Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Bioorg Med Chem ; 47: 116358, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34479103

ABSTRACT

Bruton tyrosine kinase (BTK) is a key kinase in the B cell antigen receptor signal transduction pathway, which is involved in the regulation of the proliferation, differentiation and apoptosis of B cells. BTK has become a significant target for the treatment of hematological malignancies and autoimmune diseases. Ibrutinib, the first-generation BTK inhibitor, has made a great contribution to the treatment of B cell malignant tumors, but there are still some problems such as resistance or miss target of site mutation. Therefore, there is an imperative need to develop novel BTK inhibitors to overcome these problems. Besides, proteolysis targeting chimera (PROTAC) technology has been successfully applied to the development of BTK degradation agents, which has opened a fresh way for the BTK targeted treatment. This paper reviews the biological function of BTK, the discovery and development of BTK targeted drugs as a promising cancer therapy. It mainly reviews the binding sites and structural characteristics of BTK, structure-activity relationships, activity and drug resistance of BTK inhibitors, as well as potential treatment strategies to overcome the resistance of BTK, which provides a reference for the rational design and development of new powerful BTK inhibitors.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Drug Development , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Agammaglobulinaemia Tyrosine Kinase/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
2.
Bioorg Med Chem ; 29: 115875, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33232875

ABSTRACT

NEDDylation is a post-translational modification of a protein, which transfers Ubiquitin like protein NEDD8 (Neuronal Precursor Cell-expressed Developmentally Down-regulated Protein 8) to the lysine residue of the product through a three-stage enzymatic reaction, and widely regulates many biological processes, such as cell cycle signal transduction and immune recognition. In the past ten years, we have witnessed tremendous progress in the study of protein ubiquitination modification, from modification mechanisms to drug development. Which suggests that inhibition of NEDDylation is an effective way to inhibit tumor. A variety of biological detection methods have been developed during the development of the inhibitor. In this review, we briefly introduced the modification process and substrates of NEDDylation, and discussed detection methods of NEDDylation activity in detail. This review will provide an up-to-date and comprehensive review of the methods for detecting NEDDylation activity that will contribute to NEDDylation inhibitor development.


Subject(s)
Antineoplastic Agents/pharmacology , NEDD8 Protein/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Humans , NEDD8 Protein/metabolism , Neoplasms/metabolism , Protein Processing, Post-Translational/drug effects , Ubiquitination/drug effects
3.
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
4.
J Enzyme Inhib Med Chem ; 36(1): 1715-1731, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34425716

ABSTRACT

Hippo signalling pathway plays a crucial role in tumorigenesis and cancer progression. In this work, we identified an N-aryl sulphonamide-quinazoline derivative, compound 9i as an anti-gastric cancer agent, which exhibited potent antiproliferative ability with IC50 values of 0.36 µM (MGC-803 cells), 0.70 µM (HCT-116 cells), 1.04 µM (PC-3 cells), and 0.81 µM (MCF-7 cells), respectively and inhibited YAP activity by the activation of p-LATS. Compound 9i was effective in suppressing MGC-803 xenograft tumour growth in nude mice without obvious toxicity and significantly down-regulated the expression of YAP in vivo. Compound 9i arrested cells in the G2/M phase, induced intrinsic apoptosis, and inhibited cell colony formation in MGC-803 and SGC-7901 cells. Therefore, compound 9i is to be reported as an anti-gastric cancer agent via activating the Hippo signalling pathway and might help foster a new strategy for the cancer treatment by activating the Hippo signalling pathway regulatory function to inhibit the activity of YAP.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Serine-Threonine Kinases/metabolism , Quinazolines/pharmacology , Stomach Neoplasms/drug therapy , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Gene Expression Regulation/drug effects , Hippo Signaling Pathway , Humans , Mice, Nude , Molecular Structure , Quinazolines/chemical synthesis , Signal Transduction , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Chem Biodivers ; 18(4): e2001012, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33644937

ABSTRACT

Chemical investigation of the ethanol extract of the branch and leaves of Illicium majus resulted in the isolation of four new phenylpropanoid glycosides (1-4) and one new phenolic glycoside (9), along with 13 known ones. Spectroscopic techniques were used to elucidate the structures of the new isolates such as 3-[(2R,3S)-7-hydroxy-2-(4-hydroxy-3-methoxyphenyl)-3-(hydroxymethyl)-2,3-dihydro-1-benzofuran-5-yl]propyl ß-D-glucopyranoside (1), [(2R,3S)-7-hydroxy-2-(4-hydroxy-3-methoxyphenyl)-5-(3-hydroxypropyl)-2,3-dihydro-1-benzofuran-3-yl]methyl 2-O-α-L-rhamnopyranosyl-ß-D-glucopyranoside (2), [(2R,3S)-7-hydroxy-2-(4-hydroxy-3-methoxyphenyl)-5-(3-hydroxypropyl)-2,3-dihydro-1-benzofuran-3-yl]methyl 2-O-α-L-rhamnopyranosyl-ß-D-xylopyranoside (3), 3-[(2R,3S)-3-({[2-O-(4-O-acetyl-α-L-rhamnopyranosyl)-ß-D-xylopyranosyl]oxy}methyl)-7-hydroxy-2-(4-hydroxy-3-methoxyphenyl)-2,3-dihydro-1-benzofuran-5-yl]propyl acetate (4), and 4-(2-hydroxyethyl)phenyl 3-O-ß-D-glucopyranosyl-ß-D-glucopyranoside (9). Free radical scavenging activities of the isolates were elucidated through the DPPH assay method. The most active compounds, 1-O-caffeoyl-ß-D-glucopyranose (17) and soulieana acid 1 (18), exhibited moderate radical scavenging activities (IC50 =37.7±4.4 µM and IC50 =97.2±3.4 µM, respectively). The antibacterial activities of the isolates against Staphylococcus aureus and Escherichia coli were also assessed, and no activity was shown at the measured concentration (<32 µg/mL).


Subject(s)
Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Glycosides/pharmacology , Illicium/chemistry , Propanols/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Antioxidants/chemistry , Antioxidants/isolation & purification , Biphenyl Compounds/antagonists & inhibitors , Dose-Response Relationship, Drug , Escherichia coli/drug effects , Glycosides/chemistry , Glycosides/isolation & purification , Microbial Sensitivity Tests , Molecular Structure , Picrates/antagonists & inhibitors , Propanols/chemistry , Propanols/isolation & purification , Staphylococcus aureus/drug effects
6.
Molecules ; 26(13)2021 Jul 02.
Article in English | MEDLINE | ID: mdl-34279387

ABSTRACT

Tubulin has been regarded as an attractive and successful molecular target in cancer therapy and drug discovery. Vicinal diaryl is a simple scaffold found in many colchicine site tubulin inhibitors, which is also an important pharmacophoric point of tubulin binding and anti-cancer activity. As the continuation of our research work on colchicine binding site tubulin inhibitors, we designed and synthesized a series of diarylamide N-containing heterocyclic derivatives by the combination of vicinal diaryl core and N-containing heterocyclic skeletons into one hybrid though proper linkers. Among of these compounds, compound 15b containing a 5-methoxyindole group exhibited the most potent inhibitory activity against the tested three human cancer cell lines (MGC-803, PC-3 and EC-109) with IC50 values of 1.56 µM, 3.56 µM and 14.5 µM, respectively. Besides, the SARs of these compounds were preliminarily studied and summarized. The most active compound 15b produced the inhibition of tubulin polymerization in a dose-dependent manner and caused microtubule network disruption in MGC-803 cells. Therefore, compound 15b was identified as a novel tubulin polymerization inhibitor targeting the colchicine binding site. In addition, the results of molecular docking also suggested compound 15b could tightly bind into the colchicine binding site of ß-tubulin.


Subject(s)
Heterocyclic Compounds/chemical synthesis , Tubulin Modulators/chemical synthesis , Tubulin/chemistry , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/pharmacology , Heterocyclic Compounds/pharmacology , Humans , Microtubules/drug effects , Protein Binding , Quantitative Structure-Activity Relationship , Tubulin/drug effects , Tubulin/metabolism , Tubulin Modulators/pharmacology
7.
Molecules ; 26(16)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34443487

ABSTRACT

The chalcone and quinoline scaffolds are frequently utilized to design novel anticancer agents. As the continuation of our work on effective anticancer agents, we assumed that linking chalcone fragment to the quinoline scaffold through the principle of molecular hybridization strategy could produce novel compounds with potential anticancer activity. Therefore, quinoline-chalcone derivatives were designed and synthesized, and we explored their antiproliferative activity against MGC-803, HCT-116, and MCF-7 cells. Among these compounds, compound 12e exhibited a most excellent inhibitory potency against MGC-803, HCT-116, and MCF-7 cells with IC50 values of 1.38, 5.34, and 5.21 µM, respectively. The structure-activity relationship of quinoline-chalcone derivatives was preliminarily explored in this report. Further mechanism studies suggested that compound 12e inhibited MGC-803 cells in a dose-dependent manner and the cell colony formation activity of MGC-803 cells, arrested MGC-803 cells at the G2/M phase and significantly upregulated the levels of apoptosis-related proteins (Caspase3/9 and cleaved-PARP) in MGC-803 cells. In addition, compound 12e could significantly induce ROS generation, and was dependent on ROS production to exert inhibitory effects on gastric cancer cells. Taken together, all the results suggested that directly linking chalcone fragment to the quinoline scaffold could produce novel anticancer molecules, and compound 12e might be a valuable lead compound for the development of anticancer agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Chalcones/chemical synthesis , Chalcones/pharmacology , Drug Design , Quinolines/chemical synthesis , Quinolines/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcones/chemistry , Humans , Quinolines/chemistry , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
8.
Molecules ; 26(14)2021 Jul 13.
Article in English | MEDLINE | ID: mdl-34299525

ABSTRACT

FAK is a nonreceptor intracellular tyrosine kinase which plays an important biological function. Many studies have found that FAK is overexpressed in many human cancer cell lines, which promotes tumor cell growth by controlling cell adhesion, migration, proliferation, and survival. Therefore, targeting FAK is considered to be a promising cancer therapy with small molecules. Many FAK inhibitors have been reported as anticancer agents with various mechanisms. Currently, six FAK inhibitors, including GSK-2256098 (Phase I), VS-6063 (Phase II), CEP-37440 (Phase I), VS-6062 (Phase I), VS-4718 (Phase I), and BI-853520 (Phase I) are undergoing clinical trials in different phases. Up to now, there have been many novel FAK inhibitors with anticancer activity reported by different research groups. In addition, FAK degraders have been successfully developed through "proteolysis targeting chimera" (PROTAC) technology, opening up a new way for FAK-targeted therapy. In this paper, the structure and biological function of FAK are reviewed, and we summarize the design, chemical types, and activity of FAK inhibitors according to the development of FAK drugs, which provided the reference for the discovery of new anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Focal Adhesion Protein-Tyrosine Kinases/chemistry , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Models, Molecular , Molecular Targeted Therapy , Neoplasms/metabolism , Protein Kinase Inhibitors/chemistry
9.
Bioorg Med Chem Lett ; 30(2): 126791, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31740251

ABSTRACT

Neddylation modification is often over-expressed in a variety of human tumor cells. Therefore, targeting neddylation pathway may represent a potential approach to the treatment of human tumors. Herein, we describe the discovery of a hit scaffold from our in-house library and further structure-based optimizations. In this work, compound V11 could block the neddylation and inhibit the activity of NAE (with an EC50 value of 3.56 µM), and a dose-dependent reduction of the Ubc12-NEDD8 conjugations was also observed. Molecular docking results suggest compound V11 could bind tightly to NAE via hydrogen bonds and hydrophobic interactions. Compound V11 showed the best antiproliferative ability with an IC50 value of 8.22 µM against gastric cancer MGC-803 cells. Further anticancer activity studies suggested that compound V11 inhibited MGC-803 cell growth, caused a cell cycle arrestment at G2/M phase and induced apoptosis via extrinsic and intrinsic apoptosis pathways. All the findings suggest that 1,2,4-triazine scaffold might provide a novel scaffold for the further development of neddylation inhibitors and compound V11 might be a potential neddylation inhibitor with anticancer activity.


Subject(s)
Antineoplastic Agents/chemistry , Triazines/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , M Phase Cell Cycle Checkpoints/drug effects , Molecular Docking Simulation , NEDD8 Protein/metabolism , Protein Processing, Post-Translational/drug effects , Protein Structure, Tertiary , Stomach Neoplasms , Structure-Activity Relationship , Triazines/pharmacology , Ubiquitin-Conjugating Enzymes/metabolism
10.
Pathobiology ; 87(3): 161-170, 2020.
Article in English | MEDLINE | ID: mdl-32182608

ABSTRACT

OBJECTIVE: MiR-203 has been shown to participate in multiple malignancies, but the role of miR-203 in hepatoblastoma (HB) remains unclear. The aim of our study was to investigate the effects of miR-203 in HB. METHODS: A total of 15 pairs of HB tissues and para-tumour normal tissues were collected for the experiments. RT-qPCR and Western blotting were performed to detect the expression of CRNDE, miR-203, and VEGFA at the mRNA and/or protein levels, respectively. A dual luciferase assay verified the target relationship between miR-203 and the 3'UTR of VEGFA as well as miR-203 and CRNDE. In addition, MTT, wound healing, and tube formation assays were performed to assess the effects of miR-203, VEGFA, and CRNDE on cell proliferation, migration, and angiogenesis, respectively. RESULTS: Our data revealed that miR-203 expression was decreased in HB tissues, while long non-coding RNA (lncRNA) CRNDE expression was increased. The dysregulation of miR-203 and CRNDE was closely related to tumour size and stage. Moreover, overexpression of miR-203 inhibited angiogenesis. A dual luciferase assay verified that VEGFA is a direct target of miR-203 and that CRNDE binds to miR-203. Furthermore, our results showed that miR-203 suppressed cell viability, migration, and angiogenesis by regulating VEGFA expression. Additionally, it was confirmed that CRNDE promoted angiogenesis by negatively regulating miR-203 expression. CONCLUSION: lncRNA CRNDE targets the miR-203/VEGFA axis and promotes angiogenesis in HB. These results provide insight into the underlying mechanisms of HB and indicate that CRNDE and miR-203 might be potential targets for HB therapy.


Subject(s)
Hepatoblastoma/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , RNA, Long Noncoding/genetics , Vascular Endothelial Growth Factor A/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
11.
Molecules ; 25(23)2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33255804

ABSTRACT

Chalcone is a common scaffold found in many biologically active compounds. The chalcone scaffold was also frequently utilized to design novel anticancer agents with potent biological efficacy. Aiming to continue the research of effective chalcone derivatives to treat cancers with potent anticancer activity, fourteen amino chalcone derivatives were designed and synthesized. The antiproliferative activity of amino chalcone derivatives was studied in vitro and 5-Fu as a control group. Some of the compounds showed moderate to good activity against three human cancer cells (MGC-803, HCT-116 and MCF-7 cells) and compound 13e displayed the best antiproliferative activity against MGC-803 cells, HCT-116 cells and MCF-7 cells with IC50 values of 1.52 µM (MGC-803), 1.83 µM (HCT-116) and 2.54 µM (MCF-7), respectively which was more potent than the positive control (5-Fu). Further mechanism studies were explored. The results of cell colony formatting assay suggested compound 10e inhibited the colony formation of MGC-803 cells. DAPI fluorescent staining and flow cytometry assay showed compound 13e induced MGC-803 cells apoptosis. Western blotting experiment indicated compound 13e induced cell apoptosis via the extrinsic/intrinsic apoptosis pathway in MGC-803 cells. Therefore, compound 13e might be a valuable lead compound as antiproliferative agents and amino chalcone derivatives worth further effort to improve amino chalcone derivatives' potency.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Chalcone/chemical synthesis , Chalcone/pharmacology , Chemistry Techniques, Synthetic , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chalcone/analogs & derivatives , Drug Screening Assays, Antitumor , Humans , Inhibitory Concentration 50 , Molecular Structure , Structure-Activity Relationship
12.
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
13.
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
14.
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
15.
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
16.
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
17.
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
18.
J Asian Nat Prod Res ; 15(1): 78-83, 2013.
Article in English | MEDLINE | ID: mdl-23323717

ABSTRACT

A new C(18)-diterpenoid alkaloid, kirinenine A (1), was isolated from the root of Aconitum kirinense, along with eight known diterpenoid alkaloids. The structures of all compounds were characterized on the basis of extensive NMR and MS analyses and by comparison with literature values, and the new one was further confirmed by X-ray crystallographic diffraction. All the compounds were isolated from the title plant for the first time.


Subject(s)
Aconitum/chemistry , Alkaloids/isolation & purification , Diterpenes/isolation & purification , Drugs, Chinese Herbal/isolation & purification , Aconitine/analogs & derivatives , Alkaloids/chemistry , Crystallography, X-Ray , Diterpenes/chemistry , Drugs, Chinese Herbal/chemistry , Molecular Conformation , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Plant Roots/chemistry
19.
Molecules ; 18(10): 11866-72, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24077171

ABSTRACT

Two new monoterpenes, p-mentha-1(7),8-dien-2-O-ß-D-glucoside and trans-2,4-dihydroxy-2,4-dimethyl-trans-1-acetic acid γ-lactone were isolated from the fruits of Illicium lanceolatum along with trans-2,4-dihydroxy-2,4-dimethyl-cis-1-acetic acid γ-lactone, (1R,2R,4R)-8-p-menthen-1,2-diol, trans-sobrerol, (1S,2S,4R)-p-menthane-1,2,8-triol and (1S, 2S, 4R, 8R)-p-menthane-1,2,9-triol. The structures of the isolates were confirmed by spectroscopic analysis and they showed no inhibitory effects on the in vitro growth of microbial organisms (Escherichia coli, Staphyloccocus aureus, Bacillus subtilis) at less than 1.0 mg/mL.


Subject(s)
Fruit/chemistry , Illicium/chemistry , Monoterpenes/chemistry , Plant Extracts/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , Bacillus subtilis/drug effects , Escherichia coli/drug effects , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Models, Chemical , Models, Molecular , Molecular Conformation , Molecular Weight , Monoterpenes/isolation & purification , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Spectrometry, Mass, Electrospray Ionization , Staphylococcus aureus/drug effects
20.
Shock ; 59(3): 469-476, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36579896

ABSTRACT

ABSTRACT: Aim: The purpose of this study was to investigate the effect of esmolol (ES) on LPS-induced cardiac injury and the possible mechanism. Methods: Sepsis was induced by i.p. injection of LPS (10 mg/kg) in male Sprague-Dawley rats pretreated with ES, 3-methyladenine or rapamycin. The severity of myocardial damage was analyzed by hematoxylin-eosin staining, and myocardial damage scores were calculated. The concentration of cardiac troponin was measured by enzyme-linked immunosorbent assay. The expression of autophagy-related proteins (beclin-1, LC3-II, p-AMPK, p-ULK1, p-mTOR) in myocardial tissue was detected by Western blotting. Autophagosome formation and the ultrastructural damage of mitochondria were assessed using transmission electron microscopy. Results: LPS induced an increase in myocardial damage score in a time-dependent manner, accompanied with an increase in autophagy at 3 h and decrease in autophagy at 6, 12, and 24 h. Pretreatment of LPS-treated rats with ES or rapamycin reduced myocardial injury (release of cardiac troponin, myocardial damage score) and increased autophagy (LC3-II, beclin-1, p-AMPK, and p-ULK1 levels and autophagosome numbers) at 12 and 24 h. In contrast, 3-methyladenine showed no effect. Conclusion: Esmolol alleviates LPS-induced myocardial damage through activating the AMPK/mTOR/ULK1 signal pathway-regulated autophagy.


Subject(s)
AMP-Activated Protein Kinases , Heart Injuries , Animals , Rats , Male , AMP-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Beclin-1/pharmacology , Autophagy-Related Protein-1 Homolog/metabolism , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/metabolism , Autophagy , Sirolimus/pharmacology , Troponin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL