Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 821
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Anal Chem ; 96(28): 11488-11497, 2024 07 16.
Article in English | MEDLINE | ID: mdl-38970811

ABSTRACT

Synthetic cannabinoids are a widely abused class of dangerous psychoactive substances, especially among youths and young adults. Dozens of such drugs have been identified to date, and new ones continue to emerge. The ability to detect these drugs is important for interdiction efforts and the diagnosis of drug overdose, but existing analytical methods lack broad cross-reactivity to diverse members of this drug family. Here, we have utilized library-immobilized SELEX to generate DNA aptamers that can broadly recognize various members of the indazole-3-carboxamide synthetic cannabinoid family. Using two representatives of this family, AB-FUBINACA and 5F-AMB, we identify two aptamers FUB4 and AMB2F with respective dissociation constants (KDs) of 138 ± 15 and 411 ± 20 nM for their targets. These aptamers can recognize many indazole-based synthetic cannabinoids with high affinity and excellent specificity against natural cannabinoids as well as other structurally similar interferents like serotonin and tryptophan. We use these two aptamers to develop fluorescence strand-displacement sensors that successfully detect these synthetic cannabinoids at concentrations as low as 50 nM in human serum. The sensors can also detect up to 14 different drugs from this family─a major improvement over the six recognized by an existing commercial immunoassay.


Subject(s)
Aptamers, Nucleotide , Cannabinoids , Indazoles , Aptamers, Nucleotide/chemistry , Indazoles/chemistry , Cannabinoids/chemistry , SELEX Aptamer Technique , Humans
2.
Bioconjug Chem ; 35(8): 1251-1257, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39116103

ABSTRACT

The DNA-encoded library (DEL) is a robust tool for chemical biology and drug discovery. In this study, we developed a DNA-compatible light-promoted reaction that is highly efficient and plate-compatible for DEL construction based on the formation of the indazolone scaffold. Employing this high-efficiency approach, we constructed a DEL featuring an indazolone core, which enabled the identification of a novel series of ligands specifically targeting E1A-binding protein (p300) after DEL selection. Taken together, our findings underscore the feasibility of light-promoted reactions in DEL synthesis and unveil promising avenues for developing p300-targeting inhibitors.


Subject(s)
DNA , Drug Discovery , E1A-Associated p300 Protein , Indazoles , Small Molecule Libraries , DNA/chemistry , Indazoles/chemistry , Indazoles/pharmacology , E1A-Associated p300 Protein/antagonists & inhibitors , E1A-Associated p300 Protein/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Drug Discovery/methods , Humans , Gene Library , Ligands
3.
Mol Pharm ; 21(7): 3321-3329, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38843501

ABSTRACT

Poly ADP-ribose polymerase (PARP) plays an important role in the DNA repair process and has become an attractive target for cancer therapy in recent years. Given that niraparib has good clinical efficacy as a PARP inhibitor, this study aimed to develop radiolabeled niraparib derivatives for tumor imaging to detect PARP expression and improve the accuracy of stratified patient therapy. The niraparib isonitrile derivative (CNPN) was designed, synthesized, and radiolabeled to obtain the [99mTc]Tc-CNPN complex with high radiochemical purity (>95%). It was lipophilic and stable in vitro. In HeLa cell experiments, the uptake of [99mTc]Tc-CNPN was effectively inhibited by the ligand CNPN, indicating the binding affinity for PARP. According to the biodistribution studies of HeLa tumor-bearing mice, [99mTc]Tc-CNPN has moderate tumor uptake and can be effectively inhibited, demonstrating its specificity for targeting PARP. The SPECT imaging results showed that [99mTc]Tc-CNPN had tumor uptake at 2 h postinjection. All of the results of this study indicated that [99mTc]Tc-CNPN is a promising tumor imaging agent that targets PARP.


Subject(s)
Indazoles , Piperidines , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Humans , Mice , Piperidines/chemistry , Piperidines/pharmacokinetics , Indazoles/chemistry , Indazoles/pharmacokinetics , HeLa Cells , Poly(ADP-ribose) Polymerase Inhibitors/pharmacokinetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Tissue Distribution , Tomography, Emission-Computed, Single-Photon/methods , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/chemistry , Poly (ADP-Ribose) Polymerase-1/metabolism , Female , Technetium/chemistry , Nitriles/chemistry , Nitriles/pharmacokinetics , Mice, Nude , Mice, Inbred BALB C
4.
Bioorg Chem ; 147: 107391, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677010

ABSTRACT

Apoptosis signal regulated kinase 1 (ASK1, MAP3K5) is a member of the mitogen activated protein kinase (MAPK) signaling pathway, involved in cell survival, differentiation, stress response, and apoptosis. ASK1 kinase inhibition has become a promising strategy for the treatment of Non-alcoholic steatohepatitis (NASH) disease. A series of novel ASK1 inhibitors with indazole scaffolds were designed and synthesized, and their ASK1 kinase activities were evaluated. The System Structure Activity Relationship (SAR) study discovered a promising compound 33c, which has a strong inhibitory effect on ASK1. Noteworthy observations included a discernible reduction in lipid droplets within LO2 cells stained with Oil Red O, coupled with a decrease in LDL, CHO, and TG content within the NASH model cell group. Mechanistic inquiries revealed that compound 33c could inhibit the protein expression levels of the upregulated ASK1-p38/JNK signaling pathway in TNF-α treated HGC-27 cells and regulate apoptotic proteins. In summary, these findings suggest that compound 33c may be valuable for further research as a potential candidate compound against NASH.


Subject(s)
Drug Design , Indazoles , MAP Kinase Kinase Kinase 5 , Molecular Docking Simulation , Protein Kinase Inhibitors , Humans , Apoptosis/drug effects , Dose-Response Relationship, Drug , Indazoles/pharmacology , Indazoles/chemical synthesis , Indazoles/chemistry , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase Kinase 5/metabolism , Molecular Structure , Non-alcoholic Fatty Liver Disease/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism
5.
Bioorg Chem ; 147: 107376, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38640722

ABSTRACT

The inhibition of the programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) pathway with small molecules is a promising approach for cancer immunotherapy. Herein, novel small molecules compounds bearing various scaffolds including thiophene, thiazole, tetrahydroquinoline, benzimidazole and indazole were designed, synthesized and evaluated for their inhibitory activity against the PD-1/PD-L1 interaction. Among them, compound Z13 exhibited the most potent activity with IC50 of 189.6 nM in the homogeneous time-resolved fluorescence (HTRF) binding assay. Surface plasmon resonance (SPR) assay demonstrated that Z13 bound to PD-L1 with high affinity (KD values of 231 nM and 311 nM for hPD-L1 and mPD-L1, respectively). In the HepG2/Jurkat T co-culture cell model, Z13 decreased the viability rate of HepG2 cells in a concentration-dependent manner. In addition, Z13 showed significant in vivo antitumor efficacy (TGI = 52.6 % at 40 mg/kg) without obvious toxicity in the B16-F10 melanoma model. Furthermore, flow cytometry analysis demonstrated that Z13 inhibited tumor growth in vivo by activating the tumor immune microenvironment. These findings indicate that Z13 is a promising PD-1/PD-L1 inhibitor deserving further investigation.


Subject(s)
Antineoplastic Agents , B7-H1 Antigen , Cell Proliferation , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Indazoles , Programmed Cell Death 1 Receptor , Humans , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Structure-Activity Relationship , Indazoles/chemistry , Indazoles/pharmacology , Indazoles/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Animals , Molecular Structure , Mice , Cell Proliferation/drug effects , Drug Discovery , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Mice, Inbred C57BL , Hep G2 Cells , Cell Survival/drug effects
6.
J Nanobiotechnology ; 22(1): 429, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033109

ABSTRACT

Pazopanib (PAZ), an oral multi-tyrosine kinase inhibitor, demonstrates promising cytostatic activities against various human cancers. However, its clinical utility is limited by substantial side effects and therapeutic resistance. We developed a nanoplatform capable of delivering PAZ for enhanced anti-breast cancer therapy. Nanometer-sized PAZ@Fe-MOF, compared to free PAZ, demonstrated increased anti-tumor therapeutic activities in both syngeneic murine 4T1 and xenograft human MDA-MB-231 breast cancer models. High-throughput single-cell RNA sequencing (scRNAseq) revealed that PAZ@Fe-MOF significantly reduced pro-tumorigenic M2-like macrophage populations at tumor sites and suppressed M2-type signaling pathways, such as ATF6-TGFBR1-SMAD3, as well as chemokines including CCL17, CCL22, and CCL24. PAZ@Fe-MOF reprogramed the inhibitory immune microenvironment and curbed tumorigenicity by blocking the polarization of M2 phenotype macrophages. This platform offers a promising and new strategy for improving the cytotoxicity of PAZ against breast cancers. It provides a method to evaluate the immunological response of tumor cells to PAZ-mediated treatment.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Indazoles , Macrophages , Metal-Organic Frameworks , Nanoparticles , Pyrimidines , Sulfonamides , Animals , Female , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Breast Neoplasms/drug therapy , Humans , Macrophages/drug effects , Indazoles/pharmacology , Indazoles/chemistry , Mice , Pyrimidines/pharmacology , Pyrimidines/chemistry , Cell Line, Tumor , Nanoparticles/chemistry , Sulfonamides/pharmacology , Sulfonamides/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice, Inbred BALB C , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
7.
Chem Biodivers ; 21(7): e202302085, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38666662

ABSTRACT

In this work, a highly effective synthesis technique for obtaining aryl indazole under mild circumstances is provided, using trimethyl phosphine as a powerful reagent. The procedure shows that a wide range of substrates can be investigated, yielding various 2-aryl indazole derivatives with acceptable to exceptional yields and a wide range of functional group tolerance. Additionally, based on In Silico studies tests were conducted to determine the anticancer activity In Vitro for all produced compounds (3 a-3 j) against A549, HT-29 and HepG2 cell lines. Compounds 3 c and 3 d, with IC50 values of 15, 53.55, 7.34, 7.10, 56.28, and 17.87 (µM) against A549, HT-29 and HepG2 respectively, showed significant anticancer activity.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Drug Screening Assays, Antitumor , Indazoles , Indazoles/chemical synthesis , Indazoles/pharmacology , Indazoles/chemistry , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Structure-Activity Relationship , Cell Proliferation/drug effects , Molecular Structure , Cell Line, Tumor , Dose-Response Relationship, Drug , Molecular Docking Simulation
8.
Arch Pharm (Weinheim) ; 357(6): e2400020, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38478964

ABSTRACT

Haspin and Clk4 are both understudied protein kinases (PKs), offering potential targets for the development of new anticancer agents. Thus, the identification of new inhibitors targeting these PKs is of high interest. However, the inhibitors targeting haspin or Clk4 developed to date show a poor selectivity profile over other closely related PKs, increasing the risk of side effects. Herein, we present two newly developed N1-benzyolated 5-(4-pyridinyl)indazole-based inhibitors (18 and 19), derived from a newly identified indazole hit. These inhibitors exhibit an exceptional inhibitory profile toward haspin and/or Clk4. Compound 18 (2-acetyl benzoyl) showed a preference to inhibit Clk4 and haspin over a panel of closely related kinases, with sixfold selectivity for Clk4 (IC50 = 0.088 and 0.542 µM, respectively). Compound 19 (4-acetyl benzoyl) showed high selectivity against haspin over the common off-target kinases (Dyrks and Clks) with an IC50 of 0.155 µM for haspin. Molecular docking studies explained the remarkable selectivity of 18 and 19, elucidating how the new scaffold can be modified to toggle between inhibition of haspin or Clk4, despite the high homology of the ATP-binding sites. Their distinguished profile allows these compounds to be marked as interesting chemical probes to assess the selective inhibition of haspin and/or Clk4.


Subject(s)
Indazoles , Protein Kinase Inhibitors , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Indazoles/pharmacology , Indazoles/chemistry , Indazoles/chemical synthesis , Humans , Structure-Activity Relationship , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Molecular Structure , Molecular Docking Simulation , Dose-Response Relationship, Drug , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis
9.
Molecules ; 29(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39124921

ABSTRACT

The inhibitory-kappaB kinases (IKKs) IKKα and IKKß play central roles in regulating the non-canonical and canonical NF-κB signalling pathways. Whilst the proteins that transduce the signals of each pathway have been extensively characterised, the clear dissection of the functional roles of IKKα-mediated non-canonical NF-κB signalling versus IKKß-driven canonical signalling remains to be fully elucidated. Progress has relied upon complementary molecular and pharmacological tools; however, the lack of highly potent and selective IKKα inhibitors has limited advances. Herein, we report the development of an aminoindazole-pyrrolo[2,3-b]pyridine scaffold into a novel series of IKKα inhibitors. We demonstrate high potency and selectivity against IKKα over IKKß in vitro and explain the structure-activity relationships using structure-based molecular modelling. We show selective target engagement with IKKα in the non-canonical NF-κB pathway for both U2OS osteosarcoma and PC-3M prostate cancer cells by employing isoform-related pharmacodynamic markers from both pathways. Two compounds (SU1261 [IKKα Ki = 10 nM; IKKß Ki = 680 nM] and SU1349 [IKKα Ki = 16 nM; IKKß Ki = 3352 nM]) represent the first selective and potent pharmacological tools that can be used to interrogate the different signalling functions of IKKα and IKKß in cells. Our understanding of the regulatory role of IKKα in various inflammatory-based conditions will be advanced using these pharmacological agents.


Subject(s)
Drug Design , I-kappa B Kinase , NF-kappa B , Protein Kinase Inhibitors , Signal Transduction , I-kappa B Kinase/metabolism , I-kappa B Kinase/antagonists & inhibitors , Humans , NF-kappa B/metabolism , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Structure-Activity Relationship , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Cell Line, Tumor , Pyridines/pharmacology , Pyridines/chemistry , Pyridines/chemical synthesis , Indazoles/pharmacology , Indazoles/chemistry , Indazoles/chemical synthesis , Models, Molecular
10.
Rapid Commun Mass Spectrom ; 37(13): e9524, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37062936

ABSTRACT

RATIONALE: Mass spectrometry has evolved into a highly powerful tool for qualitative and quantitative chemical analyses. However, the identification of trace amounts of previously unknown structures in complex chemical matrix environments remains challenging. The rapid emergence of new synthetic cannabinoid substances is a typical example of this. Existing laboratory techniques are mostly based on methods used for lists of known illegal compounds. This situation poses a challenge to traditional data analysis and the risk of missing the compounds. Therefore, we propose to develop and validate a statistical model to classify newly emerging synthetic cannabinoid substances into a structural class or subclass. METHODS: We obtained 70 electrospray ionization spectra of indole/indazole synthetic cannabinoids from both the actual standard analysis and the SWGDRUG mass spectral library (version 3.10). Each sample consisted of 330 m/z variables and corresponding relative intensities. We first cleared the variables with a variance below 0.1. Principal component analysis (PCA) was performed on the variance-filtered data, and the two principal components were retained to generate new data for hierarchical clustering. After hierarchical clustering, we used the receiver operating characteristic method in this cluster. RESULTS: Seventy synthetic indole/indazole cannabinoids were classified into four clusters. The side chain of cluster 1 is mainly fluorobenzyl, cluster 2 is pentyl, cluster 3 includes compounds from several structures, and cluster 4 is mainly fluoropentyl. The most relevant characteristic ions are m/z 109, m/z 252, and m/z 253 for cluster 1; m/z 144 and m/z 214 for cluster 2; and m/z 232 and m/z 233 for cluster 4. CONCLUSIONS: This study provides a more objective and less time-consuming solution for characterizing synthetic cannabinoids. And this work validates the ability of PCA to extract characteristic fragment ions.


Subject(s)
Cannabinoids , Indazoles , Gas Chromatography-Mass Spectrometry/methods , Indazoles/chemistry , Indoles/chemistry , Cannabinoids/analysis , Ions
11.
Bioorg Med Chem ; 90: 117377, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37352576

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO1) is a heme-containing enzyme mainly responsible for the metabolism of tryptophan to kynurenine. To date, the IDO1 inhibitors have been developed intensively for the re-activation of the anticancer immune response. In this report, we designed, and synthesized novel 1,3-dimethyl-6-amino indazole derivatives as IDO1 inhibitors based on the structure of IDO1 active site. We further examined their anticancer activity on hypopharyngeal carcinoma cells (FaDu), squamous cell carcinoma of the oral tongue (YD-15), breast cancer cells (MCF7), and human dental pulp stem cells (HDPSC). Of them, compound N-(4-bromobenzyl)-1,3-dimethyl-1H-indazol-6-amine (7) remarkably suppressed IDO1 expression in a concentration - dependent manner. In addition, 7 was the most potential anticancer compound with inducing apoptosis activity as well as selectively activated extracellular signal-regulated kinases (ERK) in mitogen-activated protein kinase (MAPK) pathways on FaDu cells. Finally, compound 7 suppressed cell mobility in wound healing assay with the reduced expression of matrix metalloproteinase MMP9. Taken together, we believe that 7 is the most promising compound, which may be applied to treatment of hypopharyngeal carcinoma.


Subject(s)
Antineoplastic Agents , Carcinoma , Humans , Indazoles/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Tryptophan , Indoleamine-Pyrrole 2,3,-Dioxygenase , Enzyme Inhibitors/chemistry
12.
Bioorg Chem ; 133: 106412, 2023 04.
Article in English | MEDLINE | ID: mdl-36773456

ABSTRACT

The PI3K/AKT/mTOR signaling pathway is one of the most common abnormal activation pathways in tumor cells, and has associated with multiple functions such as tumor cell growth, proliferation, migration, invasion, and tumor angiogenesis. Here, a series of 3-amino-1H-indazole derivatives were synthesized, and their antiproliferative activities against HT-29, MCF-7, A-549, HepG2 and HGC-27 cells were evaluated. Among them, W24 exhibited the broad-spectrum antiproliferative activity against four cancer cells with IC50 values of 0.43-3.88 µM. Mechanism studies revealed that W24 inhibited proliferation by affecting the DNA synthesis, induced G2/M cell cycle arrest and apoptosis by regulating Cyclin B1, BAD and Bcl-xL, meanwhile induced the change of intracellular ROS and mitochondrial membrane potential in HGC-27 cells. Moreover, W24 inhibited the migration and invasion of HGC-27 cells by decreasing EMT pathway related proteins and reducing the mRNA expression levels of Snail, Slug and HIF-1α. Furthermore, W24 displayed low tissue toxicity profile and good pharmacokinetic properties in vivo. Therefore, 3-amino-1H-indazole derivatives might serve as a new scaffold for the development of PI3K/AKT/mTOR inhibitor and anti-gastric cancer reagent.


Subject(s)
Indazoles , Neoplasms , Humans , Apoptosis , Cell Line, Tumor , Cell Proliferation , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Indazoles/chemistry , Indazoles/pharmacology
13.
Nature ; 550(7677): 534-538, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29045385

ABSTRACT

The ubiquitin system regulates essential cellular processes in eukaryotes. Ubiquitin is ligated to substrate proteins as monomers or chains and the topology of ubiquitin modifications regulates substrate interactions with specific proteins. Thus ubiquitination directs a variety of substrate fates including proteasomal degradation. Deubiquitinase enzymes cleave ubiquitin from substrates and are implicated in disease; for example, ubiquitin-specific protease-7 (USP7) regulates stability of the p53 tumour suppressor and other proteins critical for tumour cell survival. However, developing selective deubiquitinase inhibitors has been challenging and no co-crystal structures have been solved with small-molecule inhibitors. Here, using nuclear magnetic resonance-based screening and structure-based design, we describe the development of selective USP7 inhibitors GNE-6640 and GNE-6776. These compounds induce tumour cell death and enhance cytotoxicity with chemotherapeutic agents and targeted compounds, including PIM kinase inhibitors. Structural studies reveal that GNE-6640 and GNE-6776 non-covalently target USP7 12 Å distant from the catalytic cysteine. The compounds attenuate ubiquitin binding and thus inhibit USP7 deubiquitinase activity. GNE-6640 and GNE-6776 interact with acidic residues that mediate hydrogen-bond interactions with the ubiquitin Lys48 side chain, suggesting that USP7 preferentially interacts with and cleaves ubiquitin moieties that have free Lys48 side chains. We investigated this idea by engineering di-ubiquitin chains containing differential proximal and distal isotopic labels and measuring USP7 binding by nuclear magnetic resonance. This preferential binding protracted the depolymerization kinetics of Lys48-linked ubiquitin chains relative to Lys63-linked chains. In summary, engineering compounds that inhibit USP7 activity by attenuating ubiquitin binding suggests opportunities for developing other deubiquitinase inhibitors and may be a strategy more broadly applicable to inhibiting proteins that require ubiquitin binding for full functional activity.


Subject(s)
Aminopyridines/chemistry , Aminopyridines/pharmacology , Indazoles/chemistry , Indazoles/pharmacology , Phenols/chemistry , Phenols/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Ubiquitin/metabolism , Animals , Binding, Competitive , Cell Line, Tumor , Drug Synergism , Female , Humans , Mice , Mice, SCID , Models, Molecular , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/pathology , Protein Binding , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Substrate Specificity , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin/chemistry , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/deficiency , Ubiquitin-Specific Peptidase 7/metabolism
14.
Int J Mol Sci ; 24(2)2023 Jan 16.
Article in English | MEDLINE | ID: mdl-36675281

ABSTRACT

An efficient method for the synthesis of pyrazolo [4,3-b]pyridines has been developed on the basis of readily available 2-chloro-3-nitropyridines via a sequence of SNAr and modified Japp-Klingemann reactions. The method offers a number of advantages including utilization of stable arenediazonium tosylates, operational simplicity as well as combining the azo-coupling, deacylation and pyrazole ring annulation steps in a one-pot manner. An unusual rearrangement (C-N-migration of the acetyl group) was observed and a plausible mechanism was proposed based on the isolated intermediates and NMR experiments. In addition, the developed protocol was successfully applied to the synthesis of 1-arylindazoles combining the Japp-Klingemann reaction and cyclization of the resulting hydrazone as a one-pot procedure.


Subject(s)
Indazoles , Pyridines , Pyridines/chemistry , Indazoles/chemistry , Cyclization
15.
Fa Yi Xue Za Zhi ; 39(5): 457-464, 2023 Oct 25.
Article in English, Zh | MEDLINE | ID: mdl-38006265

ABSTRACT

OBJECTIVES: To establish the GC-MS qualitative and quantitative analysis methods for the synthetic cannabinoids, its main matrix and additives in suspicious electronic cigarette (e-cigarette) oil samples. METHODS: The e-cigarette oil samples were analyzed by GC-MS after diluted with methanol. Synthetic cannabinoids, its main matrix and additives in e-cigarette oil samples were qualitatively analyzed by the characteristic fragment ions and retention time. The synthetic cannabinoids were quantitatively analyzed by using the selective ion monitoring mode. RESULTS: The linear range of each compound in GC-MS quantitative method was 0.025-1 mg/mL, the matrix recovery rate was 94%-103%, the intra-day precision relative standard deviations (RSD) was less than 2.5%, and inter-day precision RSD was less than 4.0%. Five indoles or indazole amide synthetic cannabinoids were detected in 25 e-cigarette samples. The main matrixes of e-cigarette samples were propylene glycol and glycerol. Additives such as N,2,3-trimethyl-2-isopropyl butanamide (WS-23), glycerol triacetate and nicotine were detected in some samples. The content range of synthetic cannabinoids in 25 e-cigarette samples was 0.05%-2.74%. CONCLUSIONS: The GC-MS method for synthesizing cannabinoid, matrix and additive in e-cigarette oil samples has good selectivity, high resolution, low detection limit, and can be used for simultaneous qualitative and quantitative analysis of multiple components; The explored fragment ion fragmentation mechanism of the electron bombardment ion source of indole or indoxamide compounds helps to identify such substances or other compounds with similar structures in cases.


Subject(s)
Cannabinoids , Electronic Nicotine Delivery Systems , Illicit Drugs , Gas Chromatography-Mass Spectrometry/methods , Illicit Drugs/analysis , Indazoles/chemistry , Glycerol/analysis , Indoles/chemistry , Ions
16.
J Org Chem ; 87(24): 16188-16203, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36417354

ABSTRACT

A copper-promoted regiodivergent, AcOH-switchable, distal and proximal direct cyanation of N-aryl-(1H/2H)-indazoles via aerobic oxidative C(sp2)-H bond activation has been developed. The inclusion or exclusion of AcOH as an additive is the foremost cause for the positional switch in the C-CN bond formation method that results in (C-2')-cyanated 2-aryl-2H-indazoles 3a-j, (C-2')-cyanated 1-aryl-1H-indazoles 4a-j [distal], or C-3 cyanated 2-aryl-2H-indazoles 5a-i [proximal] products in good to excellent yields and showed various functional group tolerance. The cyanide (CN-) ion surrogate was generated via the unification of dimethylformamide and ammonium iodide (NH4I). The utilization of molecular oxygen (aerobic oxidative strategy) as a clean and safe oxidant is liable for generous value addition. The further pertinence of the developed protocol has been demonstrated by transforming the synthesized cyanated product into numerous other functional groups, which will, undoubtedly, accomplish utilization in the synthetic area of biologically important compounds and medicinal chemistry.


Subject(s)
Copper , Indazoles , Indazoles/chemistry , Catalysis , Oxidation-Reduction , Copper/chemistry , Oxidative Stress
17.
Org Biomol Chem ; 20(45): 8893-8897, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36317604

ABSTRACT

A new iodine(III)-mediated oxidative dearomatization of 2H-indazoles has been developed to afford N-1 indazolyl indazolones. In this methodology, PIFA plays a dual role: as an oxidant and as a carbonyl oxygen source. A series of indazolone derivatives was promptly synthesized in good to excellent yields through sequential C-heteroatom bond formation. Mechanistic studies indicate that the reaction likely takes place through an SET pathway.


Subject(s)
Indazoles , Iodine , Indazoles/chemistry , Iodine/chemistry , Oxidation-Reduction , Iodides , Oxidative Stress
18.
J Enzyme Inhib Med Chem ; 37(1): 151-167, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34894940

ABSTRACT

An efficient pathway was disclosed for the synthesis of 3-chloro-6-nitro-1H-indazole derivatives by 1,3-dipolar cycloaddition on dipolarophile compounds 2 and 3. Faced the problem of separation of two regioisomers, a click chemistry method has allowed us to obtain regioisomers of triazole-1,4 with good yields from 82 to 90% were employed. Also, the antileishmanial biological potency of the compounds was achieved using an MTT assay that reported compound 13 as a promising growth inhibitor of Leishmania major. Molecular docking demonstrated highly stable binding with the Leishmania trypanothione reductase enzyme and produced a network of hydrophobic and hydrophilic interactions. Molecular dynamics simulations were performed for TryR-13 complex to understand its structural and intermolecular affinity stability in a biological environment. The studied complex remained in good equilibrium with a structure deviation of ∼1-3 Å. MM/GBSA binding free energies illustrated the high stability of TryR-13 complex. The studied compounds are promising leads for structural optimisation to enhance the antileishmanial activity.


Subject(s)
Antiprotozoal Agents/pharmacology , Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Leishmania major/drug effects , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Indazoles/chemical synthesis , Indazoles/chemistry , Leishmania major/enzymology , Models, Molecular , Molecular Structure , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/metabolism , Parasitic Sensitivity Tests , Structure-Activity Relationship
19.
Molecules ; 27(15)2022 Aug 03.
Article in English | MEDLINE | ID: mdl-35956893

ABSTRACT

Designing new synthetic strategies for indazoles is a prominent topic in contemporary research. The transition-metal-catalyzed C-H activation/annulation sequence has arisen as a favorable tool to construct functionalized indazole derivatives with improved tolerance in medicinal applications, functional flexibility, and structural complexity. In the current review article, we aim to outline and summarize the most common synthetic protocols to use in the synthesis of target indazoles via a transition-metal-catalyzed C-H activation/annulation sequence for the one-step synthesis of functionalized indazole derivatives. We categorized the text according to the metal salts used in the reactions. Some metal salts were used as catalysts, and others may have been used as oxidants and/or for the activation of precatalysts. The roles of some metal salts in the corresponding reaction mechanisms have not been identified. It can be expected that the current synopsis will provide accessible practical guidance to colleagues interested in the subject.


Subject(s)
Indazoles , Transition Elements , Catalysis , Indazoles/chemistry , Metals/chemistry , Salts , Transition Elements/chemistry
20.
Toxicol Appl Pharmacol ; 428: 115648, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34280409

ABSTRACT

Acute lung injury (ALI) is a diffuse lung dysfunction disease characterized by high prevalence and high mortality. Thus far, no effective pharmacological treatment has been made for ALI in clinics. Inflammation is critical to the development of ALI. Therefore, anti-inflammation may be a potential therapy strategy for ALI. Indazole-containing derivatives, representing one of the most important heterocycles in drug molecules, are endowed with a broad range of biological properties, such as anti-cancer and anti-inflammation. In the current study, we investigated the biological effects of Cyy-272, a newly synthesized indazole compound, on LPS-induced ALI both in vivo and in vitro. Results show that Cyy-272 can inhibit the release of inflammatory cytokines in LPS-stimulated macrophage and alleviate LPS induced ALI. Further experiment revealed that Cyy-272 exhibit anti-inflammation activity by inhibiting JNK phosphorylation. Overall, our studies show that an indazole derivative, Cyy-272, is effective in suppressing LPS-induced JNK activation and inflammatory signaling.


Subject(s)
Acute Lung Injury/chemically induced , Acute Lung Injury/prevention & control , Indazoles/therapeutic use , Lipopolysaccharides/toxicity , MAP Kinase Kinase 4/antagonists & inhibitors , Acute Lung Injury/metabolism , Animals , Dose-Response Relationship, Drug , Indazoles/chemistry , Indazoles/pharmacology , MAP Kinase Kinase 4/chemistry , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Structure, Tertiary , RAW 264.7 Cells
SELECTION OF CITATIONS
SEARCH DETAIL