Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.146
Filter
1.
Molecules ; 29(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731442

ABSTRACT

Two series, "a" and "b", each consisting of nine chemical compounds, with 2,3-disubstituted quinazolin-4(3H)-one scaffold, were synthesized and evaluated for their anticonvulsant activity. They were investigated as dual potential positive allosteric modulators of the GABAA receptor at the benzodiazepine binding site and inhibitors of carbonic anhydrase II. Quinazolin-4(3H)-one derivatives were evaluated in vivo (D1-3 = 50, 100, 150 mg/kg, administered intraperitoneally) using the pentylenetetrazole (PTZ)-induced seizure model in mice, with phenobarbital and diazepam, as reference anticonvulsant agents. The in silico studies suggested the compounds act as anticonvulsants by binding on the allosteric site of GABAA receptor and not by inhibiting the carbonic anhydrase II, because the ligands-carbonic anhydrase II predicted complexes were unstable in the molecular dynamics simulations. The mechanism targeting GABAA receptor was confirmed through the in vivo flumazenil antagonism assay. The pentylenetetrazole experimental anticonvulsant model indicated that the tested compounds, 1a-9a and 1b-9b, present a potential anticonvulsant activity. The evaluation, considering the percentage of protection against PTZ, latency until the onset of the first seizure, and reduction in the number of seizures, revealed more favorable results for the "b" series, particularly for compound 8b.


Subject(s)
Anticonvulsants , Pentylenetetrazole , Receptors, GABA-A , Seizures , Anticonvulsants/pharmacology , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Animals , Mice , Seizures/drug therapy , Seizures/chemically induced , Receptors, GABA-A/metabolism , Quinazolinones/pharmacology , Quinazolinones/chemistry , Quinazolinones/chemical synthesis , Molecular Docking Simulation , Male , Structure-Activity Relationship , Molecular Dynamics Simulation , Computer Simulation , Disease Models, Animal , Molecular Structure , Allosteric Site
2.
Molecules ; 29(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38731603

ABSTRACT

A new quinazolinone alkaloid named peniquinazolinone A (1), as well as eleven known compounds, 2-(2-hydroxy-3-phenylpropionamido)-N-methylbenzamide (2), viridicatin (3), viridicatol (4), (±)-cyclopeptin (5a/5b), dehydrocyclopeptin (6), cyclopenin (7), cyclopenol (8), methyl-indole-3-carboxylate (9), 2,5-dihydroxyphenyl acetate (10), methyl m-hydroxyphenylacetate (11), and conidiogenone B (12), were isolated from the endophytic Penicillium sp. HJT-A-6. The chemical structures of all the compounds were elucidated by comprehensive spectroscopic analysis, including 1D and 2D NMR and HRESIMS. The absolute configuration at C-13 of peniquinazolinone A (1) was established by applying the modified Mosher's method. Compounds 2, 3, and 7 exhibited an optimal promoting effect on the seed germination of Rhodiola tibetica at a concentration of 0.01 mg/mL, while the optimal concentration for compounds 4 and 9 to promote Rhodiola tibetica seed germination was 0.001 mg/mL. Compound 12 showed optimal seed-germination-promoting activity at a concentration of 0.1 mg/mL. Compared with the positive drug 6-benzyladenine (6-BA), compounds 2, 3, 4, 7, 9, and 12 could extend the seed germination period of Rhodiola tibetica up to the 11th day.


Subject(s)
Alkaloids , Penicillium , Quinazolinones , Rhodiola , Seeds , Penicillium/chemistry , Quinazolinones/chemistry , Quinazolinones/pharmacology , Rhodiola/chemistry , Rhodiola/microbiology , Alkaloids/chemistry , Alkaloids/pharmacology , Alkaloids/isolation & purification , Germination/drug effects , Molecular Structure , Endophytes/chemistry
3.
Int Immunopharmacol ; 133: 112001, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38608443

ABSTRACT

Acute kidney injury (AKI) is a critical complication known for their extremely high mortality rate and lack of effective clinical therapy. Disorders in mitochondrial dynamics possess a pivotal role in the occurrence and progression of contrast-induced nephropathy (CIN) by activating NLRP3 inflammasome. The activation of dynamin-related protein-1 (Drp1) can trigger mitochondrial dynamic disorders by regulating excessive mitochondrial fission. However, the precise role of Drp1 during CIN has not been clarified. In vivo experiments revealed that inhibiting Drp1 through Mdivi-1 (one selective inhibitor of Drp1) can significantly decrease the expression of p-Drp1 (Ser616), mitochondrial p-Drp1 (Ser616), mitochondrial Bax, mitochondrial reactive oxygen species (mROS), NLRP3, caspase-1, ASC, TNF-α, IL-1ß, interleukin (IL)-18, IL-6, creatinine (Cr), malondialdehyde (MDA), blood urea nitrogen (BUN), and KIM-1. Moreover, Mdivi-1 reduced kidney pathological injury and downregulated the interaction between NLRP3 and thioredoxin-interacting protein (TXNIP), which was accompanied by decreased interactions between TRX and TXNIP. This resulted in increasing superoxide dismutase (SOD) and CAT activity, TRX expression, up-regulating mitochondrial membrane potential, and augmenting ATP contents and p-Drp1 (Ser616) levels in the cytoplasm. However, it did not bring impact on the expression of p-Drp1 (Ser637) and TXNIP. Activating Drp-1though Acetaldehyde abrogated the effects of Mdivi-1. In addition, the results of in vitro studies employing siRNA-Drp1 and plasmid-Drp1 intervention in HK-2 cells treated with iohexol were consistent with the in vivo experiments. Our findings revealed inhibiting Drp1 phosphorylation at Ser616 could ameliorate iohexol -induced acute kidney injury though alleviating the activation of the TXNIP-NLRP3 inflammasome pathway.


Subject(s)
Acute Kidney Injury , Carrier Proteins , Contrast Media , Dynamins , Inflammasomes , Mitochondrial Dynamics , NLR Family, Pyrin Domain-Containing 3 Protein , Quinazolinones , Reactive Oxygen Species , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Dynamins/metabolism , Animals , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/drug therapy , Mitochondrial Dynamics/drug effects , Inflammasomes/metabolism , Carrier Proteins/metabolism , Carrier Proteins/genetics , Male , Quinazolinones/pharmacology , Quinazolinones/therapeutic use , Mice , Contrast Media/adverse effects , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL , Humans , Signal Transduction/drug effects , Thioredoxins/metabolism , Thioredoxins/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Cell Line
4.
Biochem Biophys Res Commun ; 710: 149886, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38581953

ABSTRACT

Mdivi-1, Mitochondrial DIVIsion inhibitor 1, has been widely employed in research under the assumption that it exclusively influences mitochondrial fusion, but effects other than mitochondrial dynamics have been underinvestigated. This paper provides transcriptome and DNA methylome-wide analysis for Mdivi-1 treated SH-SY5Y human neuroblastoma cells using RNA sequencing (RNA-seq) and methyl capture sequencing (MC-seq) methods. Gene ontology analysis of RNA sequences revealed that p53 transcriptional gene network and DNA replication initiation-related genes were significantly up and down-regulated, respectively, showing the correlation with the arrest cell cycle in the G1 phase. MC-seq, a powerful sequencing method for capturing DNA methylation status in CpG sites, revealed that although Mdivi-1 does not induce dramatic DNA methylation change, the subtle alterations were concentrated within the CpG island. Integrative analysis of both sequencing data disclosed that the p53 transcriptional network was activated while the Parkinson's disease pathway was halted. Next, we investigated several changes in mitochondria in response to Mdivi-1. Copy number and transcription of mitochondrial DNA were suppressed. ROS levels increased, and elevated ROS triggered mitochondrial retrograde signaling rather than inducing direct DNA damage. In this study, we could better understand the molecular network of Mdivi-1 by analyzing DNA methylation and mRNA transcription in the nucleus and further investigating various changes in mitochondria, providing inspiration for studying nuclear-mitochondrial communications.


Subject(s)
Dynamins , Neuroblastoma , Humans , Dynamins/metabolism , Mitochondrial Dynamics , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Quinazolinones/pharmacology
5.
Int Immunopharmacol ; 130: 111790, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38447417

ABSTRACT

OBJECTIVE: Diabetic kidney disease (DKD) is the most common cause of the end-stage renal disease, which has limited treatment options. Rutaecarpine has anti-inflammatory effects, however, it has not been studied in DKD. Pyroptosis is a newly discovered mode of podocyte death related to inflammation. This study aimed to explore whether Rutaecarpine can ameliorate DKD and to clarify its possible mechanism. METHODS: In this study, we investigated the effects of Rutaecarpine on DKD using diabetic mice model (db/db mice) and high glucose (HG)-stimulated mouse podocyte clone 5 (MPC5) cells. Quantitative reverse transcription polymerase chain reaction and western blot were performed to detect the related gene and protein levels. We applied pharmacological prediction, co-immunoprecipitation assay, cellular thermal shift assay, surface plasmon resonance to find the target and pathway of the substances. Gene knockdown experiments confirmed this view in HG-stimulated MPC5 cells. RESULTS: Rutaecarpine significantly reduced proteinuria, histopathological damage, and pyroptosis of podocytes in a dose-dependent manner in db/db mice. Rutaecarpine also protected high glucose induced MPC5 injury in vitro experiments. Mechanistically, Rutaecarpine can inhibit pyroptosis in HG-stimulated MPC5 by reducing the expression of VEGFR2. VEGFR2 is a target of Rutaecarpine in MPC5 cells and directly binds to the pyroptosis initiation signal, NLRP3. VEGFR2-knockdown disrupted the beneficial effects of Rutaecarpine in HG-stimulated MPC5 cells. CONCLUSION: Rutaecarpine inhibits renal inflammation and pyroptosis through VEGFR2/NLRP3 pathway, thereby alleviating glomerular podocyte injury. These findings highlight the potential of Rutaecarpine as a novel drug for DKD treatment.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Indole Alkaloids , Podocytes , Pyroptosis , Quinazolinones , Animals , Mice , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/drug therapy , Glucose/metabolism , Indole Alkaloids/pharmacology , Indole Alkaloids/therapeutic use , Inflammation/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Podocytes/drug effects , Pyroptosis/drug effects , Quinazolinones/pharmacology , Quinazolinones/therapeutic use , Mice, Inbred C57BL , Male
6.
Sci Rep ; 14(1): 3530, 2024 02 12.
Article in English | MEDLINE | ID: mdl-38347004

ABSTRACT

A series of novel azepine derivatives based on quinazolinone moiety was synthesized through the reaction of quinazolinone chalcones (2a-d) either with 2-amino aniline in acidic medium to give diazepines (3a-d) or with 2-aminophenol to offer oxazepine (4a-d). The structure of the synthesized compounds was confirmed via melting points, elemental analyses, and different spectroscopic techniques. Moreover, these newly compounds mode of action was investigated in-silico using molecular docking against the outer membrane protein A (OMPA), exo-1,3-beta-glucanase for their antimicrobial activity, and against Smoothened (SMO), transcription factor glioma-associated homology (SUFU/GLI-1), the main proteins of Hedgehog signaling pathway to inspect their anticancer potential. Our results showed that, diazepine (3a) and oxazepine (4a) offered the highest binding energy against the target OMPA/ exo-1,3-beta-glucanase proteins and exhibited the potent antimicrobial activities against E. coli, P. aeruginosa, S. aureus, B. subtilis, C. Albicans and A. flavus. As well, diazepine (3a) and oxazepine (4a) achieved the best results among the other compounds, in their binding energy against the target SMO, SUFU/GLI-1 proteins. The in-vitro cytotoxic study was done for them on panel of cancer cell lines HCT-116, HepG2, and MCF-7 and normal cell line WI-38. Conclusively, it was revealed that molecular docking in-silico simulations and the in-vitro experiments were agreed. As a result, our findings elucidated that diazepine (3a) and oxazepine (4a), have the potential to be used as antimicrobial agents and as possible cancer treatment medications.


Subject(s)
Anti-Infective Agents , Antineoplastic Agents , Oxazepines , Structure-Activity Relationship , Molecular Docking Simulation , Hedgehog Proteins , Quinazolinones/pharmacology , Cell Proliferation , Escherichia coli/metabolism , Staphylococcus aureus/metabolism , Glucan 1,3-beta-Glucosidase , Oxazepines/pharmacology , Prospective Studies , Anti-Infective Agents/pharmacology , Antineoplastic Agents/chemistry , Molecular Structure , Drug Screening Assays, Antitumor
7.
Hypertens Res ; 47(5): 1338-1349, 2024 May.
Article in English | MEDLINE | ID: mdl-38383894

ABSTRACT

Mitochondrial dysfunction has been implicated in various types of cardiovascular disease including hypertension. Mitochondrial fission fusion balance is critical to mitochondrial quality control, whereas enhanced fission has been reported in several models of cardiovascular disease. However, limited information is available regarding the contribution of mitochondrial fission in hypertension. Here, we have tested the hypothesis that inhibition of mitochondrial fission attenuates the development of hypertension and associated vascular remodeling. In C57BL6 mice infused with angiotensin II for 2 weeks, co-treatment of mitochondrial fission inhibitor, mdivi1, significantly inhibited angiotensin II-induced development of hypertension assessed by radiotelemetry. Histological assessment of hearts and aortas showed that mdivi1 inhibited vessel fibrosis and hypertrophy induced by angiotensin II. This was associated with attenuation of angiotensin II-induced decline in mitochondrial aspect ratio seen in both the endothelial and medial layers of aortas. Mdivi1 also mitigated angiotensin II-induced cardiac hypertrophy assessed by heart weight-to-body weight ratio as well as by echocardiography. In ex vivo experiments, mdivi1 inhibited vasoconstriction and abolished the enhanced vascular reactivity by angiotensin II in small mesenteric arteries. Proteomic analysis on endothelial cell culture media with angiotensin II and/or mdivi1 treatment revealed that mdivi1 inhibited endothelial cell hypersecretory phenotype induced by angiotensin II. In addition, mdivi1 attenuated angiotensin II-induced protein induction of periostin, a myofibroblast marker in cultured vascular fibroblasts. In conclusion, these data suggest that mdivi1 prevented angiotensin II-induced hypertension and cardiovascular remodeling via multicellular mechanisms in the vasculature.


Subject(s)
Angiotensin II , Hypertension , Mice, Inbred C57BL , Mitochondrial Dynamics , Animals , Angiotensin II/pharmacology , Hypertension/chemically induced , Hypertension/prevention & control , Mitochondrial Dynamics/drug effects , Mice , Male , Quinazolinones/pharmacology , Vascular Remodeling/drug effects , Blood Pressure/drug effects
8.
J Biosci Bioeng ; 137(3): 165-172, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38212152

ABSTRACT

6,7-Bis-(2-methoxyethoxy)-4(3H)-quinazolinone (BMEQ) was selected from quinazolinones for its strong tyrosinase inhibitory activity (IC50 = 160 ± 6 µM). It suppressed tyrosinase activity in a competitive way and quenched the fluorescence of the enzyme through a static mechanism. The binding of BMEQ to tyrosinase increased the hydrophobicity of the latter and facilitated non-radiative energy transfer between them. The formation of BMEQ-tyrosinase complex was driven by hydrogen bonds and hydrophobic interactions, and it loosened the basic framework structure of tyrosinase, affecting the conformation of the enzyme, and leading to a decrease in tyrosinase activity. In addition, the BMEQ postponed the oxidation of phenolics and flavonoids by inhibiting polyphenol oxidase (PPO) and peroxidase (POD), which resulted in the inhibition of the browning of fresh-cut apples. This study identified a novel tyrosinase inhibitor BMEQ and verified its potential application for improving the preservation of postharvest fruits.


Subject(s)
Malus , Monophenol Monooxygenase , Quinazolinones/pharmacology , Fruit
9.
Chem Biodivers ; 21(2): e202301737, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38204291

ABSTRACT

A series of flavonol derivatives containing quinazolinone were designed and synthesized, and their antiviral activities against tobacco mosaic virus (TMV) were evaluated. The results of the half maximal effective concentration (EC50 ) test against TMV showed that the EC50 value of curative activity of K5 was 139.6 µg/mL, which was better than that of the commercial drug ningnanmycin (NNM) 296.0 µg/mL, and the EC50 value of protective activity of K5 was 120.6 µg/mL, which was superior to that of NNM 207.0 µg/mL. The interaction of K5 with TMV coat protein (TMV-CP) was investigated using microscale thermophoresis (MST) and molecular docking and the results showed that K5 can combine with TMV-CP more strongly to TMV-CP than that NNM can. Furthermore, the assay measuring malondialdehyde (MDA) content indicated that K5 had the ability to improve the disease resistance of tobacco. Hence, this study offers strong evidence that flavonol derivatives have potential as novel antiviral agents.


Subject(s)
Quinazolinones , Tobacco Mosaic Virus , Structure-Activity Relationship , Quinazolinones/pharmacology , Molecular Docking Simulation , Antiviral Agents/pharmacology , Microbial Sensitivity Tests , Drug Design
10.
Bioorg Chem ; 144: 107086, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38219478

ABSTRACT

The upregulation of RecQ helicases has been associated with cancer cell survival and resistance to chemotherapy, making them appealing targets for therapeutic intervention. In this study, twenty-nine novel quinazolinone derivatives were designed and synthesized. The anti-proliferative activity of all compounds was evaluated against 60 cancer cell lines at the National Cancer Institute Developmental Therapeutic Program, with six compounds (11f, 11g, 11k, 11n, 11p, and 11q) being promoted to a five-dose screen. Compound 11g demonstrated high cytotoxic activity against all examined cell lines. The compounds were further assayed for Bloom syndrome (BLM) helicase inhibition, where 11g, 11q, and 11u showed moderate activity. These compounds were counter-screened against WRN and RECQ1 helicases, where 11g moderately inhibited both enzymes. An ATP competition assay confirmed that the compounds bound to the ATP site of RecQ helicases, and molecular docking simulations were used to study the binding mode within the active site of BLM, WRN, and RECQ1 helicases. Compound 11g induced apoptosis in both HCT-116 and MDA-MB-231 cell lines, but also caused an G2/M phase cell cycle arrest in HCT-116 cells. This data revealed the potential of 11g as a modulator of cell cycle dynamics and supports its interaction with RecQ helicases. In addition, compound 11g displayed non-significant toxicity against FCH normal colon cells at doses up to 100 µM, which confirming its high safety margin and selectivity on cancer cells. Overall, these findings suggest compound 11g as a potential pan RecQ helicase inhibitor with high anticancer potency and a favorable safety margin and selectivity.


Subject(s)
Antineoplastic Agents , RecQ Helicases , Molecular Docking Simulation , RecQ Helicases/metabolism , Quinazolinones/pharmacology , Antineoplastic Agents/pharmacology , Adenosine Triphosphate
11.
J Biomol Struct Dyn ; 42(6): 3145-3165, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37227775

ABSTRACT

A new and highly efficient visible-light-promoted catalyst free (VLCF) strategy for neat and clean synthesis of spiro indolo-quinazolinone-pyrrolo[3,4-a]pyrrolizine hybrids (6a-d) has been introduced. We have performed visible-light triggered 1,3-Dipolar cycloaddition reaction of maleimide (5a-d) with azomethine ylide generated in situ derived from tryptanthrin (3) and L-proline (4) to obtain desired products (6a-d) in good to excellent yield. Authentication and characterization of product was done using various spectroscopic techniques such as IR, 1H NMR, 13C NMR, Mass spectrometry and single crystal XRD analysis. To explain the reaction spontaneity, product stability, reactivity as well as possible mode of the interaction a quantum chemical investigation was performed and depicted through DFT studies. The synthesized compound 6a was also evaluated for anti-proliferative activity against a panel of five cancer cell lines (MCF-7, MDA-MB-231, HeLa, PC-3 and Ishikawa) and normal human embryonic kidney (HEK-293) cell line by using MTT assay. Compound 6a showed very good in vitro anti-proliferative activity (IC50  = 6.58-17.98 µM) against four cancer cell lines and no cytotoxicity against normal HEK-293. In order to evaluate the anticancer potential of compounds 6a-d, molecular docking was performed against wild type and mutant EGFR. The results suggest that all the compounds occupied the active site of both enzymes, with a strong binding energy (-10.2 to -11.5 kcal/mol). These results have been confirmed by molecular dynamics simulation by evaluating root mean square deviation (RMSD) and root mean square fluctuation (RMSF), along with principal component analysis (PCA).Communicated by Ramaswamy H. Sarma.


Subject(s)
Antineoplastic Agents , Humans , Molecular Docking Simulation , Antineoplastic Agents/chemistry , Quinazolinones/pharmacology , HEK293 Cells , Molecular Dynamics Simulation
12.
Bioorg Chem ; 142: 106920, 2024 01.
Article in English | MEDLINE | ID: mdl-37898082

ABSTRACT

In the current investigation, a new class of quinazolinone N-acetohydrazides 9a-v was designed as type II multi-kinase inhibitors. The target quinazolinones were tailored so that the quinazolinone moiety would occupy the front pocket of the binding sites of VEGFR-2, FGFR-1 and BRAF kinases, meanwhile, the phenyl group at position 2 would act as a spacer which was functionalized at position 4 with an N-acetohydrazide linker that could achieve the key interactions with the essential gate area amino acids. The hydrazide moiety was linked to diverse aryl derivatives to occupy the hydrophobic back pocket of the DFG-out conformation of target kinases. The synthesized quinazolinone derivatives 9a-v demonstrated moderate to potent VEGFR-2 inhibitory activity with IC50 spanning from 0.29 to 5.17 µM. Further evaluation of the most potent derivatives on FGFR-1, BRAFWT and BRAFV600E showed that the quinazolinone N-acetohydrazides 9d, 9e, 9f, 9l and 9m have a potent multi-kinase inhibitory activity. Concurrently, 9b, 9d, 9e, 9k, 9l, 9o, 9q demonstrated potent growth inhibitory activity on NCI cancer cell lines with GI50 reaching 0.72 µM. In addition, compound 9e arrested the cell cycle progression in MDA-MB-231 cell line at the G2/M phase and showed the ability to induce apoptosis.


Subject(s)
Antineoplastic Agents , Vascular Endothelial Growth Factor Receptor-2 , Molecular Structure , Structure-Activity Relationship , Quinazolinones/pharmacology , Proto-Oncogene Proteins B-raf , Protein Kinase Inhibitors , Cell Proliferation , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor , Molecular Docking Simulation
13.
J Agric Food Chem ; 71(49): 19277-19287, 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38038681

ABSTRACT

A total of 35 new quinazolinone derivatives bearing the 1,2,4-triazolo[3,4-b][1,3,4]thiadiazole scaffold and the 4-piperidinyl linker were designed, prepared, and assessed for their antibacterial and antifungal activities. Among these derivatives, the chemical structure of compound F5 was clearly verified via single-crystal X-ray diffraction analysis. The experimental results revealed that some of the compounds displayed good even excellent inhibitory effects toward the tested phytopathogenic bacteria. For instance, compound F33 was capable of strongly inhibiting Xanthomonas oryzae pv. oryzae (Xoo) in vitro with an EC50 (half-maximal effective concentration) value of 4.1 µg/mL, about 16-fold more effective than the commercialized bactericide bismerthiazol. Significantly, this compound also effectively suppressed the proliferation of Xoo in the potted rice plants, showing a good in vivo protection efficacy of 47.6% at 200 µg/mL. Subsequently, the antibacterial mechanisms of compound F33 were explored by means of different biophysical and biochemical methods. Last, some of the compounds were found to possess relatively good antifungal activities in vitro, like compound F19 against Phytophthora nicotianae (with an inhibition rate of 67.2% at 50 µg/mL). In a word, the current experimental results imply that the 4-piperidinyl-bridged quinazolinone-1,2,4-triazolo[3,4-b][1,3,4]thiadiazole derivatives possess potential as lead compounds for developing more efficient anti-Xoo bactericides.


Subject(s)
Oryza , Thiadiazoles , Xanthomonas , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , X-Rays , Microbial Sensitivity Tests , Anti-Bacterial Agents/chemistry , Quinazolinones/pharmacology , Thiadiazoles/pharmacology , Thiadiazoles/chemistry , Oryza/microbiology , Plant Diseases/microbiology
14.
J Med Chem ; 66(24): 16728-16761, 2023 12 28.
Article in English | MEDLINE | ID: mdl-38100045

ABSTRACT

E3 ubiquitin ligase, Constitutive Photomorphogenic 1 (COP1) regulates turnover of Adipose Triglyceride Lipase (ATGL), the rate-limiting lipolytic enzyme. Genetic perturbation in the COP1-ATGL axis disrupts lipid homeostasis, leading to liver steatosis. Using drug development strategies, we herein report quinazolinone and quinazolinedione based modulators for COP1-ATGL axis. Systematic SAR studies and subsequent optimization were performed by incorporating relevant functional groups at the N1, N3, C5, and C6 positions of both scaffolds. Compounds' efficacy was evaluated by multiple biological assays and ADME profiling. The lead compound 86 could increase ATGL protein expression, reduce ATGL ubiquitination and COP1 autoubiquitination, and diminish lipid accumulation in hepatocytes in the nanomolar range. Oral administration of 86 abrogated triglyceride accumulation and resolved fibrosis in preclinical Nonalcoholic Fatty Liver Disease (NAFLD) model. The study thus establishes quinazolinedione as a viable chemotype to therapeutically modulate the activity of COP1 and ATGL in relevant clinical contexts.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Quinazolinones/pharmacology , Quinazolinones/metabolism , Lipase , Hepatocytes/metabolism , Triglycerides/metabolism
15.
Carbohydr Res ; 534: 108967, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37844370

ABSTRACT

A series of novel tricyclic quinazolinone-iminosugars 5 and their derivatives 7 were obtained from the tosylated sugars by three steps. Firstly, the reaction of the isopropylidene protected sugar tosylate 1 and o-aminobenzylamine 2 generated the precursor tricyclic quinazolin-iminosuar 3, which was then oxidized by KMnO4 to produce the corresponding quinazolinone 4. Finally, removal of the isopropylidene group yielded the target tricyclic quinazolinone iminosugars 5. In addition, quinazolinone-iminosugars 4ac, 4bc and 4cc who contain bromine in the aromatic region underwent Suzuki reaction with phenylboronic acid, followed with the removal of the isopropylidene group to afford the derivatives 7. This strategy will help to construct such fused multicyclic quinazolinone-iminosugars efficiently. Some compounds show certain inhibition against α-glucosidase (saccharomyce cerevisiae).


Subject(s)
Imino Sugars , Quinazolinones , Quinazolinones/pharmacology , Alkenes , alpha-Glucosidases , Imino Sugars/pharmacology , Sugars
16.
Chem Biodivers ; 20(12): e202300800, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37708234

ABSTRACT

A library of 6-(((1-(substitutedphenyl)-1H-1,2,3-triazol-4-yl)methyl) amino)-3-methylquinazolin-4(3H)-one analogues synthesized from Isatin precursor through a series of nitration, reduction, hydrolysis, cyclization and click reaction. The structures of compounds were characterized by spectral data including IR, 1 H-NMR, 13 C NMR and Mass. The novel quinazolinone - 1,2,3-triazoles were screened for their cytotoxicity against the human breast adenocarcinoma cell lines MCF-7 by MTT assay. 4-Isopropyl and 2-bromo substituted analogues executed high activity against MCF-7 cell line with IC50 value of 10.16±0.07 µM and 11.23±0.20 µM compared to the Doxorubicin whose IC50 value is 10.81±0.03 µM. The activity of remaining compounds is good to moderate. Further, the molecular docking studies against the crystal structure of Epidermal Growth Factor Receptor delivered the best binding energies and the interactions such as H-bond and hydrophobic are inevitable. The predicted pharmacokinetic properties results showed that these compounds have more drug likeness properties.


Subject(s)
Antineoplastic Agents , Triazoles , Humans , MCF-7 Cells , Molecular Structure , Structure-Activity Relationship , Triazoles/chemistry , Quinazolinones/pharmacology , Quinazolinones/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Molecular Docking Simulation , Cell Proliferation
17.
Eur J Pharmacol ; 960: 176046, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-37708985

ABSTRACT

The in vitro A549 cells, and A549 xenografts in nude mouse, were two commonly used models for anti-cancer drug discovery. However, the biological and molecular characteristics of these two classic models, and also the dynamic transcriptome changes after dacomitinib exposure remains elusive. We performed single-cell RNA sequencing to define the transcriptome profile at single-cell resolution, and processed tumor samples for bulk RNA and protein analysis to validate the differently expressed genes. Transcriptome profiling revealed that the in vitro A549 cells are heterogeneous. The minimal subpopulation of the in vitro A549 cells, which were characterized by the signature of response to unfolded protein, became the overriding subpopulation of the xenografts. The EGFR non-activating A549 cells were resistant to dacomitinib in vitro, while A549 xenografts were comparatively sensitive as EGFR-activating HCC827 xenografts. Dacomitinib inhibited MAPK signaling pathway, and increased the immune response in the A549 xenografts. A phagocytosis checkpoint stanniocalcin-1 (STC1) was significantly inhibited in dacomitinib-treated xenografts. So here our study gives the first insight of the heterogeneity of the two classic models, and the translational potential of dacomitinib being used into a broader patient population rather than EGFR common activating mutation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Mice , Humans , ErbB Receptors/genetics , ErbB Receptors/metabolism , Single-Cell Gene Expression Analysis , Protein Kinase Inhibitors/pharmacology , Quinazolinones/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation
18.
Sci Rep ; 13(1): 14461, 2023 09 02.
Article in English | MEDLINE | ID: mdl-37660139

ABSTRACT

Quinazoline derivatives, as an important category of heterocyclic compounds, have received much attention for the design and development of new drugs due to their various pharmacological properties. Besides, there is a great deal of evidence showing pyrimidine analogs as anticancer agents. Thus, in the present study, for the design of new target compounds with cytotoxic activity, we focused on various quinazolinone and pyrimidine hybrids. A new series of quinazoline-pyrimidine hybrid derivatives (6a-6n) have been designed and synthesized as novel antiproliferative agents. All the synthesized compounds characterized based on their IR, NMR and Mass spectroscopic data. Antiproliferative activities of the new compounds were evaluated against three human cancer cell lines (MCF-7, A549, SW-480). The compounds were found to have appropriate potential with IC50 values ranging from 2.3 ± 5.91 to 176.5 ± 0.7 µM against the tested cell lines. Compound 6n exerted the highest antiproliferative activity with IC50 values of 5.9 ± 1.69 µM, 2.3 ± 5.91 µM and 5.65 ± 2.33 µM against A549, SW-480 and MCF-7 respectively. The results indicated that 6n could induce apoptosis in A549 cell line in a dose dependent manner and arrest in the S phase of cell cycle. Docking studies were also done to investigate the detailed binding pattern of the synthesized compounds against EGFR. Furthermore, molecular dynamic simulation and binding free energy calculation have been done to rescore initial docking pose of the synthesized compounds using ensemble-based MMGB/PBSA free energy method. According to the results, free energy calculation confirmed biological activity of compounds and also, Arg 817 and Lys 721 residues had the pivotal role in the high potency of 6n. Finally, the drug likeness and in silico ADME study were also predicted.


Subject(s)
Antineoplastic Agents , Quinazolines , Humans , Quinazolines/pharmacology , Pyrimidines/pharmacology , Antineoplastic Agents/pharmacology , Antimetabolites , Quinazolinones/pharmacology , Antihypertensive Agents
19.
Future Med Chem ; 15(13): 1133-1147, 2023 07.
Article in English | MEDLINE | ID: mdl-37529897

ABSTRACT

Background: Targeting CDK4/6 has advanced breast cancer treatment. Herein, new quinazolinones were synthesized with acetamide linkers as potential anti-breast cancer agents. Methods & results: In vitro cytotoxic evaluation on human breast cancer cell lines (MCF7 and MDA-MB-231) identified 1,3-benzodioxole (5d) to be of the highest potency. It showed good inhibitory activity on CDK4/6. Compound 5d arrested the cell cycle at the G1-phase, caused induction of early and late apoptosis in an Annexin V-FITC assay, led to an increase in the level of caspase-3 and upregulated Bax expression and downregulated Bcl-2 in MCF7 cells. Compound 5d showed good radiosensitizing activity when combined with a single dose of 8-Gy γ-radiation. Conclusion: This study introduces quinazolinone scaffolds as new CDK4/6 inhibitors for breast cancer.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Humans , Female , Cell Proliferation , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Quinazolinones/pharmacology , Cell Cycle , Apoptosis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cyclin-Dependent Kinase 4/metabolism
20.
Molecules ; 28(14)2023 Jul 11.
Article in English | MEDLINE | ID: mdl-37513216

ABSTRACT

The synthesis of 5,6,7,8-tetrahydro-[1,2,4]triazolo[5,1-b]quinazolin-9(4H)-one (THTQ), a potentially biologically active compound, was pursued, and its structure was determined through a sequence of spectral analysis, including 1H-NMR, 13C-NMR, IR, and HRMS. Four bacterial and four fungal strains were evaluated for their susceptibility to the antibacterial and antifungal properties of the THTQ compound using the well diffusion method. The impact of THTQ on the corrosion of mild steel in a 1 M HCl solution was evaluated using various methods such as weight loss, potentiodynamic polarization (PDP), electrochemical impedance spectroscopy (EIS), and scanning electron microscopy (SEM) analysis. The study revealed that the effectiveness of THTQ as an inhibitor increased with the concentration but decreased with temperature. The PDP analysis suggested that THTQ acted as a mixed-type inhibitor, whereas the EIS data showed that it created a protective layer on the steel surface. This protective layer occurs due to the adsorption behavior of THTQ following Langmuir's adsorption isotherm. The inhibition potential of THTQ is also predicted theoretically using DFT at B3LYP and Monte Carlo simulation.


Subject(s)
Antifungal Agents , Quinazolinones , Antifungal Agents/pharmacology , Quinazolinones/pharmacology , Temperature , Steel/chemistry , Anti-Bacterial Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...