Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Molecules ; 28(14)2023 Jul 24.
Article in English | MEDLINE | ID: mdl-37513470

ABSTRACT

MSN8C, an analog of mansonone E, has been identified as a novel catalytic inhibitor of human DNA topoisomerase II that induces tumor regression and differs from VP-16(etoposide). Treatment with MSN8C showed significant antiproliferative activity against eleven human tumor cell lines in vitro. It was particularly effective against the HL-60/MX2 cell line, which is resistant to Topo II poisons. The resistance factor (RF) of MSN8C for Topo II in HL-60/MX2 versus HL-60 was 1.7, much lower than that of traditional Topo II poisons. Furthermore, in light of its potent antitumor efficacy and low toxicity, as demonstrated in the A549 tumor xenograft model, MSN8C has been identified as a promising candidate for antitumor applications.


Subject(s)
Antineoplastic Agents , DNA Topoisomerases, Type II , Humans , DNA Topoisomerases, Type II/metabolism , Topoisomerase II Inhibitors/pharmacology , Etoposide/pharmacology , Cell Line, Tumor , HL-60 Cells , Antineoplastic Agents/pharmacology
2.
J Med Chem ; 66(11): 7387-7404, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37253101

ABSTRACT

Metabolic reprogramming is a crucial hallmark of tumorigenesis. Modulating the reprogrammed energy metabolism is an attractive anticancer therapeutic strategy. We previously found a natural product, bouchardatine, modulated aerobic metabolism and inhibited proliferation in the colorectal cancer cell (CRC). Herein, we designed and synthesized a new series of bouchardatine derivatives to discover more potential modulators. We applied the dual-parametric high-content screening (HCS) to evaluate their AMP-activated protein kinase (AMPK) modulation and CRC proliferation inhibition effect simultaneously. And we found their antiproliferation activities were highly correlated to AMPK activation. Among them, 18a was identified with nanomole-level antiproliferation activities against several CRCs. Interestingly, the evaluation found that 18a selectively upregulated oxidative phosphorylation (OXPHOS) and inhibited proliferation by modulating energy metabolism. Additionally, this compound effectively inhibited the RKO xenograft growth along with AMPK activation. In conclusion, our study identified 18a as a promising candidate for CRC treatment and suggested a novel anti-CRC strategy by AMPK activating and OXPHOS upregulating.


Subject(s)
AMP-Activated Protein Kinases , Colorectal Neoplasms , Humans , AMP-Activated Protein Kinases/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Indole Alkaloids/pharmacology , Energy Metabolism , Cell Proliferation , Cell Line, Tumor
3.
J Med Chem ; 65(18): 12346-12366, 2022 09 22.
Article in English | MEDLINE | ID: mdl-36053318

ABSTRACT

The development of triple-negative breast cancer (TNBC) is highly associated with G-quadruplex (G4); thus, targeting G4 is a potential strategy for TNBC therapy. Because concomitant histone deacetylases (HDAC) inhibition could amplify the impact of G4-targeting compounds, we designed and synthesized two novel series of G4/HDAC dual-targeting compounds by connecting the zinc-binding pharmacophore of HDAC inhibitors to the G4-targeting isaindigotone scaffold (1). Among the new compounds, a6 with the potent HDAC inhibitory and G4 stabilizing activity could induce more DNA G4 formation than SAHA and 1 in TNBC cells. Remarkably, a6 caused more G4-related DNA damage and G4-related differentially expressed genes, consistent with its effect on disrupting the cell cycle, invasion, and glycolysis. Furthermore, a6 significantly suppresses the proliferation of various TNBC cells and the MDA-MB-231 xenograft model without evident toxicity. Our study suggests a novel strategy for TNBC therapeutics through dual-targeting HDAC and G4.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Cell Proliferation , DNA/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays , Zinc/pharmacology
4.
Molecules ; 27(17)2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36080326

ABSTRACT

Topo II and Hsp90 are promising targets. In this study, we first verified the structural similarities between Topo IIα ATPase and Hsp90α N-ATPase. Subsequently, 720 compounds from the Food and Drug Administration (FDA) drug library and kinase library were screened using the malachite green phosphate combination with the Topo II-mediated DNA relaxation and MTT assays. Subsequently, the antimalarial drug quinacrine was found to be a potential dual-target inhibitor of Topo II and Hsp90. Mechanistic studies showed that quinacrine could specifically bind to the Topo IIα ATPase domain and inhibit the activity of Topo IIα ATPase without impacting DNA cleavage. Furthermore, our study revealed that quinacrine could bind Hsp90 N-ATPase and inhibit Hsp90 activity. Significantly, quinacrine has broad antiproliferation activity and remains sensitive to the multidrug-resistant cell line MCF-7/ADR and the atypical drug-resistant tumor cell line HL-60/MX2. Our study identified quinacrine as a potential dual-target inhibitor of Topo II and Hsp90, depending on the ATP-binding domain, positioning it as a hit compound for further structural modification.


Subject(s)
Antineoplastic Agents , Neoplasms , Adenosine Triphosphatases/metabolism , Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA Topoisomerases, Type II/metabolism , Drug Repositioning , HSP90 Heat-Shock Proteins , Quinacrine/pharmacology
5.
J Med Chem ; 65(19): 12675-12700, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36121464

ABSTRACT

c-MYC is a key driver of tumorigenesis. Repressing the transcription of c-MYC by stabilizing the G-quadruplex (G4) structure with small molecules is a potential strategy for cancer therapy. Herein, we designed and synthesized 49 new derivatives by introducing carbohydrates to our previously developed c-MYC G4 ligand 1. Among these compounds, 19a coupled with a d-glucose 1,2-orthoester displayed better c-MYC G4 binding, stabilization, and protein binding disruption abilities than 1. Our further evaluation indicated that 19a blocked c-MYC transcription by targeting the promoter G4, leading to c-MYC-dependent cancer cell death in triple-negative breast cancer cell MDA-MB-231. Also, 19a significantly inhibited tumor growth in the MDA-MB-231 mouse xenograft model accompanied by c-MYC downregulation. Notably, the safety of 19a was dramatically improved compared to 1. Our findings indicated that 19a could become a promising anticancer candidate, which suggested that introducing carbohydrates to improve the G4-targeting and antitumor activity is a feasible option.


Subject(s)
Antineoplastic Agents , G-Quadruplexes , 14-alpha Demethylase Inhibitors , Animals , Antineoplastic Agents/chemistry , Carbohydrates , Glucose , Humans , Imidazoles , Ligands , Mice , Proto-Oncogene Proteins c-myc/metabolism , Sugars , Sweetening Agents
6.
Br J Pharmacol ; 179(7): 1411-1432, 2022 04.
Article in English | MEDLINE | ID: mdl-34783017

ABSTRACT

BACKGROUND AND PURPOSE: Non-alcoholic steatohepatitis (NASH) is the more severe form of metabolic associated fatty liver disease (MAFLD) and no pharmacological treatment as yet been approved. Identification of novel therapeutic targets and their agents is critical to overcome the current inadequacy of drug treatment for NASH. EXPERIMENTAL APPROACH: The correlation between heat shock factor 1 (HSF1) levels and the development of NASH and the target genes of HSF1 in hepatocyte were investigated by chromatin-immunoprecipitation sequencing. The effects and mechanisms of SYSU-3d in alleviating NASH were examined in relevant cell models and mouse models (the Ob/Ob mice, high-fat and high-cholesterol diet and the methionine-choline deficient diet-fed mice). The actions of SYSU-3d in vivo were evaluated. KEY RESULTS: HSF1 is progressively reduced with mitochondrial dysfunction in NASH pathogenesis and activation of this transcription factor by its newly identified activator SYSU-3d effectively inhibited all manifestations of NASH in mice. When activated, the phosphorylated HSF1 (Ser326) translocated to nucleus and bound to the promoter of PPARγ coactivator-1α (PGC-1α) to induce mitochondrial biogenesis. Thus, increasing mitochondrial adaptive oxidation and inhibiting oxidative stress. The deletion of HSF1 and PGC-1α or recovery of HSF1 in HSF1-deficiency cells showed the HSF1/PGC-1α pathway was mainly responsible for the anti-NASH effects of SYSU-3d independent of AMP-activated protein kinase (AMPK). CONCLUSION AND IMPLICATIONS: Activation of HSF1 is a practical therapeutic approach for NASH treatment via the HSF1/PGC-1α/mitochondrial pathway and SYSU-3d can be considered as a potential candidate for the treatment of NASH.


