Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Apoptosis ; 29(1-2): 210-228, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38087046

RESUMO

Epithelial ovarian cancer (EOC) is the leading cause of cancer death all over the world. USP43 functions as a tumor promoter in various malignant cancers. Nevertheless, the biological roles and mechanisms of USP43 in EOC remain unknown. In this study, USP43 was highly expressed in EOC tissues and cells, and high expression of USP43 were associated with a poor prognosis of EOC. USP43 overexpression promoted EOC cell proliferation, enhanced the ability of migration and invasion, decreased cisplatin sensitivity and inhibited apoptosis. Knockdown of USP43 in vitro effectively retarded above malignant progression of EOC. In vivo xenograft tumors, silencing USP43 slowed tumor growth and enhanced cisplatin sensitivity. Mechanistically, USP43 inhibited HDAC2 degradation and enhanced HDAC2 protein stability through its deubiquitylation function. USP43 diminished the sensitivity of EOC cells to cisplatin through activation of the Wnt/ß-catenin signaling pathway mediated by HDAC2. Taken together, the data in this study revealed the functions of USP43 in proliferation, migration, invasion, chemoresistance of EOC cells, and the mechanism of HDAC2-mediated Wnt/ß-catenin signaling pathway. Thus, USP43 might serve as a potential target for the control of ovarian cancer progression.


Assuntos
Cisplatino , Neoplasias Ovarianas , Humanos , Feminino , Carcinoma Epitelial do Ovário/tratamento farmacológico , Carcinoma Epitelial do Ovário/genética , Carcinoma Epitelial do Ovário/patologia , Cisplatino/farmacologia , Via de Sinalização Wnt , beta Catenina/genética , beta Catenina/metabolismo , Apoptose , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral , Histona Desacetilase 2/genética , Histona Desacetilase 2/metabolismo
2.
BMC Med ; 22(1): 19, 2024 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-38191448

RESUMO

BACKGROUND: The benefits of first-line, cisplatin-based chemotherapy for muscle-invasive bladder cancer are limited due to intrinsic or acquired resistance to cisplatin. Increasing evidence has revealed the implication of cancer stem cells in the development of chemoresistance. However, the underlying molecular mechanisms remain to be elucidated. This study investigates the role of LASS2, a ceramide synthase, in regulating Wnt/ß-catenin signaling in a subset of stem-like bladder cancer cells and explores strategies to sensitize bladder cancer to cisplatin treatment. METHODS: Data from cohorts of our center and published datasets were used to evaluate the clinical characteristics of LASS2. Flow cytometry was used to sort and analyze bladder cancer stem cells (BCSCs). Tumor sphere formation, soft agar colony formation assay, EdU assay, apoptosis analysis, cell viability, and cisplatin sensitivity assay were used to investigate the functional roles of LASS2. Immunofluorescence, immunoblotting, coimmunoprecipitation, LC-MS, PCR array, luciferase reporter assays, pathway reporter array, chromatin immunoprecipitation, gain-of-function, and loss-of-function approaches were used to investigate the underlying mechanisms. Cell- and patient-derived xenograft models were used to investigate the effect of LASS2 overexpression and a combination of XAV939 on cisplatin sensitization and tumor growth. RESULTS: Patients with low expression of LASS2 have a poorer response to cisplatin-based chemotherapy. Loss of LASS2 confers a stem-like phenotype and contributes to cisplatin resistance. Overexpression of LASS2 results in inhibition of self-renewal ability of BCSCs and increased their sensitivity to cisplatin. Mechanistically, LASS2 inhibits PP2A activity and dissociates PP2A from ß-catenin, preventing the dephosphorylation of ß-catenin and leading to the accumulation of cytosolic phospho-ß-catenin, which decreases the transcription of the downstream genes ABCC2 and CD44 in BCSCs. Overexpression of LASS2 combined with a tankyrase inhibitor (XAV939) synergistically inhibits tumor growth and restores cisplatin sensitivity. CONCLUSIONS: Targeting the LASS2 and ß-catenin pathways may be an effective strategy to overcome cisplatin resistance and inhibit tumor growth in bladder cancer patients.


Assuntos
Cisplatino , Esfingosina N-Aciltransferase , Neoplasias da Bexiga Urinária , Humanos , Apoptose , beta Catenina , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Esfingosina N-Aciltransferase/metabolismo
3.
Mol Cell Biochem ; 2024 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-38735913

RESUMO

Early detection and effective chemotherapy for ovarian cancer, a serious gynecological malignancy, require further progress. This study aimed to investigate the molecular mechanism of ATPase H+-Transporting V1 Subunit B1 (ATP6V1B1) in ovarian cancer development and chemoresistance. Our data show that ATP6V1B1 is upregulated in ovarian cancer and correlated with decreased progression-free survival. Gain- and loss-of-function experiments demonstrated that ATP6V1B1 promotes the proliferation, migration, and invasion of ovarian cancer cells in vitro, while ATP6V1B1 knockout inhibits tumor growth in vivo. In addition, knocking down ATP6V1B1 increases the sensitivity of ovarian cancer cells to cisplatin. Mechanistic studies showed that ATP6V1B1 regulates the activation of the mTOR/autophagy pathway. Overall, our study confirmed the oncogenic role of ATP6V1B1 in ovarian cancer and revealed that ATP6V1B1 promotes ovarian cancer progression via the mTOR/autophagy axis.

4.
Cell Biol Int ; 48(4): 521-540, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38263578

RESUMO

The ion channel two-pore channel 2 (TPC2), localised on the membranes of acidic organelles such as endo-lysosomes and melanosomes, has been shown to play a role in pathologies including cancer, and it is differently expressed in primary versus metastatic melanoma cells. Whether TPC2 plays a pro- or anti-oncogenic role in different tumour conditions is a relevant open question which we have explored in melanoma at different stages of tumour progression. The behaviour of primary melanoma cell line B16F0 and its metastatic subline B16F10 were compared in response to TPC2 modulation by silencing (by small interfering RNA), knock-out (by CRISPR/Cas9) and overexpression (by mCherry-TPC2 transfected plasmid). TPC2 silencing increased cell migration, epithelial-to-mesenchymal transition and autophagy in the metastatic samples, but abated them in the silenced primary ones. Interestingly, while TPC2 inactivation failed to affect markers of proliferation in both samples, it strongly enhanced the migratory behaviour of the metastatic cells, again suggesting that in the more aggressive phenotype TPC2 plays a specific antimetastatic role. In line with this, overexpression of TPC2 in B16F10 cells resulted in phenotype rescue, that is, a decrease in migratory ability, thus collectively resuming traits of the B16F0 primary cell line. Our research shows a novel role of TPC2 in melanoma cells that is intriguingly different in initial versus late stages of cancer progression.


Assuntos
Melanoma , Humanos , Melanoma/metabolismo , Canais de Dois Poros , Lisossomos/metabolismo , Linhagem Celular , Autofagia/fisiologia , Cálcio/metabolismo
5.
J Biochem Mol Toxicol ; 38(1): e23537, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37700640

RESUMO

Increasing evidence indicated that protein arginine methyltransferase-1 (PRMT1) is an oncogene in multiple malignant tumors, including osteosarcoma (OS). The aim of this study was to investigate the underlying mechanism of PRMT1 in OS. The effects of PRMT1 or BCAT1, branched-chain amino acid transaminase 1 (BCAT1) on OS cell proliferation, invasion, autophagy, and apoptosis in vitro were examined. Moreover, molecular control of PRMT1 on c-Myc or transactivation of BCAT1 on c-Myc was assessed by chromatin immunoprecipitation and quantitative reverse transcription PCR assays. The effects of PRMT1 in vivo were examined with a xenograft tumor model. The results showed that PRMT1 was potently upregulated in OS tissues and cells. Upregulation of PRMT1 markedly increased OS cell proliferation and invasion in vitro and reduced cell apoptosis, whereas PRMT1 silencing showed the opposite effects. Cisplatin, one of the most effective chemotherapeutic drugs, improved cell survival rate by inducing the expression of PRMT1 to downregulate the cisplatin sensitivity. Meanwhile, the cisplatin-induced upregulation of PRMT1 expression caused dramatically autophagy induction and autophagy-mediated apoptosis by inactivating the mTOR signaling pathway, which could be reversed by 3-methyladenine, an autophagy inhibitor, or PRMT1 silencing. PRMT1 could activate c-Myc transcription and increase c-Myc-mediated expression of BCAT1. Furthermore, BCAT1 overexpression counteracted the effects of PRMT1 knockdown on cell proliferation, invasion, and apoptosis. Of note, deficiency of PRMT1 suppressed tumor growth in vivo. PRMT1 facilitated the proliferation and invasion of OS cells, inhibited cell apoptosis, and decreased chemotherapy sensitivity through c-Myc/BCAT1 axis, which may become potential target in treating OS.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Humanos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Regulação para Baixo , Linhagem Celular Tumoral , Osteossarcoma/tratamento farmacológico , Osteossarcoma/genética , Osteossarcoma/metabolismo , Apoptose , Metiltransferases/metabolismo , Neoplasias Ósseas/metabolismo , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases/uso terapêutico , Proteínas Repressoras/metabolismo , Transaminases/genética , Transaminases/metabolismo , Transaminases/farmacologia
6.
J Cell Mol Med ; 27(16): 2412-2423, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37438979

RESUMO

Non-small cell lung cancer (NSCLC) accounts for approximately 85% of lung cancer. Cisplatin is commonly used in the treatment of many malignant tumours including NSCLC. The innate drug sensitivity greatly affects the clinical efficacy of cisplatin-based chemotherapy. As a plasma membrane adhesion molecule, amphoterin-induced gene and ORF-2 (AMIGO2) initially identified as a neurite outgrowth factor has been recently found to play a crucial role in cancer occurrence and progression. However, it is still unclear whether AMIGO2 is involved in innate cisplatin sensitivity. In the present study, we provided the in vitro and in vivo evidences indicating that the alteration of AMIGO2 expression triggered changes of innate cisplatin sensitivity as well as cisplatin-induced pyroptosis in NSCLC. Further results revealed that AMIGO2 might inhibit cisplatin-induced activation of (caspase-8 and caspase-9)/caspase-3 via stimulating PDK1/Akt (T308) signalling axis, resulting in suppression of GSDME cleavage and the subsequent cell pyroptosis, thereby decreasing the sensitivity of NSCLC cells to cisplatin treatment. The results provided a new insight that AMIGO2 regulated the innate cisplatin sensitivity of NSCLC through GSDME-mediated pyroptosis.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Caspase 3/metabolismo , Cisplatino/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Proteínas do Tecido Nervoso/genética , Piroptose , Transdução de Sinais , Gasderminas/efeitos dos fármacos , Gasderminas/metabolismo
7.
Biochem Biophys Res Commun ; 661: 75-81, 2023 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-37087801

RESUMO

Cisplatin resistance is the main reason for uveal melanoma (UM) treatment failure. Thus, developing strategy that increasing cisplatin sensitivity is needed. In this study, we performed drug repositioning analysis with the Connectivity Map database using a panel of previously identified cisplatin sensitivity-associated genes and cisplatin resistance-associated genes as the signature and obtained the antiparasitic drug selamectin. We demonstrated that the selamectin and cisplatin combination showed a synergistic effect on inhibiting UM cell growth. Experiments in tumor-bearing nude mice further showed that selamectin and cisplatin have synergistic effects in reducing tumor growth. Previous studies have linked increased autophagy with tumor resistance to chemotherapy. We found that selamectin inhibited the expression of the autophagy-related gene ATG9B, thus reducing autophagy. The cisplatin resistance-associated genes PDGFRB, DUSP1, MAST1 and IL11 were significantly downregulated in UM cells treated with selamectin. In summary, our study shows that selamectin enhanced the sensitivity of UM to cisplatin, through the mechanism of inhibiting cisplatin resistance-associated gene expression and autophagy. These findings may provide a new strategy for the treatment of UM.


Assuntos
Cisplatino , Neoplasias Uveais , Animais , Camundongos , Cisplatino/farmacologia , Camundongos Nus , Linhagem Celular Tumoral , Neoplasias Uveais/tratamento farmacológico , Autofagia
8.
Mol Carcinog ; 61(11): 1043-1055, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36102200

RESUMO

Esophageal squamous cell carcinoma (ESCC) is a common human malignancy characterized by late-stage diagnosis, metastasis, and poor prognosis. Cisplatin (DDP)-based chemotherapy has been the most predominant treatment for patients with ESCC. However, the high rate of DDP resistance and toxicity seriously hinder its clinical application. Then, the optimized strategy and mechanisms for ESCC to enhance DDP sensitivity are in great demand. Accumulating evidence have shown that chaperone proteins are closely related to the tumorigenesis and drug resistance of cancers. Chaperonin containing TCP1 complex 4 (CCT4) is a recent identified member of the family. However, its expression and function in ESCC have not been well illustrated. In this study, we found that CCT4 was highly expressed in human ESCC tissues and cell lines, and closely related to the poor prognosis. Moreover, CCT4 silence raised oxidative stress and inhibited glycolysis of ESCC cells, which significantly inhibited cell proliferation and migration, promoted apoptosis and caused cell cycle arrest in ESCC cells. Interestingly, CCT4 knockdown enhanced the sensitivity of KYSE150 cells to DDP by regulating AMPK/AKT/Nrf2 signaling pathway and inhibiting glycolysis ability. Taken together, our results indicate that targeting CCT4 may be a therapeutic target in ESCC patients, which provides a theoretical basis to enhance the sensitivity of DDP in ESCC.


Assuntos
Carcinoma de Células Escamosas , Chaperonina com TCP-1 , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Proteínas Quinases Ativadas por AMP/metabolismo , Apoptose , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Chaperonina com TCP-1/genética , Chaperoninas/metabolismo , Chaperoninas/uso terapêutico , Cisplatino/farmacologia , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/genética , Regulação Neoplásica da Expressão Gênica , Glicólise , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
9.
Exp Cell Res ; 409(1): 112891, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34688610

RESUMO

Cisplatin (CDDP) is widely used for chemotherapy of esophageal squamous cell carcinoma (ESCC) but the drug resistance limits its therapeutic benefit. Heterogeneous nuclear ribonucleoprotein U-like 1 (HNRNPUL1) belongs to the family of RNA-binding proteins (RBPs) and is involved in DNA damage repair. To investigate whether and how HNRNPUL1 affects CDDP resistance of ESCC, we evaluated the expression of HNRNPUL1 and found that it was associated with recurrence in ESCC patients receiving postoperative platinum-based chemotherapy and was an independent prognostic factor for disease-free survival (DFS). Besides, we showed that the reduced expression of HNRNPUL1 enhanced the CDDP sensitivity of ESCC cells. Furthermore, RNA immunoprecipitation coupled with high-throughput sequencing (RIP-seq) were performed and a range of HNRNPUL1-binding RNAs influenced by CDDP treatment were identified followed by bioinformatics analysis. In terms of mechanism, we found that HNRNPUL1 inhibited CDDP sensitivity of ESCC cells by regulating the CDDP sensitivity-inhibited circular RNA (circRNA) MAN1A2 formation. Taken together, our results first demonstrated the role of HNRNPUL1 in CDDP resistance of ESCC and suggested that HNRNPUL1 may be a potential target of ESCC chemotherapy.


Assuntos
Cisplatino/uso terapêutico , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/genética , Ribonucleoproteínas Nucleares Heterogêneas/genética , Proteínas Nucleares/genética , RNA Circular/genética , Fatores de Transcrição/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade
10.
Oral Dis ; 2022 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-36453015

RESUMO

OBJECTIVES: This study concentrates on exploring the synergistic effect of shikonin on cisplatin against oral cancer. METHODS: To analyze the IC50 value of shikonin, gradient concentrations of shikonin were added to the oral cancer cell culture medium. After the cisplatin-resistant cell line was established, the effects of cisplatin and shikonin on the survival rate, proliferation, apoptosis and related pathway protein expression of common/drug-resistant oral cancer cells were compared through MTT, clone formation, flow cytometry, and Western blot experiments. ß-catenin, which had the most significant expression changes, was overexpressed and silenced, and used to design a reverse validation. RESULTS: Shikonin inhibited the viability of oral cancer cells. Although cisplatin killed some cancer cells, its effect on drug-resistant cancer cells was significantly reduced. The addition of shikonin enhanced the sensitivity of drug-resistant cells to cisplatin. Shikonin regulated key proteins in cell proliferation and apoptosis-related pathways. Among them, shikonin generated the most evident inhibitory effect on ß-catenin. Therefore, ß-catenin overexpression plasmid/siß-catenin was transfected into the cells. Silenced ß-catenin was found to reinforce the damaging effect of cisplatin on cancer cells, and overexpressed ß-catenin reversed the effect of shikonin. CONCLUSION: By down-regulating ß-catenin expression, shikonin improves the sensitivity of drug-resistant oral cancer cells to cisplatin.

11.
Drug Dev Res ; 83(7): 1589-1599, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35903032

RESUMO

Replication protein A 3 (RPA3) is a significant component of replication protein A and has been documented to function as an oncogene in several types of cancers. However, the role and underlying mechanism of RPA3 in lung adenocarcinoma (LUAD) remains unknown. In this study, messenger expression of RPA3 and survival probability in LUAD were predicted by the UALCAN database. The combination of RPA3 with cyclin-dependent kinases regulatory subunit 2 (CKS2) were characterized by the humanbase and STRING databases and verified by co-immunoprecipitation. Cell viability was assessed by Cell Counting Kit-8 assay and colony formation assay. Flow cytometric analysis and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay were used to determine cell cycle and cell apoptosis, respectively. The expressions of protein kinase B/mammalian target of rapamycin (AKT/mTOR) pathway and autophagy-related proteins were examined by western blot assay. Significantly, we revealed that RPA3 expression was upregulated in LUAD and is associated with poor prognosis in LUAD patients. RPA3 and CKS2 expression was highly expressed in LUAD cell lines and the interaction between RPA3 and CKS2 was confirmed. RPA3 silencing inhibited A549 cell viability, blocked cell cycle and promoted cell apoptosis, as well as induction of autophagy and inhibition of AKT/mTOR signaling. CKS2 overexpression reversed the effects of RPA3 silencing on A549 cells. In addition, RPA3 knockdown enhanced cisplatin sensitivity of A549 cells through blocking the AKT/mTOR signaling. These results suggested that RPA3 might control LUAD cell autophagy and enhance cisplatin sensitivity by regulation of AKT/mTOR signaling via targeting CKS2.


Assuntos
Adenocarcinoma de Pulmão , Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Neoplasias Pulmonares , Humanos , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Autofagia , Quinases relacionadas a CDC2 e CDC28/genética , Proteínas de Ciclo Celular/genética , Proliferação de Células , Cisplatino/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo
12.
Cancer Cell Int ; 20: 198, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32514243

RESUMO

BACKGROUND: Esophageal cancer is the sixth leading cause of cancer-related mortality worldwide, which is partially due to limited progress of therapy. Apatinib, an inhibitor of VEGFR2, has a promising antitumor effect on malignancies. However, the underlying mechanism of its antitumor effect on esophageal cancer remains poorly understood. MATERIALS AND METHODS: Eighteen pairs of frozen esophageal cancer and their para-cancer samples and 25 paraffin specimens from advanced esophageal cancer patients treated with cisplatin-based regimen were collected. The effects of apatinib on cell growth, cell apoptosis, cell cycle and invasion/migration of esophageal cancer cells were assessed. Bioinformatics, luciferase reporter, immunoprecipitation and immunofluorescence assays were conducted for mechanic investigation. Quantitative RT-PCR, western blotting and immunohistochemistry were used to measure the expression of functional genes. Xenograft tumor growth of mice was performed. RESULTS: We found that VEGFR2 was highly expressed in esophageal cancer and associated with poor efficacy of cisplatin-based treatment. Apatinib displayed profound actions against tumor cell growth of human esophageal cancer via promoting cell apoptosis and cell cycle arrest. Also, apatinib displayed the inhibitory effects on cell migration and invasion. Moreover, apatinib strongly suppressed the growth of esophageal cancer xenografts in mice. The effects of apatinib on esophageal cancer were partially dependent on its block of the VEGFR2/Akt/ß-catenin pathway. Specifically, apatinib induced the degradation of ß-catenin and decreased its transcriptional activity through Akt/GSK-3ß repression. Further in vitro and in vivo studies revealed that low dose apatinib had a synergistic antitumor effect with cisplatin on esophageal cancer. CONCLUSION: Our study indicates that apatinib suppresses tumor progression and enhances cisplatin sensitivity in esophageal cancer by deactivating the Akt/ß-catenin pathway. These findings provide a theoretical foundation for using apatinib as an effective therapeutic drug for esophageal cancer.

13.
BMC Cancer ; 20(1): 349, 2020 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-32326899

RESUMO

BACKGROUND: Testicular germ cell tumours (TGCTs) are characterised by an overall high cisplatin-sensitivity which has been linked to their continued expression of pluripotency factors. Recently, the Nodal signalling pathway has been implicated in the regulation of pluripotency factor expression in fetal germ cells, and the pathway could therefore also be involved in regulating expression of pluripotency factors in malignant germ cells, and hence cisplatin-sensitivity in TGCTs. METHODS: We used in vitro culture of the TGCT-derived cell line NTera2, ex vivo tissue culture of primary TGCT specimens and xenografting of NTera2 cells into nude mice in order to investigate the consequences of manipulating Nodal and Activin signalling on pluripotency factor expression, apoptosis, proliferation and cisplatin-sensitivity. RESULTS: The Nodal signalling factors were markedly expressed concomitantly with the pluripotency factor OCT4 in GCNIS cells, seminomas and embryonal carcinomas. Despite this, inhibition of Nodal and Activin signalling either alone or simultaneously did not affect proliferation or apoptosis in malignant germ cells in vitro or ex vivo. Interestingly, inhibition of Nodal signalling in vitro reduced the expression of pluripotency factors and Nodal pathway genes, while stimulation of the pathway increased their expression. However, cisplatin-sensitivity was not affected following pharmacological inhibition of Nodal/Activin signalling or siRNA-mediated knockdown of the obligate co-receptor CRIPTO in NTera2 cells in vitro or in a xenograft model. CONCLUSION: Our findings suggest that the Nodal signalling pathway may be involved in regulating pluripotency factor expression in malignant germ cells, but manipulation of the pathway does not appear to affect cisplatin-sensitivity or tumour cell proliferation.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Linfonodos/patologia , Neoplasias Embrionárias de Células Germinativas/patologia , Células-Tronco Pluripotentes/patologia , Neoplasias Testiculares/patologia , Animais , Proliferação de Células , Humanos , Linfonodos/efeitos dos fármacos , Masculino , Camundongos , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Células-Tronco Pluripotentes/efeitos dos fármacos , Transdução de Sinais , Neoplasias Testiculares/tratamento farmacológico , Células Tumorais Cultivadas
14.
Cell Mol Biol (Noisy-le-grand) ; 66(3): 191-196, 2020 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-32538770

RESUMO

The current experiment aimed to investigate the effects of lncRNA KCNQ1OT1 on the proliferation, autophagy and drug resistance of hepatocellular carcinoma cells, as well as the potential molecular mechanism. Hepatocellular carcinoma SK-HEP-1 cells and DDP resistant SK-HEP-1/DDP cells were treated with cisplatin (DDP) of different concentrations (1 nmol/L, 2 nmol/L, 4 nmol/L, 8 nmol/L, 16 nmol/L). The survival rate of SK-HEP-1 and SK-HEP-1/DDP cells was determined by the CCK8 method. QRT-PCR was used to detect the levels of lncRNA KCNQ1OT1 and miR-338-3p in normal hepatocyte HH01, hepatocellular cell SK-HEP-1 and hepatoma cisplatin-resistant cell SK-HEP-1/DDP. Western blot was carried out to detect the expression levels of autophagy-related protein Beclin1 and proliferation-related protein P21 in cells. A dual-luciferase reporter assay system was performed to validate the relationship between KCNQ1OT1 and miR-338-3p. After the treatment of 1 nmol/L, 2 nmol/L, 4nmol/L, 8nmol/L and 16nmol/L cisplatin (DDP), the survival rate of SK-HEP-1/DDP cells is higher than that of SK-HEP-1 cells. The level of lncRNA KCNQ1OT1 was increased successively in HH01, SK-HEP-1 and SK-HEP-1/DDP cells, while miR-338-3p was decreased successively. Silencing lncRNA KCNQ1OT1 or over-expressing miR-338-3p combined with 16nmol/L DDP treatment reduced the survival rate of SK-HEP-1/DDP cells and up-regulate levels of P21 and Beclin1 proteins. LncRNA KCNQ1OT1 targeted and negatively regulated the expression of miR-338-3p. Inhibition of miR-338-3p reversed the effect of silencing lncRNA KCNQ1OT1 on survival, autophagy and cisplatin sensitivity of SK-HEP-1/DDP cell. LncRNA KCNQ1OT1 targets miR-338-3p to regulate the survival rate and autophagy of SK-HEP-1/DDP cells and improve the cisplatin sensitivity of SK-HEP-1/DDP cells.  LncRNA KCNQ1OT1 is a potential molecular target for hepatocellular carcinoma.


Assuntos
Autofagia/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Hepáticas/genética , MicroRNAs/metabolismo , Sequência de Bases , Carcinoma Hepatocelular/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , MicroRNAs/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo
15.
BMC Cancer ; 19(1): 1124, 2019 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-31744479

RESUMO

BACKGROUND: Testicular germ cell cancer (TGCC) develops from pre-malignant germ neoplasia in situ (GCNIS) cells. GCNIS originates from fetal gonocytes (POU5F1+/MAGE-A4-), which fail to differentiate to pre-spermatogonia (POU5F1-/MAGE-A4+) and undergo malignant transformation. Gankyrin is an oncogene which has been shown to prevent POU5F1 degradation and specifically interact with MAGE-A4 in hepatocellular carcinoma (HCC) cells. We aimed to investigate the role of Gankyrin in progression from gonocyte to pre-invasive GCNIS and subsequent invasive TGCC. METHODS: We determined Gankyrin expression in human fetal testicular tissue (gestational weeks 9-20; n = 38), human adult testicular tissue with active spermatogenesis (n = 9), human testicular tissue with germ cell maturation delay (n = 4), testicular tissue from patients with pre-invasive GCNIS (n = 6), and invasive TGCC including seminoma (n = 6) and teratoma (n = 7). Functional analysis was performed in-vitro by siRNA knock-down of Gankyrin in the NTera2 cells (derived from embryonal carcinoma). RESULTS: Germ cell expression of Gankyrin was restricted to a sub-population of prespermatogonia in human fetal testes. Nuclear Gankyrin was also expressed in GCNIS cells of childhood and adult pre-invasive TGCC patients, and in GCNIS from seminoma and non-seminoma patients. Cytoplasmic expression was observed in seminoma tumour cells and NTera2 cells. Gankyrin knock-down in NTera2 cells resulted in an increase in apoptosis mediated via the TP53 pathway, whilst POU5F1 expression was unaffected. Furthermore, Gankyrin knock-down in NTera2 cells increased cisplatin sensitivity with an increase in cell death (13%, p < 0.05) following Gankyrin knock-down, when compared to cisplatin treatment alone, likely via BAX and FAS. Our results demonstrate that Gankyrin expression changes in germ cells during normal transition from gonocyte to prespermatogonia. In addition, changes in Gankyrin localisation are associated with progression of pre-invasive GCNIS to invasive TGCC. Furthermore, we found that Gankyrin is involved in the regulation of NTera2 cell survival and that a reduction in Gankyrin expression can modulate cisplatin sensitivity. CONCLUSIONS: These results suggest that manipulation of Gankyrin expression may reduce the cisplatin dose required for the treatment of TGCC, with benefits in reducing dose-dependent side effects of chemotherapy. Further studies are required in order to assess the effects of modulating Gankyrin on GCNIS/TGCC using in vivo models.


Assuntos
Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Embrionárias de Células Germinativas/genética , Oncogenes , Complexo de Endopeptidases do Proteassoma/genética , Proteínas Proto-Oncogênicas/genética , Neoplasias Testiculares/genética , Apoptose/genética , Biomarcadores Tumorais , Ciclo Celular/genética , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Masculino
16.
BMC Cancer ; 19(1): 1039, 2019 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-31684899

RESUMO

BACKGROUND: Understanding mechanisms underlying specific chemotherapeutic responses in subtypes of cancer may improve identification of treatment strategies most likely to benefit particular patients. For example, triple-negative breast cancer (TNBC) patients have variable response to the chemotherapeutic agent cisplatin. Understanding the basis of treatment response in cancer subtypes will lead to more informed decisions about selection of treatment strategies. METHODS: In this study we used an integrative functional genomics approach to investigate the molecular mechanisms underlying known cisplatin-response differences among subtypes of TNBC. To identify changes in gene expression that could explain mechanisms of resistance, we examined 102 evolutionarily conserved cisplatin-associated genes, evaluating their differential expression in the cisplatin-sensitive, basal-like 1 (BL1) and basal-like 2 (BL2) subtypes, and the two cisplatin-resistant, luminal androgen receptor (LAR) and mesenchymal (M) subtypes of TNBC. RESULTS: We found 20 genes that were differentially expressed in at least one subtype. Fifteen of the 20 genes are associated with cell death and are distributed among all TNBC subtypes. The less cisplatin-responsive LAR and M TNBC subtypes show different regulation of 13 genes compared to the more sensitive BL1 and BL2 subtypes. These 13 genes identify a variety of cisplatin-resistance mechanisms including increased transport and detoxification of cisplatin, and mis-regulation of the epithelial to mesenchymal transition. CONCLUSIONS: We identified gene signatures in resistant TNBC subtypes indicative of mechanisms of cisplatin. Our results indicate that response to cisplatin in TNBC has a complex foundation based on impact of treatment on distinct cellular pathways. We find that examination of expression data in the context of heterogeneous data such as drug-gene interactions leads to a better understanding of mechanisms at work in cancer therapy response.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Genômica/métodos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Evolução Biológica , Linhagem Celular Tumoral , Sequência Conservada , Transição Epitelial-Mesenquimal/genética , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Ratos , Receptores Androgênicos/metabolismo
17.
BMC Cancer ; 18(1): 1294, 2018 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-30594176

RESUMO

BACKGROUND: MLH1 plays a critical role in maintaining the fidelity of DNA replication, and defects in human MLH1 have been reported. However, the role of MLH1 in endometrial carcinoma has not been fully investigated. Therefore, we aimed to study the role of MLH1 in the sensitivity of human endometrial carcinoma cells to cisplatin. METHODS: In this study, we detected the expression of MLH1 in Ishikawa and RL95-2 cells. MLH1-siRNA and ADV-MLH1 were adopted for the silencing and overexpression of MLH1, respectively. Real-time polymerase chain reaction, Western blotting, cell proliferation assays, and cell cycle and apoptotic analyses by flow cytometry were employed to explore the underlying mechanism. A mouse xenograft model was used to investigate the effect of MLH1 on tumor growth after treatment with cisplatin. RESULTS: Over-expression of MLH1 in Ishikawa cells dramatically increased the sensitivity of cells to cisplatin and enhanced cell apoptosis. By contrast, knockdown of MLH1 yielded the opposite effects in vitro. Mechanistically, cisplatin induced the MLH1/c-Abl apoptosis signaling pathway in ADV-MLH1-infected endometrial carcinoma cells, and these effects involved c-Abl, caspase-9, caspase-3 and PARP. Altogether, our results indicate that ADV-MLH1 might attenuate Ishikawa cell growth in vivo, resulting in increased cisplatin sensitivity. CONCLUSIONS: MLH1 may render endometrial carcinoma cells more sensitive to cisplatin by activating the MLH1/c-Abl apoptosis signaling pathway. In addition, an applicable adenovirus vector (ADV-MLH1) for MLH1 overexpression in endometrial carcinoma was generated. Thus, ADV-MLH1 might be a novel potential therapeutic target for endometrial carcinoma.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias do Endométrio/terapia , Proteína 1 Homóloga a MutL/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Adenoviridae/genética , Animais , Antineoplásicos/uso terapêutico , Apoptose/genética , Linhagem Celular Tumoral , Cisplatino/uso terapêutico , Terapia Combinada/métodos , Neoplasias do Endométrio/patologia , Feminino , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Injeções Intralesionais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteína 1 Homóloga a MutL/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/genética , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Chem Pharm Bull (Tokyo) ; 66(2): 162-169, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29386467

RESUMO

Crebanine (CN), tetrahydropalmatine (THP), O-methylbulbocapnine (OMBC) and N-methyl tetrahydropalmatine (NMTHP) are isoquinoline derived natural alkaloids isolated from tubers of Stephania venosa. We investigated chemo-sensitizing effects of these alkaloids in ovarian cancer cells and evaluated underlying molecular mechanisms involved in chemo-sensitivity. Detection of cell apoptosis was evaluated by using flow cytometry. Cell viability was analyzed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Chou-Talalay median effect principle was used to evaluate potential drug interactions. Protein analyses were performed on ovarian carcinoma cells using Western blotting upon treatment with anticancer drug and alkaloids. Aporphine alkaloids, such as CN and OMBC, enhanced cisplatin sensitivity in intrinsic cisplatin resistant SKOV3 cells, but not in cisplatin sensitive A2780 cells. Protoberberine alkaloids, such as THP and NMTHP, had no synergistic effect on cisplatin sensitivity in either cell line. Chemo-sensitizing effects of CN and OMBC in SKOV3 cells were mediated via activating apoptosis-induced cell death through caspase-3, -8 and cleaved poly ADP-ribose polymerase (PARP) and via inhibiting anti-apopotic and survival protein expression, such as Bcl-xL, Baculoviral IAP repeat-containing protein 3 (cIAP-2), survivin and interleukin (IL) -6. Cisplatin stimulated protein kinase B (Akt) and nuclear factor-kappaB (NF-κB) signaling pathways, but not mitogen-activated protein kinase (MAPK), activator protein 1 (AP-1) and signal transducer and activator of transcription 3 (STAT3) in SKOV3 cells. Akt/NF-κB signaling was blocked by CN and OMBC leading to increased sensitization to cisplatin. These findings demonstrate that CN and OMBC sensitizes SKOV3 cells to cisplatin via inhibition of Akt/NF-κB signaling and the down regulation of NF-κB mediated gene products. Our results suggest that alkaloids obtained from S. venosa could be used as chemo-sensitizers in ovarian cancer to sensitize and minimize the dose related toxicity of platinum-based chemotherapeutic drugs.


Assuntos
Alcaloides/química , Antineoplásicos/química , Neoplasias Ovarianas/tratamento farmacológico , Extratos Vegetais/química , Stephania/química , Alcaloides/isolamento & purificação , Alcaloides/farmacologia , Antineoplásicos/farmacologia , Apoptose , Alcaloides de Berberina/química , Linhagem Celular Tumoral , Sobrevivência Celular , Cisplatino/farmacologia , Regulação para Baixo , Feminino , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Extratos Vegetais/isolamento & purificação , Extratos Vegetais/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
19.
Tumour Biol ; 37(7): 8857-67, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26749283

RESUMO

Tissue inhibitors of metalloproteinases (TIMPs) inhibit matrix metalloproteinases (MMPs) to limit degradation of the extracellular matrix. Low levels of TIMP3 have been demonstrated in cancer tissues at advanced clinical stages, with positive distant metastasis and chemotherapeutic resistance. We examined the role of TIMP3 in osteosarcoma (OS) cell invasiveness and chemoresistance. TIMP3 was overexpressed or knocked down in the human OS cell lines Saos2 and MG63. Cell migration and invasion capacities were then evaluated using Transwell assays, and resistance to cisplatin was assessed by CCK-8 assay and flow cytometry. Real-time PCR and western blotting were used to investigate activation of signaling pathways downstream of TIMP3. Overexpression of TIMP3 inhibited the migration and invasion of Saos2 and MG63 cells, while knockdown of TIMP3 had the opposite effect. Cell survival after exposure to cisplatin was inhibited by TIMP3 overexpression in both Saos2 and MG63 cells. Consistently, downregulation of TIMP3 gene expression significantly decreased the sensitivity of OS cells to cisplatin treatment. MMP1, MMP2, Bcl-2, and Akt1 were all downregulated following TIMP3 overexpression, while Bax and cleaved caspase-3 were upregulated. TIMP3 knockdown had opposite effects on the regulation of these genes. Taken together, our findings suggest TIMP3 as a new target for inhibition of OS progression and chemotherapeutic resistance.


Assuntos
Movimento Celular/genética , Resistencia a Medicamentos Antineoplásicos/genética , Invasividade Neoplásica/genética , Osteossarcoma/genética , Osteossarcoma/patologia , Inibidor Tecidual de Metaloproteinase-3/genética , Caspase 3/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Cisplatino/farmacologia , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 2 da Matriz/genética , Invasividade Neoplásica/patologia , Osteossarcoma/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transdução de Sinais/genética , Regulação para Cima/genética , Proteína X Associada a bcl-2/genética
20.
Helicobacter ; 19(3): 174-81, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24628843

RESUMO

BACKGROUND: Recent studies found that gastric cancer patients with Helicobacter pylori infection had a better response to chemotherapy and had an improved overall prognosis compared with those without. However, the underlying mechanism remains unknown. METHODS: Quantitative real-time PCR (qRT-PCR) was utilized to determine the expression profile of miR-141 in H. pylori infected cells and tissues and their respective controls. qRT-PCR and Western blot were used to determine the expression level of KEAP-1. Luciferase reporter assays were used to determine whether KEAP-1 was a direct target of miR-141 in the gastric cancer cells. MTT and apoptosis assay were performed to detect the survival of cells under cisplatin treatment. RESULT: We found that H. pylori infection can significantly down-regulate miR-141 expression. Knockdown miR-141 expression in 7901/DDP and 7901 cells could significantly improve cisplatin sensitivity. Over-expression of miR-141 resulted in enhanced resistance to cisplatin in both gastric cancer cells. We also demonstrated that miR-141 directly targets KEAP1 by luciferase reporter assay, and that down-regulation of KEAP1 induces cisplatin resistance. Conversely, over-expression of KEAP1 significantly enhanced cisplatin sensitivity. Our 75 pairs of tissues also showed that KEAP1 was significantly up-regulated in H. pylori-positive tissues. CONCLUSION: Altogether, these findings demonstrated that the H. pylori infection could modulate cisplatin sensitivity through miR-141-mediated regulation of KEAP1.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Helicobacter pylori/fisiologia , Interações Hospedeiro-Patógeno , MicroRNAs/biossíntese , Neoplasias Gástricas/tratamento farmacológico , Adulto , Antineoplásicos/farmacologia , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/farmacologia , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/análise , Proteína 1 Associada a ECH Semelhante a Kelch , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA