1
|
Jin L, Jiang X, Li Y, Xiang S, Pei R, Lu Y, Jiang L. Flavokawain a induces cell cycle arrest through CDT1-dependent p27 regulation and synergizes with venetoclax in acute myeloid leukemia. Toxicol Appl Pharmacol 2025; 498:117305. [PMID: 40113097 DOI: 10.1016/j.taap.2025.117305] [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: 12/02/2024] [Revised: 03/10/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
The poor prognosis of patients with acute myeloid leukemia (AML) is largely ascribed to the deficiency of persistently effective therapies. Despite the recent approval of targeted drugs such as the BCL-2 inhibitor venetoclax, the clinical benefit is limited due to the development of resistance. The use of natural products is emerging as a feasible strategy to treat malignant diseases including AML. Flavokawain A (FKA) is a naturally occurring chalcone isolated from the root of kava and possesses extensive antitumor activities. The therapeutic potential of FKA in AML remains unknown. In the present study, we found that treatment with FKA reduced the viability in four AML cell lines in dose- and time-dependent manners. The anti-AML activity of venetoclax was significantly potentiated by FKA. Mechanistically, FKA induced G1 phase arrest in AML cells along with CDT1 downregulation and p27 upregulation. Knockdown of CDT1 increased the expression of p27, leading to the inhibition on cell viability. Both p27 upregulation and viability inhibition caused by FKA was partially rescued by CDT1 overexpression. The therapeutic effect of FKA alone or in combination with venetoclax was verified in primary blasts from AML patients, further strengthening the clinical relevance of the current study. Therefore, our data suggest that FKA can be considered as a potential therapeutic agent in the treatment of AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Sulfonamides/pharmacology
- Sulfonamides/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Drug Synergism
- Cell Line, Tumor
- Cyclin-Dependent Kinase Inhibitor p27/metabolism
- Cyclin-Dependent Kinase Inhibitor p27/genetics
- Cell Cycle Checkpoints/drug effects
- Cell Survival/drug effects
- Antineoplastic Agents/pharmacology
- Dose-Response Relationship, Drug
- Flavonoids/pharmacology
Collapse
Affiliation(s)
- Lili Jin
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Department of Pathology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China
| | - Xia Jiang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Department of Pathology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China
| | - Youhong Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Department of Pathology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China
| | - Sumeng Xiang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Department of Pathology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China
| | - Renzhi Pei
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China
| | - Ying Lu
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China.
| | - Lei Jiang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Department of Pathology, Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China.
| |
Collapse
|
2
|
Gayen S, Roy S, Laishram D, Bandyopadhyay SN, Mukherjee S. Flavokawain A Ruthenium-p-Cymene Complex-Induced Apoptosis by the Modulation of PI3K/β-Catenin/HER2/PARP Signalling in Lung Cancer. Clin Exp Pharmacol Physiol 2025; 52:e70030. [PMID: 39971480 DOI: 10.1111/1440-1681.70030] [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: 08/24/2024] [Revised: 12/12/2024] [Accepted: 01/13/2025] [Indexed: 02/21/2025]
Abstract
Lung cancer is most terrible cause of cancer-related death throughout the world. This study focused on the synthesis and characterisation of novel flavokawain A ruthenium-p-cymene complex and to investigate the chemotherapeutic activity against lung carcinoma via in silico, in vitro and in vivo approaches. The complex was characterised via several spectroscopic techniques. In vitro study including cell viability, transwell migration, Western blot and flow cytometric analysis have been executed on both A549 and NCI-H460 cells. The toxicological assessment was performed and subsequently anticancer activity of complex was evaluated in benzo[α]pyrene persuaded lung carcinoma in mice. The molecular docking study demonstrated the compound has greater binding ability with β-catenin, Akt, HER2 and PARP. Followed by the complex treatment, the downregulation of β-catenin, PI3K, Akt, HER2 and PARP were investigated by Western blot analysis and cell cycle arrest was determined through flow cytometry. The outcomes of in vivo experimentation represented fruitful restoration of typical lung architecture after complex treatment. Immunohistochemical analysis demonstrated the downstream of β-catenin/m-TOR/Akt and upstream of caspase-3 and p53 expression, thereby initiating apoptosis. The complex exhibited a potent chemotherapeutic activity via the alteration of tumour microenvironment by modulating PI3K/Akt/β-catenin/HER2/PARP transduction in correlates with apoptotic events in lung carcinoma.
Collapse
Affiliation(s)
- Sakuntala Gayen
- Department of Pharmacy, NSHM Knowledge Campus-Kolkata, Kolkata, West Bengal, India
| | - Souvik Roy
- Department of Pharmacy, NSHM Knowledge Campus-Kolkata, Kolkata, West Bengal, India
| | - Diana Laishram
- Department of Anatomy, Jagannath Gupta Institute of Medical Sciences and Hospital (JIMSH), Kolkata, India
| | | | | |
Collapse
|
3
|
Akla N, Veilleux C, Annabi B. The Chemopreventive Impact of Diet-Derived Phytochemicals on the Adipose Tissue and Breast Tumor Microenvironment Secretome. Nutr Cancer 2024; 77:9-25. [PMID: 39300732 DOI: 10.1080/01635581.2024.2401647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024]
Abstract
Cancer cells-derived extracellular vesicles can trigger the transformation of adipose-derived mesenchymal stem cells (ADMSC) into a pro-inflammatory, cancer-associated adipocyte (CAA) phenotype. Such secretome-mediated crosstalk between the adipose tissue and the tumor microenvironment (TME) therefore impacts tumor progression and metastatic processes. In addition, emerging roles of diet-derived phytochemicals, especially epigallocatechin-3-gallate (EGCG) among other polyphenols, in modulating exosome-mediated metabolic and inflammatory signaling pathways have been highlighted. Here, we discuss how selected diet-derived phytochemicals could alter the secretome signature as well as the crosstalk dynamics between the adipose tissue and the TME, with a focus on breast cancer. Their broader implication in the chemoprevention of obesity-related cancers is also discussed.
Collapse
Affiliation(s)
- Naoufal Akla
- Laboratoire d'Oncologie Moléculaire, Département de Chimie and CERMO-FC, Université du Québec à Montréal, Montreal, Canada
| | - Carolane Veilleux
- Laboratoire d'Oncologie Moléculaire, Département de Chimie and CERMO-FC, Université du Québec à Montréal, Montreal, Canada
| | - Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Département de Chimie and CERMO-FC, Université du Québec à Montréal, Montreal, Canada
| |
Collapse
|
4
|
William JNG, Dhar R, Gundamaraju R, Sahoo OS, Pethusamy K, Raj AFPAM, Ramasamy S, Alqahtani MS, Abbas M, Karmakar S. SKping cell cycle regulation: role of ubiquitin ligase SKP2 in hematological malignancies. Front Oncol 2024; 14:1288501. [PMID: 38559562 PMCID: PMC10978726 DOI: 10.3389/fonc.2024.1288501] [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: 09/04/2023] [Accepted: 02/15/2024] [Indexed: 04/04/2024] Open
Abstract
SKP2 (S-phase kinase-associated protein 2) is a member of the F-box family of substrate-recognition subunits in the SCF ubiquitin-protein ligase complexes. It is associated with ubiquitin-mediated degradation in the mammalian cell cycle components and other target proteins involved in cell cycle progression, signal transduction, and transcription. Being an oncogene in solid tumors and hematological malignancies, it is frequently associated with drug resistance and poor disease outcomes. In the current review, we discussed the novel role of SKP2 in different hematological malignancies. Further, we performed a limited in-silico analysis to establish the involvement of SKP2 in a few publicly available cancer datasets. Interestingly, our study identified Skp2 expression to be altered in a cancer-specific manner. While it was found to be overexpressed in several cancer types, few cancer showed a down-regulation in SKP2. Our review provides evidence for developing novel SKP2 inhibitors in hematological malignancies. We also investigated the effect of SKP2 status on survival and disease progression. In addition, the role of miRNA and its associated families in regulating Skp2 expression was explored. Subsequently, we predicted common miRNAs against Skp2 genes by using miRNA-predication tools. Finally, we discussed current approaches and future prospective approaches to target the Skp2 gene by using different drugs and miRNA-based therapeutics applications in translational research.
Collapse
Affiliation(s)
- Jonahunnatha Nesson George William
- Department of Medical, Oral and Biotechnological Sciences (DSMOB), Ageing Research Center and Translational Medicine-CeSI-MeT, “G. d’Annunzio” University Chieti-Pescara, Chieti, Italy
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Rohit Gundamaraju
- ER Stress and Intestinal Mucosal Biology Lab, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Om Saswat Sahoo
- Department of Biotechnology, National Institute of Technology, Durgapur, India
| | - Karthikeyan Pethusamy
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | - Subbiah Ramasamy
- Cardiac Metabolic Disease Laboratory, Department Of Biochemistry, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Mohammed S. Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, University of Leicester, Leicester, United Kingdom
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, Saudi Arabia
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
5
|
Li J, Wang R, Liu Y, Wu Y, Han L, Zheng L, Bao Z. FKA-A NPs enhances PTX-A NPs efficacy to suppress ovarian cancer via regulating Skp2/YAP pathway. Fundam Clin Pharmacol 2023; 37:125-136. [PMID: 36028983 DOI: 10.1111/fcp.12828] [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: 05/20/2022] [Revised: 07/26/2022] [Accepted: 08/26/2022] [Indexed: 01/25/2023]
Abstract
Recurrence and distant metastasis after paclitaxel (PTX)-based chemotherapy in ovarian cancer (OC) patients remains a clinical obstacle. Flavokawain A (FKA) is a novel chalcone from kava plant that can induce G2/M arrest and inhibit invasion and metastasis in different tumor cells. In this study, we examined the effects and the molecular mechanism of sodium aescinate (Aes)-stabilized nanoparticles FKA-A NPs in enhancing the efficacy of PTX-A NPs in vitro and in vivo. We showed that FKA-A NPs combined with PTX-A NPs notably inhibited the proliferation and migration and reduced the expression of EMT-related markers in OCs. YAP nuclear translocation and its downstream signaling pathway were remarkably activated after PTX-A NPs treatment in OCs. FKA-A NPs obviously inhibited YAP nuclear translocation and reduced the transcriptional activity of YAP target genes. Simultaneously, FKA-A NPs dose and time dependently inhibited Skp2 expression in A2780 and Skov3 cells. In contrast, overexpression of Skp2 significantly attenuated the inhibition of FKA-A NPs on YAP nuclear translocation. In OC homograft mice, treatment with FKA-A NPs and PTX-A NPs significantly suppressed the growth of homograft tumor compared with PTX-A NPs but did not decrease mice's body weight. In summary, we demonstrate that FKA-A NPs enhance the efficacy of PTX-A NPs against OCs in vitro and in vivo via reducing Skp2 expression, thus suppressing YAP nuclear translocation and activity of its target genes.
Collapse
Affiliation(s)
- Juan Li
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of medicine, Shandong University, Jinan, Shandong, China
| | - Rongmei Wang
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of medicine, Shandong University, Jinan, Shandong, China
| | - Yongqing Liu
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of medicine, Shandong University, Jinan, Shandong, China
| | - Yuqian Wu
- Cancer Center, The Second Hospital, Cheeloo College of medicine, Shandong University, Jinan, Shandong, China
| | - Leiqiang Han
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of medicine, Shandong University, Jinan, Shandong, China
| | - Lei Zheng
- Department of Pharmacy, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhengqiang Bao
- Cancer Center, The Second Hospital, Cheeloo College of medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
6
|
Jing J, Rui L, Junyuan S, Jinfeng Y, Zhihao H, Weiguo L, Zhenyu J. Small-molecule compounds inhibiting S-phase kinase-associated protein 2: A review. Front Pharmacol 2023; 14:1122008. [PMID: 37089937 PMCID: PMC10113621 DOI: 10.3389/fphar.2023.1122008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
S-phase kinase-associated protein 2 (Skp2) is a substrate-specific adaptor in Skp1-CUL1-ROC1-F-box E3 ubiquitin ligases and widely regarded as an oncogene. Therefore, Skp2 has remained as an active anticancer research topic since its discovery. Accordingly, the structure of Skp2 has been solved and numerous Skp2 inhibiting compounds have been identified. In this review, we would describe the structural features of Skp2, introduce the ubiquitination function of SCFSkp2, and summarize the diverse natural and synthetic Skp2 inhibiting compounds reported to date. The IC50 data of the Skp2 inhibitors or inhibiting compounds in various kinds of tumors at cellular levels implied that the cancer type, stage and pathological mechanisms should be taken into consideration when selecting Skp2-inhibiting compound for cancer treatment.
Collapse
Affiliation(s)
- Jia Jing
- Schools of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Li Rui
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Sun Junyuan
- Schools of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Yang Jinfeng
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Hong Zhihao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Lu Weiguo
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Women′s Reproductive Health Research of Zhejiang Province, Hangzhou, Zhejiang Province, China
- *Correspondence: Lu Weiguo, ; Jia Zhenyu,
| | - Jia Zhenyu
- Institute of Occupation Diseases, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
- *Correspondence: Lu Weiguo, ; Jia Zhenyu,
| |
Collapse
|
7
|
An Updated Review on the Psychoactive, Toxic and Anticancer Properties of Kava. J Clin Med 2022; 11:jcm11144039. [PMID: 35887801 PMCID: PMC9315573 DOI: 10.3390/jcm11144039] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 01/27/2023] Open
Abstract
Kava (Piper methysticum) has been widely consumed for many years in the South Pacific Islands and displays psychoactive properties, especially soothing and calming effects. This plant has been used in Western countries as a natural anxiolytic in recent decades. Kava has also been used to treat symptoms associated with depression, menopause, insomnia, and convulsions, among others. Along with its putative beneficial health effects, kava has been associated with liver injury and other toxic effects, including skin toxicity in heavy consumers, possibly related to its metabolic profile or interference in the metabolism of other xenobiotics. Kava extracts and kavalactones generally displayed negative results in genetic toxicology assays although there is sufficient evidence for carcinogenicity in experimental animals, most likely through a non-genotoxic mode of action. Nevertheless, the chemotherapeutic/chemopreventive potential of kava against cancer has also been suggested. Both in vitro and in vivo studies have evaluated the effects of flavokavains, kavalactones and/or kava extracts in different cancer models, showing the induction of apoptosis, cell cycle arrest and other antiproliferative effects in several types of cancer, including breast, prostate, bladder, and lung. Overall, in this scoping review, several aspects of kava efficacy and safety are discussed and some pertinent issues related to kava consumption are identified.
Collapse
|
8
|
Liu Z, Song L, Xie J, Simoneau AR, Uchio E, Zi X. Chemoprevention of Urothelial Cell Carcinoma Tumorigenesis by Dietary Flavokawain A in UPII-Mutant Ha-ras Transgenic Mice. Pharmaceutics 2022; 14:496. [PMID: 35335874 PMCID: PMC8955676 DOI: 10.3390/pharmaceutics14030496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 02/05/2023] Open
Abstract
Non-muscle-invasive bladder cancer (NMIBC) has one of the highest recurrence rates among all solid cancers and the highest lifetime treatment cost per patient. Therefore, the development of chemoprevention strategies for reducing the occurrence and recurrence of NMIBC as well as its burdens on the healthcare system is valuable. Our aim was to determine whether flavokawain A (FKA), a kava chalcone isolated from the kava plant, can target the in vivo activated Ha-ras pathway for prevention and treatment of NMIBC. UPII-mutant Ha-ras transgenic mice that develop papillary urothelial cell carcinoma were fed orally with vehicle control or FKA-formulated food for 6 months starting at 6 weeks of age. Seventy-nine percent (15/19) of male mice fed with 6 g FKA per kilogram (kg) of food survived beyond the 6 months of treatment, while 31.6% (6/19) of control food-fed male mice survived the 6-month treatment period (p = 0.02). The mean bladder weights in FKA vs. control food-fed mice were 0.216 ± 0.033 vs. 0.342 ± 0.039 g in male mice (p = 0.0413) and 0.043 ± 0.004 vs. 0.073 ± 0.004 g in female mice (p < 0.0001); FKA reduced bladder weight by 37% and 41%, respectively. The tumor burdens, determined by the wet bladder weight, in these mice were inversely related to plasma FKA concentrations. In addition to decreased bladder weight, FKA treatment significantly reduced the incidences of hydronephrosis and hematuria. FKA-treated mice exhibited more well-differentiated tumors in the bladder and ureter. Immunohistochemical analysis of FKA-treated tumors compared to those in the control group revealed fewer Ki-67- and survivin-positive cells and an increased number of p27- and TUNEL-positive cells, indicating that FKA inhibits proliferation and induces apoptosis. Overall, the results suggest that FKA can target the in vivo activated Ha-ras pathway for the prevention and treatment of NMIBC.
Collapse
Affiliation(s)
- Zhongbo Liu
- Department of Urology, University of California, Irvine, CA 92868, USA; (Z.L.); (L.S.); (J.X.); (A.R.S.); (E.U.)
| | - Liankun Song
- Department of Urology, University of California, Irvine, CA 92868, USA; (Z.L.); (L.S.); (J.X.); (A.R.S.); (E.U.)
| | - Jun Xie
- Department of Urology, University of California, Irvine, CA 92868, USA; (Z.L.); (L.S.); (J.X.); (A.R.S.); (E.U.)
| | - Anne R. Simoneau
- Department of Urology, University of California, Irvine, CA 92868, USA; (Z.L.); (L.S.); (J.X.); (A.R.S.); (E.U.)
| | - Edward Uchio
- Department of Urology, University of California, Irvine, CA 92868, USA; (Z.L.); (L.S.); (J.X.); (A.R.S.); (E.U.)
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92868, USA
| | - Xiaolin Zi
- Department of Urology, University of California, Irvine, CA 92868, USA; (Z.L.); (L.S.); (J.X.); (A.R.S.); (E.U.)
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92868, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92617, USA
| |
Collapse
|
9
|
Sun M, Wang Y, Yuan M, Zhao Q, Zhang Y, Yao Y, Duan Y. Angiogenesis, Anti-Tumor, and Anti-Metastatic Activity of Novel α-Substituted Hetero-Aromatic Chalcone Hybrids as Inhibitors of Microtubule Polymerization. Front Chem 2021; 9:766201. [PMID: 34900935 PMCID: PMC8652888 DOI: 10.3389/fchem.2021.766201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/05/2021] [Indexed: 11/13/2022] Open
Abstract
A library of new heteroaromatic ring-linked chalcone analogs were designed and synthesized of these, compound 7m with α-CH3 substitution and bearing a benzofuran ring, displaying the most potent activity, with IC50 values of 0.07–0.183 µM against three cancer cells. Its low cytotoxicity toward normal human cells and strong potency on drug-resistant cells revealed the possibility for cancer therapy. It also could moderately inhibit in vitro tubulin polymerization with an IC50 value of 12.23 µM, and the disruption of cellular architecture in MCF-7 cells was observed by an immunofluorescence assay. Cellular-based mechanism studies elucidated that 7m arrested the cell cycle at the G2/M phase and induced apoptosis by regulating the expression levels of caspases and PARP protein. Importantly, the compound 7 m was found to inhibit HUVEC tube formation, migration, and invasion in vitro. In vivo assay showed that 7m could effectively destroy angiogenesis of zebrafish embryos. Furthermore, our data suggested that treatment with 7m significantly reduced MCF-7 cell metastasis and proliferation in vitro and in zebrafish xenograft. Collectively, this work showed that chalcone hybrid 7m deserves further investigation as dual potential tubulin polymerization and angiogenesis inhibitor.
Collapse
Affiliation(s)
- Moran Sun
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China.,School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Yuyang Wang
- School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Minghua Yuan
- School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Qing Zhao
- School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Yixin Zhang
- School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Yongfang Yao
- School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China.,School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
10
|
de Souza PS, Bibá GCC, Melo EDDN, Muzitano MF. Chalcones against the hallmarks of cancer: a mini-review. Nat Prod Res 2021; 36:4809-4826. [PMID: 34865580 DOI: 10.1080/14786419.2021.2000980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Chalcones (1,3-diphenylpropen-1-ones) are a class of flavonoids that have been shown a broad spectrum of biological activities with therapeutic potential. Naturally occurring chalcones or synthetic chalcone derivatives have been extensively investigated as anticancer compounds. Cancer is still among the leading causes of death globally, although cancer treatments have improved over the past decades. Most of chemotherapeutic drugs target proliferating tumor cells; however, the cancer cells capabilities are also associated to tumor surround microenvironment. Thereby, the search of new compounds with a broad antitumor activity is still a great challenge. The cytotoxicity mechanisms of chalcones are beyond apoptosis induction in tumor cells, which make them promising compound for cancer therapy. In this mini-review we summarized recent studies that describe the anticancer potential of chalcones related to some of hallmarks of cancer. We shed a light on sustaining proliferative signaling, tumor-promoting inflammation, activating invasion and metastasis, inducing angiogenesis and resisting cell death.
Collapse
Affiliation(s)
- Paloma Silva de Souza
- Laboratório de Produtos Bioativos, Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Geysa Cristina Caldas Bibá
- Laboratório de Produtos Bioativos, Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Evelynn Dalila do Nascimento Melo
- Laboratório de Produtos Bioativos, Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michelle Frazão Muzitano
- Laboratório de Produtos Bioativos, Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
He M, Yu M, Li C, Meng X, Su J, Zhu Y, Yao R. Design, Synthesis and Bioactivity Evaluation of Novel Chalcone Derivatives Possessing Tryptophan Moiety with Dual Activities of Anti-cancer and Partially Restoring the Proliferation of Normal Kidney Cells Pre-treated with Cisplatin. Anticancer Agents Med Chem 2021; 22:1945-1961. [PMID: 34674626 DOI: 10.2174/1871520621666211021134626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/09/2021] [Accepted: 09/01/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Chalcone is a broad-spectrum natural product with anti-cancer and anti-inflammatory activities. However, low potency, low selectivity, and serious side effects limit its druggability. L-Tryptophan is an essential precursor molecule of an anti-cancer active substance. Also, the indole moiety inhibits the proliferation of tumor cells by binding to colchicine sites. A decrease in kidney cell activity caused by kidney inflammation is the primary side effect of cancer therapy. OBJECTIVE The purpose of this work was to design, synthesize, and perform bioactivity evaluation of novel chalcone derivatives possessing tryptophan moiety with dual activities of anti-cancer and partially restoring the proliferation of normal kidney cells pre-treated with cisplatin. METHODS A series of novel chalcone derivatives possessing tryptophan moiety (5a-5g, 6a-6o) were designed, synthesized, and evaluated for anti-cancer activity against four cancer cell lines (gastric (HGC-27), colon (HCT-116), prostate (PC-3), and lung (A549)), and a human normal cell line (gastric mucosal epithelial (GES-1)). The activity of restoring the proliferation of normal kidney cells pre-treated with cisplatin was evaluated by MTT assay. Cell cycle, apoptosis, and apoptosis proteins (Bax and Bcl-2) were used to evaluate the anti-cancer mechanism of the most potent compound. Moreover, a docking study was performed to explain the high anti-cancer activity of 6n. The expressions of TNF-α, IL-6, and MCP-1 were detected by ELISA. RESULTS Most of the compounds exhibited high anti-cancer activity against the HGC-27 cell line and exhibited low toxicity against the normal cell line. Based on three rounds of a structure optimization, 6n was discovered as the most potent compound against HGC-27 cells with an IC50 value of 2.02 μM and an SI value of 28.47. Further studies demonstrated that 6n could induce cell cycle arrest at the G2/M phase and the apoptosis of the HGC-27 cell line by reducing the expression of Bcl-2 and improving the expression level of Bax. Molecular docking result displayed 6n bound to the colchicine site. At the same time, 6n also exhibited moderate activity of restoring the proliferation of normal kidney cells pre-treated with cisplatin by reducing the expression of inflammatory substances. CONCLUSION Our findings collectively suggested that 6n should be further studied as a potential anti-cancer agent that could partially restore the proliferation of normal kidney cells pre-treated with cisplatin in gastric cancer patients by an anti-inflammatory pathway.
Collapse
Affiliation(s)
- Meng He
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230000. China
| | - Mingjun Yu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230000. China
| | - Chao Li
- School of Pharmacy, Anhui Medical University, Hefei, 230032. China
| | - Xiaoming Meng
- School of Pharmacy, Anhui Medical University, Hefei, 230032. China
| | - Jiamin Su
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230000. China
| | - Yuting Zhu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230000. China
| | - Risheng Yao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230000. China
| |
Collapse
|
12
|
Michalkova R, Mirossay L, Gazdova M, Kello M, Mojzis J. Molecular Mechanisms of Antiproliferative Effects of Natural Chalcones. Cancers (Basel) 2021; 13:cancers13112730. [PMID: 34073042 PMCID: PMC8198114 DOI: 10.3390/cancers13112730] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Despite the important progress in cancer treatment in the past decades, the mortality rates in some types of cancer have not significantly decreased. Therefore, the search for novel anticancer drugs has become a topic of great interest. Chalcones, precursors of flavonoid synthesis in plants, have been documented as natural compounds with pleiotropic biological effects including antiproliferative/anticancer activity. This article focuses on the knowledge on molecular mechanisms of antiproliferative action of chalcones and draws attention to this group of natural compounds that may be of importance in the treatment of cancer disease. Abstract Although great progress has been made in the treatment of cancer, the search for new promising molecules with antitumor activity is still one of the greatest challenges in the fight against cancer due to the increasing number of new cases each year. Chalcones (1,3-diphenyl-2-propen-1-one), the precursors of flavonoid synthesis in higher plants, possess a wide spectrum of biological activities including antimicrobial, anti-inflammatory, antioxidant, and anticancer. A plethora of molecular mechanisms of action have been documented, including induction of apoptosis, autophagy, or other types of cell death, cell cycle changes, and modulation of several signaling pathways associated with cell survival or death. In addition, blockade of several steps of angiogenesis and proteasome inhibition has also been documented. This review summarizes the basic molecular mechanisms related to the antiproliferative effects of chalcones, focusing on research articles from the years January 2015–February 2021.
Collapse
|
13
|
Wu T, Gu X, Cui H. Emerging Roles of SKP2 in Cancer Drug Resistance. Cells 2021; 10:cells10051147. [PMID: 34068643 PMCID: PMC8150781 DOI: 10.3390/cells10051147] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
More than half of all cancer patients receive chemotherapy, however, some of them easily acquire drug resistance. Resistance to chemotherapy has become a massive obstacle to achieve high rates of pathological complete response during cancer therapy. S-phase kinase-associated protein 2 (Skp2), as an E3 ligase, was found to be highly correlated with drug resistance and poor prognosis. In this review, we summarize the mechanisms that Skp2 confers to drug resistance, including the Akt-Skp2 feedback loop, Skp2-p27 pathway, cell cycle and mitosis regulation, EMT (epithelial-mesenchymal transition) property, enhanced DNA damage response and repair, etc. We also addressed novel molecules that either inhibit Skp2 expression or target Skp2-centered interactions, which might have vast potential for application in clinics and benefit cancer patients in the future.
Collapse
Affiliation(s)
- Ting Wu
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China;
| | - Xinsheng Gu
- Department of Pharmacology, College of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, China;
| | - Hongmei Cui
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China;
- Correspondence:
| |
Collapse
|
14
|
Recent advances in research of colchicine binding site inhibitors and their interaction modes with tubulin. Future Med Chem 2021; 13:839-858. [PMID: 33821673 DOI: 10.4155/fmc-2020-0376] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Microtubules have been a concerning target of cancer chemotherapeutics for decades, and several tubulin-targeted agents, such as paclitaxel, vincristine and vinorelbine, have been approved. The colchicine binding site is one of the primary targets on microtubules and possesses advantages compared with other tubulin-targeted agents, such as inhibitors of tumor vessels and overcoming P-glycoprotein overexpression-mediated multidrug resistance. This study reviews and summarizes colchicine binding site inhibitors reported in recent years with structural studies via the crystal structures of complexes or computer simulations to discover new lead compounds. We are attempting to resolve the challenge of colchicine site agent research.
Collapse
|
15
|
Peppers: A "Hot" Natural Source for Antitumor Compounds. Molecules 2021; 26:molecules26061521. [PMID: 33802144 PMCID: PMC8002096 DOI: 10.3390/molecules26061521] [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: 02/18/2021] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 12/20/2022] Open
Abstract
Piper, Capsicum, and Pimenta are the main genera of peppers consumed worldwide. The traditional use of peppers by either ancient civilizations or modern societies has raised interest in their biological applications, including cytotoxic and antiproliferative effects. Cellular responses upon treatment with isolated pepper-derived compounds involve mechanisms of cell death, especially through proapoptotic stimuli in tumorigenic cells. In this review, we highlight naturally occurring secondary metabolites of peppers with cytotoxic effects on cancer cell lines. Available mechanisms of cell death, as well as the development of analogues, are also discussed.
Collapse
|
16
|
Liu X, Xu R, Wang L, Liu Y, Chen Z, Qin W, Tian Y. Synthesis and Evaluation in vitro of Dihydrothiophenopyridine-Chalcone Derivatives as Anticancer Activity Agents. CHINESE J ORG CHEM 2021. [DOI: 10.6023/cjoc202101014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
17
|
Evaluation of the anticancer activity of enzymatically synthesized Baccatin III: an intermediate precursor of Taxol®. 3 Biotech 2020; 10:465. [PMID: 33088661 DOI: 10.1007/s13205-020-02457-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/27/2020] [Indexed: 01/05/2023] Open
Abstract
Baccatin III is an important precursor for the synthesis of clinically important anticancer drug Taxol. Previously, we have characterized a key enzyme of 10-deacetylbaccatin III-10-β-O-acetyltransferase (DBAT) which catalyses the 10-deacetylbaccatin III into baccatin III in taxol biosynthesis. Here, in the present study, we have evaluated and compared the cytotoxic properties of the enzymatically synthesized baccatin III (ESB III) with standard baccatin III in different human cancer cell lines, namely human cervical cancer (HeLa), human lung cancer (A549), human skin cancer (A431) and human liver cancer cells (HepG2). Among the various cancer lines tested, HeLa was more susceptible to ESB III with IC50 of 4.30 µM while IC50 values for A549, A431 and HepG2 ranged from 4 to 7.81 µM. Further, it showed G2/M phase cell cycle arrest, production of reactive oxygen species and depolarised mitochondrial membrane potential. In addition, annexin V-FITC staining was performed which showed the apoptotic cell death of HeLa cells, when treated with ESB III. Hence, ESB III was capable to show anticancer activities by inducing apoptotic cell death which could further be used for the semisynthesis of taxol in future.
Collapse
|
18
|
Bian T, Corral P, Wang Y, Botello J, Kingston R, Daniels T, Salloum RG, Johnston E, Huo Z, Lu J, Liu AC, Xing C. Kava as a Clinical Nutrient: Promises and Challenges. Nutrients 2020; 12:E3044. [PMID: 33027883 PMCID: PMC7600512 DOI: 10.3390/nu12103044] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 12/20/2022] Open
Abstract
Kava beverages are typically prepared from the root of Piper methysticum. They have been consumed among Pacific Islanders for centuries. Kava extract preparations were once used as herbal drugs to treat anxiety in Europe. Kava is also marketed as a dietary supplement in the U.S. and is gaining popularity as a recreational drink in Western countries. Recent studies suggest that kava and its key phytochemicals have anti-inflammatory and anticancer effects, in addition to the well-documented neurological benefits. While its beneficial effects are widely recognized, rare hepatotoxicity had been associated with use of certain kava preparations, but there are no validations nor consistent mechanisms. Major challenges lie in the diversity of kava products and the lack of standardization, which has produced an unmet need for quality initiatives. This review aims to provide the scientific community and consumers, as well as regulatory agencies, with a broad overview on kava use and its related research. We first provide a historical background for its different uses and then discuss the current state of the research, including its chemical composition, possible mechanisms of action, and its therapeutic potential in treating inflammatory and neurological conditions, as well as cancer. We then discuss the challenges associated with kava use and research, focusing on the need for the detailed characterization of kava components and associated risks such as its reported hepatotoxicity. Lastly, given its growing popularity in clinical and recreational use, we emphasize the urgent need for quality control and quality assurance of kava products, pharmacokinetics, absorption, distribution, metabolism, excretion, and foundational pharmacology. These are essential in order to inform research into the molecular targets, cellular mechanisms, and creative use of early stage human clinical trials for designer kava modalities to inform and guide the design and execution of future randomized placebo controlled trials to maximize kava's clinical efficacy and to minimize its risks.
Collapse
Affiliation(s)
- Tengfei Bian
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; (T.B.); (P.C.); (Y.W.); (J.B.)
| | - Pedro Corral
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; (T.B.); (P.C.); (Y.W.); (J.B.)
| | - Yuzhi Wang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; (T.B.); (P.C.); (Y.W.); (J.B.)
| | - Jordy Botello
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; (T.B.); (P.C.); (Y.W.); (J.B.)
| | - Rick Kingston
- College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Tyler Daniels
- Thorne Research Inc., Industrial Road, 620 Omni Dr, Summerville, SC 29483, USA;
| | - Ramzi G. Salloum
- Department of Health Outcome & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Edward Johnston
- The Association for Hawaiian Awa (kava), Pepe’ekeo, HI 96783, USA;
| | - Zhiguang Huo
- Department of Biostatistics, College of Public Health & Health Professions, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Junxuan Lu
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA 17033, USA;
| | - Andrew C. Liu
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Chengguo Xing
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; (T.B.); (P.C.); (Y.W.); (J.B.)
| |
Collapse
|
19
|
Celentano A, Yiannis C, Paolini R, Zhang P, Farah CS, Cirillo N, Yap T, McCullough M. Kava constituents exert selective anticancer effects in oral squamous cell carcinoma cells in vitro. Sci Rep 2020; 10:15904. [PMID: 32985597 PMCID: PMC7522996 DOI: 10.1038/s41598-020-73058-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022] Open
Abstract
Kava is a beverage made from the ground roots of the plant Piper Methysticum. Active compounds of Kava have previously been demonstrated to exert an antiproliferative effect through cell cycle arrest and promotion of apoptosis. Our aim was to investigate the in vitro effects of the main constituents derived from Kava on oral squamous cell carcinoma (OSCC) activity. Gas chromatography mass spectrometry (GCMS) was used to characterise the main constituents of two Kava preparations. Cell proliferation was assessed in two human OSCC cell lines (H400 and BICR56) and in normal oral keratinocytes (OKF6) treated with the identified Kava constituents, namely Flavokawain A (FKA), Flavokawain B (FKB), yangonin, kavain and methysticin using an MTS in vitro assay. Cell migration at 16 h was assessed using a Transwell migration assay. Cell invasion was measured at 22 h using a Matrigel assay. Cell adhesion was assessed at 90 min with a Cytoselect Adhesion assay. The two Kava preparations contained substantially different concentrations of the main chemical constituents. Treatment of malignant and normal oral keratinocyte cell lines with three of the identified constituents, 10 μg/ml FKA, 2.5 μg/ml FKB and 10 μg/ml yangonin, showed a significant reduction in cell proliferation in both H400 and BICR56 cancer cell lines but not in normal OKF6 cells. Remarkably, the same Kava constituents induced a significant reduction of OSCC cell migration and invasion. We have demonstrated, for the first time, that Kava constituents, FKA, FKB and yangonin have potential anticancer effects on OSCC. This highlights an avenue for further research of Kava constituents in the development of future cancer therapies to prevent and treat OSCC.
Collapse
Affiliation(s)
- Antonio Celentano
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia.
| | - Callisthenis Yiannis
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia
| | - Rita Paolini
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia
| | - Pangzhen Zhang
- School of Agriculture and Food, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, 142 Royal Parade, Parkville, VIC, 3052, Australia
| | - Camile S Farah
- Australian Centre for Oral Oncology Research and Education, Perth, WA, 6009, Australia.,Oral, Maxillofacial and Dental Surgery, Fiona Stanley Hospital, Murdoch, WA, 6150, Australia
| | - Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia
| | - Tami Yap
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia
| | - Michael McCullough
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia
| |
Collapse
|
20
|
Li J, Zheng L, Wang R, Sun D, Liang S, Wu J, Liu Y, Tian X, Li T, Yang Y, Han L. Synergistic Combination of Sodium Aescinate-Stabilized, Polymer-Free, Twin-Like Nanoparticles to Reverse Paclitaxel Resistance. Int J Nanomedicine 2020; 15:5839-5853. [PMID: 32848393 PMCID: PMC7428345 DOI: 10.2147/ijn.s259432] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The development of paclitaxel (PTX) resistance seriously restricts its clinical efficacy. An attractive option for combating resistance is inhibiting the expression of P-glycoprotein (P-gp) in tumor cells. We have reported that flavokawain A (FKA) inhibited P-gp protein expression in PTX-resistant A549 (A549/T) cells, indicating that FKA combined with PTX may reverse PTX resistance. However, due to the variable pharmacokinetics of FKA and PTX, the conventional cocktail combination in clinics may cause uncertainty of treatment efficacy in vivo. MATERIALS AND METHODS To synergistically elevate the anti-cancer activity of PTX and FKA in vivo, the national medical products administration (NMPA) approved sodium aescinate (Aes) was utilized to stabilize hydrophobic PTX and FKA to form polymer-free twin like PTX-A nanoparticles (NPs) and FKA-A NPs. RESULTS The resulting nanoparticles prepared simply by nanoprecipitation possessed similar particle size, good stability and ultrahigh drug loadings of up to 50%. With the aid of Aes, these two drugs accumulated in tumor tissue by passive targeting and were efficiently taken up by A549/T cells; this resulted in significant suppression of tumor growth in A549/T homograft mice at a low PTX dose (2.5 mg·kg-1). Synergistic effects and reversed PTX resistance were achieved by the combination of PTX-A NPs and FKA-A NPs by inhibiting P-gp expression in tumor cells. CONCLUSION Using NMPA-approved Aes to prepare twin-like nanoparticles without introducing any new materials provides an efficient platform for combination chemotherapy and clinical translation.
Collapse
Affiliation(s)
- Juan Li
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Lei Zheng
- Department of Pharmacy, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250031, People’s Republic of China
| | - Rongmei Wang
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Deqing Sun
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Shuang Liang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, People’s Republic of China
| | - Jing Wu
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Yongqing Liu
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Xiaona Tian
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Tingting Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Yang Yang
- China National Center for Biotechnology Development, Beijing100039, People’s Republic of China
| | - Leiqiang Han
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| |
Collapse
|
21
|
Yan L, Lin M, Pan S, Assaraf YG, Wang ZW, Zhu X. Emerging roles of F-box proteins in cancer drug resistance. Drug Resist Updat 2020; 49:100673. [PMID: 31877405 DOI: 10.1016/j.drup.2019.100673] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/02/2019] [Accepted: 12/04/2019] [Indexed: 12/24/2022]
Abstract
Chemotherapy continues to be a major treatment strategy for various human malignancies. However, the frequent emergence of chemoresistance compromises chemotherapy efficacy leading to poor prognosis. Thus, overcoming drug resistance is pivotal to achieve enhanced therapy efficacy in various cancers. Although increased evidence has revealed that reduced drug uptake, increased drug efflux, drug target protein alterations, drug sequestration in organelles, enhanced drug metabolism, impaired DNA repair systems, and anti-apoptotic mechanisms, are critically involved in drug resistance, the detailed resistance mechanisms have not been fully elucidated in distinct cancers. Recently, F-box protein (FBPs), key subunits in Skp1-Cullin1-F-box protein (SCF) E3 ligase complexes, have been found to play critical roles in carcinogenesis, tumor progression, and drug resistance through degradation of their downstream substrates. Therefore, in this review, we describe the functions of FBPs that are involved in drug resistance and discuss how FBPs contribute to the development of cancer drug resistance. Furthermore, we propose that targeting FBPs might be a promising strategy to overcome drug resistance and achieve better treatment outcome in cancer patients. Lastly, we state the limitations and challenges of using FBPs to overcome chemotherapeutic drug resistance in various cancers.
Collapse
Affiliation(s)
- Linzhi Yan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Min Lin
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Shuya Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Lab, Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel.
| | - Zhi-Wei Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
22
|
Li K, Zhao S, Long J, Su J, Wu L, Tao J, Zhou J, Zhang J, Chen X, Peng C. A novel chalcone derivative has antitumor activity in melanoma by inducing DNA damage through the upregulation of ROS products. Cancer Cell Int 2020; 20:36. [PMID: 32021565 PMCID: PMC6993520 DOI: 10.1186/s12935-020-1114-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 01/17/2020] [Indexed: 12/11/2022] Open
Abstract
Background Melanoma is one of the most aggressive tumors with the remarkable characteristic of resistance to traditional chemotherapy and radiotherapy. Although targeted therapy and immunotherapy benefit advanced melanoma patient treatment, BRAFi (BRAF inhibitor) resistance and the lower response rates or severe side effects of immunotherapy have been observed, therefore, it is necessary to develop novel inhibitors for melanoma treatment. Methods We detected the cell proliferation of lj-1-59 in different melanoma cells by CCK 8 and colony formation assay. To further explore the mechanisms of lj-1-59 in melanoma, we performed RNA sequencing to discover the pathway of differential gene enrichment. Western blot and Q-RT-PCR were confirmed to study the function of lj-1-59 in melanoma. Results We found that lj-1-59 inhibits melanoma cell proliferation in vitro and in vivo, induces cell cycle arrest at the G2/M phase and promotes apoptosis in melanoma cell lines. Furthermore, RNA-Seq was performed to study alterations in gene expression profiles after treatment with lj-1-59 in melanoma cells, revealing that this compound regulates various pathways, such as DNA replication, P53, apoptosis and the cell cycle. Additionally, we validated the effect of lj-1-59 on key gene expression alterations by Q-RT-PCR. Our findings showed that lj-1-59 significantly increases ROS (reactive oxygen species) products, leading to DNA toxicity in melanoma cell lines. Moreover, lj-1-59 increases ROS levels in BRAFi -resistant melanoma cells, leading to DNA damage, which caused G2/M phase arrest and apoptosis. Conclusions Taken together, we found that lj-1-59 treatment inhibits melanoma cell growth by inducing apoptosis and DNA damage through increased ROS levels, suggesting that this compound is a potential therapeutic drug for melanoma treatment.
Collapse
Affiliation(s)
- Keke Li
- 1The Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan China.,2Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,3Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Shuang Zhao
- 1The Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan China.,2Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,3Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Jing Long
- 1The Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan China.,2Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,3Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Juan Su
- 1The Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan China.,2Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,3Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Lisha Wu
- 1The Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan China.,2Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,3Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Juan Tao
- 4Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianda Zhou
- 5Department of Plastic Surgery of Third Xiangya Hospital, Central South University, Changsha, China
| | - JiangLin Zhang
- 1The Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan China.,2Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,3Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Xiang Chen
- 1The Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan China.,2Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,3Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Cong Peng
- 1The Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan China.,2Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan China.,3Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan China
| |
Collapse
|
23
|
Design, synthesis, and validation of novel nitrogen-based chalcone analogs against triple negative breast cancer. Eur J Med Chem 2019; 187:111954. [PMID: 31838326 DOI: 10.1016/j.ejmech.2019.111954] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/24/2022]
Abstract
Great strides have been made in triple negative breast cancer (TNBC) treatment, which represents 20% of total predicted annual US breast cancer (BC) cases. Despite the development of several therapeutics, TNBC patients have poor overall survival rate, compared to other BC patients, justifying the urgent need to discover new entities for use to control TNBC. Chalcones are important natural products with diverse bioactivities including anticancer effects. This study aimed to design, synthesize and validate novel chalcone leads as potential therapies for TNBC. Fourteen novel chalcone analogs were designed and synthesized comprising alicyclic amines (pyrrolidine, morpholine and piperidine) or nitrogen mustard (Bis-(2-chloroethyl) amine) substituents. Among them, compound 14((E)-3-(4-(Bis(2-chloroethyl) amino) phenyl)-1-(3-methoxyphenyl) prop-2-en-1-one) was identified as the most effective against TNBC and other BC phenotypes, with anti-proliferative IC50 values ranging between 3.94 and 9.22 μM against the TNBC cell lines MDA-MB-231 and MDA-MB-468, as well as against the estrogen positive MCF-7 cell line. Chalcone 14 effectively suppressed the colony formation capacity of MDA-MB-231, MDA-MB-468, and MCF-7 cell lines at 5 and 10 μM treatment concentrations. Furthermore, compound 14 has significantly inhibited cell invasion and migration of MDA-MB-231 and MCF-7 BC cell lines. Additionally, compound 14 had significantly promoted apoptosis by upregulating BAX and downregulating Bcl-2 proteins. Compound 14 induced significant cell cycle arrest of TNBC cells at the G2/M phase. It also induced a reversal of Epithelial Mesenchymal Transition (EMT) by upregulating the epithelial markers E-cadherin and Pan-cadherin and downregulating FAK. Furthermore, it had dramatically diminished new vessel formation (vasculogenesis) in chick chorioallantoic membrane (CAM) model by 60.20 ± 8.47%. Chalcone 14 inhibited 46.41 ± 0.71% of the TNBC MAD-MB-231 cells growth in a nude mouse orthotopic xenograft model in comparison with vehicle control treated animals. Collectively, this study results propose chalcone 14 as a promising lead molecule for the control of TNBC as well as other breast cancer phenotypes.
Collapse
|
24
|
Li J, Zheng L, Yan M, Wu J, Liu Y, Tian X, Jiang W, Zhang L, Wang R. Activity and mechanism of flavokawain A in inhibiting P-glycoprotein expression in paclitaxel resistance of lung cancer. Oncol Lett 2019; 19:379-387. [PMID: 31897150 PMCID: PMC6923923 DOI: 10.3892/ol.2019.11069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/11/2019] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is one of the most common cancers, which is the leading cause of cancer-related death among various cancers worldwide. Flavokawain A (FKA), a chalcone found in the kava plant, exerts potent anticancer activity. However, the activity and mechanisms of FKA in inhibiting the viability of paclitaxel (PTX)-resistant lung cancer A549 (A549/T) have not been investigated. In the present study, the effect of FKA on the viability of A549/T and hepatotoxicity in normal liver epithelial cells was detected by Cell Counting Kit-8 assay. Flow cytometry, western blot analysis and Annexin V-FITC/PI apoptosis detection kit were used to assess cell apoptosis. The effect of FKA on permeability-glycoprotein (P-gp) expression was measured by reverse transcription-PCR and western blot analysis. The results indicated that FKA dose-dependently inhibited cell proliferation and induced cell apoptosis in PTX-resistant A549/T cells, with an IC50 value of ~21 µM, while the IC50 value of A549/T cells to PTX was 34.64 µM. FKA had no hepatic toxicity in liver epithelial cells. P-gp, which contributes to the chemoresistant phenotype, was not expressed in A549 cells but was remarkably enhanced in A549/T cells. FKA (30 µM) decreased P-gp protein expression at 24 h by 3-fold. Furthermore, FKA downregulated P-gp expression by blocking the PI3K/Akt pathway. These findings suggest FKA as a potential candidate for the treatment of PTX-resistant lung cancer.
Collapse
Affiliation(s)
- Juan Li
- Department of Clinical Pharmacy, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Lei Zheng
- Department of Pharmacy, Shandong Provincial Third Hospital, Jinan, Shandong 250031, P.R. China
| | - Mi Yan
- Department of Clinical Pharmacy, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Jing Wu
- Department of Clinical Pharmacy, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yongqing Liu
- Department of Clinical Pharmacy, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xiaona Tian
- Department of Clinical Pharmacy, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Wen Jiang
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Lu Zhang
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Rongmei Wang
- Department of Clinical Pharmacy, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
25
|
朱 星, 杨 嘉, 张 恩, 乔 炜, 李 学. [Bioinformatic analysis of direct protein targets of aspirin against human breast cancer proliferation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:1141-1148. [PMID: 31801720 PMCID: PMC6867953 DOI: 10.12122/j.issn.1673-4254.2019.10.02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To explore the molecular mechanism underlying the inhibitory effects of aspirin against human breast cancer cell proliferation through bioinformatics analysis. METHODS Drug Bank 5.1.3 was searched to identify direct protein targets (DPTs) of aspirin, and the protein-protein interaction (PPI) network of the DPTs was constructed online using STRING and the signaling pathways involved were identified. The genetic alterations of 6 DPTs associated with human breast cancer was analyzed and visualized by cBio Portal and OncoPrint, respectively. The transcriptomic data of breast cancer and normal tissues were downloaded from TCGA database, and the overexpressed genes were analyzed by DECenter. The intersection between the genes associated with the DPTs obtained by STRING analysis and the differentially over-expressed genes in TCGA was determined to confirm the candidate DPTs as a potential target of aspirin, and GO functional enrichment analysis was performed using Gene Ontology. The potential targets of aspirin against the proliferation of human breast cancer cells were verified by Western blotting. RESULTS Eleven DPTs of aspirin were identified. KEGG pathway enrichment indicated that 6 genes (EDNRA, IKBKB, NFKB2, NFKBIA, PTGS2 and TP53) were associated with the occurrence and development of cancer. A total of 10 220 differentially expressed genes were identified from the TCGA database, and among them 4 genes (CDC25C, TPX2, CDC20, PLK1) were found to be the potential targets for aspirin. These genes were involved mostly in the regulation of cell cycle and cell division. Western blotting showed that aspirin could down-regulate the expression levels of several pivotal proteins that regulated cell cycle and cell division, including CDC25C, TPX2, CDC20 and PLK1. CONCLUSIONS CDC25C, TPX2, CDC20 and PLK1 may be potential targets for aspirin to inhibit the proliferation of human breast cancer cells, by affecting the progress of cell cycle and cell division.
Collapse
Affiliation(s)
- 星枚 朱
- 陕西中医药大学药学院药理学教研室,陕西 咸阳 712046Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, China
- 陕西省中医药管理局中药药效机制与物质基础重点研究室,陕西 咸阳 712046Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Xianyang 712046, China
- 陕西省中药基础与新药研究重点实验室,陕西 咸阳 712046Shaanxi Key Laboratory of Traditional Medicine Foundation and New Drug Research, Xianyang 712046, China
| | - 嘉妮 杨
- 陕西中医药大学药学院药理学教研室,陕西 咸阳 712046Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, China
- 陕西省中医药管理局中药药效机制与物质基础重点研究室,陕西 咸阳 712046Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Xianyang 712046, China
- 陕西省中药基础与新药研究重点实验室,陕西 咸阳 712046Shaanxi Key Laboratory of Traditional Medicine Foundation and New Drug Research, Xianyang 712046, China
| | - 恩户 张
- 陕西中医药大学药学院药理学教研室,陕西 咸阳 712046Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, China
- 陕西省中医药管理局中药药效机制与物质基础重点研究室,陕西 咸阳 712046Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Xianyang 712046, China
| | - 炜 乔
- 中国兵器工业五二一医院,陕西 西安 710065Department of Gastroenterology, 521 Hospital of Norinco Group, Shaanxi, Xi'an, 710065, China
| | - 学军 李
- 北京大学医学部药理学系,北京 100191School of Basic Medicine, Peking University Health Science Center, Beijing, 100191, China
| |
Collapse
|
26
|
Celentano A, Tran A, Testa C, Thayanantha K, Tan-Orders W, Tan S, Syamal M, McCullough MJ, Yap T. The protective effects of Kava (Piper Methysticum) constituents in cancers: A systematic review. J Oral Pathol Med 2019; 48:510-529. [PMID: 31172600 DOI: 10.1111/jop.12900] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Kava is a beverage made from the ground roots of the plant Piper Methysticum and has long-held a significant place within Pacific island communities. Active compounds were extracted from kava, and secondary metabolites include kavalactones, chalcones, cinnamic acid derivatives and flavanones. It is thought that components of kava may exert an antiproliferative effect through cell cycle arrest and promotion of apoptosis. METHODS We conducted a systematic review to summarize available evidence of the anticancer effects of kava components and investigate their potential use for oral squamous cell carcinoma (OSCC) treatment. Eligible studies were identified through a comprehensive search of OVID EMBASE, OVID MEDLINE and Web of Science, as at April 2018. RESULTS Of 39 papers that met the inclusion criteria, 32 included in vitro models and 13 included animal studies. A total of 26 different cancers were assessed with 32 studies solely assessing epithelial cancers, 6 mesenchymal cancers and 1 study including both. There was only one report assessing an OSCC cell line. Antiproliferative properties were demonstrated in 32 out of 39 papers. The most researched constituent of kava was flavokavain B followed by flavokavain A. Both were associated with increased expression of pro-apoptotic proteins and decreased expression of anti-apoptotic proteins. Further, they were associated with a dose-dependent reduction of angiogenesis. CONCLUSION There was heterogeneity of study models and methods of investigation across the studies identified. Components of kava appear to present an area of interest with chemotherapeutic potential in cancer prevention and treatment, particularly for epithelial neoplasms. To date, there is a paucity of literature of the utility of kava components in the prevention and treatment of oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Antonio Celentano
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew Tran
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Claire Testa
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Krishen Thayanantha
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| | - William Tan-Orders
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Stephanie Tan
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Mitali Syamal
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael J McCullough
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Tami Yap
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Keshavarz M, Asadi MH. Long non-coding RNA ES1 controls the proliferation of breast cancer cells by regulating the Oct4/Sox2/miR-302 axis. FEBS J 2019; 286:2611-2623. [PMID: 30927330 DOI: 10.1111/febs.14825] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 12/06/2018] [Accepted: 03/27/2019] [Indexed: 12/23/2022]
Abstract
ES1 is a long non-coding RNA (lncRNA) that regulates pluripotency of human embryonic stem cells, which is known to be a downstream target of stemness factors Oct4 and Nanog, and serves as a modular scaffold for Sox2. However, the role of ES1 in cancer biology is not fully characterized. The results of our study show that ES1 transcript is upregulated in both high-grade and P53-mutated breast tumor tissues. Knockdown experiments show that ES1 suppression in breast cancer cells restricts cancer cell proliferation and cell cycle progression. Moreover, ES1 inhibition can also induce apoptosis and cellular senescence. Additionally, our data reveal that ES1 transcript promotes cell migration as well as the epithelial to mesenchymal transition of breast cancer cells. Furthermore, loss of ES1 expression downregulates the expression of Oct4/Sox2 and consequently leads to downregulation of their targets, miR-302 and miR-106b. Altogether, for the first time, our findings reveal that ES1 controls the proliferation and death of breast cancer cells by regulating the Oct4/Sox2/miR-302/miR-106b axis.
Collapse
Affiliation(s)
- Mostafa Keshavarz
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Malek Hossein Asadi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| |
Collapse
|
28
|
Xu L, Liu S, Yang T, Shen Y, Zhang Y, Huang L, Zhang L, Ding S, Song F, Cheng W. DNAzyme Catalyzed Tyramide Depositing Reaction for In Situ Imaging of Protein Status on the Cell Surface. Theranostics 2019; 9:1993-2002. [PMID: 31037152 PMCID: PMC6485291 DOI: 10.7150/thno.31943] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/13/2019] [Indexed: 12/11/2022] Open
Abstract
Effective characterization of protein biomarkers status on the cell surface has important value in the diagnosis and treatment of diseases. Traditional immunohistochemistry can only assess the protein expression level rather than accurately reflect their interaction and oligomerization, resulting in inevitable problems for personalized therapy. Methods: Herein, we developed a novel DNAzyme-catalyzed tyramide depositing reaction (DCTDR) for in situ amplified imaging of membrane protein status. By using human epidermal growth factor receptor 2 (HER2) as model, the binding of HER2 proteins with specific aptamers induced the formation of activated hemin/G-quadruplex (G4) DNAzyme on the cell surface to catalyze the covalent deposition of fluorescent tyramide on the membrane proteins for fluorescence imaging. Results: The DCTDR-based imaging can conveniently characterize total HER2 expression and HER2 dimerization on the breast cancer cell surface with the application of aptamer-G4 probes and proximity aptamer-split G4 probes, respectively. The designed DCTDR strategy was successfully applied to quantitatively estimate total HER2 expression and HER2 homodimer on clinical breast cancer tissue sections with high specificity and accuracy. Conclusion: The DCTDR strategy provides a simple, pragmatic and enzyme-free toolbox to conveniently and sensitively analyze protein status in clinical samples for improving clinical research, cancer diagnostics and personalized treatment.
Collapse
|
29
|
Yan W, Xiangyu C, Ya L, Yu W, Feng X. An orally antitumor chalcone hybrid inhibited HepG2 cells growth and migration as the tubulin binding agent. Invest New Drugs 2019; 37:784-790. [PMID: 30740631 DOI: 10.1007/s10637-019-00737-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/31/2019] [Indexed: 01/03/2023]
Abstract
Liver cancer is a kind of high mortality cancer due to the difficulty of early diagnosis. It is necessary to develop the anticancer agents to treat liver cancer. Here, a novel chalcone derivative was synthesized and evaluated for anticancer activity in vitro against liver cancer cell lines (HepG2, SNU-423, SMMC7221, and SNU-398). The chalcone hybrid 9 displayed the antiproliferative effect against HepG2, SNU-423, SMMC7221 and SNU-398 cells with IC50 values of 0.9 μM, 2.7 μM, 6.2 μM and 4.6 μM, respectively. Cellular mechanisms showed that derivative 9 could obviously inhibit HepG2 cells growth and colony formation in a concentration-dependent manner. Analogue 9 inhibited the migration by regulating the expression levels of migration-releated markers and transcription factors (Snail and Slug). Tubulin polymerization inhibition assay illustrated that chalcone hybrid 9 might be a potent tubulin polymerization inhibitor. Importantly, compound 9 displayed the antitumor activity against liver cancer HepG2 cells in vivo with the low toxicity toward mice. Therefore, compound 9 as a novel tubulin polymerization inhibitor deserves further investigation to treat liver cancer. Graphical abstract Compound 9 displayed the antitumor activity against liver cancer HepG2 cells in vivo and low toxicity toward mice Figure: Orally antitumor chalcone hybrid 9 inhibited HepG2 cells growth and migration as the tubulin binding agent.
Collapse
Affiliation(s)
- Wang Yan
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chen Xiangyu
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Li Ya
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Wang Yu
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xu Feng
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
30
|
Elkhalifa D, Alali F, Al Moustafa AE, Khalil A. Targeting triple negative breast cancer heterogeneity with chalcones: a molecular insight. J Drug Target 2019; 27:830-838. [PMID: 30582377 DOI: 10.1080/1061186x.2018.1561889] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Triple negative breast cancers (TNBCs) are aggressive heterogeneous cancers with not yet determined conventional targeted medication. Therefore, identification of new alternatives or improved treatment options to combat this deadly disease is highly needed. On the other hand, various derived products with chalcone scaffold were historically considered excellent candidates for the development of anticancer drugs. Chalcones unique chemical structure and their substantial biological activities in cancer cells make them an extremely attractive target for the treatment of several human carcinomas including TNBCs. This review highlights the promising therapeutic role of chalcones in TNBC management.
Collapse
Affiliation(s)
- Dana Elkhalifa
- a College of Pharmacy , Qatar University , Doha , Qatar.,b Biomedical Research Centre , Qatar University , Doha , Qatar
| | - Feras Alali
- a College of Pharmacy , Qatar University , Doha , Qatar
| | - Ala-Eddin Al Moustafa
- b Biomedical Research Centre , Qatar University , Doha , Qatar.,c College of Medicine , Qatar University , Doha , Qatar.,d Oncology Department , McGill University , Montreal , Quebec , Canada
| | - Ashraf Khalil
- a College of Pharmacy , Qatar University , Doha , Qatar
| |
Collapse
|
31
|
Laka K, Makgoo L, Mbita Z. Survivin Splice Variants in Arsenic Trioxide (As₂O₃)-Induced Deactivation of PI3K and MAPK Cell Signalling Pathways in MCF-7 Cells. Genes (Basel) 2019; 10:genes10010041. [PMID: 30646589 PMCID: PMC6356383 DOI: 10.3390/genes10010041] [Citation(s) in RCA: 20] [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: 11/19/2018] [Revised: 12/30/2018] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
Several pathways are deregulated during carcinogenesis but most notably, tumour cells can lose cell cycle control and acquire resistance to apoptosis by expressing a number of anti-apoptotic proteins such as the Inhibitors of Apoptosis Protein (IAP) family of proteins that include survivin, which is implicated in cancer development. There is no study which had proven that arsenic trioxide (As2O3) has any effect on the splicing machinery of survivin and its splice variants, hence this study was aimed at determining the cytotoxic effect of As2O3 and its effect on the expression pattern of survivin splice variants in MCF-7 cells. As2O3 inhibited the growth of the MCF-7 cells in a concentration-dependent manner. The Muse® Cell Analyser showed that As2O3-induced G2/M cell cycle arrest, promoted caspase-dependent apoptosis without causing any damage to the mitochondrial membrane of MCF-7 cells. As2O3 also deactivated two survival pathways, Mitogen-Activated Protein Kinase (MAPK) and Phosphoinositide 3-Kinase (PI3K) signalling pathways in MCF-7 cells. Deactivation of the two pathways was accompanied by the upregulation of survivin 3α during As2O3-induced G2/M cell cycle arrest and apoptosis. Survivin 2B was found to be upregulated only during As2O3-induced G2/M cell cycle arrest but downregulated during As2O3-induced apoptosis. Survivin wild-type was highly expressed in the untreated MCF-7 cells, the expression was upregulated during As2O3-induced G2/M cell cycle arrest and it was downregulated during As2O3-induced apoptosis. Survivin variant ΔEx3 was undetected in both untreated and treated MCF-7 cells. Survivin proteins were localised in both the nucleus and cytoplasm in MCF-7 cells and highly upregulated during the As2O3-induced G2/M cell cycle arrest, which can be attributed to the upregulation of survivin-2B. This study has provided the first evidence showing that the novel survivin 2B splice variant may be involved in the regulation of As2O3-induced G2/M cell cycle arrest only. This splice variant can therefore, be targeted for therapeutic purposes against Luminal A breast cancer cells.
Collapse
Affiliation(s)
- Kagiso Laka
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga 0727, Polokwane, South Africa.
| | - Lilian Makgoo
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga 0727, Polokwane, South Africa.
| | - Zukile Mbita
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X1106, Sovenga 0727, Polokwane, South Africa.
| |
Collapse
|
32
|
Zhang X, Pan Y, Fu H, Zhang J. Nucleolar and Spindle Associated Protein 1 (NUSAP1) Inhibits Cell Proliferation and Enhances Susceptibility to Epirubicin In Invasive Breast Cancer Cells by Regulating Cyclin D Kinase (CDK1) and DLGAP5 Expression. Med Sci Monit 2018; 24:8553-8564. [PMID: 30476929 PMCID: PMC6278864 DOI: 10.12659/msm.910364] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Differentially expressed genes (DEGs) of IBC were selected from the Gene Expression Omnibus (GEO) chip data: GSE21422 and GSE21974. Network analysis of the DEGs and IBC-related genes was performed in STRING database to find the core gene. Thus, this study aimed to determine the role of NUSAP1 in invasive breast cancer (IBC) and to investigate its effect on drug susceptibility to epirubicin (E-ADM). MATERIAL AND METHODS The mRNA expression of NUSAP1 was determined by quantitative polymerase chain reaction (q-PCR). The protein expression was detected by Western blotting. Cell growth and growth cycle were detected by MTT assay and flow cytometry, respectively. Cell migration and invasion were tested by Transwell assay. RESULTS Through use of gene network analysis, we found that NUSAP1 interacts with IBC-related genes. NUSAP1 presented high expression in IBC tissue samples and MCF-7 cells. NUSAP1 overexpression promoted the growth, migration, and invasion of MCF-7 cells. While NUSAP1 gene silencing downregulated the expression of genes associated with cell cycle progression in G2/M phase, cyclin D kinase (CDK1) and DLGAP5 arrested cells in G2/M phase and significantly inhibited the growth, migration, and invasion of MCF-7 cells. si-NUSAP1 increased the susceptibility of MCF-7 cells to E-ADM-induced apoptosis. CONCLUSIONS Our study provides evidence that downregulation of NUSAP1 can inhibit the proliferation, migration, and invasion of IBC cells by regulating CDK1 and DLGAP5 expression and enhances the drug susceptibility to E-ADM.
Collapse
|
33
|
Kahweol inhibits proliferation and induces apoptosis by suppressing fatty acid synthase in HER2-overexpressing cancer cells. Food Chem Toxicol 2018; 121:326-335. [DOI: 10.1016/j.fct.2018.09.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/30/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022]
|
34
|
Down-regulation of Skp2 expression inhibits invasion and lung metastasis in osteosarcoma. Sci Rep 2018; 8:14294. [PMID: 30250282 PMCID: PMC6155331 DOI: 10.1038/s41598-018-32428-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 09/04/2018] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma (OS), the most common primary cancer of bone, exhibits a high propensity for local invasion and distant metastasis. This study sought to elucidate the role of S phase kinase-associated protein (Skp2) in osteosarcoma invasion and metastasis and to explore flavokawain A (FKA), a natural chalcone from kava extract, as a potential Skp2 targeting agent for preventing osteosarcoma progression. Skp2 was found to be overexpressed in multiple osteosarcoma cell lines, including 5 standard and 8 primary patient-derived cell lines. Patients whose tumors expressed high levels of Skp2 sustained a significantly worse metastasis-free (p = 0.0095) and overall survival (p = 0.0013) than those with low Skp2. Skp2 knockdown markedly reduced in vitro cellular invasion and in vivo lung metastasis in an orthotopic mouse model of osteosarcoma. Similar to Skp2 knockdown, treatment with FKA also reduced Skp2 expression in osteosarcoma cell lines and blocked the invasion of osteosarcoma cells in vitro and lung metastasis in vivo. Together, our findings suggest that Skp2 is a promising therapeutic target in osteosarcoma, and that FKA may be an effective Skp2-targeted therapy to reduce osteosarcoma metastasis.
Collapse
|
35
|
Iqbal J, Abbasi BA, Batool R, Mahmood T, Ali B, Khalil AT, Kanwal S, Shah SA, Ahmad R. Potential phytocompounds for developing breast cancer therapeutics: Nature’s healing touch. Eur J Pharmacol 2018. [DOI: 10.1016/j.ejphar.2018.03.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Ye D, Li Z, Wei C. Genistein inhibits the S-phase kinase-associated protein 2 expression in breast cancer cells. Exp Ther Med 2017; 15:1069-1075. [PMID: 29434697 PMCID: PMC5772955 DOI: 10.3892/etm.2017.5489] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/14/2017] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is one of the most lethal cancers affecting women worldwide and was estimated to account for ~30% of all new cancer diagnoses in women. Although available evidence has proved the tumor suppressor role of genistein in cancer, the underling mechanisms have remained to be fully elucidated. S-phase kinase-associated protein 2 (Skp2) has been revealed to critically enhance the pathogenesis of multiple human cancers. The present study determined whether genistein exerts its anti-tumor function by suppressing Skp2 in breast cancer cells. Genistein significantly inhibited the proliferation, invasion and migration of breast cancer cells. Furthermore, genistein treatment also induced marked apoptosis and a typical cell cycle arrest in G2/M phase. Mechanistically, genistein treatment was identified to cause a significant downregulation of Skp2. Two crucial tumor suppressors, p21 and p27, were upregulated in genistein-treated breast cancer cells. The present results revealed that genistein exerted its tumor suppressor effect at least partially via inhibition of Skp2 and promotion of its downstream targets p21 and p27. Therefore, inactivation of Skp2 by genistein may be a promising approach for breast cancer treatment.
Collapse
Affiliation(s)
- Dengfeng Ye
- Department of Oncological Surgery, The Second Hospital of Shaoxing, Shaoxing, Zhejiang 312000, P.R. China
| | - Zhian Li
- Department of Oncological Surgery, The Second Hospital of Shaoxing, Shaoxing, Zhejiang 312000, P.R. China
| | - Chunshou Wei
- Department of Oncological Surgery, The Second Hospital of Shaoxing, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|