1
|
Zigová M, Michalková R, Mojžiš J. Anticancer Potential of Indole Phytoalexins and Their Analogues. Molecules 2024; 29:2388. [PMID: 38792249 PMCID: PMC11124384 DOI: 10.3390/molecules29102388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Indole phytoalexins, found in economically significant Cruciferae family plants, are synthesized in response to pathogen attacks or stress, serving as crucial components of plant defense mechanisms against bacterial and fungal infections. Furthermore, recent research indicates that these compounds hold promise for improving human health, particularly in terms of potential anticancer effects that have been observed in various studies. Since our last comprehensive overview in 2016 focusing on the antiproliferative effects of these substances, brassinin and camalexin have been the most extensively studied. This review analyses the multifaceted pharmacological effects of brassinin and camalexin, highlighting their anticancer potential. In this article, we also provide an overview of the antiproliferative activity of new synthetic analogs of indole phytoalexins, which were synthesized and tested at our university with the aim of enhancing efficacy compared to the parent compound.
Collapse
Affiliation(s)
| | - Radka Michalková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia;
| | - Ján Mojžiš
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia;
| |
Collapse
|
2
|
Kwon HH, Ahn CH, Lee HJ, Sim DY, Park JE, Park SY, Kim B, Shim BS, Kim SH. The Apoptotic and Anti-Warburg Effects of Brassinin in PC-3 Cells via Reactive Oxygen Species Production and the Inhibition of the c-Myc, SIRT1, and β-Catenin Signaling Axis. Int J Mol Sci 2023; 24:13912. [PMID: 37762214 PMCID: PMC10530901 DOI: 10.3390/ijms241813912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Though Brassinin is known to have antiangiogenic, anti-inflammatory, and antitumor effects in colon, prostate, breast, lung, and liver cancers, the underlying antitumor mechanism of Brassinin is not fully understood so far. Hence, in the current study, the apoptotic mechanism of Brassinin was explored in prostate cancer. Herein, Brassinin significantly increased the cytotoxicity and reduced the expressions of pro-Poly ADP-ribose polymerase (PARP), pro-caspase 3, and B-cell lymphoma 2 (Bcl-2) in PC-3 cells compared to DU145 and LNCaP cells. Consistently, Brassinin reduced the number of colonies and increased the sub-G1 population and terminal deoxynucleotidyl transferase (TdT) dUTP Nick-End Labeling (TUNEL)-positive cells in the PC-3 cells. Of note, Brassinin suppressed the expressions of pyruvate kinase-M2 (PKM2), glucose transporter 1 (GLUT1), hexokinase 2 (HK2), and lactate dehydrogenase (LDH) as glycolytic proteins in the PC-3 cells. Furthermore, Brassinin significantly reduced the expressions of SIRT1, c-Myc, and β-catenin in the PC-3 cells and also disrupted the binding of SIRT1 with β-catenin, along with a protein-protein interaction (PPI) score of 0.879 and spearman's correlation coefficient of 0.47 being observed between SIRT1 and β-catenin. Of note, Brassinin significantly increased the reactive oxygen species (ROS) generation in the PC-3 cells. Conversely, ROS scavenger NAC reversed the ability of Brassinin to attenuate pro-PARP, pro-Caspase3, SIRT1, and β-catenin in the PC-3 cells. Taken together, these findings support evidence that Brassinin induces apoptosis via the ROS-mediated inhibition of SIRT1, c-Myc, β-catenin, and glycolysis proteins as a potent anticancer candidate.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (H.H.K.); (C.-H.A.); (H.-J.L.); (D.Y.S.); (J.E.P.); (S.-Y.P.); (B.K.); (B.-S.S.)
| |
Collapse
|
3
|
Yin Z, Lv Y, Deng L, Li G, Ou R, Chen L, Zhu Y, Zhong Q, Liu Z, Huang J, Wu H, Zhang Q, Fei J, Liu S. Targeting ABCB6 with nitidine chloride inhibits PI3K/AKT signaling pathway to promote ferroptosis in multiple myeloma. Free Radic Biol Med 2023; 203:86-101. [PMID: 37044150 DOI: 10.1016/j.freeradbiomed.2023.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/19/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023]
Abstract
Since multiple myeloma (MM) remains a cureless malignancy of plasma cells to date, it becomes imperative to develop novel drugs and therapeutic targets for MM. We screened a small molecule library comprising 3633 natural product drugs, which demonstrated that Nitidine Chloride (NC), an extract from traditional Chinese medicine Zanthoxylum nitidum. We used Surface Plasmon Resonance-High Performance Liquid Chromatography-Protein Mass Spectrometry (SPR-HPLC-MS), Cellular Thermal Shift Assay (CETSA), molecular docking, and SPR assay to identify the potential targets of NC, in which ABCB6 was the unique target of NC. The effects of ABCB6 on cellular proliferation and drug resistance were determined by CCK8, western blot, flow cytometry, site-mutation cells, transmission electron microscopy, immunohistochemistry staining and xenograft model in vitro and in vivo. NC induced MM cell death by promoting ferroptosis. ABCB6 is the direct target of NC. ABCB6 expression was increased in MM samples compared to normal controls, which was significantly associated with MM relapse and poor outcomes. VGSK was the inferred binding epitope of NC on the ABCB6 protein. In the ABCB6-mutated MM cells, NC did not display cancer resistance, implying the vital role of ABCB6 in NC's bioactivity. Moreover, the silencing of ABCB6 significantly inhibited MM cell growth. Mechanistically, the direct binding of NC to ABCB6 suppressed PI3K/AKT signaling pathway to promote ferroptosis. In conclusion, ABCB6 can be a potential therapeutic target and prognostic biomarker in MM, while NC can be considered a novel drug for MM treatment.
Collapse
Affiliation(s)
- Zhao Yin
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Yiwen Lv
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Li Deng
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Guangchao Li
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Ruiming Ou
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Lizhi Chen
- Department of Science and Education, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Yangmin Zhu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Qi Zhong
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Zhi Liu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Jing Huang
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Hong Wu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Qing Zhang
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Jia Fei
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, Guangzhou, 510632, China
| | - Shuang Liu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong Province, 510317, China.
| |
Collapse
|
4
|
Brassinin Induces Apoptosis, Autophagy, and Paraptosis via MAPK Signaling Pathway Activation in Chronic Myelogenous Leukemia Cells. BIOLOGY 2023; 12:biology12020307. [PMID: 36829581 PMCID: PMC9953140 DOI: 10.3390/biology12020307] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/31/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
Brassinin (BSN), a potent phytoalexin found in cruciferous vegetables, has been found to exhibit diverse anti-neoplastic effects on different cancers. However, the impact of BSN on chronic myelogenous leukemia (CML) cells and the possible mode of its actions have not been described earlier. We investigated the anti-cytotoxic effects of BSN on the KBM5, KCL22, K562, and LAMA84 CML cells and its underlying mechanisms of action in inducing programmed cell death. We noted that BSN could induce apoptosis, autophagy, and paraptosis in CML cells. BSN induced PARP cleavage, subG1 peak increase, and early apoptosis. The potential action of BSN on autophagy activation was confirmed by an LC3 expression and acridine orange assay. In addition, BSN induced paraptosis through increasing the reactive oxygen species (ROS) production, mitochondria damage, and endoplasmic reticulum (ER) stress. Moreover, BSN promoted the activation of the MAPK signaling pathway, and pharmacological inhibitors of this signaling pathway could alleviate all three forms of cell death induced by BSN. Our data indicated that BSN could initiate the activation of apoptosis, autophagy, and paraptosis through modulating the MAPK signaling pathway.
Collapse
|
5
|
Marianna B, Radka M, Martin K, Janka V, Jan M. Design, Synthesis and Antiproliferative Evaluation of Bis-Indole Derivatives with a Phenyl Linker: Focus on Autophagy. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010251. [PMID: 36615444 PMCID: PMC9822133 DOI: 10.3390/molecules28010251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022]
Abstract
This work deals with the study of the synthesis of new bis-indole analogues with a phenyl linker derived from indole phytoalexins. Synthesis of target bis-indole thiourea linked by a phenyl linker was achieved by the reaction of [1-(tert-butoxycarbonyl)indol-3-yl]methyl isothiocyanate with p-phenylenediamine. By replacing the sulfur of the thiocarbonyl group in bis-indole thiourea with oxygen using mesityl nitrile oxide, a bis-indole homodimer with a urea group was obtained. A cyclization protocol utilizing bis-indole thiourea and methyl bromoacetate was applied to synthesize a bis-indole homodimer with a thiazolidin-4-one moiety. Bis-indole homodimers derived from 1-methoxyspirobrassinol methyl ether were prepared by bromospirocyclization methodology. Among the synthesized analogues, compound 49 was selected for further study. To evaluate the mode of the mechanism of action, we used flow cytometry, Western blot, and spectroscopic analyses. Compound 49 significantly inhibited the proliferation of lung cancer cell line A549 with minimal effects on the non-cancer cells. We also demonstrated that compound 49 induced autophagy through the upregulation of Beclin-1, LC3A/B, Atg7 and AMPK and ULK1. Furthermore, chloroquine (CQ; an autophagy inhibitor) in combination with compound 49 decreased cell proliferation and induced G1 cell cycle arrest and apoptosis. Compound 49 also caused GSH depletion and significantly potentiated the antiproliferative effect of cis-platin.
Collapse
Affiliation(s)
- Budovska Marianna
- Department of Organic Chemistry, Institute of Chemistry, Faculty of Science, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Michalkova Radka
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Kello Martin
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Vaskova Janka
- Department of Medical and Clinical Biochemistry, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Mojzis Jan
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
- Correspondence:
| |
Collapse
|
6
|
Assessing the Mechanism of Action of “Fructus Ligustri Lucidi-Cuscutae Semen” in Prostate Cancer Treatment Using Network Pharmacology and Molecular Docking. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7543619. [PMID: 36330452 PMCID: PMC9626213 DOI: 10.1155/2022/7543619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/18/2022] [Accepted: 08/27/2022] [Indexed: 11/25/2022]
Abstract
Objective To explore the mechanism of action of “Fructus Ligustri Lucidi-Cuscutae Semen” in the treatment of prostate cancer using network pharmacology and molecular docking. Methods The active ingredients and targets of “Fructus Ligustri Lucidi-Cuscutae Semen” were obtained by searching the TCMSP and DrugBank databases. These were matched and corrected using the UniProt platform. A drug “active ingredient-target” network map was constructed using Cytoscape 3.8.0. Prostate cancer-related targets were acquired from GeneCards, Disgenet, DrugBank, and other databases. The protein-protein interaction (PPI) network between the drug and prostate cancer was constructed with BioGenet; the crossover network of the two targets was extracted derive the key targets of “Fructus Ligustri Lucidi-Cuscutae Semen” for prostate cancer treatment. We used the Metascape platform for GO and KEGG enrichment analysis of the key targets. AutoDockTools1.5.6 and PyMOL software were used to perform molecular docking. Results We obtained 13 active ingredients, 221 drug targets, 1511 prostate cancer targets (including 221 key targets), and 305 KEGG pathways from “Fructus Ligustri Lucidi-Cuscutae Semen.” Paclitaxel, quercetin, kaempferol, TP53, β-sitosterol, EGFR, and ESR1 in “Fructus Ligustri Lucidi-Cuscutae Semen” showed good docking activity. Conclusion “Fructus Ligustri Lucidi-Cuscutae Semen” is a valuable clinical guide for the treatment of prostate cancer with multicomponent, multitarget, and multipathway characteristics.
Collapse
|
7
|
FAM107A Inactivation Associated with Promoter Methylation Affects Prostate Cancer Progression through the FAK/PI3K/AKT Pathway. Cancers (Basel) 2022; 14:cancers14163915. [PMID: 36010909 PMCID: PMC9405870 DOI: 10.3390/cancers14163915] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 12/01/2022] Open
Abstract
Simple Summary Prostate cancer (PCa) is a common male malignancy. FAM107A, or actin-associated protein, is commonly downregulated in PCa and is associated with a poor patient prognosis. We investigated the role of FAM107A in PCa and found that downregulation of FAM107A expression was caused by hypermethylation of CpG islands, and DNA methyltransferase 1 (DNMT1) was involved in maintaining hypermethylation. Mechanistically, FAM107A regulated PCa cell growth through the FAK/PI3K/AKT signaling pathway. Therefore, FAM107A overexpression may represent a potential treatment for PCa, while therapies targeting epigenetic events that regulate FAM107A expression may also be an effective strategy for PCa treatment. Abstract Prostate cancer (PCa) is one of the most common cancers and is the second leading cause of mortality in men. Studies exploring novel therapeutic methods are urgently needed. FAM107A, a coding gene located in the short arm of chromosome3, is generally downregulated in PCa and is associated with a poor prognosis. However, the downregulation of FAM107A in PCa and the mechanism of its action remain challenging to determine. This investigation found that downregulation of FAM107A expression in PCa was caused by hypermethylation of CpG islands. Furthermore, DNA methyltransferase 1 (DNMT1) was involved in maintaining hypermethylation. Mechanistically, overexpression of FAM107A inhibits tumor cell proliferation, migration, invasion and promotes apoptosis through the FAK/PI3K/AKT signaling pathway, indicating that FAM107A may be a molecular brake of FAK/PI3K/AKT signaling, thus limiting the active state of the FAK/PI3K/AKT pathway. These findings will contribute to a better understanding of the effect of FAM107A in PCa, and FAM107A may represent a new therapeutic target for PCa.
Collapse
|
8
|
Brassinin Promotes the Degradation of Tie2 and FGFR1 in Endothelial Cells and Inhibits Triple-Negative Breast Cancer Angiogenesis. Cancers (Basel) 2022; 14:cancers14143540. [PMID: 35884601 PMCID: PMC9318525 DOI: 10.3390/cancers14143540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Brassinin is a natural compound enriched in several commonly consumed vegetables, such as broccoli and cabbages. It shows potent anti-cancer activity against several cancers. However, its effects on triple-negative breast cancer (TNBC), an aggressive subtype with limited treatment options, remain elusive so far. Therefore, we investigated the effects of brassinin on TNBC angiogenesis and growth. Our results demonstrate that brassinin inhibits TNBC growth preferentially through inhibiting the angiogenic activity of endothelial cells (ECs). Additional in-vitro analyses revealed that this effect may be mediated by brassinin-stimulated degradation of two pivotal angiogenesis-related receptors in ECs: Tie2 and fibroblast growth factor receptor 1. These findings provide novel insights into the cellular and molecular mechanisms underlying the anti-cancer activity of brassinin and indicate that this phytochemical may be a promising lead compound or drug candidate for TNBC treatment. Abstract Brassinin, a phytoalexin derived from cruciferous vegetables, has been reported to exhibit anti-cancer activity in multiple cancer types. However, its effects on triple-negative breast cancer (TNBC) development and the underlying mechanisms have not been elucidated so far. In this study, we demonstrated in vitro that brassinin preferentially reduces the viability of endothelial cells (ECs) when compared to other cell types of the tumor microenvironment, including TNBC cells, pericytes, and fibroblasts. Moreover, brassinin at non-cytotoxic doses significantly suppressed the proliferation, migration, tube formation, and spheroid sprouting of ECs. It also efficiently inhibited angiogenesis in an ex-vivo aortic ring assay and an in-vivo Matrigel plug assay. Daily intraperitoneal injection of brassinin significantly reduced tumor size, microvessel density, as well as the perfusion of tumor microvessels in a dorsal skinfold chamber model of TNBC. Mechanistic analyses showed that brassinin selectively stimulates the degradation of Tie2 and fibroblast growth factor receptor 1 in ECs, leading to the down-regulation of the AKT and extracellular signal-regulated kinase pathways. These findings demonstrate a preferential and potent anti-angiogenic activity of brassinin, which may be the main mechanism of its anti-tumor action. Accordingly, this phytochemical represents a promising candidate for the future anti-angiogenic treatment of TNBC.
Collapse
|
9
|
Budovská M, Krochtová K, Michalková R, Mojžiš J. Aminoanalogues of isobrassinin, erucalexin and isocyclobrassinin: Synthesis and evaluation of the antiproliferative and cytotoxic properties. Tetrahedron 2022. [DOI: 10.1016/j.tet.2022.132898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
10
|
Brassinin Enhances Apoptosis in Hepatic Carcinoma by Inducing Reactive Oxygen Species Production and Suppressing the JAK2/STAT3 Pathway. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12094733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Plants from the family Brassicaceae produce brassinin (BSN), which is an essential indole phytoalexin. BSN can kill certain types of cancer cells. Using hepatocarcinoma (HCC) cells, we examined the molecular mechanisms of BSN. We found that HCC cell growth was suppressed and apoptosis was induced by BSN via the downregulation of the JAK/STAT3 pathway. The cytoplasmic latent transcription factor STAT3, belonging to the STAT family, acted as both a signal transducer and an activator and was linked to tumor progression and decreased survival. BSN incubation caused HCC cells to produce reactive oxygen species (ROS). By activating caspase-9/-3 and PARP cleavage, Bcl-2 was reduced, and apoptosis was increased. BSN inhibited constitutive STAT3, JAK2, and Src phosphorylation. The JAK/STAT signaling cascade was confirmed by siRNA silencing STAT3 in HCC cells. BSN also suppressed apoptosis by Z-Val-Ala-Asp-Fluoromethylketone (Z-VAD-FMK), an apoptotic inhibitor. N-acetylcysteine (NAC) inhibited the production of ROS and diminished BSN-induced apoptosis. Our findings suggested that BSN has potential as a treatment for cancer.
Collapse
|
11
|
Annie-Mathew AS, Prem-Santhosh S, Jayasuriya R, Ganesh G, Ramkumar KM, Sarada DVL. The pivotal role of Nrf2 activators in adipocyte biology. Pharmacol Res 2021; 173:105853. [PMID: 34455076 DOI: 10.1016/j.phrs.2021.105853] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 01/07/2023]
Abstract
Adipose tissue is instrumental in maintaining metabolic homeostasis by regulating energy storage in the form of triglycerides. In the case of over-nutrition, adipocytes favorably regulate lipogenesis over lipolysis and accumulate excess triglycerides, resulting in increased adipose tissue mass. An abnormal increase in hypertrophic adipocytes is associated with chronic complications such as insulin resistance, obesity, diabetes, atherosclerosis and nonalcoholic fatty liver disease. Experimental studies indicate the occurrence of oxidative stress in the pathogenesis of obesity. A common underlying link between increasing adipose tissue mass and oxidative stress is the Nuclear Factor Erythroid 2-related factor 2 (Nrf2), Keap1-Nrf2-ARE signaling, which plays an indispensable role in metabolic homeostasis by regulating oxidative and inflammatory responses. Additionally, Nrf2 also activates CCAAT/enhancer-binding protein α, (C/EBP-α), C/EBP-β and peroxisome proliferator-activated receptor γ (PPARγ) the crucial pro-adipogenic factors that promote de novo adipogenesis. Hence, at the forefront of research is the quest for prospecting novel compounds to modulate Nrf2 activity in the context of adipogenesis and obesity. This review summarizes the molecular mechanism behind the activation of the Keap1-Nrf2-ARE signaling network and the role of Nrf2 activators in adipocyte pathophysiology.
Collapse
Affiliation(s)
- A S Annie-Mathew
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Subramanian Prem-Santhosh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Ravichandran Jayasuriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India; SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Goutham Ganesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India; SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India; SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| | - D V L Sarada
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
12
|
Kazemi T, Huang S, Avci NG, Akay YM, Akay M. Investigating the effects of chronic perinatal alcohol and combined nicotine and alcohol exposure on dopaminergic and non-dopaminergic neurons in the VTA. Sci Rep 2021; 11:8706. [PMID: 33888815 PMCID: PMC8062589 DOI: 10.1038/s41598-021-88221-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 04/06/2021] [Indexed: 02/02/2023] Open
Abstract
The ventral tegmental area (VTA) is the origin of dopaminergic neurons and the dopamine (DA) reward pathway. This pathway has been widely studied in addiction and drug reinforcement studies and is believed to be the central processing component of the reward circuit. In this study, we used a well-established rat model to expose mother dams to alcohol, nicotine-alcohol, and saline perinatally. DA and non-DA neurons collected from the VTA of the rat pups were used to study expression profiles of miRNAs and mRNAs. miRNA pathway interactions, putative miRNA-mRNA target pairs, and downstream modulated biological pathways were analyzed. In the DA neurons, 4607 genes were differentially upregulated and 4682 were differentially downregulated following nicotine-alcohol exposure. However, in the non-DA neurons, only 543 genes were differentially upregulated and 506 were differentially downregulated. Cell proliferation, differentiation, and survival pathways were enriched after the treatments. Specifically, in the PI3K/AKT signaling pathway, there were 41 miRNAs and 136 mRNAs differentially expressed in the DA neurons while only 16 miRNAs and 20 mRNAs were differentially expressed in the non-DA neurons after the nicotine-alcohol exposure. These results depicted that chronic nicotine and alcohol exposures during pregnancy differentially affect both miRNA and gene expression profiles more in DA than the non-DA neurons in the VTA. Understanding how the expression signatures representing specific neuronal subpopulations become enriched in the VTA after addictive substance administration helps us to identify how neuronal functions may be altered in the brain.
Collapse
Affiliation(s)
- Tina Kazemi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Shuyan Huang
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Naze G Avci
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Yasemin M Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Metin Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
13
|
Yang MH, Baek SH, Ha IJ, Um JY, Ahn KS. Brassinin enhances the anticancer actions of paclitaxel by targeting multiple signaling pathways in colorectal cancer cells. Phytother Res 2021; 35:3875-3885. [PMID: 33792984 DOI: 10.1002/ptr.7095] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/22/2022]
Abstract
Brassinin (BSN), a precursor of phytoalexins, extracted from Chinese cabbage has been reported to act as a promising anti-neoplastic agent. However, the effects of BSN on colon cancer cells and its underlying mechanisms have not been fully elucidated. This study aimed at investigating the anti-neoplastic impact of BSN and its possible synergistic effect with paclitaxel on colon cancer cells. The effect of BSN on Janus-activated kinases (JAKs)/signal transducer and activator of transcription 3 (STAT3) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathways and its downstream functions was deciphered using diverse assays in colon carcinoma cells. We found that BSN displayed significant cytotoxic effect and suppressed cell proliferation on colon carcinoma cells. Additionally, it was noted that BSN modulated oncogenic gene expression and induced apoptosis through down regulating multiple oncogenic signaling cascades such as JAKs/STAT3 and PI3K/Akt/mTOR simultaneously. Besides, BSN-paclitaxel combination significantly increased cytotoxicity and induced apoptosis synergistically as compared with individual treatment of both the agents. Overall, our findings indicate that BSN may be a novel candidate for anti-colon cancer targeted therapy.
Collapse
Affiliation(s)
- Min Hee Yang
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, South Korea
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Seung Ho Baek
- College of Korean Medicine, Dongguk University, Goyang-si, South Korea
| | - In Jin Ha
- Korean Medicine Clinical Trial Center (K-CTC), Korean Medicine Hospital, Kyung Hee University, Seoul, South Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Kwang Seok Ahn
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, South Korea
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
14
|
Design, synthesis and biological evaluation of 3,5-diaryl isoxazole derivatives as potential anticancer agents. Bioorg Med Chem Lett 2020; 30:127427. [DOI: 10.1016/j.bmcl.2020.127427] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 12/11/2022]
|
15
|
Jiang G, Su Z, Liang X, Huang Y, Lan Z, Jiang X. Long non-coding RNAs in prostate tumorigenesis and therapy (Review). Mol Clin Oncol 2020; 13:76. [PMID: 33005410 DOI: 10.3892/mco.2020.2146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 06/18/2020] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PCa) is one of the most frequently diagnosed malignancy. Although there have been many advances in PCa diagnosis and therapy, the concrete mechanism remains unknown. Long non-coding RNAs (lncRNAs) are novel biomarkers associated with PCa, and their dysregulated expression is closely associated with risk stratification, diagnosis and carcinogenesis. Accumulating evidence has suggested that lncRNAs play important roles in prostate tumorigenesis through relevant pathways, such as androgen receptor interaction and PI3K/Akt. The present review systematically summarized the potential clinical utility of lncRNAs and provided a novel guide for their function in PCa.
Collapse
Affiliation(s)
- Ganggang Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China.,Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Zhengming Su
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China.,Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Xue Liang
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Yiqiao Huang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Ziquan Lan
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| | - Xianhan Jiang
- Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China.,Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510700, P.R. China
| |
Collapse
|
16
|
Wang B, Li H, Wang X, Zhu X. The association of aberrant expression of NLRP3 and p-S6K1 in colorectal cancer. Pathol Res Pract 2019; 216:152737. [PMID: 31757663 DOI: 10.1016/j.prp.2019.152737] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/26/2019] [Accepted: 11/10/2019] [Indexed: 01/07/2023]
Abstract
Colorectal carcinoma (CRC) is one of the most common malignancies worldwide, and except for surgery, treatments effects are limited. Recently, NLRP3 has been reported as an activator in the tumorigenesis of CRC. However, the function and mechanism of NLRP3 in CRC remains elusive. In this study, we revealed that NLRP3 was elevated in CRC tissues and related to clinical factors, such as lymph node invasion and tumor-node-metastasis (TNM) stage. Moreover, NLRP3-positive patients had a poor prognosis. Furthermore, univariate and multivariate analysis revealed that NLRP3 expression was an independent prognostic factor for the survival of CRC patients. We investigated whether NLRP3 linked with the mTOR-S6K1 pathway. The expression of p-S6K1 was upregulated in CRC tissues and NLRP3 expression level was positively associated with the p-S6K1 level. Thus, targeting NLRP3 may be promising for targeted therapy of CRC, especially for mTORC1-targeted resistant patients.
Collapse
Affiliation(s)
- Bin Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hao Li
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xuchao Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xinguo Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
17
|
Olender J, Wang BD, Ching T, Garmire LX, Garofano K, Ji Y, Knox T, Latham P, Nguyen K, Rhim J, Lee NH. A Novel FGFR3 Splice Variant Preferentially Expressed in African American Prostate Cancer Drives Aggressive Phenotypes and Docetaxel Resistance. Mol Cancer Res 2019; 17:2115-2125. [PMID: 31266816 DOI: 10.1158/1541-7786.mcr-19-0415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/19/2019] [Accepted: 06/21/2019] [Indexed: 01/08/2023]
Abstract
Alternative splicing (AS) has been shown to participate in prostate cancer development and progression; however, a link between AS and prostate cancer health disparities has been largely unexplored. Here we report on the cloning of a novel splice variant of FGFR3 that is preferentially expressed in African American (AA) prostate cancer. This novel variant (FGFR3-S) omits exon 14, comprising 123 nucleotides that encode the activation loop in the intracellular split kinase domain. Ectopic overexpression of FGFR3-S in European American (EA) prostate cancer cell lines (PC-3 and LNCaP) led to enhanced receptor autophosphorylation and increased activation of the downstream signaling effectors AKT, STAT3, and ribosomal S6 compared with FGFR3-L (retains exon 14). The increased oncogenic signaling imparted by FGFR3-S was associated with a substantial gain in proliferative and antiapoptotic activities, as well as a modest but significant gain in cell motility. Moreover, the FGFR3-S-conferred proliferative and motility gains were highly resistant to the pan-FGFR small-molecule inhibitor dovitinib and the antiapoptotic gain was insensitive to the cytotoxic drug docetaxel, which stands in marked contrast with dovitinib- and docetaxel-sensitive FGFR3-L. In an in vivo xenograft model, mice injected with PC-3 cells overexpressing FGFR3-S exhibited significantly increased tumor growth and resistance to dovitinib treatment compared with cells overexpressing FGFR3-L. In agreement with our in vitro and in vivo findings, a high FGFR3-S/FGFR3-L expression ratio in prostate cancer specimens was associated with poor patient prognosis. IMPLICATIONS: This work identifies a novel FGFR3 splice variant and supports the hypothesis that differential AS participates in prostate cancer health disparities.
Collapse
Affiliation(s)
- Jacqueline Olender
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, GW Cancer Center, Washington, D. C
| | - Bi-Dar Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, Maryland
| | - Travers Ching
- Cancer Epidemiology Program, University of Hawaii, Honolulu, Hawaii
| | - Lana X Garmire
- Department of Computational Medicine and Bioinformatics, School of Medicine, University of Michigan, Ann Arbor, Michigan
| | - Kaitlin Garofano
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, GW Cancer Center, Washington, D. C
| | - Youngmi Ji
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - Tessa Knox
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, GW Cancer Center, Washington, D. C
| | - Patricia Latham
- Department of Pathology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Kenneth Nguyen
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, GW Cancer Center, Washington, D. C
| | - Johng Rhim
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of Health Sciences, Bethesda, Maryland
| | - Norman H Lee
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, GW Cancer Center, Washington, D. C.
| |
Collapse
|
18
|
Sun C, Gu Y, Chen G, Du Y. Bioinformatics Analysis of Stromal Molecular Signatures Associated with Breast and Prostate Cancer. J Comput Biol 2019; 26:1130-1139. [PMID: 31180245 DOI: 10.1089/cmb.2019.0045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
This study aimed to identify stromal molecular signatures associated with breast and prostate cancer. The microarray data GSE26910 was downloaded from Gene Expression Omnibus database, including six invasive breast tumor stroma, six matched normal controls, six invasive prostate tumor stroma, and six matched controls. The differentially expressed genes (DEGs) in invasive breast and prostate tumors stroma were, respectively, identified. Then common stromal genes (B_P.DEGs) were further screened. Protein-protein interaction (PPI) network was constructed and Gene Ontology analysis was performed. Besides, gene-chemical interactions were mapped in Comparative Toxicogenomics Database to screen the chemicals related to feature genes. The results showed that, in total, 16 B_P.DEGs were identified. Thereinto, only seven B_P.DEGs were mapped into PPI, and only four functional modules (adenylate cyclase activating polypeptide 1 (pituitary) receptor type I (ADCYAP1R1) module, aspartoacylase (ASPA) module, glutathione S-transferase mu 5 (GSTM5) module, and periplakin (PPL) module) were involved in important biological processes associated with cancer progression. In addition, the chemicals, such as dihydrotestosterone, apocarotenal, testosterone, and progesterone, were screened for the roles of feature genes in the progression of breast and prostate cancer. In conclusion, ADCYAP1R1, GSTM5, and PPL were stromal molecular signatures and might play a key role in the progression of breast and prostate cancer.
Collapse
Affiliation(s)
- Chao Sun
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifan Gu
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoqing Chen
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yibao Du
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Brassinin Represses Invasive Potential of Lung Carcinoma Cells through Deactivation of PI3K/Akt/mTOR Signaling Cascade. Molecules 2019; 24:molecules24081584. [PMID: 31013639 PMCID: PMC6514890 DOI: 10.3390/molecules24081584] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 04/19/2019] [Accepted: 04/19/2019] [Indexed: 12/22/2022] Open
Abstract
The epithelial–mesenchymal transition (EMT) is a phenomenon that facilitates epithelial cells to acquire invasive potential to induce the initiation the metastatic spread of tumor cells. Here, we determined if brassinin (BSN) can affect the EMT process and deciphered its anti-cancer effects. BSN attenuated the levels of EMT linked genes and suppressed transforming growth factor beta (TGF-β)-mediated regulation of diverse mesenchymal markers. Additionally, BSN did increase the expression of various epithelial marker proteins in lung cancer cells. TGF-β-induced morphological changes and induction of invasive ability of tumor cells was also found to be abrogated by BSN treatment. Finally, BSN not only suppressed constitutive, but also inducible phosphoinositide-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) phosphorylation in tumor cells.
Collapse
|
20
|
Wu YQ, Ju CL, Wang BJ, Wang RG. PABPC1L depletion inhibits proliferation and migration via blockage of AKT pathway in human colorectal cancer cells. Oncol Lett 2019; 17:3439-3445. [PMID: 30867782 PMCID: PMC6396114 DOI: 10.3892/ol.2019.9999] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 01/22/2019] [Indexed: 12/20/2022] Open
Abstract
Numerous studies have demonstrated that PABPC1 participates in the process of carcinogenesis and its function is inconsistent in different types of cancers. PABPC1-like (PABPC1L) is an important paralog of PABPC1 and few studies are available on the roles of PABPC1L in colorectal cancer (CRC) development. Hence, we explored the biological function and prognostic impact of PABPC1L in CRC. The mRNA expression of PABPC1L in CRC was determined based on the data obtained from The Cancer Genome Atlas (TCGA) database. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was utilized to determine the PABPC1L mRNA expression level in CRC HT-29 and LS-174T cell lines. Kaplan-Meier method and Cox proportional-hazards model were utilized to conduct the survival and prognosis analyses. HT-29 cells with silenced PABPC1L were constructed to explore the effect of PABPC1L on cell proliferation, invasion and migration capacities using cell counting kit-8 (CCK-8), clone formation, wound-healing and Transwell assays, respectively. To uncover the potential mechanisms of how PABPC1L influences CRC proliferation and migration, we analyzed the expression of AKT, p-AKT, PI3K, and p-PI3K in HT-29 cells using western blotting. Our results revealed that PABPC1L was overexpressed in CRC tissues compared with normal tissues based on the data obtained from TCGA database. Similarly, the mRNA expression of PABPC1L was higher in HT-29 and LS-174T cells than that in CCD-18Co cells. The expression of PABPC1L in CRC was found to be significantly related to age, pathologic stage, pathologic-node, pathologic-metastasis, and death. In univariate and multivariate analyses, pathologic-tumor and pathologic-metastasis were identified as independent prognostic factors for CRC. After PABPC1L depletion, cell proliferation rate, colony numbers, and the invasive and migratory capacity of HT-29 cells were all reduced. Western blot analysis showed that reduction of PABPC1L significantly inhibited p-AKT, and p-PI3K expression level in HT-29 cells. Collectively, our results suggested that PABPC1L is a potential novel candidate oncogene in CRC, and targeting PABPC1L may provide clinical utility in CRC.
Collapse
Affiliation(s)
- Yue-Qin Wu
- Department of Integration of Traditional Chinese Medicine and Western Medicine, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Chao-Long Ju
- Anorectal Department of Traditional Chinese Medicine, Central Hospital of Tongchuan Mining Bureau, Tongchuan, Shanxi 727000, P.R. China
| | - Bao-Juan Wang
- Department of Nephropathy, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300150, P.R. China
| | - Ruo-Gu Wang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, Shandong 250031, P.R. China
| |
Collapse
|
21
|
Bakar-Ates F, Ozkan E. The combined treatment of brassinin and imatinib synergistically downregulated the expression of MMP-9 in SW480 colon cancer cells. Phytother Res 2018; 33:397-402. [PMID: 30450754 DOI: 10.1002/ptr.6233] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/17/2018] [Accepted: 10/20/2018] [Indexed: 11/09/2022]
Abstract
In cancer treatment, which is a major cause of mortality today, combination studies with clinically used chemotherapeutics are becoming increasingly important as much as investigating the effects of novel natural compounds. In this context, phytoalexins constitute an important group due to their unique structure. Brassinin is an essential indole phytoalexin and is a biosynthetic precursor for other phytoalexins. The purpose of this study was to evaluate the anticancer effects of brassinin in combination with imatinib in SW480 cells. In the study, it was observed that brassinin-imatinib combination significantly increased cytotoxicity compared with the single treatment of both compounds and inhibited cell cycle at G0/G1 phase. Annexin V binding and fluorescence imaging assays showed that the combination of brasinin-imatinib induces apoptosis in a dose-dependent manner. Furthermore, the effect of brassinin on the activity of MMP-9 in SW480 cells was evaluated for the first time, and it was detected that MMP-9 activity was significantly reduced. The combination of brassinin-imatinib was found to inhibit MMP-9 activity as well as relative MMP-9 gene expression on a higher level compared with control and compounds alone. Our findings have revealed that the combination of brassinin-imatinib synergistically induces cytotoxicity and apoptosis in SW480 cells. The findings on MMP-9 downregulation have also revealed the anti-metastatic potential of treatment.
Collapse
Affiliation(s)
- Filiz Bakar-Ates
- Faculty of Pharmacy, Department of Biochemistry, Ankara University, Ankara, Turkey
| | - Erva Ozkan
- Faculty of Pharmacy, Department of Biochemistry, Ankara University, Ankara, Turkey
| |
Collapse
|
22
|
Xu X, Jiang J, Yao L, Ji B. Silencing the FOLR2 Gene Inhibits Cell Proliferation and Increases Apoptosis in the NCI-H1650 Non-Small Cell Lung Cancer Cell Line via Inhibition of AKT/Mammalian Target of Rapamycin (mTOR)/Ribosomal Protein S6 Kinase 1 (S6K1) Signaling. Med Sci Monit 2018; 24:8064-8073. [PMID: 30415267 PMCID: PMC6240851 DOI: 10.12659/msm.911384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background The FOLR2 gene encodes folate receptor-beta (FR-beta), which is expressed by tumor-associated macrophages. The effects of FOLR2 gene expression in non-small cell lung cancer (NSCLC) remains unknown. This study aimed to investigate the effects of FOLR2 gene expression and gene silencing in human NSCLC cell lines and normal human bronchial epithelial (HBE) cells in vitro. Material/Methods Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot were performed to detect the expression of the FOLR2 gene, cell cycle and apoptosis-associated genes in normal HBE cells and the NSCLC cell lines, A549, NCI-H1299, NCI-H1650, and NCI-H460. Using small interfering RNA (siRNA), or silencing RNA, FOLR2 gene silencing was performed for NCI-H1650 cells. Cell counting kit-8 (CCK-8) was used to measure cell viability. Cell cycle and apoptosis were determined using flow cytometry. Western blot evaluated the expression of Akt, mTOR, and S6K1 signaling. Results Expression of the FOLR2 gene was increased in NSCLC cells compared with normal HBE cells. Silencing of the expression of the FOLR2 gene in NCI-H1650 cells reduced cell viability, increased cell apoptosis, and arrested cells in the G1 phase of the cell cycle, decreased the expression of cyclin D1, upregulated expression of cell cycle inhibitors, p21 and p27, upregulated the expression of Bax/Bcl-2, and inhibited phosphorylation of AKT, mTOR, and S6K1. Conclusions Silencing of the FOLR2 gene inhibited phosphorylation of AKT, mTOR, and S6K1, inhibited cell proliferation and increased apoptosis in the NCI-H1650 human NSCLC cell line.
Collapse
Affiliation(s)
- Xiaohua Xu
- Department of Respiration, Quzhou Peoples' Hospital, Quzhou, Zhejiang, China (mainland)
| | - Jianyang Jiang
- Department of Respiration, Quzhou Peoples' Hospital, Quzhou, Zhejiang, China (mainland)
| | - Lijuan Yao
- Department of Urinary Surgery, Affiliated Huzhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medicine University, Huzhou, Zhejiang, China (mainland)
| | - Bing Ji
- Department of Medicine, Affiliated Huzhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medicine University, Huzhou, Zhejiang, China (mainland)
| |
Collapse
|
23
|
Qian B, Zhao L, Wang X, Xu J, Teng F, Gao L, Shen R. RETRACTED: miR-149 regulates the proliferation and apoptosis of cervical cancer cells by targeting GIT1. Biomed Pharmacother 2018; 105:1106-1116. [DOI: 10.1016/j.biopha.2018.06.075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/07/2018] [Accepted: 06/13/2018] [Indexed: 12/31/2022] Open
|
24
|
Liu X, Wang Z, Xie R, Tang P, Yuan Q. Design, synthesis and biological evaluation of novel carbamodithioates as anti-proliferative agents against human cancer cells. Eur J Med Chem 2018; 157:1526-1540. [PMID: 30282324 DOI: 10.1016/j.ejmech.2018.07.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/06/2018] [Accepted: 07/15/2018] [Indexed: 11/19/2022]
Abstract
A series of new carbamodithioates compounds has been successfully synthesized. All the carbamodithioate derivatives of SFE and SFA with benzenethiols (substituted or unsubstituted) exhibited, in general, higher percentages of inhibition than their parent compounds: SFE and SFA. A number of carbamodithioate derivatives with benzenethiols (substituted or unsubstituted) (1l, 1m, 1n, 1o, 1q, 1s, 2l, 2n, 2p, 2q, 2r and 2s) were investigated for in vitro anti-proliferative activities against five cancer cell lines: SMMC-7721, A549, A375, HCT 116 and Hela. The carbamodithioate compounds (1l, 1m, 1n, 1o, 1q and 1s) derived from SFE and the carbamodithioate compounds (2l, 2n, 2p, 2q, 2r and 2s) derived from SFA are more sensitive toward SMMC-7721and A549 cancer cells than toward other cancer cells in that their IC50 values are appreciably lower. Moreover, they exhibited stronger inhibitory activities than their parent compound SFE and SFE. Further investigation indicated that these carbamodithioate derivatives inhibited colony formation of SMMC-7721 and remarkably induced the G2/M or G0/G1 phase cell cycle arrest and apoptosis in SMMC-7721 cancer cells. More important, these carbamodithioate derivatives are stable in protic solvent media than their parent compounds. By virtue of the simplicity of the preparation of these carbamodithioate derivatives and their stability, compounds 1m and 2s could be the promising candidates for replacement for their parent SFE and SFA for cancer prevention agents.
Collapse
Affiliation(s)
- Xia Liu
- State Key Laboratory of Chemical Resource Engineering, Organic and Medicinal Chemistry Division, College of Life Science and Technology, Beijing University of Chemical Technology, 15 Beisanhuan East Road, Beijing, 100029, China
| | - Zhijun Wang
- State Key Laboratory of Chemical Resource Engineering, Organic and Medicinal Chemistry Division, College of Life Science and Technology, Beijing University of Chemical Technology, 15 Beisanhuan East Road, Beijing, 100029, China
| | - Rui Xie
- State Key Laboratory of Chemical Resource Engineering, Organic and Medicinal Chemistry Division, College of Life Science and Technology, Beijing University of Chemical Technology, 15 Beisanhuan East Road, Beijing, 100029, China
| | - Pingwah Tang
- State Key Laboratory of Chemical Resource Engineering, Organic and Medicinal Chemistry Division, College of Life Science and Technology, Beijing University of Chemical Technology, 15 Beisanhuan East Road, Beijing, 100029, China.
| | - Qipeng Yuan
- State Key Laboratory of Chemical Resource Engineering, Organic and Medicinal Chemistry Division, College of Life Science and Technology, Beijing University of Chemical Technology, 15 Beisanhuan East Road, Beijing, 100029, China.
| |
Collapse
|
25
|
Design, synthesis, and biological evaluation of thiomatrine derivatives as potential anticancer agents. Med Chem Res 2018. [DOI: 10.1007/s00044-018-2205-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
26
|
Synthesis, cytotoxic evaluation and target identification of thieno[2,3- d ]pyrimidine derivatives with a dithiocarbamate side chain at C2 position. Eur J Med Chem 2018; 154:324-340. [DOI: 10.1016/j.ejmech.2018.05.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/30/2018] [Accepted: 05/20/2018] [Indexed: 01/08/2023]
|
27
|
Zhu L, Hao J, Cheng M, Zhang C, Huo C, Liu Y, Du W, Zhang X. Hyperglycemia-induced Bcl-2/Bax-mediated apoptosis of Schwann cells via mTORC1/S6K1 inhibition in diabetic peripheral neuropathy. Exp Cell Res 2018; 367:186-195. [PMID: 29621478 DOI: 10.1016/j.yexcr.2018.03.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/27/2018] [Accepted: 03/26/2018] [Indexed: 12/31/2022]
Abstract
Schwann cell apoptosis is one of the characteristics of diabetic peripheral neuropathy (DPN). The mammalian target of rapamycin (mTOR) is a multifunctional signaling pathway that regulates cell apoptosis in various types of tissues and cells. To investigate whether the mTOR pathway is involved in cell apoptosis in the Schwann cells of DPN, diabetic mice and rat Schwann cells (RSC96) were chosen to detect phospho-mTOR (Ser 2448), phospho-S6K1 (Thr 389), phospho-4EBP1 (Thr 37/46), Bcl-2, Bax and cleaved caspase-3 by diverse pathological and biological techniques. The results showed that phospho-mTOR (Ser 2448) was decreased in the sciatic nerves of diabetic mice, concomitant with decreased Bcl-2, increased Bax, cleaved caspase-3 and cell apoptosis. In addition, high glucose treatment for 72 h caused a 35.95% decrease in the phospho-mTOR (Ser 2448)/mTOR ratio, a 65.50% decrease in the phospho-S6K1 (Thr 389)/S6K1 ratio, a 3.67-fold increase in the Bax/Bcl-2 ratio and a 1.47-fold increase in the cleaved caspase-3/caspase-3 ratio. Furthermore, mTORC1 inhibition, rather than mTORC2 inhibition, resulted in mitochondrial controlled apoptosis in RSC96 cells by silencing RAPTOR or RICTOR. Again, suppression of the mTORC1 pathway by a chemical inhibitor led to mitochondrial controlled apoptosis in cultured RSC96 cells in vitro. By contrast, activation of the mTORC1 pathway with MHY1485 prevented decreased phospho-S6K1 (Thr 389) levels caused by high glucose and cell apoptosis. Additionally, constitutive activation of S6K1 avoided high glucose-induced cell apoptosis in RSC96 cells. In summary, our findings suggest that activating mTORC1/S6K1 signaling in Schwann cells may be a promising strategy for the prevention and treatment of DPN.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China; Department of Electromyogram, Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Jun Hao
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China
| | - Meijuan Cheng
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China
| | - Cuihong Zhang
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China; Department of Radiation Oncology, Bethune International Peace Hospital, Shijiazhuang 050051, China
| | - Chunxiu Huo
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yaping Liu
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China
| | - Wei Du
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xianghong Zhang
- Department of Pathology, Hebei Medical University, Shijiazhuang 050017, China; Lab of Pathology, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
28
|
Resveratrol induces mitochondria-mediated, caspase-independent apoptosis in murine prostate cancer cells. Oncotarget 2017; 8:20895-20908. [PMID: 28157696 PMCID: PMC5400554 DOI: 10.18632/oncotarget.14947] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 01/10/2017] [Indexed: 12/29/2022] Open
Abstract
Found in the skins of red fruits, including grapes, resveratrol (RES) is a polyphenolic compound with cancer chemopreventive activity. Because of this activity, it has gained interest for scientific investigations. RES inhibits tumor growth and progression by targeting mitochondria-dependent or -independent pathways. However, further investigations are needed to explore the underlying mechanisms. The present study is focused on examining the role of RES-induced, mitochondria-mediated, caspase-independent apoptosis of prostate cancer cells, namely transgenic adenocarcinoma of mouse prostate (TRAMP) cells. These cells were exposed to RES for various times, and cell killing, cell morphology, mitochondrial membrane potential (Δψm), expression of Bax and Bcl2 proteins, the role of caspase-3, and DNA fragmentation were analyzed. TRAMP cells exposed to RES showed decreased cell viability, altered cell morphology, and disrupted Δψm, which led to aberrant expression of Bax and Bcl2 proteins. Furthermore, since the caspase-3 inhibitor, z-VAD-fmk (benzyloxycarbonyl-valine-alanine-aspartic acid-fluoromethyl ketone), had no appreciable impact on RES-induced cell killing, the killing was evidently caspase-independent. In addition, RES treatment of TRAMP-C1, TRAMP-C2, and TRAMP-C3 cells caused an appreciable breakage of genomic DNA into low-molecular-weight fragments. These findings show that, in inhibition of proliferation of TRAMP cells, RES induces mitochondria-mediated, caspase-independent apoptosis. Therefore, RES may be utilized as a therapeutic agent to control the proliferation and growth of cancer cells.
Collapse
|
29
|
Ramu A, Kathiresan S, Ali Ahmed B. Gramine inhibits angiogenesis and induces apoptosis via modulation of TGF-β signalling in 7,12 dimethylbenz[a]anthracene (DMBA) induced hamster buccal pouch carcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 33:69-76. [PMID: 28887922 DOI: 10.1016/j.phymed.2017.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 04/21/2017] [Accepted: 05/12/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Transforming growth factor-β (TGF-β) and its receptors are considered as a novel target in cancer chemotherapy. Gramine, an indole alkaloid, possesses various pharmacological properties including antiproliferative and anticancer. However, the anti-angiogenic property remains unexplored. PURPOSE The present study was designed to evaluate the anti-angiogenic and apoptosis induction properties of gramine through inhibiting TGF-β on DMBA induced oral squamous cell carcinoma (OSCC) in the hamster buccal pouch (HBP). METHODS The effects of gramine on TGF-β signalling in DMBA induced carcinogenic events such as angiogenesis and apoptosis were analysed by studying the mRNA expression using RT-PCR, protein expression by western blot and histopathological analysis using haematoxylin and eosin (H & E) staining. RESULTS Gramine significantly inhibited phosphorylation and nuclear translocation of Smad2 and Smad4 by blocking activity of the TGFβ-RII, RI and activation of inhibitory Smad7. Gramine inhibited angiogenic markers such as MMP-2, MMP-9, HIF-1α, VEGF, and VEGF-R2 as well as increased TIMP-2 expression. Furthermore, gramine induced apoptosis in DMBA induced tumour bearing animals by up regulating the pro apoptotic proteins Bax, cytochrome C, apaf-1, caspase-9 caspase-3 and PARP. CONCLUSION In this study, we clearly demonstrated that gramine treatment diminishes angiogenesis and induces apoptosis in hamster buccal pouch (HBP) carcinogenesis by modulating TGF-β signals.
Collapse
Affiliation(s)
- Arunkumar Ramu
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalai Nagar 608002, Tamil Nadu, India
| | - Suresh Kathiresan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalai Nagar 608002, Tamil Nadu, India.
| | - Bakrudeen Ali Ahmed
- Faculty of Applied Science, Ton Duc Thang University, Ho Chi Minh, Vietnam; University of Malaya, Institute of Biological Sciences, Faculty of Science, Kuala Lumpur, Malaysia
| |
Collapse
|
30
|
Antiproliferative Effect of Indole Phytoalexins. Molecules 2016; 21:molecules21121626. [PMID: 27898039 PMCID: PMC6274154 DOI: 10.3390/molecules21121626] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/15/2016] [Accepted: 11/22/2016] [Indexed: 12/20/2022] Open
Abstract
Indole phytoalexins from crucifers have been shown to exhibit significant anti-cancer, chemopreventive, and antiproliferative activity. Phytoalexins are natural low molecular antimicrobial compounds that are synthesized and accumulated in plants after their exposure to pathogenic microorganisms. Most interestingly, crucifers appear to be the only plant family producing sulfur-containing indole phytoalexins. The mechanisms underlying its anti-cancer properties are unknown. Isolation from cruciferous plants does not provide sufficient quantities of indole phytoalexins and, for biological screening, they are usually obtainable through synthesis. Understanding the molecular mechanism of the action of these substances and their structure-activity relationships is quite important in the development of new analogs with a more favorable profile of biological activities. In this review, we present the key features of indole phytoalexins, mainly their antiproliferative ativities.
Collapse
|
31
|
Lovely CB, Fernandes Y, Eberhart JK. Fishing for Fetal Alcohol Spectrum Disorders: Zebrafish as a Model for Ethanol Teratogenesis. Zebrafish 2016; 13:391-8. [PMID: 27186793 PMCID: PMC5035362 DOI: 10.1089/zeb.2016.1270] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Fetal Alcohol Spectrum Disorders (FASD) describes a wide array of ethanol-induced developmental defects, including craniofacial dysmorphology and cognitive impairments. It affects ∼1 in 100 children born in the United States each year. Due to the pleiotropic effects of ethanol, animal models have proven critical in characterizing the mechanisms of ethanol teratogenesis. In this review, we focus on the utility of zebrafish in characterizing ethanol-induced developmental defects. A growing number of laboratories have focused on using zebrafish to examine ethanol-induced defects in craniofacial, cardiac, ocular, and neural development, as well as cognitive and behavioral impairments. Growing evidence supports that genetic predisposition plays a role in these ethanol-induced defects, yet little is understood about these gene-ethanol interactions. With a high degree of genetic amenability, zebrafish is at the forefront of identifying and characterizing the gene-ethanol interactions that underlie FASD. Because of the conservation of gene function between zebrafish and humans, these studies will directly translate to studies of candidate genes in human populations and allow for better diagnosis and treatment of FASD.
Collapse
Affiliation(s)
| | - Yohaan Fernandes
- Molecular Biosciences, University of Texas at Austin , Austin, Texas
| | - Johann K Eberhart
- Molecular Biosciences, University of Texas at Austin , Austin, Texas
| |
Collapse
|
32
|
Brassinin inhibits STAT3 signaling pathway through modulation of PIAS-3 and SOCS-3 expression and sensitizes human lung cancer xenograft in nude mice to paclitaxel. Oncotarget 2016; 6:6386-405. [PMID: 25788267 PMCID: PMC4467444 DOI: 10.18632/oncotarget.3443] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/21/2015] [Indexed: 12/14/2022] Open
Abstract
Persistent phosphorylation of signal transducers and activators of transcription 3 (STAT3) is frequently observed in tumor cells. We found that brassinin (BSN) suppressed both constitutive and IL-6-inducible STAT3 activation in lung cancer cells. Moreover, BSN induced PIAS-3 protein and mRNA, whereas the expression of SOCS-3 was reduced. Knockdown of PIAS-3 by small interfering RNA prevented inhibition of STAT3 and cytotoxicity by BSN. Overexpression of SOCS-3 in BSN-treated cells increased STAT3 phosphorylation and cell viability. BSN down-regulated STAT3-regulated gene products, inhibited proliferation, invasion, as well as induced apoptosis. Most importantly, when administered intraperitoneally, combination of BSN and paclitaxel significantly decreased the tumor development in a xenograft lung cancer mouse model associated with down-modulation of phospho-STAT3, Ki-67 and CD31. We suggest that BSN inhibits STAT3 signaling through modulation of PIAS-3 and SOCS-3, thereby attenuating tumor growth and increasing sensitivity to paclitaxel.
Collapse
|
33
|
Kim SM, Oh EY, Lee JH, Nam D, Lee SG, Lee J, Kim SH, Shim BS, Ahn KS. Brassinin Combined with Capsaicin Enhances Apoptotic and Anti-metastatic Effects in PC-3 Human Prostate Cancer Cells. Phytother Res 2015; 29:1828-36. [DOI: 10.1002/ptr.5478] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 08/14/2015] [Accepted: 08/29/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Sung-Moo Kim
- College of Korean Medicine; Kyung Hee University; 1 Hoegidong Dongdaemungu Seoul 130-701 Republic of Korea
| | - Eun Young Oh
- College of Korean Medicine; Kyung Hee University; 1 Hoegidong Dongdaemungu Seoul 130-701 Republic of Korea
| | - Jong Hyun Lee
- College of Korean Medicine; Kyung Hee University; 1 Hoegidong Dongdaemungu Seoul 130-701 Republic of Korea
| | - Dongwoo Nam
- College of Korean Medicine; Kyung Hee University; 1 Hoegidong Dongdaemungu Seoul 130-701 Republic of Korea
| | - Seok Geun Lee
- College of Korean Medicine; Kyung Hee University; 1 Hoegidong Dongdaemungu Seoul 130-701 Republic of Korea
| | - Junhee Lee
- College of Korean Medicine; Kyung Hee University; 1 Hoegidong Dongdaemungu Seoul 130-701 Republic of Korea
| | - Sung-Hoon Kim
- College of Korean Medicine; Kyung Hee University; 1 Hoegidong Dongdaemungu Seoul 130-701 Republic of Korea
| | - Bum Sang Shim
- College of Korean Medicine; Kyung Hee University; 1 Hoegidong Dongdaemungu Seoul 130-701 Republic of Korea
| | - Kwang Seok Ahn
- College of Korean Medicine; Kyung Hee University; 1 Hoegidong Dongdaemungu Seoul 130-701 Republic of Korea
| |
Collapse
|
34
|
Two cytochromes P450 catalyze S-heterocyclizations in cabbage phytoalexin biosynthesis. Nat Chem Biol 2015; 11:837-9. [PMID: 26389737 PMCID: PMC4731101 DOI: 10.1038/nchembio.1914] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 08/19/2015] [Indexed: 12/15/2022]
Abstract
Phytoalexins are abundant in edible crucifers and have important biological activities, yet no dedicated gene for their biosynthesis is known. Here, we report two new cytochromes P450 from Brassica rapa (Chinese cabbage) that catalyze unprecedented S-heterocyclizations in cyclobrassinin and spirobrassinin biosynthesis. Our results reveal the first genetic and biochemical insights into the biosynthesis of a prominent pair of dietary metabolites, and have implications for pathway discovery across >20 recently sequenced crucifers.
Collapse
|
35
|
Zhang X, Shi H, Tang H, Fang Z, Wang J, Cui S. miR-218 inhibits the invasion and migration of colon cancer cells by targeting the PI3K/Akt/mTOR signaling pathway. Int J Mol Med 2015; 35:1301-8. [PMID: 25760926 DOI: 10.3892/ijmm.2015.2126] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/26/2015] [Indexed: 11/06/2022] Open
Abstract
Colon cancer is one of the most common and lethal malignancies worldwide. Despite major advances in the treatment of colon cancer, the prognosis remains very poor. Thus, novel and effective therapies for colon cancer are urgently needed. In the present study, the expression status of miR-218 and the role of the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway were investigated in colon cancer samples. Firstly, we observed that miR-218 expression was significantly reduced, while PI3K/Akt/mTOR pathway activity was enhanced. The overexpression of miR-218 suppressed the proliferation, migration and invasion of LoVo colon cancer cells, whereas the inhibition of miR-218 promoted these processes. Furthermore, the PI3K/Akt/mTOR signaling pathway was identified as a direct target of miR-218. The upregulation of miR-218 inhibited the activation of the PI3K/Akt/mTOR signaling pathway, as well as the expression of matrix metalloproteinase (MMP)9. The downregulation of miR-218 activated the PI3K/Akt/mTOR signaling pathway and promoted MMP9 expression. Taken together, our results demonstrate that miR-218 suppresses the proliferation, migration and invasion of LoVo colon cancer cells by targeting the PI3K/Akt/mTOR signaling pathway and MMP9. Our data indicate that miR-218 is a potential target in the treatment of colon cancer.
Collapse
Affiliation(s)
- Xiangliang Zhang
- Department of Abdominal Surgery (Section 2), The Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Huijuan Shi
- Department of Pathology, The First Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hongsheng Tang
- Department of Abdominal Surgery (Section 2), The Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Zhiyuan Fang
- Department of Abdominal Surgery (Section 2), The Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Jiping Wang
- Department of Surgery, Brigham and Women's Hospital affiliated to Harvard Medical School, Boston, MA 02115, USA
| | - Shuzhong Cui
- Department of Abdominal Surgery (Section 2), The Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| |
Collapse
|
36
|
Murthy KNC, Jayaprakasha GK, Patil BS. Cytotoxicity of obacunone and obacunone glucoside in human prostate cancer cells involves Akt-mediated programmed cell death. Toxicology 2015; 329:88-97. [PMID: 25592883 DOI: 10.1016/j.tox.2015.01.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 12/24/2014] [Accepted: 01/10/2015] [Indexed: 10/24/2022]
Abstract
Obacunone and obacunone glucoside (OG) are naturally occurring triterpenoids commonly found in citrus and other plants of the Rutaceae family. The current study reports the mechanism of cytotoxicity of citrus-derived obacunone and OG on human androgen-dependent prostate cancer LNCaP cells. Both limonoids exhibited time- and dose-dependent inhibition of cell proliferation, with more than 60% inhibition of cell viability at 100 μM, after 24 and 48 h. Analysis of fragmentation of DNA, activity of caspase-3, and cytosolic cytochrome-c in the cells treated with limonoids provided evidence for activation of programmed cell death by limonoids. Treatment of LNCaP cells with obacunone and OG resulted in dose-dependent changes in expression of proteins responsible for the induction of programmed cell death through the intrinsic pathway and down-regulation of Akt, a key molecule in cell signaling pathways. In addition, obacunone and OG also negatively regulated an inflammation-associated transcription factor, androgen receptor, and prostate-specific antigen, and activated proteins related to the cell cycle, confirming the ability of limonoids to induce cytotoxicity through multiple pathways. The results of this study provided, for the first time, an evidence of the cytotoxicity of obacunone and OG in androgen-dependent human prostate cancer cells.
Collapse
Affiliation(s)
- Kotamballi N Chidambara Murthy
- Vegetable and Fruit Improvement Center, Department of Horticultural Sciences, Texas A&M University, College Station, TX 77845-2119, United States
| | | | - Bhimanagouda S Patil
- Vegetable and Fruit Improvement Center, Department of Horticultural Sciences, Texas A&M University, College Station, TX 77845-2119, United States.
| |
Collapse
|
37
|
Lee J, Lee S, Kim SL, Choi JW, Seo JY, Choi DJ, Park YI. Corn silk maysin induces apoptotic cell death in PC-3 prostate cancer cells via mitochondria-dependent pathway. Life Sci 2014; 119:47-55. [PMID: 25445226 DOI: 10.1016/j.lfs.2014.10.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 10/02/2014] [Accepted: 10/20/2014] [Indexed: 12/18/2022]
Abstract
AIMS Despite recent advances in prostate cancer diagnostics and therapeutics, the overall survival rate still remains low. This study was aimed to assess potential anti-cancer activity of maysin, a major flavonoid of corn silk (CS, Zea mays L.), in androgen-independent human prostate cancer cells (PC-3). MAIN METHODS Maysin was isolated from CS of Kwangpyeongok, a Korean hybrid corn, via methanol extraction and preparative C18 reverse phase column chromatography. Maysin cytotoxicity was determined by either monitoring cell viability in various cancer cell lines by MTT assay or morphological changes. Apoptotic cell death was assessed by annexin V-FITC/PI double staining, depolarization of mitochondrial membrane potential (MMP), expression levels of Bcl-2 and pro-caspase-3 and by terminal transferase mediated dUTP-fluorescein nick end labeling (TUNEL) staining. Underlying mechanism in maysin-induced apoptosis of PC-3 cells was explored by evaluating its effects on Akt and ERK pathway. KEY FINDINGS Maysin dose-dependently reduced the PC-3 cell viability, with an 87% reduction at 200 μg/ml. Maysin treatment significantly induced apoptotic cell death, DNA fragmentation, depolarization of MMP, and reduction in Bcl-2 and pro-caspase-3 expression levels. Maysin also significantly attenuated phosphorylation of Akt and ERK. A combined treatment with maysin and other known anti-cancer agents, including 5-FU, etoposide, cisplatin, or camptothecin, synergistically enhanced PC-3 cell death. SIGNIFICANCE These results suggested for the first time that maysin inhibits the PC-3 cancer cell growth via stimulation of mitochondria-dependent apoptotic cell death and may have a strong therapeutic potential for the treatment of either chemo-resistant or androgen-independent human prostate cancer.
Collapse
Affiliation(s)
- Jisun Lee
- Department of Biotechnology, The CUK Agromedical Research Center, The Catholic University of Korea, Bucheon, Gyeonggi-do 420-743, Republic of Korea
| | - Seul Lee
- Department of Biotechnology, The CUK Agromedical Research Center, The Catholic University of Korea, Bucheon, Gyeonggi-do 420-743, Republic of Korea
| | - Sun-Lim Kim
- National Institute of Crop Science, RDA, Suwon, Gyeonggi-do 441-857, Republic of Korea
| | - Ji Won Choi
- Department of Biotechnology, The CUK Agromedical Research Center, The Catholic University of Korea, Bucheon, Gyeonggi-do 420-743, Republic of Korea
| | - Jeong Yeon Seo
- Department of Biotechnology, The CUK Agromedical Research Center, The Catholic University of Korea, Bucheon, Gyeonggi-do 420-743, Republic of Korea
| | - Doo Jin Choi
- Department of Biotechnology, The CUK Agromedical Research Center, The Catholic University of Korea, Bucheon, Gyeonggi-do 420-743, Republic of Korea
| | - Yong Il Park
- Department of Biotechnology, The CUK Agromedical Research Center, The Catholic University of Korea, Bucheon, Gyeonggi-do 420-743, Republic of Korea.
| |
Collapse
|
38
|
Retinoblastoma binding protein 2 (RBP2) promotes HIF-1α-VEGF-induced angiogenesis of non-small cell lung cancer via the Akt pathway. PLoS One 2014; 9:e106032. [PMID: 25162518 PMCID: PMC4146555 DOI: 10.1371/journal.pone.0106032] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 07/27/2014] [Indexed: 01/25/2023] Open
Abstract
Background Pathological angiogenesis plays an essential role in tumor aggressiveness and leads to unfavorable prognosis. The aim of this study is to detect the potential role of Retinoblastoma binding protein 2 (RBP2) in the tumor angiogenesis of non-small cell lung cancer (NSCLC). Methods Immunohistochemical staining was used to detect the expression of RBP2, hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF) and CD34. Two pairs of siRNA sequences and pcDNA3-HA-RBP2 were used to down-regulate and up-regulate RBP2 expression in H1975 and SK-MES-1 cells. An endothelial cell tube formation assay, VEGF enzyme-linked immunosorbent assay, real-time PCR and western blotting were performed to detect the potential mechanisms mediated by RBP2 in tumor angiogenesis. Results Of the 102 stage I NSCLC specimens analyzed, high RBP2 protein expression is closely associated with tumor size (P = 0.030), high HIF-1α expression (P = 0.028), high VEGF expression (P = 0.048), increased tumor angiogenesis (P = 0.033) and poor prognosis (P = 0.037); high MVD was associated with high HIF-1α expression (P = 0.034), high VEGF expression (P = 0.001) and poor prognosis (P = 0.040). Multivariate analysis indicated that RBP2 had an independent influence on the survival of patients with stage I NSCLC (P = 0.044). By modulating the expression of RBP2, our findings suggested that RBP2 protein depletion decreased HUVECs tube formation by down-regulating VEGF in a conditioned medium. RBP2 stimulated the up-regulation of VEGF, which was dependent on HIF-1α, and activated the HIF-1α via phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. Moreover, VEGF increased the activation of Akt regulated by RBP2. Conclusions The RBP2 protein may stimulate HIF-1α expression via the activation of the PI3K/Akt signaling pathway under normoxia and then stimulate VEGF expression. These findings indicate that RBP2 may play a critical role in tumor angiogenesis and serve as an attractive therapeutic target against tumor aggressiveness for early-stage NSCLC patients.
Collapse
|
39
|
ROS-dependent antiproliferative effect of brassinin derivative homobrassinin in human colorectal cancer Caco2 cells. Molecules 2014; 19:10877-97. [PMID: 25068784 PMCID: PMC6271261 DOI: 10.3390/molecules190810877] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/17/2014] [Accepted: 07/18/2014] [Indexed: 12/20/2022] Open
Abstract
This study was designed to examine the in vitro antiproliferative effect of brassinin and its derivatives on human cancer cell lines. Among seven tested compounds, homobrassinin (K1; N-[2-(indol-3-yl)ethyl]-S-methyldithiocarbamate) exhibited the most potent activity with IC50 = 8.0 μM in human colorectal Caco2 cells and was selected for further studies. The flow cytometric analysis revealed a K1-induced increase in the G2/M phase associated with dysregulation of α-tubulin, α1-tubulin and β5-tubulin expression. These findings suggest that the inhibitory effect of K1 can be mediated via inhibition of microtubule formation. Furthermore, simultaneously with G2/M arrest, K1 also increased population of cells with sub-G1 DNA content which is considered to be a marker of apoptotic cell death. Apoptosis was also confirmed by annexin V/PI double staining, DNA fragmentation assay and chromatin condensation assay. The apoptosis was associated with the loss of mitochondrial membrane potential (MMP), caspase-3 activation as well as intracellular reactive oxygen species (ROS) production. Moreover, the antioxidant Trolox blocked ROS production, changes in MMP and decreased K1 cytotoxicity, which confirmed the important role of ROS in cell apoptosis. Taken together, our data demonstrate that K1 induces ROS-dependent apoptosis in Caco2 cells and provide the rationale for further in vivo anticancer investigation.
Collapse
|
40
|
McCarthy N, Eberhart JK. Gene-ethanol interactions underlying fetal alcohol spectrum disorders. Cell Mol Life Sci 2014; 71:2699-706. [PMID: 24554057 PMCID: PMC11114006 DOI: 10.1007/s00018-014-1578-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/14/2014] [Accepted: 01/27/2014] [Indexed: 12/16/2022]
Abstract
Fetal alcohol spectrum disorders (FASD) is an umbrella term that describes a diverse set of ethanol-induced defects. The phenotypic variation is generated by numerous factors, including timing and dosage of ethanol exposure as well as genetic background. We are beginning to learn about how the concentration, duration, and timing of ethanol exposure mediate variability within ethanol teratogenesis. However, little is known about the genetic susceptibilities in FASD. Studies of FASD animal models are beginning to implicate a number of susceptibility genes that are involved in various pathways. Here we review the current literature that focuses on the genetic predispositions in FASD.
Collapse
Affiliation(s)
- Neil McCarthy
- Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research, Institute for Cellular and Molecular Biology, University of Texas at Austin,
Austin, TX 78713
USA
| | - Johann K. Eberhart
- Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research, Institute for Cellular and Molecular Biology, University of Texas at Austin,
Austin, TX 78713
USA
| |
Collapse
|
41
|
Brassinin and its derivatives as potential anticancer agents. Toxicol In Vitro 2014; 28:909-15. [PMID: 24747292 DOI: 10.1016/j.tiv.2014.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 03/17/2014] [Accepted: 04/01/2014] [Indexed: 01/12/2023]
Abstract
The aim of the study was to investigate the anti-proliferative activity of brassinin and its derivatives on human cancer cell lines. We found that among twenty-one tested compounds, 1- methoxybrassinin exerted the most potent anti-proliferative activity in Caco-2 cells with IC₅₀ 8.2 (±1.2)μmoll(-1). The flow cytometric analysis revealed a 1-methoxybrassinin-induced increase in the sub-G1 DNA content fraction which is considered to be a marker of apoptotic cell death. Apoptosis was also confirmed by DNA fragmentation assay. Moreover, quantitative real-time PCR showed that 1-methoxybrassinin upregulated the expression of pro-apoptotic Bax and downregulated the expression of anti-apoptotic genes Bcl-2 and Bcl-xL. The compound also increased activity of caspase-3, -7, cleaved PARP and decreased intracellular GSH content. The present study has assessed the in vitro anti-proliferative potential of 1-methoxybrassinin. The results generate a rationale for in vivo efficacy studies with this compound in preclinical cancer models.
Collapse
|
42
|
Ting H, Deep G, Agarwal C, Agarwal R. The strategies to control prostate cancer by chemoprevention approaches. Mutat Res 2014; 760:1-15. [PMID: 24389535 DOI: 10.1016/j.mrfmmm.2013.12.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 11/23/2013] [Accepted: 12/12/2013] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PCA) is the most commonly diagnosed cancer in men in the United States with growing worldwide incidence. Despite intensive investment in improving early detection, PCA often escapes timely detection and mortality remains high; this malignancy being the second highest cancer-associated mortality in American men. Collectively, health care costs of PCA results in an immense financial burden that is only expected to grow. Additionally, even in cases of successful treatment, PCA is associated with long-term and pervasive effects on patients. A proactive alternative to treat PCA is to prevent its occurrence and progression prior to symptomatic malignancy. This may serve to address the issue of burgeoning healthcare costs and increasing number of sufferers. One potential regimen in service of this alternative is PCA chemoprevention. Here, chemical compounds with cancer preventive efficacy are identified on the basis of their potential in a host of categories: their historical medicinal use, correlation with reduced risk in population studies, non-toxicity, their unique chemical properties, or their role in biological systems. PCA chemopreventive agents are drawn from multiple broad classes of chemicals, themselves further subdivided based on source or potential effect, with most derived from natural products. Many such compounds have shown efficacy, varying from inhibiting deregulated PCA cell signaling, proliferation, epithelial to mesenchymal transition (EMT), invasion, metastasis, tumor growth and angiogenesis and inducing apoptosis. Overall, these chemopreventive agents show great promise in PCA pre-clinical models, though additional work remains to be done in effectively translating these findings into clinical use.
Collapse
Affiliation(s)
- Harold Ting
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States
| | - Gagan Deep
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States; University of Colorado Cancer Center, University of Colorado, Aurora, CO, United States
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States; University of Colorado Cancer Center, University of Colorado, Aurora, CO, United States
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States; University of Colorado Cancer Center, University of Colorado, Aurora, CO, United States.
| |
Collapse
|