Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.335
Filter
1.
Eur J Med Chem ; 278: 116794, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39226707

ABSTRACT

Alzheimer's disease (AD, also known as dementia) has become a serious global health problem along with population aging, and neuroinflammation is the underlying cause of cognitive impairment in the brain. Nowadays, the development of multitarget anti-AD drugs is considered to be one effective approach. Imidazolylacetophenone oxime ethers or esters (IOEs) were multifunctional agents with neuroinflammation inhibition, metal chelation, antioxidant and neuroprotection properties against Alzheimer's disease. In this study, IOEs derivatives 1-8 were obtained by structural modifications of the oxime and imidazole groups, and the SARs showed that (Z)-oxime ether (derivative 2) had stronger anti-neuroinflammatory and neuroprotective ability than (E)-congener. Then, IOEs derivatives 9-30 were synthesized based on target-directed ligands and activity-based groups hybridization strategy. In vitro anti-AD activity screening revealed that some derivatives exhibited potentially multifunctional effects, among which derivative 28 exhibited the strongest inhibitory activity on NO production with EC50 value of 0.49 µM, and had neuroprotective effects on 6-OHDA-induced cell damage and RSL3-induced ferroptosis. The anti-neuroinflammatory mechanism showed that 28 could inhibit the release of pro-inflammatory factors PGE2 and TNF-α, down-regulate the expression of iNOS and COX-2 proteins, and promote the polarization of BV-2 cells from pro-inflammatory M1 phenotype to anti-inflammatory M2 phenotype. In addition, 28 can dose-dependently inhibit acetylcholinesterase (AChE) and Aß42 aggregation. Moreover, the selected nuclide [18F]-labeled 28 was synthesized to explore its biodistribution by micro-PET/CT, of which 28 can penetrate the blood-brain barrier (BBB). These results shed light on the potential of 28 as a new multifunctional candidate for AD treatment.


Subject(s)
Acetophenones , Alzheimer Disease , Drug Design , Imidazoles , Neuroprotective Agents , Oximes , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Oximes/chemistry , Oximes/pharmacology , Oximes/chemical synthesis , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/chemical synthesis , Animals , Structure-Activity Relationship , Imidazoles/pharmacology , Imidazoles/chemistry , Imidazoles/chemical synthesis , Acetophenones/chemistry , Acetophenones/pharmacology , Acetophenones/chemical synthesis , Molecular Structure , Humans , Brain/metabolism , Brain/drug effects , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Acetylcholinesterase/metabolism , Dose-Response Relationship, Drug , Rats , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry
2.
Bull Exp Biol Med ; 177(3): 344-348, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39126546

ABSTRACT

The neuroprotective activity of tryptanthrin and its oxime was compared in male Wistar rats with a model of intraluminal occlusion of the middle cerebral artery. Neurobehavioral tests were performed 4, 24, and 48 h after focal cerebral infarction (FCI) using a modified neurological severity score (mNSS); additionally, the horizontal stability test, the plantar sensitivity test of the fore and hind limbs, holding on the tilted cage top test, and negative geotaxis test were performed. The size of FCI and the severity of brain tissue swelling were examined on day 2 after occlusion. Tryptanthrin and its oxime were administered at a dose of 10 mg/kg intraperitoneally during FCI, then daily for 2 days. In the control group, the mean score of neurological deficit remained at a high level for 2 days. FCI size was 43.8±3.4% of hemisphere area, and the hemisphere volume increased by 18.5±2.0% due to brain tissue swelling and edema. Administration of tryptanthrin and its oxime significantly decreased neurological deficits at all control points and reduced FCI size (by 24.2 and 30.4%, respectively) and brain tissue swelling of the affected hemisphere (by 64.9 and 62.7%, respectively). Therefore, the neuroprotective effect of tryptanthrine and its oxime in the acute period of FCI is largely determined by their anti-inflammatory activity.


Subject(s)
Infarction, Middle Cerebral Artery , Neuroprotective Agents , Oximes , Quinazolines , Rats, Wistar , Animals , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Male , Rats , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Quinazolines/pharmacology , Quinazolines/therapeutic use , Oximes/pharmacology , Oximes/therapeutic use , Brain Edema/drug therapy , Brain Edema/pathology , Disease Models, Animal , Brain/drug effects , Brain/pathology
3.
Cell Rep Med ; 5(8): 101663, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39094577

ABSTRACT

The current targeted therapy for BRAFV600E-mutant lung cancer consists of a dual blockade of RAF/MEK kinases often combining dabrafenib/trametinib (D/T). This regimen extends survival when compared to single-agent treatments, but disease progression is unavoidable. By using whole-genome CRISPR screening and RNA sequencing, we characterize the vulnerabilities of both persister and D/T-resistant cellular models. Oxidative stress together with concomitant induction of antioxidant responses is boosted by D/T treatment. However, the nature of the oxidative damage, the choice of redox detoxification systems, and the resulting therapeutic vulnerabilities display stage-specific differences. Persister cells suffer from lipid peroxidation and are sensitive to ferroptosis upon GPX4 inhibition in vivo. Biomarkers of lipid peroxidation are detected in clinical samples following D/T treatment. Acquired alterations leading to mitogen-activated protein kinase (MAPK) reactivation enhance cystine transport to boost GPX4-independent antioxidant responses. Similarly to BRAFV600E-mutant melanoma, histone deacetylase (HDAC) inhibitors decrease D/T-resistant cell viability and extend therapeutic response in vivo.


