JAG1 silencing were evaluated using in vitro and in vivo assays. Prognostic implications were assessed using GEO datasets. RESULTS: Using GEO datasets and Western blot analysis we detected significantly higher JAG1 mRNA and protein expression levels in pancreatic cancer compared to normal pancreatic tissues. JAG1 silencing significantly restrained the growth, migration and invasion of pancreatic cancer cells through the induction of apoptosis and blockade of various kinases independent of the Notch1 pathway. Combined JAG1 silencing and gemcitabine treatment showed synergistic anti-viability effects in human pancreatic cancer cells. JAG1 silencing also resulted in significant anti-cancer effects in vivo and high JAG1 expression was found to be associated with an adverse prognosis in pancreatic cancer patients. CONCLUSIONS: From our data we conclude that JAG1 may be a promising therapeutic target in pancreatic cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteína Jagged-1/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Humanos , Proteína Jagged-1/antagonistas & inibidores , Proteína Jagged-1/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Pancreáticas/tratamento farmacológico , Prognóstico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Gencitabina
16.
Anticancer Res ; 40(3): 1335-1344, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32132030

RESUMO

BACKGROUND/AIM: The prognosis of pancreatic cancer has not improved due to its migratory feature and refractory potential to chemo-resistance with absence of effective diagnosis. Despite continuous efforts, its underlying mechanisms of malignant nature remain ambiguous. The objective of this study was to investigate delta-like 1 (DLL1) as a tumor suppressor in the metastasic ability of human pancreatic cancer cells. MATERIALS AND METHODS: Cellular expression of DLL1 was demonstrated using the GEO public database and western blot analysis. The biological function of DLL1 was validated by biological behavior analysis. Prognosis to DLL1 expression was demonstrated using analysis of the GEO public database. RESULTS: Analysis using the GEO database and western blotting showed higher DLL1 mRNA and protein expression levels in pancreatic cancer compared to those in normal pancreas. DLL1 was uniquely expressed in seven human pancreatic cancer cell lines compared to human pancreatic duct epithelial H6c7 cells. Ablation of DLL1 expression stimulated migration and invasion by activating Src and p38 phosphorylation, but not viability and chemo-resistance of human pancreatic cancer cells. In addition, expression of DLL1 was correlated with migratory features of pancreatic cancer in vivo. Moreover, high DLL1 expression was associated with a favorable prognosis in pancreatic cancer patients. CONCLUSION: DLL1 is a potent suppressor of pancreatic cancer metastasis. Understanding correlation between expression and function of DLL1 might contribute to our knowledge of the complicated mechanism of pancreatic cancer metastasis.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/metabolismo , Neoplasias Pancreáticas/metabolismo , Quinases da Família src/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Prognóstico , Transdução de Sinais , Taxa de Sobrevida
17.
Cancers (Basel) ; 12(2)2020 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-31979216

RESUMO

Even though the tumour suppressive role of PTEN is well-known, its prognostic implications are ambiguous. The objective of this study was to further explore the function of PTEN expression in human pancreatic cancer. The expression of PTEN has been dominant in various human cancers including pancreatic cancer when compared with their matched normal tissues. The pancreatic cancer cells have been divided into PTEN blockade-susceptible and PTEN blockade-impassible groups dependent on targeting PTEN by altering intracellular signaling. The expression of PTEN has led to varying clinical outcomes of pancreatic cancer based on GEO Series (GSE) data analysis and Liptak's z analysis. Differential dependency to PTEN blockade has been ascertained based on the expression of polo-like kinase1 PLK1 in pancreatic cancer cells. The prognostic value of PTEN also depends on PLK1 expression in pancreatic cancer. Collectively, the present study provides a rationale for targeting PTEN as a promising therapeutic strategy dependent on PLK1 expressions using a companion biomarker discovery platform.

18.
Cancer Lett ; 456: 1-12, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30902562

RESUMO

Pancreatic cancer remains one of the most dangerous cancers with a grave prognosis. We have reported that dual specificity phosphatise 28 (DUSP28) could be secreted in pancreatic cancer cells. However, its biological function is poorly understood. Here, we distinguish the function of scattered DUSP28 in human pancreatic cancer. DUSP28 was specifically secreted to cultured medium in metastatic pancreatic cancer cells. Treatment with recombinant DUSP28 significantly increased the migration, invasion, and viability of metastatic pancreatic cancer cells through the activation of CREB, AKT, and ERK1/2 signaling pathways. In addition, administration of recombinant DUSP28 elicited pro-angiogenic effects in human umbilical vein endothelial cells. Injection of recombinant DUSP28 also produced tumor growth in vivo. Of interest, DUSP28 formed an autocrine loop with integrin α1 (ITGα1) by transcriptional regulation and recombinant DUSP28 acted as an oncogenic reagent through the interaction with ITGα1. Notably, scattered DUSP28 could be detected in whole blood samples of pancreatic cancer patients by accessible immunoassay. These results provide the basis for DUSP28 as a promising therapeutic target and a biomarker for metastatic pancreatic cancer.


Assuntos
Comunicação Autócrina , Movimento Celular , Fosfatases de Especificidade Dupla/metabolismo , Neoplasias Pancreáticas/enzimologia , Comunicação Parácrina , Animais , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/metabolismo , Fosfatases de Especificidade Dupla/genética , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Integrina alfa1/genética , Integrina alfa1/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Neovascularização Fisiológica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Carga Tumoral
19.
Anticancer Res ; 39(11): 5963-5971, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31704821

RESUMO

BACKGROUND/AIM: The aim of this study was to investigate matrix metalloproteinase 11 (MMP11) as a promising biomarker in human pancreatic cancer. MATERIALS AND METHODS: A consecutive eliminating method was used to select biomarker candidates in pancreatic cancer. mRNA and protein expression levels of candidates were determined in tissues and whole blood samples of healthy donors and pancreatic cancer patients. The prognostic value of MMP11 was determined using various data-sets and Liptak's Z analysis. RESULTS: Analysis using Gene Expression Omnibus (GEO) database showed significantly higher MMP11 mRNA expression in pancreatic cancer tissues compared to that in various normal tissues. MMP11 protein was specifically expressed in pancreatic cancer tissues, but not in various normal or other cancer tissues. Secreted MMP11 levels could be measured using easily accessible techniques and whole blood samples of pancreatic cancer. In addition, high levels of MMP11 were associated with poor prognosis of pancreatic cancer patients. CONCLUSION: MMP11 may be a promising prognostic biomarker for pancreatic cancer patients.


Assuntos
Biomarcadores Tumorais/metabolismo , Metaloproteinase 11 da Matriz/metabolismo , Neoplasias Pancreáticas/patologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Estudos de Casos e Controles , Proliferação de Células , Seguimentos , Humanos , Metaloproteinase 11 da Matriz/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Pancreáticas/enzimologia , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA