Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Biomarkers ; 27(2): 138-150, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34927500

ABSTRACT

INTRODUCTION: Bladder cancer is still of unknown initiation and progression, it is difficult to treat the patient once bladder cancer have a distant metastasis. MATERIALS AND METHODS: In the present study, propolis extract was evaluated against bladder cancer cells (T24). Two independent pathways were investigated, apoptosis and angiogenesis, Bax, Bcl-2, P53, and caspase-3 for apoptosis, vascular endothelial growth factor receptor and protein kinase A as angiogenesis potential targets. OBJECTIVES: Molecular docking studies will be conducted for the major known constituents of Egyptian propolis into apoptotic and angiogenic protein targets, to give better insights to the possible binding mode and interactions and investigate the ability of propolis constituents to target both apoptotic and angiogenic pathways. RESULTS: Propolis showed anti-proliferative activity against T24 cancer cell line, the IC50 value was 6.36 µg/ml. Also significant effects of propolis on Bax, Bcl-2, P53, and caspase-3 were observed. DISCUSSION: These obtained results proved the ability of propolis to induce cell death. Also it has revealed noticeable effects on protein kinase A and vascular endothelial growth factor receptor. CONCLUSION: The obtained results can encourage us to say that propolis extract can induce a programmed cell death in human bladder cancer cells, and also affect angiogenesis.


Subject(s)
Antineoplastic Agents , Propolis , Urinary Bladder Neoplasms , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Early Detection of Cancer , Humans , Molecular Docking Simulation , Propolis/pharmacology , Propolis/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Vascular Endothelial Growth Factor A
2.
Molecules ; 25(14)2020 Jul 17.
Article in English | MEDLINE | ID: mdl-32709143

ABSTRACT

Quercetin (QU), a hyperthermic sensitizer, when combined with cisplatin (CP) affects tumor growth. To determine the effects of QU and CP and their interactions, multimodal treatment in vitro and in vivo models under physiological and hyperthermic conditions was performed. In vitro, different sensitivity of T24 and UMUC human bladder cancer cells was observed after short-term exposure to QU (2 h) and CP (1 h). Effects of both compounds were investigated at low and high micromolar concentrations (1 and 50 µM, respectively) under both thermal conditions. QU acted in additive or synergistic manner in combination with CP between physiological condition and hyperthermia. As determined by 3-(4,5-dimethylthiazol2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, short-term application of QU and CP reduced cell viability. Clonal assay also indicated that combined treatment with QU and CP is lethal to bladder cancer cells in both conditions. In vivo, CP (5 or 10 mg kg-1) and QU (50 mg kg-1) acted synergistically with hyperthermia (43 °C) and inhibited tumor growth, activated immune effectors and increased mice survival. Our results demonstrate that combined treatment with CP and QU may increase death of tumor cells in physiological and hyperthermic conditions which could be clinically relevant in locoregional chemotherapy.


Subject(s)
Cell Proliferation/drug effects , Cisplatin/pharmacology , Quercetin/pharmacology , Urinary Bladder Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Heterografts , Humans , Hyperthermia, Induced/methods , Mice , Urinary Bladder Neoplasms/pathology
3.
J Sci Food Agric ; 99(6): 3106-3116, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30516283

ABSTRACT

BACKGROUND: Allyl isothiocyanate (AITC), a volatile and water-insoluble compound present in several cruciferous vegetables, has been shown to possess several biological qualities such as anti-bacterial, anti-fungal, and anti-cancer activity. In this study, water-soluble allyl isothiocyanate nanoparticles (AITC-NPs) were prepared by oil dispersed in water (O/W) microemulsion and complex coacervation techniques and evaluated for their anti-inflammatory activity towards macrophage cell RAW 264.7 and anti-cancer effect on human bladder cancer cell HT1376. RESULTS: The AITC-NPs with a particle size of 9.4 nm were stable during heating up to 110 °C or three freeze-thawing cycles. No significant cytotoxicity was shown on Caco-2 and intestine epithelial IEC-6 cells at AITC-NP doses ranging from 0.25 to 2 g L-1 (8.75-70 mg L-1 AITC). However, at 2 g L-1 dosage, AITC-NPs could inhibit the growth of human bladder cancer cells HT1376 by 90%, while their low dosage at 0.25 g L-1 could inhibit migration ability by 83.7, 71.3, 58.4 and 31.4% after 4, 8, 12, and 24 h of incubation, respectively. Compared to AITC and NPs, AITC-NPs showed a better inhibition on lipopolysaccharide (LPS)-induced TNF-α, IL-6, NO and iNOS production in RAW 264.7 macrophage cells. CONCLUSION: The results demonstrate the potential of AITC-NPs as therapeutic agents for the treatment of bladder cancer and the enhancement of immune function. © 2018 Society of Chemical Industry.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Isothiocyanates/pharmacology , Nanoparticles/chemistry , Urinary Bladder Neoplasms/physiopathology , Animals , Anti-Inflammatory Agents/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Humans , Isothiocyanates/chemistry , Macrophages/drug effects , Macrophages/immunology , Mice , RAW 264.7 Cells , Urinary Bladder Neoplasms/drug therapy
4.
J Biol Chem ; 292(38): 15952-15963, 2017 09 22.
Article in English | MEDLINE | ID: mdl-28794159

ABSTRACT

Bladder cancer (BC) is the sixth most common cancer in the United States and is the number one cause of death among patients with urinary system malignancies. This makes the identification of invasive regulator(s)/effector(s) as the potential therapeutic targets for managing BC a high priority. p63 is a member of the p53 family of tumor suppressor genes/proteins, plays a role in the differentiation of epithelial tissues, and is believed to function as a tumor suppressor. However, it remains unclear whether and how p63 functions in BC cell invasion after tumorigenesis. Here, we show that p63α protein levels were much higher in mouse high-invasive BC tissues than in normal tissues. Our results also revealed that p63α is crucial for heat shock protein 70 (Hsp70) expression and subsequently increases the ability of BC invasion. Mechanistic experiments demonstrated that p63α can transcriptionally up-regulate Hsp70 expression, thereby promoting BC cell invasion via the Hsp70/Wasf3/Wave3/MMP-9 axis. We further show that E2F transcription factor 1 (E2F1) mediates p63α overexpression-induced Hsp70 transcription. We also found that p63α overexpression activates E2F1 transcription, which appears to be stimulated by p63α together with E2F1. Collectively, our results demonstrate that p63α is a positive regulator of BC cell invasion after tumorigenesis, providing significant insights into the biological function of p63α in BC and supporting the notion that p63α might be a potential target for invasive BC therapy.


Subject(s)
E2F1 Transcription Factor/metabolism , HSP70 Heat-Shock Proteins/metabolism , Matrix Metalloproteinase 9/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation , Urinary Bladder Neoplasms/pathology , Wiskott-Aldrich Syndrome Protein Family/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , E2F1 Transcription Factor/genetics , HSP70 Heat-Shock Proteins/genetics , Humans , Male , Mice , Neoplasm Invasiveness , Signal Transduction , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
5.
Molecules ; 23(10)2018 Oct 10.
Article in English | MEDLINE | ID: mdl-30309003

ABSTRACT

Bladder cancer has become the most common malignant urinary carcinoma. Studies have shown that significant antioxidant and bladder cancer-fighting properties of several plant-based diets like Psidium guajava, ginger and amomum, are associated with their high kaempferol content. In this paper, we evaluated the antioxidant and anticancer activities of kaempferol and its mechanism of induction to apoptosis on bladder cancer cells. Our findings demonstrated that kaempferol showed an obvious radical scavenging activity in erythrocytes damaged by oxygen. Kaempferol promoted antioxidant enzymes, inhibited ROS generation and lipid peroxidation and finally prevented the occurrence of hemolysis. Additionally, kaempferol exhibited a strong inhibitory effect on bladder cancer cells and high safety on normal bladder cells. At the molecular level, kaempferol suppressed EJ bladder cancer cell proliferation by inhibiting the function of phosphorylated AKT (p-AKT), CyclinD1, CDK4, Bid, Mcl-1 and Bcl-xL, and promoting p-BRCA1, p-ATM, p53, p21, p38, Bax and Bid expression, and finally triggering apoptosis and S phase arrest. We found that Kaempferol exhibited strong anti-oxidant activity on erythrocyte and inhibitory effects on the growth of cancerous bladder cells through inducing apoptosis and S phase arrest. These findings suggested that kaempferol might be regarded as a bioactive food ingredient to prevent oxidative damage and treat bladder cancer.


Subject(s)
Apoptosis/drug effects , Hemolysis/drug effects , Kaempferols/pharmacology , Reactive Oxygen Species/metabolism , Urinary Bladder Neoplasms/pathology , Amidines/chemistry , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythrocytes/ultrastructure , Humans , Kaempferols/chemistry , Models, Biological , Oxidative Stress/drug effects , Protective Agents/pharmacology , S Phase/drug effects , Signal Transduction/drug effects
6.
Zhonghua Yi Xue Za Zhi ; 98(6): 457-459, 2018 Feb 06.
Article in Zh | MEDLINE | ID: mdl-29429260

ABSTRACT

Objective: To observe the effect of purple sweet potato anthocyanins on the proliferation of bladder cancer cell line BIU87 and to investigate the molecular mechanisms. Methods: Bladder cancer BIU87 cells were cultured and exposed to anthocyanins at the different concentrations of 100, 200, 400, and 800 µg/ml respectively. The growth inhibition of anthocyanins on BIU87 cells were evaluated by morphometry and cell counting kit-8 (CCK-8) assay, and the cell apoptosis rate was detected by Flow cytometry (FCM). Results: Morphometry showed that the number of BIU87 cells decreased, the volume shrank, the intercellular space enlarged, the ability of cell adherence weakened, and the cell shape changed when the concentration of anthocyanins increased. CCK-8 assay showed that when 100, 200, 400, 800 µg/ml anthocyanins treated BIU87 cells for 48 h, the absorbance was 24 ± 0.07, 1.15 ± 0.11, 0.90 ± 0.08, 0.56 ± 0.09, respectively. Compared with the control group, anthocyanins-treated groups significantly inhibited the proliferation of BIU87 cells (P<0.05). FCM test showed that after treatment with different doses of anthocyanins, the apoptosis rate was 7.31%, 11.11%, 25.96%, 36.28%, respectively, and with the concentration of anthocyanins being higher, the apoptosis rate of BIU87 cells was being higher. Conclusion: Purple sweet potato anthocyanins can inhibit the growth of bladder cancer BIU87 cells through inducing cell apoptosis in a dose-dependent manner.


Subject(s)
Urinary Bladder Neoplasms , Anthocyanins , Cell Line, Tumor , Humans , Ipomoea batatas
7.
Bioorg Med Chem Lett ; 26(20): 5082-5086, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27599742

ABSTRACT

A novel series of arylurea and arylamide derivatives 1a-z, 2a-d having aminoquinazoline scaffold was designed and synthesized. Their in vitro antiproliferative activities against RT112 bladder cancer cell line and inhibitory activities against FGFR3 kinase were tested. Most compounds showed good antiproliferative activities against RT112 bladder cancer cell line, and arylurea compounds 1a-z were more potent than arylamide compounds 2a-d. Among them, eight compounds 1a, 1d-g, 1l, 1y, and 1z showed potent activities with GI50 values below submicromolar range. Especially, arylurea compounds 1d and 1g possessing 2,3-dimethyl and 3,4-dimethyl moieties exhibited superior or similar antiproliferative activity (GI50=8.8nM and 30.2nM, respectively) to AZD4547 (GI50=29.2nM) as a reference standard.


Subject(s)
Cell Proliferation/drug effects , Quinazolines/chemical synthesis , Quinazolines/pharmacology , Urea/chemical synthesis , Urea/pharmacology , Urinary Bladder Neoplasms/pathology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Quinazolines/chemistry , Urea/chemistry
8.
Asian Pac J Cancer Prev ; 25(3): 921-930, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38546074

ABSTRACT

OBJECTIVE: Brucein D (BrD), a quassinoid isolated from Brucea javanica fruit, reportedly demonstrates anti-cancer activity. This study's objective is to evaluate the cytotoxicity of Brucein D and its ability to induce apoptosis in T24 bladder cancer cells. METHODS: We investigated the cytotoxic activity of BrD against the T24 cell through the induction of apoptosis in vitro. This cytotoxic activity was evaluated with ΜΤΤ assay and followed by Calcein-AM/PI viability staining. Apoptotic activity was determined with Hoechst 33342 nuclear staining and DNA fragmentation. Doxorubicin and docetaxel were used as a positive control. Evaluation of apoptotic-related gene expression, Bax, Bak, Bcl2, and p53 was also performed using semi-quantitative PCR analysis. Statistical analysis was conducted using One-way ANOVA followed by post hoc test Turkey's HSD (Honestly Significance Difference). RESULTS: Results show that BrD had high toxicity against T24 bladder cancer cells with an IC50 value of 7.65 ± 1.2 µg/mL but relatively less toxic to 1BR3 normal skin fibroblast cells compared to the doxorubicin and docetaxel treated cells. The viability assay shows that BrD significantly increases the percentage of dead cells relative to control in a dose-dependent manner. Furthermore, the percentage of cells with apoptotic appearance was significantly higher in group treated with BrD IC50 (56.04±3.09%) compared to control (9.42±2.88). The result was similar to doxorubicin IC50 (58.97±12.31) but lower than docetaxel IC50 (74.42±9.79). DNA fragmentation in gel electrophoresis was also observed in T24 cells treated with BrD. Apoptosis was also verified by an alteration in the expression of apoptosis-related genes, upregulation of Bax, Bak, and p53, and downregulation of Bcl-2. CONCLUSION: BrD has shown a cytotoxic effect against T24 bladder cancer cells. Hence, it is a promising natural compound for the management of bladder cancer by induction of apoptosis through activation of the intrinsic pathway, with low toxicity to normal cells.


Subject(s)
Antineoplastic Agents , Urinary Bladder Neoplasms , Humans , Docetaxel/therapeutic use , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/metabolism , Urinary Bladder Neoplasms/genetics , Apoptosis , Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Cell Line, Tumor
9.
Biomedicines ; 11(11)2023 Nov 01.
Article in English | MEDLINE | ID: mdl-38001959

ABSTRACT

Three-dimensional tumor models have gained significant importance in bladder cancer (BCa) research. Organoids consisting of different cell types better mimic solid tumors in terms of 3D architecture, proliferation, cell-cell interaction and drug responses. We developed four organoids from human BCa cell lines with fibroblasts and smooth muscle cells of the bladder, aiming to find models for BCa research. The organoids were characterized in terms of cytokeratins, vimentin, α-actin and KI67 by immunoreactivity. Further, we studied ligand-dependent activation of the Wnt/ß-catenin pathway and investigated the responses to anti-tumor therapies. The organoids mimicked the structure of an inverse bladder wall, with outside urothelial cells and a core of supportive cells. The cytokeratin staining patterns and proliferation rate were in conjunction with the origins of the BCa cells. RT-112 even showed stratification of the epithelium. Treatment with Wnt10B led to increased ß-catenin (active) levels in high-grade organoids, but not in low-grade BCa cells. Doxorubicin treatment resulted in clearly reduced viability (10-30% vs. untreated). In contrast, the effectivity of radiotherapy depended on the proliferation status of BCa cells. In conclusion, cell-line-based organoids can form bladder-like structures and reproduce in vivo features such as urothelial differentiation and stratification. Thus, they can be useful tools for functional studies in BCa and anti-cancer drug development.

10.
Front Mol Biosci ; 8: 789658, 2021.
Article in English | MEDLINE | ID: mdl-35252345

ABSTRACT

Bladder cancer (BC) ranks the fourth in incidence in cancers of men and is a common malignant tumor in women. 4-Methoxydalbergione (4MOD), which is purified from Dalbergia sissoo Roxb, has been shown to have anticancer capacity for osteosarcoma and astroglioma. The role of 4MOD in bladder cancer has not been investigated. This study aims to evaluate the anticancer effect of 4MOD in BC cells and its possible mechanisms. The two human bladder cancer cell lines J82 and UMUC3 were used to evaluate the proliferation inhibitory effect of 4MOD by CCK8 and clonogenic assays. The migratory and invasive ability of tumor cells was examined by scratch test and transwell assay. Apoptosis was detected by flow cytometry and TUNEL assays. The autophagy-related molecules including Beclin-1 and LC3 were examined by Western blotting analysis. Furthermore, the RT-PCR was used to detect the mRNA expression of LC3. 4MOD repressed cell proliferation, migration, invasion and induced cell apoptosis in a concentration-dependent manner. The IC50 values of J82 and UMUC3 were 8.17 and 14.50 µM respectively. The mRNA and protein expression ratio of light chain 3-II (LC3-II)/LC3-I and the protein expression of Beclin-1 were increased when the BC cells were treated with 4MOD. The treatment of 4MOD attenuated the phosphorylation of Akt and ERK in the BC cells. We revealed that the 4MOD inhibits BC cells growth by inducing autophagy and inhibiting Akt/ERK signaling pathway. Our study provides new insights into the mechanism by which 4MOD weakens the proliferation of BC cells. This study demonstrates that 4MOD provided a lead compound for the development of novel compound with potent anticancer effect on BC cells.

11.
Cancers (Basel) ; 13(1)2020 Dec 30.
Article in English | MEDLINE | ID: mdl-33396795

ABSTRACT

Bladder cancer is among the top ten most common cancer types in the world. Around 25% of all cases are muscle-invasive bladder cancer, for which the gold standard treatment in the absence of metastasis is the cystectomy. In recent years, trimodality treatment associating maximal transurethral resection and radiotherapy combined with concurrent chemotherapy is increasingly used as an organ-preserving alternative. However, the use of this treatment is still limited by the lack of biomarkers predicting tumour response and by a lack of targeted radiosensitising drugs that can improve the therapeutic index, especially by limiting side effects such as bladder fibrosis. In order to improve the bladder-preserving treatment, experimental studies addressing these main issues ought to be considered (both in vitro and in vivo studies). Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for systematic reviews, we conducted a literature search in PubMed on experimental studies investigating how to improve bladder cancer radiotherapy with different radiosensitising agents using a comprehensive search string. We made comments on experimental model selection, experimental design and results, formulating the gaps of knowledge still existing: such as the lack of reliable predictive biomarkers of tumour response to chemoradiation according to the molecular tumour subtype and lack of efficient radiosensitising agents specifically targeting bladder tumour cells. We provided guidance to improve forthcoming studies, such as taking into account molecular characteristics of the preclinical models and highlighted the value of using patient-derived xenografts as well as syngeneic models. Finally, this review could be a useful tool to set up new radiation-based combined treatments with an improved therapeutic index that is needed for bladder preservation.

12.
Biomed Pharmacother ; 124: 109830, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31972359

ABSTRACT

OBJECTIVE: Our study is to examine the citron rho-interacting, serine/threonine kinase 21 (CIT) in bladder cancer. METHODS: We examined CIT level in human bladder cancer tissues by immunohistochemical staining. To explore the impact of CIT on cell proliferation and apoptosis, we down-regulated its expression in two human bladder cancer cell lines, 5367 and T24. We examined cell growth in 5367 and T24. We also performed in vivo analysis using T24 cells. We further used microarray expression profiling to investigate genes differentially expressed in T24 cells with CIT down-regulated. RESULTS: In 100 human samples, CIT was expressed by only 2 of 30 (6.7 %) controls in bladder tissues, whereas by 64 of 70 (91.4 %) cancer patients in tumor tissues (p < 0.001). in vitro analysis demonstrated that CIT knockdown represses cell proliferation by 50 % in both cells and colony formation (77 ± 5 vs. 13 ± 2, p = 0.001 for T24, 58 ± 3 vs. 1 ± 1, p < 0.001 for 5637). We also found CIT knockdown could induce cell cycle arrest, and promote apoptosis in both cells. Tumor-volume monitoring and live in vivo bladder cancer imaging in human xenograft model confirmed that CIT knockdown reduces tumor volume (668.4 ± 333.0 vs. 305.7 ± 170.4 mm3, p = 0.02) and weight (0.27 ± 0.15 vs. 0.57 ± 0.32 g, p = 0.02). Microarray analysis revealed that CIT may regulate cell cycle signalling pathway through various cell cycle regulators. CONCLUSIONS: In summary, we provided clinical and experimental evidence that CIT may promote bladder cancer through regulation of cell cycle pathway.


Subject(s)
Apoptosis/genetics , Cell Proliferation/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Urinary Bladder Neoplasms/pathology , Aged , Animals , Down-Regulation , Female , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microarray Analysis , Middle Aged , Urinary Bladder Neoplasms/genetics , Xenograft Model Antitumor Assays
13.
J Clin Med ; 9(8)2020 Aug 03.
Article in English | MEDLINE | ID: mdl-32756427

ABSTRACT

The treatment of solid cancers with pharmacological all-trans retinoic acid (ATRA) concentrations, even if it is a gold standard therapy for the acute promyelocytic leukaemia (APL), is not always effective due to some resistance mechanisms. Here the resistance to ATRA treatment of T24 cell line, bladder cancer, was investigated. T24 was not only resistant to cell death when treated at concentrations up to 20 µM of ATRA, but it was also able to stimulate the cellular proliferation. An over-expression of the fatty acid binding protein 5 (FABP5) in conjunction with the cellular retinol-binding protein-II (CRABP-II) down-expression was found. However, the direct inhibition of the peroxisome proliferator-activated receptor ß/δ (PPARß/δ) did not abolish T24 proliferation, but rather potentiated it. Moreover, considering the ability of the long-chain fatty acids (LCFAs) to displace ATRA from FABP5, the actions of the saturated palmitic acid (PA), unsaturated omega-6 linoleic acid (LA) and omega-3 docosahexaenoic acid (DHA) were evaluated to counteract ATRA-related proliferation. ATRA-PA co-treatment induces cellular growth inhibition, while ATRA-LA co-treatment induces cellular growth enhancement. However, even if DHA is unsaturated LCFA as LA, it was able to reverse the ATRA-induced cellular proliferation of T24, bringing the viability percentages at the levels of the control.

14.
Chem Biol Interact ; 292: 65-75, 2018 Aug 25.
Article in English | MEDLINE | ID: mdl-29981725

ABSTRACT

BACKGROUND: Thymoquinone (TQ), the major active compound isolated from black seed oil (Nigella sativa), has been reported to exhibit anti-inflammatory and anticancer abilities. However, the exact molecular mechanism underlying the anticancer effect of TQ is still poorly understood, especially in regard to TQ's effect on endoplasmic reticulum stress-mediated apoptosis. METHODS: The cytotoxicity of TQ on T24 and 253J bladder cancer cells was analyzed by MTT assay, colony formation assay and Annexin V-FITC/PI staining, while SV-HUC-1, the SV-40 immortalized human uroepithelial cell line, was used as a normal control. The change in mitochondrial membrane potential was assessed by JC-1 staining. mRNA expression and protein expression were detected by qPCR and western blotting. Caspase-3 activity was detected by colorimetric assay. KEY FINDING: TQ has a significant cytotoxicity on bladder cancer cells and can inhibit their proliferation and induce apoptosis. The protein changes of Bcl-2, Bax, cytochrome c and endoplasmic reticulum stress-related proteins (GRP78, CHOP, and caspase-12) revealed that the anticancer effect of TQ was associated with mitochondrial dysfunction and the endoplasmic reticulum stress pathway. Pretreatment with a pan-caspase inhibitor, Z-VAD-fmk, or an ER stress inhibitor, 4-PBA, or knockdown of CHOP by shRNA can partly reverse the pro-apoptotic effect of TQ by enhancing the expression of the anti-apoptotic protein Bcl-2, blocking the release of cytochrome c and the translocation of Bax from the cytoplasm to mitochondria. SIGNIFICANCE: Our findings provide the first demonstration of the anticancer effect of TQ on bladder cancer, and the relationship between ER stress and mitochondrial dysfunction was clearly understood when the apoptosis progressed is revealed.


Subject(s)
Apoptosis/drug effects , Benzoquinones/pharmacology , Endoplasmic Reticulum Stress/drug effects , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Endoplasmic Reticulum Chaperone BiP , Humans , Models, Biological , Polymerase Chain Reaction , Signal Transduction/drug effects , Urinary Bladder Neoplasms/physiopathology
15.
Oncol Lett ; 15(3): 3814-3819, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29467898

ABSTRACT

The present study aimed to investigate the role of microRNA-96 (miR-96) in the proliferation, invasion and apoptosis of bladder cancer cell lines, and the associated mechanisms. The expression of miR-96 and human ether-à-go-go-related (HERG1) potassium channel in the normal uroepithelium SV-HUC-1 cell line, and bladder cancer T24 and 5637 cell lines were examined using reverse transcription-polymerase chain reaction or/and western blotting. Transfection with miR-96 inhibitor or scrambled control (SC) was used to study the biological activities of miR-96 in bladder cancer cell lines. MTT, flow cytometric and Transwell assays were applied to detect cell viability, apoptosis and invasion, respectively. A dual-luciferase reporter assay was applied to determine the association between miR-96 and HERG1 expression. As demonstrated, miR-96 was highly expressed in the two bladder cancer cell lines, particularly in T24 cells. Following transfection with miR-96 inhibitor, miR-96 expression was significantly reduced in the T24 cell line, compared with SC. The miR-96 inhibitor suppressed cell proliferation and invasion, promoted apoptosis and arrested the cell cycle at the G1 phase. Consistently, HERG1 was also highly expressed in the two bladder cancer cell lines at the mRNA and protein level, but not in the normal uroepithelium cell line. The miR-96 inhibitor also significantly decreased HERG1 expression compared with SC. The results of the dual-luciferase reporter assay indicated that miR-96 directly targeted wild-type HERG1. In conclusion, miR-96 inhibitor exhibited anticancer effects on bladder cancer cells by inhibiting proliferation and invasion of cells, and promoting their apoptosis. HERG1 was an important target of miR-96. These results provided experimental evidence supporting miR-96 as a therapeutic target for patients with bladder cancer.

16.
Oncol Lett ; 16(3): 2851-2856, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30127871

ABSTRACT

Past studies have confirmed that tumors can impair the function of dendritic cells (DCs) and promote tumor evasion. AG490, a Janus kinase 2/signal transducer and activator of transcription 3 inhibitor, has been shown to induce maturation of DCs and inhibit the growth of tumor cells. In the present study, DCs were generated from healthy human peripheral blood mononuclear cells. On day 5 of culture, the DCs were co-cultured with human bladder cancer pumc-91 cells for 24 h, and then purified using magnetic beads. The maturation of the DCs was induced by lipopolysaccharide. Subsequent to co-culture with pumc-91 cells, the expression of human leukocyte antigen-antigen D related (HLA-DR), cluster of differentiation (CD)86 and CD80 was found to be reduced in the DCs, accompanied by increased production of interleukin (IL)-10, but decreased production of IL-12p70. Furthermore, the DCs co-cultured with pumc-91 inhibited the proliferation of allogeneic T cells. Finally, AG490 restored the expression of HLA-DR, CD86 and CD80. These data identified that bladder cancer cells could inhibit the antigen-presenting function of the DCs and induce anergy in T cells. AG490 may partly reverse this inhibitory effect of bladder cancer cells on DCs, activate immunogenicity and induce the antitumor immunity response of DCs.

17.
Biomaterials ; 113: 93-104, 2017 01.
Article in English | MEDLINE | ID: mdl-27810645

ABSTRACT

Nonmuscle-invasive bladder cancer (NMIBC) is treated with transurethral resection followed by intravesical chemotherapy. However, drug-resistant tumorigenic cells cannot be eliminated, leading to half of the treated cancers recur with increased stage and grade. Innovative approaches to enhance drug sensitivity and eradicate tumorigenic cells in NMIBC treatment are urgently needed. Here, we show that pre-instillation of tumor cell-derived microparticles (T-MP) as natural biomaterials markedly enhance the inhibitory effects of intravesical chemotherapy on growth and hematuria occurrence of orthotropic bladder cancer in mice. We provide evidence that T-MPs enter and increase the pH value of lysosomes from 4.6 to 5.6, leading to the migration of drug-loaded lysosomes along microtubule tracks toward the nucleus and discharging the drugs whereby for the entry of the nucleus. We propose that T-MPs may function as a potent sensitizer for augmenting NMIBC chemotherapy with unprecedented clinical benefits.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell-Derived Microparticles/metabolism , Drug Carriers/metabolism , Lysosomes/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Urinary Bladder/drug effects , Animals , Antineoplastic Agents/therapeutic use , Biocompatible Materials/metabolism , Cell Line, Tumor , Cell-Derived Microparticles/pathology , Female , Humans , Lysosomes/drug effects , Lysosomes/pathology , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology
18.
Anticancer Res ; 37(4): 1737-1745, 2017 04.
Article in English | MEDLINE | ID: mdl-28373436

ABSTRACT

BACKGROUND/AIM: This study aimed to evaluate the in vitro efficacy of carboplatin and piroxicam, both in isolation and combined, against T24 and 5637 human urinary bladder cancer cell lines. MATERIALS AND METHODS: Cell viability, drug interaction, cell morphology, cell proliferation, apoptosis and autophagy were analyzed after 72 h of drug exposure. Statistical analysis was performed and values of p<0.05 were considered statistically significant. RESULTS: Drug exposure in combination led to a significant reduction of cell viability comparatively to single-drug exposure. These combinations resulted in a synergistic interaction in the T24 (combination index for 50% effect (CI50)=0.65) and 5637 (CI50=0.17) cell lines. Notable increase of morphological alterations, a marked decrease of Ki-67 expression, a considerable enhancement of autophagic vacuoles and a minimal effect on apoptosis was observed in both cell lines treated with combined drugs. CONCLUSION: Data showed that in vitro combination of carboplatin and piroxicam produced a more potent antiproliferative effect when compared to single drugs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carboplatin/pharmacology , Drug Synergism , Piroxicam/pharmacology , Urinary Bladder Neoplasms/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Drug Therapy, Combination , Humans , Tumor Cells, Cultured , Urinary Bladder Neoplasms/drug therapy
19.
Oncol Lett ; 14(5): 5149-5156, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29113153

ABSTRACT

Previous in vitro studies have demonstrated that LAG1 longevity assurance homolog 2 (LASS2) is a novel tumor suppressor gene that is significantly associated with the proliferation and invasion ability of tumor cells. However, the role LASS2 serves in regulating bladder cancer cell tumorigenicity and tumor growth in vivo has not yet been elucidated in animal or clinical studies. In the present study, LASS2 knockdown in human bladder cancer EJ-M3 cells significantly promoted the growth of xenografts in nude mice compared with the control group, while overexpression of LASS2 suppressed tumor growth; however, this was not statistically significant. Furthermore, LASS2 knockdown resulted in more apparent heteromorphism and a higher activity of matrix metalloproteinase (MMP)-2 and MMP-9 in xenograft tumors. The data from the present study demonstrated that LASS2 knockdown significantly promoted the tumorigenicity and growth of EJ-M3 xenograft tumors in nude mice, and that LASS2 overexpression has a tendency to inhibit the growth of xenografts, suggesting that it may be a potential therapeutic target for bladder cancer.

20.
Nanomedicine (Lond) ; 12(6): 597-613, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28186432

ABSTRACT

AIM: To develop a novel anti-CD47 single-chain variable fragment (scFv) functionalized magnetic nanoparticles (MNPs) for targeting bladder cell lines and its applicability in thermotherapy. MATERIAL & METHODS: An immunized murine antibody phage display library was constructed and screened to isolate anti-CD47 binders. A scFv was selected and conjugated to MNPs which was then utilized to discriminate CD47+ bladder cells along with assessing its efficacy in thermotherapy. RESULTS: An scFv with high affinity to bladder cells was efficiently conjugated to MNPs. Following a hyperthermia treatment, the function of scFv-MNP conjugates led to a considerable reduction in cell viability. CONCLUSION: The anti-CD47 scFv-MNP conjugate was an effective cancer cell thermotherapy tool that might pave the way for development of bionano-based targeting techniques in both early detection and treatment of cancer.


Subject(s)
CD47 Antigen/immunology , Magnetite Nanoparticles/administration & dosage , Single-Chain Antibodies/immunology , Urinary Bladder Neoplasms/drug therapy , Animals , CD47 Antigen/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Humans , Magnetite Nanoparticles/chemistry , Mice , Single-Chain Antibodies/therapeutic use , Urinary Bladder Neoplasms/immunology
SELECTION OF CITATIONS
SEARCH DETAIL