Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Bioorg Chem ; 136: 106546, 2023 07.
Article in English | MEDLINE | ID: mdl-37098288

ABSTRACT

Various diseases are deeply associated with aberrations in HDAC8 functions. These aberrations can be assigned to either structural functions or catalytic functions of HDAC8. Therefore, development of HDAC8 degradation inducers might be more promising than HDAC8 inhibitors. We employed the proteolysis targeting chimera (PROTAC) strategy to develop a selective and potent HDAC8 degradation inducer CT-4 with single-digit nanomolar DC50 values and over 95% Dmax in both triple-negative breast cancer MDA-MB-231 cells and T-cell leukemia cells. Notably, CT-4 demonstrated potent anti-migration activity and limited anti-proliferative activity in MDA-MB-231 cells. In contrast, CT-4 effectively induced apototic cell death in Jurkat cells, as assessed by a caspase 3/7 activity assay and flow cytometry. Our findings suggest that the development of HDAC8 degradation inducers holds great potential for the treatment of HDAC8-related diseases.


Subject(s)
Proteolysis Targeting Chimera , Repressor Proteins , Humans , Cell Line, Tumor , Histone Deacetylases/metabolism , Jurkat Cells , Proteolysis , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/chemistry
2.
Bioorg Chem ; 97: 103648, 2020 04.
Article in English | MEDLINE | ID: mdl-32065882

ABSTRACT

Natural protoberberine alkaloids were first identified and characterized as potent, selective and cellular active lysine specific demethylase 1 (LSD1) inhibitors. Due to our study, isoquinoline-based tetracyclic scaffold was identified as the key structural element for their anti-LSD1 activity, subtle changes of substituents attached to the core structure led to dramatic changes of the activity. Among these protoberberine alkaloids, epiberberine potently inhibited LSD1 (IC50 = 0.14 ± 0.01 µM) and was highly selective to LSD1 over MAO-A/B. Furthermore, epiberberine could induce the expression of CD86, CD11b and CD14 in THP-1 and HL-60 cells, confirming its cellular activity of inducing acute myeloid leukemia (AML) cells differentiation. Moreover, epiberberine prolonged the survival of THP-1 cells bearing mice and inhibited the growth of AML cells in vivo without obvious global toxicity. These findings give the potential application of epiberberine in AML treatment, and the isoquinoline-based tetracyclic scaffold could be used for further development of LSD1 inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Berberine Alkaloids/therapeutic use , Histone Demethylases/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Animals , Antineoplastic Agents/chemistry , Berberine Alkaloids/pharmacology , Cell Differentiation/drug effects , Cell Survival/drug effects , Female , HL-60 Cells , Histone Demethylases/metabolism , Humans , Mice , Mice, SCID
3.
J Cell Biochem ; 120(5): 7657-7666, 2019 May.
Article in English | MEDLINE | ID: mdl-30485491

ABSTRACT

USP28, a member of the deubiquitinating enzymes family, plays a vital role in the physiological process of cell proliferation, differentiation and apoptosis, DNA repair, immune response, and stress response. USP28 has been reported to be overexpressed in bladder cancer, colon cancer, breast carcinomas, and so on. Nevertheless, the role of USP28 in gastric cancer has not yet been investigated. In our study, we examined the USP28 expression in 87 paired samples of gastric cancer and normal gastric tissues. We found that USP28 was overexpressed in gastric cancer compared with normal gastric tissues (P < 0.01), and its overexpression was related to the degree of differentiation and metastases. Inhibiting USP28 expression in vitro suppressed the proliferation and invasion of gastric cancer cells by downregulating lysine specific demethylase 1. On the basis of our data, it can be concluded that USP28 may be a novel therapeutic target for gastric cancer.

4.
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
5.
Bioorg Chem ; 87: 688-698, 2019 06.
Article in English | MEDLINE | ID: mdl-30953888

ABSTRACT

Sanggenon O (SO) is a Diels-Alder type adduct extracted fromMorus alba, which has been used for its anti-inflammatory action in the Oriental medicine. However, whether it has regulatory effect on human cancer cell proliferation and what the underlying mechanism remains unknown. Here, we found that SO could significantly inhibit the growth and proliferation of A549 cells and induce its pro-apoptotic action through a caspase-dependent pathway. It could also impair the mitochondria which can be reflected by mitochondrial membrane permeabilization. Besides, SQSTM1 up-regulation and autophagic flux measurement demonstrated that exposure to SO led to autophagosome accumulation, which plays a protective role in SO-treated cells. In addition, knocking down of LC3B increased SO triggered apoptotic cell rates. These results indicated that SO has great potential as a promising candidate combined with autophagy inhibitor for the treatment of NSCLC. In conclusion, our results identified a novel mechanism by which SO exerts potent anticancer activity.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Flavonoids/pharmacology , Protective Agents/pharmacology , A549 Cells , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Flavonoids/chemical synthesis , Flavonoids/chemistry , Humans , Membrane Potential, Mitochondrial/drug effects , Molecular Conformation , Molecular Docking Simulation , Protective Agents/chemical synthesis , Protective Agents/chemistry , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
6.
J Cell Biochem ; 119(6): 4957-4966, 2018 06.
Article in English | MEDLINE | ID: mdl-29384217

ABSTRACT

B vitamins play an essential role in the biosynthesis of nucleotides, replication of DNA, supply of methyl-groups, growth and repair of cells, aberrancies of which have all been implicated in carcinogenesis. Although the potential role of vitamin B in relation to the risk of cancer, including breast, and colorectal cancer, has been investigated in several observational studies, the mechanism of action is still unclear. In this study, vitamin B2 exhibited efficient activation of LSD1 by occupying the active sites where FAD stands. Interestingly, vitamin B2 significantly downregulated expression of CD86, a sensitive surrogate biomarker of LSD1 inhibition, and showed marked activation of gastric cancer cell migration and invasion. Meanwhile, vitamin B2 induced activation of LSD1 may attenuate the proliferation inhibition, and anti-migration effects of apatinib in gastric cancer cells. These findings suggested that vitamin B supplementation may interfere with the efficacy of apatinib in patients with gastric cancer.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Histone Demethylases/metabolism , Neoplasm Proteins/metabolism , Pyridines/pharmacology , Riboflavin/pharmacology , Stomach Neoplasms/enzymology , Cell Line, Tumor , Enzyme Activation/drug effects , Histone Demethylases/genetics , Humans , Neoplasm Proteins/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
7.
Bioorg Med Chem ; 26(23-24): 6000-6014, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30448189

ABSTRACT

LSD1 is implicated in a number of malignancies and has emerged as an exciting target. As part of our sustained efforts to develop novel reversible LSD1 inhibitors for epigenetic therapy of cancers, in this study, we reported a series of stilbene derivatives and evaluated their LSD1 inhibitory activities, obtaining several compounds as potent LSD1 inhibitors with IC50 values in submicromolar range. Enzyme kinetics studies and SPR assay suggested that compound 8c, the most active LSD1 inhibitor (IC50 = 283 nM), potently inhibited LSD1 in a reversible and FAD competitive manner. Consistent with the kinetics data, molecular docking showed that compound 8c can be well docked into the FAD binding site of LSD1. Flow cytometry analysis showed that compound 8c was capable of up-regulating the expression of the surrogate cellular biomarker CD86 in THP-1 human leukemia cells, suggesting the ability to block LSD1 activity in cells. Compound 8c showed good inhibition against THP-1 and MOLM-13 cells with IC50 values of 5.76 and 8.34 µM, respectively. Moreover, compound 8c significantly inhibited colony formation of THP-1 cells dose dependently.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Stilbenes/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Histone Demethylases/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Molecular Docking Simulation , Molecular Structure , Stilbenes/chemical synthesis , Stilbenes/chemistry , Structure-Activity Relationship , THP-1 Cells
8.
Bioorg Med Chem Lett ; 27(22): 5036-5039, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29037950

ABSTRACT

Lysine specific demethylase 1 (LSD1), the first identified histone demethylase, plays an important role in epigenetic regulation of gene activation and repression, has been reported to be up-regulated and involved in numbers of solid malignant tumors. In this study, we identified a series of phenylalanyl hydrazones based LSD1 inhibitors, and the most potent one, compound 4q, can inactivate LSD1 with IC50 = 91.83 nM. In cellular level, compound 4q can induce the accumulation of CD86 as well as H3K4me2, and inhibit gastric cancer cell proliferation by inactivating LSD1. Our findings indicated that compound 4q may serve as a potential leading compound to target LSD1 overexpressed gastric cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Histone Demethylases/metabolism , Hydrazones/chemistry , Tranylcypromine/analogs & derivatives , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Histone Demethylases/antagonists & inhibitors , Humans , Inhibitory Concentration 50 , Structure-Activity Relationship , Tranylcypromine/chemical synthesis , Tranylcypromine/toxicity
9.
Pharmaceutics ; 14(1)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35057080

ABSTRACT

The tumor necrosis factor (TNF) ligand family has nine ligands that show promiscuity in binding multiple receptors. As different receptors transduce into diverse pathways, the study on the functional role of natural ligands is very complex. In this review, we discuss the TNF ligands engineering for receptor specificity and summarize the performance of the ligand variants in vivo and in vitro. Those variants have an increased binding affinity to specific receptors to enhance the cell signal conduction and have reduced side effects due to a lowered binding to untargeted receptors. Refining receptor specificity is a promising research strategy for improving the application of multi-receptor ligands. Further, the settled variants also provide experimental guidance for engineering receptor specificity on other proteins with multiple receptors.

10.
Pharmaceutics ; 14(2)2022 Feb 10.
Article in English | MEDLINE | ID: mdl-35214127

ABSTRACT

Artemisinin, the most famous anti-malaria drug initially extracted from Artemisia annua L., also exhibits anti-tumor properties in vivo and in vitro. To improve its solubility and bioavailability, multiple derivatives have been synthesized. However, to reveal the anti-tumor mechanism and improve the efficacy of these artemisinin-type drugs, studies have been conducted in recent years. In this review, we first provide an overview of the effect of artemisinin-type drugs on the regulated cell death pathways, which may uncover novel therapeutic approaches. Then, to overcome the shortcomings of artemisinin-type drugs, we summarize the recent advances in two different therapeutic approaches, namely the combination therapy with biologics influencing regulated cell death, and the use of nanocarriers as drug delivery systems. For the former approach, we discuss the superiority of combination treatments compared to monotherapy in tumor cells based on their effects on regulated cell death. For the latter approach, we give a systematic overview of nanocarrier design principles used to deliver artemisinin-type drugs, including inorganic-based nanoparticles, liposomes, micelles, polymer-based nanoparticles, carbon-based nanoparticles, nanostructured lipid carriers and niosomes. Both approaches have yielded promising findings in vitro and in vivo, providing a strong scientific basis for further study and upcoming clinical trials.

11.
Eur J Med Chem ; 176: 1-10, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31082759

ABSTRACT

A class of vanadium complexes were prepared and investigated for their antiproliferative effects by MTT assay. The structure-activity relationship was extensively studied through the ligand variation. The results showed that the synthetic vanadium complexes demonstrated moderate to good antiproliferative activities against the four cancer cell lines including MGC803, EC109, MCF7 and HepG2, respectively. Of note was that most of the complexes showed preferential growth inhibitory activity to some degree toward gastric cancer line MGC803. Among them, complex 19 exhibited the most and broad-spectrum proliferative inhibition against the tested cell lines. In addition, mechanism studies illustrated that complex 19 could prevent the colony formation, migration and EMT process, as well as induce apoptosis of MGC803 cells. Furthermore, Western blot experiments revealed that the expression of apoptosis-related proteins changed, including up-regulation of Bax, PARP and caspase-3/9, as well as down-regulation of Bcl-2.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Vanadium/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Down-Regulation , Humans , Molecular Structure , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Structure-Activity Relationship , Up-Regulation , bcl-2-Associated X Protein/metabolism
12.
Eur J Med Chem ; 166: 432-444, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30739825

ABSTRACT

Phenelzine was first employed to design new aryl hydrazine-based LSD1 inhibitors based on the experience-based discovery (EBD) strategy. Among these compounds, D8 potently inhibited LSD1 (IC50 = 882.30 nM) in a reversible manner. Compound D8 was selective to LSD1 over MAO-A/B and showed H3K4me2 competitive binding to LSD1. The interaction between H3K4me2 and LSD1 was also confirmed by the Co-IP assay. In LSD1 overexpressed A549 cells, compound D8 dose-dependently induced accumulation of LSD1 substrates H3K4me1/2 and H3K9me1/2, showed cellular target engagement to LSD1 and significantly inhibited cell migration of A549 cells. Docking studies suggested that compound D8 occupied the peptide binding region and therefore blocked the access of the peptide substrate to the FAD, finally leading to the demethylase activity inhibition of LSD1. The findings indicate that aryl hydrazines are new scaffolds for the design of LSD1 inhibitors, the identification of D8 provides further evidence for our previously proposed general principle that fused heterocycles with an amine group are potentially active toward LSD1 by competitive binding to LSD1 with H3 peptide substrates.


Subject(s)
Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Hydrazines/chemistry , Hydrazines/pharmacology , A549 Cells , Binding, Competitive , Cell Survival/drug effects , Enzyme Inhibitors/metabolism , Histone Demethylases/metabolism , Histones/metabolism , Humans , Hydrazines/metabolism , Methylation/drug effects , Structure-Activity Relationship
13.
Eur J Med Chem ; 167: 388-401, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30780087

ABSTRACT

The histone lysine specific demethylase 1 (LSD1/KDM1A) is implicated in the development of cancers, targeting LSD1 has been recognized as a promising strategy for cancer therapy. To date, some small-molecule inhibitors are currently being investigated in clinical trials. Herein we report the design, synthesis and biochemical characterization of [1,2,4]triazolo[1,5-a]pyrimidine derivatives as new LSD1 inhibitors. Of these compounds, compound C26 inhibited LSD1 in a reversible manner (IC50 = 1.72 µM) and showed selectivity to LSD1 over MAO-A/B. Besides, compound C26 displayed FAD-competitive binding to LSD1. Interestingly, C26 did not inhibit horseradish peroxidase (HRP) and quench H2O2, thus excluding the possibility that LSD1 inhibition by C26 was due to the HRP inhibition and consumption of H2O2. In LSD1 overexpressed A549 cells, compound C26 concentration-dependently induced accumulation of H3K4me1/me2 and H3K9me2 and showed cellular target engagement to LSD1. Additionally, compound C26 significantly inhibited migration of A549 cells in a concentration-dependent manner, further western blot analysis showed that C26 increased expression levels of epithelial cell markers E-Cadherin and Claudin-1, down-regulated mesenchymal cell marker N-Cadherin and the upstream transcription factors Snail and Slug. Docking studies were also performed to rationalize the potency of C26 toward LSD1. To conclude, the [1,2,4]triazolo[1,5-a]pyrimidine could serve as a promising scaffold for the development of new LSD1 inhibitors.


Subject(s)
Drug Design , Enzyme Inhibitors/chemical synthesis , Histone Demethylases/antagonists & inhibitors , Pyrimidines/pharmacology , Triazoles/pharmacology , Binding, Competitive , Cadherins/metabolism , Claudin-1/metabolism , Enzyme Inhibitors/pharmacology , Humans , Molecular Docking Simulation , Pyrimidines/chemical synthesis , Structure-Activity Relationship , Transcription Factors/metabolism , Triazoles/chemical synthesis
14.
J Pharm Biomed Anal ; 162: 9-15, 2019 Jan 05.
Article in English | MEDLINE | ID: mdl-30219599

ABSTRACT

N6-methyladenosine (m6A) is the most prevalent internal modification of eukaryotic messenger RNA (mRNA). Until now, two RNA demethylases have been identified, including FTO (fat mass and obesity-associated protein) and ALKBH5 (α-ketoglutarate-dependent dioxygenase alkB homologue 5). As a mammalian m6A demethylase, ALKBH5 significantly affects mRNA export and RNA metabolism as well as the assembly of mRNA processing factors in nuclear speckles, and ALKBH5 may play a significant role in these biological processes. Nevertheless, no modulator of ALKBH5 has been reported. The reason for that may be the lack of in vitro assays for ALKBH5 inhibitor screening. Herein, we describe the development of two homogeneous assays for ALKBH5 using N6-methyladenosine as substrate with different principles. Using ALKBH5 recombinant, we developed a formaldehyde dehydrogenase coupled fluorescence based assay and an antibody based assay for the activity evaluation of ALKBH5. These robust coupled assays are suitable for screening ALKBH5 inhibitors in 384-well format (Z' factors of 0.74), facilitating the discovery of modulators in the quest for the regulation of biological processes.


Subject(s)
Adenosine/analogs & derivatives , Aldehyde Oxidoreductases/metabolism , AlkB Homolog 5, RNA Demethylase/metabolism , Drug Discovery/methods , Fluorescent Antibody Technique , Formaldehyde/metabolism , High-Throughput Screening Assays/methods , RNA, Messenger/metabolism , Adenosine/metabolism , Aldehyde Oxidoreductases/antagonists & inhibitors , AlkB Homolog 5, RNA Demethylase/antagonists & inhibitors , Demethylation , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Reproducibility of Results , Spectrometry, Fluorescence
15.
Acta Pharm Sin B ; 9(4): 794-808, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31384539

ABSTRACT

Histone lysine specific demethylase 1 (LSD1) has been recognized as an important modulator in post-translational process in epigenetics. Dysregulation of LSD1 has been implicated in the development of various cancers. Herein, we report the discovery of the hit compound 8a (IC50 = 3.93 µmol/L) and further medicinal chemistry efforts, leading to the generation of compound 15u (IC50 = 49 nmol/L, and K i = 16 nmol/L), which inhibited LSD1 reversibly and competitively with H3K4me2, and was selective to LSD1 over MAO-A/B. Docking studies were performed to rationalize the potency of compound 15u. Compound 15u also showed strong antiproliferative activity against four leukemia cell lines (OCL-AML3, K562, THP-1 and U937) as well as the lymphoma cell line Raji with the IC50 values of 1.79, 1.30, 0.45, 1.22 and 1.40 µmol/L, respectively. In THP-1 cell line, 15u significantly inhibited colony formation and caused remarkable morphological changes. Compound 15u induced expression of CD86 and CD11b in THP-1 cells, confirming its cellular activity and ability of inducing differentiation. The findings further indicate that targeting LSD1 is a promising strategy for AML treatment, the triazole-fused pyrimidine derivatives are new scaffolds for the development of LSD1/KDM1A inhibitors.

16.
Eur J Med Chem ; 175: 357-372, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31096156

ABSTRACT

Lysine-specific demethylase 1 (LSD1), demethylase against mono- and di - methylated histone3 lysine 4, has emerged as a promising target in oncology. More specifically, it has been demonstrated as a key promoter in acute myeloid leukemia (AML), and several LSD1 inhibitors have already entered into clinical trials for the treatment of AML. In this paper, a series of new indole derivatives were designed and synthesized based on a lead compound obtained by a high-throughput screening with our in-house compound library. Among the synthetic compounds, 9e was characterized as a potent LSD1 inhibitor with an IC50 of 1.230 µM and can inhibit the proliferation of THP-1 cells effectively. And most importantly, this is the first irreversible LSD1 inhibitor that is not derived from monoamine oxidase inhibitors. Hence, the discovery of 9e may serve as a proof of concept work for AML treatment.


Subject(s)
Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Indoles/chemical synthesis , Indoles/pharmacology , Cell Line , Cell Proliferation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Furans/chemistry , High-Throughput Screening Assays , Histone Demethylases/metabolism , Humans , Indoles/chemistry , Indoles/metabolism , Inhibitory Concentration 50 , Leukemia, Myeloid, Acute/pathology , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Structure-Activity Relationship
17.
Curr Med Chem ; 24(23): 2496-2504, 2017.
Article in English | MEDLINE | ID: mdl-28486922

ABSTRACT

BACKGROUND: In the past few years, great of attention has been paid to the identification and characterization of selective and potent inhibitors of the first identified histone demethylase LSD1, which may erase mono- and di-methylated histone 3 lysine 4 and 9. As the aberrant overexpression of LSD1 is involved in various pathological processes, especially cancer, obtaining selective and potent LSD1 inhibitors has emerged as a crucial issue in medicinal chemistry research. METHOD: Until now, several LSD1 inhibitor screening models have been established, including enzyme coupled assay, LC-MS based assay, and FRET based assay. Nevertheless, due to some special instrument requirement and additional costs of LC-MS and FRET, the enzyme coupled assay is the most widely applied method for LSD1 inhibitor screening. RESULT: We summarized and compared several reported in vitro LSD1 inhibitor screening models. Each of them has distinct advantages and disadvantages, and none of these methods is perfect. In order to exclude the false positive results, at least one additional method should be applied to screen LSD1 inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Histone Demethylases/metabolism , Humans , Structure-Activity Relationship
18.
ACS Med Chem Lett ; 8(4): 384-389, 2017 Apr 13.
Article in English | MEDLINE | ID: mdl-28435523

ABSTRACT

Lysine specific demethylase 1 (LSD1) plays a pivotal role in regulating the lysine methylation. The aberrant overexpression of LSD1 has been reported to be involved in the progression of certain human malignant tumors. Abrogation of LSD1 with RNAi or small molecule inhibitors may lead to the inhibition of cancer proliferation and migration. Herein, a series of [1,2,3]triazolo[4,5-d]pyrimidine derivatives were synthesized and evaluated for their LSD1 inhibitory effects. The structure-activity relationship studies (SARs) were conducted by exploring three regions of this scaffold, leading to the discovery of compound 27 as potent LSD1 inhibitor (IC50 = 0.564 µM). Compound 27 was identified as a reversible LSD1 inhibitor and showed certain selectivity to LSD1 over monoamine oxidase A/B (MAO-A/B). When MGC-803 cells were treated with compound 27, the activity of LSD1 can be significantly inhibited, and the cell migration ability was also suppressed. Docking studies indicated that the hydrogen interaction between the nitrogen atom in the pyridine ring and Met332 could be responsible for the improved activity of 2-thiopyridine series. The [1,2,3]triazolo[4,5-d]pyrimidine scaffold can be used as the template for designing new LSD1 inhibitors.

SELECTION OF CITATIONS
SEARCH DETAIL