1
|
Emerging roles of hnRNP A2B1 in cancer and inflammation. Int J Biol Macromol 2022; 221:1077-1092. [PMID: 36113587 DOI: 10.1016/j.ijbiomac.2022.09.104] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/27/2022] [Accepted: 09/11/2022] [Indexed: 11/05/2022]
Abstract
Heterogeneous nuclear ribonucleoproteins (hnRNPs) are a group of RNA-binding proteins with important roles in multiple aspects of nucleic acid metabolism, including the packaging of nascent transcripts, alternative splicing, transactivation of gene expression, and regulation of protein translation. As a core component of the hnRNP complex in mammalian cells, heterogeneous nuclear ribonucleoprotein A2B1 (hnRNP A2B1) participates in and coordinates various molecular events. Given its regulatory role in inflammation and cancer progression, hnRNP A2B1 has become a novel player in immune response, inflammation, and cancer development. Concomitant with these new roles, a surprising number of mechanisms deemed to regulate hnRNP A2B1 functions have been identified, including post-translational modifications, changes in subcellular localization, direct interactions with multiple DNAs, RNAs, and proteins or the formation of complexes with them, which have gradually made hnRNP A2B1 a molecular target for multiple drugs. In light of the rising interest in the intersection between cancer and inflammation, this review will focus on recent knowledge of the biological roles of hnRNP A2B1 in cancer, immune response, and inflammation.
Collapse
|
2
|
Che L, Wu JS, Du ZB, He YQ, Yang L, Lin JX, Lei Z, Chen XX, Guo DB, Li WG, Lin YC, Lin ZN. Targeting Mitochondrial COX-2 Enhances Chemosensitivity via Drp1-Dependent Remodeling of Mitochondrial Dynamics in Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14030821. [PMID: 35159089 PMCID: PMC8834292 DOI: 10.3390/cancers14030821] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/27/2022] [Accepted: 02/03/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary New therapeutic strategies are urgently needed to improve the anti-cancer effect for hepatocellular carcinoma (HCC). Overexpression of cyclooxygenase-2 (COX-2) is found in several types of cancers and correlates with a poor prognosis. However, it remains unclear how the mitochondrial translocation of COX-2 is involved in mitochondrial dynamics and sensitizes HCC cells to multipattern anti-tumor therapy. We explored the impact of targeting mitochondrial COX-2 (mito-COX-2) intervention toward mitochondrial dynamics on platinum-based chemotherapeutics in HCC cells and xenograft nude mouse models. Our study indicates that the mito-COX-2 represents a candidate predictive biomarker and potential target to regulate anti-cancer sensitization of HCC, and possibly for other types of COX-2-high-expression cancers. Abstract Mitochondria are highly dynamic organelles and undergo constant fission and fusion, which are both essential for the maintenance of cell physiological functions. Dysregulation of dynamin-related protein 1 (Drp1)-dependent mitochondrial dynamics is associated with tumorigenesis and the chemotherapeutic response in hepatocellular carcinoma (HCC). The enzyme cyclooxygenase-2 (COX-2) is overexpressed in most cancer types and correlates with a poor prognosis. However, the roles played by the translocation of mitochondrial COX-2 (mito-COX-2) and the interaction between mito-COX-2 and Drp1 in chemotherapeutic responses remain to be elucidated in the context of HCC. Bioinformatics analysis, paired HCC patient specimens, xenograft nude mice, immunofluorescence, transmission electron microscopy, molecular docking, CRISPR/Cas9 gene editing, proximity ligation assay, cytoplasmic and mitochondrial fractions, mitochondrial immunoprecipitation assay, and flow cytometry analysis were performed to evaluate the underlying mechanism of how mito-COX-2 and p-Drp1Ser616 interaction regulates the chemotherapeutic response via mitochondrial dynamics in vitro and in vivo. We found that COX-2 and Drp1 were frequently upregulated and confer a poor prognosis in HCC. We also found that the proportion of mito-COX-2 and p-Drp1Ser616 was increased in HCC cell lines. In vitro, we demonstrated that the enhanced mitochondrial translocation of COX-2 promotes its interaction with p-Drp1Ser616 via PTEN-induced putative kinase 1 (PINK1)-mediated Drp1 phosphorylation activation. This increase was associated with higher colony formation, cell proliferation, and mitochondrial fission. These findings were confirmed by knocking down COX-2 in HCC cells using CRISPR/Cas9 technology. Furthermore, inhibition of Drp1 using pharmacologic inhibitors (Mdivi-1) or RNA interference (siDNM1L) decreased mito-COX-2/p-Drp1Ser616 interaction-mediated mitochondrial fission, and increased apoptosis in HCC cells treated with platinum drugs. Moreover, inhibiting mito-COX-2 acetylation with the natural phytochemical resveratrol resulted in reducing cell proliferation and mitochondrial fission, occurring through upregulation of mitochondrial deacetylase sirtuin 3 (SIRT3), which, in turn, increased the chemosensitivity of HCC to platinum drugs in vitro and in vivo. Our results suggest that targeting interventions to PINK1-mediated mito-COX-2/p-Drp1Ser616-dependent mitochondrial dynamics increases the chemosensitivity of HCC and might help us to understand how to use the SIRT3-modulated mito-COX-2/p-Drp1Ser616 signaling axis to develop an effective clinical intervention in hepatocarcinogenesis.
Collapse
Affiliation(s)
- Lin Che
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China; (L.C.); (J.-S.W.); (Z.-B.D.); (Y.-Q.H.); (L.Y.); (J.-X.L.); (Z.L.); (X.-X.C.); (D.-B.G.)
| | - Jia-Shen Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China; (L.C.); (J.-S.W.); (Z.-B.D.); (Y.-Q.H.); (L.Y.); (J.-X.L.); (Z.L.); (X.-X.C.); (D.-B.G.)
| | - Ze-Bang Du
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China; (L.C.); (J.-S.W.); (Z.-B.D.); (Y.-Q.H.); (L.Y.); (J.-X.L.); (Z.L.); (X.-X.C.); (D.-B.G.)
| | - Yu-Qiao He
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China; (L.C.); (J.-S.W.); (Z.-B.D.); (Y.-Q.H.); (L.Y.); (J.-X.L.); (Z.L.); (X.-X.C.); (D.-B.G.)
| | - Lei Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China; (L.C.); (J.-S.W.); (Z.-B.D.); (Y.-Q.H.); (L.Y.); (J.-X.L.); (Z.L.); (X.-X.C.); (D.-B.G.)
| | - Jin-Xian Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China; (L.C.); (J.-S.W.); (Z.-B.D.); (Y.-Q.H.); (L.Y.); (J.-X.L.); (Z.L.); (X.-X.C.); (D.-B.G.)
| | - Zhao Lei
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China; (L.C.); (J.-S.W.); (Z.-B.D.); (Y.-Q.H.); (L.Y.); (J.-X.L.); (Z.L.); (X.-X.C.); (D.-B.G.)
| | - Xiao-Xuan Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China; (L.C.); (J.-S.W.); (Z.-B.D.); (Y.-Q.H.); (L.Y.); (J.-X.L.); (Z.L.); (X.-X.C.); (D.-B.G.)
| | - Dong-Bei Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China; (L.C.); (J.-S.W.); (Z.-B.D.); (Y.-Q.H.); (L.Y.); (J.-X.L.); (Z.L.); (X.-X.C.); (D.-B.G.)
| | - Wen-Gang Li
- Department of Hepatobiliary Surgery and Pancreatic & Organ Transplantation Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China;
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yu-Chun Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China; (L.C.); (J.-S.W.); (Z.-B.D.); (Y.-Q.H.); (L.Y.); (J.-X.L.); (Z.L.); (X.-X.C.); (D.-B.G.)
- Correspondence: (Y.-C.L.); (Z.-N.L.); Tel.: +86-592-2880615 (Y.-C.L.); Fax: +86-592-2881578 (Y.-C.L.)
| | - Zhong-Ning Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China; (L.C.); (J.-S.W.); (Z.-B.D.); (Y.-Q.H.); (L.Y.); (J.-X.L.); (Z.L.); (X.-X.C.); (D.-B.G.)
- Correspondence: (Y.-C.L.); (Z.-N.L.); Tel.: +86-592-2880615 (Y.-C.L.); Fax: +86-592-2881578 (Y.-C.L.)
| |
Collapse
|
3
|
Afsharirad T, Tahmasvand R, Amini M, Daraei B, Salimi M. Two novel anticancer compounds with minimum cardiotoxic property. BMC Pharmacol Toxicol 2020; 21:79. [PMID: 33213519 PMCID: PMC7678303 DOI: 10.1186/s40360-020-00457-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/02/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although two novel synthesized compounds with tri-aryl structures; 3-(4-chlorophenyl)-5-(4-fluorophenyl)-4-phenyl-4,5-dihydro-1,2,4-oxadiazole (A) and 3,5-bis-(4-chlorophenyl)-4-phenyl-4,5-dihydro-1,2,4-oxadiazole (B) have been previously demonstrated to possess remarkable anti-breast cancer activity, their cardiotoxicity remains a major concern due to their mechanism of action. To address this concern, we assessed the ability of these compounds to cause toxicity towards H9c2 cardiomyocytes as an in vitro model of cardiotoxicity. METHODS Cytotoxic activity of both compounds was explored in vitro on H9c2 cells using MTT assay. Annexin V/PI method, intracellular ROS determination and mitochondrial membrane potential assay were applied to elucidate the mechanism of action of the cell death. RESULTS MTT assay revealed a concentration- and time-dependent cardiotoxicity. Findings of apoptosis by double staining with annexin V and propidium iodide divulged no cell death including apoptosis and necrosis at the concentration that were effective to inhibit cancer cells proliferation (10 μM) at 24 and 48 h. Furthermore, flow cytometric measurement of membrane potential and ROS determination using DCFH-DA verified the safe concentration of the compounds against H9c2 cells with no cardiotoxic effect. However, the higher concentration of the compounds could induce cell death through ROS-mediated mitochondrial dysfunction. CONCLUSIONS Altogether, the results represented two novel chemical molecules possessing anti-breast cancer activity with minimum cardiac side effect.
Collapse
Affiliation(s)
- Tayebeh Afsharirad
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Physiology and Pharmacology Department, Pasteur Institute of Iran, P.O. Box 13164, Tehran, Iran
| | - Raheleh Tahmasvand
- Physiology and Pharmacology Department, Pasteur Institute of Iran, P.O. Box 13164, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Teharn, Iran
| | - Bahram Daraei
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mona Salimi
- Physiology and Pharmacology Department, Pasteur Institute of Iran, P.O. Box 13164, Tehran, Iran.
| |
Collapse
|
4
|
Tahmasvand R, Bayat P, Vahdaniparast SM, Dehghani S, Kooshafar Z, Khaleghi S, Almasirad A, Salimi M. Design and synthesis of novel 4-thiazolidinone derivatives with promising anti-breast cancer activity: Synthesis, characterization, in vitro and in vivo results. Bioorg Chem 2020; 104:104276. [PMID: 32992280 DOI: 10.1016/j.bioorg.2020.104276] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/08/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022]
Abstract
Novel lead compounds as anticancer agents with the ability to circumvent emerging drug resistance have recently gained a great deal of interest. Thiazolidinones are among such compounds with well-established biological activity in the field of oncology. Here, we designed, synthesized and characterized a series of thiazolidinone structures (8a-8k). The results of anti-proliferative assay led to the discovery of compound 8j with a high potent cytotoxic effect using colon, liver and breast cancer cells. Furthermore, MDA-MB-231 and 4T1 cell lines were used to represent triple negative breast cancer (TNBC). Next, a number of in vitro and in vivo evaluations were carried out to demonstrate the potential activity against TNBC and also elucidate the possible mechanism of cell death induction. Our in vitro outcomes exhibited an impressive anticancer activity for compound 8j toward MDA-MB-231 cells through inducing apoptosis and a remarkable anti-metastatic feature via suppressing MMP-9 expression as well. Consistently, the in vivo and immunohistopathologic evaluations demonstrated that this compound significantly inhibited the 4T1 induced tumor growth and its metastasis to the lung. Altogether, among numerous thiazolidinone derivatives, compound 8j might represent a promising anticancer agent for TNBC, which is a major concern in the developed and developing countries.
Collapse
Affiliation(s)
- Raheleh Tahmasvand
- Department of Medical Biotechnology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Peyman Bayat
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Seyyed Mahmood Vahdaniparast
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Soudeh Dehghani
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Kooshafar
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Sepideh Khaleghi
- Department of Medical Biotechnology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Almasirad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mona Salimi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
5
|
Afsharinasab A, Moayer F, Amini M, Choopani S, Tahmasvand R, dehghani S, Mousavi SZ, Salimi M. Two Novel Compounds with Tri-aryl Structures as Effective Anti-Breast Cancer Candidates In-vivo. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 19:145-152. [PMID: 33224219 PMCID: PMC7667539 DOI: 10.22037/ijpr.2019.111802.13366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Prognosis of metastatic breast cancer is very poor which urges the necessity to develop novel potential drug candidates. We assessed two compounds with tri-aryl structures (A and B) for their potency to reduce primary breast tumor growth and lung metastasis in 4T1 mice model. MTT assay, 4T1 mammary mouse model, and immunohistochemistry experiments were used in this study. In-vitro results exhibited an anti-proliferative effect for compounds A and B towards MDA-MB-231 cancer cells. Our in-vivo results displayed that administered compounds A and B could suppress the size of the primary tumor and the number of lung metastatic foci in 4T1 BALB/c mice model. Histopathological analysis revealed that treatment of both compounds resulted in necrosis. Our findings provide new evidence that compound B may be promising for slowing the growth of tumor along with metastatic foci via COX-2 independent pathway.
Collapse
Affiliation(s)
- Ahoo Afsharinasab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran (IAUPS).
- Department of Physiology and pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| | - Fariborz Moayer
- Department of Pathobiology, College of Veterinary Medicine, Karaj Branch, Islamic Azad University, Alborz, Iran.
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Samira Choopani
- Department of Physiology and pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| | - Raheleh Tahmasvand
- Department of Physiology and pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| | - Soudeh dehghani
- Department of Physiology and pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| | - Seyede Zahra Mousavi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran (IAUPS).
| | - Mona Salimi
- Department of Physiology and pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
6
|
Moradi K, Barneh F, Irian S, Amini M, Moradpoor R, Amanzadeh A, Choopani S, Rahimi H, Ghodselahi T, Boujar MM, Salimi M. Two Novel Tri-Aryl Derivatives Attenuate the Invasion-Promoting Effects of Stromal Mesenchymal Stem Cells on Breast Cancer. Anticancer Agents Med Chem 2019; 19:1002-1011. [DOI: 10.2174/1871520619666190212123912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/28/2018] [Accepted: 02/04/2019] [Indexed: 01/06/2023]
Abstract
Background:The concept of Epithelial-Mesenchymal Transition (EMT) to promote carcinoma progression has been recognized as a venue for research on novel anticancer drugs. Triaryl template-based structures are one of the pivotal structural features found in a number of compounds with a wide variety of biological properties including anti-breast cancer. Among the various factors triggering EMT program, cyclooxygenase-2 (COX-2), NF-κB as well as the transforming growth factor-beta (TGF-β) have been widely investigated.Objective:Here, we aim to investigate the effect of two novel compounds A and B possessing triaryl structures, which interact with both COX-2 and TGF-β active sites and suppress NF-κB activation, on EMT in a co-culture system with breast cancer and stromal cells.Methods:MDA-MB-231 and bone-marrow mesenchymal stem (BM-MS) cells were co-cultured in a trans-well plate. Migration, matrigel-based invasion and colony formation in soft agar assays along with Real- time PCR and Western blot analysis were performed to examine the effect of compounds A and B on the invasive properties of MDA-MB-231 cells after 72 hours of co-culturing with BM-MSCs. In addition, TGF-beta interaction was investigated by Localized Surface Plasmon Resonance (LSPR).Results:BM-MSCs enhanced migration, invasion and anchorage-independent growth of the co-cultured MDAMB- 231 cells. A reduction in E-cadherin level concomitant with an increase in vimentin and N-cadherin levels following the co-culture implied EMT as the underlying process. Compounds A and B inhibited invasion and anchorage-independent growth of breast cancer cells co-cultured with BM-MSCs at 10µM. The observed inhibitory effects along with an increase in E-cadherin and a reduction in vimentin and ZEB2 levels suggest that the anti-invasive properties of compounds A and B might proceed through the blockade of stromal cell-induced EMT, mediated by their interaction with TGF-beta.Conclusion:These findings introduce compounds A and B as novel promising agents, which prevent EMT in invasive breast cancer cells.