Subject(s)
Heat Shock Transcription Factors , Mitochondria , Non-alcoholic Fatty Liver Disease , AMP-Activated Protein Kinases/metabolism , Animals , Heat Shock Transcription Factors/agonists , Heat Shock Transcription Factors/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
7.
Bioorg Chem ; 114: 105097, 2021 09.
Article in English | MEDLINE | ID: mdl-34171594

ABSTRACT

Novel mansonone F derivative MSN54 (9-bromo-2,3-diethylbenzo[de]chromene-7,8-dione) exhibited significant cytotoxicity against twelve human tumor cell lines in vitro, with particularly strong potency against HL-60/MX2 cell line resistant to Topo II poisons. MSN54 was found to have IC50 of 0.69 and 1.43 µM against HL-60 and HL-60/MX2 cells, respectively. The resistance index is 10 times lower than that of the positive control VP-16 (etoposide). Various biological assays confirmed that MSN54 acted as a Topo IIα specific non-intercalative catalytic inhibitor. Furthermore, MSN54 exhibited good antitumor efficacy and low toxicity at a dose of 5 mg/kg in A549 tumor xenograft models. Thus, compound MSN54 is a promising candidate for the development of novel antitumor agents.


Subject(s)
Antineoplastic Agents/pharmacology , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Topoisomerase II Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Topoisomerases, Type II/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Poly-ADP-Ribose Binding Proteins/metabolism , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry
8.
J Med Chem ; 63(17): 9752-9772, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32697083

ABSTRACT

DNA damage response (DDR) pathways are crucial for the survival of cancer cells and are attractive targets for cancer therapy. Bloom syndrome protein (BLM) is a DNA helicase that performs important roles in DDR pathways. Our previous study discovered an effective new BLM inhibitor with a quinazolinone scaffold by a screening assay. Herein, to better understand the structure-activity relationship (SAR) and biological roles of the BLM inhibitor, a series of new derivatives were designed, synthesized, and evaluated based on this scaffold. Among them, compound 9h exhibited nanomolar inhibitory activity and binding affinity for BLM. 9h could effectively disrupt BLM recruitment to DNA in cells. Furthermore, 9h inhibited the proliferation of the colorectal cell line HCT116 by significantly triggering DNA damage in the telomere region and inducing apoptosis, especially in combination with a poly (ADP-ribose) polymerase (PARP) inhibitor. This result suggested a synthetic lethal effect between the BLM and PARP inhibitors in DDR pathways.


Subject(s)
DNA Damage , Drug Design , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Quinazolinones/chemical synthesis , Quinazolinones/pharmacology , RecQ Helicases/antagonists & inhibitors , Telomere/genetics , Apoptosis/drug effects , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Drug Synergism , HCT116 Cells , Humans , Models, Molecular , Protein Conformation , Quinazolinones/chemistry , RecQ Helicases/chemistry , Structure-Activity Relationship
9.
Eur J Med Chem ; 192: 112172, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32163815

ABSTRACT

Discovery of novel anti-obesity agents is a challenging and promising research area. Based on our previous works, we synthesized 40 novel ß-indoloquinazoline analogues by altering the skeleton and introducing preferential side chains, evaluated their lipid-lowering activity and summarized the structure-activity relationships. In combination with an evaluation of the lipid-lowering efficacies, AMP-dependent activated protein kinase (AMPK) activating ability and liver microsomal stability, compound 23 (named as IQZ23) was selected for further studies. IQZ23 exerted a high efficacy in decreasing the triglyceride level (EC50 = 0.033 µM) in 3T3-L1 adipocytes. Mechanistic studies revealed the lipid-lowering activity of IQZ23 was dependent on the AMPK pathway by modulating ATP synthase activity. This activation was accompanied by mitochondrial biogenesis and oxidation capacity increased, and insulin sensitivity enhanced in pertinent cell models by various interventions. Correspondingly, IQZ23 (20 mg/kg, i.p.) treatment significantly reversed high fat and cholesterol diet (HFC)- induced body weight increases and accompanying clinical symptoms of obesity in mice but without indicative toxicity. These results indicate that IQZ23 could be a useful candidate for the treatment of obesity and related metabolic disorders.


Subject(s)
Anti-Obesity Agents/pharmacology , Drug Discovery , Metabolic Diseases/drug therapy , Obesity/drug therapy , 3T3-L1 Cells , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/chemistry , Cell Differentiation/drug effects , Cells, Cultured , Cholesterol , Diet, High-Fat , Dose-Response Relationship, Drug , Male , Metabolic Diseases/chemically induced , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Molecular Structure , Obesity/chemically induced , Obesity/metabolism , Structure-Activity Relationship
10.
Bioorg Chem ; 91: 103131, 2019 10.
Article in English | MEDLINE | ID: mdl-31377387

ABSTRACT

For the development of novel anticancer agents, we designed and synthesized a total of 37 perimidine o-quinone derivatives containing the o-quinone group at the A or B ring and different substituents (alkyl groups, aryl groups or heterocycles) at the C ring of the compounds. The structure-activity relationships (SARs) were established based on the cytotoxicity data of compounds from the HL-60, Huh7, Hct116, and Hela cell lines. The cytotoxicity results showed that most compounds exhibited potent cytotoxicity. In particular, compound b-12 showed the best anti-proliferative activity (IC50 ≤ 1 µM) against four cancer cell lines and strong potency against the HL-60/MX2 (0.47 µM) cell line, which is resistant to Topo II poisons. Further studies showed that b-12 exhibited potent Topo IIα inhibitory activity (IC50 = 7.54 µM) compared with Topo I, which acted as a class of non-intercalative Topo IIα catalytic inhibitor by inhibiting the ATP binding site of Topo II. Cell apoptosis and cell cycle assays confirmed that b-12 could induce the apoptosis of Huh7 cells in a dose-dependent manner.


Subject(s)
Antineoplastic Agents/pharmacology , Quinazolines/pharmacology , Quinones/pharmacology , Topoisomerase II Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/metabolism , Drug Design , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Molecular Structure , Protein Binding , Quinazolines/chemical synthesis , Quinazolines/metabolism , Quinones/chemical synthesis , Quinones/metabolism , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/metabolism
11.
J Med Chem ; 62(6): 3147-3162, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30827110

ABSTRACT

Homologous recombination repair (HRR), a crucial approach in DNA damage repair, is an attractive target in cancer therapy and drug design. The Bloom syndrome protein (BLM) is a 3'-5' DNA helicase that performs an important role in HRR regulation. However, limited studies about BLM inhibitors and their biological effects have been reported. Here, we identified a class of isaindigotone derivatives as novel BLM inhibitors by synthesis, screening, and evaluating. Among them, compound 29 was found as an effective BLM inhibitor with a high binding affinity and good inhibitory effect on BLM. Cellular evaluation indicated that 29 effectively disrupted the recruitment of BLM at DNA double-strand break sites, promoted an accumulation of RAD51, and regulated the HRR process. Meanwhile, 29 significantly induced DNA damage responses, as well as apoptosis and proliferation arrest in cancer cells. Our finding provides a potential anticancer strategy based on interfering with BLM via small molecules.


Subject(s)
Alkaloids/pharmacology , DNA/metabolism , Drug Discovery , Enzyme Inhibitors/pharmacology , Quinazolines/pharmacology , RecQ Helicases/antagonists & inhibitors , Recombinational DNA Repair , Alkaloids/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , DNA Breaks, Double-Stranded/drug effects , Enzyme Inhibitors/chemistry , HCT116 Cells , Humans , Quinazolines/chemistry , Rad51 Recombinase/metabolism , RecQ Helicases/metabolism
12.
Medicine (Baltimore) ; 97(36): e12175, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30200119

ABSTRACT

BACKGROUND: There is no consensus regarding the therapeutic effect of nasointestinal tubes (NITs) versus nasogastric tubes (NGTs) in the management of small-bowel obstruction (SBO). This study aimed to compare the clinical outcomes between the use of NITs and NGTs in the management of SBO. METHODS: Published studies on comparing NITs with NGTs in the treatment of SBO were searched from electronic databases. Two investigators independently extracted the data; any discrepancies were adjudicated by a third investigator. Pooled odds ratio (OR) and 95% confidence interval (CI) were calculated using Review Manager 5.0. RESULTS: An extensive literature search identified 268 relevant publications, 4 of which met the inclusion criteria. There were no significant differences in the nonrequirement of operative intervention between NITs and NGTs groups (OR: 1.79; 95% CI: 0.55, 5.84). Compared with the NGTs, the NITs, which successfully passed through the pylorus, did not decrease the rate of operation in patients with SBO (OR: 2.19; 95% CI: 0.59, 8.15). There was no advantage of NITs over NGTs in patients with partial SBO (P-SBO) (OR: 1.04; 95% CI: 0.23, 4.60). Postoperative complications were compared between the groups (OR: 2.13; 95% CI: 1.09, 4.15). CONCLUSION: The result of this meta-analysis showed no advantage of NITs over NGTs in the management of patients with SBO.


Subject(s)
Intestinal Obstruction/therapy , Intubation, Gastrointestinal/methods , Disease Management , Humans , Intestine, Small
13.
Bioorg Med Chem ; 26(16): 4693-4705, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30107970

ABSTRACT

Amyloid-ß (Aß) and tau protein are two crucial hallmarks in Alzheimer's disease (AD). Their aggregation forms are thought to be toxic to the neurons in the brain. A series of new 1,2,3,4-tetrahydro-1-acridone analogues were designed, synthesized, and evaluated as potential dual inhibitors for Aß and tau aggregation. In vitro studies showed that compounds 25-30 (20 µM) with N-methylation of the quinolone ring effectively inhibited Aß1-42 aggregation by 84.7%-99.5% and tau aggregation by 71.2%-101.8%. Their structure-activity relationships are discussed. In particular, 30 could permeate the blood-brain barrier, bind to Aß1-42 and tau, inhibit Aß1-42 ß-sheets formation, and prevent tau aggregation in living cells.


Subject(s)
Acridones/chemistry , Amyloid beta-Peptides/metabolism , Central Nervous System Agents/chemical synthesis , Peptide Fragments/metabolism , tau Proteins/metabolism , Acridones/metabolism , Acridones/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Blood-Brain Barrier/metabolism , Central Nervous System Agents/metabolism , Central Nervous System Agents/pharmacology , Drug Design , HEK293 Cells , Humans , Microscopy, Confocal , Microscopy, Electron, Transmission , Peptide Fragments/antagonists & inhibitors , Protein Aggregates/drug effects , Structure-Activity Relationship , Surface Plasmon Resonance , Swine , Tacrine/chemistry , tau Proteins/antagonists & inhibitors
14.
Dalton Trans ; 47(6): 2073-2078, 2018 Feb 06.
Article in English | MEDLINE | ID: mdl-29355255

ABSTRACT

The newly synthesized ionic triple salt Ru-Er, {[RuII(bpy)2(dbim)][ErIII(hfac)4][CF3COO]·H2O} (bpy = 2,2'-bipyridine; hfac- = hexafluoroacetylacetonate; dbim = 2,2'-dibenzimidazole) exhibits near-infrared (NIR) emission at 1535 nm by intermolecular Ru → Er (d → f) energy transfer across supramolecular interactions when pumped within the Ru(ii) 3MLCT band. It is the first such observation for a transition metal-lanthanide ionic pair.

15.
Org Lett ; 19(21): 5868-5871, 2017 11 03.
Article in English | MEDLINE | ID: mdl-29047270

ABSTRACT

A novel cascade Cp*Rh(III)-catalyzed C-H alkylation/Cu(II)-promoted α-oxygenation which enabled a three-component carboxygenation of activated alkene is reported. Mild reaction conditions, broad substrate scope, and good functional group tolerance were observed. The synthetic utility of the protocol was showcased by the facile transformations of the product to a variety of structurally diverse molecules. Preliminary mechanistic studies were conducted.

16.
Eur J Med Chem ; 130: 139-153, 2017 Apr 21.
Article in English | MEDLINE | ID: mdl-28242549

ABSTRACT

A series of 2-arylethenyl-N-methylquinolinium derivatives were designed and synthesized based on our previous research of 2-arylethenylquinoline analogues as multifunctional agents for the treatment of Alzheimer's disease (AD) (Eur. J. Med. Chem. 2015, 89, 349-361). The results of in vitro biological activity evaluation, including ß-amyloid (Aß) aggregation inhibition, cholinesterase inhibition, and antioxidant activity, showed that introduction of N-methyl in quinoline ring significantly improved the anti-AD potential of compounds. The optimal compound, compound a12, dramatically attenuated the cell death of glutamate-induced HT22 cells by preventing the generation of ROS and increasing the level of GSH. Most importantly, intragastric administration of a12•HAc was well tolerated at doses up to 2000 mg/kg and could traverse blood-brain barrier.


Subject(s)
Alzheimer Disease/drug therapy , Quinolines/chemistry , Amyloid beta-Peptides/drug effects , Antioxidants/chemistry , Antioxidants/pharmacology , Blood-Brain Barrier/metabolism , Cell Death/drug effects , Cell Line , Cholinesterase Inhibitors/chemistry , Drug Design , Glutathione/metabolism , Humans , Quinolines/pharmacology , Reactive Oxygen Species/metabolism
17.
J Med Chem ; 59(1): 238-52, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26649766

ABSTRACT

Novel topoisomerase II (Topo II) inhibitors have gained considerable interest for the development of anticancer agents. In this study, a series of 1,3-benzoazolyl-substituted pyrrolo[2,3-b]pyrazine derivatives were designed, synthesized, and evaluated as potential Topo II catalytic inhibitors. It was found that some of derivatives had good antiproliferative activity on seven cancer cell lines, especially on HL-60/MX2, a cancer cell line derivative from HL-60 that is resistant to Topo II poison. Topo II mediated DNA relaxation assay results showed that derivatives could significantly inhibit the activity of Topo II, and the structure-activity relationship studies indicated the importance of the alkylamino side chain and the benzoazolyl group. Further mechanism studies revealed that derivatives function as Topo II nonintercalative catalytic inhibitors and may block the ATP binding site of Topo II. Moreover, flow cytometric analysis showed that this class of compounds could induce apoptosis of HL-60 cells.


Subject(s)
Pyrazines/chemical synthesis , Pyrazines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Catalysis , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/drug effects , DNA Helicases/drug effects , DNA Topoisomerases, Type II/chemistry , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Docking Simulation , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2/metabolism , Structure-Activity Relationship
18.
J Med Chem ; 58(23): 9395-413, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26572282

ABSTRACT

Our recent study has shown that the natural product bouchardatine (1) can reduce the triglyceride (TG) content in 3T3-L1 adipocytes (EC50 ≈ 25 µM). Here, we synthesized two series of compounds by introducing amine side chains at the 5 or 8 position of 1 and evaluated the lipid-lowering activity of derivatives. It was found that some of the compounds had significant lipid-lowering effects, and the most active compound 3d showed better activity (EC50 = 0.017 µM) than 2 (EC50 = 0.086 µM), a compound reported by us. Further, the mechanism studies revealed that 3d blocked TG accumulation via activation of the LKB1-AMPK signaling pathway, efficiently down-regulating the expression of key regulators of adipogenesis/lipogenesis. Cell uptake assay and confocal imaging of 3d in cells indicated that compound 3d had favorable cell permeability. Our results suggest that 3d may be a promising agent for the treatment of obesity and related metabolic disorders.


Subject(s)
Adipogenesis/drug effects , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/pharmacology , Indole Alkaloids/chemistry , Indole Alkaloids/pharmacology , Lipogenesis/drug effects , Quinazolines/chemistry , Quinazolines/pharmacology , Triglycerides/metabolism , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacokinetics , Cell Cycle Checkpoints/drug effects , Cholesterol/metabolism , Hep G2 Cells , Humans , Indole Alkaloids/chemical synthesis , Indole Alkaloids/pharmacokinetics , Mice , Protein Serine-Threonine Kinases/metabolism , Quinazolines/chemical synthesis , Quinazolines/pharmacokinetics , Signal Transduction/drug effects
19.
Bioorg Med Chem ; 23(15): 4719-4727, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26088335

ABSTRACT

Bouchardatine (1), a naturally occurring ß-indoloquinazoline alkaloid, was synthesized. For the first time, the lipid-lowering effect and mechanism of 1 was investigated in 3T3-L1 adipocytes. Our study showed that 1 could significantly reduce lipid accumulation without cytotoxicity and mainly inhibited early differentiation of adipocyte through proliferation inhibition and cell cycle arrested in dose-dependent manner. Furthermore, the inhibition of early differentiation was reflected by down-regulation of key regulators of adipogenesis/lipogenesis, including CCAAT enhancer binding proteins (C/EBPß, C/EBPδ, C/EBPα), peroxisome proliferator-activated receptors γ (PPARγ) and sterol-regulatory element binding protein-1c (SREBP-1c), in both of mRNA and protein levels. Subsequently decreasing the protein levels of acetyl CoA carboxylase (ACC), fatty acid synthase (FAS), and stearyl coenzyme A desaturated enzyme 1 (SCD-1), the rate-limited metabolic enzymes of fatty acid synthesis, were also observed. Further studies revealed that 1 persistently activated adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) during differentiation, suggesting that the AMPK may be an upstream mechanism for the effect of 1 on adipogenesis and lipogenesis. Our data suggest that 1 can be a candidate for the development of new therapeutic drugs against obesity and related metabolic disorders.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/pharmacology , Indole Alkaloids/chemistry , Lipogenesis/drug effects , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-delta/genetics , CCAAT-Enhancer-Binding Protein-delta/metabolism , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Indole Alkaloids/pharmacology , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
20.
Curr Alzheimer Res ; 12(5): 403-14, 2015.
Article in English | MEDLINE | ID: mdl-25938868

ABSTRACT

A series of new asymmetric curcumin analogues were synthesized and evaluated as potential multifunctional agents for the treatment of Alzheimer's disease. Our results showed that most of these synthetic compounds had better inhibitory properties against Aß aggregation compared with curcumin, and better anti-oxidative properties compared with the reference compound Trolox through the study of oxygen radical absorbance capacity (ORAC). Some compounds showed good properties in selectively chelating metal ions such as copper and iron. Besides, some compounds were found to be able to dissociate Aß protein which had already aggregated. The structure-activity relationships (SAR) of these synthetic compounds were studied. The present investigation indicated that our synthetic asymmetric curcumin derivatives could be potential multifunctional agents for the treatment of Alzheimer's disease (AD).


Subject(s)
Alzheimer Disease/drug therapy , Curcumin/chemical synthesis , Curcumin/pharmacology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Amyloid/drug effects , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Antioxidants/chemical synthesis , Antioxidants/pharmacology , Cell Line, Tumor , Chelating Agents/chemical synthesis , Chelating Agents/pharmacology , Chromans/pharmacology , Copper/metabolism , Curcumin/analogs & derivatives , Drug Evaluation , Humans , Microscopy, Electron, Transmission , Molecular Structure , Oxygen/chemistry , Peptide Fragments/metabolism , Protein Aggregates/drug effects , Spectrum Analysis , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...