1
|
Potential Role of Curcumin and Its Nanoformulations to Treat Various Types of Cancers. Biomolecules 2021; 11:biom11030392. [PMID: 33800000 PMCID: PMC8001478 DOI: 10.3390/biom11030392] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
Cancer is a major burden of disease globally. Each year, tens of millions of people are diagnosed with cancer worldwide, and more than half of the patients eventually die from it. Significant advances have been noticed in cancer treatment, but the mortality and incidence rates of cancers are still high. Thus, there is a growing research interest in developing more effective and less toxic cancer treatment approaches. Curcumin (CUR), the major active component of turmeric (Curcuma longa L.), has gained great research interest as an antioxidant, anticancer, and anti-inflammatory agent. This natural compound shows its anticancer effect through several pathways including interfering with multiple cellular mechanisms and inhibiting/inducing the generation of multiple cytokines, enzymes, or growth factors including IκB kinase β (IκKβ), tumor necrosis factor-alpha (TNF-α), signal transducer, and activator of transcription 3 (STAT3), cyclooxygenase II (COX-2), protein kinase D1 (PKD1), nuclear factor-kappa B (NF-κB), epidermal growth factor, and mitogen-activated protein kinase (MAPK). Interestingly, the anticancer activity of CUR has been limited primarily due to its poor water solubility, which can lead to low chemical stability, low oral bioavailability, and low cellular uptake. Delivering drugs at a controlled rate, slow delivery, and targeted delivery are other very attractive methods and have been pursued vigorously. Multiple CUR nanoformulations have also been developed so far to ameliorate solubility and bioavailability of CUR and to provide protection to CUR against hydrolysis inactivation. In this review, we have summarized the anticancer activity of CUR against several cancers, for example, gastrointestinal, head and neck, brain, pancreatic, colorectal, breast, and prostate cancers. In addition, we have also focused on the findings obtained from multiple experimental and clinical studies regarding the anticancer effect of CUR in animal models, human subjects, and cancer cell lines.
Collapse
|
2
|
Zhang Q, Mohammed EAH, Wang Y, Bai Z, Zhao Q, He D, Wang Z. Synthesis and anti-hepaticfibrosis of glycyrrhetinic acid derivatives with inhibiting COX-2. Bioorg Chem 2020; 99:103804. [PMID: 32272365 DOI: 10.1016/j.bioorg.2020.103804] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/24/2020] [Accepted: 03/27/2020] [Indexed: 02/08/2023]
Abstract
Many tests have shown cyclooxygenase-2 (COX-2) was closely related to the activation of hepatic stellate cells (HSCs), which further promoting the onset and development of hepatic fibrosis. According to these research findings, a series of glycyrrhetinic acid derivatives were designed and synthesized. Meanwhile, their anti-hepaticfibrotic activities were evaluated in vitro and in vivo. Firstly, in the tests of the cell models, all the compounds displayed anti-proliferative effect on the HSC-T6 activated by (transforming growth factor beta) TGF-β1 (10 ng/mL). Among them, compounds 2 and 16 exhibited a stronger activity than the others, and their IC50 values were 17.6 µM and 30.3 µM, respectively; both of them were low toxicity to normal HSC-T6 cells and WI38 cells, and they inhibited the activated HSC-T6 cells proliferation by promoting apoptosis and resting them at the G0/G1 phase. Secondly, compounds 2 and 16 displayed strong inhibitory effect on activation of HSCs; they not only inhibited the expression of α-SMA and Col1 in the activated HSC-T6 cells, but also decreased the levels of COX-2, TGF-β1 and (reactive oxygen species) ROS in a concentration-dependent manner; they down-regulated the levels of three biomarkers in the process of test, but this decrease did not change linearly with the action time of compound. Thirdly, for the rats which induced with (carbontetrachloride) CCl4, the symptoms of liver fibrosis in rats were significantly alleviated after successive administration the tested compound for 14d; the α-SMA level in liver tissue decreased in a concentration dependent manner; and the liver cell necrosis and the fat collagen fiber decreased significantly compared with the positive control group; furthermore, inflammatory infiltration was significantly lower than that of the control. This suggests the compounds possibly are candidates for hepatic fibrosis with promising application in clinic.
Collapse
Affiliation(s)
- Qiuping Zhang
- Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou 730000, China
| | | | - Yanni Wang
- Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou 730000, China
| | - Zhongjie Bai
- Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou 730000, China
| | - Quanyi Zhao
- Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou 730000, China.
| | - Dian He
- Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou 730000, China
| | - Zhen Wang
- Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
3
|
Chen K, Lai K, Zhang X, Qin Z, Fu Q, Luo C, Jin X, Hu J, Liu S, Yao K. Bromfenac Inhibits TGF-β1-Induced Fibrotic Effects in Human Pterygium and Conjunctival Fibroblasts. Invest Ophthalmol Vis Sci 2019; 60:1156-1164. [PMID: 30908581 DOI: 10.1167/iovs.18-24743] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Nonsteroidal anti-inflammatory drugs (NSAIDs) have shown antifibrotic effects on several diseases. The aims of the present in vitro study were to investigate the antifibrotic effects of bromfenac (a kind of NSAID) on primary human pterygium fibroblasts (HPFs) and primary human conjunctival fibroblasts (HConFs), as well as to explore the possible mechanisms of these effects. Methods The cells used in this study were primary HPFs and HConFs, and profibrotic activation was induced by transforming growth factor-beta1 (TGF-β1). Western blot, quantitative real-time PCR, and immunofluorescence (IF) assays were used to detect the effects of TGF-β1 and bromfenac on the synthesis of fibronectin (FN), type III collagen (COL3), and alpha-smooth muscle actin (α-SMA) in HPFs and HConFs; the changes of signaling pathways were detected by Western blot; cell migration ability was detected by wound healing assay; cell proliferation ability was detected by CCK-8 assay; and pharmaceutical inhibitions of the downstream signaling pathways of TGF-β1 were used to assess their possible associations with the effects of bromfenac. Results Bromfenac suppressed the TGF-β1-induced protein expression of FN (0.59 ± 0.07 folds, P = 0.008), COL3 (0.48 ± 0.08 folds, P = 0.001), and α-SMA (0.61 ± 0.03 folds, P = 0.008) in HPFs. Bromfenac also attenuated TGF-β1-induced cell migration (0.30 ± 0.07 folds, P < 0.001), cell proliferation (0.64 ± 0.03 folds, P = 0.002) and the expression levels of p-AKT (0.66 ± 0.08 folds, P = 0.032), p-ERK1/2 (0.69 ± 0.11 folds, P = 0.003), and p-GSK-3β-S9 (0.65 ± 0.10 folds, P = 0.002) in HPFs. PI3K/AKT inhibitor (wortmannin) and MEK/ERK inhibitor (U0126) reduced the TGF-β1-induced synthesis of FN, COL3, and α-SMA in HPFs. All the results were similar in HConFs. Conclusions Bromfenac protects against TGF-β1-induced synthesis of FN, α-SMA, and COL3 in HPFs and HConFs at least in part by inactivating the AKT and ERK pathways.
Collapse
Affiliation(s)
- Kailin Chen
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Kairan Lai
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Xiaobo Zhang
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Zhenwei Qin
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Qiuli Fu
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Chenqi Luo
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Xiuming Jin
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Jianghua Hu
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Siyu Liu
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| | - Ke Yao
- Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Ophthalmology of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
4
|
Álvares PR, de Arruda JAA, Oliveira Silva LV, da Silva LP, do Nascimento GJF, da Silveira MMF, Sobral APV. Immunohistochemical Analysis of Cyclooxygenase-2 and Tumor Necrosis Factor Alpha in Periapical Lesions. J Endod 2018; 44:1783-1787. [DOI: 10.1016/j.joen.2018.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/22/2018] [Accepted: 09/06/2018] [Indexed: 12/19/2022]
|
5
|
Silva LP, Gonzaga AKG, Serpa MS, Nascimento GJF, Sobral APV. Immunohistochemical expression of cyclooxygenase-2 and cyclin-D1 in oral squamous cell carcinoma. JOURNAL OF ORAL AND MAXILLOFACIAL SURGERY MEDICINE AND PATHOLOGY 2017. [DOI: 10.1016/j.ajoms.2016.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
6
|
Quercetin as an Antiviral Agent Inhibits Influenza A Virus (IAV) Entry. Viruses 2015; 8:v8010006. [PMID: 26712783 PMCID: PMC4728566 DOI: 10.3390/v8010006] [Citation(s) in RCA: 242] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 11/25/2015] [Accepted: 11/30/2015] [Indexed: 12/22/2022] Open
Abstract
Influenza A viruses (IAVs) cause seasonal pandemics and epidemics with high morbidity and mortality, which calls for effective anti-IAV agents. The glycoprotein hemagglutinin of influenza virus plays a crucial role in the initial stage of virus infection, making it a potential target for anti-influenza therapeutics development. Here we found that quercetin inhibited influenza infection with a wide spectrum of strains, including A/Puerto Rico/8/34 (H1N1), A/FM-1/47/1 (H1N1), and A/Aichi/2/68 (H3N2) with half maximal inhibitory concentration (IC50) of 7.756 ± 1.097, 6.225 ± 0.467, and 2.738 ± 1.931 μg/mL, respectively. Mechanism studies identified that quercetin showed interaction with the HA2 subunit. Moreover, quercetin could inhibit the entry of the H5N1 virus using the pseudovirus-based drug screening system. This study indicates that quercetin showing inhibitory activity in the early stage of influenza infection provides a future therapeutic option to develop effective, safe and affordable natural products for the treatment and prophylaxis of IAV infections.
Collapse
|
7
|
Naska S, Yuzwa SA, Johnston APW, Paul S, Smith KM, Paris M, Sefton MV, Datti A, Miller FD, Kaplan DR. Identification of Drugs that Regulate Dermal Stem Cells and Enhance Skin Repair. Stem Cell Reports 2015; 6:74-84. [PMID: 26724904 PMCID: PMC4719140 DOI: 10.1016/j.stemcr.2015.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 12/01/2015] [Accepted: 12/01/2015] [Indexed: 01/24/2023] Open
Abstract
Here, we asked whether we could identify pharmacological agents that enhance endogenous stem cell function to promote skin repair, focusing on skin-derived precursors (SKPs), a dermal precursor cell population. Libraries of compounds already used in humans were screened for their ability to enhance the self-renewal of human and rodent SKPs. We identified and validated five such compounds, and showed that two of them, alprostadil and trimebutine maleate, enhanced the repair of full thickness skin wounds in middle-aged mice. Moreover, SKPs isolated from drug-treated skin displayed long-term increases in self-renewal when cultured in basal growth medium without drugs. Both alprostadil and trimebutine maleate likely mediated increases in SKP self-renewal by moderate hyperactivation of the MEK-ERK pathway. These findings identify candidates for potential clinical use in human skin repair, and provide support for the idea that pharmacological activation of endogenous tissue precursors represents a viable therapeutic strategy. Small-molecule screens identify compounds that enhance SKP self-renewal Alprostadil and trimebutine maleate both increase SKP self-renewal Both compounds likely act by promoting activation of the MEK-ERK pathway Both compounds activated dermal precursors in vivo to enhance wound healing
Collapse
Affiliation(s)
- Sibel Naska
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Scott A Yuzwa
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Adam P W Johnston
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Smitha Paul
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Kristen M Smith
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Maryline Paris
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Michael V Sefton
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Alessandro Datti
- S.M.A.R.T. Laboratory for High-Throughput Screening Programs, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Agricultural, Food, and Environmental Sciences, University of Perugia, 06121 Perugia, Italy
| | - Freda D Miller
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada; Department of Physiology, University of Toronto, Toronto, ON M5G 1X5, Canada.
| | - David R Kaplan
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada.
| |
Collapse
|
8
|
Zhong D, Gu C, Shi L, Xun T, Li X, Liu S, Yu L. Obatoclax induces G1/G0-phase arrest via p38/p21(waf1/Cip1) signaling pathway in human esophageal cancer cells. J Cell Biochem 2015; 115:1624-35. [PMID: 24788582 DOI: 10.1002/jcb.24829] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 04/29/2014] [Indexed: 12/15/2022]
Abstract
Pan-Bcl-2 family inhibitor obatoclax has been demonstrated to be effective against various cancers, of which the mechanism of action is not fully understood. In this study, we demonstrate that obatoclax suppressed esophageal cancer cell viability with concomitant G1/G0-phase cell cycle arrest. At the tested concentrations (1/2 IC50 and IC50), obatoclax neither induced PARP cleavage nor increased the Annexin V-positive population, suggesting G1/G0-phase arrest rather than apoptosis accounts for most of the reduction of cell viability produced by obatoclax. Double knockdown of Bak and Bax by small interference RNA failed to block obatoclax-induced G1/G0-phase arrest, implying its role in cell cycle progression is Bak/Bax-independent. The cell cycle arresting effect of obatoclax was associated with up-regulation of p21(waf1/Cip1). Knockdown of p21(waf1/Cip1) significantly attenuated obatoclax-induced G1/G0-phase arrest. Although obatoclax stimulated phosphorylation of Erk, p38, and JNK, pharmacological inhibition of p38 but not Erk or JNK blocked obatoclax-induced G1/G0-phase arrest. Moreover, knockdown of p38 abolished the cell cycle arresting effect of obatoclax. In consistent with this finding, inhibition of p38 blocked obatoclax-induced p21(waf1/Cip1) expression while inhibition of Erk or JNK failed to exert similar effect. To conclude, these findings suggest that obatoclax induced cell cycle arrest via p38/p21(waf1/Cip1) signaling pathway. This study may shed a new light on the anti-cancer activity of obatoclax in relation to cell cycle arrest.
Collapse
Affiliation(s)
- Desheng Zhong
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | | | | | | | | | | | | |
Collapse
|
9
|
Diakowska D, Markocka-Mączka K, Nienartowicz M, Lewandowski A, Grabowski K. Increased level of serum prostaglandin-2 in early stage of esophageal squamous cell carcinoma. Arch Med Sci 2014; 10:956-61. [PMID: 25395947 PMCID: PMC4223123 DOI: 10.5114/aoms.2013.34985] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 05/26/2012] [Accepted: 09/26/2012] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Prostaglandin-2 (PGE-2), one of the products of cyclooxygenase-2 (COX-2) induced catalysis, may play a critical role in the carcinogenesis of esophageal squamous cell carcinoma (ESCC). We investigated the efficacy of using serum PGE-2 concentration as a biomarker for this cancer type. MATERIAL AND METHODS Prostaglandin-2 levels were analyzed in the serum of 65 ESCC patients and in 47 healthy individuals. The concentrations of cyclooxygenase-2 (COX-2) were measured in tumor tissues and normal tissues obtained from 31 surgically treated ESCC patients. RESULTS Serum PGE-2 concentration was significantly higher in ESCC patients than in control patients (p = 0.004), especially in the early stages (I + II) of cancer (p < 0.0001). We observed significant inverse relationships between serum PGE-2 levels and: tumor stage, primary tumor progression, lymph and distant metastasis. The COX-2 concentration was significantly elevated in tumors as compared to normal tissues (p = 0.008). A significant correlation between serum PGE-2 and tumor COX-2 was observed (rho = 0.46, p = 0.009). However, ROC analysis showed that serum PGE-2 may be a weak prognostic factor for ESCC. CONCLUSIONS Our results suggest that an elevated concentration of serum PGE-2 in the early stages of cancer may possibly be associated with tumor initiation and cancer development in ESCC. The exact role of these findings in early detection of this highly lethal cancer requires further research.
Collapse
Affiliation(s)
- Dorota Diakowska
- Department of Gastrointestinal and General Surgery, Medical University of Wroclaw, Wroclaw, Poland
| | - Krystyna Markocka-Mączka
- Department of Gastrointestinal and General Surgery, Medical University of Wroclaw, Wroclaw, Poland
| | - Mirosław Nienartowicz
- Department of Gastrointestinal and General Surgery, Medical University of Wroclaw, Wroclaw, Poland
| | - Andrzej Lewandowski
- Department of Gastrointestinal and General Surgery, Medical University of Wroclaw, Wroclaw, Poland
| | - Krzysztof Grabowski
- Department of Gastrointestinal and General Surgery, Medical University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
10
|
Xu KK, Tian F, Chang D, Gong M, Fan JQ, Wang TY. Clinical effect of E-series of prostaglandin receptor 2 and epidermal growth factor receptor signal pathways in the development of esophageal squamous cell carcinoma. Dis Esophagus 2014; 27:388-95. [PMID: 24883443 DOI: 10.1111/dote.12143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Signal pathways mediated by epidermal growth factor receptor (EGFR) and E-series of prostaglandin receptors (EPs) are closely correlated to the pathogenesis of tumor. This experiment was designed to investigate the expression and clinical significance of EP2 and EGFR in esophageal squamous cell carcinoma (ESCC). Tissue samples were collected reterospectively from 87 patients with ESCC (first diagnosed). The patients were followed up for 5 years after radical surgery. The expression of EP-2 and EGFR were examined by tissue chip technology and immunohistochemistry methods. Clinicopathological and prognostic impact were evaluated. Overexpression of EGFR and EP-2 was more observed in ESCC than the control group (58.6% vs. 13.9%; 52.9% vs. 4.88%, P < 0.001, respectively); which correlated with tumor infiltration depth, lymph node metastasis, and tumor-lymph node-metastasis staging. Both the EP-2 and EGFR overexpression were detected in 39 specimens and exhibited the positive correlation (P < 0.001, r = 0.404). Overexpression of EP2 and EGFR exhibited significant correlation with worse 5-year overall survival than those with negative result (17.6% vs. 27.8%, P = 0.011; 10.9% vs. 34.1%, P < 0.001, respectively). Cox proportional hazard model showed that the T-staging, lymph node metastasis, and EGFR overexpression were the independent risk factors of the prognosis. The present study exhibited that the overexpression of EP2 and EGFR in ESCC tissues might play an important role in carcinogenesis and the progression of ESCC.
Collapse
|
11
|
Subramaniam D, Ponnurangam S, Ramamoorthy P, Standing D, Battafarano RJ, Anant S, Sharma P. Curcumin induces cell death in esophageal cancer cells through modulating Notch signaling. PLoS One 2012; 7:e30590. [PMID: 22363450 PMCID: PMC3281833 DOI: 10.1371/journal.pone.0030590] [Citation(s) in RCA: 179] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 12/19/2011] [Indexed: 12/21/2022] Open
Abstract
Background Curcumin inhibits the growth of esophageal cancer cell lines; however, the mechanism of action is not well understood. It is becoming increasingly clear that aberrant activation of Notch signaling has been associated with the development of esophageal cancer. Here, we have determined that curcumin inhibits esophageal cancer growth via a mechanism mediated through the Notch signaling pathway. Methodology/Principal Findings In this study, we show that curcumin treatment resulted in a dose and time dependent inhibition of proliferation and colony formation in esophageal cancer cell lines. Furthermore, curcumin treatment induced apoptosis through caspase 3 activation, confirmed by an increase in the ratio of Bax to Bcl2. Cell cycle analysis demonstrated that curcumin treatment induced cell death and down regulated cyclin D1 levels. Curcumin treatment also resulted in reduced number and size of esophagospheres. Furthermore, curcumin treatment led to reduced Notch-1 activation, expression of Jagged-1 and its downstream target Hes-1. This reduction in Notch-1 activation was determined to be due to the down-regulation of critical components of the γ-secretase complex proteins such as Presenilin 1 and Nicastrin. The combination of a known γ-secretase inhibitor DAPT and curcumin further decreased proliferation and induced apoptosis in esophageal cancer cells. Finally, curcumin treatment down-regulate the expressions of Notch-1 specific microRNAs miR-21 and miR-34a, and upregulated tumor suppressor let-7a miRNA. Conclusion/Significance Curcumin is a potent inhibitor of esophageal cancer growth that targets the Notch-1 activating γ-secretase complex proteins. These data suggest that Notch signaling inhibition is a novel mechanism of action for curcumin during therapeutic intervention in esophageal cancers.
Collapse
Affiliation(s)
- Dharmalingam Subramaniam
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * (D. Subramaniam); (PS)
| | - Sivapriya Ponnurangam
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Prabhu Ramamoorthy
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - David Standing
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Richard J. Battafarano
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Shrikant Anant
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Prateek Sharma
- Division of Gastroenterology and Hepatology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * (D. Subramaniam); (PS)
| |
Collapse
|
12
|
Woodward DF, Jones RL, Narumiya S. International Union of Basic and Clinical Pharmacology. LXXXIII: classification of prostanoid receptors, updating 15 years of progress. Pharmacol Rev 2011; 63:471-538. [PMID: 21752876 DOI: 10.1124/pr.110.003517] [Citation(s) in RCA: 332] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
It is now more than 15 years since the molecular structures of the major prostanoid receptors were elucidated. Since then, substantial progress has been achieved with respect to distribution and function, signal transduction mechanisms, and the design of agonists and antagonists (http://www.iuphar-db.org/DATABASE/FamilyIntroductionForward?familyId=58). This review systematically details these advances. More recent developments in prostanoid receptor research are included. The DP(2) receptor, also termed CRTH2, has little structural resemblance to DP(1) and other receptors described in the original prostanoid receptor classification. DP(2) receptors are more closely related to chemoattractant receptors. Prostanoid receptors have also been found to heterodimerize with other prostanoid receptor subtypes and nonprostanoids. This may extend signal transduction pathways and create new ligand recognition sites: prostacyclin/thromboxane A(2) heterodimeric receptors for 8-epi-prostaglandin E(2), wild-type/alternative (alt4) heterodimers for the prostaglandin FP receptor for bimatoprost and the prostamides. It is anticipated that the 15 years of research progress described herein will lead to novel therapeutic entities.
Collapse
Affiliation(s)
- D F Woodward
- Dept. of Biological Sciences RD3-2B, Allergan, Inc., 2525 Dupont Dr., Irvine, CA 92612, USA.
| | | | | |
Collapse
|
13
|
Pink RC, Bailey TA, Iputo JE, Sammon AM, Woodman AC, Carter DRF. Molecular basis for maize as a risk factor for esophageal cancer in a South African population via a prostaglandin E2 positive feedback mechanism. Nutr Cancer 2011; 63:714-21. [PMID: 21667399 DOI: 10.1080/01635581.2011.570893] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The incidence of squamous cancer of the esophagus varies up to a hundredfold in different regions of the world. In Transkei, South Africa, a particularly high incidence of the disease is observed. We have previously proposed an association between a maize-rich diet and elevated levels of intragastric prostaglandin E2 production (PGE(2)). Here we investigate the molecular mechanisms by which a high-maize diet could lead to increased incidence of squamous cancer of the esophagus. We confirm that levels of PGE(2) are high (606.8 pg/ml) in the gastric fluid of individuals from Transkei. We also show that treatment of esophageal cells with linoleic acid, which is found at high levels in maize and is a precursor to PGE(2), leads to increased cell proliferation. Similarly, treatment of cells with PGE(2) or with gastric fluid from Transkeians also leads to increased proliferation. Our data suggest that the high levels of PGE(2) associated with a maize-rich diet stimulate cell division and induce the enzyme COX 2, resulting in a positive feedback mechanism that predisposes the esophagus to carcinoma.
Collapse
Affiliation(s)
- Ryan C Pink
- Cranfield Health, Cranfield University, Cranfield, Bedfordshire, United Kingdom.
| | | | | | | | | | | |
Collapse
|
14
|
Rajasekaran SA. Therapeutic potential of curcumin in gastrointestinal diseases. World J Gastrointest Pathophysiol 2011; 2:1-14. [PMID: 21607160 PMCID: PMC3097964 DOI: 10.4291/wjgp.v2.i1.1] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 11/25/2010] [Accepted: 12/02/2010] [Indexed: 02/06/2023] Open
Abstract
Curcumin, also known as diferuloylmethane, is derived from the plant Curcuma longa and is the active ingredient of the spice turmeric. The therapeutic activities of curcumin for a wide variety of diseases such as diabetes, allergies, arthritis and other chronic and inflammatory diseases have been known for a long time. More recently, curcumin’s therapeutic potential for preventing and treating various cancers is being recognized. As curcumin’s therapeutic promise is being explored more systematically in various diseases, it has become clear that, due to its increased bioavailability in the gastrointestinal tract, curcumin may be particularly suited to be developed to treat gastrointestinal diseases. This review summarizes some of the current literature of curcumin’s anti-inflammatory, anti-oxidant and anti-cancer potential in inflammatory bowel diseases, hepatic fibrosis and gastrointestinal cancers.
Collapse
|
15
|
Abrahao AC, Castilho RM, Squarize CH, Molinolo AA, dos Santos-Pinto D, Gutkind JS. A role for COX2-derived PGE2 and PGE2-receptor subtypes in head and neck squamous carcinoma cell proliferation. Oral Oncol 2010; 46:880-7. [PMID: 20951077 DOI: 10.1016/j.oraloncology.2010.09.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Revised: 09/03/2010] [Accepted: 09/06/2010] [Indexed: 10/18/2022]
Abstract
The overexpression of cyclooxygenase (COX)-2 is a frequent event in squamous cell carcinomas of the head and neck (HNSCC), and non-steroidal anti-inflammatory drugs, which are potent inhibitors of COX-1 and COX-2, exert chemopreventive effects on HNSCC cancer development. COX-2 promotes the release of the pro-inflammatory mediator prostaglandin E2 (PGE2), which acts on its cell surface G protein-coupled receptors EP1, EP2, EP3, and EP4. Here, we investigated the role of PGE2 and its receptors in cellular proliferation in HNSCC. The expression of COX-2 and EP1-4 was examined in immortalized oral epithelial cells and in a representative panel of HNSCC cell lines, and based on these data EP1-EP3 and COX-2 expression were evaluated by immunohistochemistry in a large clinical sample collection using HNSCC tissue microarrays. The ability of selective COX-2 inhibition to block PGE2 secretion was measured by ELISA specific assays. The effects of PGE2 on cell proliferation were evaluated using PGE2, its stable analog, and EP2 and EP3-specific synthetic agonists. The results presented here show that HNSCC tumoral lesions and their derived cell lines constitutively express COX-2 and the EP1, EP2 and EP3 receptors for PGE2. HNSCC cells secrete PGE2, which can be suppressed by low concentrations of COX-2 selective inhibitors, without inhibiting cell proliferation. Exogenously added stable PGE2 and EP3-specific agonists induce DNA synthesis in all HNSCC cell lines tested. Overall, our study supports the emerging notion that PGE2 produced in the tumor microenvironment by the overexpression of COX-2 in tumoral and inflammatory cells may promote the growth of HNSCC cells in an autocrine and paracrine fashion by acting on PGE2 receptors that are widely expressed in most HNSCC cancer cells. In particular, our findings suggest that EP3 receptor may play a more prominent role in HNSCC cell growth promotion, thus providing a rationale for the future evaluation of this PGE2 receptor as a target for HNSCC prevention strategies.
Collapse
Affiliation(s)
- Aline Correa Abrahao
- Department of Oral Pathology, Dental School, University of São Paulo, Avenida Lineu Prestes, 2227, São Paulo, SP 05509-000, Brazil
| | | | | | | | | | | |
Collapse
|
16
|
Gao F, Brant KA, Ward RM, Cattley RT, Barchowsky A, Fabisiak JP. Multiple protein kinase pathways mediate amplified IL-6 release by human lung fibroblasts co-exposed to nickel and TLR-2 agonist, MALP-2. Toxicol Appl Pharmacol 2010; 247:146-57. [PMID: 20600219 DOI: 10.1016/j.taap.2010.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 06/03/2010] [Accepted: 06/10/2010] [Indexed: 12/16/2022]
Abstract
Microbial stimuli and atmospheric particulate matter (PM) interact to amplify the release of inflammatory and immune-modulating cytokines. The basis of this interaction, however, is not known. Cultured human lung fibroblasts (HLF) were used to determine whether various protein kinase pathways were involved in the release of IL-6 following combined exposure to the PM-derived metal, Ni, and M. fermentans-derived macrophage-activating lipopeptide 2 (MALP-2), a toll-like receptor 2 agonist. Synergistic release of IL-6 by MALP-2 and NiSO4 was obvious after 8h of co-stimulation and correlated with a late phase accumulation of IL-6 mRNA. Ni and MALP-2, alone or together, all led to rapid and transient phosphorylations of ERK(1/2) and JNK/SAPK of similar magnitude. p38 phosphorylation, however, was observed only after prolonged treatment of cells with both stimuli together. A constitutive level of PI3K-dependent Akt phosphorylation remained unchanged by Ni and/or MALP-2 exposure. IL-6 induced by Ni/MALP-2 co-exposure was partially dependent on activity of HIF-1alpha and COX-2 as shown by targeted knockdown using siRNA. IL-6 release in response to Ni/MALP-2 was partially sensitive to pharmacological inhibition of ERK(1/2), p38, and PI3K signaling. The protein kinase inhibitors had minimal or no effects on Ni/MALP-2-induced accumulation of HIF-1alpha protein, however, COX-2 expression and, more markedly PGE(2) production, were suppressed by LY294002, SB203580, and U0126. Thus, Ni/MALP-2 interactions involve multiple protein kinase pathways (ERK(1/2), p38, and PI3K) that modulate events downstream from the early accumulation of HIF-1alpha to promote IL-6 gene expression directly or secondarily, through COX-2-derived autocrine products like PGE(2).
Collapse
Affiliation(s)
- Fei Gao
- Department of Environmental & Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15219, USA
| | | | | | | | | | | |
Collapse
|
17
|
Cyclooxygenase-2 in tumorigenesis of gastrointestinal cancers: an update on the molecular mechanisms. Cancer Lett 2010; 295:7-16. [PMID: 20381235 DOI: 10.1016/j.canlet.2010.03.015] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Revised: 03/18/2010] [Accepted: 03/22/2010] [Indexed: 12/20/2022]
Abstract
The use of nonsteroidal anti-inflammatory drugs (NSAIDs) is associated with lower risks for esophageal, gastric and colon cancers as well as other solid tumors. The antitumor effect of NSAIDs is mediated through cyclooxygenase-2 (COX-2)-dependent and -independent regulation of oncogenic and tumor-suppressive pathways. Recent discoveries have shed new light on the regulation of COX-2 at the molecular level in these cancers. Moreover, prostaglandin E(2) (PGE(2)), a COX-2-derived eicosanoid, has been found to affect numerous tumorigenic processes. In this connection, PGE(2) activates multiple intracellular signaling pathways, including (1) transactivation of epidermal growth factor receptor (EGFR); (2) protein kinase C-dependent, EGFR-independent activation of extracellular signal-regulated kinase (ERK) and the transcription factors activator protein-1 and c-Myc; (3) G-protein-mediated activation of beta-catenin/TCF-dependent transcription. Activation of these signaling pathways by PGE(2) is mediated by EP receptors whose inhibitors suppress gastrointestinal carcinogenesis. Taken together, COX-2 expression is dysregulated in many types of cancer and COX-2-derived PGE(2) elicits multiple oncogenic signals to promote carcinogenesis. Targeting PGE(2) signaling by EP receptor antagonists holds promise for the development of targeted therapy for the treatment of cancer.
Collapse
|
18
|
Mauchley D, Meng X, Johnson T, Fullerton DA, Weyant MJ. Modulation of growth in human esophageal adenocarcinoma cells by group IIa secretory phospholipase A2. J Thorac Cardiovasc Surg 2010; 139:591-9; discussion 599. [DOI: 10.1016/j.jtcvs.2009.10.061] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 09/04/2009] [Accepted: 10/03/2009] [Indexed: 12/20/2022]
|
19
|
Prostaglandin E2promotes cell proliferationviaprotein kinase C/extracellular signal regulated kinase pathway-dependent induction of c-Myc expression in human esophageal squamous cell carcinoma cells. Int J Cancer 2009; 125:2540-6. [DOI: 10.1002/ijc.24607] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
20
|
Ge JH, Zhu YY, Liu YR, Jiang JJ, Dong J. siRNA-mediated downregulation of COX-2 gene expression alters the proliferation of hepatocellular carcinoma HepG2 cells. Shijie Huaren Xiaohua Zazhi 2009; 17:2244-2250. [DOI: 10.11569/wcjd.v17.i22.2244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the effects of siRNA-mediated downregulation of COX-2 gene expression on the proliferation of hepatocellular carcinoma HepG2 cells.
METHODS: HepG2 cells were divided into four groups: COX-2 siRNA intervention group, control siRNA intervention group, empty liposome group and blank control group. COX-2 siRNA was transfected into HepG2 cells by lipofection. Cell proliferation inhibition was measured at 24, 48 and 72 h after transfection by MTT (thiazolyl blue) assay. The changes in cell cycle distribution were detected at 24 h after transfection by flow cytometry. The expression levels of COX-2 and ERK1/2 mRNAs and proteins were detected at 24 h after transfection by semiquantitative RT-PCR and Western blot, respectively.
RESULTS: The proliferation inhibition rate achieved in the COX-2 siRNA intervention group was higher than those in the control siRNA intervention group and empty liposome group (67.08% vs 2.45% and 1.56%, both P < 0.01). The percentage of HepG2 cells arrested at G1 phase (DNA presynthetic phase) was increased in the COX-2 siRNA intervention group as compared to the control siRNA intervention group, empty liposome group and blank control group (72.80% vs 50.27%, 50.97% and 53.13%, all P < 0.05). Semiquantitative RT-PCR analysis showed that the expression levels of COX-2, ERK1 and ERK2 mRNAs in the COX-2 siRNA intervention group were significantly decreased as compared to the control siRNA intervention group, empty liposome group and blank control group (0.58 vs 0.83, 0.89 and 0.93; 0.32 vs 0.71, 0.64 and 0.76; 0.48 vs 0.65, 0.67 and 0.72; all P < 0.05). Similar results were obtained for protein expression as revealed by Western blot analysis.
CONCLUSION: siRNA can downregulate the expression of COX-2 gene in hepatocellular carcinoma HepG2 cells and inhibit their proliferation. COX-2 promotes the development of primary hepatic carcinoma perhaps through regulation of ERK1/2 pathway.
Collapse
|
21
|
Kuo KT, Wang HW, Chou TY, Hsu WH, Hsu HS, Lin CH, Wang LS. Prognostic role of PGE2 receptor EP2 in esophageal squamous cell carcinoma. Ann Surg Oncol 2008; 16:352-60. [PMID: 19050969 DOI: 10.1245/s10434-008-0242-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 10/27/2008] [Accepted: 10/28/2008] [Indexed: 12/18/2022]
Abstract
Prostaglandin E2 (PGE2), a major cyclooxygenase-2 (COX-2) product, has been shown to affect numerous tumorigenic processes. PGE2 acts through G-protein-coupled receptors designated as EPs. Recently it has been documented that PGE2 promotes colon cancer cell growth via EP2. However, the expression and the prognostic role of EP2 in esophageal squamous cell carcinoma (ESCC) remained unknown. From January 1995 to January 2001, tissue samples from 226 patients with ESCC who underwent esophagectomies at our institutions were collected and made into tissue core arrays for study. EP2 expression was examined by immunohistochemical staining and confirmed by Western blot. The clinicopathologic data were then analyzed. EP2 overexpression was observed in 43.4% (98/226) of ESCC. Overexpression of EP2 correlated positively with depth of tumor invasion (T status) (P = 0.016) and was associated with worse overall survival (P = 0.047). In patients without regional or distant lymph node metastasis (N0 or M0), EP2 overexpression was associated with worse overall survival (P = 0.033 and P = 0.003, respectively). Using Cox regression analysis, T status, N status, and M status were the independent factors of overall survival, but EP2 expression was not. However, when focusing on patients with T1-3N0M0 status, EP2 expression became an independent factor of overall survival (P = 0.048). Our results show that EP2 overexpression was associated with worse prognosis, and correlated positively with T status in ESCC. Meanwhile, among those patients at earlier stages, EP2 overexpression significantly disclosed patients at high risks for poor prognosis.
Collapse
Affiliation(s)
- Kuang-Tai Kuo
- Department of Surgery, Buddhist Tzu Chi General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|