Collapse
Affiliation(s)
- Khadijeh Moradi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Farnaz Barneh
- Department of Basic Sciences, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Irian
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mohsen Amini
- Medicinal Chemistry Department, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Raheleh Moradpoor
- Department of Basic Sciences, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Amanzadeh
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Samira Choopani
- Physiology and Pharmacology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Hamzeh Rahimi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | - Massoud M. Boujar
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mona Salimi
- Physiology and Pharmacology Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
7
|
Ren SZ, Wang ZC, Zhu XH, Zhu D, Li Z, Shen FQ, Duan YT, Cao H, Zhao J, Zhu HL. Design and biological evaluation of novel hybrids of 1, 5-diarylpyrazole and Chrysin for selective COX-2 inhibition. Bioorg Med Chem 2018; 26:4264-4275. [DOI: 10.1016/j.bmc.2018.07.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/28/2018] [Accepted: 07/12/2018] [Indexed: 12/20/2022]
|
8
|
Abdelazeem AH, El-Saadi MT, Said EG, Youssif BGM, Omar HA, El-Moghazy SM. Novel diphenylthiazole derivatives with multi-target mechanism: Synthesis, docking study, anticancer and anti-inflammatory activities. Bioorg Chem 2017; 75:127-138. [PMID: 28938224 DOI: 10.1016/j.bioorg.2017.09.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/07/2017] [Accepted: 09/11/2017] [Indexed: 12/22/2022]
Abstract
Over the last few decades, a growing body of studies addressed the anticancer activity of NSAIDs, particularly selective COX-2 inhibitors. However, their exact molecular mechanism is still unclear and is not fully investigated. In this regard, a novel series of compounds bearing a COXs privilege scaffold, diphenyl thiazole, was synthesized and evaluated for their anticancer activity against a panel of cancer cell lines. The most active compounds 10b, 14a,b, 16a, 17a,b and 18b were evaluated in vitro for COX-1/COX-2 inhibitory activity. These compounds were suggested to exert their anticancer activity through a multi-target mechanism based on their structural features. Thus, compounds 10b and 17b with the least IC50 values in MTT assay were tested against three known anticancer targets; EGFR, BRAF and tubulin. Compounds 10b and 17b showed remarkable activity against EGFR with IC50 values of 0.4 and 0.2μM, respectively and good activity against BRAF with IC50 values of 1.3 and 1.7μM, respectively. In contrast, they showed weak activity in tubulin polymerization assay. The in vivo anti-inflammatory potential was assessed and interestingly, compound 17b was the most potent compound. Together, this study offers some important insights into the correlation between COXs inhibition and cancer treatment. Additionally, the results demonstrated the promising activity of these compounds with a multi-target mechanism as good candidates for further development into potential anticancer agents.
