1
|
Zhen X, Zhang M, Hao S, Sun J. Glucose-6-phosphate dehydrogenase and transketolase: Key factors in breast cancer progression and therapy. Biomed Pharmacother 2024; 176:116935. [PMID: 38876050 DOI: 10.1016/j.biopha.2024.116935] [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: 04/06/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/16/2024] Open
Abstract
Breast cancer is one of the most common malignant tumors in women and is a serious threat to women's health. The pentose phosphate pathway (PPP) is a mode of oxidative breakdown of glucose that can be divided into oxidative (oxPPP) and non-oxidative (non-oxPPP) stages and is necessary for cell and body survival. However, abnormal activation of PPP often leads to proliferation, migration, invasion, and chemotherapy resistance in breast cancer. Glucose-6-phosphate dehydrogenase (G6PD) is the rate-limiting enzyme in PPP oxidation. Nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) produced by G6PD is the raw material for cholesterol and lipid synthesis and can resist the production of oxygen species (ROS) and reduce oxidative stress damage to tumor cells. Transketolase (TKT) is a key enzyme in non-oxPPP. Ribose 5-phosphate (R5P), produced by TKT, is a raw material for DNA and RNA synthesis, and is essential for tumor cell proliferation and DNA damage repair. In this review, we describe the role and specific mechanism of the PPP and the two most important enzymes of the PPP, G6PD and TKT, in the malignant progression of breast cancer, providing strategies for future clinical treatment of breast cancer and a theoretical basis for breast cancer research.
Collapse
Affiliation(s)
- Xin Zhen
- Department of Physical examination center, China-Japan Union Hospital of Jilin University, 126 Xiantai Blvd, Changchun 130033, People's Republic of China
| | - Mingyu Zhang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Blvd, Changchun 130033, People's Republic of China
| | - Shiming Hao
- Department of Pathology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, People's Republic of China.
| | - Jing Sun
- Department of Physical examination center, China-Japan Union Hospital of Jilin University, 126 Xiantai Blvd, Changchun 130033, People's Republic of China.
| |
Collapse
|
2
|
Hao S, Meng Q, Sun H, Yang X, Liu B, Zhang Y, Zhou H, Xu Z, Wang Y. Human papillomavirus type 16 E6 promotes cervical cancer proliferation by upregulating transketolase enzymatic activity through the activation of protein kinase B. Mol Carcinog 2024; 63:339-355. [PMID: 37988232 DOI: 10.1002/mc.23656] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 10/16/2023] [Accepted: 10/30/2023] [Indexed: 11/23/2023]
Abstract
Over 99% of precancerous cervical lesions are associated with human papillomavirus (HPV) infection, with HPV types 16 and 18 (especially type 16) found in over 70% of cervical cancer cases globally. E6, a critical HPV gene, triggers malignant proliferation by degrading p53; however, this mechanism alone cannot fully explain the oncogenic effects of HPV16 E6. Therefore, we aimed to investigate new targets of HPV oncogenic mechanisms. Our results revealed significant changes in nonoxidative pentose phosphate pathway (PPP) metabolites in HPV16-positive cells. However, the role of nonoxidative PPP in HPV-associated cell transformation and tumor development remained unexplored. In this study, we investigated the impact and mechanisms of HPV16 E6 on cervical cancer proliferation using the HPV-negative cervical cancer cell line (C33A). HPV16 E6 was found to promote cervical cancer cell proliferation both in vitro and in vivo, activating the nonoxidative PPP. Transketolase (TKT), a key enzyme in the nonoxidative PPP, is highly expressed in cervical cancer tissues and associated with poor prognosis. HPV16 E6 promotes cervical cancer cell proliferation by upregulating TKT activity through the activation of AKT. In addition, oxythiamine (OT), a TKT inhibitor, hindered tumor growth, with enhanced effects when combined with cisplatin (DDP). In conclusion, HPV16 E6 promotes cervical cancer proliferation by upregulating TKT activity through the activation of AKT. OT demonstrates the potential to inhibit HPV16-positive cervical cancer growth, and when combined with DDP, could further enhance the tumor-suppressive effect of DDP.
Collapse
Affiliation(s)
- Shiming Hao
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin Province, China
| | - Qingfei Meng
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin Province, China
| | - Huihui Sun
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin Province, China
| | - Xiangzhe Yang
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin Province, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin Province, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Zhixiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin Province, China
- Department of Urology, The First Hospital of Jilin University, Changchun, China
- Department of Medicine, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
3
|
Shao L, González-Cardenete MA, Prieto-Garcia JM. In Vitro Cytotoxic Effects of Ferruginol Analogues in Sk-MEL28 Human Melanoma Cells. Int J Mol Sci 2023; 24:16322. [PMID: 38003511 PMCID: PMC10671721 DOI: 10.3390/ijms242216322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Ferruginol is a promising abietane-type antitumor diterpene able to induce apoptosis in SK-Mel-28 human malignant melanoma. We aim to increase this activity by testing the effect of a small library of ferruginol analogues. After a screening of their antiproliferative activity (SRB staining, 48 h) on SK-Mel-28 cells the analogue 18-aminoferruginol (GI50 ≈ 10 µM) was further selected for mechanistic studies including induction of apoptosis (DAPI staining, p < 0.001), changes in cell morphology associated with the treatment (cell shrinkage and membrane blebbing), induction of caspase-3/7 activity (2.5 at 48 h, 6.5 at 72 h; p < 0.0001), changes in the mitochondrial membrane potential (not significant) and in vitro effects on cell migration and cell invasion (Transwell assays, not significant). The results were compared to those of the parent molecule (ferruginol, GI50 ≈ 50 µM, depolarisation of mitochondrial membrane p < 0.01 at 72 h; no caspases 3/7 activation) and paclitaxel (GI50 ≈ 10 nM; caspases 3/7 activation p < 0.0001) as a reference drug. Computational studies of the antiproliferative activity of 18-aminoferruginol show a consistent improvement in the activity over ferruginol across a vast majority of cancer cells in the NCI60 panel. In conclusion, we demonstrate here that the derivatisation of ferruginol into 18-aminoferruginol increases its antiproliferative activity five times in SK-MEL-28 cells and changes the apoptotic mechanism of its parent molecule, ferruginol.
Collapse
Affiliation(s)
- Luying Shao
- School of Pharmacy, University College London, London WC1E 6HX, UK;
| | - Miguel A. González-Cardenete
- Instituto de Tecnología Química (UPV-CSIC), Universitat Politècnica de Valencia-Consejo Superior de Investigaciones Científicas, Avda de los Naranjos s/n, 46022 Valencia, Spain;
| | - Jose M. Prieto-Garcia
- School of Pharmacy, University College London, London WC1E 6HX, UK;
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 5UX, UK
| |
Collapse
|
4
|
Bakhsh T, Abuzahrah SS, Qahl SH, Akela MA, Rather IA. Sugiol Masters Apoptotic Precision to Halt Gastric Cancer Cell Proliferation. Pharmaceuticals (Basel) 2023; 16:1528. [PMID: 38004394 PMCID: PMC10675088 DOI: 10.3390/ph16111528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/12/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Sugiol, a natural compound with anticancer properties, has shown promise in various cancer types, but its potential in preventing gastric cancer remains uncertain. In this study, we aimed to examine the inhibitory effect of sugiol on human gastric cancer cell proliferation. Our findings demonstrate that sugiol effectively suppresses the proliferation of SNU-5 human gastric cancer cells, leading to apoptotic cell death. We assessed the chemo-preventive potential of sugiol via an MTT assay and confirmed the induction of oxidative stress using the H2DCFDA fluorescent dye. Treatment with sugiol at concentrations higher than 25 µM for 24 h resulted in an increase in intracellular levels of reactive oxygen species (ROS). This elevation of ROS levels inhibited cell-cycle progression and induced cell-cycle arrest at the G1 phase. Furthermore, our study revealed that sugiol reduces the viability and proliferation of SNU-5 cells in a dose-dependent manner. Importantly, ADME and toxicity analyses revealed that sugiol was effective and nontoxic at low doses. In parallel, we utilized the Swiss target prediction tool to identify potential targets for sugiol. Enzymes and nuclear receptors were identified as major targets. To gain insights into the molecular interactions, we performed structure-based molecular docking studies, focusing on the interaction between sugiol and STAT3. The docking results revealed strong binding interactions within the active site pocket of STAT3, with a binding affinity of -12.169 kcal/mole. Sugiol's -OH group, carbonyl group, and phenyl ring demonstrated hydrogen-bonding interactions with specific residues of the target protein, along with Vander Waals and hydrophobic interactions. These data suggest that sugiol has the potential to inhibit the phosphorylation of STAT3, which is known to play a crucial role in promoting the growth and survival of cancer cells. Targeting the dysregulated STAT3 signaling pathway holds promise as a therapeutic strategy for various human tumors. In combination with interventions that regulate cell cycle progression and mitigate the DNA damage response, the efficacy of these therapeutic approaches can be further enhanced. The findings from our study highlight the antiproliferative and apoptotic potential of sugiol against human gastric cancer cells (SNU-5). Moreover, the result underpins that sugiol's interactions with STAT3 may contribute to its inhibitory effects on cancer cell growth and proliferation. Further research is warranted to explore the full potential of sugiol as a therapeutic agent and its potential application in treating gastric cancer and other malignancies characterized by dysregulated STAT3 activity.
Collapse
Affiliation(s)
- Tahani Bakhsh
- Department of Biological Science, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Samah Sulaiman Abuzahrah
- Department of Biological Science, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Safa H. Qahl
- Department of Biological Science, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Mohamed A. Akela
- Department of Biology, College of Sciences and Humanities in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jedddah 21589, Saudi Arabia
| |
Collapse
|
5
|
Naes SM, Ab-Rahim S, Mazlan M, Amir Hashim NA, Abdul Rahman A. Increased ENT2 expression and its association with altered purine metabolism in cell lines derived from different stages of colorectal cancer. Exp Ther Med 2023; 25:212. [PMID: 37123217 PMCID: PMC10133795 DOI: 10.3892/etm.2023.11911] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/21/2023] [Indexed: 05/02/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignant cancer types worldwide. Although the purine metabolism pathway is vital for cancer cell survival, little is known about the role of equilibrative nucleoside transporter 2 (ENT2) in CRC development and its association with purine metabolites. The aim of the present study was to evaluate the levels of hypoxanthine phosphoribosyl transferase (HPRT), hypoxanthine and uric acid (UA), as well as xanthine oxidase (XO) activity, and investigate their association with ENT2 expression levels in a normal human colon cell line and CRC cell lines derived from different stages of CRC. These analyses were performed using the normal colon CCD-841CoN cell line and a panel of human CRC cell lines comprising SW480, HCT15 and HCT116, which represent Dukes' B, C and D stages, respectively. Reverse transcription-quantitative PCR was performed to determine the level of ENT2 mRNA expression. In cells of all CRC stages, the levels of HPRT and hypoxanthine were significantly higher (P<0.05), while XO activity and UA levels were significantly decreased (P<0.05), compared with those in the CCD-841CoN cell line. ENT2 expression was found to be elevated in cells derived from all stages of CRC. The Dukes' D stage cell line had higher levels of HPRT and hypoxanthine, although its ENT2 level was not significantly lower than that of the Dukes' B and C stage cell lines. Increased levels of HPRT and hypoxanthine in various stages of CRC may indicate an increase in the activity of the salvage pathway. The increased expression of ENT2 implies the importance of the ENT2 protein in facilitating hypoxanthine transport, which is required for enhanced DNA synthesis via hypoxanthine recycling. In conclusion, ENT2 may have potential as a target in the development of CRC therapeutics.
Collapse
Affiliation(s)
- Safaa M. Naes
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital Sungai Buloh, Selangor 47000, Malaysia
- Institute of Medical and Molecular Biotechnology, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital Sungai Buloh, Selangor 47000, Malaysia
| | - Sharaniza Ab-Rahim
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital Sungai Buloh, Selangor 47000, Malaysia
| | - Musalmah Mazlan
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital Sungai Buloh, Selangor 47000, Malaysia
| | - Nurul Azmir Amir Hashim
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital Sungai Buloh, Selangor 47000, Malaysia
| | - Amirah Abdul Rahman
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital Sungai Buloh, Selangor 47000, Malaysia
- Correspondence to: Dr Amirah Abdul Rahman, Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA, Cawangan Selangor, Kampus Sungai Buloh, Jalan Hospital, Sungai Buloh, Selangor 47000, Malaysia
| |
Collapse
|
6
|
Kang YE, Yi HS, Yeo MK, Kim JT, Park D, Jung Y, Kim OS, Lee SE, Kim JM, Joung KH, Lee JH, Ku BJ, Lee M, Kim HJ. Increased Pro-Inflammatory T Cells, Senescent T Cells, and Immune-Check Point Molecules in the Placentas of Patients With Gestational Diabetes Mellitus. J Korean Med Sci 2022; 37:e338. [PMID: 36513052 PMCID: PMC9745681 DOI: 10.3346/jkms.2022.37.e338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/22/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is the most common metabolic complication of pregnancy. To define the altered pathway in GDM placenta, we investigated the transcriptomic profiles from human placenta between GDM and controls. METHODS Clinical parameters and postpartum complications were reviewed in all participants. Differentially expressed canonical pathways were analyzed between the GDM and control groups based on transcriptomic analysis. CD4+ T, CD8+ T, and senescent T cell subsets were determined by flow cytometry based on staining for specific intracellular cytokines. RESULTS Gene ontology analysis revealed that the placenta of GDM revealed upregulation of diverse mitochondria or DNA replication related pathways and downregulation of T-cell immunity related pathways. The maternal placenta of the GDM group had a higher proportion of CD4+ T and CD8+ T cells than the control group. Interestingly, senescent CD4+ T cells tended to increase and CD8+ T cells were significantly increased in GDM compared to controls, along with increased programmed cell death-1 (CD274+) expression. Programmed death-ligand 1 expression in syncytotrophoblasts was also significantly increased in patients with GDM. CONCLUSION This study demonstrated increased proinflammatory T cells, senescent T cells and immune-check point molecules in GDM placentas, suggesting that changes in senescent T cells and immune-escape signaling might be related to the pathophysiology of GDM.
Collapse
Affiliation(s)
- Yea Eun Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hyon-Seung Yi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
- Laboratory of Endocrinology and Immune System, Chungnam National University College of Medicine, Daejeon, Korea
| | - Min-Kyung Yeo
- Department of Pathology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Jung Tae Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Danbit Park
- Department of Obstetrics and Gynecology, Chungnam National University Hospital, Daejeon, Korea
| | - Yewon Jung
- Department of Obstetrics and Gynecology, Chungnam National University Sejong Hospital, Sejong, Korea
| | - Ok Soon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Seong Eun Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Ji Min Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Kyong Hye Joung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Ju Hee Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Bon Jeong Ku
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Mina Lee
- Department of Obstetrics and Gynecology, Chungnam National University College of Medicine, Daejeon, Korea.
| | - Hyun Jin Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea.
| |
Collapse
|
7
|
Baker R, Hontecillas R, Tubau-Juni N, Leber AJ, Kale S, Bassaganya-Riera J. Computational modeling of complex bioenergetic mechanisms that modulate CD4+ T cell effector and regulatory functions. NPJ Syst Biol Appl 2022; 8:45. [DOI: 10.1038/s41540-022-00263-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 11/11/2022] [Indexed: 11/24/2022] Open
Abstract
AbstractWe built a computational model of complex mechanisms at the intersection of immunity and metabolism that regulate CD4+ T cell effector and regulatory functions by using coupled ordinary differential equations. The model provides an improved understanding of how CD4+ T cells are shaping the immune response during Clostridioides difficile infection (CDI), and how they may be targeted pharmacologically to produce a more robust regulatory (Treg) response, which is associated with improved disease outcomes during CDI and other diseases. LANCL2 activation during CDI decreased the effector response, increased regulatory response, and elicited metabolic changes that favored Treg. Interestingly, LANCL2 activation provided greater immune and metabolic modulation compared to the addition of exogenous IL-2. Additionally, we identified gluconeogenesis via PEPCK-M as potentially responsible for increased immunosuppressive behavior in Treg cells. The model can perturb immune signaling and metabolism within a CD4+ T cell and obtain clinically relevant outcomes that help identify novel drug targets for infectious, autoimmune, metabolic, and neurodegenerative diseases.
Collapse
|
8
|
Jin Y, Jung SN, Lim MA, Oh C, Piao Y, Kim HJ, Nguyena Q, Kang YE, Chang JW, Won HR, Koo BS. SHMT2 Induces Stemness and Progression of Head and Neck Cancer. Int J Mol Sci 2022; 23:ijms23179714. [PMID: 36077112 PMCID: PMC9456418 DOI: 10.3390/ijms23179714] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Various enzymes in the one-carbon metabolic pathway are closely related to the development of tumors, and they can all be potential targets for cancer therapy. Serine hydroxymethyltransferase2 (SHMT2), a key metabolic enzyme, is very important for the proliferation and growth of cancer cells. However, the function and mechanism of SHMT2 in head and neck cancer (HNC) are not clear. An analysis of The Cancer Genome Atlas (TCGA) data showed that the expression of SHMT2 was higher in tumor tissue than in normal tissue, and its expression was significantly associated with male sex, aggressive histological grade, lymph node metastasis, distant metastasis, advanced TNM stage, and lymphovascular invasion in HNC. SHMT2 knockdown in FADU and SNU1041 cell lines significantly inhibited cell proliferation, colony formation, migration, and invasion. Additionally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses using TCGA data revealed that SHMT2 was closely related to cancer stem cell regulation and maintenance. Furthermore, we found that silencing SHMT2 inhibited the expression of stemness markers and tumor spheroid formation compared with a control group. On the contrary, stemness markers were significantly increased after SHMT2 overexpression in HEP-2 cells. Interestingly, we found that knocking down SHMT2 reduced the expression of genes related to the Notch and Wnt pathways. Finally, silencing SHMT2 significantly reduced tumor growth and decreased stemness markers in a xenograft model. Taken together, our study suggests that targeting SHMT2 may play an important role in inhibiting HNC progression.
Collapse
Affiliation(s)
- Yanli Jin
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Seung-Nam Jung
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Mi Ae Lim
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Chan Oh
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Yudan Piao
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Hae Jong Kim
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - QuocKhanh Nguyena
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Yea Eun Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Jae Won Chang
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Ho-Ryun Won
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Bon Seok Koo
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Correspondence: ; Tel.: +82-42-280-7690
| |
Collapse
|
9
|
Hao S, Meng Q, Sun H, Li Y, Li Y, Gu L, Liu B, Zhang Y, Zhou H, Xu Z, Wang Y. The role of transketolase in human cancer progression and therapy. Biomed Pharmacother 2022; 154:113607. [PMID: 36030587 DOI: 10.1016/j.biopha.2022.113607] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/02/2022] Open
Abstract
Transketolase (TKT) is an enzyme that is ubiquitously expressed in all living organisms and has been identified as an important regulator of cancer. Recent studies have shown that the TKT family includes the TKT gene and two TKT-like (TKTL) genes; TKTL1 and TKTL2. TKT and TKTL1 have been reported to be involved in the regulation of multiple cancer-related events, such as cancer cell proliferation, metastasis, invasion, epithelial-mesenchymal transition, chemoradiotherapy resistance, and patient survival and prognosis. Therefore, TKT may be an ideal target for cancer treatment. More importantly, the levels of TKTL1 were detected using EDIM technology for the early detection of some malignancies, and TKTL1 was more sensitive and specific than traditional tumor markers. Detecting TKTL1 levels before and after surgery could be used to evaluate the surgery's effect. While targeted TKT suppresses cancer in multiple ways, in some cases, it has detrimental effects on the organism. In this review, we discuss the role of TKT in different tumors and the detailed mechanisms while evaluating its value and limitations in clinical applications. Therefore, this review provides a basis for the clinical application of targeted therapy for TKT in the future, and a strategy for subsequent cancer-related research.
Collapse
Affiliation(s)
- Shiming Hao
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Qingfei Meng
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Huihui Sun
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yao Li
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Liting Gu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Zhixiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| |
Collapse
|
10
|
Alharthy SA, Tabrez S, Mirza AA, Zughaibi TA, Firoz CK, Dutta M. Sugiol Suppresses the Proliferation of Human U87 Glioma Cells via Induction of Apoptosis and Cell Cycle Arrest. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:7658899. [PMID: 35677372 PMCID: PMC9168084 DOI: 10.1155/2022/7658899] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/09/2022] [Indexed: 11/24/2022]
Abstract
The diterpenoid, sugiol, has been reported to exert anticancer effects against a number of human cancers. However, the anticancer effects of sugiol have not been evaluated against the human glioma cells. The present study was designed to examine the effects of sugiol on the proliferation of human U87 glioma cells. The results showed that sugiol significantly (P < 0.05) suppressed the viability of the U87 cells in a concentration dependent manner and exhibited an IC50 value of 15 μM. On the other hand, the growth inhibitory effects of sugiol were minimal on the normal human astrocytes. Acridine orange and ethidium bromide staining (AO/EB) staining revealed that sugiol induces apoptosis which was further confirmed by Western blot analysis, wherein upregulation of Bax and downregulation of Bcl-2 were observed in U87 cells. Flow cytometry showed that sugiol causes cell cycle arrest at the G 0/G 1 stage. The relative percentage of G1 phase was found to be increased from 26.58% at 0 μM to 70.96% at 30 μM sugiol. Taken together, the results suggest sugiol inhibits the growth of glioma cells and may prove to be a lead molecule in the management of human glioma.
Collapse
Affiliation(s)
- Saif A. Alharthy
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shams Tabrez
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed A. Mirza
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Torki A. Zughaibi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Chelapram K. Firoz
- Department of Medical Laboratory Technology, MIMS College of Allied Health Sciences, ASTER MIMS Academy, Kerala University of Health Sciences, Kerala, India
| | - Mycal Dutta
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| |
Collapse
|
11
|
Role of Plant-Derived Active Constituents in Cancer Treatment and Their Mechanisms of Action. Cells 2022; 11:cells11081326. [PMID: 35456005 PMCID: PMC9031068 DOI: 10.3390/cells11081326] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 03/31/2022] [Accepted: 04/11/2022] [Indexed: 02/07/2023] Open
Abstract
Despite significant technological advancements in conventional therapies, cancer remains one of the main causes of death worldwide. Although substantial progress has been made in the control and treatment of cancer, several limitations still exist, and there is scope for further advancements. Several adverse effects are associated with modern chemotherapy that hinder cancer treatment and lead to other critical disorders. Since ancient times, plant-based medicines have been employed in clinical practice and have yielded good results with few side effects. The modern research system and advanced screening techniques for plants’ bioactive constituents have enabled phytochemical discovery for the prevention and treatment of challenging diseases such as cancer. Phytochemicals such as vincristine, vinblastine, paclitaxel, curcumin, colchicine, and lycopene have shown promising anticancer effects. Discovery of more plant-derived bioactive compounds should be encouraged via the exploitation of advanced and innovative research techniques, to prevent and treat advanced-stage cancers without causing significant adverse effects. This review highlights numerous plant-derived bioactive molecules that have shown potential as anticancer agents and their probable mechanisms of action and provides an overview of in vitro, in vivo and clinical trial studies on anticancer phytochemicals.
Collapse
|
12
|
Cytotoxic and Antimigration Activity of Etlingera alba (A.D.) Poulsen Rhizome. Adv Pharmacol Pharm Sci 2022; 2021:6597402. [PMID: 34993485 PMCID: PMC8727096 DOI: 10.1155/2021/6597402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/13/2021] [Accepted: 11/16/2021] [Indexed: 01/04/2023] Open
Abstract
Etlingera alba is one of the Etlingera plants that might have anticancer activity. This study aims to investigate the cytotoxic and antimetastatic activity of E. alba rhizome fractions and migration cell assay against MDA-MB-231 cell lines, which are used for triple-negative breast cancer (TNBC) treatment assay. The cytotoxic activity was assayed using CCK-8 assay, while the antimetastatic was assayed using migration cell assay for the fractions A–F. They were followed by LCMS/MS profiling to determine the chemical contents in the most active fraction. According to results obtained, fraction B was the most active fraction for cytotoxic activity with an IC50 value of 65.43 μg/mL, while fraction E was the most active fraction for antimetastasis activity against migration rate doses of 50, 100, and 200 ppm which were 6.80, 3.66, and 3.00%, respectively. Several compounds in fraction B, such as rengyolone, licochalcone A, sugiol, and spinasterol, might have been known to have activity against cancer cells, as well as aschantin and lirioresinol B dimethyl ether from fraction E. In conclusion, the chemical components from E. alba rhizome fractions provided potency for discovering new agents for cancer treatment, specifically for TNBC.
Collapse
|
13
|
Su T, Wang T, Zhang N, Shen Y, Li W, Xing H, Yang M. Long non-coding RNAs in gastrointestinal cancers: implications for protein phosphorylation. Biochem Pharmacol 2022; 197:114907. [PMID: 35007523 DOI: 10.1016/j.bcp.2022.114907] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/30/2021] [Accepted: 01/01/2022] [Indexed: 12/24/2022]
Abstract
Phosphorylation of proteins is one of the most extensively investigated post-translational protein modifications. Threonine, serine and tyrosine in proteins are the most commonly phosphorylated amino acids. Dysregulated cancer-related signaling pathways due to aberrant phosphorylation status of the key protein(s) in these pathways exist in most malignancies. Intensive studies in the recent decade have implicated long non-coding RNAs (lncRNAs) in the precise regulation of protein phosphorylation in cancers. In this review, we systematically delve into recent advance that underlines the multidimensional role of lncRNAs in modulating protein phosphorylation, regulating cancerous signaling and impacting prognosis of gastrointestinal (GI) cancers including hepatocellular carcinoma, colorectal cancer, gastric cancer, esophageal cancer, and pancreatic cancer. LncRNAs regulate protein phosphorylation via directly binding to the target protein(s), interacting with the partner protein(s) of the target protein(s) or lncRNAs-encoded small peptides. Although there are still extensive studies on disclosing the intricate interactions between lncRNAs and proteins and their impacts on protein phosphorylation, we believe that targeting lncRNAs controlling phosphorylation of key protein(s) in cancerous signaling pathways might provide novel paths for precision therapeutics of GI cancers in the future.
Collapse
Affiliation(s)
- Tao Su
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Teng Wang
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Nasha Zhang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Jinan, Shandong Province, China
| | - Yue Shen
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenwen Li
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Jinan, Shandong Province, China
| | - Huaixin Xing
- Department of Anesthesiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China.
| | - Ming Yang
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Jinan, Shandong Province, China.
| |
Collapse
|
14
|
Jin Y, Jung SN, Lim MA, Oh C, Piao Y, Kim HJ, Liu L, Kang YE, Chang JW, Won HR, Song K, Koo BS. Transcriptional Regulation of GDF15 by EGR1 Promotes Head and Neck Cancer Progression through a Positive Feedback Loop. Int J Mol Sci 2021; 22:ijms222011151. [PMID: 34681812 PMCID: PMC8538541 DOI: 10.3390/ijms222011151] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 01/01/2023] Open
Abstract
Growth and differentiation factor 15 (GDF15), a divergent member of the transforming growth factor-β (TGF-β) superfamily, has been reported to be overexpressed in different kinds of cancer types. However, the function and mechanism of GDF15 in head and neck cancer (HNC) remains unclear. The Cancer Genome Atlas (TCGA) data show that the expression of GDF15 is significantly associated with tumor AJCC stage, lymph vascular invasion and tumor grade in HNC. In this study, we confirmed that knockdown of GDF15 attenuated: cell proliferation, migration and invasion via regulation of EMT through a canonical pathway; SMAD2/3 and noncanonical pathways; PI3K/AKT and MEK/ERK in HNC cell lines. Furthermore, we found that early growth response 1 (EGR1) was a transcription factor of GDF15. Interestingly, we also demonstrated that GDF15 could regulate the expression of EGR1, which meant a positive feedback loop occurred between these two factors. Moreover, combined inhibition of both GDF15 and EGR1 in a HNC mouse xenograft model showed significantly decreased tumor volume compared to inhibition of EGR1 or GDF15 alone. Our study showed that the GDF15–EGR1 signaling axis may be a good target in HNC patients.
Collapse
Affiliation(s)
- Yanli Jin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (Y.J.); (C.O.); (Y.P.); (H.J.K.); (L.L.); (J.W.C.); (H.-R.W.)
| | - Seung-Nam Jung
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon 35015, Korea; (S.-N.J.); (M.A.L.); (K.S.)
| | - Mi Ae Lim
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon 35015, Korea; (S.-N.J.); (M.A.L.); (K.S.)
| | - Chan Oh
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (Y.J.); (C.O.); (Y.P.); (H.J.K.); (L.L.); (J.W.C.); (H.-R.W.)
| | - Yudan Piao
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (Y.J.); (C.O.); (Y.P.); (H.J.K.); (L.L.); (J.W.C.); (H.-R.W.)
| | - Hae Jong Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (Y.J.); (C.O.); (Y.P.); (H.J.K.); (L.L.); (J.W.C.); (H.-R.W.)
| | - Lihua Liu
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (Y.J.); (C.O.); (Y.P.); (H.J.K.); (L.L.); (J.W.C.); (H.-R.W.)
| | - Yea Eun Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon 35015, Korea;
| | - Jae Won Chang
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (Y.J.); (C.O.); (Y.P.); (H.J.K.); (L.L.); (J.W.C.); (H.-R.W.)
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon 35015, Korea; (S.-N.J.); (M.A.L.); (K.S.)
| | - Ho-Ryun Won
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (Y.J.); (C.O.); (Y.P.); (H.J.K.); (L.L.); (J.W.C.); (H.-R.W.)
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon 35015, Korea; (S.-N.J.); (M.A.L.); (K.S.)
| | - Kunho Song
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon 35015, Korea; (S.-N.J.); (M.A.L.); (K.S.)
| | - Bon Seok Koo
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea; (Y.J.); (C.O.); (Y.P.); (H.J.K.); (L.L.); (J.W.C.); (H.-R.W.)
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon 35015, Korea; (S.-N.J.); (M.A.L.); (K.S.)
- Correspondence: ; Tel.: +82-42-280-7690
| |
Collapse
|
15
|
Nile A, Nile SH, Shin J, Park G, Oh JW. Quercetin-3-Glucoside Extracted from Apple Pomace Induces Cell Cycle Arrest and Apoptosis by Increasing Intracellular ROS Levels. Int J Mol Sci 2021; 22:10749. [PMID: 34639090 PMCID: PMC8509831 DOI: 10.3390/ijms221910749] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 11/17/2022] Open
Abstract
Cervical cancer is a life-threatening disease and the fourth most common cancer among women worldwide. Apple pomace is a multifunctional phenolic compound possessing effective biological activity against cervical cancer cells. This study aimed to investigate the anticancer effects of quercetin-3-glucoside (Q3G) extracted from apple pomace in HeLa cell lines and analyze its molecular mechanisms. High-performance liquid chromatography revealed that Q3G, coumaric acid, phloridzin, quercetin, and phloretin are the major polyphenolic compounds constituting apple pomace. Among them, Q3G possessed the greatest antioxidant and anti-inflammatory effects in vitro and exhibited significant cytotoxic effects in HeLa cells in a dose-and time-dependent manner. Flow cytometric analysis indicated that Q3G induced cell cycle arrest at the S phase in a time-dependent manner by altering cyclin-dependent kinase 2. Moreover, it induced apoptosis via chromosomal DNA degradation and increased reactive oxygen species generation. Furthermore, Q3G treatment altered the apoptosis-associated protein expression in the cells by activating caspase-9/-3, downregulating anti-apoptosis protein B-cell lymphoma (Bcl)-2 expressions and up regulating the pro-apoptotic Bcl-2-associated X protein. BH3-interacting domain death agonist cleavage occurred prior to the degradation of an anti-apoptotic Mu-2-related death-inducing gene involved in cell death signaling. Consequently, apple pomace Q3G holds promise as an anti-inflammatory and anticancer agent for treating cervical cancer.
Collapse
Affiliation(s)
- Arti Nile
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (A.N.); (J.S.); (G.P.)
| | - Shivraj Hariram Nile
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Juhyun Shin
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (A.N.); (J.S.); (G.P.)
| | - Gyunseok Park
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (A.N.); (J.S.); (G.P.)
| | - Jae-Wook Oh
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (A.N.); (J.S.); (G.P.)
| |
Collapse
|
16
|
Bai B, Chen Q, Jing R, He X, Wang H, Ban Y, Ye Q, Xu W, Zheng C. Molecular Basis of Prostate Cancer and Natural Products as Potential Chemotherapeutic and Chemopreventive Agents. Front Pharmacol 2021; 12:738235. [PMID: 34630112 PMCID: PMC8495205 DOI: 10.3389/fphar.2021.738235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer is the second most common malignant cancer in males. It involves a complex process driven by diverse molecular pathways that closely related to the survival, apoptosis, metabolic and metastatic characteristics of aggressive cancer. Prostate cancer can be categorized into androgen dependent prostate cancer and castration-resistant prostate cancer and cure remains elusive due to the developed resistance of the disease. Natural compounds represent an extraordinary resource of structural scaffolds with high diversity that can offer promising chemical agents for making prostate cancer less devastating and curable. Herein, those natural compounds of different origins and structures with potential cytotoxicity and/or in vivo anti-tumor activities against prostate cancer are critically reviewed and summarized according to the cellular signaling pathways they interfere. Moreover, the anti-prostate cancer efficacy of many nutrients, medicinal plant extracts and Chinese medical formulations were presented, and the future prospects for the application of these compounds and extracts were discussed. Although the failure of conventional chemotherapy as well as involved serious side effects makes natural products ideal candidates for the treatment of prostate cancer, more investigations of preclinical and even clinical studies are necessary to make use of these medical substances reasonably. Therefore, the elucidation of structure-activity relationship and precise mechanism of action, identification of novel potential molecular targets, and optimization of drug combination are essential in natural medicine research and development.
Collapse
Affiliation(s)
- Bingke Bai
- Department of Chinese Medicine Authentication, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Qianbo Chen
- Department of Anesthesiology, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Rui Jing
- Department of Chinese Medicine Authentication, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Xuhui He
- Department of Chinese Medicine Authentication, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Hongrui Wang
- Department of Chinese Medicine Authentication, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yanfei Ban
- Department of Chinese Medicine Authentication, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Qi Ye
- Department of Biological Science, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Weiheng Xu
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Chengjian Zheng
- Department of Chinese Medicine Authentication, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
17
|
Qiu L, Ma Y, Chen X, Zhou L, Zhang H, Zhong G, Zhang L, Tang J. Heparin-binding growth factor (HDGF) drives radioresistance in breast cancer by activating the STAT3 signaling pathway. J Transl Med 2021; 19:344. [PMID: 34376200 PMCID: PMC8353798 DOI: 10.1186/s12967-021-03021-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022] Open
Abstract
Although reports implicate radioresistance as an important obstacle for the management of breast cancer, its molecular mechanism is elusive. Herein, we found that high HDGF levels are expressed significantly in breast cancer and exhibit a positive association with poor survival prognosis. Heparin-binding growth factor (HDGF) was upregulated in radioresistant breast cancer cells, however, its knockdown could reduce breast cancer radioresistant both in vitro and in vivo. Additionally, the binding of RXRα to HDGF promoter blocked HDGF transcriptional activity, consequently inhibiting breast cancer radioresistance. The enhanced radioresistant activity of HDGF is induced by TKT and STAT3, impacting the STAT3-Tyr705 and STAT3-Ser727 phosphorylation and STAT3 transcriptional activity. Notably, HDGF depletion renders radioresistant hypersensitive to the drug that targets STAT3 phosphorylation. This article demonstrates the novel function of HDGF as a promising molecular target for predicting radioresistance in breast cancer.
Collapse
Affiliation(s)
- Lingyun Qiu
- Oncology Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Yan Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Xiaohua Chen
- Department of Radiation Oncology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Liheng Zhou
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Haibo Zhang
- Oncology Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Guansheng Zhong
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, People's Republic of China
| | - Lei Zhang
- Department of Radiation Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China.
| | - Jianming Tang
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
18
|
Mohan CD, Rangappa S, Nayak SC, Sethi G, Rangappa KS. Paradoxical functions of long noncoding RNAs in modulating STAT3 signaling pathway in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2021; 1876:188574. [PMID: 34062154 DOI: 10.1016/j.bbcan.2021.188574] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/03/2021] [Accepted: 05/27/2021] [Indexed: 12/20/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the lethal and leading types of cancer threatening the globe with a high mortality rate. STAT3 is an oncogenic transcription factor that is aberrantly activated in several human malignancies including HCC. Many STAT3-driven genes control cell proliferation and survival, apoptotic resistance, cell cycle progression, metastasis, and chemotherapeutic resistance. STAT3 signaling is regulated by endogenous modulators such as protein tyrosine phosphatase (PTP), suppressor of cytokine signaling (SOCS), protein inhibitor of activated STAT (PIAS), and various long noncoding RNAs (lncRNAs). Interestingly, lncRNAs have been reported to exhibit oncogenic and tumor suppressor functions, and these effects are mediated through diverse molecular mechanisms including sponging of microRNAs (miRs), transcription activation/inhibition, and epigenetic modifications. In this article, we have discussed the possible role of STAT3 signaling in hepatocarcinogenesis and various mechanisms by which lncRNAs impart their oncogenic or tumor suppressive action by modulating the STAT3 pathway in HCC.
Collapse
Affiliation(s)
| | - Shobith Rangappa
- Adichunchanagiri Institute for Molecular Medicine, Adichunchanagiri University, BG Nagara 571448, Nagamangala Taluk, India
| | - S Chandra Nayak
- Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore.
| | | |
Collapse
|
19
|
Sugiol suppresses the growth, migration, and invasion of human endometrial cancer cells via induction of apoptosis and autophagy. 3 Biotech 2021; 11:234. [PMID: 33968578 DOI: 10.1007/s13205-020-02625-3] [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: 11/04/2020] [Accepted: 12/26/2020] [Indexed: 12/15/2022] Open
Abstract
Recently, diterpenoids have been shown to exhibit several health benefits including cancer prevention. In the present study, we examined the anticancer effects of sugiol diterpene against the endometrial carcinoma and attempted to explore the underlying mechanisms. The results showed that sugiol significantly (P < 0.05) inhibited the proliferation of the endometrial carcinoma cell lines (HEC-1-A, HEC-1-B, and KLE) as compared to the normal THESCs cells. The IC50 of sugiol against all the three endometrial carcinoma cell lines ranged between 14 and 18 µM as against an IC50 of 110 μM against the normal THESCs cells. Sugiol caused several changes in the morphology of the HEC-1-B cells characteristic of apoptosis. The DAPI and annexin PI assays confirmed the induction of apoptosis in HEC-1-B cells. Sugiol also triggered increase in Bax and decrease in Bcl-2 expression. The acridine orange staining revealed that the formation of autolysosomes in HEC-1-B cells upon treatment with sugiol suggestive of autophagy. The autophagy was further confirmed by increase in the expression of LC3B-II, Beclin-1, Atg5, and Atg12 and decrease in the expression of P62. The transwell assay showed that relative to the untreated HEC-1-B cells, the migration and invasion of the sugiol-treated HEC-1-B cells was significantly (P < 0.05) inhibited. Collectively, the finding of the present study revealed that sugiol suppresses the growth of human endometrial cells via induction of apoptosis and autophagy. Consistently, sugiol may prove to be an important lead molecule in the development of chemotherapy for endometrial carcinoma.
Collapse
|
20
|
Ma LT, Wang CH, Hon CY, Lee YR, Chu FH. Discovery and characterization of diterpene synthases in Chamaecyparis formosensis Matsum. which participated in an unprecedented diterpenoid biosynthesis route in conifer. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2021; 304:110790. [PMID: 33568294 DOI: 10.1016/j.plantsci.2020.110790] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/24/2020] [Accepted: 12/06/2020] [Indexed: 05/20/2023]
Abstract
Chamaecyparis formosensis Matsum. is an endemic and precious coniferous species of Taiwan, and is known for a high abundance of specialized metabolites, which contributes to the excellent timber durability. Several terpenoids were identified and isolated from C. formosensis wood and needles, and exhibit anti-fungal and anti-bacterial bioactivities, which may participate in plant defense against pathogens. In various identified compounds, not only cadinene and ferruginol, were identified in C. formosensis extracts but also unique diterpenoids, which include pisferal, totarol, and derivates of isoabienol. To understand the biosynthesis of these specific diterpenoids, we conducted a series of functional characterization of the C. formosensis diterpene synthases (CfdiTPSs), which participate in skeleton formation and differentiation of diterpenes. In this study, we identified eight diTPSs from C. formosensis transcriptome, and they all contain either class I or class II motif, which indicates they are all monofunctional enzymes. These candidates consist of three class II diTPSs and five class I diTPSs, and after conducting in vivo and in vitro assays, class II diTPS CfCPS1 was characterized as a (+)-copalyl diphosphate synthase ((+)-CPS), and class I diTPSs CfKSL1 could further convert (+)-copalyl diphosphate ((+)-CPP) to levopimaradiene. Meanwhile, CfKSL1 also accepted labda-13-en-8-ol diphosphate (LPP) as substrate and formed monoyl oxide. Another class I diTPS, CfKSL4, exhibits a strong enzymatic ability of isoabienol synthase, which is firstly reported in conifer. This finding provides potential participants in the biosynthesis of unique diterpenoids, and with this knowledge, we can further expand our understanding of diterpenoid metabolism in Cupressaceae and their potential role in plant defense.
Collapse
Affiliation(s)
- Li-Ting Ma
- School of Forestry and Resource Conservation, National Taiwan University, Taipei, 10617, Taiwan
| | - Chang-Hsin Wang
- School of Forestry and Resource Conservation, National Taiwan University, Taipei, 10617, Taiwan
| | - Chong-Yao Hon
- School of Forestry and Resource Conservation, National Taiwan University, Taipei, 10617, Taiwan
| | - Yi-Ru Lee
- School of Forestry and Resource Conservation, National Taiwan University, Taipei, 10617, Taiwan
| | - Fang-Hua Chu
- School of Forestry and Resource Conservation, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
21
|
Biological Profiling of Semisynthetic C19-Functionalized Ferruginol and Sugiol Analogues. Antibiotics (Basel) 2021; 10:antibiotics10020184. [PMID: 33673350 PMCID: PMC7918733 DOI: 10.3390/antibiotics10020184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/10/2021] [Accepted: 02/10/2021] [Indexed: 12/17/2022] Open
Abstract
The abietane-type diterpenoids are significant bioactive compounds exhibiting a varied range of pharmacological properties. In this study, the first synthesis and biological investigation of the new abietane-diterpenoid (+)-4-epi-liquiditerpenoid acid (8a) together with several of its analogs are reported. The compounds were generated from the readily available methyl callitrisate (7), which was obtained from callitrisic acid present in Moroccan Sandarac resin. A biological evaluation was conducted to determine the effects of the different functional groups present in these molecules, providing basic structure-activity relationship (SAR) elements. In particular, the ferruginol and sugiol analogs compounds 10-16 were characterized by the presence of a phenol moiety, higher oxidization states at C-7 (ketone), and the hydroxyl, methyl ester or free carboxylic acid at C19. The biological profiling of these compounds was investigated against a panel of six human solid tumor cell lines (HBL-100, A549, HeLa, T-47D, SW1573 and WiDr), four parasitic Leishmania species (L. donovani, L. infantum, L. guyanensis and L. amazonensis) and two malaria strains (3D7 and K1). Furthermore, the capacity of the compounds to modulate gamma-aminobutyric acid type A (GABAA) receptors (α1β2γ2s) is also described. A comparison of the biological results with those previously reported of the corresponding C18-functionalized analogs was conducted.
Collapse
|
22
|
EGR1/GADD45α Activation by ROS of Non-Thermal Plasma Mediates Cell Death in Thyroid Carcinoma. Cancers (Basel) 2021; 13:cancers13020351. [PMID: 33477921 PMCID: PMC7833439 DOI: 10.3390/cancers13020351] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/11/2021] [Accepted: 01/15/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Recent studies have identified new anti-cancer mechanisms of nonthermal plasma (NTP) in several cancers. However, the molecular mechanisms underlying its therapeutic effect on thyroid cancer have not been elucidated. The objective of this study was to understand the anticancer effects of NTP-activated medium (NTPAM) on thyroid cancer cells and elucidate the signaling mechanisms responsible for NTPAM-induced thyroid cancer cell death. Abstract (1) Background: Nonthermal plasma (NTP) induces cell death in various types of cancer cells, providing a promising alternative treatment strategy. Although recent studies have identified new mechanisms of NTP in several cancers, the molecular mechanisms underlying its therapeutic effect on thyroid cancer (THCA) have not been elucidated. (2) Methods: To investigate the mechanism of NTP-induced cell death, THCA cell lines were treated with NTP-activated medium -(NTPAM), and gene expression profiles were evaluated using RNA sequencing. (3) Results: NTPAM upregulated the gene expression of early growth response 1 (EGR1). NTPAM-induced THCA cell death was enhanced by EGR1 overexpression, whereas EGR1 small interfering RNA had the opposite effect. NTPAM-derived reactive oxygen species (ROS) affected EGR1 expression and apoptotic cell death in THCA. NTPAM also induced the gene expression of growth arrest and regulation of DNA damage-inducible 45α (GADD45A) gene, and EGR1 regulated GADD45A through direct binding to its promoter. In xenograft in vivo tumor models, NTPAM inhibited tumor progression of THCA by increasing EGR1 levels. (4) Conclusions: Our findings suggest that NTPAM induces apoptotic cell death in THCA through a novel mechanism by which NTPAM-induced ROS activates EGR1/GADD45α signaling. Furthermore, our data provide evidence that the regulation of the EGR1/GADD45α axis can be a novel strategy for the treatment of THCA.
Collapse
|
23
|
Bajpai VK, Sonwal S, Hwang SK, Shukla S, Khan I, Dey DK, Chen L, Simal-Gandara J, Xiao J, Huh YS, Han YK. Sugiol, a diterpenoid: Therapeutic actions and molecular pathways involved. Pharmacol Res 2021; 163:105313. [PMID: 33246173 DOI: 10.1016/j.phrs.2020.105313] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/06/2020] [Accepted: 11/13/2020] [Indexed: 02/08/2023]
Abstract
Understanding how the natural products structural diversity interacts with cellular metabolism and infectious disease targets remains a challenge. Inflammation is an important process in the human healing response in which the tissues respond to injuries induced by many agents, including pathogens. In recent years, several drugs derived from plant products have been developed, and current drug research is actively investigating the pharmacotherapeutic role of natural products in advanced multimodal inflammatory disease targeting. Sugiol, a diterpenoid, can act as an antimicrobial, antioxidant, anti-inflammatory, anti-carcinoma, antiviral, and cardiovascular agent. Until now, there have been no updates on the pharmacotherapeutic advancement of sugiol. Herein, we correlate the diverse molecular pathways in disease prevention involving sugiol. We also discuss the origins of its structural diversity and summarize new research directions toward exploring its novel effective future uses. Despite much evidence of its efficacy and safety, the sugiol has not yet been approved as a therapeutic agent due to its low bioavailability, and insolubility in an aqueous environment. The aim of this review is to renew and update noteworthy information on the pharmacotherapeutic characteristics of sugiol to approach different advanced strategies employed in the context of natural nurturing-based biomedicine.
Collapse
Affiliation(s)
- Vivek K Bajpai
- Department of Energy and Materials Engineering, Dongguk University, 30 Pildong-ro 1-gil, Seoul, 04620, South Korea
| | - Sonam Sonwal
- Department of Biological Engineering, NanoBio High-Tech Materials Research Center, Inha University, Incheon 22212, Republic of Korea
| | - Seung-Kyu Hwang
- Department of Biological Engineering, NanoBio High-Tech Materials Research Center, Inha University, Incheon 22212, Republic of Korea
| | - Shruti Shukla
- Department of Food Science and Technology, National Institute of Food Technology Entrepreneurship and Management (NIFTEM), Sonipat, Haryana, 131028, India
| | - Imran Khan
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Jillyang, Naeri-ri, Gyeongsan, Gyeongbuk, 38453, South Korea
| | - Lei Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, China.
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, E-32004 Ourense, Spain
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, E-32004 Ourense, Spain.
| | - Yun Suk Huh
- Department of Biological Engineering, NanoBio High-Tech Materials Research Center, Inha University, Incheon 22212, Republic of Korea.
| | - Young-Kyu Han
- Department of Energy and Materials Engineering, Dongguk University, 30 Pildong-ro 1-gil, Seoul, 04620, South Korea.
| |
Collapse
|
24
|
Yoon YJ, Kwon BM. Cinnamomum cassia, apoptosis, STAT3 inactivation and reactive oxygen species in cancer studies. Cancer 2021. [DOI: 10.1016/b978-0-12-819547-5.00029-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
25
|
Lee YJ, Choi J, Yoon YJ, Sim Y, Ryu HW, Oh SR, Kim DY, Hwang J, Chi SW, Han DC, Kwon BM. 8-Epi-xanthatin induces the apoptosis of DU145 prostate carcinoma cells through signal transducer and activator of transcription 3 inhibition and reactive oxygen species generation. Phytother Res 2020; 35:1508-1520. [PMID: 33164240 DOI: 10.1002/ptr.6918] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/11/2020] [Accepted: 10/01/2020] [Indexed: 12/15/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is aberrantly activated in many human cancers. We tried to find STAT3 inhibitors from natural sources and found that Xanthium fruit extracts decreased phosphorylation of STAT3-Y705. 8-Epi-xanthatin (EXT) was isolated from the extracts. When DU145 cancer cells were treated with EXT, p-STAT3-Y705 was decreased with an IC50 of 3.2 μM. EXT decreased the expression of STAT3 target genes, such as cyclin A, cyclin D1, and BCL-2, and induced PARP cleavage, indicating apoptotic cell death. Downregulation of EXT-induced p-STAT3-Y705 was rescued by pretreating DU145 cells with antioxidants, such as N-acetyl-L-cysteine (NAC), indicating that reactive oxygen species (ROS) were involved in the EXT-induced inhibition of STAT3 activation. Furthermore, we proved the association of EXT with STAT3 protein by using a drug affinity responsive target stability (DARTS) assay and a cellular thermal shift assay (CETSA). EXT inhibited proliferation of DU145 cells with a GI50 of 6 μM and reduced tumor growth in mice xenografted with DU145 cells. Immunoblotting showed that phosphorylation of STAT3-Y705 was lower in EXT-treated tumor tissue than in control tissues. Collectively, we found that EXT binds to, and inhibits, STAT3 activation and could be a lead compound for anticancer therapy.
Collapse
Affiliation(s)
- Yu-Jin Lee
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Jiyeon Choi
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Yae Jin Yoon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Yugyeong Sim
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,University of Science and Technology in Korea, Daejeon
| | - Hyung Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Doo-Young Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
| | - Jihyun Hwang
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Seung-Wook Chi
- University of Science and Technology in Korea, Daejeon.,Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Dong Cho Han
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,University of Science and Technology in Korea, Daejeon
| | - Byoung-Mog Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,University of Science and Technology in Korea, Daejeon
| |
Collapse
|
26
|
Jung SN, Kang YE, Lee GH, Liu L, Oh C, Jin YL, Lim MA, Lee K, Oh T, Won HR, Chang JW, Koo BS. Brn3a/Pou4f1 Functions as a Tumor Suppressor by Targeting c-MET/STAT3 Signaling in Thyroid Cancer. J Clin Endocrinol Metab 2020; 105:5849340. [PMID: 32474599 DOI: 10.1210/clinem/dgaa316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 05/22/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Brn3a/Pou4f1 is a class IV POU domain-containing transcription factor and has been found to be expressed in a variety of cancers. However, the mechanism and action of Brn3a in thyroid cancer has not been investigated. PURPOSE To investigate the role of Brn3a in thyroid cancer progression and its clinical implication. METHODS We examined Brn3a expression status in patients with thyroid cancer and analyzed relationships between Brn3a expression and clinicopathological findings using The Cancer Genome Atlas (TCGA) database. For functional in vitro analysis, proliferation, migration, invasion assay, and Western blotting were performed after overexpression or suppression of Brn3a. RESULTS The promoter hypermethylation of Brn3a was found in patients with aggressive thyroid cancer and Brn3a was downregulated in tissues of patients with thyroid cancer. In TCGA database, the low-Brn3a-expression group revealed a more aggressive phenotype, including T stage and extrathyroid extension when compared with the high-Brn3a-expression group. Overexpression of Brn3a suppressed cell migration and invasion via regulation of epithelial-mesenchymal transition (EMT)-associated proteins in thyroid cancer cell lines. Brn3a overexpression also downregulated signal transducer and activator of transcription 3 (STAT3) signaling through suppression of tyrosine-protein kinase Met (c-MET). In contrast, knockdown of Brn3a by small interfering ribonucleic acid (siRNA) significantly increased cell migration and invasion through upregulation of c-MET/STAT3. These results imply that Brn3a suppresses tumor metastasis via c-MET/STAT3 inhibition and EMT suppression in thyroid cancer. CONCLUSIONS Our findings show that Brn3a is a potential tumor suppressor that leads to reduced cancer cell migration and invasion in thyroid cancer. Elucidation of the Brn3a-regulated cancer pathways may therefore provide novel therapeutic strategies to control thyroid cancer metastasis.
Collapse
Affiliation(s)
- Seung-Nam Jung
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Yea Eun Kang
- Department of Endocrinology and Metabolism, Chungnam National University College of Medicine, Daejeon, Korea
| | - Gun Ho Lee
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Lihua Liu
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Chan Oh
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Yan Li Jin
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Mi Ae Lim
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Kyungmin Lee
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Taejeong Oh
- Research and Development Center, Genomictree Inc., Daejeon, Republic of Korea
| | - Ho-Ryun Won
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Jae Won Chang
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| | - Bon Seok Koo
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
27
|
Lee YJ, Song H, Yoon YJ, Park SJ, Kim SY, Cho Han D, Kwon BM. Ethacrynic acid inhibits STAT3 activity through the modulation of SHP2 and PTP1B tyrosine phosphatases in DU145 prostate carcinoma cells. Biochem Pharmacol 2020; 175:113920. [DOI: 10.1016/j.bcp.2020.113920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/17/2020] [Indexed: 01/17/2023]
|
28
|
Wang Y, Shi LY, Qi WH, Yang J, Qi Y. Anticancer activity of sugiol against ovarian cancer cell line SKOV3 involves mitochondrial apoptosis, cell cycle arrest and blocking of the RAF/MEK/ERK signalling pathway. Arch Med Sci 2020; 16:428-435. [PMID: 32190154 PMCID: PMC7069438 DOI: 10.5114/aoms.2017.71420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 09/09/2017] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Ovarian cancer is one of the leading causes of cancer-related deaths in women. Treatments for ovarian cancer include surgery followed by chemotherapy. However, the survival rate for ovarian cancer is still not satisfactory. Moreover, the current chemotherapy has numerous associated side effects. Therefore there is an urgent need to look for novel and more viable treatment options. Against this backdrop the present study was designed to evaluate the anticancer activity of sugiol against ovarian cancer cells. MATERIAL AND METHODS Cell viability was assessed by CCK8 assay, apoptosis by DAPI, AO/ER and annexin V/PI staining. Mitochondrial membrane potential and cell cycle analysis was performed by flow cytometry. Cell migration was investigated by wound healing assay. Protein expression was monitored by western blotting. RESULTS The results of the present study indicated that sugiol exerts significant (p < 0.0001) anticancer effects on SKOV3 cancer cells with an IC50 of 25 μM. However, sugiol exhibited less cytotoxicity against normal ovarian cells with an IC50 of 62.5 μM. The anticancer effects of sugiol were found to be due to G0/G1 cell cycle arrest and mitochondrial apoptosis. Sugiol also inhibited cell migration of SKOV3 cells dose dependently. Moreover, the results showed that sugiol could inhibit the RAF/MEK/ERK signalling pathway in a dose-dependent manner. CONCLUSIONS The results of the present study indicate that sugiol exerts potent anticancer effects on SKOV3 cells via induction of cell cycle arrest, mitochondrial apoptosis and inhibition of the RAF/MEK/ERK signalling pathway.
Collapse
Affiliation(s)
- Yan Wang
- Department of Gynaecology, Hubei Maternity and Child Health Hospital, Wuhan, China
| | - Liang-Yan Shi
- Department of Gynaecology, Hubei Maternity and Child Health Hospital, Wuhan, China
| | - Wei-Hong Qi
- Department of Obstetrics and Gynaecology, Beijing Hospital, Beijing, China
| | - Jing Yang
- Department of Obstetrics and Gynaecology, Haidian Woman’s and Children’s Hospital, Beijing, China
| | - Yue Qi
- Department of Obstetrics and Gynaecology, Dongguan Hospital of Traditional Chinese Medicine, Dongguan Guangdong, China
| |
Collapse
|
29
|
Yang L, Lin S, Xu L, Lin J, Zhao C, Huang X. Novel activators and small-molecule inhibitors of STAT3 in cancer. Cytokine Growth Factor Rev 2019; 49:10-22. [PMID: 31677966 DOI: 10.1016/j.cytogfr.2019.10.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022]
Abstract
Excessive activation of signal transducer and activator of transcription 3 (STAT3) signaling is observed in a subset of many cancers, making activated STAT3 a highly promising potential therapeutic target supported by multiple preclinical and clinical studies. However, early-phase clinical trials have produced mixed results with STAT3-targeted cancer therapies, revealing substantial complexity to targeting aberrant STAT3 signaling. This review discusses the diverse mechanisms of oncogenic activation of STAT3, and the small molecule inhibitors of STAT3 in cancer treatment.
Collapse
Affiliation(s)
- Lehe Yang
- Department of Respiratory Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, Wenzhou, Zhejiang 325035, China; Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Shichong Lin
- Department of Respiratory Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, Wenzhou, Zhejiang 325035, China
| | - Lingyuan Xu
- Department of Respiratory Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, Wenzhou, Zhejiang 325035, China
| | - Jiayuh Lin
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Chengguang Zhao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, Wenzhou, Zhejiang 325035, China.
| | - Xiaoying Huang
- Department of Respiratory Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
30
|
Scariot DB, Volpato H, Fernandes NDS, Soares EFP, Ueda-Nakamura T, Dias-Filho BP, Din ZU, Rodrigues-Filho E, Rubira AF, Borges O, Sousa MDC, Nakamura CV. Activity and Cell-Death Pathway in Leishmania infantum Induced by Sugiol: Vectorization Using Yeast Cell Wall Particles Obtained From Saccharomyces cerevisiae. Front Cell Infect Microbiol 2019; 9:208. [PMID: 31259161 PMCID: PMC6587907 DOI: 10.3389/fcimb.2019.00208] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 05/28/2019] [Indexed: 12/22/2022] Open
Abstract
Visceral leishmaniasis, caused by Leishmania infantum, is a neglected tropical disease, to which efforts in the innovation of effective and affordable treatments remain limited, despite the rising incidence in several regions of the world. In this work, the antileishmanial effects of sugiol were investigated in vitro. This compound was isolated from the bark of Cupressus lusitanica and showed promising activity against L. infantum. In spite of the positive results, it is known that the compound is a poorly water-soluble diterpene molecule, which hinders further investigation, especially in preclinical animal studies. Thus, in an alternative delivery method, sugiol was entrapped in glucan-rich particles obtained from Saccharomyces cerevisiae yeast cell walls (YCWPs). To evaluate the activity of sugiol, the experiments were divided into two parts: (i) the in vitro investigation of antileishmanial activity of free sugiol against L. infantum promastigotes after 24, 48, and 72 h of treatment and (ii) the evaluation of antileishmanial activity of sugiol entrapped in glucan-rich particles against intracellular L. infantum amastigotes. Free sugiol induced the cell-death process in promastigotes, which was triggered by enhancing cytosolic calcium level and promoting the autophagy up to the first 24 h. Over time, the presence of autophagic vacuoles became rarer, especially after treatment with lower concentrations of sugiol, but other cellular events intensified, like ROS production, cell shrinkage, and phosphatidylserine exposure. Hyperpolarization of mitochondrial membrane potential was found at 72 h, induced by the mitochondria calcium uptake, causing an increase in ROS production and lipid peroxidation as a consequence. These events resulted in the cell death of promastigotes by secondary necrosis. Sugiol entrapped in glucan-rich particles was specifically recognized by dectin-1 receptor on the plasma membrane of macrophages, the main host cell of Leishmania spp. Electron micrographs revealed particles containing sugiol within the infected macrophages and these particles were active against the intracellular L. infantum amastigotes without affecting the host cell. Therefore, the YCWPs act like a Trojan horse to successfully deliver sugiol into the macrophage, presenting an interesting strategy to deliver water-insoluble drugs to parasitized cells.
Collapse
Affiliation(s)
- Débora Botura Scariot
- Laboratory of Technological Innovation in Drugs and Cosmetics Development, State University of Maringá, Maringá, Brazil
| | - Hélito Volpato
- Laboratory of Technological Innovation in Drugs and Cosmetics Development, State University of Maringá, Maringá, Brazil
| | - Nilma de Souza Fernandes
- Laboratory of Technological Innovation in Drugs and Cosmetics Development, State University of Maringá, Maringá, Brazil
| | | | - Tânia Ueda-Nakamura
- Laboratory of Technological Innovation in Drugs and Cosmetics Development, State University of Maringá, Maringá, Brazil
| | - Benedito Prado Dias-Filho
- Laboratory of Technological Innovation in Drugs and Cosmetics Development, State University of Maringá, Maringá, Brazil
| | - Zia Ud Din
- Chemistry Department, Federal University of São Carlos, São Carlos, Brazil
| | | | | | - Olga Borges
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,CNC - Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Maria Do Céu Sousa
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,CNC - Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Celso Vataru Nakamura
- Laboratory of Technological Innovation in Drugs and Cosmetics Development, State University of Maringá, Maringá, Brazil
| |
Collapse
|
31
|
Hamulić D, Stadler M, Hering S, Padrón JM, Bassett R, Rivas F, Loza-Mejía MA, Dea-Ayuela MA, González-Cardenete MA. Synthesis and Biological Studies of (+)-Liquiditerpenoic Acid A (Abietopinoic Acid) and Representative Analogues: SAR Studies. JOURNAL OF NATURAL PRODUCTS 2019; 82:823-831. [PMID: 30840453 DOI: 10.1021/acs.jnatprod.8b00884] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The first semisynthesis and biological profiling of the new abietane diterpenoid (+)-liquiditerpenoic acid A (abietopinoic acid) (7) along with several analogues are reported. The compounds were obtained from readily available methyl dehydroabietate (8), which was derived from (-)-abietic acid (1). Biological comparison was conducted according to the different functional groups, leading to some basic structure-activity relationships (SAR). In particular, the ferruginol and sugiol analogues 7 and 10-16 were characterized by the presence of an acetylated phenolic moiety, an oxidized C-7 as a carbonyl, and a different functional group at C-18 (methoxycarbonyl, carboxylic acid, and hydroxymethyl). The biological properties of these compounds were investigated against a panel of six representative human tumor solid cells (A549, HBL-100, HeLa, SW1573, T-47D, and WiDr), five leukemia cellular models (NALM-06, KOPN-8, SUP-B15, UoCB1, and BCR-ABL), and four Leishmania species ( L. infantum, L. donovani, L. amazonensis, and L. guyanensis). A molecular docking study pointed out some targets in these Leishmania species. In addition, the ability of the compounds to modulate GABAA receptors (α1β2γ2s) is also reported. The combined findings indicate that these abietane diterpenoids offer a source of novel bioactive molecules with promising pharmacological properties from cheap chiral-pool building blocks.
Collapse
Affiliation(s)
- Damir Hamulić
- Instituto de Tecnología Química (UPV-CSIC) , Universitat Politècnica de Valencia-Consejo Superior de Investigaciones Científicas , Avenida de los Naranjos s/n , 46022 Valencia , Spain
| | - Marco Stadler
- Department of Pharmacology and Toxicology , University of Vienna , Althanstrasse 14 , A-1090 Vienna , Austria
| | - Steffen Hering
- Department of Pharmacology and Toxicology , University of Vienna , Althanstrasse 14 , A-1090 Vienna , Austria
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Centro de Investigaciones Biomédicas de Canarias (CIBICAN) , Universidad de La Laguna , C/Astrofísico Francisco Sanchez 2 , La Laguna 38200 , Tenerife , Spain
| | - Rachel Bassett
- Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 , United States
| | - Fatima Rivas
- Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 , United States
| | - Marco A Loza-Mejía
- Facultad de Ciencias Químicas , Universidad La Salle México , Avenue Benjamín Franklin 45 , Condesa , 06140 Ciudad de México , Mexico
| | - M Auxiliadora Dea-Ayuela
- Departamento de Farmacia , Universidad CEU Cardenal Herrera , Avenida Seminario s/n , 46113 Moncada (Valencia) , Spain
| | - Miguel A González-Cardenete
- Instituto de Tecnología Química (UPV-CSIC) , Universitat Politècnica de Valencia-Consejo Superior de Investigaciones Científicas , Avenida de los Naranjos s/n , 46022 Valencia , Spain
| |
Collapse
|
32
|
Yoon YJ, Kim YH, Lee YJ, Choi J, Kim CH, Han DC, Kwon BM. 2'-Hydroxycinnamaldehyde inhibits proliferation and induces apoptosis via signal transducer and activator of transcription 3 inactivation and reactive oxygen species generation. Cancer Sci 2018; 110:366-378. [PMID: 30375708 PMCID: PMC6317917 DOI: 10.1111/cas.13852] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/18/2018] [Accepted: 10/23/2018] [Indexed: 12/25/2022] Open
Abstract
Inhibition of the signal transducer and activator of transcription 3 (STAT3) signaling pathway is a novel therapeutic strategy to treat human cancers with constitutively active STAT3. During the screening of natural products to find STAT3 inhibitors, we identified 2′‐hydroxycinnamaldehyde (HCA) as a STAT3 inhibitor, which was isolated from the stem bark of Cinnamomum cassia. In this study, we found that HCA inhibited constitutive and inducible STAT3 activation in STAT3‐activated DU145 prostate cancer cells. HCA selectively inhibited the STAT3 activity by direct binding to STAT3, which was confirmed by biochemical methods, including a pull‐down assay with biotin‐conjugated HCA, a drug affinity responsive target stability (DARTS) experiment and a cellular thermal shift assay (CETSA). HCA inhibited STAT3 phosphorylation at the tyrosine 705 residue, dimer formation, and nuclear translocation in DU145 cells, which led to a downregulation of STAT3 target genes. The downregulation of cell cycle progression and antiapoptosis‐related gene expression by HCA induced the accumulation of cells in the G0/G1 phase of the cell cycle and then induced apoptosis. We also found that reactive oxygen species (ROS) were involved in the HCA‐induced inhibition of STAT3 activation and cell proliferation because the suppressed p‐STAT3 level was rescued by glutathione or N‐acetyl‐L‐cysteine treatment, which are general ROS inhibitors. These results suggest that HCA could be a potent anticancer agent targeting STAT3‐activated tumor cells.
Collapse
Affiliation(s)
- Yae Jin Yoon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Young-Hwan Kim
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,Department of Biology, Chungnam National University, Daejeon, Korea
| | - Yu-Jin Lee
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Jiyeon Choi
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,Department of Biology, Chungnam National University, Daejeon, Korea
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, Korea
| | - Dong Cho Han
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,Korea University of Science and Technology in Korea, Daejeon, Korea
| | - Byoung-Mog Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,Korea University of Science and Technology in Korea, Daejeon, Korea
| |
Collapse
|
33
|
Zhou P, Chen Y, Lu Q, Qin H, Ou H, He B, Ye J. Cellular metabolism network of Bacillus thuringiensis related to erythromycin stress and degradation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 160:328-341. [PMID: 29857237 DOI: 10.1016/j.ecoenv.2018.05.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/15/2018] [Accepted: 05/20/2018] [Indexed: 06/08/2023]
Abstract
Erythromycin is one of the most widely used macrolide antibiotics. To present a system-level understanding of erythromycin stress and degradation, proteome, phospholipids and membrane potentials were investigated after the erythromycin degradation. Bacillus thuringiensis could effectively remove 77% and degrade 53% of 1 µM erythromycin within 24 h. The 36 up-regulated and 22 down-regulated proteins were mainly involved in spore germination, chaperone and nucleic acid binding. Up-regulated ribose-phosphate pyrophosphokinase and ribosomal proteins confirmed that the synthesis of protein, DNA and RNA were enhanced after the erythromycin degradation. The reaction network of glycolysis/gluconeogenesis was activated, whereas, the activity of spore germination was decreased. The increased synthesis of phospholipids, especially, palmitoleic acid and oleic acid, altered the membrane permeability for erythromycin transport. Ribose-phosphate pyrophosphokinase and palmitoleic acid could be biomarkers to reflect erythromycin exposure. Lipids, disease, pyruvate metabolism and citrate cycle in human cells could be the target pathways influenced by erythromycin. The findings presented novel insights to the interaction among erythromycin stress, protein interaction and metabolism network, and provided a useful protocol for investigating cellular metabolism responses under pollutant stress.
Collapse
Affiliation(s)
- Pulin Zhou
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China
| | - Ya Chen
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China
| | - Qiying Lu
- College of Biology and Food Engineering, Guangdong University of Education, Guangzhou 510303, Guangdong, China
| | - Huaming Qin
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China
| | - Huase Ou
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China
| | - Baoyan He
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China
| | - Jinshao Ye
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China.
| |
Collapse
|
34
|
Tavares WR, Seca AML. The Current Status of the Pharmaceutical Potential of Juniperus L. Metabolites. MEDICINES 2018; 5:medicines5030081. [PMID: 30065158 PMCID: PMC6165314 DOI: 10.3390/medicines5030081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/16/2018] [Accepted: 07/20/2018] [Indexed: 12/27/2022]
Abstract
Background: Plants and their derived natural compounds possess various biological and therapeutic properties, which turns them into an increasing topic of interest and research. Juniperus genus is diverse in species, with several traditional medicines reported, and rich in natural compounds with potential for development of new drugs. Methods: The research for this review were based in the Scopus and Web of Science databases using terms combining Juniperus, secondary metabolites names, and biological activities. This is not an exhaustive review of Juniperus compounds with biological activities, but rather a critical selection taking into account the following criteria: (i) studies involving the most recent methodologies for quantitative evaluation of biological activities; and (ii) the compounds with the highest number of studies published in the last four years. Results: From Juniperus species, several diterpenes, flavonoids, and one lignan were emphasized taking into account their level of activity against several targets. Antitumor activity is by far the most studied, being followed by antibacterial and antiviral activities. Deoxypodophyllotoxin and one dehydroabietic acid derivative appears to be the most promising lead compounds. Conclusions: This review demonstrates the Juniperus species value as a source of secondary metabolites with relevant pharmaceutical potential.
Collapse
Affiliation(s)
- Wilson R Tavares
- Faculty of Sciences and Technology, University of Azores, 9501-801 Ponta Delgada, Portugal.
| | - Ana M L Seca
- Department of Chemistry & QOPNA-Organic Chemistry, Natural Products and Food Stuffs, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal.
- cE3c-Centre for Ecology, Evolution and Environmental Changes/Azorean Biodiversity Group & Faculty of Sciences and Technology, University of Azores, Rua Mãe de Deus, 9501-321 Ponta Delgada, Portugal.
| |
Collapse
|
35
|
Nunes-Xavier CE, Mingo J, López JI, Pulido R. The role of protein tyrosine phosphatases in prostate cancer biology. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:102-113. [PMID: 30401533 DOI: 10.1016/j.bbamcr.2018.06.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/18/2018] [Accepted: 06/28/2018] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PCa) is the most frequent malignancy in the male population of Western countries. Although earlier detection and more active surveillance have improved survival, it is still a challenge how to treat advanced cases. Since androgen receptor (AR) and AR-related signaling pathways are fundamental in the growth of normal and neoplastic prostate cells, targeting androgen synthesis or AR activity constitutes the basis of the current hormonal therapies in PCa. However, resistance to these treatments develops, both by AR-dependent and -independent mechanisms. Thus, alternative therapeutic approaches should be developed to target more efficiently advanced disease. Protein tyrosine phosphatases (PTPs) are direct regulators of the protein- and residue-specific phosphotyrosine (pTyr) content of cells, and dysregulation of the cellular Tyr phosphorylation/dephosphorylation balance is a major driving event in cancer, including PCa. Here, we review the current knowledge on the role of classical PTPs in the growth, differentiation, and survival of epithelial prostate cells, and their potential as important players and therapeutic targets for modulation in PCa.
Collapse
Affiliation(s)
- Caroline E Nunes-Xavier
- Department of Tumor Biology, Institute of Cancer Research, Oslo University Hospital Radiumhospitalet, N-0310 Oslo, Norway; Biomarkers in Cancer Unit, Biocruces Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - Janire Mingo
- Biomarkers in Cancer Unit, Biocruces Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - José I López
- Biomarkers in Cancer Unit, Biocruces Health Research Institute, 48903 Barakaldo, Bizkaia, Spain; Department of Pathology, Cruces University Hospital, University of the Basque Country (UPV/EHU), 48903 Barakaldo, Bizkaia, Spain
| | - Rafael Pulido
- Biomarkers in Cancer Unit, Biocruces Health Research Institute, 48903 Barakaldo, Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, 48011 Bilbao, Spain.
| |
Collapse
|
36
|
Jung SN, Lim HS, Liu L, Chang JW, Lim YC, Rha KS, Koo BS. LAMB3 mediates metastatic tumor behavior in papillary thyroid cancer by regulating c-MET/Akt signals. Sci Rep 2018; 8:2718. [PMID: 29426928 PMCID: PMC5807368 DOI: 10.1038/s41598-018-21216-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 02/01/2018] [Indexed: 01/22/2023] Open
Abstract
Laminin subunit beta-3 (LAMB3) encodes one of the three subunits of LM-332, a protein of the extracellular matrix secreted by cultured human keratinocytes. While LAMB3 is involved in the invasive and metastatic abilities of several tumor types, including those found in the colon, pancreas, lung, cervix, stomach, and prostate, its mechanism of action in thyroid cancer has not been investigated previously. Our results show that LAMB3 is up-regulated in papillary thyroid cancer, and that its suppression reduces cell migration/invasion via down-regulation of epithelial‒mesenchymal transition-associated proteins (N-cadherin, vimentin, slug) and inhibition of matrix metalloproteinase 9. LAMB3 suppression also significantly decreases Akt phosphorylation and inhibits the transcription of c-MET, reducing its activation. These results suggest that LAMB3 leads to tumor invasion via Akt activation induced by the HGF/c-MET axis in papillary thyroid cancer cells. Our findings reveal a novel mechanism of action for LAMB3 in papillary thyroid cancer cells.
Collapse
Affiliation(s)
- Seung-Nam Jung
- Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Hyun Sil Lim
- Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Lihua Liu
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jae Won Chang
- Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Young Chang Lim
- Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Konkuk University College of Medicine, Seoul, Republic of Korea
| | - Ki Sang Rha
- Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Bon Seok Koo
- Department of Otolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
37
|
Jin Y, Kim YH, Park JY, Lee YJ, Oh HM, Choi SK, Han DC, Kwon BM. Methyllucidone inhibits STAT3 activity by regulating the expression of the protein tyrosine phosphatase MEG2 in DU145 prostate carcinoma cells. Bioorg Med Chem Lett 2018; 28:853-857. [PMID: 29456111 DOI: 10.1016/j.bmcl.2018.02.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 01/13/2023]
Abstract
During the search for signal transducer and activator of transcription 3 (STAT3) inhibitors from natural products, methyllucidone, isolated from Lindera species (Lauraceae), was identified as a STAT3 inhibitor. Methyllucidone inhibited STAT3 phosphorylation at tyrosine 705 in a dose- and time dependent manner in DU145 prostate cancer cells and suppressed IL-6-induced STAT3 phosphorylation at Tyr-705 in LNCaP cells. Methyllucidone decreased the expression levels of STAT3 target genes, such as cyclin D1, cyclin A, Bcl-2, Mcl-1, and survivin. Methyllucidone inhibited DU145 cell growth and induced apoptosis by arresting the cell cycle at G1 phase. Notably, knockdown of the MEG2 gene by small interfering RNA suppressed the ability of methyllucidone to inhibit STAT3 activation. Methyllucidone regulates STAT3 activity by modulating MEG2 expression, and our results suggest that this compound is a novel inhibitor of the STAT3 pathway and may be a useful lead molecule for the development of a therapeutic STAT3 inhibitor.
Collapse
Affiliation(s)
- Yena Jin
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Young Hwan Kim
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Jin Yong Park
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Yu-Jin Lee
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Hyun-Mi Oh
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Sung-Kyu Choi
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Dong Cho Han
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Korea University of Science and Technology, Daejeon, Republic of Korea.
| | - Byoung-Mog Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Korea University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
38
|
Pharmacotherapeutic potential of phytochemicals: Implications in cancer chemoprevention and future perspectives. Biomed Pharmacother 2018; 97:564-586. [DOI: 10.1016/j.biopha.2017.10.124] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/14/2017] [Accepted: 10/23/2017] [Indexed: 12/17/2022] Open
|
39
|
Zhang J, Li Z, Liu L, Wang Q, Li S, Chen D, Hu Z, Yu T, Ding J, Li J, Yao M, Huang S, Zhao Y, He X. Long noncoding RNA TSLNC8 is a tumor suppressor that inactivates the interleukin-6/STAT3 signaling pathway. Hepatology 2018; 67:171-187. [PMID: 28746790 DOI: 10.1002/hep.29405] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/26/2017] [Accepted: 07/23/2017] [Indexed: 02/06/2023]
Abstract
UNLABELLED Long noncoding RNAs can serve as oncogenes or tumor suppressors in human cancer; however, their biological functions and underlying mechanism in hepatocarcinogenesis are largely unknown. Here, we report a novel tumor suppressor long noncoding RNA on chromosome 8p12 (termed TSLNC8) that is frequently deleted and down-regulated in hepatocellular carcinoma (HCC) tissues. The loss of TSLNC8 is highly associated with the malignant features of HCC and serves as a prognostic indicator for HCC patients. TSLNC8 significantly suppresses the proliferation and metastasis of HCC cells in vitro and in vivo. TSLNC8 exerts its tumor suppressive activity by competitively interacting with transketolase and signal transducer and activator of transcription 3 (STAT3) and modulating the STAT3-Tyr705 and STAT3-Ser727 phosphorylation levels and STAT3 transcriptional activity, thus resulting in inactivation of the interleukin-6-STAT3 signaling pathway in HCC cells. CONCLUSION TSLNC8 is a promising prognostic predictor for patients with HCC, and the TSLNC8-transketolase-STAT3 axis is a potential therapeutic target for HCC treatment. (Hepatology 2018;67:171-187).
Collapse
Affiliation(s)
- Jiwei Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhe Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Longzi Liu
- Liver Cancer Institute, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qifeng Wang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shengli Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Di Chen
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhixiang Hu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tao Yu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Ding
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinjun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shenglin Huang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yingjun Zhao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xianghuo He
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Collaborative Innovation Center for Cancer Medicine, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
40
|
Cho HD, Lee JH, Moon KD, Park KH, Lee MK, Seo KI. Auriculasin-induced ROS causes prostate cancer cell death via induction of apoptosis. Food Chem Toxicol 2017; 111:660-669. [PMID: 29217266 DOI: 10.1016/j.fct.2017.12.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 01/11/2023]
Abstract
Recent studies have demonstrated that natural agents targeting the accumulation of reactive oxygen species (ROS) that selectively kill, leaving normal cells undamaged, can suppress prostate cancer. Here, we show that auriculasin, derived from Flemingia philippinensis, induces significant cell death and apoptosis via ROS generation in prostate cancer cells. Auriculasin treatment resulted in selective apoptotic cell death in LNCaP prostate cancer cells, characterized by DNA fragmentation, accumulation of sub-G1 cell population, cleavage of poly (ADP-ribose) polymerase (PARP), regulation of Bax/Bcl-2 ratio, increase of cytosolic apoptosis-inducing factor (AIF) and endonuclease G (EndoG), in addition to inhibiting tumor growth in a xenograft mouse model. Interestingly, auriculasin-induced apoptosis did not result in caspase-3, -8, and -9 activations. We found that auriculasin treatment decreased phosphorylation of AKT/mTOR/p70s6k in a dose- and time-dependent manner. Further, cellular ROS levels increased in LNCaP cells treated with auriculasin and blocking ROS accumulation with ROS scavengers resulted in inhibition of auriculasin-induced PARP cleavage, AIF increase, upregulation of Bax/Bcl-2 ratio, and decrease in AKT/mTOR phosphorylation. Taken together, these data suggest that auriculasin targets ROS-mediated caspase-independent pathways and suppresses PI3K/AKT/mTOR signaling, which leads to apoptosis and decreased tumor growth.
Collapse
Affiliation(s)
- Hyun-Dong Cho
- Department of Food Science and Technology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ju-Hye Lee
- Functional Food and Nutrition Division, Department of Agro-Food Resource, National Academy of Agricultural Science, Rural Development Administration, Wanju, 55365, Republic of Korea
| | - Kwang-Deog Moon
- Department of Food Science and Technology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ki-Hun Park
- Division of Applied Life Science (BK21 Plus), IALS, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Mi-Kyung Lee
- Department of Food and Nutrition, Sunchon National University, Suncheon, 57922, Republic of Korea
| | - Kwon-Il Seo
- Department of Biotechnology, Dong-A University, Busan, 49315, Republic of Korea.
| |
Collapse
|
41
|
Jin Y, Yoon YJ, Jeon YJ, Choi J, Lee YJ, Lee J, Choi S, Nash O, Han DC, Kwon BM. Geranylnaringenin (CG902) inhibits constitutive and inducible STAT3 activation through the activation of SHP-2 tyrosine phosphatase. Biochem Pharmacol 2017; 142:46-57. [DOI: 10.1016/j.bcp.2017.06.131] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/23/2017] [Indexed: 12/19/2022]
|
42
|
Lu H, Zhu H. Effect of siRNA-mediated gene silencing of transketolase on A549 lung cancer cells. Oncol Lett 2017; 14:5906-5912. [PMID: 29113225 PMCID: PMC5661397 DOI: 10.3892/ol.2017.6916] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 05/04/2017] [Indexed: 01/25/2023] Open
Abstract
The aim of the present study was to investigate the effects of transketolase (TKT) on cell proliferation, cell migration and interaction with other metabolism-associated genes in A549 lung cancer cells. A549 cells were transfected with three TKT-specific small interfering (si)RNAs, screened for the optimal transfection concentration, and sequenced with flow cytometry and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Cell viability was evaluated using Cell Counting Kit-8 (CCK-8), cell cycle was assessed by flow cytometric analysis. Cell migration was determined by scratch-wound and Transwell chamber assays. The changes in mRNA expression levels of glucose-6-phosphate dehydrogenase (G6PDH), transaldolase (TAL), sorbitol dehydrogenase (SORD), phosphoribosyl pyrophosphate synthetase 1 (PRPS1) and hexokinase 1 (HK1) were detected by RT-qPCR. siRNA-C at 50 nmol/l was selected for the subsequent experiments. Compared with the negative control, cell proliferation of the TKT-siRNA-C group was inhibited dramatically (CCK-8 24 h, 0.2984±0.0371 vs. 0.0952±0.0063; P<0.0001), the cell cycle was arrested at the G1/G0 cell cycle phase (58±2.0% vs. 70±2.5%; P=0.002), and cell migration ability was decreased [wound size, 254.71±34.96 vs. 349.12±37.43 µm (P=0.0001); Transwell migration, 250±47.8/field vs. 150±49.0/field (P<0.0001)]. The mRNA expression levels of G6PDH, TAL, SORD, PRPS1 and HK1 were downregulated in the TKT-siRNA-C group compared with the negative control. The present study revealed that synthetic TKT-siRNA can inhibit A549 cell viability and migration, which may be due to arrest of the cell cycle and downregulation of relevant metabolic enzymes.
Collapse
Affiliation(s)
- Huan Lu
- Department of Respiratory Medicine, Huadong Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Huili Zhu
- Department of Respiratory Medicine, Huadong Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
43
|
Berwin Singh SV, Kim J, Park H, Khang G, Lee D. Novel chemi-dynamic nanoparticles as a light-free photodynamic therapeutic system for cancer treatment. Macromol Res 2017. [DOI: 10.1007/s13233-017-5078-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
44
|
Qiu HY, Zhu X, Luo YL, Lin HY, Tang CY, Qi JL, Pang YJ, Yang RW, Lu GH, Wang XM, Yang YH. Identification of New Shikonin Derivatives as Antitumor Agents Targeting STAT3 SH2 Domain. Sci Rep 2017; 7:2863. [PMID: 28588262 PMCID: PMC5460289 DOI: 10.1038/s41598-017-02671-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/13/2017] [Indexed: 12/13/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is hyper-activated in diversiform human tumors and has been validated as an attractive therapeutic target. Current research showed that a natural product, shikonin, along with its synthetic analogues, is able to inhibit the activity of STAT3 potently. The potential space of shikonin in developing novel anti-cancer agents encouraged us to carry out the investigation of the probable binding mode with STAT3. From this foundation, we have designed new types of STAT3 SH2 inhibitors. Combined simulations were performed to filter for the lead compound, which was then substituted, synthesized and evaluated by a variety of bioassays. Among the entities, PMM-172 exhibited the best anti-proliferative activity against MDA-MB-231 cells with IC50 value 1.98 ± 0.49 μM. Besides, it was identified to decrease luciferase activity, induce cell apoptosis and reduce mitochondrial transmembrane potential in MDA-MB-231 cells. Also, PMM-172 inhibited constitutive/inducible STAT3 activation without affecting STAT1 and STAT5 in MDA-MB-231 cells, and had no effect in non-tumorigenic MCF-10A cells. Moreover, PMM-172 suppressed STAT3 nuclear localization and STAT3 downstream target genes expression. Overall, these results indicate that the antitumor activity of PMM-172 is at least partially due to inhibition of STAT3 in breast cancer cells.
Collapse
Affiliation(s)
- Han-Yue Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, NJU-NJFU Joint Institute of Plant Molecular Biology, Nanjing University, Nanjing, 210023, China.,Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037, China
| | - Xiang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, NJU-NJFU Joint Institute of Plant Molecular Biology, Nanjing University, Nanjing, 210023, China.,Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037, China
| | - Yue-Lin Luo
- State Key Laboratory of Pharmaceutical Biotechnology, NJU-NJFU Joint Institute of Plant Molecular Biology, Nanjing University, Nanjing, 210023, China.,Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037, China
| | - Hong-Yan Lin
- State Key Laboratory of Pharmaceutical Biotechnology, NJU-NJFU Joint Institute of Plant Molecular Biology, Nanjing University, Nanjing, 210023, China.,Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037, China
| | - Cheng-Yi Tang
- State Key Laboratory of Pharmaceutical Biotechnology, NJU-NJFU Joint Institute of Plant Molecular Biology, Nanjing University, Nanjing, 210023, China.,Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037, China
| | - Jin-Liang Qi
- State Key Laboratory of Pharmaceutical Biotechnology, NJU-NJFU Joint Institute of Plant Molecular Biology, Nanjing University, Nanjing, 210023, China.,Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037, China
| | - Yan-Jun Pang
- State Key Laboratory of Pharmaceutical Biotechnology, NJU-NJFU Joint Institute of Plant Molecular Biology, Nanjing University, Nanjing, 210023, China
| | - Rong-Wu Yang
- State Key Laboratory of Pharmaceutical Biotechnology, NJU-NJFU Joint Institute of Plant Molecular Biology, Nanjing University, Nanjing, 210023, China
| | - Gui-Hua Lu
- State Key Laboratory of Pharmaceutical Biotechnology, NJU-NJFU Joint Institute of Plant Molecular Biology, Nanjing University, Nanjing, 210023, China. .,Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037, China.
| | - Xiao-Ming Wang
- State Key Laboratory of Pharmaceutical Biotechnology, NJU-NJFU Joint Institute of Plant Molecular Biology, Nanjing University, Nanjing, 210023, China. .,Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037, China.
| | - Yong-Hua Yang
- State Key Laboratory of Pharmaceutical Biotechnology, NJU-NJFU Joint Institute of Plant Molecular Biology, Nanjing University, Nanjing, 210023, China. .,Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing, 210037, China.
| |
Collapse
|
45
|
STAT3 mediates multidrug resistance of Burkitt lymphoma cells by promoting antioxidant feedback. Biochem Biophys Res Commun 2017; 488:182-188. [PMID: 28483518 DOI: 10.1016/j.bbrc.2017.05.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 05/05/2017] [Indexed: 12/13/2022]
Abstract
Burkitt lymphoma (BL) is a highly aggressive B-cell neoplasm. Although BL is relatively sensitive to chemotherapy, some patients do not respond to initial therapy or relapse after standard therapy, which leads to poor prognosis. The mechanisms underlying BL chemoresistance remain poorly defined. Here, we report a mechanism for the relationship between the phosphorylation of STAT3 on Tyr705 and BL chemoresistance. In chemoresistant BL cells, STAT3 was activated and phosphorylated on Tyr705 in response to the generation of the reactive oxygen species (ROS), which induced Src Tyr416 phosphorylation after multi-chemotherapeutics treatment. As a transcription factor, the elevated phosphorylation level of STAT3Y705 increased the expression of GPx1 and SOD2, both of which protected cells against oxidative damage. Our findings revealed that the ROS-Src-STAT3-antioxidation pathway mediated negative feedback inhibition of apoptosis induced by chemotherapy. Thus, the phosphorylation of STAT3 on Tyr705 might be a target for the chemo-sensitization of BL.
Collapse
|
46
|
Discovery of monocarbonyl curcumin-BTP hybrids as STAT3 inhibitors for drug-sensitive and drug-resistant breast cancer therapy. Sci Rep 2017; 7:46352. [PMID: 28397855 PMCID: PMC5387716 DOI: 10.1038/srep46352] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/15/2017] [Indexed: 01/05/2023] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a well-known antitumor target. Exogenous ROS insult can lead to selective cytotoxicity against cancer cells. A combination of STAT3 inhibition and “oxidation therapy” may be a new strategy to address the multidrug-resistance issue due to their important roles in the survival and drug resistance of cancer cells. Here, a series of novel curcumin-BTP hybrids were designed and evaluated as STAT3 inhibitors with ROS production activity. Compound 6b exerted the best antitumor activity and selectivity for MCF-7 and MCF-7/DOX cells (IC50 = 0.52 μM and 0.40 μM, respectively), while its IC50 value for MCF-10A breast epithelial cells was 7.72 μM. Furthermore, compound 6b suppressed STAT3 phosphorylation, nuclear translocation and DNA-binding activity and the expression of STAT3 specific oncogenes. Increases in the level of IL-6-induced p-STAT3 were also inhibited by 6b without influencing IFN-γ-induced p-STAT1 expression. Additionally, 6b effectively promoted intracellular ROS accumulation, induced cancer cell apoptosis and cell cycle arrest, abolished the colony formation ability of breast cancer cells, and inhibited P-gp expression in MCF-7/DOX cells. Finally, 6b suppressed the growth of implanted human breast cancer in vivo. Our findings highlight that 6b may be a promising therapeutic agent for drug-sensitive and drug-resistant breast cancers.
Collapse
|
47
|
Han E, Kwon B, Yoo D, Kang C, Khang G, Lee D. Dual Stimuli-Activatable Oxidative Stress Amplifying Agent as a Hybrid Anticancer Prodrug. Bioconjug Chem 2017; 28:968-978. [DOI: 10.1021/acs.bioconjchem.6b00683] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Eunji Han
- Department of BIN Convergence Technology and ‡Department of Polymer·Nano Science
and Technology, Chonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 567-756, Republic of Korea
| | - Byeongsu Kwon
- Department of BIN Convergence Technology and ‡Department of Polymer·Nano Science
and Technology, Chonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 567-756, Republic of Korea
| | - Donghyuck Yoo
- Department of BIN Convergence Technology and ‡Department of Polymer·Nano Science
and Technology, Chonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 567-756, Republic of Korea
| | - Changsun Kang
- Department of BIN Convergence Technology and ‡Department of Polymer·Nano Science
and Technology, Chonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 567-756, Republic of Korea
| | - Gilson Khang
- Department of BIN Convergence Technology and ‡Department of Polymer·Nano Science
and Technology, Chonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 567-756, Republic of Korea
| | - Dongwon Lee
- Department of BIN Convergence Technology and ‡Department of Polymer·Nano Science
and Technology, Chonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 567-756, Republic of Korea
| |
Collapse
|
48
|
Characteristics, chemical compositions and biological activities of propolis from Al-Bahah, Saudi Arabia. Sci Rep 2017; 7:41453. [PMID: 28165013 PMCID: PMC5292687 DOI: 10.1038/srep41453] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/16/2016] [Indexed: 01/19/2023] Open
Abstract
Propolis has been used to treat several diseases since ancient times, and is an important source of bioactive natural compounds and drug derivatives. These properties have kept the interest of investigators around the world, leading to the investigation of the chemical and biological properties and application of propolis. In this report, the chemical constituents that are responsible for the anticancer activities of propolis were analyzed. The propolis was sourced from Al-Baha in the southern part of the Kingdom of Saudi Arabia. Standard protocols for chemical fractionation and bioactivity-guided chemical analysis were used to identify the bio-active ethyl acetate fraction. The extraction was performed in methanol and then analyzed by gas chromatography-mass spectrometry (GC-MS). The major compounds are triterpenoids, with a relative concentration of 74.0%; steroids, with a relative concentration of 9.8%; and diterpenoids, with a relative concentration of 7.9%. The biological activity was characterized using different approaches and cell-based assays. Propolis was found to inhibit the proliferation of cancer cells in a concentration-dependent manner through apoptosis. Immunofluorescence staining with anti-α-tubulin antibodies and cell cycle analysis indicated that tubulin and/or microtubules are the cellular targets of the L-acetate fraction. This study demonstrates the importance of Saudi propolis as anti-cancer drug candidates.
Collapse
|
49
|
Gersey ZC, Rodriguez GA, Barbarite E, Sanchez A, Walters WM, Ohaeto KC, Komotar RJ, Graham RM. Curcumin decreases malignant characteristics of glioblastoma stem cells via induction of reactive oxygen species. BMC Cancer 2017; 17:99. [PMID: 28160777 PMCID: PMC5292151 DOI: 10.1186/s12885-017-3058-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/11/2017] [Indexed: 01/14/2023] Open
Abstract
Background Glioblastoma Multiforme (GBM) is the most common and lethal form of primary brain tumor in adults. Following standard treatment of surgery, radiation and chemotherapy, patients are expected to survive 12–14 months. Theorized cause of disease recurrence in these patients is tumor cell repopulation through the proliferation of treatment-resistant cancer stem cells. Current research has revealed curcumin, the principal ingredient in turmeric, can modulate multiple signaling pathways important for cancer stem cell self-renewal and survival. Methods Following resection, tumor specimens were dissociated and glioblastoma stem cells (GSCs) were propagated in neurosphere media and characterized via immunocytochemistry. Cell viability was determined with MTS assay. GSC proliferation, sphere forming and colony forming assays were conducted through standard counting methods. Reactive oxygen species (ROS) production was examined using the fluorescent molecular probe CM-H2DCFA. Effects on cell signaling pathways were elucidated by western blot. Results We evaluate the effects of curcumin on patient-derived GSC lines. We demonstrate a curcumin-induced dose-dependent decrease in GSC viability with an approximate IC50 of 25 μM. Treatment with sub-toxic levels (2.5 μM) of curcumin significantly decreased GSC proliferation, sphere forming ability and colony forming potential. Curcumin induced ROS, promoted MAPK pathway activation, downregulated STAT3 activity and IAP family members. Inhibition of ROS with the antioxidant N-acetylcysteine reversed these effects indicating a ROS dependent mechanism. Conclusions Discoveries made in this investigation may lead to a non-toxic intervention designed to prevent recurrence in glioblastoma by targeting glioblastoma stem cells. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3058-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zachary C Gersey
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Gregor A Rodriguez
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Eric Barbarite
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Anthony Sanchez
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Winston M Walters
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kelechi C Ohaeto
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ricardo J Komotar
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Regina M Graham
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, Florida, USA. .,Department of Neurological Surgery, University of Miami Brain Tumor Initiative (UMBTI) Research Laboratory, Lois Pope LIFE Center, 2nd Floor, 1095 NW 14th Terrace, Miami, Florida, 33136, USA.
| |
Collapse
|
50
|
d,l-Sulforaphane Induces ROS-Dependent Apoptosis in Human Gliomablastoma Cells by Inactivating STAT3 Signaling Pathway. Int J Mol Sci 2017; 18:ijms18010072. [PMID: 28054986 PMCID: PMC5297707 DOI: 10.3390/ijms18010072] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/11/2016] [Accepted: 12/26/2016] [Indexed: 12/25/2022] Open
Abstract
d,l-Sulforaphane (SFN), a synthetic analogue of broccoli-derived isomer l-SFN, exerts cytotoxic effects on multiple tumor cell types through different mechanisms and is more potent than the l-isomer at inhibiting cancer growth. However, the means by which SFN impairs glioblastoma (GBM) cells remains poorly understood. In this study, we investigated the anti-cancer effect of SFN in GBM cells and determined the underlying molecular mechanisms. Cell viability assays, flow cytometry, immunofluorescence, and Western blot results revealed that SFN could induced apoptosis of GBM cells in a dose- and time-dependent manner, via up-regulation of caspase-3 and Bax, and down-regulation of Bcl-2. Mechanistically, SFN treatment led to increase the intracellular reactive oxygen species (ROS) level in GBM cells. Meanwhile, SFN also suppressed both constitutive and IL-6-induced phosphorylation of STAT3, and the activation of upstream JAK2 and Src tyrosine kinases, dose- and time-dependently. Moreover, blockage of ROS production by using the ROS inhibitor N-acetyl-l-cysteine totally reversed SFN-mediated down-regulation of JAK2/Src-STAT3 signaling activation and the subsequent effects on apoptosis by blocking the induction of apoptosis-related genes in GBM cells. Taken together, our data suggests that SFN induces apoptosis in GBM cells via ROS-dependent inactivation of STAT3 phosphorylation. These findings motivate further evaluation of SFN as a cancer chemopreventive agent in GBM treatment.
Collapse
|