1
|
Klischan MT, Mazzone F, Berning L, Greb J, Schlamkow M, Haase M, Frey W, Stork B, Pfeffer K, Pietruszka J. Modular Approach for the Synthesis and Bioactivity Profiling of 8,8'-Biflavones. ACS OMEGA 2023; 8:41816-41834. [PMID: 37970025 PMCID: PMC10634270 DOI: 10.1021/acsomega.3c06503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 11/17/2023]
Abstract
In this work, we report the scalable and modular synthesis of a library of 55 monomeric and dimeric flavonoids including 14 8,8'-biflavones. The sterically demanding tetra-ortho-substituted axis of an acetophenone dimer key intermediate was constructed in a regioselective manner using Fe-mediated oxidative coupling. This step was systematically optimized and performed on up to multigram scale. The biological activities of this compound library were evaluated, including cytotoxicity against healthy and malignant human cell lines, antimicrobial activity against the apicomplexan parasite Toxoplasma gondii, and antioxidant capacity. A marked increase in activity for the 8,8'-dimeric structures compared to that of their monomeric counterparts was observed. Several biflavones were identified with high selectivity indices (low cytotoxicity and high antiprotozoal activity), showing that this class of natural products may serve as lead structures for further investigations.
Collapse
Affiliation(s)
- Moritz
K. T. Klischan
- Institute
of Bioorganic Chemistry, Heinrich Heine
University Düsseldorf, Forschungszentrum Jülich, Stetternicher Forst, Geb.15.8, 52426 Jülich, Germany
| | - Flaminia Mazzone
- Institute
of Medical Microbiology and Hospital Hygiene, Medical Faculty and
University Hospital Düsseldorf, Heinrich
Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Lena Berning
- Institute
of Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Julian Greb
- Institute
of Bioorganic Chemistry, Heinrich Heine
University Düsseldorf, Forschungszentrum Jülich, Stetternicher Forst, Geb.15.8, 52426 Jülich, Germany
| | - Max Schlamkow
- Institute
of Bioorganic Chemistry, Heinrich Heine
University Düsseldorf, Forschungszentrum Jülich, Stetternicher Forst, Geb.15.8, 52426 Jülich, Germany
- Institut
für Bio- und Geowissenschaften (IBG-1: Bioorganische Chemie)
Forschungszentrum, 52428 Jülich, Germany
| | - Mona Haase
- Institute
of Bioorganic Chemistry, Heinrich Heine
University Düsseldorf, Forschungszentrum Jülich, Stetternicher Forst, Geb.15.8, 52426 Jülich, Germany
| | - Wolfgang Frey
- Institute
of Organic Chemistry, University of Stuttgart, 70569 Stuttgart, Germany
| | - Björn Stork
- Institute
of Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Klaus Pfeffer
- Institute
of Medical Microbiology and Hospital Hygiene, Medical Faculty and
University Hospital Düsseldorf, Heinrich
Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Jörg Pietruszka
- Institute
of Bioorganic Chemistry, Heinrich Heine
University Düsseldorf, Forschungszentrum Jülich, Stetternicher Forst, Geb.15.8, 52426 Jülich, Germany
- Institut
für Bio- und Geowissenschaften (IBG-1: Bioorganische Chemie)
Forschungszentrum, 52428 Jülich, Germany
| |
Collapse
|
2
|
Synthesis and Investigation of Flavanone Derivatives as Potential New Anti-Inflammatory Agents. Molecules 2022; 27:molecules27061781. [PMID: 35335145 PMCID: PMC8953636 DOI: 10.3390/molecules27061781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 11/17/2022] Open
Abstract
Flavonoids are polyphenols with broad known pharmacological properties. A series of 2,3-dihydroflavanone derivatives were thus synthesized and investigated for their anti-inflammatory activities. The target flavanones were prepared through cyclization of 2'-hydroxychalcone derivatives, the later obtained by Claisen-Schmidt condensation. Since nitric oxide (NO) represents an important inflammatory mediator, the effects of various flavanones on the NO production in the LPS-induced RAW 264.7 macrophage were assessed in vitro using the Griess test. The most active compounds were flavanone (4G), 2'-carboxy-5,7-dimethoxy-flavanone (4F), 4'-bromo-5,7-dimethoxy-flavanone (4D), and 2'-carboxyflavanone (4J), with IC50 values of 0.603, 0.906, 1.030, and 1.830 µg/mL, respectively. In comparison, pinocembrin achieved an IC50 value of 203.60 µg/mL. Thus, the derivatives synthesized in this work had a higher NO inhibition capacity compared to pinocembrin, demonstrating the importance of pharmacomodulation to improve the biological potential of natural molecules. SARs suggested that the use of a carboxyl-group in the meta-position of the B-ring increases biological activity, whereas compounds carrying halogen substituents in the para-position were less active. The addition of methoxy-groups in the meta-position of the A-ring somewhat decreased the activity. This study successfully identified new bioactive flavanones as promising candidates for the development of new anti-inflammatory agents.
Collapse
|
3
|
Biological Activity, Hepatotoxicity, and Structure-Activity Relationship of Kavalactones and Flavokavins, the Two Main Bioactive Components in Kava ( Piper methysticum). EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6851798. [PMID: 34471418 PMCID: PMC8405297 DOI: 10.1155/2021/6851798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/13/2021] [Indexed: 11/25/2022]
Abstract
Kava (Piper methysticum Forst) is a popular and favorable edible medicinal herb which was traditionally used to prepare a nonfermented beverage with relaxant beneficial for both social and recreational purposes. Numerous studies conducted on kava have confirmed the presence of kavalactones and flavokawains, two major groups of bioactive ingredients, in this miraculous natural plant. Expectedly, both kavalactone and flavokawain components exhibited potent antianxiety and anticancer activities, and their structure-activity relationships were also revealed. However, dozens of clinical data revealed the hepatotoxicity effect which is indirectly or directly associated with kava consumption, and most of the evidence currently seems to point the compounds of flavokawains in kava were responsible. Therefore, our aim is to conduct a systematic review of kavalactones and flavokawains in kava including their biological activities, structure-activity relationships, and toxicities, and as a result of our systematic investigations, suggestions on kava and its compounds are supplied for future research.
Collapse
|
4
|
Belkadi A, Kenouche S, Melkemi N, Daoud I, Djebaili R. K-means clustering analysis, ADME/pharmacokinetic prediction, MEP, and molecular docking studies of potential cytotoxic agents. Struct Chem 2021. [DOI: 10.1007/s11224-021-01796-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
5
|
Peppers: A "Hot" Natural Source for Antitumor Compounds. Molecules 2021; 26:molecules26061521. [PMID: 33802144 PMCID: PMC8002096 DOI: 10.3390/molecules26061521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 12/20/2022] Open
Abstract
Piper, Capsicum, and Pimenta are the main genera of peppers consumed worldwide. The traditional use of peppers by either ancient civilizations or modern societies has raised interest in their biological applications, including cytotoxic and antiproliferative effects. Cellular responses upon treatment with isolated pepper-derived compounds involve mechanisms of cell death, especially through proapoptotic stimuli in tumorigenic cells. In this review, we highlight naturally occurring secondary metabolites of peppers with cytotoxic effects on cancer cell lines. Available mechanisms of cell death, as well as the development of analogues, are also discussed.
Collapse
|
6
|
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: 3.4] [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
|
7
|
Melanogenic Inhibition and Toxicity Assessment of Flavokawain A and B on B16/F10 Melanoma Cells and Zebrafish ( Danio rerio). Molecules 2020; 25:molecules25153403. [PMID: 32731323 PMCID: PMC7436045 DOI: 10.3390/molecules25153403] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/14/2022] Open
Abstract
Excessive production of melanin implicates hyperpigmentation disorders. Flavokawain A (FLA) and flavokawain B (FLB) have been reported with anti-melanogenic activity, but their melanogenic inhibition and toxicity effects on the vertebrate model of zebrafish are still unknown. In the present study, cytotoxic as well as melanogenic effects of FLA and FLB on cellular melanin content and tyrosinase activity were evaluated in α-MSH-induced B16/F10 cells. Master regulator of microphthalmia-associated transcription factor (Mitf) and the other downstream melanogenic-related genes were verified via quantitative real time PCR (qPCR). Toxicity assessment and melanogenesis inhibition on zebrafish model was further observed. FLA and FLB significantly reduced the specific cellular melanin content by 4.3-fold and 9.6-fold decrement, respectively in α-MSH-induced B16/F10 cells. Concomitantly, FLA significantly reduced the specific cellular tyrosinase activity by 7-fold whilst FLB by 9-fold. The decrement of melanin production and tyrosinase activity were correlated with the mRNA suppression of Mitf which in turn down-regulate Tyr, Trp-1 and Trp-2. FLA and FLB exhibited non-toxic effects on the zebrafish model at 25 and 6.25 µM, respectively. Further experiments on the zebrafish model demonstrated successful phenotype-based depigmenting activity of FLA and FLB under induced melanogenesis. To sum up, our findings provide an important first key step for both of the chalcone derivatives to be further studied and developed as potent depigmenting agents.
Collapse
|
8
|
Yang Y, Wei Z, Teichmann AT, Wieland FH, Wang A, Lei X, Zhu Y, Yin J, Fan T, Zhou L, Wang C, Chen L. Development of a novel nitric oxide (NO) production inhibitor with potential therapeutic effect on chronic inflammation. Eur J Med Chem 2020; 193:112216. [PMID: 32208222 DOI: 10.1016/j.ejmech.2020.112216] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/07/2020] [Accepted: 03/07/2020] [Indexed: 02/08/2023]
Abstract
Inflammation is a complex biological response to stimuli. Activated macrophages induced excessively release of pro-inflammatory cytokines and mediators such as endogenous radical nitric oxide (NO) play a significant role in the progression of multiple inflammatory diseases. Both natural and synthetic chalcones possess a wide range of bioactivities. In this work, thirty-nine chalcones and three related compounds, including several novel ones, based on bioactive kava chalcones were designed, synthesized and their inhibitory effects on NO production in RAW 264.7 cells were evaluated. The novel compound (E)-1-(2'-hydroxy-4',6'-dimethoxyphenyl)-3-(3-methoxy-4-(3-morpholinopropoxy)phenyl)prop-2-en-1-one (53) exhibited a better inhibitory activity (84.0%) on NO production at 10 μM (IC50 = 6.4 μM) with the lowest cytotoxicity (IC50 > 80 μM) among the tested compounds. Besides, western blot analysis indicated that compound 53 was a potent down-regulator of inducible nitric oxide synthase (iNOS) protein. Docking study revealed that compound 53 also can dock into the active site of iNOS. Furthermore, at the dose of 10 mg/kg/day, compound 53 could both significantly suppress the progression of inflammation on collagen-induced arthritis (CIA) and adjuvant-induced arthritis (AIA) models. In addition, the structure-activity relationship (SAR) of the kava chalcones based analogs was also depicted.
Collapse
Affiliation(s)
- Youzhe Yang
- Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, PR China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, PR China; Natural Products Research Center, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, PR China.
| | - Zhe Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, PR China
| | - Alexander Tobias Teichmann
- Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, PR China
| | - Frank Heinrich Wieland
- Sichuan Provincial Center for Gynaecology and Breast Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, PR China
| | - Amu Wang
- School of Science, Xihua University, Chengdu, 610039, PR China
| | - Xiangui Lei
- School of Science, Xihua University, Chengdu, 610039, PR China
| | - Yue Zhu
- School of Science, Xihua University, Chengdu, 610039, PR China
| | - Jinxiang Yin
- School of Science, Xihua University, Chengdu, 610039, PR China
| | - Tiantian Fan
- Natural Products Research Center, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, PR China
| | - Li Zhou
- Natural Products Research Center, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, PR China
| | - Chao Wang
- Natural Products Research Center, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, PR China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, PR China.
| |
Collapse
|
9
|
Almeida JR, Moreira J, Pereira D, Pereira S, Antunes J, Palmeira A, Vasconcelos V, Pinto M, Correia-da-Silva M, Cidade H. Potential of synthetic chalcone derivatives to prevent marine biofouling. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 643:98-106. [PMID: 29936172 DOI: 10.1016/j.scitotenv.2018.06.169] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/06/2018] [Accepted: 06/13/2018] [Indexed: 06/08/2023]
Abstract
Biofouling represents a major economic, environmental and health concern for which new eco-friendly solutions are needed. International legislation has restricted the use of biocidal-based antifouling coatings, and increasing efforts have been applied in the search for environmentally friendly antifouling agents. This research work deals with the assessment of the interest of a series of synthetic chalcone derivatives for antifouling applications. Sixteen chalcone derivatives were synthesized with moderate yields (38-85%). Antifouling bioactivity of these compounds was assessed at different levels of biological organization using both anti-macrofouling and anti-microfouling bioassays, namely an anti-settlement assay using mussel (Mytilus galloprovincialis) larvae, as well as marine bacteria and microalgal biofilms growth inhibition bioassays. Results showed that three compounds (11, 12, and 16) were particularly active against the settlement of mussel larvae (EC50 7.24-34.63 μM), being compounds 12 and 16 also able to inhibit the growth of microfouling species (EC50 4.09-20.31 μM). Moreover, the most potent compounds 12 and 16 were found to be non-toxic to the non-target species Artemia salina (<10% mortality at 25 μM). A quantitative structure-activity relationship model predicted that descriptors describing the ability of molecules to form hydrogen bonds and encoding the shape, branching ratio and constitutional diversity of the molecule were implied in the antifouling activity against the settlement of mussel larvae. This work elucidates for the first time the relevance of synthesizing chalcone derivatives to generate new non-toxic products to prevent marine biofouling.
Collapse
Affiliation(s)
- J R Almeida
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal
| | - J Moreira
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - D Pereira
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - S Pereira
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal
| | - J Antunes
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal; Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, P 4069-007 Porto, Portugal
| | - A Palmeira
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal; Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - V Vasconcelos
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal; Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, P 4069-007 Porto, Portugal
| | - M Pinto
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal; Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - M Correia-da-Silva
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal; Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| | - H Cidade
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal; Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
10
|
Brandão P, Loureiro JB, Carvalho S, Hamadou MH, Cravo S, Moreira J, Pereira D, Palmeira A, Pinto M, Saraiva L, Cidade H. Targeting the MDM2-p53 protein-protein interaction with prenylchalcones: Synthesis of a small library and evaluation of potential antitumor activity. Eur J Med Chem 2018; 156:711-721. [PMID: 30041135 DOI: 10.1016/j.ejmech.2018.07.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/23/2018] [Accepted: 07/15/2018] [Indexed: 12/27/2022]
Abstract
Prenylation of several bioactive scaffolds is a very interesting strategy used in Medicinal Chemistry in order to improve biological/pharmacological effects. A small library of prenylchalcones was synthesized and evaluated for the ability to inhibit the MDM2-p53 interaction using a yeast-based assay. The capacity of all synthesized prenylchalcones and their non-prenylated precursors to inhibit the growth of human colon tumor HCT116 cells was also evaluated. The obtained results led to the identification of a hit compound, prenylchalcone 2e, which behaved as potential inhibitor of the MDM2-p53 interaction in yeast, and showed improved cytotoxicity against human tumor cells expressing wild-type p53, including liver hepatocellular carcinoma HepG2, breast adenocarcinoma MCF-7, and malignant melanoma A375 cells. In colon cancer cells, it was also shown that the growth inhibitory effect of prenylchalcone 2e was associated with the induction of cell cycle arrest, apoptosis, and increased protein expression levels of p53 transcriptional targets. Moreover, computational docking studies were performed in order to predict docking poses and residues involved in the MDM2-p53 potential interaction.
Collapse
Affiliation(s)
- Pedro Brandão
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Joana B Loureiro
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Sylvie Carvalho
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Meriem Hadjer Hamadou
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Sara Cravo
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/nm, 4450-208, Matosinhos, Portugal
| | - Joana Moreira
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Daniela Pereira
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Andreia Palmeira
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/nm, 4450-208, Matosinhos, Portugal
| | - Madalena Pinto
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/nm, 4450-208, Matosinhos, Portugal
| | - Lucília Saraiva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| | - Honorina Cidade
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal; Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/nm, 4450-208, Matosinhos, Portugal.
| |
Collapse
|
11
|
Takac P, Kello M, Pilatova MB, Kudlickova Z, Vilkova M, Slepcikova P, Petik P, Mojzis J. New chalcone derivative exhibits antiproliferative potential by inducing G2/M cell cycle arrest, mitochondrial-mediated apoptosis and modulation of MAPK signalling pathway. Chem Biol Interact 2018; 292:37-49. [PMID: 29981726 DOI: 10.1016/j.cbi.2018.07.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/04/2018] [Accepted: 07/03/2018] [Indexed: 02/07/2023]
Abstract
In the present study, we investigated antiproliferative activity of seven newly synthesized chalcone derivatives. Among tested compounds, (2 E)-3-(acridin-9-yl)-1-(2,6-dimethoxyphenyl)prop-2-en-1-one (1C) was the most potent with IC50 = 4.1 μmol/L in human colorectal HCT116 cells and was selected for further studies. Inhibition of cell proliferation was associated with cell cycle arrest in G2/M phase and dysregulation of α, α1 and β5 tubulins. Moreover, 1C caused disruption of the mitochondrial membrane potential and increased number of cells with sub G0/G1 DNA content which is considered as marker of apoptosis. Apoptosis was confirmed by annexin V/PI and AO/PI staining. Furthermore, we found increased concentration of cytochrome c, Smac/Diablo and increased caspase-3 and caspase-9 activity, cleavage of PARP as well as activation of DNA repair mechanisms in 1C-treated HCT116 cancer cells. Moreover this chalcone derivative up-regulated proapoptotic Bax expression and down-regulated antiapoptotic Bcl-2 and Bcl-xL expression. Additionally, 1C treatment led to modulation of MAPKs and Akt signalling pathways. In conclusion, our data showed ability of 1C to suppress cancel cell growth and provide the rationale for further in vivo study.
Collapse
Affiliation(s)
- Peter Takac
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Safarik University, 04011, Kosice, Slovak Republic
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Safarik University, 04011, Kosice, Slovak Republic
| | - Martina Bago Pilatova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Safarik University, 04011, Kosice, Slovak Republic
| | - Zuzana Kudlickova
- Department of Organic Chemistry, Faculty of Science, Pavol Jozef Safarik University, 040 01, Kosice, Slovak Republic; Department of Chemistry, Biochemistry and Biophysics, University of Veterinary Medicine and Pharmacy, 04181, Košice, Slovak Republic
| | - Maria Vilkova
- Department of Organic Chemistry, Faculty of Science, Pavol Jozef Safarik University, 040 01, Kosice, Slovak Republic
| | - Pavlina Slepcikova
- Department of Organic Chemistry, Faculty of Science, Pavol Jozef Safarik University, 040 01, Kosice, Slovak Republic
| | - Peter Petik
- Department of Pathology, Pavol Jozef Safarik University, 040 01, Kosice, Slovak Republic
| | - Jan Mojzis
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Safarik University, 04011, Kosice, Slovak Republic.
| |
Collapse
|