Collapse
Affiliation(s)
- Ahmed H Abdelazeem
- Department of Medicinal Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Mohammed T El-Saadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Eman G Said
- Department of Medicinal Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Bahaa G M Youssif
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt; Department of Pharmaceutical Chemistry, College of Pharmacy, Aljouf University, Aljouf, Sakaka 2014, Saudi Arabia
| | - Hany A Omar
- Sharjah Institute for Medical Research and College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Pharmacology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Samir M El-Moghazy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| |
Collapse
|
9
|
Lu XY, Wang ZC, Ren SZ, Shen FQ, Man RJ, Zhu HL. Coumarin sulfonamides derivatives as potent and selective COX-2 inhibitors with efficacy in suppressing cancer proliferation and metastasis. Bioorg Med Chem Lett 2016; 26:3491-8. [PMID: 27349331 DOI: 10.1016/j.bmcl.2016.06.037] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/12/2016] [Accepted: 06/15/2016] [Indexed: 12/21/2022]
Abstract
Cyclooxygenase-2 is frequently overexpression in malignant tumors and the product PGE2 promotes cancer cell progression and metastasis. We designed novel series of coumarin sulfonamides derivatives to improve biological activities of COX-2 inhibition and anticancer. Among them, compound 7t showed most powerful selective inhibitory and antiproliferative activity (IC50=0.09μM for COX-2, IC50=48.20μM for COX-1, IC50=0.36μM against HeLa cells), comparable to the control positive compound Celecoxib (0.31μM, 43.37μM, 7.79μM). Cancer cell apoptosis assay were performed and results indicated that compound 7t effectively fuels HeLa cells apoptosis in a dose and time-dependent manner. Moreover, 7t could significantly suppress cancer cell adhesion, migration and invasion which were essential process of cancer metastasis. Docking simulations results was further indicated that compound 7t could bind well to the COX-2 active site and guided a reasonable design of selective COX-2 inhibitor with anticancer activities in future.
Collapse
Affiliation(s)
- Xiao-Yuan Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, People's Republic of China
| | - Zhong-Chang Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, People's Republic of China
| | - Shen-Zhen Ren
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, People's Republic of China
| | - Fa-Qian Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, People's Republic of China
| | - Ruo-Jun Man
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, People's Republic of China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, People's Republic of China.
| |
Collapse
|
10
|
Norouzi S, Norouzi M, Amini M, Amanzadeh A, Nabiuni M, Irian S, Salimi M. Two COX-2 inhibitors induce apoptosis in human erythroleukemia K562cells by modulating NF-κB and FHC pathways. ACTA ACUST UNITED AC 2016; 24:1. [PMID: 26739353 PMCID: PMC4704250 DOI: 10.1186/s40199-015-0139-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/18/2015] [Indexed: 11/20/2022]
Abstract
Background Leukemia is distinguished by abnormal proliferation of leukocytes. Although there has been some progress in developing novel cancer therapies, no significant improvement was observed in the overall survival rate over the last decade. Selective cyclooxygenase-2 (COX-2) inhibitors are known to inhibit tumor growth by exerting antimetastatic and antiangiogenic effects through inhibition of COX –dependent and independent pathways. The ability of two new triaryl-oxadiazole derivatives, compounds A (3-(4-chlorophenyl) -5-(4-flurophenyl)-4-Phenyl-4,5-dihydro-1,2,4-oxadiazole) and B (3,5-bis(4-chlorophenyl)-4-Phenyl-4,5-dihydro-1,2,4-oxadiazole), to induce apoptosis in human erythroleukemia K562 cells was evaluated and the upstream mechanism was investigated. Methods K562 cells were treated with compounds A and B at their IC50 concentrations and analyzed by DAPI staining and Annexin-V-FLUOS labelling solution. Nuclear factor kappa-B (NF-κB) activation was evaluated by TransAM kit. Cyclooxygenase-2 (COX-2), Caspase-3, Bax, Bcl-2, ferritin heavy chain (FHC), extra cellular signal-regulated kinase (ERK), p-ERK and early growth response protein-1 (Egr1) levels were determined using Western blotting, while c-Myc mRNA level was investigated by RT-PCR. Results Changes in nuclear morphology and the increased annexin-V/PI staining revealed the apoptotic cell death in compounds A- and B-treated K562 cells. A significant reduction in NF-κB activity as well as FHC and p-ERK levels were detected in these cells. No change was observed in the levels of Bax, Bcl-2, Caspase-3, COX-2, c-Myc and Egr1, following treatment with the two compounds. Collectively, compounds A and B potentiate apoptosis as shown by DAPI staining, flowcytometry, FHC and p-ERK downregulation and NF-κB inactivation. Conclusion Two compounds induce apoptosis in a COX-2-independent manner which also appears to be independent from mitochondria, caspase and c-Myc/Egr1 pathways.
Collapse
Affiliation(s)
- Shaghayegh Norouzi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, P.O. Box 1481765544, Tehran, Iran
| | - Mahnaz Norouzi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, P.O. Box 1481765544, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Amanzadeh
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Mohamad Nabiuni
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, P.O. Box 1481765544, Tehran, Iran
| | - Saeed Irian
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, P.O. Box 1481765544, Tehran, Iran.
| | - Mona Salimi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, P.O. Box 13164, Tehran, Iran.
| |
Collapse
|
11
|
Xuan Y, Wang J, Ban L, Lu JJ, Yi C, Li Z, Yu W, Li M, Xu T, Yang W, Tang Z, Tang R, Xiao X, Meng S, Chen Y, Liu Q, Huang W, Guo W, Cui X, Deng W. hnRNPA2/B1 activates cyclooxygenase-2 and promotes tumor growth in human lung cancers. Mol Oncol 2015; 10:610-24. [PMID: 26774881 DOI: 10.1016/j.molonc.2015.11.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/20/2015] [Accepted: 11/19/2015] [Indexed: 12/23/2022] Open
Abstract
Cyclooxygenase-2 (COX-2) is highly expressed in tumor cells and has been regarded as a hallmarker for cancers, but the excise regulatory mechanism of COX-2 in tumorigenesis remains largely unknown. Here, we pulled down and identified a novel COX-2 regulator, heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2/B1), which could specifically bind to COX-2 core promoter and regulate tumor growth in non-small-cell lung cancers (NSCLCs). Knockdown of hnRNPA2/B1 by shRNA or siRNA downregulated COX-2 expression and prostaglandin E2 (PGE2) production, and suppressed tumor cell growth in NSCLC cells in vitro and in vivo. Conversely, overexpression of hnRNPA2/B1 up-regulated the levels of COX-2 and PGE2 and promoted tumor cell growth. We also showed that hnRNPA2/B1 expression was positively correlated with COX-2 expression in NSCLC cell lines and tumor tissues, and the up-regulated expression of hnRNPA2/B1 and COX-2 predicted worse prognosis in NSCLC patients. Furthermore, we demonstrated that the activation of COX-2 expression by hnRNPA2/B1 was mediated through the cooperation with p300, a transcriptional co-activator, in NSCLC cells. The hnRNPA2/B1 could interact with p300 directly and be acetylated by p300. Exogenous overexpression of p300, but not its histone acetyltransferase (HAT) domain deletion mutation, augmented the acetylation of hnRNPA2/B1 and enhanced its binding on COX-2 promoter, thereby promoted COX-2 expression and lung cancer cell growth. Collectively, our results demonstrate that hnRNPA2/B1 promotes tumor cell growth by activating COX-2 signaling in NSCLC cells and imply that the hnRNPA2/B1/COX-2 pathway may be a potential therapeutic target for human lung cancers.
Collapse
Affiliation(s)
- Yang Xuan
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China; The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Jingshu Wang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Liying Ban
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Jian-Jun Lu
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Canhui Yi
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Zhenglin Li
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Wendan Yu
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Mei Li
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Tingting Xu
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Wenjing Yang
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Zhipeng Tang
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Ranran Tang
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Xiangsheng Xiao
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Songshu Meng
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Yiming Chen
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Quentin Liu
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China; The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China
| | - Wenlin Huang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China; State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, PR China
| | - Wei Guo
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China.
| | - Xiaonan Cui
- The First Affiliated Hospital & Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China.
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China; State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, PR China.
| |
Collapse
|