Subject(s)
Adenocarcinoma of Lung , Drug Resistance, Neoplasm , Histone Deacetylase Inhibitors , Lung Neoplasms , Phospholipid Hydroperoxide Glutathione Peroxidase , Proto-Oncogene Proteins B-raf , Humans , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Cell Line, Tumor , Animals , Histone Deacetylase Inhibitors/pharmacology , Ferroptosis/drug effects , Ferroptosis/genetics , Mice , Oxidative Stress/drug effects , Oximes/pharmacology , Imidazoles/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Lipid Peroxidation/drug effects , Mutation/genetics , Xenograft Model Antitumor Assays
4.
Exp Cell Res ; 442(1): 114215, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39182666

ABSTRACT

Mutant BRAF is a critical oncogenic driver in melanoma, making it an attractive therapeutic target. However, the success of targeted therapy using BRAF inhibitors vemurafenib and dabrafenib has been limited due to development of resistance, restricting their clinical efficacy. A prior knowledge of resistance mechanisms to BRAFi or any cancer drug can lead to development of drugs that overcome resistance thus improving clinical outcomes. In vitro cellular models are powerful systems that can be utilized to mimic and study resistance mechanisms. In this study, we employed a multi-omics approach to characterize a panel of BRAF mutant melanoma cell lines to develop and systematically characterize BRAFi persister and resistant cells using exome sequencing, proteomics and phosphoproteomics. Our datasets revealed frequently observed intrinsic and acquired, genetic and non-genetic mechanisms of BRAFi resistance that have been studied in patients who developed resistance. In addition, we identified proteins that can be potentially targeted to overcome BRAFi resistance. Overall, we demonstrate that in vitro systems can be utilized not only to predict resistance mechanisms but also to identify putative therapeutic targets.


Subject(s)
Drug Resistance, Neoplasm , Melanoma , Protein Kinase Inhibitors , Proteomics , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Proteomics/methods , Protein Kinase Inhibitors/pharmacology , Cell Line, Tumor , Mutation , Vemurafenib/pharmacology , Oximes/pharmacology , Multiomics , Imidazoles
5.
Molecules ; 29(16)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39202899

ABSTRACT

IQ-1 (11H-indeno[1,2-b]quinoxalin-11-one oxime) is a specific c-Jun N-terminal kinase (JNK) inhibitor with anticancer and neuro- and cardioprotective properties. Because aryloxime derivatives undergo cytochrome P450-catalyzed oxidation to nitric oxide (NO) and ketones in liver microsomes, NO formation may be an additional mechanism of IQ-1 pharmacological action. In the present study, electron paramagnetic resonance (EPR) of the Fe2+ complex with diethyldithiocarbamate (DETC) as a spin trap and hemoglobin (Hb) was used to detect NO formation from IQ-1 in the liver and blood of rats, respectively, after IQ-1 intraperitoneal administration (50 mg/kg). Introducing the spin trap and IQ-1 led to signal characteristics of the complex (DETC)2-Fe2+-NO in rat liver. Similarly, the introduction of the spin trap components and IQ-1 resulted in an increase in the Hb-NO signal for both the R- and the T-conformers in blood samples. The density functional theory (DFT) calculations were in accordance with the experimental data and indicated that the NO formation of IQ-1 through the action of superoxide anion radical is thermodynamically favorable. We conclude that the administration of IQ-1 releases NO during its oxidoreductive bioconversion in vivo.


