1
|
Guesmi F, Tahri W, Mehrez A, Barkaoui T, Prasad S, Giuffrè AM, Landoulsi A. Colorectal carcinoma cell targeting aromatherapy with Teucrium ramosissimum essential oil to sensitize TRAIL/Apo2L-induced HCT-116 cell death. Int Immunopharmacol 2024; 136:112405. [PMID: 38850792 DOI: 10.1016/j.intimp.2024.112405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/14/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
This report drives insights for the investigation of the underlying mechanisms of antitumor effects of Teucrium ramosissimum (TrS) essential oil (EO) that elicits colon tumor protection via activation of cell death machinery. A study of the aerial part phytocomplex was performed by FTIR spectra and GC/MS. In vivo colon carcinogenesis induced by LPS was carried out using mouse model. HCT-116 cells were coincubated with TrS EO and TRAIL-resistant cancer cells, and then cell lysates were assessed using Western blotting technique for death and decoy receptor expression. TrS essential oil potentiates TRAIL-mediated apoptosis cell death of HCT-116 as detected by PARP cleavage and caspase activation. Further data suggest that TrS up-regulates DR 5/4 expression, and down-regulates DcRs expression. Additionally, TrS potentiates apoptosis in TRAIL-resistant tumor cells through induction of MAPK signalling components, including ERK, p38 kinase, JNK, and activation of CHOP, and SP1, involved in DR5 expression. Moreover, Teucrium EO phytoconstituents mediate HCT-116 cells apoptosis by evoking cell cycle arrest at the G1 and G2/M phase through diminishing the expression of cyclin D1 acting as a potent multitargeted factors of inhibition of JAK/STAT oncogenic signaling pathway. These results demonstrate that TRAIL-induced apoptosis enhancing effect of TrS mediated through proto-oncogene expression in HCT-116. TrS administered intragastrically is able to prevent tumor of colon by stopping carcinogenesis process and impede tumor cell growth in in vivo analysis promoted by LPS. On the whole, our results revealed that TrS is an effective antitcancer agent through the induction of transcription factor and kinases, either are needed to trigger Apo2L receptors.
Collapse
Affiliation(s)
- Fatma Guesmi
- Laboratory of Risks Related to Environmental Stresses: Fight and Prevention, Unit UR03ES06, Faculty of Sciences of Bizerte, University of Carthage, Tunisia; Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| | - Wiem Tahri
- Laboratory of Risks Related to Environmental Stresses: Fight and Prevention, Unit UR03ES06, Faculty of Sciences of Bizerte, University of Carthage, Tunisia
| | - Amel Mehrez
- Laboratory of Risks Related to Environmental Stresses: Fight and Prevention, Unit UR03ES06, Faculty of Sciences of Bizerte, University of Carthage, Tunisia
| | - Taha Barkaoui
- Laboratory of Risks Related to Environmental Stresses: Fight and Prevention, Unit UR03ES06, Faculty of Sciences of Bizerte, University of Carthage, Tunisia
| | - Sahdeo Prasad
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA; Research and Development, Noble Pharma LLC, Menomonie, WI 54751, USA
| | - Angelo Maria Giuffrè
- Department AGRARIA, University of Studies "Mediterranea" of Reggio Calabria, Via dell'Università, 25 - 89124 Reggio Calabria, Italy.
| | - Ahmed Landoulsi
- Laboratory of Risks Related to Environmental Stresses: Fight and Prevention, Unit UR03ES06, Faculty of Sciences of Bizerte, University of Carthage, Tunisia
| |
Collapse
|
2
|
CCL3 Promotes Proliferation of Colorectal Cancer Related with TRAF6/NF-κB Molecular Pathway. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:2387192. [PMID: 35935327 PMCID: PMC9296340 DOI: 10.1155/2022/2387192] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/28/2022] [Accepted: 06/16/2022] [Indexed: 11/25/2022]
Abstract
Chemokine C-C motif chemokine ligand 3 (CCL3) plays an important role in the invasion and metastasis of malignant tumors. For developing new therapeutic targets and antitumor drugs, the effect of chemokine CCL3 and the related cytokine network on colorectal cancer should be investigated. This study used cell, tissue, and animal experiments to prove that CCL3 is highly expressed in colorectal cancer and confirmed that CCL3 can promote the proliferation of cancer cells, and its expression is closely related to TRAF6/NF-κB molecular pathway. In addition, protein chip technology was used to examine colorectal cancer tissue samples and identify the key factors of chemokine CCL3 and the toll-like receptors/nuclear factor-κB (TLR/NF-κB) pathway in cancer and metastatic lymph nodes. Furthermore, the lentiviral vector technology was employed for transfection to construct interference and overexpression cell lines. The experimental results reveal the mechanism of CCL3 and TNF receptor-associated factor 6 (TRAF6)/NF-κB pathway-related factors and their effects on the proliferation of colon cancer cells. Finally, the expression and significance of CCL3 in colorectal cancer tissues and its correlation with clinical pathology were studied by immunohistochemistry. Also, the results confirmed that CCL3 and C-C motif chemokine receptor 5 (CCR5) were expressed in adjacent tissues, colorectal cancer tissues, and metastatic cancer. The expression level was correlated with the clinical stage and nerve invasion. The expression of chemokine CCL3 and receptor CCR5 was positively correlated with the expression of TRAF6 and NF-κB and could promote the proliferation, invasion, and migration of colorectal cancer cells through TRAF6 and NF-κB.
Collapse
|
3
|
The effects of centipedegrass extract on hair growth via promotion of anagen inductive activity. PLoS One 2022; 17:e0265532. [PMID: 35320304 PMCID: PMC8942214 DOI: 10.1371/journal.pone.0265532] [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: 06/04/2021] [Accepted: 03/03/2022] [Indexed: 12/02/2022] Open
Abstract
To investigate the CGE on hair growth and to explore the mechanism that is involved in the acceleration of anagen induction, we investigated the effects of CGE studied on cell proliferation and molecular mechanism in human hair dermal papilla cells (hDPCs) and keratinocytes (HaCaT cells). Additionally, hair growth evaluation was carried out following topical treatment of the dorsal skin of telogen C57BL/6 mice with CGE for 14 days. As result, CGE increased cell viability and ALP activity in hDPCs. Moreover, CGE increased the expression of catenin beta 1 (CTNNB1), ALP, sex-determining region Y-box 2 (SOX2), insulin-like growth factor 1 (IGF1), and vascular endothelial growth factor A (VEGFA) genes in hDPCs. CGE increased the expression of proteins such as ALP, β-catenin, and phosphorylation of glycogen synthase kinase 3β (pGSK3β), and protein kinase B (pAKT) in hDPCs. Furthermore, CGE induced the proliferation of HaCaT cells and up-regulated AKT-ERK-GSKβ-β-catenin signaling in HaCaT cells. Additionally, the anagen induction effects of CGE were confirmed on the telogen-anagen transition mice model. these findings demonstrated that CGE promoted the entering the growth phase of hair follicle via activation of β-catenin signaling pathways in vivo. Thus, this study suggests that CGE might be a potential therapeutic reagent for hair growth.
Collapse
|
4
|
Guesmi F, Prasad S, Ali MB, Ismail IA, Landoulsi A. Thymus hirtus sp. algeriensis Boiss. and Reut. volatile oil enhances TRAIL/Apo2L induced apoptosis and inhibits colon carcinogenesis through upregulation of death receptor pathway. Aging (Albany NY) 2021; 13:21975-21990. [PMID: 34543231 PMCID: PMC8507293 DOI: 10.18632/aging.203552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/11/2021] [Indexed: 11/25/2022]
Abstract
Background: The aim of the study is to determine the anticancer activity of Thymus algeriensis (TS) and its underlying mechanisms using in vitro and in animal models. Methods: HCT116 cells were treated with TS essential oil alone or with TRAIL, and then its anticancer effect was determined by using MTT assay, live dead assay, caspase activation and PARP cleavage. Further mechanisms of its anticancer effects was determined by analyzing expression of death receptor signaling pathway using Western blotting. A mouse model was also used to assess the antitumor potential of thyme essential oil. Results: TS oily fraction showed tumor growth inhibitory effect even at lower concentration. TS induces apoptotic cell death as indicated by cleavage of PARP, and activation of the initiator and effector caspases (caspase-3, -8 and -9). Further, results showed that TS increases the expression of death receptors (DRs) and reduces the expression of TRAIL decoy receptors (DcRs). In addition, upregulation of signaling molecules of MAPK pathway (p38 kinase, ERK, JNK), down-regulation of c-FLIP, and overexpression of SP1 and CHOP were observed by TS. Further in animal model, intragastric administration of TS (12.5 mg/ml and 50 mg/ml) prevented colorectal carcinogenesis by blocking multi-steps in carcinoma. Conclusion: Overall, these results indicate that thymus essential oil promotes apoptosis in HCT116 cells and impedes tumorigenesis in animal model. Moreover, thyme potentiates TRAIL-induced cell death through upregulation of DRs, CHOP and SP1 as well as downregulation of antiapoptotic proteins in HCT116 cells. However, therapeutic potential of TS needs to be further explored.
Collapse
Affiliation(s)
- Fatma Guesmi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Laboratory of Biochemistry and Molecular Biology, University of Carthage, Faculty of Sciences of Bizerte, Zarzouna, Bizerte 7021, Tunisia
| | - Sahdeo Prasad
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Research and Development, Noble Pharma LLC, Menomonie, WI 54751, USA
| | - Manel Ben Ali
- Department of Biology, College of Science, Taif University, Taif 21944, Saudi Arabia
| | - Ismail A Ismail
- Department of Biology, College of Science, Taif University, Taif 21944, Saudi Arabia
| | - Ahmed Landoulsi
- Laboratory of Biochemistry and Molecular Biology, University of Carthage, Faculty of Sciences of Bizerte, Zarzouna, Bizerte 7021, Tunisia
| |
Collapse
|
5
|
Effect of heating on the digestibility of isolated hempseed (Cannabis sativa L.) protein and bioactivity of its pepsin-pancreatin digests. Food Chem 2020; 314:126198. [PMID: 31954285 DOI: 10.1016/j.foodchem.2020.126198] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/07/2020] [Accepted: 01/10/2020] [Indexed: 01/13/2023]
Abstract
The objective was to investigate the effects of heat pretreatment and simulated gastrointestinal digestion on potential antioxidant, anticancer and anti-inflammatory activities of hempseed (Cannabis sativa L.) proteins. Unheated isolated hempseed protein (IHP) and its heated counterparts (100 °C, 15 min and 30 min, termed as HP15D and HP30D) were hydrolyzed sequentially with pepsin and pancreatin and analyzed for digestibility and bioactivity (antioxidant, anti-proliferative and anti-inflammatory properties). Heat pretreatment led to an increase of low molecular weight proteins and degree of hydrolysis, and decrease of concentration of soluble protein, which means heat pretreated can significantly improve the digestibility of IHP. Pepsin-pancreatin digests released from heat pretreated IHP possessed less antioxidant, antiproliferative and anti-inflammatory properties than digests from unheated IHP. In conclusion, heat pre-treatment improved the digestibility of IHP but the resulting digests from heated IHP had lower bioactivity.
Collapse
|
6
|
Bai HW, Park CH, Jang DM, Kawala RA, Lee SS, Chung BY. Centipedegrass extracts regulate LPS-mediated aberrant immune responses by inhibiting Janus kinase. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 55:172-178. [PMID: 30668427 DOI: 10.1016/j.phymed.2018.06.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/20/2018] [Accepted: 06/19/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Centipedegrass extract (CGE) is rich in several polyphenolic compounds including C-glycosylflavonoids, such as maysin and its derivatives, and exerts antioxidant, anti-adipogenic and anticancer effects. However, the effect of CGE on the immune system is unclear. PURPOSE CGE might inhibit NO production induced by lipopolysaccharide (LPS). In this study, we propose a molecular mechanism for regulation of aberrant immune responses by CGE in LPS-stimulated RAW264.7 cells. STUDY DESIGN We will preparation of Centipedegrass extract and purify partially in rich of maysin and its derivatives. And examine the effect of the CGE on immune system using LPS-induced RAW cells and animals. METHODS LPS-induced nitric oxide (NO) and interleukin-6 levels were measured by enzyme-linked immunosorbent assay. The mRNA and protein levels of immune mediators were analyzed by reverse-transcription polymerase chain reaction and immunoblotting, respectively. RESULTS CGE inhibited LPS-induced NO production in a concentration-dependent manner by suppressing inducible nitric oxide synthase (iNOS) expression in LPS-stimulated cells; this effect was mediated by inhibition of the JAK/STAT pathway. However, CGE did not regulate the expression of other factors, including phosphorylated p38, c-jun N-terminal kinase, or extracellular signal-regulated kinase 1/2. In addition, CGE increased T cells percentage in peripheral blood after oral administration. CONCLUSION These results indicate that CGE suppresses LPS-induced production of NO and expression of iNOS by directly inhibiting JAK2 kinase activity and enhancing effects on the immune system in mice.
Collapse
Affiliation(s)
- Hyoung-Woo Bai
- Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup 56212, Republic of Korea; Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Chul-Hong Park
- Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup 56212, Republic of Korea
| | - Dong-Min Jang
- Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup 56212, Republic of Korea
| | - Remigius Ambrose Kawala
- Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup 56212, Republic of Korea; Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Seung Sik Lee
- Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup 56212, Republic of Korea; Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Byung Yeoup Chung
- Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), Jeongeup 56212, Republic of Korea.
| |
Collapse
|
7
|
Lin Y, Li B, Zhao J, Wei L, Wang Y, Wang M, Dia VP, Meng X. Combinatorial effect of blueberry extracts and oxaliplatin in human colon cancer cells. J Cell Physiol 2019; 234:17242-17253. [DOI: 10.1002/jcp.28341] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/23/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Yang Lin
- College of Food Science Shenyang Agricultural University Shenyang Liaoning China
- Department of Food Science The University of Tennessee Institute of Agriculture Knoxville Tennessee
| | - Bin Li
- College of Food Science Shenyang Agricultural University Shenyang Liaoning China
| | - Jin Zhao
- College of Food Science Shenyang Agricultural University Shenyang Liaoning China
| | - Lulu Wei
- College of Food Science Shenyang Agricultural University Shenyang Liaoning China
| | - Yuehua Wang
- College of Food Science Shenyang Agricultural University Shenyang Liaoning China
| | - Mingyue Wang
- College of Food Science Shenyang Agricultural University Shenyang Liaoning China
| | - Vermont P. Dia
- Department of Food Science The University of Tennessee Institute of Agriculture Knoxville Tennessee
| | - Xianjun Meng
- College of Food Science Shenyang Agricultural University Shenyang Liaoning China
| |
Collapse
|
8
|
Liang X, Kong P, Wang J, Xu Y, Gao C, Guo G. Effects of metformin on proliferation and apoptosis of human megakaryoblastic Dami and MEG-01 cells. J Pharmacol Sci 2017; 135:14-21. [PMID: 28927780 DOI: 10.1016/j.jphs.2017.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/11/2017] [Accepted: 08/02/2017] [Indexed: 01/07/2023] Open
Abstract
Metformin has received increasing attention for its potential anticancer activity against certain human leukemia cells, but its effects on human megakaryoblastic cells are unclear. This study aimed to investigate the effects of metformin on proliferation and apoptosis of human megakaryoblastic cells (Dami and MEG-01) and the underlying molecular mechanisms. CCK8 assay was employed to measure cell proliferation. Flow cytometry was adopted to detect cell apoptosis. Western blot was further employed to measure apoptosis-related proteins. In Dami and MEG-01 cells, metformin significantly inhibited proliferation and promoted apoptosis in a dose- and time-dependent manner, and metformin (4 mM) was selected for subsequent experiments. Metformin inhibited ERK1/2, JNK, and PI3K/Akt, but activated p38 pathway in these two cells. Moreover, inhibition of ERK1/2, JNK or PI3K/Akt pathway alone induced cell apoptosis compared to the control group. The combination of specific inhibitors of ERK1/2, JNK or PI3K/Akt pathway and metformin further promoted cell apoptosis and the up-regulation of p21, Bax, Bad, cleaved caspase-3 and -9 as well as the down-regulation of Bcl-2 mediated by metformin alone, but inhibition of p38 pathway exhibited the opposite results. These findings support the possibility of metformin treatment as a new therapeutic strategy against acute megakaryoblastic leukemia (AMKL).
Collapse
Affiliation(s)
- Xue Liang
- Department of Hematology, No. 150 Central Hospital of Chinese People's Liberation Army, Luoyang, China
| | - Peiyan Kong
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Jin Wang
- Department of Hematology, No. 150 Central Hospital of Chinese People's Liberation Army, Luoyang, China
| | - Yulin Xu
- Department of Hematology, No. 150 Central Hospital of Chinese People's Liberation Army, Luoyang, China
| | - Chunfang Gao
- Institute of Anal-Colorectal Surgery, No. 150 Central Hospital of Chinese People's Liberation Army, No. 2 Huaxia Road, Luoyang, 471000, China.
| | - Guozhen Guo
- Department of Radiation Medicine, School of Public Health, The Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
9
|
Wang C, Guo L, Wang S, Wang J, Li Y, Dou Y, Wang R, Shi H, Ke Y, Liu H. Anti-proliferative effect of Jesridonin on paclitaxel-resistant EC109 human esophageal carcinoma cells. Int J Mol Med 2017; 39:645-653. [PMID: 28204832 PMCID: PMC5360389 DOI: 10.3892/ijmm.2017.2867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/13/2017] [Indexed: 12/19/2022] Open
Abstract
Chemoresistance to anticancer drugs is a major obstacle in the efforts to develop a successful treatment strategy for esophageal squamous carcinoma (ESCC). Thus, the exploration of new drugs and treatment strategies for combating resistance are of utmost importance. In this study, we investigated the antitumor drug resistance activity of Jesridonin, a new ent-kaurene diterpenoid, and its possible mechanisms of action using the resistant cancer cell line, EC109/Taxol. MTT assay revealed that Jesridonin had similar IC50 values against EC109 paclitaxel-sensitive cells and drug-resistant EC109/Taxol cells in vitro. In mice, Jesridonin effectively prevented the growth of EC109/Taxol tumor xenografts without exerting any significant toxicity. In addition, Jesridonin significantly inhibited the proliferation of EC109/Taxol cells, induced apoptosis and arrested the cell cycle at the G2/M phase. Furthermore, western blot analysis revealed that Jesridonin upregulated the expression of p53, p53 upregulated modulator of apoptosis (PUMA), cleaved-caspase-9 and cleaved-caspase-3 in EC109/Taxol cells, and downregulated the expression of procaspase-3, procaspase-9 and Bcl-2 in the EC109/Taxol cells in a concentration-dependent manner. Overall, our results demonstrate that Jesridonin may have potential for use in the treatment of paclitaxel-resistant ESCC. The data of the present study may lead to the development of novel treatment strategies for paclitaxel-resistant tumors.
Collapse
Affiliation(s)
- Cong Wang
- Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Zhengzhou, Henan 450001, P.R. China
| | - Liubin Guo
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Saiqi Wang
- Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Zhengzhou, Henan 450001, P.R. China
| | - Junwei Wang
- Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Zhengzhou, Henan 450001, P.R. China
| | - Yongmei Li
- Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Zhengzhou, Henan 450001, P.R. China
| | - Yinhui Dou
- Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Zhengzhou, Henan 450001, P.R. China
| | - Ran Wang
- Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Zhengzhou, Henan 450001, P.R. China
| | - Hongge Shi
- Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Zhengzhou, Henan 450001, P.R. China
| | - Yu Ke
- Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Zhengzhou, Henan 450001, P.R. China
| | - Hongmin Liu
- Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
10
|
Pan LL, Wang XL, Luo XL, Liu SY, Xu P, Hu JF, Liu XH. Boehmenan, a Lignan From the Chinese Medicinal Plant Clematis armandii, Inhibits A431 Cell Growth via Blocking p70S6/S6 Kinase Pathway. Integr Cancer Ther 2016; 16:351-359. [PMID: 27698262 PMCID: PMC5759931 DOI: 10.1177/1534735416669803] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Previously, we have shown that boehmenan, a natural product isolated from the dried stem of Caulis clematidis armandii, exhibits various biological activities. The current study investigated the effects of boehmenan on the growth of human epidermoid carcinoma A431 cells. Cell viability and 50% inhibiting concentration (IC50) were assessed by CellTiter-Glo luminescent cell viability assay. Cell cycle arrest was measured by flow cytometry. Intracellular reactive oxygen species production and mitochondrial membrane potential (ΔΨm) collapse were analyzed by a fluorescence spectrophotometer. The activation of epidermal growth factor receptor signaling pathway was evaluated by Western blot. The results showed that boehmenan significantly inhibited the growth of A431 cells (IC50 = 1.6 µM) in a concentration- and time-dependent manner. This compound also blocked cell cycle progression at G2/M phase and modulated mitochondrial apoptosis-related proteins, as evidenced by upregulating p21, cleaved caspase-3, and cleaved poly (ADP-ribose) polymerase protein levels and by downregulating Bcl-2, pro-caspase-9 levels. In addition, boehmenan also markedly induced intracellular reactive oxygen species production and ΔΨm depolarization in a concentration-dependent manner. Furthermore, boehmenan-attenuated epidermal growth factor mediated the phosphorylation of signal transducer and activator of transcription 3 (STAT3), p70 ribosomal protein S6 kinase (p70S6)/S6 in a concentration-dependent manner. Taken together, our results suggest that boehmenan-mediated antiproliferative property in A431 cells was mediated partially by modulation of mitochondrial function and inhibition of STAT3 and p70S6 signal pathways.
Collapse
Affiliation(s)
| | | | | | | | - Peng Xu
- 1 Fudan University, Shanghai, China
| | | | | |
Collapse
|
11
|
León-González AJ, Sharif T, Kayali A, Abbas M, Dandache I, Etienne-Selloum N, Kevers C, Pincemail J, Auger C, Chabert P, Alhosin M, Schini-Kerth VB. Delphinidin-3-O-glucoside and delphinidin-3-O-rutinoside mediate the redox-sensitive caspase 3-related pro-apoptotic effect of blackcurrant juice on leukaemia Jurkat cells. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.06.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
12
|
Su C, Fan M, Lu L, Li P. Role of epidermal growth factor in pathogenesis of uterine leiomyomas. ASIAN PAC J TROP MED 2015; 8:378-81. [PMID: 26003597 DOI: 10.1016/s1995-7645(14)60347-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate the role of epidermal growth factor (EGF) in the pathogenesis of uterine leiomyomas. METHODS Human myometrial smooth muscle cells (HM-SMCs) and smooth muscle cells of human uterine leiomyomas (HL-SMCs) were separated from patients' specimens and cultured. After processed by EGF or PD98059 (inhibitor of MKK/MEK) +EGF, the proliferation rate of both SMCs was detected by BrdU method and the phosphorylation level of p44/42 mitogen-activated protein kinase (MAPK) was determined by Western-blot. After different processing time by EGF, the phosphorylation levels of p44/42 MAPK and AKT and p27 expression level in both SMCs were detected by Western-blot. RESULTS EGF could significantly promote HL-SMCs proliferation and PD98059 could inhibit this effect (P<0.05); besides, PD98059 could inhibit the increase of the phosphorylation level of p44/42 MAPK in both SMCs induced by EGF. When the processing time by EGF was over 15min, the phosphorylation levels of p44/42 MAPK and AKT in both SMCs decreased sharply and were close to zero; p27 expression in HM-SMCs raised significantly while the upregulation in HL-SMCs was little. CONCLUSIONS EGF could not cause activation of EGFR because of the dephosphorylation of p44/42 MAPK and AKT in HL-SMCs, which caused p27 expression insufficiently and cell cycle dysregulation.
Collapse
Affiliation(s)
- Chun Su
- Department of Gynaecology and Obstetrics, The Fifth Affiliated Hospital of Zhengzhou University, Kangfu Qian Street No. 3, 450052, Zhengzhou, China
| | - Mei Fan
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Jianshe Dong Street No.1, 450052, Zhengzhou, China
| | - Lin Lu
- Department of Ultrasound, The Third Affiliated Hospital of Zhengzhou University, Kangfu Qian Street No. 7, 450052, Zhengzhou, China
| | - Pei Li
- Department of Pathophysiology, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|