Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.168
Filter
1.
Chem Biol Drug Des ; 104(1): e14599, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39039616

ABSTRACT

In this study, we synthesized 15 novel quinazoline-morpholinobenzylideneamino hybrid compounds from methyl anthranilate and we assessed their cytotoxicity via in vitro assays against A549 and BEAS-2B cell lines. Molecular docking studies were conducted to evaluate the protein-ligand interactions and inhibition mechanisms on nine different molecular targets, while molecular dynamics (MD) simulations were carried out to assess the stability of the best docked ligand-protein complexes. Additionally, ADME prediction was carried out to determine physicochemical parameters and drug likeness. According to the cytotoxicity assays, compound 1 (IC50 = 2.83 µM) was found to be the most active inhibitor against A549 cells. While the selectivity index (SI) of compound 1 is 29, the SI of the reference drugs paclitaxel and sorafenib, used in this study, are 2.40 and 4.92, respectively. Among the hybrid compounds, 1 has the best docking scores against VEGFR1 (-11.744 kcal/mol), VEGFR2 (-12.407 kcal/mol) and EGFR (-10.359 kcal/mol). During MD simulations, compound 1 consistently exhibited strong hydrogen bond interactions with the active sites of VEGFR1 and 2, and these interactions were maintained for more than 90% of the simulation time. Additionally, the RMSD and RMSF values of the ligand-protein complexes exhibited high stability at their minimum levels around 1-2 Å. In conclusion, these findings suggest that compound 1 may be a potent and selective inhibitor candidate for lung cancer treatment and inhibition of VEGFR2, especially.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Molecular Docking Simulation , Molecular Dynamics Simulation , Morpholines , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Morpholines/chemistry , Morpholines/pharmacology , Cell Line, Tumor , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , A549 Cells , Quinazolinones/chemistry , Quinazolinones/pharmacology , Quinazolinones/metabolism , Quinazolinones/chemical synthesis , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Quinazolines/chemistry , Quinazolines/pharmacology , Quinazolines/chemical synthesis , Quinazolines/metabolism , Binding Sites , Drug Screening Assays, Antitumor , Hydrogen Bonding
2.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000168

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an extremely complex neurodegenerative disease involving different cell types, but motoneuronal loss represents its main pathological feature. Moreover, compensatory plastic changes taking place in parallel to neurodegeneration are likely to affect the timing of ALS onset and progression and, interestingly, they might represent a promising target for disease-modifying treatments. Therefore, a simplified animal model mimicking motoneuronal loss without the other pathological aspects of ALS has been established by means of intramuscular injection of cholera toxin-B saporin (CTB-Sap), which is a targeted neurotoxin able to kill motoneurons by retrograde suicide transport. Previous studies employing the mouse CTB-Sap model have proven that spontaneous motor recovery is possible after a subtotal removal of a spinal motoneuronal pool. Although these kinds of plastic changes are not enough to counteract the functional effects of the progressive motoneuron degeneration, it would nevertheless represent a promising target for treatments aiming to postpone ALS onset and/or delay disease progression. Herein, the mouse CTB-Sap model has been used to test the efficacy of mitochondrial division inhibitor 1 (Mdivi-1) as a tool to counteract the CTB-Sap toxicity and/or to promote neuroplasticity. The homeostasis of mitochondrial fission/fusion dynamics is indeed important for cell integrity, and it could be affected during neurodegeneration. Lesioned mice were treated with Mdivi-1 and then examined by a series of behavioral test and histological analyses. The results have shown that the drug may be capable of reducing functional deficits after the lesion and promoting synaptic plasticity and neuroprotection, thus representing a putative translational approach for motoneuron disorders.


Subject(s)
Amyotrophic Lateral Sclerosis , Disease Models, Animal , Mitochondrial Dynamics , Motor Neurons , Animals , Motor Neurons/drug effects , Motor Neurons/metabolism , Motor Neurons/pathology , Mitochondrial Dynamics/drug effects , Mice , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Cholera Toxin/metabolism , Saporins , Quinazolinones/pharmacology , Neuronal Plasticity/drug effects , Male , Mitochondria/drug effects , Mitochondria/metabolism
3.
Mol Biol Rep ; 51(1): 870, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080104

ABSTRACT

BACKGROUND: Pediatric postoperative cognitive dysfunction (POCD) is a prevalent complication following anesthesia and surgery. Hypoxia and propofol are the primary risk factors contributing to pediatric POCD. Our previous in vivo animal research has demonstrated that cognitive dysfunction in immature Sprague-Dawley (SD) rats, induced by hypoxia combined with propofol (HCWP), is closely associated with hippocampal neuron ferroptosis. METHODS AND RESULTS: In vivo transcriptome sequencing and KEGG functional analysis revealed significant enrichment of the mitophagy pathway. To further elucidate the relationship between mitophagy and ferroptosis, HT22 cells were selected to construct an in vitro HCWP model. Our findings indicate that HCWP activates excessive mitophagy in HT22 cells, leading to decreased mitochondrial membrane potential (ΔΨm), reactive oxygen species (ROS) burst, mitochondrial fragmentation, and the induction of ferroptosis. To explore this causal relationship further, we employed Mdivi-1, a mitophagy inhibitor. Notably, low-dose Mdivi-1 (10 µM) effectively suppressed excessive mitophagy in HT22 cells, improved mitochondrial function and morphology, and mitigated markers associated with ferroptosis. The mechanism by which Mdivi-1 alleviates HCWP-induced ferroptosis in HT22 cells is likely due to its inhibition of excessive mitophagy, thereby promoting mitochondrial homeostasis. CONCLUSIONS: Our study suggests that mitophagy may be an upstream event in HCWP-induced ferroptosis in HT22 cells. Consequently, targeted regulation of mitophagy by Mdivi-1 may represent a promising approach to prevent cognitive dysfunction following HCWP exposure.


Subject(s)
Ferroptosis , Membrane Potential, Mitochondrial , Mitophagy , Propofol , Quinazolinones , Reactive Oxygen Species , Mitophagy/drug effects , Propofol/pharmacology , Ferroptosis/drug effects , Animals , Quinazolinones/pharmacology , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Rats , Mitochondria/drug effects , Mitochondria/metabolism , Cell Line , Hippocampus/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Mice , Hypoxia/metabolism , Hypoxia/complications , Neurons/metabolism , Neurons/drug effects , Rats, Sprague-Dawley , Cell Hypoxia/drug effects , Postoperative Cognitive Complications/metabolism
4.
Molecules ; 29(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38999000

ABSTRACT

In this study, a new series of cis and trans 5-substituted-3-(dibenzyloxyphosphoryl)isoxazolidines 16a-g were synthesized by the 1,3-dipolar cycloaddition reaction of N-benzyl-C-(dibenzyloxyphosphoryl)nitrone and selected N1-allyl-N3-benzylquinazoline-2,4-diones. All the obtained trans-isoxazolidines 16a-g and the samples enriched in respective cis-isomers were evaluated for anticancer activity against three tumor cell lines. All the tested compounds exhibited high activity against the prostate cancer cell line (PC-3). Isoxazolidines trans-16a and trans-16b and diastereoisomeric mixtures of isoxazolidines enriched in cis-isomer using HPLC, namely cis-16a/trans-16a (97:3) and cis-16b/trans-16b (90:10), showed the highest antiproliferative properties towards the PC-3 cell line (IC50 = 9.84 ± 3.69-12.67 ± 3.45 µM). For the most active compounds, induction apoptosis tests and an evaluation of toxicity were conducted. Isoxazolidine trans-16b showed the highest induction of apoptosis. Moreover, the most active compounds turned out safe in vitro as none affected the cell viability in the HEK293, HepG2, and HSF cellular models at all the tested concentrations. The results indicated isoxazolidine trans-16b as a promising new lead structure in the search for effective anticancer drugs.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Structure-Activity Relationship , Isoxazoles/chemistry , Isoxazoles/pharmacology , PC-3 Cells , Drug Screening Assays, Antitumor , Quinazolinones/chemistry , Quinazolinones/pharmacology , Quinazolinones/chemical synthesis , Molecular Structure , Cell Survival/drug effects , Apoptosis/drug effects
5.
Nat Commun ; 15(1): 5244, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898000

ABSTRACT

Methaqualone, a quinazolinone marketed commercially as Quaalude, is a central nervous system depressant that was used clinically as a sedative-hypnotic, then became a notorious recreational drug in the 1960s-80s. Due to its high abuse potential, medical use of methaqualone was eventually prohibited, yet it persists as a globally abused substance. Methaqualone principally targets GABAA receptors, which are the major inhibitory neurotransmitter-gated ion channels in the brain. The restricted status and limited accessibility of methaqualone have contributed to its pharmacology being understudied. Here, we use cryo-EM to localize the GABAA receptor binding sites of methaqualone and its more potent derivative, PPTQ, to the same intersubunit transmembrane sites targeted by the general anesthetics propofol and etomidate. Both methaqualone and PPTQ insert more deeply into subunit interfaces than the previously-characterized modulators. Binding of quinazolinones to this site results in widening of the extracellular half of the ion-conducting pore, following a trend among positive allosteric modulators in destabilizing the hydrophobic activation gate in the pore as a mechanism for receptor potentiation. These insights shed light on the underexplored pharmacology of quinazolinones and further elucidate the molecular mechanisms of allosteric GABAA receptor modulation through transmembrane binding sites.


Subject(s)
Cryoelectron Microscopy , Receptors, GABA-A , Receptors, GABA-A/metabolism , Receptors, GABA-A/chemistry , Binding Sites , Humans , Animals , Etomidate/pharmacology , Etomidate/analogs & derivatives , Propofol/pharmacology , Propofol/chemistry , Quinazolinones/pharmacology , Quinazolinones/chemistry , Allosteric Regulation/drug effects , HEK293 Cells , Hypnotics and Sedatives/pharmacology
6.
Bioorg Med Chem Lett ; 109: 129855, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38908766

ABSTRACT

The role of G-quadruplex (G4) in cellular processes can be investigated by the covalent modification of G4-DNA using alkylating reagents. Controllable alkylating reagents activated by external stimuli can react elegantly and selectively. Herein, we report a chemical activation system that can significantly boost the reaction rate of methylamine-protected vinyl-quinazolinone (VQ) derivative for the alkylation of G4-DNA. The two screened activators can transform low-reactive VQ-NHR' to highly reactive intermediates following the Michael addition mechanism. This approach expands the toolbox of activable G4 alkylating reagents.


Subject(s)
G-Quadruplexes , Methylamines , Quinazolinones , Alkylation , G-Quadruplexes/drug effects , Methylamines/chemistry , Methylamines/pharmacology , Methylamines/chemical synthesis , Quinazolinones/chemistry , Quinazolinones/pharmacology , Quinazolinones/chemical synthesis , Humans , Molecular Structure , DNA/chemistry , Vinyl Compounds/chemistry , Vinyl Compounds/pharmacology
7.
Bioorg Chem ; 150: 107574, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38936049

ABSTRACT

Mitochondrial dynamics have pillar roles in several diseases including cancer. Cancer cell survival is monitored by mitochondria which impacts several cellular functions such as cell metabolism, calcium signaling, and ROS production. The equilibrium of death and survival rate of mitochondria is important for healthy cellular processes. Whereas inhibition of mitochondrial metabolism and dynamics can have crucial regulatory decisions between cell survival and death. The steady rate of physiological flux of both mitochondrial fission and fusion is strongly related to the preservation of cellular bioenergetics. Dysregulation of mitochondrial dynamics including fission and fusion is a critical machinery in cells accompanied by crosstalk in cancer progression and resistance. Many cancer cells express high levels of Drp-1 to induce cancer cell invasion, metastasis and chemoresistance including breast cancer, liver cancer, pancreatic cancer, and colon cancer. Targeting Drp-1 by inhibitors such as Midivi-1 helps to enhance the responsiveness of cancer cells towards chemotherapy. The review showed Drp-1 linked processes such as mitochondrial dynamics and relationship with cancer, invasion, and chemoresistance along with computational assessing of all publicly available Drp-1 inhibitors. Drp1-IN-1, Dynole 34-2, trimethyloctadecylammonium bromide, and Schaftoside showed potential inhibitory effects on Drp-1 as compared to standard Mdivi- 1. This emerging approach may have extensive strength in the context of cancer development and chemoresistance and further work is needed to aid in more effective cancer management.


Subject(s)
Antineoplastic Agents , Drug Resistance, Neoplasm , Dynamins , Neoplasms , Humans , Dynamins/antagonists & inhibitors , Dynamins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Mitochondrial Dynamics/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Animals , Quinazolinones/pharmacology , Quinazolinones/chemistry , Quinazolinones/chemical synthesis
8.
Sci Rep ; 14(1): 14178, 2024 06 19.
Article in English | MEDLINE | ID: mdl-38898058

ABSTRACT

Increasing evidence supports the hypothesis that cancer progression is under mitochondrial control. Mitochondrial fission plays a pivotal role in the maintenance of cancer cell homeostasis. The inhibition of DRP1, the main regulator of mitochondrial fission, with the mitochondrial division inhibitor (mdivi-1) had been associated with cancer cell sensitivity to chemotherapeutics and decrease proliferation. Here, using breast cancer cells we find that mdivi-1 induces the detachment of the cells, leading to a bulk of floating cells that conserved their viability. Despite a decrease in their proliferative and clonogenic capabilities, these floating cells maintain the capacity to re-adhere upon re-seeding and retain their migratory and invasive potential. Interestingly, the cell detachment induced by mdivi-1 is independent of DRP1 but relies on inhibition of mitochondrial complex I. Furthermore, mdivi-1 induces cell detachment rely on glucose and the pentose phosphate pathway. Our data evidence a novel DRP1-independent effect of mdivi-1 in the attachment of cancer cells. The generation of floating viable cells restricts the use of mdivi-1 as a therapeutic agent and demonstrates that mdivi-1 effect on cancer cells are more complex than anticipated.


Subject(s)
Breast Neoplasms , Dynamins , Extracellular Matrix , Mitochondrial Dynamics , Quinazolinones , Humans , Dynamins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Female , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Cell Line, Tumor , Quinazolinones/pharmacology , Mitochondrial Dynamics/drug effects , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Cell Proliferation/drug effects , Mitochondria/metabolism , Mitochondria/drug effects
9.
Int J Pharm ; 661: 124384, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38917957

ABSTRACT

Postoperative distant metastasis and high recurrence rate causes a dilemma in treating triple-negative breast cancer (TNBC) owing to its unforeseeable invasion into various organs or tissues. The wealth of nutrition provided by vascular may facilitate the proliferation and angiogenesis of cancer cells, which further enhance the rates of postoperative metastasis and recurrence. Chemotherapy, as a systemic postoperative adjuvant therapy, is generally applied to diminish recurrence and metastasis of TNBC. Herein, an halofuginone-silver nano thermosensitive hydrogel (HTPM&AgNPs-gel) was prepared via a physical swelling method. The in vitro anticancer efficacy of HTPM&AgNPs-gel was analyzed by investigating cell proliferation, migration, invasion, and angiogenesis capacity. Furthermore, the in vivo anti-cancer activity of HTPM&AgNPs-gel was further appraised through the tumor suppression, anti-metastatic, anti-angiogenic, and anti-inflammatory ability. The optimized HTPM&AgNPs-gel, a thermosensitive hydrogel, showed excellent properties, including syringeability, swelling behavior, and a sustained release effect without hemolysis. In addition, HTPM&AgNPs-gel was confirmed to effectively inhibit the proliferation, migration, invasion, and angiogenesis of MDA-MB-231 cells. An evaluation of the in vivo anti-tumor efficacy demonstrated that HTPM&AgNPs-gel showed a stronger tumor inhibition rate (68.17%) than did HTPM-gel or AgNPs-gel used alone and exhibited outstanding biocompatibility. Notably, HTPM&AgNPs-gel also inhibited lung metastasis induced by residual tumor tissue after surgery and further blocked angiogenesis-related inflammatory responses. Taken together, the suppression of inflammation by interdicting the blood vessels adjoining the tumor and inhibiting angiogenesis is a potential strategy to attenuate the recurrence and metastasis of TNBC. HTPM&AgNPs-gel is a promising anticancer agent for TNBC as a local postoperative treatment.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Hydrogels , Piperidines , Quinazolinones , Silver , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/drug therapy , Hydrogels/administration & dosage , Hydrogels/chemistry , Animals , Female , Silver/chemistry , Silver/administration & dosage , Humans , Cell Line, Tumor , Piperidines/pharmacology , Piperidines/administration & dosage , Piperidines/chemistry , Cell Proliferation/drug effects , Quinazolinones/chemistry , Quinazolinones/administration & dosage , Quinazolinones/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice, Inbred BALB C , Mice , Cell Movement/drug effects , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Neovascularization, Pathologic/drug therapy , Xenograft Model Antitumor Assays , Mice, Nude
10.
Acta Chim Slov ; 71(2): 334-352, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38919105

ABSTRACT

The Re(I) organometallic compounds [(Re(CO)3L1-6 )Cl], where Ligand(L) = Tryptanthrin derivatives were prepared and characterized by various spectroscopic techniques. To assess the binding capacities and binding manner, tests of Calf thymus DNA under the impact of organometallic complexes were conducted using absorption titration and viscosity measuring techniques. Data from the research mentioned above point to an intercalation type of binding, which was verified by the docking study. Swiss ADME tools carried out an ADME study. The work focuses on computing the molecular orbital energies for the synthesized compounds using the density functional theory (DFT). The compounds were tested against the MCF-7 cell line to determine their anticancer effects. It was observed that their IC50 values were equivalent to those of the standard medication, indicating that they had a similar antiproliferative impact.


Subject(s)
Antineoplastic Agents , Rhenium , Rhenium/chemistry , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , MCF-7 Cells , Cell Proliferation/drug effects , Organometallic Compounds/pharmacology , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Molecular Docking Simulation , DNA/metabolism , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Drug Screening Assays, Antitumor , Density Functional Theory , Cattle , Quinazolinones/chemical synthesis , Quinazolinones/pharmacology , Quinazolinones/chemistry , Molecular Structure , Animals , Indoles/pharmacology , Indoles/chemistry , Indoles/chemical synthesis
11.
Eur J Pharmacol ; 977: 176736, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38878877

ABSTRACT

Mitochondrial dysfunction and the activation of multiple programmed cell death (PCD) have been shown to aggravate the severity and mortality associated with the progression of myocardial infarction (MI). Although pharmacological modulation of mitochondrial dynamics, including treatment with the fusion promoter (M1) and the fission inhibitor (Mdivi-1), exerted cardioprotection against several cardiac complications, their roles in the post-MI model have never been investigated. Using a MI rat model instigated by permanent left-anterior descending (LAD) coronary artery occlusion, post-MI rats were randomly assigned to receive one of 4 treatments (n = 10/group): vehicle (DMSO 3%V/V), enalapril (10 mg/kg), Mdivi-1 (1.2 mg/kg) and M1 (2 mg/kg), while a control group of sham operated rats underwent surgery without LAD occlusion (n = 10). After 32-day treatment, cardiac and mitochondrial function, and histopathological morphology were investigated and molecular analysis was performed. Treatment with enalapril, Mdivi-1, and M1 significantly mitigated cardiac pathological remodeling, reduced myocardial injury, and improved left ventricular (LV) function in post-MI rats. Importantly, all interventions also attenuated mitochondrial dynamic imbalance and mitigated activation of apoptosis, necroptosis, and pyroptosis after MI. This investigation demonstrated for the first time that chronic mitochondrial dynamic-targeted therapy mitigated mitochondrial dysfunction and activation of PCD, leading to improved LV function in post-MI rats.


Subject(s)
Apoptosis , Enalapril , Mitochondrial Dynamics , Myocardial Infarction , Ventricular Dysfunction, Left , Animals , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Mitochondrial Dynamics/drug effects , Male , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/physiopathology , Rats , Enalapril/pharmacology , Enalapril/therapeutic use , Apoptosis/drug effects , Rats, Sprague-Dawley , Quinazolinones/pharmacology , Quinazolinones/therapeutic use , Disease Models, Animal , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology
12.
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
13.
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
14.
Bioorg Med Chem Lett ; 108: 129800, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38763480

ABSTRACT

In a quest to discover new antimalarial and antitubercular drugs, we have designed and synthesized a series of novel triazole-quinazolinone hybrids. The in vitro screening of the triazole-quinazolinone hybrid entities against the plasmodium species P. falciparum offered potent antimalarial molecules 6c, 6d, 6f, 6g, 6j & 6k owing comparable activity to the reference drugs. Furthermore, the target compounds were evaluated in vitro against Mycobacterium tuberculosis (MTB) H37Rv strain. Among the screened compounds, 6c, 6d and 6l were found to be the most active molecules with a MIC values of 19.57-40.68 µM. The cytotoxicity of the most active compounds was studied against RAW 264.7 cell line by MTT assay and no toxicity was observed. The computational study including drug likeness and ADMET profiling, DFT, and molecular docking study was done to explore the features of target molecules. The compounds 6a, 6g, and 6k exhibited highest binding affinity of -10.3 kcal/mol with docked molecular targets from M. tuberculosis. Molecular docking study indicates that all the molecules are binding to the falcipain 2 protease (PDB: 6SSZ) of the P. falciparum. Our findings indicated that these new triazole-quinazolinone hybrids may be considered hit molecules for further optimization studies.


Subject(s)
Antimalarials , Antitubercular Agents , Drug Design , Microbial Sensitivity Tests , Molecular Docking Simulation , Mycobacterium tuberculosis , Plasmodium falciparum , Quinazolinones , Triazoles , Antitubercular Agents/pharmacology , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Antimalarials/pharmacology , Antimalarials/chemical synthesis , Antimalarials/chemistry , Triazoles/chemistry , Triazoles/pharmacology , Triazoles/chemical synthesis , Mycobacterium tuberculosis/drug effects , Plasmodium falciparum/drug effects , Quinazolinones/chemistry , Quinazolinones/pharmacology , Quinazolinones/chemical synthesis , Mice , Structure-Activity Relationship , Animals , Molecular Structure , Dose-Response Relationship, Drug , RAW 264.7 Cells
15.
Cancer Lett ; 597: 216996, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38815797

ABSTRACT

Relapsed or refractory diffuse large B cell lymphoma (DLBCL) and mantle cell lymphoma (MCL) patients still faced with poor survival, representing an unmet clinical need. In-depth research into the disease's pathogenesis and the development of targeted treatment strategies are urgently needed. Here, we conducted a comprehensive bioinformatic analysis of gene mutation and expression using data from our center and public databases. Cell cycle-related genes especially for CDKN2A/B-CDK4/6/CCND1 machinery altered frequently in DLBCL and MCL. Clinically, high CDK4 and CDK6 expression were correlated with poor prognosis of DLBCL and MCL patients. Furthermore, we also validated the pharmacological efficacy of CDK4/6 inhibitor palbociclib and its synergy effect with PI3K inhibitor idelalisib utilizing in vitro cell lines and in vivo cell-derived xenograft (CDX) and patient-derived xenograft (PDX) mouse models. Our results provided sufficient pre-clinical evidence to support the potential combination of palbociclib and idelalisib for DLBCL and MCL patients.


Subject(s)
Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Drug Synergism , Piperazines , Purines , Pyridines , Quinazolinones , Xenograft Model Antitumor Assays , Humans , Purines/pharmacology , Animals , Piperazines/pharmacology , Pyridines/pharmacology , Quinazolinones/pharmacology , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Mice , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/pathology , Lymphoma, Mantle-Cell/genetics , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Proliferation/drug effects , Female , Protein Kinase Inhibitors/pharmacology
16.
Phytomedicine ; 129: 155670, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38704915

ABSTRACT

BACKGROUND: Anaplastic thyroid carcinoma (ATC) is recognized as the most aggressive and malignant form of thyroid cancer, underscoring the critical need for effective therapeutic strategies to curb its progression and improve patient prognosis. Halofuginone (HF), a derivative of febrifugine, has displayed antitumor properties across various cancer types. However, there is a paucity of published research focused on the potential of HF to enhance the clinical efficacy of treating ATC. OBJECTIVE: In this study, we thoroughly investigated the antitumor effects and mechanisms of HF in ATC, aiming to discover lead compounds for treating ATC and reveal novel therapeutic targets for ATC tumors. METHODS: A series of assays, including CCK8, colony formation, tumor xenograft models, and ATC tumor organoid experiments, were conducted to evaluate the anticancer properties of HF both in vitro and in vivo. Techniques such as drug affinity responsive target stability (DARTS), western blot, immunofluorescence, and immunohistochemistry were employed to pinpoint HF target proteins within ATC. Furthermore, we harnessed the GEPIA and GEO databases and performed immunohistochemistry to validate the therapeutic potential of the glutamyl-prolyl-tRNA-synthetase (EPRS)- activating transcription factor 4 (ATF4)- type I collagen (COLI) pathway axis in the context of ATC. The study also incorporated RNA sequencing analysis, confocal imaging, and flow cytometry to delve into the molecular mechanisms of HF in ATC. RESULTS: HF exhibited a substantial inhibitory impact on cell proliferation in vitro and on tumor growth in vivo. The DARTS results highlighted HF's influence on EPRS within ATC cells, triggering an amino acid starvation response (AASR) by suppressing EPRS expression, consequently leading to a reduction in COLI expression in ATC cells. The introduction of proline mitigated the effect of HF on ATF4 and COLI expression, indicating that the EPRS-ATF4-COLI pathway axis was a focal target of HF in ATC. Analysis of the expression levels of the EPRS, ATF4, and COLI proteins in thyroid tumors, along with an examination of the relationship between COLI expression and thyroid tumor stage, revealed that HF significantly inhibited the growth of ATC tumor organoids, demonstrating the therapeutic potential of targeting the EPRS-ATF4-COLI pathway axis in ATC. RNA sequencing analysis revealed significant differences in the pathways associated with metastasis and apoptosis between control and HF-treated cells. Transwell assays and flow cytometry experiments provided evidence of the capacity of HF to impede cell migration and induce apoptosis in ATC cells. Furthermore, HF hindered cell metastasis by suppressing the epithelial-mesenchymal transition (EMT) pathway, acting through the inhibition of FAK-AKT-NF-κB/Wnt-ß-catenin signaling and restraining angiogenesis via the VEGF pathway. HF also promoted apoptosis through the mitochondrial apoptotic pathway. CONCLUSION: This study provided inaugural evidence suggesting that HF could emerge as a promising therapeutic agent for the treatment of ATC. The EPRS-ATF4-COLI pathway axis stood out as a prospective biomarker and therapeutic target for ATC.


Subject(s)
Activating Transcription Factor 4 , Piperidines , Quinazolinones , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms , Thyroid Carcinoma, Anaplastic/drug therapy , Activating Transcription Factor 4/metabolism , Humans , Animals , Cell Line, Tumor , Thyroid Neoplasms/drug therapy , Piperidines/pharmacology , Quinazolinones/pharmacology , Mice , Mice, Nude , Xenograft Model Antitumor Assays , Signal Transduction/drug effects , Cell Proliferation/drug effects , Mice, Inbred BALB C
17.
Protein Sci ; 33(6): e5028, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38757396

ABSTRACT

Prolyl-tRNA synthetase (ProRS), belonging to the family of aminoacyl-tRNA synthetases responsible for pairing specific amino acids with their respective tRNAs, is categorized into two distinct types: the eukaryote/archaeon-like type (E-type) and the prokaryote-like type (P-type). Notably, these types are specific to their corresponding cognate tRNAs. In an intriguing paradox, Thermus thermophilus ProRS (TtProRS) aligns with the E-type ProRS but selectively charges the P-type tRNAPro, featuring the bacterium-specific acceptor-stem elements G72 and A73. This investigation reveals TtProRS's notable resilience to the inhibitor halofuginone, a synthetic derivative of febrifugine emulating Pro-A76, resembling the characteristics of the P-type ProRS. Furthermore, akin to the P-type ProRS, TtProRS identifies its cognate tRNA through recognition of the acceptor-stem elements G72/A73, along with the anticodon elements G35/G36. However, in contrast to the P-type ProRS, which relies on a strictly conserved R residue within the bacterium-like motif 2 loop for recognizing G72/A73, TtProRS achieves this through a non-conserved sequence, RTR, within the otherwise non-interacting eukaryote-like motif 2 loop. This investigation sheds light on the adaptive capacity of a typically conserved housekeeping enzyme to accommodate a novel substrate.


Subject(s)
Amino Acyl-tRNA Synthetases , RNA, Transfer, Pro , Thermus thermophilus , Thermus thermophilus/chemistry , Thermus thermophilus/enzymology , Thermus thermophilus/genetics , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , RNA, Transfer, Pro/chemistry , RNA, Transfer, Pro/genetics , RNA, Transfer, Pro/metabolism , Amino Acid Motifs , Amino Acid Sequence , Inverted Repeat Sequences , Evolution, Molecular , Yeasts/enzymology , Protein Synthesis Inhibitors/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Piperidines/pharmacology , Quinazolinones/pharmacology
18.
Arch Pharm (Weinheim) ; 357(8): e2400057, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38775630

ABSTRACT

Quinazoline and quinazolinone derivatives piqued medicinal chemistry interest in developing novel drug candidates owing to their pharmacological potential. They are important chemicals for the synthesis of a variety of physiologically significant and pharmacologically useful molecules. Quinazoline and quinazolinone derivatives have anticancer, anti-inflammatory, antidiabetic, anticonvulsant, antiviral, and antimicrobial potential. The increased understanding of quinazoline and quinazolinone derivatives in biological activities provides opportunities for new medicinal products. The present review focuses on novel advances in the synthesis of these important scaffolds and other medicinal aspects involving drug design, structure-activity relationship, and action mechanisms of quinazoline and quinazolinone derivatives to help in the development of new quinazoline and quinazolinone derivatives.


Subject(s)
Molecular Docking Simulation , Quinazolines , Quinazolinones , Quinazolines/pharmacology , Quinazolines/chemical synthesis , Quinazolines/chemistry , Quinazolinones/pharmacology , Quinazolinones/chemical synthesis , Quinazolinones/chemistry , Humans , Structure-Activity Relationship , Drug Design , Animals , Molecular Structure
19.
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 , Inflammasomes , Mitochondrial Dynamics , NLR Family, Pyrin Domain-Containing 3 Protein , Quinazolinones , Animals , Humans , Male , Mice , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/drug therapy , Carrier Proteins/metabolism , Carrier Proteins/genetics , Cell Line , Contrast Media/adverse effects , Dynamins/metabolism , Inflammasomes/metabolism , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Quinazolinones/pharmacology , Quinazolinones/therapeutic use , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Thioredoxins/metabolism , Thioredoxins/genetics
20.
J Cell Biochem ; 125(6): e30558, 2024 06.
Article in English | MEDLINE | ID: mdl-38577900

ABSTRACT

The complex impacts of prolonged morphine exposure continue to be a significant focus in the expanding area of addiction studies. This research investigates the effectiveness of a combined treatment using Cabergoline and Mdivi-1 to counteract the neuroadaptive changes caused by in vitro morphine treatment. The impact of Methadone, Cabergoline, and a combination of Cabergoline and Mdivi-1 on the cellular and molecular responses associated with Morphine-induced changes was studied in human Neuroblastoma (SK-N-MC) and Glioblastoma (U87-MG) cell lines that were exposed to prolong Morphine treatment. Cabergoline and Mdivi-1 combined treatment effectively influenced the molecular alterations associated with neuroadaptation in chronic morphine-exposed neural cells. This combination therapy normalized autophagy and reduced oxidative stress by enhancing total-antioxidant capacity, mitigating apoptosis, restoring BDNF expression, and balancing apoptotic elements. Our research outlines morphine's dual role in modulating mitochondrial dynamics via the dysregulation of the autophagy-apoptosis axis. This emphasizes the significant involvement of DRP1 activity in neurological adaptation processes, as well as disturbances in the dopaminergic pathway during in vitro chronic exposure to morphine in neural cells. This study proposes a novel approach by recommending the potential effectiveness of combining Cabergoline and Mdivi-1 to modulate the neuroadaptations caused by morphine. Additionally, we identified BDNF and PCNA in neural cells as potential neuroprotective markers for assessing the effectiveness of drugs against opioid toxicity, emphasizing the need for further validation. The study uncovers diverse effects observed in pretreated morphine glioblastoma cells under treatment with Cabergoline and methadone. This highlights the potential for new treatments in the DRD2 pathway and underscores the importance of investigating the interplay between autophagy and apoptosis to advance research in managing cancer-related pain. The study necessitates an in-depth investigation into the relationship between autophagy and apoptosis, with a specific emphasis on protein interactions and the dynamics of cell signaling.


Subject(s)
Apoptosis , Autophagy , Cabergoline , Morphine , Quinazolinones , Humans , Autophagy/drug effects , Apoptosis/drug effects , Morphine/pharmacology , Cabergoline/pharmacology , Cell Line, Tumor , Quinazolinones/pharmacology , Oxidative Stress/drug effects , Mitochondrial Dynamics/drug effects , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Brain-Derived Neurotrophic Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL