Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters











Publication year range
1.
Bioorg Chem ; 151: 107687, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39096559

ABSTRACT

Increasing the levels of antiapoptotic Bcl-2 proteins is an important way that cancer cells utilize to get out of apoptosis, underscoring their significance as promising targets for anticancer therapies. Lately, a primary compound 1 bearing thiazolidine-2,4-dione was discovered to exhibit comparable Mcl-1 inhibitory activity in comparison to WL-276. Herein, thirty-nine thiazolidine-2,4-dione analogs were yielded through incorporating different biphenyl moieties (R1), amino acid side chains (R2) and sulfonamides (R3) on 1. The findings indicated that certain compounds exhibited favorable inhibitory effects against Bcl-2/Mcl-1, while demonstrating limited or negligible binding affinity towards Bcl-xL. In particular, compounds 16 and 20 exhibited greater Bcl-2/Mcl-1 inhibition compared to AT-101, WL-276 and 1. Moreover, they demonstrated notable antiproliferative effects and significantly induced apoptosis in U937 cells. The western blot and co-immunoprecipitation assays confirmed that 20 could induce alterations in the expression of apoptosis-associated proteins to result in apoptosis through on-target Bcl-2 and Mcl-1 inhibition. In addition, 20 exhibited favorable stability profiles in both rat plasma and rat liver microsomes. In total, 20 could be used as a promising compound to discover Bcl-2/Mcl-1 dual inhibitors with favorable therapeutic properties.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Proliferation , Dose-Response Relationship, Drug , Drug Discovery , Drug Screening Assays, Antitumor , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2 , Thiazolidinediones , Humans , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Structure-Activity Relationship , Cell Proliferation/drug effects , Molecular Structure , Apoptosis/drug effects , Thiazolidinediones/pharmacology , Thiazolidinediones/chemistry , Thiazolidinediones/chemical synthesis , Animals , Rats , Drug Development
2.
J Colloid Interface Sci ; 670: 519-529, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38776687

ABSTRACT

The high theoretical energy density and specific capacity of lithium-sulfur (Li-S) batteries have garnered considerable attention in the prospective market. However, ongoing research on Li-S batteries appears to have encountered a bottleneck, with unresolved key technical challenges such as the significant shuttle effect and sluggish reaction kinetics. This investigation explores the catalytic efficacy of three catalysts for Li-S batteries and elucidates the correlation between their structure and catalytic impacts. The results suggest that the combined utilization of lithium-insertion technology and a proton exchange approach for δ-MnO2 can optimize its electronic structure, resulting in an optimal catalyst (H/Li inserted δ-MnO2, denoted as HLM) for the sulfur reduction reaction. The replacement of Mn sites in δ-MnO2 with Li atoms can enhance the structural stability of the catalyst, while the introduction of H atoms between transition metal layers contributes to the satisfactory catalytic performance of HLM. Theoretical calculations demonstrate that the bond length of Li2S4 adsorbed by the HLM molecule is elongated, thereby facilitating the dissociation process of Li2S4 and enhancing the reaction kinetics in Li-S batteries. Consequently, the Li-S battery utilizing HLM as a catalyst achieves a high areal specific capacity of 4.2 mAh cm-2 with a sulfur loading of 4.1 mg cm-2 and a low electrolyte/sulfur (E/S) ratio of 8 µL mg-1. This study introduces a methodology for designing effective catalysts that could significantly advance practical developments in Li-S battery technology.

3.
Eur J Med Chem ; 258: 115621, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37423125

ABSTRACT

Currently, heterocycles have occupied an important position in the fields of drug design. Among them, azaindole moiety is regarded as one privileged scaffold to develop therapeutic agents. Since two nitrogen atoms of azaindole increase the possibility to form hydrogen bonds in the adenosine triphosphate (ATP)-binding site, azaindole derivatives are important sources of kinase inhibitors. Moreover, some of them have been on the market or in clinical trials for the treatment of some kinase-related diseases (e.g., vemurafenib, pexidartinib, decernotinib). In this review, we focused on the recent development of azaindole derivatives as potential kinase inhibitors based on kinase targets, such as adaptor-associated kinase 1 (AAK1), anaplastic lymphoma kinase (ALK), AXL, cell division cycle 7 (Cdc7), cyclin-dependent kinases (CDKs), dual-specificity tyrosine (Y)-phosphorylation regulated kinase 1A (DYRK1A), fibroblast growth factor receptor 4 (FGFR4), phosphatidylinositol 3-kinase (PI3K) and proviral insertion site in moloney murine leukemia virus (PIM) kinases. Meanwhile, the structure-activity relationships (SARs) of most azaindole derivatives were also elucidated. In addition, the binding modes of some azaindoles complexed with kinases were also investigated during the SARs elucidation. This review may offer an insight for medicinal chemists to rationally design more potent kinase inhibitors bearing the azaindole scaffold.


Subject(s)
Drug Design , Phosphatidylinositol 3-Kinases , Mice , Animals , Structure-Activity Relationship , Binding Sites , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry
4.
Curr Med Chem ; 30(4): 372-389, 2023.
Article in English | MEDLINE | ID: mdl-35792127

ABSTRACT

BACKGROUND: Benzoxazine is one of the most important privileged scaffolds in medicinal chemistry. Compounds bearing benzoxazine moiety usually have a variety of biological activities, such as anti-inflammatory, anti-microbial, anti-tuberculosis, anti- oxidant and anti-cancer activities. The fascinating bioactivity profile of benzoxazine scaffold in various fields has prompted medicinal chemists to design and discover novel benzoxazine derivatives as potential therapeutic candidates with the desired biological properties. OBJECTIVE: This review aimed to provide a comprehensive elucidation on the recent advances of benzoxazine derivatives in medicinal chemistry. METHODS: We have searched the recent literature about benzoxazine derivatives from the online resources and databases, such as PubMed, SciFinder and Google Scholar. RESULTS: Many benzoxazine derivatives with a wide range of bioactivities, such as anti- microbial, anti-cancer, anti-tuberculosis, anti-oxidant and anti-inflammatory, were summed up. Many compounds displayed good biological activities. CONCLUSION: Benzoxazine is a versatile structure and building block in medicinal chemistry. Benzoxazine derivatives have gained considerable attention from medicinal chemists due to their various pharmacological properties and multiple modification sites. This review might help medicinal chemists to seek new drug candidates with better bioactivities and pharmacokinetics properties.


Subject(s)
Benzoxazines , Chemistry, Pharmaceutical , Humans , Benzoxazines/pharmacology , Anti-Inflammatory Agents/pharmacology , Structure-Activity Relationship
5.
Bioorg Med Chem Lett ; 54: 128447, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34767915

ABSTRACT

A series of (Z)-3-(2-(1,3,4-thiadiazol-2-yl)hydrazono)-1-substituted indolin-2-ones derivatives (3a-3m) were designed and synthesized. All newly synthesized compounds were evaluated for their a-glucosidase inhibitory activity with resveratrol as positive control in vitro. Except for 3i and 3j, all of the compounds showed a potent inhibitory activity against a-glucosidase with IC50 values in the range of 3.12 ± 1.25 to 45.95 ± 1.26 µM and the purity of these compounds was greater than 95%. The IC50 values were being compared to the standard resveratrol (IC50 = 22.00 ± 1.15 µM) and it was found that compounds 3b, 3d-3h were found to be more active than resveratrol. Specifically, (Z)-3-(2-(1,3,4-thiadiazol-2-yl)hydrazono)-1-(4-chlorobenzyl)indolin-2-one (3d) exhibited the most potent a-glucosidase inhibitory activity with IC50 value of 3.12 ± 1.25 µM. The kinetic analysis revealed that compound (3d) is noncompetitive inhibitor. Structure activity relationship has been established for all compounds. Furthermore, the binding interactions of compound 3d with the active site of a-glucosidase were confirmed through molecular docking. This study has identified a new class of potent a-glucosidase inhibitors for further investigation.


Subject(s)
Glycoside Hydrolase Inhibitors/pharmacology , Thiadiazoles/pharmacology , alpha-Glucosidases/metabolism , Dose-Response Relationship, Drug , Glycoside Hydrolase Inhibitors/chemical synthesis , Glycoside Hydrolase Inhibitors/chemistry , Humans , Molecular Structure , Structure-Activity Relationship , Thiadiazoles/chemical synthesis , Thiadiazoles/chemistry
6.
Eur J Med Chem ; 226: 113837, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34530384

ABSTRACT

Currently, the arise of drug resistance and undesirable off-target effects of anti-cancer agents are major challenges for cancer treatment, which energizes medicinal chemists to develop more anti-cancer agents with high efficiency and low toxicity continuously. Sulfonamide derivatives are a class of promising compounds with diverse biological activities including anti-cancer, and parts of them have been marketed for cancer therapy, such as Belinostat, ABT-199 and Amsacrine. In this review, we summed up the recent advances of sulfonamide derivatives as potential anti-cancer agents based on the anti-cancer targets, such as aromatase, carbonic anhydrase (CA), anti-apoptotic B-cell lymphoma-2 (Bcl-2) proteins, topoisomerase and phosphatidylinositol 3-kinase (PI3K), and elucidated the corresponding structure-activity relationships (SARs) of most sulfonamide derivatives. We hope this review could provide a clear insight for medicinal chemists in the rational design of more potent and bio-target specific anti-cancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Sulfonamides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
7.
Eur J Med Chem ; 210: 112953, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33148490

ABSTRACT

Cancer has been the second heath killer being next only to cardiovascular diseases in human society. Although many efforts have been taken for cancer therapy and many achievements have been yielded in the diagnosis and treatment of cancer, the current first-line anti-cancer agents are insufficient owing to the emergence of multi-drug resistance and side effects. Therefore, it is urgent to develop new anti-cancer agents with high activity and low toxicity. 2-Aminothiazole is a class of important scaffold which widely distributes in many natural and synthetic compounds with many pharmacological effects including the potential anti-cancer activity. In this review, we summarized the recent progress of 2-aminothiazole as a privileged scaffold for the discovery of anti-cancer agents based on biological targets, such as tubulin protein, histone acetylase/histone deacetylase (HAT/HDAC), phosphatidylinositol 3-kinases (PI3Ks), Src/Abl kinase, BRAF kinase, epidermal growth factor receptor (EGFR) kinase and sphingosine kinase (SphK), and also investigated the structure-activity relationships (SARs) of most compounds. It is believed that this review could be helpful for medicinal chemists in the discovery of more anti-cancer agents bearing 2-aminothiazole scaffold with excellent activity and high therapeutic index.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Thiazoles/chemistry , Thiazoles/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Drug Design , Drug Discovery , Humans , Neoplasms/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Structure-Activity Relationship , Thiazoles/therapeutic use , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use
8.
Curr Med Chem ; 28(9): 1775-1795, 2021.
Article in English | MEDLINE | ID: mdl-32520674

ABSTRACT

BACKGROUND: Currently, cancer continues being a dramatically increasing and serious threat to public health. Although many anti-tumor agents have been developed in recent years, the survival rate of patients is not satisfactory. The poor prognosis of cancer patients is closely related to the occurrence of drug resistance. Therefore, it is urgent to develop new strategies for cancer treatment. Multi-target therapies aim to have additive or synergistic effects and reduce the potential for the development of resistance by integrating different pharmacophores into a single drug molecule. Given the fact that majority of diseases are multifactorial in nature, multi-target therapies are being exploited with increasing intensity, which has brought improved outcomes in disease models and obtained several compounds that have entered clinical trials. Thus, it is potential to utilize this strategy for the treatment of BRD4 related cancers. This review focuses on the recent research advances of dual-target inhibitors based on BRD4 in the aspect of anti-tumor. METHODS: We have searched the recent literatures about BRD4 inhibitors from the online resources and databases, such as pubmed, elsevier and google scholar. RESULTS: In the recent years, many efforts have been taken to develop dual-target inhibitors based on BRD4 as anti-cancer agents, such as HDAC/BRD4 dual inhibitors, PLK1/BRD4 dual inhibitors and PI3K/BRD4 dual inhibitors and so on. Most compounds display good anti-tumor activities. CONCLUSION: Developing new anti-cancer agents with new scaffolds and high efficiency is a big challenge for researchers. Dual-target inhibitors based on BRD4 are a class of important bioactive compounds. Making structural modifications on the active dual-target inhibitors according to the corresponding structure-activity relationships is of benefit to obtain more potent anti-cancer leads or clinical drugs. This review will be useful for further development of new dual-target inhibitors based on BRD4 as anti-cancer agents.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins , Humans , Neoplasms/drug therapy , Nuclear Proteins , Transcription Factors
9.
Bioorg Chem ; 104: 104217, 2020 11.
Article in English | MEDLINE | ID: mdl-32911192

ABSTRACT

Overexpressing myeloid cell leukemia sequence 1 (Mcl-1) protein is an important way to confer the resistance of cancer cells to conventional anti-cancer treatments. Therefore, developing Mcl-1 inhibitors has become an attractive strategy for cancer therapy. In the studies, a series of new indazole-acylsulfonamide hybrids were designed, synthesized and evaluated as potent Mcl-1 inhibitors. Among them, the most potent compound 17 (Ki = 0.43 µM) showed a little better inhibitory activity against Mcl-1 protein than positive control AT-101 (Ki = 0.45 µM). Pleasingly, it displayed > 40-fold selectivity over Bcl-2 (Ki = 18 µM) and Bcl-xL (no activity). Furthermore, compound 17 had good inhibitory activities against PC-3, MDA-MB-231 and K562 cells (IC50 = 12.3, 10.6 and 6.62 µM, respectively) and could effectively induce apoptosis and the activation of caspase-3 in a dose-dependent manner in K562 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Indazoles/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Sulfonamides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Indazoles/chemistry , Molecular Docking Simulation , Molecular Structure , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Structure-Activity Relationship , Sulfonamides/chemistry
10.
J Med Chem ; 63(17): 8977-9002, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32320239

ABSTRACT

Histone deacetylases (HDACs) play an important role in regulating target gene expression. They have been highlighted as a novel category of anticancer targets, and their inhibition can induce apoptosis, differentiation, and growth arrest in cancer cells. In view of the fact that HDAC inhibitors and other antitumor agents, such as BET inhibitors, topoisomerase inhibitors, and RTK pathway inhibitors, exert a synergistic effect on cellular processes in cancer cells, the combined inhibition of two targets is regarded as a rational strategy to improve the effectiveness of these single-target drugs for cancer treatment. In this review, we discuss the theoretical basis for designing HDAC-involved dual-target drugs and provide insight into the structure-activity relationships of these dual-target agents.


Subject(s)
Antineoplastic Agents/chemistry , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , DNA Damage/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/chemistry , Humans , Neoplasms/drug therapy , Protein Kinases/chemistry , Protein Kinases/metabolism , Protein Kinases/pharmacology , Proteins/antagonists & inhibitors , Proteins/metabolism , Structure-Activity Relationship , Topoisomerase Inhibitors/chemistry , Topoisomerase Inhibitors/pharmacology , Topoisomerase Inhibitors/therapeutic use
11.
J Biol Inorg Chem ; 25(4): 597-607, 2020 06.
Article in English | MEDLINE | ID: mdl-32232583

ABSTRACT

Cyclometalated iridium(III) complexes represent a promising approach to developing new anticancer metallodrugs. In this work, three phosphorescent cyclometalated iridium(III) complexes Ir1-Ir3 have been explored as mitochondria-targeted anticancer agents. All three complexes display higher antiproliferative activity than cisplatin against the cancer cells screened, and with the IC50 values ranging from 0.23 to 5.6 µM. Colocalization studies showed that these complexes are mainly localized in the mitochondria. Mechanism studies show that these complexes exert their anticancer efficacy through initiating a series of events related to mitochondrial dysfunction, including depolarization of mitochondrial membrane potential (MMP), elevation of intracellular reactive oxygen species (ROS) levels, and induction of apoptosis. Mitochondria-targted cyclometalated iridium complexes induce apoptosis through depolarized mitochondria, elevation of intracellular ROS and activated caspase.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Iridium/pharmacology , Mitochondria/drug effects , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Iridium/chemistry , Mitochondria/metabolism , Molecular Structure , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
12.
Future Med Chem ; 12(10): 915-938, 2020 05.
Article in English | MEDLINE | ID: mdl-32270707

ABSTRACT

Proteolysis-targeting chimera (PROTAC) is a new technology to selectively degrade target proteins via ubiquitin-proteasome system. PROTAC molecules (PROTACs) are a class of heterobifunctional molecules, which contain a ligand targeting the protein of interest, a ligand recruiting an E3 ligase and a linker connecting these two ligands. They provide several advantages over traditional inhibitors in potency, selectivity and drug resistance. Thus, many promising PROTACs have been developed in the recent two decades, especially small-molecule PROTACs. In this review, we briefly introduce the mechanism of PROTACs and focus on the progress of small-molecule PROTACs based on different E3 ligases. In addition, we also introduce the opportunities and challenges of small-molecule PROTACs for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Small Molecule Libraries/pharmacology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Humans , Ligands , Molecular Structure , Proteolysis/drug effects , Small Molecule Libraries/chemistry , Ubiquitin-Protein Ligases/metabolism
13.
J Hazard Mater ; 394: 121635, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32289620

ABSTRACT

Organoarsenicals remediation requires degrading organoarsenicals and simultaneously immobilizing the resulted inorganic arsenic, and is thus a great challenge. In this study, a simulated solar light driven Fe(III)/Fe(II) cycle strategy was developed to degrade roxarsone and immobilize the generated inorganic arsenic via tuning the degree of Fe(III) hydrolysis. At pH values of 2.0 and 3.0, the hydrolysis of Fe(III) in the solution was suppressed to produce photoreactive Fe(III)-hydroxyl complexes, which could be excited by simulated solar light to generate OH for 85.3 % of roxarsone degradation into arsenate within 60 min. Density functional theory calculations suggested that Fe(OH)(H2O)52+ with lower energy separation gap was the most photoactive Fe(III)-hydroxyl complex for OH generation. With further increasing pH value to 6.0, the hydrolysis of Fe(III) was promoted to precipitate the arsenate for its immobilization, accompanying with the decrease of final iron ions and arsenate concentrations to 0.012 mmol L-1 and 58 µg L-1, respectively. Meanwhile, the undegraded roxarsone was also adsorbed by the precipitate, increasing the overall roxarsone removal efficiency to 99.0 %. This study offers a promising strategy for the efficient organoarsenicals treatment, and also sheds light on the dual effects of iron based materials in organic pollutants degradation and heavy metal ions immobilization.

14.
Chem Commun (Camb) ; 56(9): 1349-1352, 2020 Jan 30.
Article in English | MEDLINE | ID: mdl-31904042

ABSTRACT

Herein, a novel two-photon ratiometric fluorescence assay was proposed for monitoring endogenous steroid sulfatase (STS) activity, which could be applied for the ratiometric imaging of STS activity in the endoplasmic reticulum of living cells and tissues and also could be used to distinguish estrogen-dependent tumor cells from other types of cells.


Subject(s)
Fluorescent Dyes/chemistry , Naphthalimides/chemistry , Steryl-Sulfatase/analysis , Animals , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Fluorescent Dyes/metabolism , Fluorescent Dyes/toxicity , HEK293 Cells , Helix, Snails/enzymology , Humans , Limit of Detection , Microscopy, Fluorescence/methods , Molecular Docking Simulation , Naphthalimides/metabolism , Naphthalimides/toxicity , Photons , Protein Binding , Steryl-Sulfatase/metabolism
15.
Drug Dev Res ; 81(4): 402-418, 2020 06.
Article in English | MEDLINE | ID: mdl-31904877

ABSTRACT

Tuberculosis (TB), a chronic infectious disease, is one of the greatest risks to human beings and 10 million people were diagnosed with TB and 1.6 million died from this disease in 2017. In addition, with the emergence of multidrug-resistant tuberculosis (MDR-TB) and extensively drug-resistant tuberculosis (XDR-TB), the TB situation has become even worse, which has aggravated the mortality and spread of this disease. To overcome this problem, research into novel antituberculosis agents with enhanced activities against MDR-TB, reduced toxicity, and shortened duration of therapy is of great importance. Fortunately, many novel potential anti-TB drug candidates with five-membered rings, which are most likely to be effective against sensitive and resistant strains, have recently entered clinical trials. Different five-membered rings such as furans, pyranoses, thiazoles, pyrazolines, imidazoles, oxazolidinone, thiazolidins, isoxazoles, triazoles, oxadiazoles, thiadiazoles, and tetrazoles have been designed, prepared, and evaluated for their antimycobacterial activity against Mycobacterium tuberculosis. In this article, we highlight the recent advances made in the discovery of novel five-membered ring compounds and focus on their antitubercular activities, toxicity, structure-activity relationships, and mechanisms of action.


Subject(s)
Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy , Antitubercular Agents/adverse effects , Antitubercular Agents/chemistry , Drug Design , Drug Discovery , Humans , Structure-Activity Relationship , Tuberculosis/epidemiology , Tuberculosis/microbiology
16.
Curr Med Chem ; 27(22): 3753-3769, 2020.
Article in English | MEDLINE | ID: mdl-30556497

ABSTRACT

Osteoarthritis (OA) is an age-related degenerative disease, which is characterized by chronic joint pain, inflammation and the damage of joint cartilage. At present, steroidal drugs and nonsteroidal anti-inflammatory drugs (NSAIDS), selective cyclooxygenase-2 (COX-2) inhibitors, are the first-line drugs for the treatment of OA. However, these drugs could lead to some cardiovascular side effects. Therefore, it is urgent to develop novel agents for the treatment of OA. Matrix metalloproteinase-13 (MMP-13), an important member of matrix metalloproteinases (MMPs) family, plays a vital role by degrading type II collagen in articular cartilage and bone in OA. It is noted that MMP-13 is specially expressed in the OA patients, and not in normal adults. In addition, broadspectrum MMP inhibitors could result in some painful and joint-stiffening side effects, called musculoskeletal syndrome (MSS) in the clinical trials. Thus, developing selective MMP-13 inhibitors is a potential strategy for the therapy of OA. In this review, we summarize the recent progress of selective MMP-13 inhibitors including two subfamilies, namely zinc-binding and non-zinc-binding selective MMP-13 inhibitors.


Subject(s)
Osteoarthritis , Cartilage, Articular , Humans , Matrix Metalloproteinase 13 , Matrix Metalloproteinase Inhibitors , Osteoarthritis/drug therapy
17.
Eur J Med Chem ; 183: 111691, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31536895

ABSTRACT

In general, heterocyclic compounds are a significant source of pharmacologically active compounds. Among them, the indole scaffold widely distributes in natural products and bioactive molecules including anti-cancer agents. In view of its unique physic-chemical and biological properties, it has been used as a privileged scaffold in the anti-cancer agents design. So far, many natural and synthetic indole derivatives have been discovered as promising anti-cancer agents used in clinic or clinical evaluations, suggesting its prominent place in anti-cancer drugs development. This review aimed to provide a clear knowledge on the recent development of indoles as anti-cancer agents, such as myeloid cell leukemia-1 (Mcl-1) inhibitors, proviral insertion site in moloney murine leukemia virus (Pim) inhibitors, histone deacetylase (HDAC) inhibitors, silent mating type information regulation 2 homolog (SIRT) inhibitors and tubulin inhibitors, and made an insight into the corresponding structure-activity relationships (SARs). We hope the review could give a guide to develop new anti-cancer agents with greater potency against drug-sensitive and drug-resistant cancers in the future.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Indoles/pharmacology , Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor , Humans , Indoles/chemical synthesis , Indoles/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/metabolism , Tubulin/metabolism
18.
Spectrochim Acta A Mol Biomol Spectrosc ; 223: 117265, 2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31234021

ABSTRACT

Glutathione (GSH) is the most abundant low-molecular-weight cysteine-containing thiol in cells, which plays an essential role in many biological processes. Most reported fluorescent probes towards GSH possess short excitation and emission wavelength, which could result in low tissue penetration, high background fluorescence and photodamage to biological samples. Herein, a novel turn-on fluorescent probe (ADS) with the xanthene skeleton for GSH detection was developed based on a fluorophore, ACF-NH2. The probe had a red light emission (λem = 630 nm) and exhibited a good linear relationship for exogenous GSH (1-6 mM) and a good limit of detection (LOD: 13.1 µM, based on S/N = 3), which implied that it was possible to detect the change of GSH in the living cells (0.5-10 mM) by further structural modification. The probe displayed excellent selectivity for GSH over other analytes and good anti-interference ability. Moreover, cell viability assay indicated that ADS was biocompatible and exhibited very low cytotoxicity. A combination of mass spectrum analysis and density functional theory calculation was performed to explain the sensing mechanism of the probe. In addition, it was applied to image GSH in living cells successfully.


Subject(s)
Fluorescent Dyes/chemistry , Glutathione/analysis , Light , Xanthenes/chemistry , Cell Death , Cell Survival , Fluorescence , Fluorescent Dyes/chemical synthesis , HeLa Cells , Humans , Hydrogen-Ion Concentration , Molecular Probes/chemistry , Time Factors
19.
Bioorg Med Chem ; 27(12): 2572-2578, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30910475

ABSTRACT

A series of novel 2-hydroxyphenyl substituted aminoacetamides was designed by molecular hybridization of the aminoacetamide scaffold and 2-hydroxyphenyl motif. The target compounds were synthesized and their fungicidal activities were evaluated. Some of the target compounds showed excellent antifungal activities against S. sclerotiorum and P. capsici. Significantly, compounds 5e displayed the most potent activity against S. sclerotiorum with EC50 = 2.89 µg/mL, which was lower than that of commercial chlorothalonil. The systematic studies provided strong confidence that the hydroxyl group and the carbonyl group are crucial for the fungicidal activity. Molecular docking studies suggest that SDH enzyme could be one of the potential action targets of our compounds.


Subject(s)
Acetamides/chemistry , Antifungal Agents/chemical synthesis , Drug Design , Molecular Docking Simulation , Acetamides/chemical synthesis , Acetamides/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Ascomycota/drug effects , Binding Sites , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/metabolism , Structure-Activity Relationship , Succinate Dehydrogenase/antagonists & inhibitors , Succinate Dehydrogenase/metabolism
20.
Bioorg Med Chem ; 26(17): 4907-4915, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30170927

ABSTRACT

Anti-apoptotic Bcl-2 family proteins are vital for cancer cells to escape apoptosis, which make them attractive targets for cancer therapy. Recently, a lead compound 1 was found to modestly inhibit the binding of BH3 peptide to Bcl-2 protein with a Ki value of 5.2 µM. Based on this, a series of substituted tyrosine derivatives were developed and tested for their binding affinities to Bcl-2 protein. Results indicated that these compounds exhibited potent binding affinities to Bcl-2 and Mcl-1 protein but not to Bcl-XL protein. Promisingly, compound 6i inhibited the binding of BH3 peptide to Bcl-2 and Mcl-1 protein with a Ki value of 450 and 190 nM respectively, and showed obvious anti-proliferative activities against tested cancer cells.


Subject(s)
Drug Discovery , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Tyrosine/chemistry , Tyrosine/pharmacology , Carbon-13 Magnetic Resonance Spectroscopy , Cell Proliferation/drug effects , Humans , Jurkat Cells , Mass Spectrometry , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Binding , Proto-Oncogene Proteins c-bcl-2/metabolism , Proton Magnetic Resonance Spectroscopy , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL