1
|
Terzi E, Oz-Bedir BE, Ercan E, Ozdemir-Sanci T, Jafarova S, Aydin T. β-Arbutin and cisplatin: A combined approach to modulating apoptosis, cell viability, and migration in bladder cancer cells. Toxicol In Vitro 2025; 104:105985. [PMID: 39638161 DOI: 10.1016/j.tiv.2024.105985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
One of the preferred treatments for bladder cancer, one of the most common cancers worldwide, is cisplatin-based chemotherapy. Since most tumor cells show cisplatin resistance, it is very important to discover new agents without adverse side effects. β-arbutin, a hydroquinone-β-D-glucopyranoside, has biological properties such as antioxidant, antimicrobial, anti-inflammatory, and anticancer, and is a phytochemical widely used as a skin whitener. In this study, β-arbutin was purified from the animal feed plant Onobrychis buhseana Boiss. (sainfoin). The study aimed to investigate the combined effects of cisplatin, a clinically used chemotherapeutic agent, and β-arbutin on HT-1376 bladder cancer cells for apoptosis, cell viability, and migration. In the study, after HT-1376 bladder cancer cells were cultured, optimum β-arbutin and cisplatin doses were determined on HT-1376 cells using the WST-1 test. To determine the apoptotic and migratory effects, flow cytometry and wound healing assays were performed. In HT-1376 cells, β-Arbutin led to greater apoptotoic and migratory effects when used alone and combined with Cisplatin (p < 0.0001 for apoptotic and migratory effects treated with β-Arbutin alone, p < 0.0001 for apoptotic and migratory effects when combined with Cisplatin). As a result, it can be suggested that β-arbutin may be a good drug candidate for treating bladder cancer.
Collapse
Affiliation(s)
- Emine Terzi
- Department of Medical Biology, Faculty of Medicine, Ankara Yildirim Beyazit University, Ankara, Türkiye; Ankara Yildirim Beyazit, University Yenimahalle Training and Research Hospital, Ankara, Türkiye
| | - Beyza Ecem Oz-Bedir
- Department of Medical Biology, Faculty of Medicine, Ankara Yildirim Beyazit University, Ankara, Türkiye; Ankara Yildirim Beyazit, University Yenimahalle Training and Research Hospital, Ankara, Türkiye
| | - Elif Ercan
- Department of Medical Biology, Faculty of Medicine, Ankara Yildirim Beyazit University, Ankara, Türkiye; Ankara Yildirim Beyazit, University Yenimahalle Training and Research Hospital, Ankara, Türkiye
| | - Tuba Ozdemir-Sanci
- Department of Histology and Embriyology, Faculty of Medicine, Ankara Yildirim Beyazit University, Ankara, Türkiye; Ankara Yildirim Beyazit, University Yenimahalle Training and Research Hospital, Ankara, Türkiye.
| | - Shahla Jafarova
- Department of Pharmaceutics, Faculty of Veterninary Medicine, Azerbaijan Dövlet Aqrar University, Ganja, Azerbaijan
| | - Tuba Aydin
- Department of Pharmacognosy, Faculty of Pharmacy, Agri İbrahim Cecen University, Agri, Türkiye
| |
Collapse
|
2
|
Liu T, Ul-Haq W, Tang Q, Li W, Wang Z, Shan Q, Serfraz S, Shakir Y, Ullah Kakar M, Sun L. Novel integrated Omics based computational approach for drug repurposing for non-muscle invasive bladder cancer (NMIBC). J Biomol Struct Dyn 2025; 43:2560-2570. [PMID: 38247255 DOI: 10.1080/07391102.2024.2302343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/04/2023] [Indexed: 01/23/2024]
Abstract
Non-muscle invasive bladder cancer (NMIBC) refers to a subtype of bladder carcinoma where cancer is localized in the inner lining of bladder. NMIBC consider as one of most costly malignancy and requires significant surgical and therapeutic measure. However, recurrence and progression of tumor is common in treated patients. Here we presented an integrated OMICs approach for the identification and inhibition of NMIBC specific genes. We utilized a case study where three group of patients were compared: 1) Relapsed tumors 2) recurrent tumors and 3) tumor in progression. Common transcriptome signature between patients facing recurrence and progression allowed us to identify three NMIBC specific genes FLT-1, WHSC-1 and CD34. We further utilized novel approach of Co-expressed gene-set enrichment analysis (COGENA) on the differentially expressed genes of this case study. Three drugs (paroxetine, adiphenine and H-89) with role of receptors inhibition were identified and predicted as repurposed drugs for the inhibition NMIBC specific genes. We further tested this hypothesis by performing molecular docking and simulation analysis between cancer specific proteins and drugs. FLT-1 have shown significant stable interaction with both drugs paroxetine and adiphenine whereas WHSC-1 have shown compact interaction with adiphenine and H-89. In the light of these evidence, we suggest that adiphenine could be repositioned as alternate targeted medicine for the treatment of NMIBC. In the future, this study will help for strengthening the strategies development at the molecular level for the control of carcinomas at early as well as detection of active and binding site, receptor-ligand interaction and also make drug repurposing for the early treatment of the carcinomas.
Collapse
Affiliation(s)
- Tiantian Liu
- The Department of Oncology, Affiliated with Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Waqar- Ul-Haq
- Evolutionary Biology Lab, CABB, University of Agriculture Faisalabad, Pakistan
| | - Qing Tang
- The Department of Oncology, Affiliated with Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Wei Li
- The Department of Oncology, Affiliated with Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Zhixia Wang
- The Department of Oncology, Affiliated with Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Qiujie Shan
- The Department of Oncology, Affiliated with Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Saad Serfraz
- Evolutionary Biology Lab, CABB, University of Agriculture Faisalabad, Pakistan
| | - Yasmeen Shakir
- Department of Biochemistry, Hazara University, Mansehra, Khyber Pakhtunkhua, Pakistan
| | - Mohib Ullah Kakar
- Faculty of Marine Sciences, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, Balochistan, Pakistan
| | - Lizhu Sun
- The Department of Oncology, Affiliated with Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| |
Collapse
|
3
|
Akakpo JY, Abbott E, Woolbright BL, Ramachandran A, Rick SG, Wallace DP, Taylor JA. 4-Methylpyrazole-mediated inhibition of Cytochrome P450 2E1 protects renal epithelial cells, but not bladder cancer cells, from cisplatin toxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.10.622845. [PMID: 39605334 PMCID: PMC11601237 DOI: 10.1101/2024.11.10.622845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Cisplatin is an effective chemotherapeutic drug for the treatment of bladder cancer, though cisplatin-induced nephrotoxicity (CIN) occurs in approximately 20-30% of patients, limiting its clinical use. Evidence has shown that cytochrome P450 2E1 (CYP2E1), a drug metabolism enzyme expressed in proximal tubules, mediates the production of reactive oxygen species (ROS) during cisplatin-induced injury. Previously, we showed that the repurposed drug 4-methylpyrazole (4MP; fomepizole) blocks CYP2E1 activity and prevents acetaminophen-induced liver injury. Here, we investigated the potential protective effects of 4MP against CIN. Male and female C57BL/6J mice were treated with a single 20 mg/kg dose of cisplatin for 3 days (acute) or 9 mg/kg/week for 4 weeks (repeated dosing regimen) with or without a co-treatment of 50 mg/kg 4MP. Our findings revealed that acute treatment with cisplatin induced severe histological tubular damage and elevated plasma BUN and creatinine levels in male mice, but not in female mice. This difference correlated with higher basal CYP2E1 expression in the kidneys of male mice compared to female mice. We also found that cisplatin increased renal CYP2E1 activity and that inhibition of CYP2E1 with 4MP significantly reduced cisplatin induced cell death in male mice and primary normal human kidney cells. By contrast, human bladder cancer cells do not express CYP2E1, and treatment with 4MP did not interfere with cisplatin anti-cancer effects in human bladder cancer HTB9 cells. This study highlights the critical role of CYP2E1 in CIN and suggests that its inhibition with 4MP in the kidney is a potential prophylactic therapeutic option to prevent CIN in bladder cancer patients without affecting its anti-neoplastic effect.
Collapse
|
4
|
Priya S, Islam MM, Kasana S, Kurmi BD, Gupta GD, Patel P. Therapeutic potential of chalcone-1,2,3-triazole hybrids as anti-tumour agents: a systematic review and SAR studies. Future Med Chem 2025; 17:449-465. [PMID: 39886772 PMCID: PMC11834451 DOI: 10.1080/17568919.2025.2458450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/06/2025] [Indexed: 02/01/2025] Open
Abstract
The study of chalcone-1,2,3-triazole hybrids for anticancer activity is quite a recent area of focus, primarily because of the increasing demand for developing new drugs to treat cancer. The chalcones and 1,2,3-triazole rings in hybrid compounds has recently emerged as a promising strategy for developing novel anticancer agents. The 1,2,3-triazole ring, known for its stability and hydrogen bonding capabilities, enhances the target binding affinity of these hybrids. Chalcones possess an α,β-unsaturated carbonyl system crucial for their anticancer activity The synergistic effect of these two moieties results in compounds with potent anticancer properties. This review explores the structure-activity relationship studies which revealed that the electronic and lipophilic properties of substituents on the phenyl rings of chalcones significantly influence their anticancer activity. Electron-donating and electron-withdrawing groups can affect cellular uptake and target engagement. Incorporating various substituents into the 1,2,3-triazole ring can improve selectivity and potency against specific cancer cell lines. These hybrids often exert their anticancer effects through apoptosis and cell cycle disruption. Recent research indicates 1,2,3-triazole chalcone hybrids hold therapeutic promise as anticancer agents. Further optimization through SAR studies and in-depth mechanistic investigations could lead to the development of highly potent and selective anticancer agents with minimal toxicity.
Collapse
Affiliation(s)
- Sakshi Priya
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| | - Md Mustahidul Islam
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| | - Shivani Kasana
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
- IK Gujral Punjab Technical University, Kapurthala, India
| | - Balak Das Kurmi
- IK Gujral Punjab Technical University, Kapurthala, India
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, India
| | | | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
- IK Gujral Punjab Technical University, Kapurthala, India
| |
Collapse
|
5
|
Kobylka P, Bakun P, Kuzminska J, Goslinski T, Murias M, Kucinska M. Insights into the Mode of Action of Novel Morpholinated Curcumin Derivatives Exhibiting Potent Antitumor Activity in Bladder Cancer Cells In Vitro. Molecules 2025; 30:295. [PMID: 39860164 PMCID: PMC11767817 DOI: 10.3390/molecules30020295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Although curcumin is a well-known natural polyphenol with many biological activities, its clinical application has been limited by low aqueous solubility and stability. Therefore, curcumin derivatives have been proposed to overcome these limitations and increase anticancer activity. This study tested curcumin derivatives with modified feruloyl moieties (2a and 2a-B) and the β-diketo moiety (2a-B) to better understand their anticancer mechanism against human bladder cancer cells. The anticancer activity of 2a and 2a-B was determined using MTT (hypoxic conditions) and LDH (normoxic conditions) assays. An ELISA-based protein panel was used to find the potential molecular targets, while flow cytometric, colorimetric, fluorescent, and luminescent assays were used to investigate the cell death mechanism. It was shown that compound 2a exerted a more potent cytotoxic effect under hypoxic conditions, while compound 2a-B demonstrated a comparable effect in normoxic and hypoxic conditions. The potential molecular targets modified by 2a and 2a-B depending on oxygen concentration were also proposed. Both compounds alter cell cycle progression by blocking the cell cycle in the G2/M phase and decreasing the percentage of cells in the G0/G1 phase. Compound 2a-B led to phosphatidylserine translocation, increased caspase 3/7 activity, and decreased mitochondrial membrane potential, suggesting a mitochondrial apoptosis pathway. We found that the Akt signaling pathway may modulate the activity of compound 2a-B, as evidenced by enhanced cytotoxic activity in combination with MK-2206, an Akt 1/2/3 inhibitor. Thus, our results provide new insights into the anticancer activity of compounds 2a and 2a-B; however, further studies are needed to better understand their therapeutic potential.
Collapse
Affiliation(s)
- Paulina Kobylka
- Department of Toxicology, Poznan University of Medical Sciences, Rokietnicka 3 Street, 60-806 Poznan, Poland; (P.K.); (M.M.)
| | - Pawel Bakun
- Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3 Street, 60-806 Poznan, Poland; (P.B.); (T.G.)
| | - Joanna Kuzminska
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3 Street, 60-806 Poznan, Poland;
| | - Tomasz Goslinski
- Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3 Street, 60-806 Poznan, Poland; (P.B.); (T.G.)
| | - Marek Murias
- Department of Toxicology, Poznan University of Medical Sciences, Rokietnicka 3 Street, 60-806 Poznan, Poland; (P.K.); (M.M.)
| | - Malgorzata Kucinska
- Department of Toxicology, Poznan University of Medical Sciences, Rokietnicka 3 Street, 60-806 Poznan, Poland; (P.K.); (M.M.)
| |
Collapse
|
6
|
Deng Q, Huang Y, Zeng J, Li X, Zheng X, Guo L, Shi J, Bai L. Recent advancements in the small-molecule drugs for hepatocellular carcinoma (HCC): Structure-activity relationships, pharmacological activities, and the clinical trials. Biomed Pharmacother 2024; 179:117343. [PMID: 39180795 DOI: 10.1016/j.biopha.2024.117343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND AND AIMS: Hepatocellular carcinoma (HCC) is one of the most common malignancies in the world and the sixth leading cause of cancer death worldwide, and it is urgent to find safe and effective drugs for treatment. As an important therapeutic method, small-molecule drugs are continually being updated to achieve improved therapeutic effects. The purpose of this study was to investigate the structural effects of various FDA-listed small-molecule drugs sorafenib, cabozantinib, lenvatinib, and regorafenib on the corresponding HCC targets and possible structural optimization methods, and to explore the mechanism for identifying potential therapeutic drugs that offer better efficacy and fewer side effects. METHODS The structure-activity relationship, pharmacological actions, and clinical applications of small-molecule drugs were reviewed by referencing MEDLINE, Web of Science, CNKI, and other databases, summarizing and integrating the relevant content. RESULTS The results showed that small-molecule drugs can inhibit HCC primarily by forming hydrogen bonds with Glu885, Asp1046, and Cys919 on the HCC target. HCC can be targeted by inhibiting the activation of multiple pathways, blocking the conduction of downstream signaling, and reducing the formation of tumor blood vessels. In general, small-molecule drugs primarily target four key receptors in HCC: VEGFR, PDGFR, EGFR, and FGFR, to achieve effective treatment. CONCLUSIONS By revealing their structure-activity relationships, pharmacological actions, and clinical trials, small-molecule drugs can offer broad prospects for the development of new medications.
Collapse
Affiliation(s)
- Qichuan Deng
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Jing Zeng
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Xinyu Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xianyi Zheng
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Guo
- The State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Lan Bai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; The State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
7
|
Liu T, Tang Q, Shan Q, Wang Z, Shakir Y, Waqar-Ul-Haq, Serfraz S, Sun L. Identification of potential phytochemical for the inhibition of non-muscle invasive bladder cancer (NMIBC). J Biomol Struct Dyn 2024; 42:8772-8780. [PMID: 37962848 DOI: 10.1080/07391102.2023.2280914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/07/2023] [Indexed: 11/15/2023]
Abstract
Non-muscle invasive bladder cancer (NMIBC) is one of the most common type of bladder cancer. Here, we have utilized an integrated transcriptomic-computational approach to identify alternate treatments to the NMIBC. In this study, we have performed the comprehensive comparative analysis between three groups of 36 patients with non-relapsed (NR), recurrence and progressive symptoms. Differentially expressed genes involved in the pathways associated with the NMIBC were identified. In silico protein-protein interaction (PPI) network was performed to create the network of the hub genes associated with NMIBC. Further, we compared NR individuals with two cohorts of patients with recurrent and progressive symptoms that lead to the identification of three major biomarkers CD34, FLT1 and WHSC1 genes. Concurrently, PPI also suggests that they are significant hub genes responsible for disease recurrence and progression. Furthermore, targeted genes WHSC-1 and FLT-1 were subjected to virtual screening for identification phytochemical inhibitors. Docking and molecular dynamics simulations concluded that the phytochemicals anonymously named 'UNK' and '6-hydroxycyanidin' are suitable for the inhibition of the proteins causing the NMIBC. In the future, this study will help for strengthening the strategies development at the molecular level for the control of carcinomas at early as well as detection of active and binding site, receptor-ligand interaction and also make drug designing for the early treatment of the carcinomas.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Tiantian Liu
- The Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, China
| | - Qing Tang
- The Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, China
| | - Qiujie Shan
- The Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, China
| | - Zhixia Wang
- The Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, China
| | - Yasmeen Shakir
- Department of Biochemistry, Hazara University, Mansehra, KPK, Pakistan
| | - Waqar-Ul-Haq
- Evolutionary Biology Lab, CABB, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Saad Serfraz
- Evolutionary Biology Lab, CABB, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Lizhu Sun
- The Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, China
| |
Collapse
|
8
|
Wu J, Zhang F, Zheng X, Chen D, Li Z, Bi Q, Qiu X, Sun Z, Wang W. Identification of bladder cancer subtypes and predictive signature for prognosis, immune features, and immunotherapy based on immune checkpoint genes. Sci Rep 2024; 14:14431. [PMID: 38910160 PMCID: PMC11194261 DOI: 10.1038/s41598-024-65198-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 06/18/2024] [Indexed: 06/25/2024] Open
Abstract
Immunotherapy based on immune checkpoint genes (ICGs) has recently made significant progress in the treatment of bladder cancer patients, but many patients still cannot benefit from it. In the present study, we aimed to perform a comprehensive analysis of ICGs in bladder cancer tissues with the aim of evaluating patient responsiveness to immunotherapy and prognosis. We scored ICGs in each BLCA patient from TCGA and GEO databases by using ssGSEA and selected genes that were significantly associated with ICGs scores by using the WCGNA algorithm. NMF clustering analysis was performed to identify different bladder cancer molecular subtypes based on the expression of ICGs-related genes. Based on the immune related genes differentially expressed among subgroups, we further constructed a novel stratified model containing nine genes by uni-COX regression, LASSO regression, SVM algorithm and multi-COX regression. The model and the nomogram constructed based on the model can accurately predict the prognosis of bladder cancer patients. Besides, the patients classified based on this model have large differences in sensitivity to immunotherapy and chemotherapy, which can provide a reference for individualized treatment of bladder cancer.
Collapse
Affiliation(s)
- Jiyue Wu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gong Ti Nan Road, Chaoyang District, Beijing, 100020, China
| | - Feilong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gong Ti Nan Road, Chaoyang District, Beijing, 100020, China
| | - Xiang Zheng
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gong Ti Nan Road, Chaoyang District, Beijing, 100020, China
| | - Dongshan Chen
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gong Ti Nan Road, Chaoyang District, Beijing, 100020, China
| | - Zhen Li
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gong Ti Nan Road, Chaoyang District, Beijing, 100020, China
| | - Qing Bi
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gong Ti Nan Road, Chaoyang District, Beijing, 100020, China
| | - Xuemeng Qiu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gong Ti Nan Road, Chaoyang District, Beijing, 100020, China
| | - Zejia Sun
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gong Ti Nan Road, Chaoyang District, Beijing, 100020, China.
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gong Ti Nan Road, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
9
|
Yang K, Li X, Jiang Z, Li J, Deng Q, He J, Chen J, Li X, Xu S, Jiang Z. Tumour suppressor ABCA8 inhibits malignant progression of colorectal cancer via Wnt/β-catenin pathway. Dig Liver Dis 2024; 56:880-893. [PMID: 37968146 DOI: 10.1016/j.dld.2023.10.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/23/2023] [Accepted: 10/28/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most commonly diagnosed malignant tumours of the digestive tract, and new therapeutic targets and prognostic markers are still urgently required. However, the role and molecular mechanisms of ATP binding cassette subfamily A member 8 (ABCA8) in CRC remain unclear. METHODS Databases and clinical specimens were analysed to determine the expression level of ABCA8 in CRC. Colony formation, CCK-8 and Transwell assays were conducted to determine cell proliferation, viability, migration and invasion. Flow cytometry was used to detect cell cycle progression and apoptosis. Western blot and rescue experiments were performed to determine the specific mechanisms of action of ABCA8. RESULTS ABCA8 expression is dramatically down-regulated in CRC tissues and cell lines. Ectopic expression of ABCA8 induced apoptosis and cell cycle arrest in vitro, inhibited cell growth, suppressed migration and invasion, reversed epithelial-mesenchymal transition and suppressed xenograft tumour growth and metastasis in vivo. Mechanistically, ABCA8 inhibited CRC cell proliferation and metastasis through the Wnt/β-catenin signalling pathway, both in vitro and in vivo. CONCLUSION We verified that ABCA8 inhibits the malignant progression of CRC through the Wnt/β-catenin pathway. This newly discovered ABCA8-Wnt-β-catenin signalling axis is probably helpful in guiding the clinical diagnosis and treatment of CRC.
Collapse
Affiliation(s)
- Kun Yang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaolu Li
- Department of Respiratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Zhongxiang Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Junfeng Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qianxi Deng
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jin He
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jun Chen
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaoqing Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shuman Xu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zheng Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
10
|
Wang X, Luo L, Xu J, Lu Q, Xia H, Huang Y, Zhang L, Xie L, Jiwa H, Liang S, Luo X, Luo J. Echinatin inhibits tumor growth and synergizes with chemotherapeutic agents against human bladder cancer cells by activating p38 and suppressing Wnt/β-catenin pathways. Genes Dis 2024; 11:1050-1065. [PMID: 37692489 PMCID: PMC10491917 DOI: 10.1016/j.gendis.2023.03.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/13/2023] [Accepted: 03/29/2023] [Indexed: 09/12/2023] Open
Abstract
Bladder cancer (BC) is one of the most common malignant tumors in the urinary system. Due to the poor prognosis and high mortality rate of the disease, it is urgent to develop new drugs with high efficacy and low toxicity to treat BC. Echinatin (Ecn) is a bioactive natural flavonoid oflicorice that has attracted special attention for its promising anti-tumor potential. Herein, we explored the inhibitory effects of Echinatin on BC cells and probed the possible molecular mechanism. We found that Ecnin vitro inhibited the proliferation, migration, and invasion, arrested the cell cycle at the G2/M phase, and promoted apoptosis in BC cells. Besides, Ecn had no notable cytotoxicity towards human normal cells. We subsequently confirmed that Ecn restrained xenograft tumor growth and metastasis of BC cells in vivo. Mechanistically, Ecn activated the p38 signaling pathway but inactivated the Wnt/β-catenin signaling pathway, while over-expression of β-catenin and the p38 inhibitor both attenuated the inhibitory effects of Ecn on BC cells. Remarkably, Ecn combined with cisplatin (DDP) or gemcitabine (Gem) had synergistic inhibitory effects on BC cells. In summary, our results validate that Ecn inhibits the tumor growth of human BC cells via p38 and Wnt/β-catenin signaling pathways. More meaningfully, our results suggest a potential strategy to enhance DDP- or Gem-induced inhibitory effects on BC cells by combining with Ecn.
Collapse
Affiliation(s)
- Xiaoxuan Wang
- Key Laboratory of Diagnostic Medicine Designated By the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lijuan Luo
- Key Laboratory of Diagnostic Medicine Designated By the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jingtao Xu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Qiuping Lu
- Key Laboratory of Diagnostic Medicine Designated By the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Haichao Xia
- Key Laboratory of Diagnostic Medicine Designated By the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yanran Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Lulu Zhang
- Key Laboratory of Diagnostic Medicine Designated By the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Liping Xie
- Key Laboratory of Diagnostic Medicine Designated By the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Habu Jiwa
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Shiqiong Liang
- Key Laboratory of Diagnostic Medicine Designated By the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Jinyong Luo
- Key Laboratory of Diagnostic Medicine Designated By the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
11
|
Zhou X, Xue F, Li T, Xue J, Yue S, Zhao S, Lu H, He C. Exploration of potential biomarkers for early bladder cancer based on urine proteomics. Front Oncol 2024; 14:1309842. [PMID: 38410113 PMCID: PMC10894981 DOI: 10.3389/fonc.2024.1309842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024] Open
Abstract
Background Bladder cancer is a common malignant tumor of the urinary system. The progression of the condition is associated with a poor prognosis, so it is necessary to identify new biomarkers to improve the diagnostic rate of bladder cancer. Methods In this study, 338 urine samples (144 bladder cancer, 123 healthy control, 32 cystitis, and 39 upper urinary tract cancer samples) were collected, among which 238 samples (discovery group) were analyzed by LC-MS. The urinary proteome characteristics of each group were compared with those of bladder cancer, and the differential proteins were defined by bioinformatics analysis. The pathways and functional enrichments were annotated. The selected proteins with the highest AUC score were used to construct a diagnostic panel. One hundred samples (validation group) were used to test the effect of the panel by ELISA. Results Compared with the healthy control, cystitis and upper urinary tract cancer samples, the number of differential proteins in the bladder cancer samples was 325, 158 and 473, respectively. The differentially expressed proteins were mainly related to lipid metabolism and iron metabolism and were involved in the proliferation, metabolism and necrosis of bladder cancer cells. The AUC of the panel of APOL1 and ITIH3 was 0.96 in the discovery group. ELISA detection showed an AUC of 0.92 in the validation group. Conclusion This study showed that urinary proteins can reflect the pathophysiological changes in bladder cancer and that important molecules can be used as biomarkers for bladder cancer screening. These findings will benefit the application of the urine proteome in clinical research.
Collapse
Affiliation(s)
- Xu Zhou
- Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Fei Xue
- Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Tingmiao Li
- Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiangshan Xue
- Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Siqi Yue
- Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shujie Zhao
- Department of Laboratory Medicine, Changchun Infectious Diseases Hospital, Changchun, China
| | - Hezhen Lu
- Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chengyan He
- Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
12
|
Gill E, Perks CM. Mini-Review: Current Bladder Cancer Treatment-The Need for Improvement. Int J Mol Sci 2024; 25:1557. [PMID: 38338835 PMCID: PMC10855537 DOI: 10.3390/ijms25031557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Bladder cancer is the tenth most common cancer and is a significant burden on health care services worldwide, as it is one of the most costly cancers to treat per patient. This expense is due to the extensive treatment and follow-ups that occur with costly and invasive procedures. Improvement in both treatment options and the quality of life these interventions offer has not progressed at the rates of other cancers, and new alternatives are desperately needed to ease the burden. A more modern approach needs to be taken, with urinary biomarkers being a positive step in making treatments more patient-friendly, but there is still a long way to go to make these widely available and of a comparable standard to the current treatment options. New targets to hit the major signalling pathways that are upregulated in bladder cancer, such as the PI3K/AkT/mTOR pathway, are urgently needed, with only one drug approved so far, Erdafitinib. Immune checkpoint inhibitors also hold promise, with both PD-1 and CDLA-4 antibody therapies approved for use. They effectively block ligand/receptor binding to block the immune checkpoint used by tumour cells. Other avenues must be explored, including drug repurposing and novel biomarkers, which have revolutionised this area in other cancers.
Collapse
Affiliation(s)
| | - Claire M. Perks
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK;
| |
Collapse
|
13
|
Li Z, Yang J, Chen L, Chen P, Liu C, Long X, Chen B, Long J. Moscatilin Reverses EMT Progression and its Resulting Enhanced Invasion and Migration by Affecting the TGF-β Signaling Pathway in Bladder Cancer. Anticancer Agents Med Chem 2024; 24:1074-1084. [PMID: 38808719 DOI: 10.2174/0118715206307769240522075729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/28/2024] [Accepted: 05/10/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Bladder cancer metastasis is an essential process in the progression of muscle-invasive bladder cancer. EMT plays a crucial role in facilitating the spread of cancer cells. Identifying compounds that can inhibit these abilities of cancer cells is a significant international endeavor. OBJECTIVE To explore the migration and invasion effect of Moscatilin on the bladder and clarify the mechanism of action Methods: The anti-bladder cancer effect of Moscatilin was observed by a cell proliferation experiment. The migration and invasion of bladder cancer cells inhibited by Moscatilin were detected by Transwell and Wound healing. The effects of Moscatilin on EMT-related proteins E-cadherin, N-cadherin, Snail1, Vimentin, and TGF-β signaling pathways were detected by Western blot, and nucleic acid levels were verified by qPCR. RESULTS Our study revealed that Moscatilin reduced the viability of bladder cancer cells in vitro and impeded their migration and invasion in experimental settings. Furthermore, we observed that Moscatilin decreased the activation levels of active proteins, specifically Smad3, Samd2, and MMP2. Additionally, we found that moscatilin significantly reduced the expression level of TGF-β and was also capable of reversing the overexpression effect of TGF-β. Treatment with Moscatilin also led to significant inhibition of interstitial cell markers Ncadherin and Snail1, which are associated with EMT. CONCLUSION These findings indicate that Moscatilin impedes the migration and invasion of bladder cancer cells by influencing cell survival, modulating TGF-β/Smad signaling, and inhibiting EMT.
Collapse
Affiliation(s)
- Zhihao Li
- College of Pharmacy, Chengdu University, Chengdu, China
| | - Jin Yang
- Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, China
| | - Lin Chen
- Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, China
| | - Pei Chen
- Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, China
| | - Chenhuan Liu
- Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, China
| | - Xiaoming Long
- Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, China
| | - Bo Chen
- Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, China
| | - Jun Long
- Clinical Medical College and Affiliated Hospital, Chengdu University, Chengdu, China
| |
Collapse
|
14
|
Celik S, Aktas T, Gokbayrak O, Erol A, Yorukoglu K, Yilmaz B, Sari H, Altun Z, Mungan MU, Celebi I, Aslan G, Aktas S. Genomic Alterations of Signaling and DNA Damage Repair Pathways in Non-Muscle Invasive Bladder Cancer. Cancer Invest 2023; 41:848-857. [PMID: 37997757 DOI: 10.1080/07357907.2023.2288640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 11/23/2023] [Indexed: 11/25/2023]
Abstract
The aim of the study was to demonstrate the most common genetic alterations and evaluate possible targets involving phosphatidylinositol-3-OH kinase (PIK3)/AKT/mammalian target of rapamycin (mTOR) signaling and DNA damage repair (DDR) pathways for personalized treatment in patients with non-muscle invasive bladder cancer (NMIBC). Alterations of these pathways were observed in 89.5% and 100% of patients, respectively. Among them, BARD1 was more frequently altered in low/intermediate-risk cases, but PARP4 was more frequently affected in intermediate/high-risk patients. The possible target feasibility of BARD1 and PARP4 alterations should be evaluated for personalized treatment using PARP-inhibitors in NMIBC. It is important to detect high tumor mutation burden (TMB) in patients in terms of immunotherapy.
Collapse
Affiliation(s)
- Serdar Celik
- Department of Urology, Izmir Faculty of Medicine, Health Sciences University, Izmir Bozyaka Education and Research Hospital, Izmir, Turkey
- Department of Basic Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Turkey
| | - Tekincan Aktas
- Department of Basic Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Turkey
| | - Ozde Gokbayrak
- Department of Basic Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Turkey
| | - Aylin Erol
- Department of Basic Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Turkey
| | - Kutsal Yorukoglu
- Department of Pathology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Batuhan Yilmaz
- Department of Urology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Hilmi Sari
- Department of Urology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Zekiye Altun
- Department of Basic Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Turkey
| | - Mehmet Ugur Mungan
- Department of Urology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Ilhan Celebi
- Department of Urology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Guven Aslan
- Department of Urology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Safiye Aktas
- Department of Basic Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
15
|
Bharathiraja P, Yadav P, Sajid A, Ambudkar SV, Prasad NR. Natural medicinal compounds target signal transduction pathways to overcome ABC drug efflux transporter-mediated multidrug resistance in cancer. Drug Resist Updat 2023; 71:101004. [PMID: 37660590 PMCID: PMC10840887 DOI: 10.1016/j.drup.2023.101004] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/11/2023] [Accepted: 08/19/2023] [Indexed: 09/05/2023]
Abstract
ATP-binding cassette (ABC) transporters such as ABCB1, ABCG2, and ABCC1 are the major players in drug efflux-mediated multidrug resistance (MDR), which severely affects the efficacy of chemotherapy. Several synthetic compounds block the drug transport by ABC transporters; however, they exhibit a narrow therapeutic window, and produce side effects in non-target normal tissues. Conversely, the downregulation of the expression of ABC drug transporters seems to be a promising strategy to reverse MDR in cancer cells. Several signaling pathways, such as NF-κB, STAT3, Gli, NICD, YAP/TAZ, and Nrf2 upregulate the expression of ABC drug transporters in drug-resistant cancers. Recently, natural medicinal compounds have gained importance to overcome the ABC drug-efflux pump-mediated MDR in cancer. These compounds target transcription factors and the associated signal transduction pathways, thereby downregulating the expression of ABC transporters in drug-resistant cancer cells. Several potent natural compounds have been identified as lead candidates to synergistically enhance chemotherapeutic efficacy, and a few of them are already in clinical trials. Therefore, modulation of signal transduction pathways using natural medicinal compounds for the reversal of ABC drug transporter-mediated MDR in cancer is a novel approach for improving the efficiency of the existing chemotherapeutics. In this review, we discuss the modulatory role of natural medicinal compounds on cellular signaling pathways that regulate the expression of ABC transporters in drug-resistant cancer cells.
Collapse
Affiliation(s)
- Pradhapsingh Bharathiraja
- Department of Biochemistry & Biotechnology, Annamalai University, Annamalai Nagar 608 002, Tamil Nadu, India
| | - Priya Yadav
- Department of Biochemistry & Biotechnology, Annamalai University, Annamalai Nagar 608 002, Tamil Nadu, India
| | - Andaleeb Sajid
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD 20892-4256, USA
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD 20892-4256, USA.
| | - N Rajendra Prasad
- Department of Biochemistry & Biotechnology, Annamalai University, Annamalai Nagar 608 002, Tamil Nadu, India.
| |
Collapse
|
16
|
Zuo M, Chen H, Liao Y, He P, Xu T, Tang J, Zhang N. Sulforaphane and bladder cancer: a potential novel antitumor compound. Front Pharmacol 2023; 14:1254236. [PMID: 37781700 PMCID: PMC10540234 DOI: 10.3389/fphar.2023.1254236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/07/2023] [Indexed: 10/03/2023] Open
Abstract
Bladder cancer (BC) is a common form of urinary tract tumor, and its incidence is increasing annually. Unfortunately, an increasing number of newly diagnosed BC patients are found to have advanced or metastatic BC. Although current treatment options for BC are diverse and standardized, it is still challenging to achieve ideal curative results. However, Sulforaphane, an isothiocyanate present in cruciferous plants, has emerged as a promising anticancer agent that has shown significant efficacy against various cancers, including bladder cancer. Recent studies have demonstrated that Sulforaphane not only induces apoptosis and cell cycle arrest in BC cells, but also inhibits the growth, invasion, and metastasis of BC cells. Additionally, it can inhibit BC gluconeogenesis and demonstrate definite effects when combined with chemotherapeutic drugs/carcinogens. Sulforaphane has also been found to exert anticancer activity and inhibit bladder cancer stem cells by mediating multiple pathways in BC, including phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR), mitogen-activated protein kinase (MAPK), nuclear factor kappa-B (NF-κB), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), zonula occludens-1 (ZO-1)/beta-catenin (β-Catenin), miR-124/cytokines interleukin-6 receptor (IL-6R)/transcription 3 (STAT3). This article provides a comprehensive review of the current evidence and molecular mechanisms of Sulforaphane against BC. Furthermore, we explore the effects of Sulforaphane on potential risk factors for BC, such as bladder outlet obstruction, and investigate the possible targets of Sulforaphane against BC using network pharmacological analysis. This review is expected to provide a new theoretical basis for future research and the development of new drugs to treat BC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Neng Zhang
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
17
|
Surendran H, Palaniyandi T, Natarajan S, Hari R, Viwanathan S, Baskar G, Abdul Wahab MR, Ravi M, Rajendran BK. Role of homeobox d10 gene targeted signaling pathways in cancers. Pathol Res Pract 2023; 248:154643. [PMID: 37406379 DOI: 10.1016/j.prp.2023.154643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/07/2023]
Abstract
Homeobox D10 (HOXD10) is a transcription factor from the homeobox gene family that controls cell differentiation and morphogenesis throughout development.Due to their functional interaction, changes in HOXD10 gene expression might induce tumors. This narrative review focuses on how and why the dysregulation in the signaling pathways linked with HOXD10 contributes to the metastatic development of cancer. Organ development and tissue homeostasis need highly conserved homeotic transcription factors from homeobox (HOX) genes. Their dysregulation disrupts regulatory molecule action, causing tumors. The HOXD10 gene is upregulated in breast, gastric, hepatocellular, colorectal, bladder, cholangiocellular carcinoma and prostate cancer. Tumor signaling pathways are affected by HOXD10 gene expression changes. This study examines HOXD10-associated signaling pathway dysregulation, which may alter metastatic cancer signaling. In addition, the theoretical foundations that alter HOXD10-mediated therapeutic resistance in malignancies has been presented. New cancer therapy methods will be simpler to develop with the newly discovered knowledge. This review showed that HOXD10 may be a tumor suppressor gene and a new cancer treatment target signaling pathway.
Collapse
Affiliation(s)
- Hemapreethi Surendran
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to be University, Chennai 600095 Tamil Nadu, India
| | - Thirunavukkarasu Palaniyandi
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to be University, Chennai 600095 Tamil Nadu, India; Department of Anatomy, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College and Hospital, SIMATS, Saveetha University, Chennai, Tamilnadu, India.
| | - Sudhakar Natarajan
- Department of Virology and Biotechnology, ICMR - National institute for Research in Tuberculosis (NIRT), Chetpet, Chennai 600031 Tamil Nadu, India
| | - Rajeswary Hari
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to be University, Chennai 600095 Tamil Nadu, India
| | - Sandhiya Viwanathan
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to be University, Chennai 600095 Tamil Nadu, India
| | - Gomathy Baskar
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to be University, Chennai 600095 Tamil Nadu, India
| | - Mugip Rahaman Abdul Wahab
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to be University, Chennai 600095 Tamil Nadu, India
| | - Maddaly Ravi
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116 Tamil Nadu, India
| | | |
Collapse
|
18
|
Bhattacharyya S, Ghosh H, Covarrubias-Zambrano O, Jain K, Swamy KV, Kasi A, Hamza A, Anant S, VanSaun M, Weir SJ, Bossmann SH, Padhye SB, Dandawate P. Anticancer Activity of Novel Difluorinated Curcumin Analog and Its Inclusion Complex with 2-Hydroxypropyl-β-Cyclodextrin against Pancreatic Cancer. Int J Mol Sci 2023; 24:ijms24076336. [PMID: 37047307 PMCID: PMC10093935 DOI: 10.3390/ijms24076336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/14/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the primary reason for cancer-related deaths in the US. Genetic mutations, drug resistance, the involvement of multiple signaling pathways, cancer stem cells (CSCs), and desmoplastic stroma, which hinders drug penetrance, contribute to poor chemotherapeutic efficacy. Hence, there is a need to identify novel drugs with improved delivery to improve treatment outcomes. Curcumin is one such compound that can inhibit multiple signaling pathways and CSCs. However, curcumin’s clinical applicability for treating PDAC is limited because of its poor solubility in water and metabolic instability. Hence, we developed a difluorinated curcumin (CDF) analog that accumulates selectively in the pancreas and inhibits PDAC growth in vitro and in vivo. In the present work, we developed its 2-hydroxy-propyl-β-cyclodextrin (HCD) inclusion complex to increase its water solubility and hydrolytic stability. The CDFHCD inclusion complex was characterized by spectroscopic, thermal, and microscopic techniques. The inclusion complex exhibited increased aqueous solubility, hydrolytic stability, and antiproliferative activity compared to parent CDF. Moreover, CDF and CDFHCD inhibited colony and spheroid formation, and induced cell cycle and apoptosis in PDAC cell lines. Hence, CDFHCD self-assembly is an efficient approach to increase water solubility and anticancer therapeutic efficacy, which now warrants advancement towards a clinical proof of concept in PDAC patients.
Collapse
Affiliation(s)
- Sangita Bhattacharyya
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Hindole Ghosh
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | | | - Krishan Jain
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - K. Venkateswara Swamy
- MIT School of Bioengineering, Sciences & Research, MIT Art, Design and Technology University, Pune 412201, India
| | - Anup Kasi
- Division of Medical Oncology, University of Kansas, Kansas City, KS 66160, USA
| | - Ameer Hamza
- Pathology and Laboratory Medicine, University of Kansas, Kansas City, KS 66160, USA
| | - Shrikant Anant
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Michael VanSaun
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Scott J. Weir
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
- Division of Medical Oncology, University of Kansas, Kansas City, KS 66160, USA
- Institute for Advancing Medical Innovation, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Stefan H. Bossmann
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Subhash B. Padhye
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
- Interdisciplinary Science & Technology Research Academy (ISTRA), Azam Campus, University of Pune, Pune 411001, India
| | - Prasad Dandawate
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
- Correspondence: ; Tel.: +1-913-945-6336
| |
Collapse
|
19
|
Lu M, Liu B, Li D, Gao Z, Li W, Zhou X, Zhan H. PXDNL activates the motility of urothelial bladder carcinoma cells through the Wnt/β-catenin pathway and has a prognostic value. Life Sci 2023; 312:121270. [PMID: 36493879 DOI: 10.1016/j.lfs.2022.121270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/25/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022]
Abstract
AIMS Although aberrant expression of peroxidasin-like (PXDNL) has been associated with carcinogenesis, its potential role in the Urothelial Carcinoma of the Bladder (UCB) remains unknown. The present study aimed to explore the role of PXDNL in UCB carcinogenesis and its potential clinical value. MAIN METHODS Based on The Cancer Genome Atlas (TCGA) data, bioinformatics was used to explore the potential clinical value of PXDNL. Wound healing and Transwell invasion assays were employed for the purpose of assessing the cell motility, while the Western Blotting experiments were utilized for investigating the protein expression pattern of PXDNL in UCB and investigating the Epithelial-to-Mesenchymal Transition (EMT) and Wnt/β-catenin pathways for understanding the probable mechanisms involved. KEY FINDS PXDNL mRNA was overexpressed in UCB tissues and indicated a poor prognosis. High PXDNL mRNA levels were also associated with advanced clinicopathological features and were regarded as independent prognostic factors for UCB. However, PXDNL showed a weak correlation with immune cell infiltration in UCB. In addition, the findings of the study verified that the existing form of the PXDNL protein was 57-kDa and it was upregulated in the UCB cell lines and tissue samples. Furthermore, silencing PXDNL inhibited, while overexpressing PXDNL promoted EMT and motility of UCB cells in vitro. Mechanistic studies showed that PXDNL activated UCB cell motility via the Wnt/β-catenin pathway. SIGNIFICANCE The results reveal a novel molecular target that could be further explored for developing preventive, predictive, and individualized treatment strategies for UCB.
Collapse
Affiliation(s)
- Miaolong Lu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University Lingnan Hospital, Guangzhou 510700, Guangdong, China.
| | - Bolong Liu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University Lingnan Hospital, Guangzhou 510700, Guangdong, China.
| | - Dongyang Li
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University Lingnan Hospital, Guangzhou 510700, Guangdong, China.
| | - Zhentao Gao
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University Lingnan Hospital, Guangzhou 510700, Guangdong, China.
| | - Wenbiao Li
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University Lingnan Hospital, Guangzhou 510700, Guangdong, China.
| | - Xiangfu Zhou
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University Lingnan Hospital, Guangzhou 510700, Guangdong, China.
| | - Hailun Zhan
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University Lingnan Hospital, Guangzhou 510700, Guangdong, China.
| |
Collapse
|
20
|
Cheng Z, Yu R, Li L, Mu J, Gong Y, Wu F, Liu Y, Zhou X, Zeng X, Wu Y, Sun R, Xiang T. Disruption of ZNF334 promotes triple-negative breast carcinoma malignancy through the SFRP1/ Wnt/β-catenin signaling axis. Cell Mol Life Sci 2022; 79:280. [PMID: 35507080 PMCID: PMC11072843 DOI: 10.1007/s00018-022-04295-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/20/2022] [Accepted: 04/06/2022] [Indexed: 11/03/2022]
Abstract
Zinc-finger proteins (ZNFs) constitute the largest transcription factor family in the human genome. The family functions in many important biological processes involved in tumorigenesis. In our research, we identified ZNF334 as a novel tumor suppressor of triple-negative breast cancer (TNBC). ZNF334 expression was usually reduced in breast cancerv (BrCa) tissues and TNBC cell lines MDA-MB-231 (MB231) and YCCB1. We observed that promoter hypermethylation of ZNF334 was common in BrCa cell lines and tissues, which was likely responsible for its reduced expression. Ectopic expression of ZNF334 in TNBC cell lines MB231 and YCCB1 could suppress their growth and metastatic capacity both in vitro and in vivo, and as well induce cell cycle arrest at S phase and cell apoptosis. Moreover, re-expression of ZNF334 in TNBC cell lines could rescue Epithelial-Mesenchymal Transition (EMT) process and restrain stemness, due to up-regulation of SFRP1, which is an antagonist of Wnt/β-catenin signaling. In conclusion, we verified that ZNF334 had a suppressive function of TNBC cell lines by targeting the SFRP1/Wnt/β-catenin signaling axis, which might have the potentials to become a new biomarker for diagnosis and treatment of TNBC patients.
Collapse
Affiliation(s)
- Zhaobo Cheng
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Renjie Yu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junhao Mu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yijia Gong
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fan Wu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yujia Liu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangyi Zhou
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaohua Zeng
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yongzhong Wu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Ran Sun
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Tingxiu Xiang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| |
Collapse
|
21
|
Chiu YJ, Yang JS, Tsai FJ, Chiu HY, Juan YN, Lo YH, Chiang JH. Curcumin suppresses cell proliferation and triggers apoptosis in vemurafenib-resistant melanoma cells by downregulating the EGFR signaling pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:868-879. [PMID: 34994998 DOI: 10.1002/tox.23450] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/24/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
Melanoma is a malignant tumor with aggressive behavior. Vemurafenib, a BRAF inhibitor, is clinically used in melanoma, but resistance to melanoma cytotoxic therapies is associated with BRAF mutations. Curcumin can effectively inhibit numerous types of cancers. However, there are no reports regarding the correlation between curcumin and vemurafenib-resistant melanoma cells. In this study, vemurafenib-resistant A375.S2 (A375.S2/VR) cells were established, and the functional mechanism of the epidermal growth factor receptor (EGFR), serine-threonine kinase (AKT), and the extracellular signal-regulated kinase (ERK) signaling induced by curcumin was investigated in A375.S2/VR cells in vitro. Our results indicated that A375.S2/VR cells had a higher IC50 concentration of vemurafenib than the parental A375.S2 cells. Moreover, curcumin reduced the viability and confluence of A375.S2/VR cells. Curcumin triggered apoptosis via reactive oxygen species (ROS) production, disruption of mitochondrial membrane potential (ΔΨm), and intrinsic signaling (caspase-9/-3-dependent) pathways in A375.S2/VR cells. Curcumin-induced apoptosis was also mediated by the EGFR signaling pathway. Combination treatment with curcumin and gefitinib (an EGFR inhibitor) synergistically potentiated the inhibitory effect of cell viability in A375.S2/VR cells. The present study provides new insights into the therapy of vemurafenib-resistant melanoma and suggests that curcumin might be an encouraging therapeutic candidate for its drug-resistant treatment.
Collapse
Affiliation(s)
- Yu-Jen Chiu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- Human Genetic Center, China Medical University Hospital, Taichung, Taiwan
- Department of Medical Genetics, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Hong-Yi Chiu
- Department of Pharmacy, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Graduate Institute of Clinical Pharmacy, College of Medicine, Tzu Chi University, Hualien, Taiwan
- Holistic Education Center, Tzu Chi University of Science and Technology, Hualien, Taiwan
| | - Yu-Ning Juan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yu-Hsiang Lo
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Jo-Hua Chiang
- Department of Nursing, Chung-Jen Junior College of Nursing, Health Sciences and Management, Chiayi, Taiwan
| |
Collapse
|
22
|
Bryś M, Urbańska K, Olas B. Novel Findings regarding the Bioactivity of the Natural Blue Pigment Genipin in Human Diseases. Int J Mol Sci 2022; 23:902. [PMID: 35055094 PMCID: PMC8776187 DOI: 10.3390/ijms23020902] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/08/2022] [Accepted: 01/12/2022] [Indexed: 12/16/2022] Open
Abstract
Genipin is an important monoterpene iridoid compound isolated from Gardenia jasminoides J.Ellis fruits and from Genipa americana fruits, or genipap. It is a precursor of a blue pigment which may be attractive alternative to existing food dyes and it possesses various potential therapeutic properties such as anti-cancer, anti-diabetic and hepatoprotective activity. Biomedical studies also show that genipin may act as a neuroprotective drug. This review describes new aspects of the bioactivity of genipin against various diseases, as well as its toxicity and industrial applications, and presents its potential mechanism of action.
Collapse
Affiliation(s)
- Magdalena Bryś
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/3, 90-236 Lodz, Poland;
| | - Karina Urbańska
- Faculty of Medicine, Medical University of Lodz, 90-419 Lodz, Poland;
| | - Beata Olas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/3, 90-236 Lodz, Poland
| |
Collapse
|
23
|
Ju PC, Ho YC, Chen PN, Lee HL, Lai SY, Yang SF, Yeh CB. Kaempferol inhibits the cell migration of human hepatocellular carcinoma cells by suppressing MMP-9 and Akt signaling. ENVIRONMENTAL TOXICOLOGY 2021; 36:1981-1989. [PMID: 34156145 DOI: 10.1002/tox.23316] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Accepted: 06/15/2021] [Indexed: 06/13/2023]
Abstract
Metastasis is the most prevalent cause of cancer-related deaths and treatment failure in patients with hepatocellular carcinoma (HCC). Kaempferol is a natural flavonol belonging to the subgroup of flavonoids and exhibits potent anticancer activities. This study provides molecular evidence on the anti-invasive and anti-migratory effects of kaempferol on human HCC cells. The anti-invasive effect was investigated by applying kaempferol on two human HCC cell lines (Huh-7 and SK-Hep-1). Kaempferol reduced the invasion and migration of Huh-7 and SK-Hep-1 cells by Boyden chamber invasion assay and wound healing assay, respectively. A protease array analysis showed that Matrix Metalloproteinase-9 (MMP-9) was dramatically downregulated in HCC cells after kaempferol treatment. Gelatin zymography and Western blot assay showed that kaempferol reduced the activities and protein expression of MMP-9, respectively. Kaempferol also sufficiently suppressed the phosphorylation of the Akt expression. Overall, kaempferol inhibited the invasive properties of human HCC cells by targeting MMP-9 and Akt pathways. Hence, kaempferol could be used as an adjuvant therapeutic agent for the treatment of human HCC cells.
Collapse
Affiliation(s)
- Po-Chung Ju
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yung-Chuan Ho
- School of Medical Applied Chemistry, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Ni Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hsiang-Lin Lee
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Szu-Yu Lai
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chao-Bin Yeh
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Emergency Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
24
|
Hussain Y, Islam L, Khan H, Filosa R, Aschner M, Javed S. Curcumin-cisplatin chemotherapy: A novel strategy in promoting chemotherapy efficacy and reducing side effects. Phytother Res 2021; 35:6514-6529. [PMID: 34347326 DOI: 10.1002/ptr.7225] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/08/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022]
Abstract
The efficacy of chemotherapy in cancer therapy is limited due to resistance, treatment selectivity, and severe adverse effects. Immunotherapy, chemotherapy, targeted therapy, radiation, and surgery are the most common therapeutic strategies for treatment, with chemotherapy being the most successful. Nonetheless, these treatments exhibit poor effectiveness due to toxicity and resistance. Therefore, combination therapies of natural products may be used as an effective and novel strategy to overcome such barriers. Cisplatin is a platinum-based chemotherapy agent, and when administered alone, it can lead to severe adverse effects and resistance mechanism resulting in therapeutic failure. Curcumin is a polyphenolic compound extracted from turmeric (Curcuma longa) exhibiting anticancer potential with minimal adverse effects. The combination therapy of curcumin and cisplatin is a novel strategy to mitigate/attenuate cisplatin-related adverse effects and improve the barrier of resistance reducing unwanted effects. However, there are uncertainties on the efficacy of curcumin, and more in depth and high-quality studies are needed. This review aims to explain the adverse effects related to individual cisplatin delivery, the positive outcome of individual curcumin delivery, and the combination therapy of curcumin and cisplatin from nano platform as a novel strategy for cancer therapy.
Collapse
Affiliation(s)
- Yaseen Hussain
- Lab of Controlled Release and Drug Delivery System, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Lubna Islam
- Department of Pharmacy, University of Malakand, Dir Lower Chakdara, KPK, Pakistan
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Rosanna Filosa
- Department of Experimental Medicine, University of Campania, "L. Vanvitelli", Naples, Italy
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Saba Javed
- Department of Zoology, Hazara University, Mansehra, Pakistan
| |
Collapse
|
25
|
Fosciclopirox suppresses growth of high-grade urothelial cancer by targeting the γ-secretase complex. Cell Death Dis 2021; 12:562. [PMID: 34059639 PMCID: PMC8166826 DOI: 10.1038/s41419-021-03836-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 12/21/2022]
Abstract
Ciclopirox (CPX) is an FDA-approved topical antifungal agent that has demonstrated preclinical anticancer activity in a number of solid and hematologic malignancies. Its clinical utility as an oral anticancer agent, however, is limited by poor oral bioavailability and gastrointestinal toxicity. Fosciclopirox, the phosphoryloxymethyl ester of CPX (Ciclopirox Prodrug, CPX-POM), selectively delivers the active metabolite, CPX, to the entire urinary tract following parenteral administration. We characterized the activity of CPX-POM and its major metabolites in in vitro and in vivo preclinical models of high-grade urothelial cancer. CPX inhibited cell proliferation, clonogenicity and spheroid formation, and increased cell cycle arrest at S and G0/G1 phases. Mechanistically, CPX suppressed activation of Notch signaling. Molecular modeling and cellular thermal shift assays demonstrated CPX binding to γ-secretase complex proteins Presenilin 1 and Nicastrin, which are essential for Notch activation. To establish in vivo preclinical proof of principle, we tested fosciclopirox in the validated N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN) mouse bladder cancer model. Once-daily intraperitoneal administration of CPX-POM for four weeks at doses of 235 mg/kg and 470 mg/kg significantly decreased bladder weight, a surrogate for tumor volume, and resulted in a migration to lower stage tumors in CPX-POM treated animals. This was coupled with a reduction in the proliferation index. Additionally, there was a reduction in Presenilin 1 and Hes-1 expression in the bladder tissues of CPX-POM treated animals. Following the completion of the first-in-human Phase 1 trial (NCT03348514), the pharmacologic activity of fosciclopirox is currently being characterized in a Phase 1 expansion cohort study of muscle-invasive bladder cancer patients scheduled for cystectomy (NCT04608045) as well as a Phase 2 trial of newly diagnosed and recurrent urothelial cancer patients scheduled for transurethral resection of bladder tumors (NCT04525131).
Collapse
|
26
|
Tian H, Chen X, Zhang Y, Wang Y, Fu X, Gu W, Wen Y. Dioscin inhibits SCC15 cell proliferation via the RASSF1A/MST2/YAP axis. Mol Med Rep 2021; 23:414. [PMID: 33786612 PMCID: PMC8025490 DOI: 10.3892/mmr.2021.12053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/25/2021] [Indexed: 12/24/2022] Open
Abstract
Dioscin, an extract from traditional Chinese herbal plants, displays various biological and pharmacological effects on tumors, including inhibition of cell proliferation and induction of DNA damage. However, the effects of dioscin on oral squamous cell carcinoma (OSCC) cells are not completely understood. The present study aimed to evaluate the impact of dioscin on OSCC cell proliferation. Cell Counting Kit-8 and 5-ethynyl-2′-deoxyuridine incorporation assays were performed to assess cell proliferation. Flow cytometry was conducted to detect alterations in the cell cycle and cell apoptosis. Western blotting and coimmunoprecipitation were performed to determine protein expression levels. In SCC15 cells, dioscin treatment significantly induced cell cycle arrest, increased apoptosis and inhibited proliferation compared with the control group. Mechanistically, the tumor suppressor protein Ras association domain-containing protein 1A (RASSF1A) was activated and oncoprotein yes-associated protein (YAP) was phosphorylated by dioscin. Furthermore, YAP overexpression and knockdown reduced and enhanced the inhibitory effects of dioscin on SCC15 cells, respectively. In summary, the results demonstrated that, compared with the control group, dioscin upregulated RASSF1A expression in OSCC cells, which resulted in YAP phosphorylation, thus weakening its transcriptional coactivation function, enhancing cell cycle arrest and apoptosis, and inhibiting cell proliferation. The present study indicated that dioscin may serve as a therapeutic agent for OSCC.
Collapse
Affiliation(s)
- Hui Tian
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiyan Chen
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yafei Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Ying Wang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xucheng Fu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Weiting Gu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yong Wen
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|