Subject(s)
Nitric Oxide , Oximes , Quinoxalines , Electron Spin Resonance Spectroscopy/methods , Animals , Nitric Oxide/metabolism , Oximes/chemistry , Oximes/pharmacology , Rats , Quinoxalines/chemistry , Quinoxalines/pharmacology , Liver/metabolism , Liver/drug effects , Male , Hemoglobins/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/chemistry , Ditiocarb/pharmacology , Ditiocarb/chemistry
6.
Mol Ther ; 32(9): 3145-3162, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39097773

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has a survival rate of 12%, and multiple clinical trials testing anti-PD-1 therapies against PDAC have failed, suggesting a need for a novel therapeutic strategy. In this study, we evaluated the potential of milbemycin oxime (MBO), an antiparasitic compound, as an immunomodulatory agent in PDAC. Our results show that MBO inhibited the growth of multiple PDAC cell lines by inducing apoptosis. In vivo studies showed that the oral administration of 5 mg/kg MBO inhibited PDAC tumor growth in both subcutaneous and orthotopic models by 49% and 56%, respectively. Additionally, MBO treatment significantly increased the survival of tumor-bearing mice by 27 days as compared to the control group. Interestingly, tumors from MBO-treated mice had increased infiltration of CD8+ T cells. Notably, depletion of CD8+ T cells significantly reduced the anti-tumor efficacy of MBO in mice. Furthermore, MBO significantly augmented the efficacy of anti-PD-1 therapy, and the combination treatment resulted in a greater proportion of active cytotoxic T cells within the tumor microenvironment. MBO was safe and well tolerated in all our preclinical toxicological studies. Overall, our study provides a new direction for the use of MBO against PDAC and highlights the potential of repurposing MBO for enhancing anti-PD-1 immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Programmed Cell Death 1 Receptor , Tumor Microenvironment , Animals , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Mice , Humans , Cell Line, Tumor , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Xenograft Model Antitumor Assays , Macrolides/pharmacology , Macrolides/therapeutic use , Apoptosis/drug effects , Cell Proliferation/drug effects , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Oximes/pharmacology , Disease Models, Animal , Female
7.
Nutrients ; 16(16)2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39203879

ABSTRACT

Sulforaphane is considered the bioactive metabolite of glucoraphanin after dietary consumption of broccoli sprouts. Although both molecules pass through the gut lumen to the large intestine in stable form, their biological impact on the first intestinal tract is poorly described. In celiac patients, the function of the small intestine is affected by celiac disease (CD), whose severe outcomes are controlled by gluten-free dietary protocols. Nevertheless, pathological signs of inflammation and oxidative stress may persist. The aim of this study was to compare the biological activity of sulforaphane with its precursor glucoraphanin in a cellular model of gliadin-induced inflammation. Human intestinal epithelial cells (CaCo-2) were stimulated with a pro-inflammatory combination of cytokines (IFN-γ, IL-1ß) and in-vitro-digested gliadin, while oxidative stress was induced by H2O2. LC-MS/MS analysis confirmed that sulforaphane from broccoli sprouts was stable after simulated gastrointestinal digestion. It inhibited the release of all chemokines selected as inflammatory read-outs, with a more potent effect against MCP-1 (IC50 = 7.81 µM). On the contrary, glucoraphanin (50 µM) was inactive. The molecules were unable to counteract the oxidative damage to DNA (γ-H2AX) and catalase levels; however, the activity of NF-κB and Nrf-2 was modulated by both molecules. The impact on epithelial permeability (TEER) was also evaluated in a Transwell® model. In the context of a pro-inflammatory combination including gliadin, TEER values were recovered by neither sulforaphane nor glucoraphanin. Conversely, in the context of co-culture with activated macrophages (THP-1), sulforaphane inhibited the release of MCP-1 (IC50 = 20.60 µM) and IL-1ß (IC50 = 1.50 µM) only, but both molecules restored epithelial integrity at 50 µM. Our work suggests that glucoraphanin should not merely be considered as just an inert precursor at the small intestine level, thus suggesting a potential interest in the framework of CD. Its biological activity might imply, at least in part, molecular mechanisms different from sulforaphane.


Subject(s)
Brassica , Celiac Disease , Glucosinolates , Imidoesters , Isothiocyanates , Oxidative Stress , Oximes , Sulfoxides , Humans , Isothiocyanates/pharmacology , Sulfoxides/pharmacology , Glucosinolates/pharmacology , Glucosinolates/metabolism , Celiac Disease/drug therapy , Celiac Disease/diet therapy , Celiac Disease/metabolism , Caco-2 Cells , Oximes/pharmacology , Oxidative Stress/drug effects , Imidoesters/pharmacology , Brassica/chemistry , Gliadin/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Interleukin-1beta/metabolism , Chemokine CCL2/metabolism , Cytokines/metabolism , Interferon-gamma/metabolism
8.
J Biochem Mol Toxicol ; 38(7): e23750, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38952032

ABSTRACT

The treatment of organophosphate (OP) anticholinesterases currently lacks an effective oxime reactivator of OP-inhibited acetylcholinesterase (AChE) which can penetrate the blood-brain barrier (BBB). Our laboratories have synthesized novel substituted phenoxyalkyl pyridinium oximes and tested them for their ability to promote survival of rats challenged with lethal doses of nerve agent surrogates. These previous studies demonstrated the ability of some of these oximes to promote 24-h survival to rats challenged with a lethal level of highly relevant surrogates for sarin and VX. The reactivation of OP-inhibited AChE in peripheral tissues was likely to be a major contributor to their efficacy in survival of lethal OP challenges. In the present study, twenty of these novel oximes were screened in vitro for reactivation ability for AChE in rat skeletal muscle and serum using two nerve agent surrogates: phthalimidyl isopropyl methylphosphonate (PIMP, a sarin surrogate) and 4-nitrophenyl ethyl methylphosphonate (NEMP, a VX surrogate). The oximes demonstrated a range of 23%-102% reactivation of AChE in vitro across both tissue types. Some of the novel oximes tested in the present study demonstrated the ability to more effectively reactivate AChE in serum than the currently approved oxime, 2-PAM. Therefore, some of these novel oximes have the potential to reverse AChE inhibition in peripheral target tissues and contribute to survival efficacy.


Subject(s)
Acetylcholinesterase , Cholinesterase Inhibitors , Cholinesterase Reactivators , Muscle, Skeletal , Organophosphates , Oximes , Animals , Oximes/pharmacology , Oximes/chemistry , Rats , Acetylcholinesterase/metabolism , Acetylcholinesterase/blood , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/toxicity , Organophosphates/toxicity , Male , Cholinesterase Reactivators/pharmacology , Cholinesterase Reactivators/chemistry , Pyridinium Compounds/pharmacology , Rats, Sprague-Dawley
9.
Eur J Med Chem ; 276: 116630, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-38972081

ABSTRACT

We report here on the structure-activity relationships of hybrids combining 3-descladinosyl clarithromycin with quinolones linked by extended diamine connectors. Several hybrids, exemplified by 23Bc, 23Be, 23Bf, 26Be, and 30Bc, not only restored potency against inducibly resistant pathogens but also exhibited significantly enhanced activities against constitutively resistant strains of Staphylococcus pneumoniae and Staphylococcus pyogenes, which express high-level resistance independent of clarithromycin or erythromycin induction. Additionally, the novel hybrids showed susceptibility against Gram-negative Haemophilus influenzae. Notably, hybrid 23Be demonstrated dual modes of action by inhibiting both protein synthesis and DNA replication in vitro and in vivo. Given these promising characteristics, 23Be emerges as a potential candidate for the treatment of community-acquired bacterial pneumonia.


Subject(s)
Anti-Bacterial Agents , Clarithromycin , Drug Design , Microbial Sensitivity Tests , Structure-Activity Relationship , Clarithromycin/pharmacology , Clarithromycin/chemistry , Clarithromycin/chemical synthesis , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Molecular Structure , Diamines/chemistry , Diamines/pharmacology , Diamines/chemical synthesis , Haemophilus influenzae/drug effects , Oximes/chemistry , Oximes/pharmacology , Oximes/chemical synthesis , Dose-Response Relationship, Drug , Humans , Animals , Streptococcus pyogenes/drug effects , Drug Resistance, Bacterial/drug effects
10.
FEBS Lett ; 598(16): 2011-2027, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38977937

ABSTRACT

Malignant melanoma, an aggressive skin cancer with a poor prognosis, frequently features BRAFV600E mutation resulting in activation of the MAPK pathway and melanocyte proliferation and survival. BRAFV600E inhibitors like vemurafenib and dabrafenib have enhanced patient survival, yet drug resistance remains a significant challenge. We investigated the role of the ERK5 pathway in BRAFV600E melanoma cells and cells with acquired resistance to PLX4720 (vemurafenib) and dabrafenib. In BRAFV600E melanoma, ERK5 inhibition minimally affected viability compared to ERK1/2 inhibition. In vemurafenib-resistant cells, ERK5 inhibition alone didn't impact viability or restore drug sensitivity to vemurafenib. However, in dabrafenib-resistant cells, ERK5 inhibition reduced viability and enhanced the anti-proliferative effect of MEK1/2 inhibition. Targeting the ERK5 pathway may represent a therapeutic opportunity in dabrafenib-resistant melanoma.


Subject(s)
Drug Resistance, Neoplasm , Imidazoles , Melanoma , Mitogen-Activated Protein Kinase 7 , Oximes , Proto-Oncogene Proteins B-raf , Vemurafenib , Oximes/pharmacology , Humans , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Melanoma/metabolism , Imidazoles/pharmacology , Vemurafenib/pharmacology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Mitogen-Activated Protein Kinase 7/metabolism , Mitogen-Activated Protein Kinase 7/genetics , Indoles/pharmacology , Sulfonamides/pharmacology , Cell Proliferation/drug effects , MAP Kinase Signaling System/drug effects , Cell Survival/drug effects , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Mutation
11.
Pharmacol Res ; 207: 107298, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39032840

ABSTRACT

Acquired drug resistance is the major cause for disease recurrence in cancer patients, and this is particularly true for patients with metastatic melanoma that carry a BRAF V600E mutation. To address this problem, we investigated cyclic membrane-active peptides as an alternative therapeutic modality to kill drug-tolerant and resistant melanoma cells to avoid acquired drug resistance. We selected two stable cyclic peptides (cTI and cGm), previously shown to have anti-melanoma properties, and compared them with dabrafenib, a drug used to treat cancer patients with the BRAF V600E mutation. The peptides act via a fast membrane-permeabilizing mechanism and kill metastatic melanoma cells that are sensitive, tolerant, or resistant to dabrafenib. Melanoma cells do not become resistant to long-term treatment with cTI, nor do they evolve their lipid membrane composition, as measured by lipidomic and proteomic studies. In vivo studies in mice demonstrated that the combination treatment of cTI and dabrafenib resulted in fewer metastases and improved overall survival. Such cyclic membrane-active peptides are thus well suited as templates to design new anticancer therapeutic strategies.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Drug Resistance, Neoplasm , Imidazoles , Melanoma , Oximes , Peptides, Cyclic , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Animals , Melanoma/drug therapy , Melanoma/pathology , Humans , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Imidazoles/pharmacology , Imidazoles/therapeutic use , Cell Proliferation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Oximes/pharmacology , Oximes/therapeutic use , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/therapeutic use , Mice , Female , DNA-Binding Proteins
12.
Int Immunopharmacol ; 138: 112653, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38996664

ABSTRACT

As one of the main pathmechanisms of Alzheimer's disease (AD), amyloid-ß (Aß) is widely considered to be the prime target for the development of AD therapy. Recently, imidazolylacetophenone oxime ethers or esters (IOEs) have shown neuroprotective effects against neuronal cells damage, suggesting their potential use in the prevention and treatment of AD. Thirty IOEs compounds from our lab in-house library were constructed and screened for the inhibitory effects on Aß42-induced cytotoxicity. Among them, TJ1, as a new IOEs hit, preliminarily showed the effect on inhibiting Aß42-induced cytotoxicity. Furthermore, the inhibitory effects of TJ1 on Aß42 aggregation were tested by ThT assays and TEM. The neuroprotective effects of TJ1 were evaluated in Aß42-stimulated SH-SY5Y cells, LPS-stimulated BV-2 cells, and H2O2- and RSL3-stimulated PC12 cells. The cognitive improvement of TJ1 was assessed in 5xFAD (C57BL/6J) transgenic mouse. These results showed that TJ1 had strong neuroprotective effects and high blood-brain barrier (BBB) permeability without obvious cytotoxicity. TJ1 impeded the self-accumulation process of Aß42 by acting on Aß oligomerization and fibrilization. Besides, TJ1 reversed Aß-, H2O2- and RSL3-induced neuronal cell damage and decreased neuroinflammation. In 5xFAD mice, TJ1 improved cognitive impairment, increased GSH level, reduced the level of Aß42 and Aß plaques, and attenuated the glia reactivation and inflammatory response in the brain,. Taken together, our results demonstrate that TJ1 improves cognitive impairments as a new neuroprotective candidate via targeting amyloidogenesis, which suggests the potential of TJ1 as a treatment for AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Blood-Brain Barrier , Disease Models, Animal , Mice, Inbred C57BL , Mice, Transgenic , Neuroprotective Agents , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Humans , Mice , Rats , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Peptide Fragments/metabolism , PC12 Cells , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oximes/pharmacology , Oximes/therapeutic use , Cell Line, Tumor , Male
13.
Chem Biol Interact ; 399: 111131, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-38964639

ABSTRACT

Dabrafenib is a BRAF inhibitor that has been demonstrated to be efficacious in the treatment of melanoma and non-small-cell lung cancer patients with BRAF V600E mutations. The objective of this study was to investigate the effects of 51 traditional Chinese medicines on the metabolism of dabrafenib and to further investigate the inhibitory effect of imperatorin. The quantification of dabrafenib and its metabolite hydroxy-dabrafenib was carried out using a sensitive, rapid, and accurate assay method based on ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). The results of in vitro experiments showed that 20 drugs inhibited the metabolism of dabrafenib by more than 80 %. In a further study of imperatorin on dabrafenib, the half-maximal inhibitory concentration (IC50) values of imperatorin on dabrafenib were 0.22 µM and 3.68 µM in rat liver microsomes (RLM) and human liver microsomes (HLM), respectively, while the inhibition mechanisms were non-competitive and mixed type inhibition, respectively. The results of in vivo experiments demonstrated that in the presence of imperatorin, the AUC(0-t), AUC(0-∞), Cmax, and Tmax of dabrafenib were increased by 2.38-, 2.26-, 1.05-, and 6.10-fold, respectively, while CLz/F was decreased by 67.9 %. In addition, Tmax of hydroxy-dabrafenib was increased by 1.4-fold. The results of the research showed that imperatorin had a consistent inhibitory effect on dabrafenib in vitro and in vivo. When the concurrent use of dabrafenib and imperatorin is unavoidable, clinicians should closely monitor for potential adverse events and make timely adjustments to the administered dosage.


Subject(s)
Furocoumarins , Imidazoles , Microsomes, Liver , Oximes , Rats, Sprague-Dawley , Oximes/pharmacology , Imidazoles/pharmacology , Imidazoles/metabolism , Animals , Furocoumarins/pharmacology , Furocoumarins/metabolism , Microsomes, Liver/metabolism , Humans , Rats , Male , Tandem Mass Spectrometry , Chromatography, High Pressure Liquid
14.
Chem Biol Interact ; 399: 111138, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-38992768

ABSTRACT

Oxidative stress status, as a disruption of redox homeostasis, in the blood sera of Wistar rats caused by repeated application of selected acetylcholinesterase reactivators - asoxime, obidoxime, K027, K048, K074, and K075 were evaluated. Throughout this study, each oxime in a dose of 0.1 of LD50/kg im was given 2x/week for 4 weeks. Then, seven days after the last oximes' application, markers of lipid peroxidation (malondialdehyde, MDA), and protein oxidation (advanced oxidation protein products, AOPP), as well as the activity of antioxidant enzymes (catalase, CAT, superoxide dismutase, SOD, reduced glutathione, GSH, and oxidized glutathione, GSSG), were determined. Oxidative stress parameters, MDA and AOPP were significantly highest in the K048-, K074- and K075-treated groups (p < 0.001). The activity of CAT was significantly elevated in the obidoxime-treated group (p < 0.05), while treatment with K027, K048, and K074 induced high elevation in SOD levels (p < 0.01, p < 0.001). Interestingly, the activity of GSH in each oxime-treated group was significantly elevated. Unlike, treatment with obidoxime caused elevation in GSSG levels (p < 0.01). As a continuation of our previously published data, these results assure that applied oximes following subacute treatment ameliorated the oxidative status and further adverse systemic toxic effects in rats.


Subject(s)
Biomarkers , Glutathione , Oxidative Stress , Oximes , Rats, Wistar , Animals , Oxidative Stress/drug effects , Oximes/pharmacology , Biomarkers/blood , Rats , Male , Glutathione/blood , Glutathione/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase/blood , Lipid Peroxidation/drug effects , Catalase/metabolism , Catalase/blood , Malondialdehyde/blood , Malondialdehyde/metabolism , Cholinesterase Reactivators/pharmacology , Advanced Oxidation Protein Products/blood , Antioxidants/metabolism , Antioxidants/pharmacology
15.
Int J Biol Macromol ; 275(Pt 2): 133741, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38986985

ABSTRACT

BACKGROUND: Sarcopenia, characterized by progressive muscle dysfunction, is a common complication of chronic obstructive pulmonary disease (COPD). Our previous study revealed serum Lipoprotein-associated phospholipaseA2 (Lp-PLA2) level significantly increased in COPD and associated with exercise tolerance. This study further investigated the functions and target potential of Lp-PLA2 for sarcopenia in COPD. METHODS: The circulating Lp-PLA2 level/enzyme activity in COPD patients and age-matched healthy volunteers were measured. Clinical parameters on skeletal muscle were measured and their correlations with Lp-PLA2 were analyzed. We explored the involvement of Lp-PLA2 in vivo and treatment effectiveness of darapladib (a specific Lp-PLA2 inhibitor) in CS-induced muscle dysfunction models. RESULTS: Circulating Lp-PLA2 level/enzyme activity was elevated in COPD patients compared with healthy controls, negatively associated with skeletal muscle mass and function. In CS-induced muscle dysfunction murine models, up-regulated serum Lp-PLA2 level/enzyme activity was verified again. In CS-exposed mouse models, darapladib treatment reversed muscle mass loss and muscle dysfunction, meanwhile rescued upregulation of MuRF1 and atrogin-1, and activation of inflammatory factors, oxidant enzymes and NF-κB signaling. CONCLUSIONS: Lp-PLA2 could be a potential indicator for sarcopenia in COPD. Darapladib, a Lp-PLA2 inhibitor, can alleviate CS-induced skeletal muscle dysfunction and represents a potential therapeutic for sarcopenia in COPD.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase , Pulmonary Disease, Chronic Obstructive , Sarcopenia , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/complications , Humans , Sarcopenia/drug therapy , Sarcopenia/etiology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/blood , Male , Animals , Mice , Female , Aged , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Benzaldehydes/pharmacology , Benzaldehydes/therapeutic use , Oximes/therapeutic use , Oximes/pharmacology , Biomarkers/blood , Disease Models, Animal , Biphenyl Compounds , Diethylamines , Pyrimidinones , Sulfides
16.
J Virol ; 98(8): e0061824, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39023323

ABSTRACT

Zika virus (ZIKV) is a re-emerging mosquito-borne flavivirus that has been associated with congenital neurological defects in fetuses born to infected mothers. At present, no vaccine or antiviral therapy is available to combat this devastating disease. Repurposing drugs that target essential host factors exploited by viruses is an attractive therapeutic approach. Here, we screened a panel of clinically approved small-molecule kinase inhibitors for their antiviral effects against a clinical isolate of ZIKV and thoroughly characterized their mechanisms of action. We found that the Raf kinase inhibitors Dabrafenib and Regorafenib potently impair the replication of ZIKV, but not that of its close relative dengue virus. Time-of-addition experiments showed that both inhibitors target ZIKV infection at post-entry steps. We found that Dabrafenib, but not Regorafenib, interfered with ZIKV genome replication by impairing both negative- and positive-strand RNA synthesis. Regorafenib, on the other hand, altered steady-state viral protein levels, viral egress, and blocked NS1 secretion. We also observed Regorafenib-induced ER fragmentation in ZIKV-infected cells, which might contribute to its antiviral effects. Because these inhibitors target different steps of the ZIKV infection cycle, their use in combination therapy may amplify their antiviral effects which could be further explored for future therapeutic strategies against ZIKV and possibly other flaviviruses. IMPORTANCE: There is an urgent need to develop effective therapeutics against re-emerging arboviruses associated with neurological disorders like Zika virus (ZIKV). We identified two FDA-approved kinase inhibitors, Dabrafenib and Regorafenib, as potent inhibitors of contemporary ZIKV strains at distinct stages of infection despite overlapping host targets. Both inhibitors reduced viral titers by ~1 to 2 log10 (~10-fold to 100-fold) with minimal cytotoxicity. Furthermore, we show that Dabrafenib inhibits ZIKV RNA replication whereas Regorafenib inhibits ZIKV translation and egress. Regorafenib has the added benefit of limiting NS1 secretion, which contributes to the pathogenesis and disease progression of several flaviviruses. Because these inhibitors affect distinct post-entry steps of ZIKV infection, their therapeutic potential may be amplified by combination therapy and likely does not require prophylactic administration. This study provides further insight into ZIKV-host interactions and has implications for the development of novel antivirals against ZIKV and possibly other flaviviruses.


Subject(s)
Antiviral Agents , Imidazoles , Oximes , Phenylurea Compounds , Protein Kinase Inhibitors , Pyridines , Virus Replication , Zika Virus Infection , Zika Virus , Virus Replication/drug effects , Oximes/pharmacology , Zika Virus/drug effects , Pyridines/pharmacology , Humans , Imidazoles/pharmacology , Zika Virus Infection/virology , Zika Virus Infection/drug therapy , Chlorocebus aethiops , Animals , Phenylurea Compounds/pharmacology , Vero Cells , Antiviral Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Cell Line , Dengue Virus/drug effects
17.
Arch Toxicol ; 98(10): 3397-3407, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39004640

ABSTRACT

The risk of the use of toxic chemicals for unlawful acts has been a matter of concern for different governments and multilateral agencies. The Organisation for the Prohibition of Chemical Weapons (OPCW), which oversees the implementation of the Chemical Weapons Convention (CWC), considering recent events employing chemical warfare agents as means of assassination, has recently included in the CWC "Annex on Chemicals" some organophosphorus compounds that are regarded as acting in a similar fashion to the classical G- and V-series of nerve agents, inhibiting the pivotal enzyme acetylcholinesterase. Therefore, knowledge of the activity of the pyridinium oximes, the sole class of clinically available acetylcholinesterase reactivators to date, is plainly justified. In this paper, continuing our research efforts in medicinal chemistry on this class of toxic chemicals, we synthesized an A-230 nerve agent surrogate and applied a modified Ellman's assay to evaluate its ability to inhibit our enzymatic model, acetylcholinesterase from Electrophorus eel, and if the clinically available antidotes are able to rescue the enzyme activity for the purpose of relating the findings to the previously disclosed in silico data for the authentic nerve agent and other studies with similar A-series surrogates. Our experimental data indicates that pralidoxime is the most efficient compound for reactivating acetylcholinesterase inhibited by A-230 surrogate, which is the opposite of the in silico data previously disclosed.


Subject(s)
Acetylcholinesterase , Chemical Warfare Agents , Cholinesterase Inhibitors , Cholinesterase Reactivators , Nerve Agents , Oximes , Pyridinium Compounds , Oximes/pharmacology , Acetylcholinesterase/metabolism , Cholinesterase Reactivators/pharmacology , Cholinesterase Inhibitors/toxicity , Pyridinium Compounds/pharmacology , Chemical Warfare Agents/toxicity , Nerve Agents/toxicity , Pralidoxime Compounds/pharmacology , Organothiophosphorus Compounds/toxicity , Animals , Antidotes/pharmacology
18.
Toxicol In Vitro ; 100: 105892, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38996828

ABSTRACT

Targeting cancer cells through drug-based treatment or combination therapy protocols involving chemical compounds can be challenging due to multiple factors, including their resistance to bioactive compounds and the potential of drugs to damage healthy cells. This study aims to investigate the relationship between the structure of novel sulfur-containing shikonin oxime compounds and the corresponding cytotoxicity against four cancer types, namely colon, gastric, liver, and breast cancers, through computational chemistry tools. This investigation is suggested to help build insights into how the structure of the compounds influences their activity and understand the mechanisms behind it and subsequently might be used in multi-cancer drug design process to propose novel optimized compounds that potentially exhibit the desired activity. The findings showed that the cytotoxic activity against the four cancer types was accurately predictable (R2 > 0.7, NRMSE <20%) by a combination of search and machine learning algorithms, based on the information on the structure of the compounds, including their lipophilicity, surface area, and volume. Overall, this study is supposed to play a crucial role in effective multi-cancer drug design in cancer research areas.


Subject(s)
Antineoplastic Agents , Naphthoquinones , Oximes , Quantitative Structure-Activity Relationship , Humans , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Naphthoquinones/toxicity , Oximes/chemistry , Oximes/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Sulfur/chemistry , Machine Learning , Neoplasms/drug therapy
19.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38839106

ABSTRACT

Targeted therapies against mutant BRAF are effectively used in combination with MEK inhibitors (MEKi) to treat advanced melanoma. However, treatment success is affected by resistance and adverse events (AEs). Approved BRAF inhibitors (BRAFi) show high levels of target promiscuity, which can contribute to these effects. The blood vessel lining is in direct contact with high plasma concentrations of BRAFi, but effects of the inhibitors in this cell type are unknown. Hence, we aimed to characterize responses to approved BRAFi for melanoma in the vascular endothelium. We showed that clinically approved BRAFi induced a paradoxical activation of endothelial MAPK signaling. Moreover, phosphoproteomics revealed distinct sets of off-targets per inhibitor. Endothelial barrier function and junction integrity were impaired upon treatment with vemurafenib and the next-generation dimerization inhibitor PLX8394, but not with dabrafenib or encorafenib. Together, these findings provide insights into the surprisingly distinct side effects of BRAFi on endothelial signaling and functionality. Better understanding of off-target effects could help to identify molecular mechanisms behind AEs and guide the continued development of therapies for BRAF-mutant melanoma.


Subject(s)
Melanoma , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Signal Transduction , Vemurafenib , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Humans , Protein Kinase Inhibitors/pharmacology , Melanoma/drug therapy , Melanoma/metabolism , Signal Transduction/drug effects , Vemurafenib/pharmacology , Oximes/pharmacology , Sulfonamides/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Imidazoles/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , MAP Kinase Signaling System/drug effects , Carbamates/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Cell Line, Tumor , Mutation
20.
Molecules ; 29(12)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38930832

ABSTRACT

In this research, with an aim to develop novel pyrazole oxime ether derivatives possessing potential biological activity, thirty-two pyrazole oxime ethers, including a substituted pyridine ring, have been synthesized and structurally identified through 1H NMR, 13C NMR, and HRMS. Bioassay data indicated that most of these compounds owned strong insecticidal properties against Mythimna separata, Tetranychus cinnabarinus, Plutella xylostella, and Aphis medicaginis at a dosage of 500 µg/mL, and some title compounds were active towards Nilaparvata lugens at 500 µg/mL. Furthermore, some of the designed compounds had potent insecticidal effects against M. separata, T. cinnabarinus, or A. medicaginis at 100 µg/mL, with the mortalities of compounds 8a, 8c, 8d, 8e, 8f, 8g, 8o, 8s, 8v, 8x, and 8z against A. medicaginis, in particular, all reaching 100%. Even when the dosage was lowered to 20 µg/mL, compound 8s also expressed 50% insecticidal activity against M. separata, and compounds 8a, 8e, 8f, 8o, 8v, and 8x displayed more than 60% inhibition rates against A. medicaginis. The current results provided a significant basis for the rational design of biologically active pyrazole oxime ethers in future.


Subject(s)
Drug Design , Insecticides , Oximes , Pyrazoles , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrazoles/chemical synthesis , Oximes/chemistry , Oximes/pharmacology , Oximes/chemical synthesis , Insecticides/chemistry , Insecticides/chemical synthesis , Insecticides/pharmacology , Animals , Structure-Activity Relationship , Ethers/chemistry , Molecular Structure , Pyridines/chemistry , Pyridines/pharmacology , Pyridines/chemical synthesis , Moths/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL