1
|
Hwang B, Kim J, Park S, Chung HJ, Kim H, Choi YH, Kim WJ, Myung SC, Jeong TB, Kim KM, Jung JC, Lee MW, Kim JW, Moon SK. Ethanol Extracts of Cornus alba Improve Benign Prostatic Hyperplasia by Inhibiting Prostate Cell Proliferation through Modulating 5 Alpha-Reductase/Androgen Receptor Axis-Mediated Signaling. World J Mens Health 2024; 42:830-841. [PMID: 38606866 PMCID: PMC11439794 DOI: 10.5534/wjmh.230200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/12/2023] [Accepted: 11/12/2023] [Indexed: 04/13/2024] Open
Abstract
PURPOSE The aim of this study was to investigate the efficacy of ethanol extracts of Cornus alba (ECA) against benign prostatic hyperplasia (BPH) in vitro and in vivo. MATERIALS AND METHODS The prostate stromal cells (WPMY-1) and epithelial cells (RWPE-1) were used to examine the action mechanism of ECA in BPH in vitro. ECA efficacy was evaluated in vivo using a testosterone propionate (TP)-induced BPH rat model. RESULTS Treatment with ECA inhibited the proliferation of prostate cells by inducing G1-phase cell cycle arrest through the regulation of positive and negative proteins. Treatment of prostate cells with ECA resulted in alterations in the mitogen-activated protein kinases and protein kinase B signaling pathways. The transcriptional binding activity of the NF-κB motif was suppressed in both ECA-treated prostate cells. In addition, treatment with ECA altered the level of BPH-associated axis markers (5α-reductase, fibroblast growth factor-2, androgen receptor, epidermal growth factor, Bcl-2, and Bax) in both cell lines. Finally, the administration of ECA attenuated the enlargement of prostatic tissues in the TP-induced BPH rat model, accompanied by histology, immunoblot, and serum dihydrotestosterone levels. CONCLUSIONS These results demonstrated that ECA exerted beneficial effects on BPH both in vitro and in vivo and might provide valuable information in the development of preventive or therapeutic agents for improving BPH.
Collapse
Affiliation(s)
- Byungdoo Hwang
- Department of Food and Nutrition, Chung-Ang University, Anseong, Korea
| | - Jongyeob Kim
- Department of Food and Nutrition, Chung-Ang University, Anseong, Korea
| | - Solbi Park
- Department of Food and Nutrition, Chung-Ang University, Anseong, Korea
| | - Hyun Joo Chung
- Department of Urology, Chung-Ang University College of Medicine, Seoul, Korea
- Molecular Biodesign Research Center, Chung-Ang University College of Medicine, Seoul, Korea
| | - Hoon Kim
- Department of Food and Nutrition, Chung-Ang University, Anseong, Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dong-Eui University, Busan, Korea
| | | | - Soon Chul Myung
- Department of Urology, Chung-Ang University College of Medicine, Seoul, Korea
- Molecular Biodesign Research Center, Chung-Ang University College of Medicine, Seoul, Korea
| | - Tae-Bin Jeong
- Life Science Research Institute, Novarex Co., Ltd., Cheongju, Korea
| | - Kyung-Mi Kim
- Life Science Research Institute, Novarex Co., Ltd., Cheongju, Korea
| | - Jae-Chul Jung
- Life Science Research Institute, Novarex Co., Ltd., Cheongju, Korea
| | - Min-Won Lee
- Laboratory of Pharmacognosy and Natural Product Derived Medicine, College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Jin Wook Kim
- Department of Medical Informatics, Chung-Ang University College of Medicine, Seoul, Korea
- Department of Urology, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, Korea.
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong, Korea
- Molecular Biodesign Research Center, Chung-Ang University College of Medicine, Seoul, Korea.
| |
Collapse
|
2
|
Kim JH, Ahn DH, Moon JS, Han HJ, Bae K, Yoon KA. Longitudinal assessment of B-RAF V595E levels in the peripheral cell-free tumor DNA of a 10-year-old spayed female Korean Jindo dog with unresectable metastatic urethral transitional cell carcinoma for monitoring the treatment response to a RAF inhibitor (sorafenib). Vet Q 2021; 41:153-162. [PMID: 33764261 PMCID: PMC8118428 DOI: 10.1080/01652176.2021.1905194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Transitional cell carcinoma (TCC) is the most common malignant tumor of the canine urinary tract. In this case study, a dog with metastatic urethral TCC was treated with sorafenib. The tumor expression levels of receptor tyrosine kinase genes, including VEGFR-1, VEGFR-2, PDGFR-α, PDGFR-β, ALK, EGFR, ErbB2, and B-RAF, were analyzed. VEGFR was overexpressed in tumor tissues compared to the normal tissues. Considering the high frequency of B-RAF mutation in canine urological tumors, the B-RAF gene was examined, and the B-RAF V595E mutation was detected in the tumor tissue. Therefore, the antitumor effect of sorafenib, a multi-tyrosine kinase inhibitor, on unresectable metastatic urethral TCC characterized by B-RAF V595E was evaluated and circulating cell-free tumor DNA (ctDNA) was assessed for monitoring the treatment response. After the initiation of oral sorafenib therapy (4 mg/kg/day escalated to 10 mg/kg/day), the dysuria was alleviated gradually, and the patient remained stable for 3 months. During that treatment period, the patient showed various levels of changes associated with B-RAF V595E mutation in ctDNA as evident from longitudinal plasma samples after initiation of sorafenib therapy. The findings of this study suggest that ctDNA may serve as a useful non-invasive tool for monitoring the treatment response to anticancer drugs.
Collapse
Affiliation(s)
- Jung-Hyun Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Dana Hyunjung Ahn
- Department of Veterinary Internal Medicine, Konkuk University Veterinary Medical Teaching Hospital, Seoul, South Korea
| | - Je-Sung Moon
- Veterinary Emergency Medicine and Critical Care, Konkuk University Veterinary Medical Teaching Hospital, Seoul, South Korea
| | - Hyun-Jung Han
- Veterinary Emergency Medicine and Critical Care, Konkuk University Veterinary Medical Teaching Hospital, Seoul, South Korea
| | - Kieun Bae
- Department of Veterinary Biochemistry, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Kyong-Ah Yoon
- Department of Veterinary Biochemistry, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| |
Collapse
|
3
|
Gupta I, Jabeen A, Vranic S, Al Moustafa AE, Al-Thawadi H. Oncoproteins of High-Risk HPV and EBV Cooperate to Enhance Cell Motility and Invasion of Human Breast Cancer Cells via Erk1/Erk2 and β-Catenin Signaling Pathways. Front Oncol 2021; 11:630408. [PMID: 33777781 PMCID: PMC7994530 DOI: 10.3389/fonc.2021.630408] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is a leading cause of death in women around the world. Most breast cancer-related deaths are a result of complications from the metastatic spread. Several recent studies reported that high-risk human papillomaviruses (HPVs) and Epstein-Barr virus (EBV) are co-presented in different types of human carcinomas including breast; however, the cooperative effects between high-risk HPVs and EBV oncoproteins in human breast cancer have not been investigated yet. Thus, we herein explored the cooperation outcome between E6/E7 and latent membrane protein 1 (LMP1) oncoproteins of high-risk HPV type 16 and EBV, respectively, in two human breast cancer cell lines, MCF7 and MDA-MB-231. Our data revealed that the cooperation of E6/E7 and LMP1 oncoproteins stimulates cell proliferation and deregulates cell cycle progression of human breast cancer and normal mammary cells; in parallel, we noted that E6/E7/LMP1 incite colony formation of both breast cancer cell lines but not normal cells. More significantly, our results point out that the co-expression of E6/E7 and LMP1 oncoproteins enhances cell motility and invasion of MCF7 and MDA-MB-231 cell lines; this is accompanied by deregulation of epithelial-mesenchymal transition biomarkers including E-cadherin, β-catenin, fascin, and vimentin. The molecular pathway analysis of HPV and EBV oncoproteins cooperation shows that it can enhance the phosphorylation of extracellular signal-regulated kinases (Erk1/Erk2) in addition to β-catenin, which could be behind the effect of this cooperation in our cell models. The study clearly suggests that high-risk HPV and EBV coinfection can play an important role in breast cancer progression via Erk1/Erk2 and β-catenin signaling pathways.
Collapse
Affiliation(s)
- Ishita Gupta
- College of Medicine, Qatar University Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University Health, Qatar University, Doha, Qatar
- Biomedical Research Centre, Qatar University, Doha, Qatar
| | - Ayesha Jabeen
- College of Medicine, Qatar University Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University Health, Qatar University, Doha, Qatar
- Biomedical Research Centre, Qatar University, Doha, Qatar
| | - Semir Vranic
- College of Medicine, Qatar University Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University Health, Qatar University, Doha, Qatar
| | - Ala-Eddin Al Moustafa
- College of Medicine, Qatar University Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University Health, Qatar University, Doha, Qatar
- Biomedical Research Centre, Qatar University, Doha, Qatar
| | - Hamda Al-Thawadi
- College of Medicine, Qatar University Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University Health, Qatar University, Doha, Qatar
| |
Collapse
|
4
|
Abdo GG, Gupta I, Kheraldine H, Rizeq B, Zagho MM, Khalil A, Elzatahry A, Al Moustafa AE. Mesoporous silica coated carbon nanofibers reduce embryotoxicity via ERK and JNK pathways. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111910. [PMID: 33641906 DOI: 10.1016/j.msec.2021.111910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 11/19/2022]
Abstract
Carbon nanofibers (CNFs) have been implicated in biomedical applications, yet, they are still considered as a potential hazard. Conversely, mesoporous silica is a biocompatible compound that has been used in various biomedical applications. In this regard, we recently reported that CNFs induce significant toxicity on the early stage of embryogenesis in addition to the inhibition of its angiogenesis. Thus, we herein use mesoporous silica coating of CNFs (MCNFs) in order to explore their outcome on normal development and angiogenesis using avian embryos at 3 days and its chorioallantoic membrane (CAM) at 6 days of incubation. Our data show that mesoporous silica coating of CNFs significantly reduces embryotoxicity provoked by CNFs. However, MCNFs exhibit slight increase in angiogenesis inhibition in comparison with CNFs. Further investigation revealed that MCNFs slightly deregulate the expression patterns of key controller genes involved in cell proliferation, survival, angiogenesis, and apoptosis as compared to CNFs. We confirmed these data using avian primary normal embryonic fibroblast cells established in our lab. Regarding the molecular pathways, we found that MCNFs downregulate the expression of ERK1/ERK2, p-ERK1/ERK2 and JNK1/JNK2/JNK3, thus indicating a protective role of MCNFs via ERK and JNK pathways. Our data suggest that coating CNFs with a layer of mesoporous silica can overcome their toxicity making them suitable for use in biomedical applications. Nevertheless, further investigations are required to evaluate the effects of MCNFs and their mechanisms using different in vitro and in vivo models.
Collapse
Affiliation(s)
- Ghada G Abdo
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar; Biomedical Research Centre, Qatar University, PO Box 2713, Doha, Qatar.
| | - Ishita Gupta
- Biomedical Research Centre, Qatar University, PO Box 2713, Doha, Qatar; College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar.
| | - Hadeel Kheraldine
- Biomedical Research Centre, Qatar University, PO Box 2713, Doha, Qatar; College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar.
| | - Balsam Rizeq
- Biomedical Research Centre, Qatar University, PO Box 2713, Doha, Qatar; College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar.
| | - Moustafa M Zagho
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS 39406, United States of America.
| | - Ashraf Khalil
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar.
| | - Ahmed Elzatahry
- Department of Materials Science and Technology Program, College of Arts and Sciences, Qatar University, PO Box 2713, Doha, Qatar.
| | - Ala-Eddin Al Moustafa
- Biomedical Research Centre, Qatar University, PO Box 2713, Doha, Qatar; College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar.
| |
Collapse
|
5
|
Zheng G, Sundquist J, Sundquist K, Ji J. Association of post-diagnostic use of cholera vaccine with survival outcome in breast cancer patients. Br J Cancer 2020; 124:506-512. [PMID: 33024264 PMCID: PMC7852596 DOI: 10.1038/s41416-020-01108-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/30/2020] [Accepted: 09/16/2020] [Indexed: 12/24/2022] Open
Abstract
Background Expensive cancer treatment calls for alternative ways such as drug repurposing to develop effective drugs. The aim of this study was to analyse the effect of post-diagnostic use of cholera vaccine on survival outcome in breast cancer patients. Methods Cancer diagnosis and cholera vaccination were obtained by linkage of several Swedish national registries. One vaccinated patient was matched with maximum two unvaccinated individuals based on demographic, clinical and socioeconomic factors. We performed proportional Cox regression model to analyse the differences in overall and disease-specific survivals between the matched patients. Results In total, 617 patients received cholera vaccine after breast cancer diagnosis. The median (interquartile range) time from diagnosis to vaccination was 30 (15–51) months and from vaccination to the end of follow-up it was 62 (47–85) months. Among them, 603 patients were matched with 1194 unvaccinated patients. Vaccinated patients showed favourable overall survival (hazard ratio (HR): 0.54, 95% confidence interval (CI): 0.37–0.79) and disease-specific survival (HR: 0.53, 95% CI: 0.33–0.84), compared to their unvaccinated counterpart. The results were still significant in multiple sensitivity analyses. Conclusions Post-diagnostic use of cholera vaccine is associated with a favourable survival rate in breast cancer patients; this provides evidence for repurposing it against breast cancer.
Collapse
Affiliation(s)
- Guoqiao Zheng
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden.
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden.,Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Center for Community-based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Shimane, Japan
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden.,Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Center for Community-based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Shimane, Japan
| | - Jianguang Ji
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| |
Collapse
|
6
|
Song JH, Hwang B, Chung HJ, Moon B, Kim JW, Ko K, Kim BW, Kim WR, Kim WJ, Myung SC, Moon SK. Peanut Sprout Extracts Cultivated with Fermented Sawdust Medium Inhibits Benign Prostatic Hyperplasia In Vitro and In Vivo. World J Mens Health 2020; 38:385-396. [PMID: 32202087 PMCID: PMC7308230 DOI: 10.5534/wjmh.190173] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/13/2020] [Accepted: 02/15/2020] [Indexed: 12/19/2022] Open
Abstract
PURPOSE In this study, we tested whether the resveratrol-enriched peanut sprout extracts cultivated with fermented sawdust medium (PSEFS) could suppress benign prostatic hyperplasia (BPH) in vitro and in vivo. MATERIALS AND METHODS The mode of action of PSEFS was estimated by employing high-performance liquid chromatography analysis, MTT assay, cell counting, cell cycle analysis, immunoblots, and immunoprecipitation and electrophoretic mobility shift assay. In vivo efficacy of PSEFS was analyzed in BPH animal model via immunostaining and enzyme-linked immunosorbent assay. RESULTS We selected the Yesan peanut sprout variety, which contains the highest level of resveratrol. The resveratrol levels in PSEFS were higher than those obtained with hydroponic technology. PSEFS treatment induced cell cycle arrest at the G1-phase by downregulating CDK4 and cyclin D1 via p21WAF1 induction in the RWPE-1 and WPMY prostate cells, thereby decreasing their proliferation. Treatment with PSEFS decreased ERK1/2 phosphorylation and increased JNK phosphorylation. The levels of DNA-bound transcription factors associated with proliferation (nuclear factor-κB, Sp-1, and AP-1) decreased upon PSEFS treatment in both prostate cells. Additionally, the levels of the molecular markers of BPH development (5α-reductase, androgen receptor, fibroblast growth factor, Bcl-2, and Bax) also changed by the addition of PSEFS. Finally, in a testosterone propionate-induced BPH model in rats, PSEFS administration attenuated the size, weight, and thickness of prostate tissues with no signs of death. CONCLUSIONS These results showed that PSEFS inhibited BPH both in vitro and in vivo and might be useful in the development of a potential BPH therapy.
Collapse
Affiliation(s)
- Jun Hui Song
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Korea
| | - Byungdoo Hwang
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Korea
| | - Hyun Joo Chung
- Department of Urology, Chung-Ang University College of Medicine, Seoul, Korea
- Bio-Integration Research Center for Nutra-Pharmaceutical Epigenetics, Chung-Ang University, Seoul, Korea
| | - BoKyung Moon
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Korea
- Bio-Integration Research Center for Nutra-Pharmaceutical Epigenetics, Chung-Ang University, Seoul, Korea
| | - Jin Wook Kim
- Department of Urology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Kisung Ko
- Department of Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | | | | | - Wun Jae Kim
- Department of Urology, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Soon Chul Myung
- Department of Urology, Chung-Ang University College of Medicine, Seoul, Korea
- Bio-Integration Research Center for Nutra-Pharmaceutical Epigenetics, Chung-Ang University, Seoul, Korea.
| | - Sung Kwon Moon
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Korea
- Bio-Integration Research Center for Nutra-Pharmaceutical Epigenetics, Chung-Ang University, Seoul, Korea.
| |
Collapse
|
7
|
Shin SS, Ko MC, Park YJ, Hwang B, Park SL, Kim WJ, Moon SK. Hydrangenol inhibits the proliferation, migration, and invasion of EJ bladder cancer cells via p21 WAF1-mediated G1-phase cell cycle arrest, p38 MAPK activation, and reduction in Sp-1-induced MMP-9 expression. EXCLI JOURNAL 2018; 17:531-543. [PMID: 30034317 PMCID: PMC6046626 DOI: 10.17179/excli2018-1361] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 06/04/2018] [Indexed: 01/07/2023]
Abstract
Hydrangenol is a dihydroisocoumarin that is mainly obtained from Hydrangea macrophylla. Recently, hydrangenol has garnered attention since several studies have reported that it has anti-inflammatory, anti-allergic, anti-diabetic, and anti-malarial activities. However, there have been few studies on the effect of hydrangenol on oncogenesis. In this study, we evaluated the anti-cancer activity of hydrangenol against the EJ bladder cancer cell line. Hydrangenol significantly inhibited the proliferation of EJ cells in a dose-dependent manner with an IC50 of 100 µM. Flow cytometry and immunoblotting experiments indicated that EJ cells were arrested in the G1-phase of the cell cycle and showed reduced expression of CDK2, CDK4, cyclin D1, and cyclin E mediated via the upregulation of p21WAF1. Hydrangenol increased the phosphorylation of p38 MAPK without affecting the phosphorylation of ERK and JNK. In addition, hydrangenol significantly inhibited the migratory and invasive activities of EJ cells by suppressing the enzymatic activity of MMP-9. Electrophoretic mobility shift assays suggested that the inhibition of MMP-9 activity by hydrangenol was attributable to its suppression of the Sp-1 transcription factor binding activity. This study is the first report on the mode of action of hydrangenol as an inhibitor of bladder cancer. We believe that these results provide novel insights that could aid the development of hydrangenol-based chemotherapeutic agents.
Collapse
Affiliation(s)
- Seung-Shick Shin
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, South Korea
| | - Myeong-Cheol Ko
- Department of ICT Convergence Engineering, College of Science and Technology, Konkuk University, Chungju, Chungbuk 27478, South Korea
| | - Yu-Jin Park
- Department of Food and Nutrition, Chung-Ang University, Anseong, Kyung-gi 17546, South Korea
| | - Byungdoo Hwang
- Department of Food and Nutrition, Chung-Ang University, Anseong, Kyung-gi 17546, South Korea
| | - Sung Lyea Park
- Department of Food and Nutrition, Chung-Ang University, Anseong, Kyung-gi 17546, South Korea
| | - Wun-Jae Kim
- Department of Urology, Chungbuk National University, Cheongju, Chungbuk 28644, South Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong, Kyung-gi 17546, South Korea
| |
Collapse
|
8
|
Shin SS, Noh DH, Hwang B, Lee JW, Park SL, Park SS, Moon B, Kim WJ, Moon SK. Inhibitory effect of Au@Pt-NSs on proliferation, migration, and invasion of EJ bladder carcinoma cells: involvement of cell cycle regulators, signaling pathways, and transcription factor-mediated MMP-9 expression. Int J Nanomedicine 2018; 13:3295-3310. [PMID: 29910616 PMCID: PMC5987858 DOI: 10.2147/ijn.s158463] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Although the diverse biological properties of nanoparticles have been studied intensively, research into their mechanism of action is relatively rare. In this study, we investigated the molecular mechanisms of the anticancer activity of heterometallic Au@Pt-nanoseeds (NSs) against bladder cancers. Materials and methods Mode of action of Au@Pt-NSs was investigated through MTT assay, flow cytometry analysis, Western immunoblots, real-time qPCR, wound-healing migration and invasion assays, zymography, and electrophoretic mobility shift assay (EMSA). Results Treatment with Au@Pt-NSs significantly inhibited the proliferation of EJ cells in a dose-dependent manner by inducing G1 phase cell cycle arrest. Among the regulators associated with the G1 cell cycle phase, CDK2, CDK4, cyclin D1, cyclin E, and p21WAF1 were shown to participate in the inhibitory pathways of Au@Pt-NSs. In addition, treatment with Au@Pt-NSs led to upregulation of phospho-p38 MAPK and downregulation of phospho-AKT in EJ cells. Interestingly, Au@Pt-NSs inhibited the migratory and invasive potential of the cells, which was attributed to the suppression of the enzymatic activity of matrix metalloproteinase-9 (MMP-9). Using MMP-9-specific oligonucleotides, we showed that transcription factors such as NF-κB and Sp-1 were responsible for the MMP-9-mediated metastatic potential of EJ cells. Conclusion Au@Pt-NSs significantly limited the progression, migration, and invasion of bladder cancer EJ cells. Our data represent a novel insight into developing cisplatin-like chemotherapeutic reagents with fewer side effects and provide useful information on molecular markers to monitor patients under Au@Pt-NSs-based chemotherapy.
Collapse
Affiliation(s)
- Seung-Shick Shin
- Department of Food Science and Nutrition, Jeju National University, Jeju, South Korea
| | - Dae-Hwa Noh
- Department of Food and Nutrition, Chung-Ang University, Anseong, South Korea
| | - Byungdoo Hwang
- Department of Food and Nutrition, Chung-Ang University, Anseong, South Korea
| | - Jo-Won Lee
- Department of Food and Nutrition, Chung-Ang University, Anseong, South Korea
| | - Sung Lyea Park
- Department of Food and Nutrition, Chung-Ang University, Anseong, South Korea
| | - Sung-Soo Park
- Department of Food Science and Nutrition, Jeju National University, Jeju, South Korea
| | - Bokyung Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong, South Korea
| | - Wun-Jae Kim
- Department of Urology, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong, South Korea
| |
Collapse
|
9
|
Shin SS, Hwang B, Lee SB, Kim WJ, Moon SK. Ethanol extract of loquat fruit skin inhibits the proliferation and metastatic potential of EJ human bladder carcinoma cells. Anim Cells Syst (Seoul) 2017. [DOI: 10.1080/19768354.2017.1358665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Seung-Shick Shin
- Department of Food Science and Nutrition, Jeju National University, Jeju, South Korea
| | - Byungdoo Hwang
- Department of Food and Nutrition, Chung-Ang University, Anseong, South Korea
| | - Soo-Bok Lee
- Department of Food and Nutrition, Yonsei University, Seoul, South Korea
| | - Wun-Jae Kim
- Department of Urology, Chungbuk National University, Cheongju, South Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong, South Korea
| |
Collapse
|
10
|
El-Aouar Filho RA, Nicolas A, De Paula Castro TL, Deplanche M, De Carvalho Azevedo VA, Goossens PL, Taieb F, Lina G, Le Loir Y, Berkova N. Heterogeneous Family of Cyclomodulins: Smart Weapons That Allow Bacteria to Hijack the Eukaryotic Cell Cycle and Promote Infections. Front Cell Infect Microbiol 2017; 7:208. [PMID: 28589102 PMCID: PMC5440457 DOI: 10.3389/fcimb.2017.00208] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/09/2017] [Indexed: 12/13/2022] Open
Abstract
Some bacterial pathogens modulate signaling pathways of eukaryotic cells in order to subvert the host response for their own benefit, leading to successful colonization and invasion. Pathogenic bacteria produce multiple compounds that generate favorable conditions to their survival and growth during infection in eukaryotic hosts. Many bacterial toxins can alter the cell cycle progression of host cells, impairing essential cellular functions and impeding host cell division. This review summarizes current knowledge regarding cyclomodulins, a heterogeneous family of bacterial effectors that induce eukaryotic cell cycle alterations. We discuss the mechanisms of actions of cyclomodulins according to their biochemical properties, providing examples of various cyclomodulins such as cycle inhibiting factor, γ-glutamyltranspeptidase, cytolethal distending toxins, shiga toxin, subtilase toxin, anthrax toxin, cholera toxin, adenylate cyclase toxins, vacuolating cytotoxin, cytotoxic necrotizing factor, Panton-Valentine leukocidin, phenol soluble modulins, and mycolactone. Special attention is paid to the benefit provided by cyclomodulins to bacteria during colonization of the host.
Collapse
Affiliation(s)
- Rachid A El-Aouar Filho
- STLO, Agrocampus Ouest Rennes, Institut National de la Recherche AgronomiqueRennes, France.,Departamento de Biologia Geral, Laboratório de Genética Celular e Molecular (LGCM), Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Aurélie Nicolas
- STLO, Agrocampus Ouest Rennes, Institut National de la Recherche AgronomiqueRennes, France
| | - Thiago L De Paula Castro
- Departamento de Biologia Geral, Laboratório de Genética Celular e Molecular (LGCM), Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Martine Deplanche
- STLO, Agrocampus Ouest Rennes, Institut National de la Recherche AgronomiqueRennes, France
| | - Vasco A De Carvalho Azevedo
- Departamento de Biologia Geral, Laboratório de Genética Celular e Molecular (LGCM), Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Pierre L Goossens
- HistoPathologie et Modèles Animaux/Pathogénie des Toxi-Infections Bactériennes, Institut PasteurParis, France
| | - Frédéric Taieb
- CHU Purpan USC INRA 1360-CPTP, U1043 Institut National de la Santé et de la Recherche Médicale, Pathogénie Moléculaire et Cellulaire des Infections à Escherichia coliToulouse, France
| | - Gerard Lina
- International Center for Infectiology ResearchLyon, France.,Centre National de la Recherche Scientifique, UMR5308, Institut National de la Santé et de la Recherche Médicale U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1Lyon, France.,Département de Biologie, Institut des Agents Infectieux, Hospices Civils de LyonLyon, France
| | - Yves Le Loir
- STLO, Agrocampus Ouest Rennes, Institut National de la Recherche AgronomiqueRennes, France
| | - Nadia Berkova
- STLO, Agrocampus Ouest Rennes, Institut National de la Recherche AgronomiqueRennes, France
| |
Collapse
|
11
|
Xie JY, Chen PC, Zhang JL, Gao ZS, Neves H, Zhang SD, Wen Q, Chen WD, Kwok HF, Lin Y. The prognostic significance of DAPK1 in bladder cancer. PLoS One 2017; 12:e0175290. [PMID: 28388658 PMCID: PMC5384764 DOI: 10.1371/journal.pone.0175290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 03/23/2017] [Indexed: 12/18/2022] Open
Abstract
Bladder cancer is one of the leading causes of cancer-related death in men, however, there was only limited effective treatment for invasive bladder cancer. DAPK1 has been shown to play important role in apoptosis and autophagy to suppress cancer progression. Previous results have shown that DAPK1 promoter was hypermethylated in the majority of bladder cancer specimens, however, the prognostic significance of DAPK1 in bladder cancer has yet to be demonstrated. In the present study, we found that DAPK1 expression was negatively associated with tumor stage and a low level expression of DAPK1 in bladder cancer specimens were associated with shorter survival in bladder cancer patients in 3 independent bladder cancer datasets (n = 462). Further investigation showed that FGFR3 knockdown resulted in downregulation of DAPK1 in bladder cancer cell line, suggesting that FGFR3 may be an upstream factor of DAPK1. Further analysis of the 3 independent bladder cancer datasets have identified ACOX1, UPK2, TRAK1, PLEKHG6 and MT1X genes had their expression significantly correlated with that of DAPK1. Knockdown of DAPK1 in bladder cancer T24 cells resulted in downregulation of ACOX1, UPK2 and TRAK1. Interestingly, TRAK1, by itself, was a favorable prognostic marker in the 3 independent bladder cancer datasets. Importantly, by using connectivity mapping with DAPK1-associated gene signature, we found that vemurafenib and trametinib could possibly reverse DAPK1-associated gene signature, suggesting that inhibition of Raf/MEK pathway may be a potential therapeutic approach for bladder cancer. Indeed, treatment of vemurafenib in T24 bladder cancer cells resulted in upregulation of DAPK1 confirming our connectivity mapping, while knockdown of DAPK1 resulted in reduced sensitivity towards inhibition of Braf signaling by vemurafenib. Together, our results suggest that DAPK1 is an important prognostic marker and therapeutic target for bladder cancer and have identified possible therapeutic agents for future testing in bladder cancer models with low DAPK1 expression.
Collapse
Affiliation(s)
- Jian-Yun Xie
- Department of Urology, The Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, People’s Republic of China
| | - Peng-Chen Chen
- College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, People’s Republic of China
| | - Jia-Li Zhang
- Department of Urology, The Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, People’s Republic of China
| | - Ze-Shou Gao
- Department of Urology, The Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, People’s Republic of China
| | - Henrique Neves
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR
| | - Shu-Dong Zhang
- Northern Ireland Centre for Stratified Medicine, University of Ulster, C-TRIC Building, Altnagelvin Hospital Campus, Glenshane Road, Londonderry, United Kingdom
| | - Qing Wen
- Centre for Cancer Research & Cell Biology, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Wei-Dong Chen
- Department of Urology, The Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, People’s Republic of China
- * E-mail: (YL); (HFK); (WDC)
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR
- * E-mail: (YL); (HFK); (WDC)
| | - Yao Lin
- College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, People’s Republic of China
- * E-mail: (YL); (HFK); (WDC)
| |
Collapse
|
12
|
Hildebrand D, Heeg K, Kubatzky KF. Pasteurella multocida Toxin Manipulates T Cell Differentiation. Front Microbiol 2015; 6:1273. [PMID: 26635744 PMCID: PMC4652077 DOI: 10.3389/fmicb.2015.01273] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/30/2015] [Indexed: 12/20/2022] Open
Abstract
Pasteurella multocida causes various diseases in a broad range of wild and domestic animals. Toxigenic strains of the serotypes A and D produce an AB protein toxin named Pasteurella multocida toxin (PMT). PMT constitutively activates the heterotrimeric G protein subunits Gαq, Gα13, and Gαi through deamidation of a glutamine residue, which results in cytoskeletal rearrangements as well as increased proliferation and survival of the host cell. In human monocytes, PMT alters the lipopolysaccharide (LPS)-induced activation toward a phenotype that suppresses T cell activation. Here we describe that the toxin also modulates CD4-positive T helper (Th) cells directly. PMT amplifies the expansion of Th cells through enhanced cell cycle progression and suppression of apoptosis and manipulates the differentiation of Th subclasses through activation of Signal Transducers and Activators of Transcription (STAT) family members and induction of subtype-specific master transcription factors. A large population of toxin-treated T cells is double-positive for Foxp3 and RORγt, the transcription factors expressed by Treg and Th17 cells, respectively. This suggests that these cells could have the potential to turn into Th17 cells or suppressive Treg cells. However, in terms of function, the PMT-differentiated cells behave as inflammatory Th17 cells that produce IL-17 and trigger T cell proliferation.
Collapse
Affiliation(s)
- Dagmar Hildebrand
- Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg Heidelberg, Germany
| | - Klaus Heeg
- Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg Heidelberg, Germany
| | - Katharina F Kubatzky
- Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Heidelberg Heidelberg, Germany
| |
Collapse
|