Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Enzyme Inhib Med Chem ; 38(1): 166-175, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36330714

ABSTRACT

Although various dual-target tubulin inhibitors have been designed and synthesised, no dual tubulin-NEDDylation inhibitors as antiproliferative agents were reported so far. In this work, a series of trimethoxyphenyl analogues as potential dual tubulin-NEDDylation inhibitors were synthesised and evaluated for their antiproliferative activity. Among them, compound C11 exhibited the most potent inhibitory activity with IC50 values of 1.17, 2.48, and 1.47 µM against HepG2, PC3, and MCF7 cells, respectively. In addition, it displayed the potent inhibitory activity against tubulin with an IC50 value of 2.40 µM and obviously inhibited tubulin polymerisation in HepG2 cells. Furthermore, C11 inhibited NEDDylation by a ATP-dependent manner. Molecular docking studies revealed that the methoxy group and dithiocarbamate group of C11 could form hydrogen bonds with residues of tubulin and E1 NEDD8-activating enzyme (NAE). These results suggested that compound C11 was a dual tubulin-NEDDylation inhibitor with antiproliferative activity.


Subject(s)
Antineoplastic Agents , Tubulin Modulators , Humans , Tubulin Modulators/pharmacology , Tubulin Modulators/chemistry , Tubulin/metabolism , Drug Screening Assays, Antitumor , Molecular Docking Simulation , Structure-Activity Relationship , Molecular Structure , Dose-Response Relationship, Drug , Cell Proliferation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor
2.
Bioorg Chem ; 107: 104634, 2021 02.
Article in English | MEDLINE | ID: mdl-33476867

ABSTRACT

A series of novel indole derivatives were synthesized and evaluated for their antiproliferative activity against three selected cancer cell lines (MGC803, EC-109 and PC-3). Among these analogues, 2-(5-methoxy-1H-indol-1-yl)-N-(4-methoxybenzyl)-N-(3,4,5-trimethoxyphenyl)acetamide (V7) showed the best inhibitory activity against MGC803 cells with an IC50 value of 1.59 µM. Cellular mechanisms elucidated that V7 inhibited colony formation, induced apoptosis and arrested cell cycle at G2/M phase. Importantly, indole analogue V7 inhibited NEDDylation pathway and MAPK pathway against MGC803 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/chemistry , Signal Transduction/drug effects , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Drug Design , Drug Evaluation, Preclinical , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Indoles/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Structure-Activity Relationship , Ubiquitin-Conjugating Enzymes/metabolism
3.
Environ Toxicol ; 36(5): 984-993, 2021 May.
Article in English | MEDLINE | ID: mdl-33381906

ABSTRACT

Pesticide residues have become a healthy threaten of human beings. Among the pesticides, many of them have neurotoxicity. Extracellular Regulated Protein Kinases (ERK) pathway is an important signaling pathway that regulates a variety of downstream progress. In this work, peach (PRUNUS persica) and cherry (PRUNUS cerasus) were sampled from over 300 plantations in China and assessed for the residue risk. In mechanism studies, high-risk pesticide Avermectin showed a high activity inhibiting three neurotoxicity models, SH-SY5Y, PC-12 and SK-N-SH cells. At protein levels, ERK pathway proteins and their downstream proteins were obviously down-regulated. Moreover, the effects of low-dose Avermectin can be accumulated at protein levels in the low-dose long-term chronic toxicology detection.


Subject(s)
Pesticide Residues , raf Kinases , China , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Ivermectin/analogs & derivatives , MAP Kinase Kinase Kinases , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , raf Kinases/metabolism
4.
Mol Carcinog ; 59(2): 193-201, 2020 02.
Article in English | MEDLINE | ID: mdl-31782573

ABSTRACT

Gartanin, a 4-prenylated xanthone, has been identified from the purple mangosteen fruit as a potent growth inhibitor of various cancer cell lines, including prostate cancer. However, much of Gartanin's anticancer mechanism remains unknown. We have discovered that Gartanin docked onto the regulatory subunit of the precursor cell-expressed developmentally downregulated 8 (NEDD8)-activating enzyme (NAE) complex and next to the NEDD8 binding complex, which leads to inhibit NEDD8 conjugations to both Cullin1 and Ubc12 in prostate cancer cell lines and Ubc12 NEDDylation in an in vitro assay. The S phase kinase-associated protein (Skp2) and F-box and WD-repeat domain-containing 2 (FBXW2), the NEDD8 family members of E3 ubiqutin ligases, were also downregulated and upregulated by Gartainin, respectively. Knock-down of NEDD8 expression by short harpin (Sh) RNAs blocked or attenuated these effects of Gartainin. Finally, Gartanin demonstrated its ability to inhibit growth of prostate cancer lines via autophagy initiation. Our data support that Gartanin is a naturally occurring NEDDylation inhibitor and deserves further investigation for prostate cancer prevention and treatment.


Subject(s)
Autophagy/drug effects , F-Box Proteins/metabolism , NEDD8 Protein/antagonists & inhibitors , Prostatic Neoplasms/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Xanthones/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , F-Box Proteins/genetics , Humans , Male , NEDD8 Protein/metabolism , PC-3 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA Interference
5.
J Enzyme Inhib Med Chem ; 35(1): 1403-1413, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32588683

ABSTRACT

Tubulin polymerisation inhibitors that target colchicine binding site were powerful anticancer agents. Although along the years many colchicine binding site inhibitors (CBSIs) have been reported, few piperidine derivatives were identified as CBSIs. In this regard, we focussed efforts on the piperidine as a promising chemotype to develop potent CBSIs. Herein, novel piperidine derivatives were synthesised and evaluated for their antiproliferative activities. Among them, compound 17a displayed powerful anticancer activity with the IC50 value of 0.81 µM against PC3 cells, which was significantly better than 5-fluorouracil. It could inhibit tubulin polymerisation binding at the colchicine site and inhibit the tumour growth in vitro and in vivo. Further biological studies depicted that 17a suppressed the colony formation, induced apoptosis, and inhibited epithelial-mesenchymal transition against PC3 cells. These results revealed that compound 17a is a promising colchicine binding site inhibitor for the treatment of cancer and it is worthy of further exploitation.


Subject(s)
Apoptosis/drug effects , Colchicine/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Prostatic Neoplasms/pathology , Binding Sites , Carbon-13 Magnetic Resonance Spectroscopy , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Mass Spectrometry , Piperidines/pharmacology , Proton Magnetic Resonance Spectroscopy
6.
J Enzyme Inhib Med Chem ; 35(1): 1050-1059, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32299262

ABSTRACT

Tubulin polymerisation inhibitors exhibited an important role in the treatment of patients with prostate cancer. Herein, we reported the medicinal chemistry efforts leading to a new series of benzothiazoles by a bioisosterism approach. Biological testing revealed that compound 12a could significantly inhibit in vitro tubulin polymerisation of a concentration dependent manner, with an IC50 value of 2.87 µM. Immunofluorescence and EBI competition assay investigated that compound 12a effectively inhibited tubulin polymerisation and directly bound to the colchicine-binding site of ß-tubulin in PC3 cells. Docking analysis showed that 12a formed hydrogen bonds with residues Tyr357, Ala247 and Val353 of tubulin. Importantly, it displayed the promising antiproliferative ability against C42B, LNCAP, 22RV1 and PC3 cells with IC50 values of 2.81 µM, 4.31 µM, 2.13 µM and 2.04 µM, respectively. In summary, compound 12a was a novel colchicine site tubulin polymerisation inhibitor with potential to treat prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Colchicine/pharmacology , Tubulin Modulators/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzothiazoles/chemical synthesis , Benzothiazoles/chemistry , Cell Proliferation/drug effects , Colchicine/chemical synthesis , Colchicine/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Structure , PC-3 Cells , Structure-Activity Relationship , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Tumor Cells, Cultured
7.
Bioorg Chem ; 92: 103190, 2019 11.
Article in English | MEDLINE | ID: mdl-31465969

ABSTRACT

A series of novel indoline derivatives were synthesized and evaluated for antiproliferative activity against four selected cancer cell lines (Hela, A549, HepG2 and KYSE30). Among them, compound 20 displayed the potent inhibition activity against esophageal cancer cells (Kyse30, Kyse450, Kyse510 and EC109). Cellular mechanism studies in esophageal squamous cell carcinoma (ESCC) cells elucidated compound 20 inhibited cell growths in vitro and in vivo, reduced colony formation, arrested cell cycle at M phase, and induced Noxa-dependent apoptosis in ESCC. Importantly, compound 20 was identified as a novel Noxa mediated apoptosis inducer. These results suggested that compound 20 might be a promising anticancer agent with potential for development of further clinical applications.


Subject(s)
Antineoplastic Agents/chemistry , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Indoles/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Indoles/pharmacology , Molecular Structure , RNA, Small Interfering/metabolism , Structure-Activity Relationship
8.
Bioorg Chem ; 86: 375-385, 2019 05.
Article in English | MEDLINE | ID: mdl-30763884

ABSTRACT

Novel chalcone-dithiocarbamate hybrids were designed, synthesized and evaluated for antiproliferative activity against selected cancer cell lines (MGC803, MCF7, and PC3). Among these analogues, (E)-2-oxo-2-((4-(3-(3,4,5-trimethoxyphenyl)acryloyl)phenyl)amino)ethyl-4-(2-hydroxyethyl)piperazine-1-carbodithioate (12d) showed the best inhibitory activity against PC3 cells (IC50 = 1.05 µM). Cellular mechanism studies elucidated 12d could inhibit colony formation, arrest cell cycle at G2/M phase and induce DNA damage against PC3 cells. Compound 12d also induced mitochondrial apoptosis by caspase activation, MMP decrease, ROS production and catalase (CAT) inhibition. Importantly, 12d inhibited epithelial-mesenchymal transition (EMT) process by regulating EMT-related proteins (E-cadherin, N-cadherin, Vimentin, MMP2, MMP9). These results indicated that 12d is a promising lead compound and deserves further investigation for prevention and treatment of human prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Catalase/antagonists & inhibitors , Chalcone/pharmacology , Enzyme Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Thiocarbamates/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Catalase/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Chalcone/chemical synthesis , Chalcone/chemistry , Dose-Response Relationship, Drug , Drug Discovery , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , MCF-7 Cells , Molecular Docking Simulation , Molecular Structure , PC-3 Cells , Reactive Oxygen Species/analysis , Structure-Activity Relationship , Thiocarbamates/chemical synthesis , Thiocarbamates/chemistry , Tumor Cells, Cultured
9.
Bioorg Chem ; 84: 164-169, 2019 03.
Article in English | MEDLINE | ID: mdl-30502627

ABSTRACT

Osimertinib (AZD9291) is a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) that has been approved for the treatment of EGFR-mutated non-small cell lung cancer (NSCLC). In this study, osimertinib was characterized as a LSD1 inhibitor for the first time with an IC50 of 3.98 ±â€¯0.3 µM and showed LSD1 inhibitory effect at cellular level. These findings provide new molecular skeleton for dual inhibitor for LSD1 and EGFR. Osimertinib could serve as a lead compound for further development for anti-NSCLC drug discovery with dual targeting.


Subject(s)
Acrylamides/pharmacology , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Histone Demethylases/antagonists & inhibitors , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Acrylamides/chemistry , Aniline Compounds/chemistry , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Histone Demethylases/pharmacology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
10.
Mol Divers ; 21(4): 933-942, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28785928

ABSTRACT

Novel phenothiazine-dithiocarbamate analogues were designed by molecular hybridization strategy and synthesized and evaluated for their anticancer activity in vitro against three selected cancer cell lines (EC-109, MGC-803, and PC-3). The preliminary structure-activity relationship (SAR) for this phenothiazine-dithiocarbamate hybrids is explored. Among all analogues, 2-oxo-2-(10H-phenothiazin-10-yl)ethyl 4-ethylpiperazine-1-carbodithioate (8a) showed the most potent inhibitory activity with an [Formula: see text] value of [Formula: see text] against PC-3 cells. In addition, compound 8a could arrest the cell cycle at the G1 phase and regulate the expression of G1 checkpoint-related proteins, suggesting that phenothiazine-dithiocarbamate hybrids might be useful as cell cycle blockers.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Drug Design , Phenothiazines/chemical synthesis , Phenothiazines/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Drug Screening Assays, Antitumor , Humans , Phenothiazines/chemistry , Structure-Activity Relationship
11.
Molecules ; 22(9)2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28872607

ABSTRACT

A series of sulfanilamide-1,2,3-triazole hybrids were designed by a molecular hybridization strategy and evaluated for antiproliferative activity against three selected cancer cell lines (MGC-803, MCF-7 and PC-3). The detailed structure-activity relationships for these sulfanilamide-1,2,3-triazole hybrids were investigated. All these sulfanilamide-1,2,3-triazole hybrids exhibited moderate to potent activity against all cell lines. In particular 4-methyl-N-((1-(3-phenoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl)benzenesulfonamide (11f) showed the most potent inhibitory effect against PC-3 cells, with an IC50 value of 4.08 µM. Furthermore, the tubulin polymerization inhibitory activity in vitro of compound 11f was 2.41 µM. These sulfanilamide hybrids might serve as bioactive fragments for developing more potent antiproliferative agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Sulfanilamides/chemical synthesis , Sulfanilamides/pharmacology , Tubulin Modulators/chemical synthesis , Tubulin Modulators/pharmacology , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , Humans , Structure-Activity Relationship , Sulfanilamides/administration & dosage , Triazoles/administration & dosage , Triazoles/chemical synthesis , Triazoles/pharmacology , Tubulin/metabolism , Tubulin Modulators/administration & dosage
12.
Bioorg Med Chem Lett ; 26(16): 3918-22, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27423479

ABSTRACT

A series of novel dithiocarbamate-chalcone derivates were designed, synthesized and evaluated for antiproliferative activity against three selected cancer cell lines (EC-109, SK-N-SH and MGC-803). Majority of the synthesized compounds exhibited moderate to potent activity against all the cancer cell lines assayed. Particularly, compounds II2 and II5 exhibited the excellent growth inhibition against SK-N-SH with IC50 values of 2.03µM and 2.46µM, respectively. Further mechanism studies revealed that compound II2 could obviously inhibit the proliferation of SK-N-SH cells by inducing apoptosis and arresting the cell cycle at G0/G1 phase.


Subject(s)
Antineoplastic Agents/chemical synthesis , Chalcone/analogs & derivatives , Drug Design , Thiocarbamates/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcone/chemical synthesis , Chalcone/toxicity , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Structure-Activity Relationship , Thiocarbamates/chemical synthesis , Thiocarbamates/toxicity , Tumor Suppressor Protein p53/metabolism
13.
Molecules ; 21(5)2016 May 19.
Article in English | MEDLINE | ID: mdl-27213317

ABSTRACT

A series of novel chalcone-1,2,3-triazole-azole hybrids were designed, synthesized and evaluated for their antiproliferative activity against three selected cancer cell lines (SK-N-SH, EC-109 and MGC-803). Most of the synthesized compounds exhibited moderate to good activity against all the cancer cell lines selected. Particularly, compound I-21 showed the most excellent antiproliferative activity with an IC50 value of 1.52 µM against SK-N-SH cancer cells. Further mechanism studies revealed that compound I-21 induced morphological changes of SK-N-SH cancer cells possibly by inducing apoptosis. Novel chalcone-1,2,3-triazole-azole derivatives in this work might be a series of promising lead compounds to develop anticancer agents for treating neuroblastoma.


Subject(s)
Azoles/chemistry , Azoles/pharmacology , Cell Line, Tumor , Humans , Structure-Activity Relationship
14.
J Hematol Oncol ; 16(1): 87, 2023 07 31.
Article in English | MEDLINE | ID: mdl-37525282

ABSTRACT

NEDDylation, a post-translational modification through three-step enzymatic cascades, plays crucial roles in the regulation of diverse biological processes. NEDD8-activating enzyme (NAE) as the only activation enzyme in the NEDDylation modification has become an attractive target to develop anticancer drugs. To date, numerous inhibitors or agonists targeting NAE have been developed. Among them, covalent NAE inhibitors such as MLN4924 and TAS4464 currently entered into clinical trials for cancer therapy, particularly for hematological tumors. This review explains the relationships between NEDDylation and cancers, structural characteristics of NAE and multistep mechanisms of NEDD8 activation by NAE. In addition, the potential approaches to discover NAE inhibitors and detailed pharmacological mechanisms of NAE inhibitors in the clinical stage are explored in depth. Importantly, we reasonably investigate the challenges of NAE inhibitors for cancer therapy and possible development directions of NAE-targeting drugs in the future.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Ubiquitins/therapeutic use , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , NEDD8 Protein
15.
Anticancer Agents Med Chem ; 23(16): 1848-1859, 2023.
Article in English | MEDLINE | ID: mdl-37139673

ABSTRACT

BACKGROUND: Cancer remains one of the major health issues globally, where chemotherapy forms the main treatment mode for different types of cancers. Due to cancer cell ability to develop resistance, decreased clinical effectiveness of anticancer drugs can occur. Therefore, the need to synthesize novel antitumor drugs remains important. OBJECTIVE: The aim of our work consisted of synthesizing S-2-phenylchromane derivatives containing the tertiary amide or 1,2,3-triazole fragments with promising anticancer activity. METHODS: A series of S-2-phenylchromane derivatives were synthesized and evaluated for cytotoxic activity against three selected cancer cell lines (HGC-27 human gastric carcinoma cell line, Huh-7 epithelial-like tumorigenic cells, and A549 adenocarcinomic human alveolar basal epithelial cells) using the 3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide (MTT) assay. Hoechst staining was used to detect the effects of S-2-phenylchromane derivatives on apoptosis. The apoptosis percentages were detected by annexin V-fluoresceine isothiocyanate/propidium iodide (Annexin V-FITC/PI) double staining assay with flow cytometry. Expression levels of apoptosis-related proteins were detected by western blot. RESULTS: Cell line A549, consisting of adenocarcinomic human alveolar basal epithelial cells, displayed the highest sensitivity to the S-2-phenylchromane derivatives. Among these compounds, E2 showed the most potent antiproliferative activity against A549 cells with an IC50 value of 5.60 µM. Hoechst staining and flow cytometry analysis revealed apoptosis in A549 cells by compound E2. In addition, activation of the expression levels of caspase-3, caspase-7, and their substrate poly (ADP-ribose) polymerase (PARP) by E2 was detected by western blot. CONCLUSION: In summary, results point towards compound E2, an S-2-phenylchromane derivative, as a potential lead molecule in anticancer agents for human adenocarcinomic alveolar basal cells based on the induction of apoptosis.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Apoptosis , Antineoplastic Agents/chemistry , Apoptosis Regulatory Proteins/pharmacology , A549 Cells , Cell Proliferation , Cell Line, Tumor
16.
Eur J Med Chem ; 214: 113254, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33581557

ABSTRACT

Lysine-specific demethylase 1 (LSD1/KDM1A) has emerged as a promising target for the discovery of specific inhibitors as antitumor drugs. Based on the source of compounds, all LSD1 inhibitors in this review are divided into two categories: natural LSD1 inhibitors and synthetic LSD1 inhibitors. This review highlights the research progress of LSD1 inhibitors with the potential to treat cancer covering articles published in 2020. Design strategies, structure-activity relationships, co-crystal structure analysis and action mechanisms are also highlighted.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Histone Demethylases/metabolism , Humans , Molecular Structure , Neoplasms/metabolism
17.
Eur J Med Chem ; 201: 112510, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32592915

ABSTRACT

ß-Lactam, commonly referred as azetidin-2-one, is a multifunctional building block for synthesizing ß-amino ketones, γ-amino alcohols, and other compounds. Besides its well known antibiotic activity, this ring system exhibits a wide range of activities, attracting the attention of researchers. However, the structurally diverse ß-lactam analogues as anticancer agents and their different molecular targets are poorly discussed. The purpose of this review is 3-fold: (1) to explore the molecular hybridization approach to design ß-lactams hybrids as anticancer agents; (2) the structure activity relationship of the most active anticancer ß-lactams and (3) to summarize their antitumor mechanisms.


Subject(s)
Antineoplastic Agents/pharmacology , beta-Lactams/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Structure , Structure-Activity Relationship , beta-Lactams/chemistry
18.
Eur J Med Chem ; 192: 112153, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32135407

ABSTRACT

NEDDylation pathway regulates multiple physiological process, unlike inhibitors, NEDDylation activators are rarely studied. Novel amide derivatives were synthesized and evaluated for antiproliferative activity against MGC803, MCF-7 and PC-3 cells. Among them, Ⅶ-31 displayed the most potent activity with an IC50 value of 94 nmol/L against MGC803 cells. Cellular mechanisms elucidated that Ⅶ-31 inhibited the cell viability, arrested cell cycle at G2/M phase and induced apoptosis via intrinsic and extrinsic pathways against MGC803 cells. In addition, Ⅶ-31 activated NAE1-Ubc12-Cullin1 NEDDylation via interacting with NAE1 directly. Furthermore, the activation of NEDDylation resulted in the degradation of inhibitor of apoptosis proteins (IAPs). Importantly, Ⅶ-31 inhibited tumor growth in xenograft models in vivo without the apparent toxicity. In summary, it is the first time to reveal that Ⅶ-31 deserves consideration for cancer therapy as a NEDDylation activator.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/pharmacology , Drug Discovery , NEDD8 Protein/metabolism , Amides/chemical synthesis , Amides/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Structure-Activity Relationship , Tumor Cells, Cultured
19.
Eur J Med Chem ; 203: 112618, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32682200

ABSTRACT

On the basis and continuation of our previous studies on anti-tubulin and anti-gastric cancer agents, novel tertiary amide derivatives incorporating benzothiazole moiety were synthesized and the antiproliferative activity was studied in vitro. Preliminary structure activity relationships (SARs) were explored according to the in vitro antiproliferative activity results. Some of compounds could significantly inhibit the proliferation of three cancer cells (HCT-116, MGC-803 and PC-3 cells) and compound F10 exhibited excellent antiproliferative activity against HCT-116 cells (IC50 = 0.182 µM), MGC-803 cells (IC50 = 0.035 µM), PC-3 cells(IC50 = 2.11 µM) and SGC-7901 cells (IC50 = 0.049 µM). Compound F10 effectively inhibited tubulin polymerization (IC50 = 1.9 µM) and bound to colchicine binding site of tubulin. Molecular docking results suggested compound F10 could bind tightly into the colchicine binding site of ß-tubulin. Moreover, compound F10 could regulate the Hippo/YAP signaling pathway. Compound F10 activated Hippo signaling pathway from its very beginning MST1/2, as the result of Hippo cascade activation YAP were inhibited. And then it led to a decrease of c-Myc and Bcl-2 expression. Further molecular experiments showed that compound F10 arrested at G2/M phase, inhibited cell colony formatting and induced extrinsic and intrinsic apoptosis in MGC-803 and SGC-7901 cells. Collectively, compound F10 was the first to be reported as a new anticancer agent in vitro via inhibiting tubulin polymerization and activating the Hippo signaling pathway.


Subject(s)
Amides/chemistry , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/pathology , Tubulin/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Drug Design , G2 Phase Cell Cycle Checkpoints/drug effects , Hippo Signaling Pathway , Humans , M Phase Cell Cycle Checkpoints/drug effects , Protein Multimerization/drug effects , Protein Structure, Quaternary , Transcription Factors/metabolism , YAP-Signaling Proteins
20.
Toxicol In Vitro ; 54: 338-344, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30385350

ABSTRACT

Multi-component pesticide residues, especially pesticide residues with synergistic toxicity, are a serious threat to food safety. With risk assessment, we found that Chlorpyrifos (CPF) and Carbofuran (CBF) are 2 pesticide residues with highest risk for Actinidia chinensis planch. The results showed CPF and CBF have a synergistic neurotoxicity on neural cell SK-N-SH. The toxicity was partly depending on oxidative stress (OS) and had effects on cell apoptosis and cell cycle arrest. Furthermore, the toxicity remained on long-term low-dose condition.


Subject(s)
Carbofuran/toxicity , Chlorpyrifos/toxicity , Cholinesterase Inhibitors/toxicity , Insecticides/toxicity , Pesticide Residues/toxicity , Actinidia , Apoptosis/drug effects , Carbofuran/analysis , Cell Line, Tumor , Chlorpyrifos/analysis , Cholinesterase Inhibitors/analysis , Drug Synergism , Food Contamination/analysis , Humans , Insecticides/analysis , Oxidative Stress/drug effects , Pesticide Residues/analysis , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL