Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
J Hazard Mater ; 476: 134945, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38905984

ABSTRACT

The escalating introduction of pesticides/veterinary drugs into the environment has necessitated a rapid evaluation of their potential risks to ecosystems and human health. The developmental toxicity of pesticides/veterinary drugs was less explored, and much less the large-scale predictions for untested pesticides, veterinary drugs and bio-pesticides. Alternative methods like quantitative structure-activity relationship (QSAR) are promising because their potential to ensure the sustainable and safe use of these chemicals. We collected 133 pesticides and veterinary drugs with half-maximal active concentration (AC50) as the zebrafish embryo developmental toxicity endpoint. The QSAR model development adhered to rigorous OECD principles, ensuring that the model possessed good internal robustness (R2 > 0.6 and QLOO2 > 0.6) and external predictivity (Rtest2 > 0.7, QFn2 >0.7, and CCCtest > 0.85). To further enhance the predictive performance of the model, a quantitative read-across structure-activity relationship (q-RASAR) model was established using the combined set of RASAR and 2D descriptors. Mechanistic interpretation revealed that dipole moment, the presence of C-O fragment at 10 topological distance, molecular size, lipophilicity, and Euclidean distance (ED)-based RA function were main factors influencing toxicity. For the first time, the established QSAR and q-RASAR models were combined to prioritize the developmental toxicity of a vast array of true external compounds (pesticides/veterinary drugs/bio-pesticides) lacking experimental values. The prediction reliability of each query molecule was evaluated by leverage approach and prediction reliability indicator. Overall, the dual computational toxicology models can inform decision-making and guide the design of new pesticides/veterinary drugs with improved safety profiles.

2.
Int J Biol Macromol ; 274(Pt 2): 133404, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38925197

ABSTRACT

As a clinical anti-glioma agent, the therapeutic effect of carmustine (BCNU) was largely decreased because of the drug resistance mediated by O6-alkylguanine-DNA alkyltransferase (AGT) and the blood-brain barrier (BBB). To overcome these obstacles, we synthesized a BCNU-loaded hypoxia/esterase dual stimulus-activated nanomicelle, abbreviated as T80-HACB/BCNU NPs. In this nano-system, Tween 80 acts as the functional coating on the surface of the micelle to facilitate transport across the BBB. Hyaluronic acid (HA) with active tumor-targeting capability was linked with the hypoxia-sensitive AGT inhibitors (O6-azobenzyloxycarbonyl group) via an esterase-activated ester bond. The obtained T80-HACB/BCNU NPs had an average particle size of 232.10 ± 10.66 nm, the zeta potential of -18.13 ± 0.91 mV, and it showed high drug loading capacity, eximious biocompatibility and dual activation of hypoxia/esterase drug release behavior. The obtained T80-HACB/BCNU NPs showed enhanced cytotoxicity against hypoxic T98G and SF763 cells with IC50 at 132.2 µM and 133.1 µM, respectively. T80 modification improved the transportation of the micelle across an in vitro BBB model. The transport rate of the T80-HACB/Cou6 NPs group was 12.37 %, which was 7.6-fold (p<0.001) higher than the micelle without T80 modification. T80-HACB/BCNU NPs will contribute to the development of novel CENUs chemotherapies with high efficacy.

3.
Curr Res Food Sci ; 8: 100779, 2024.
Article in English | MEDLINE | ID: mdl-38939611

ABSTRACT

Whitespotted conger (Conger myriaster) muscle proteins were susceptible to oxidative denaturation during frozen storage. The objective of this study was to investigate the alterations in quality through physicochemical analysis and proteomics after whitespotted conger stored at temperatures of -18 °C and -60 °C. The microstructural observation revealed the noticeable variations such as increased interstitial space and fractured muscle fibre with extension of frozen storage time, and the muscle fibre of whitespotted conger stored at -60 °C were more intact than those stored at -18 °C. The raised TVB-N value indicated that the freshness of whitespotted conger decreased during 120-day frozen storage period. Analysis of myofibrillar protein content and SDS-PAGE demonstrated that compared to -18 °C, lower storage temperature (-60 °C) could better maintain the structure of whitespotted conger muscle by inhibiting protein degradation and oxidation. To reveal the mechanism of protein degradation, label-free quantitative proteomic analysis was performed through LC-MS/MS. The structural proteins including domain-associated proteins and actin-related proteins were up-regulated during frozen storage, but the phosphoglycerate kinase, phosphoglycerate mutase, and fructose-bisphosphate aldolase were down-regulated. Storage at -18 °C accelerated the up- or down-regulation of those differentially abundant proteins. According to KEGG analysis, up- or down-regulated pathways such as glycolysis/gluconeogenesis, carbon metabolism, biosynthesis of amino acids, and calcium signalling pathway mainly accounted for the protein degradation and quality reduction of whitespotted conger at low temperature. These results provided a theoretical basis for improving the quality stability of whitespotted conger during frozen storage.

4.
ACS Pharmacol Transl Sci ; 7(5): 1518-1532, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38751635

ABSTRACT

Tumor resistance seriously hinders the clinical application of chloroethylnitrosoureas (CENUs), such as O6-methylguanine-DNA methylguanine (MGMT), which can repair O6-alkyl lesions, thereby inhibiting the formation of cytotoxic DNA interstrand cross-links (ICLs). Metabolic differences between tumor and normal cells provide a biochemical basis for novel therapeutic strategies aimed at selectively inhibiting tumor energy metabolism. In this study, the energy blocker lonidamine (LND) was selected as a chemo-sensitizer of nimustine (ACNU) to explore its potential effects and underlying mechanisms in human glioblastoma in vitro and in vivo. A series of cell-level studies showed that LND significantly increased the cytotoxic effects of ACNU on glioblastoma cells. Furthermore, LND plus ACNU enhanced the energy deficiency by inhibiting glycolysis and mitochondrial function. Notably, LND almost completely downregulated MGMT expression by inducing intracellular acidification. The number of lethal DNA ICLs produced by ACNU increased after the LND pretreatment. The combination of LND and ACNU aggravated cellular oxidative stress. In resistant SF763 mouse tumor xenografts, LND plus ACNU significantly inhibited tumor growth with fewer side effects than ACNU alone. Finally, we proposed a new "HMAGOMR" chemo-sensitizing mechanism through which LND may act as a potential chemo-sensitizer to reverse ACNU resistance in glioblastoma: moderate inhibition of hexokinase (HK) activity (H); mitochondrial dysfunction (M); suppressing adenosine triphosphate (ATP)-dependent drug efflux (A); changing redox homeostasis to inhibit GSH-mediated drug inactivation (G) and increasing intracellular oxidative stress (O); downregulating MGMT expression through intracellular acidification (M); and partial inhibition of energy-dependent DNA repair (R).

5.
Arch Toxicol ; 98(7): 2213-2229, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38627326

ABSTRACT

All areas of the modern society are affected by fluorine chemistry. In particular, fluorine plays an important role in medical, pharmaceutical and agrochemical sciences. Amongst various fluoro-organic compounds, trifluoromethyl (CF3) group is valuable in applications such as pharmaceuticals, agrochemicals and industrial chemicals. In the present study, following the strict OECD modelling principles, a quantitative structure-toxicity relationship (QSTR) modelling for the rat acute oral toxicity of trifluoromethyl compounds (TFMs) was established by genetic algorithm-multiple linear regression (GA-MLR) approach. All developed models were evaluated by various state-of-the-art validation metrics and the OECD principles. The best QSTR model included nine easily interpretable 2D molecular descriptors with clear physical and chemical significance. The mechanistic interpretation showed that the atom-type electro-topological state indices, molecular connectivity, ionization potential, lipophilicity and some autocorrelation coefficients are the main factors contributing to the acute oral toxicity of TFMs against rats. To validate that the selected 2D descriptors can effectively characterize the toxicity, we performed the chemical read-across analysis. We also compared the best QSTR model with public OPERA tool to demonstrate the reliability of the predictions. To further improve the prediction range of the QSTR model, we performed the consensus modelling. Finally, the optimum QSTR model was utilized to predict a true external set containing many untested/unknown TFMs for the first time. Overall, the developed model contributes to a more comprehensive safety assessment approach for novel CF3-containing pharmaceuticals or chemicals, reducing unnecessary chemical synthesis whilst saving the development cost of new drugs.


Subject(s)
Quantitative Structure-Activity Relationship , Toxicity Tests, Acute , Animals , Rats , Administration, Oral , Toxicity Tests, Acute/methods , Algorithms , Hydrocarbons, Fluorinated/toxicity , Linear Models
6.
J Chem Inf Model ; 64(8): 3411-3429, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38511939

ABSTRACT

Chloroethylnitrosoureas (CENUs) are important chemotherapies applied in the treatment of cancer. They exert anticancer activity by inducing DNA interstrand cross-links (ICLs) via the formation of two O6-alkylguanine intermediates, O6-chloroethylguanine (O6-ClEtG) and N1,O6-ethanoguanine (N1,O6-EtG). However, O6-alkylguanine-DNA alkyltransferase (AGT), a DNA-repair enzyme, can restore the O6-alkylguanine damages and thereby obstruct the formation of ICLs (dG-dC cross-link). In this study, the inhibitory mechanism of ICL formation was investigated to elucidate the drug resistance of CENUs mediated by AGT in detail. Based on the structures of the substrate-enzyme complexes obtained from docking and MD simulations, two ONIOM (QM/MM) models with different sizes of the QM region were constructed. The model with a larger QM region, which included the substrate (O6-ClEtG or N1,O6-EtG), a water molecule, and five residues (Tyr114, Cys145, His146, Lys165, and Glu172) in the active pocket of AGT, accurately described the repairing reaction and generated the results coinciding with the experimental outcomes. The repair process consists of two sequential steps: hydrogen transfer to form a thiolate anion on Cys145 and alkyl transfer from the O6 site of guanine (the rate-limiting step). The repair of N1,O6-EtG was more favorable than that of O6-ClEtG from both kinetics and thermodynamics aspects. Moreover, the comparison of the repairing process with the formation of dG-dC cross-link and the inhibition of AGT by O6-benzylguanine (O6-BG) showed that the presence of AGT could effectively interrupt the formation of ICLs leading to drug resistance, and the inhibition of AGT by O6-BG that was energetically more favorable than the repair of O6-ClEtG could not prevent the repair of N1,O6-EtG. Therefore, it is necessary to completely eliminate AGT activity before CENUs medication to enhance the chemotherapeutic effectiveness. This work provides reasonable explanations for the supposed mechanism of AGT-mediated drug resistance of CENUs and will assist in the development of novel CENU chemotherapies and their medication strategies.


Subject(s)
DNA Repair , Molecular Docking Simulation , Molecular Dynamics Simulation , O(6)-Methylguanine-DNA Methyltransferase , O(6)-Methylguanine-DNA Methyltransferase/metabolism , O(6)-Methylguanine-DNA Methyltransferase/chemistry , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Humans , Quantum Theory , Drug Resistance, Neoplasm/drug effects , Nitrosourea Compounds/chemistry , Nitrosourea Compounds/pharmacology , Nitrosourea Compounds/metabolism
7.
8.
J Agric Food Chem ; 72(6): 3055-3065, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38298105

ABSTRACT

Alginate lyase degrades alginate by the ß-elimination mechanism to produce unsaturated alginate oligosaccharides (UAOS), which have better bioactivities than saturated AOS. Enhancing the thermal stability of alginate lyases is crucial for their industrial applications. In this study, a feasible and efficient rational design strategy was proposed by combining the computer-aided ΔΔG value calculation with the B-factor analysis. Two thermal stability-enhanced mutants, Q246V and K249V, were obtained by site-directed mutagenesis. Particularly, the t1/2, 50 °C for mutants Q246V and K249V was increased from 2.36 to 3.85 and 3.65 h, respectively. Remarkably, the specific activities of Q246V and K249V were enhanced to 2.41- and 2.96-fold that of alginate lyase AlyMc, respectively. Structural analysis and molecular dynamics simulations suggested that mutations enhanced the hydrogen bond networks and the overall rigidity of the molecular structure. Notably, mutant Q246V exhibited excellent thermal stability among the PL-7 alginate lyase family, especially considering the heightened enzymatic activity. Moreover, the rational design strategy used in this study can effectively improve the thermal stability of enzymes and has important significance in advancing applications of alginate lyase.


Subject(s)
Alginates , Polysaccharide-Lyases , Polysaccharide-Lyases/chemistry , Alginates/chemistry , Oligosaccharides/chemistry , Substrate Specificity , Hydrogen-Ion Concentration
9.
Heliyon ; 10(2): e24209, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38293468

ABSTRACT

The outbreak of the novel coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused great harm to all countries worldwide. This disease can be prevented by vaccination and managed using various treatment methods, including injections, oral medications, or aerosol therapies. However, the selection of suitable compounds for the research and development of anti-SARS-CoV-2 drugs is a daunting task because of the vast databases of available compounds. The traditional process of drug research and development is time-consuming, labour-intensive, and costly. The application of chemometrics can significantly expedite drug R&D. This is particularly necessary and important for drug development against pandemic public emergency diseases, such as COVID-19. Through various chemometric techniques, such as quantitative structure-activity relationship (QSAR) modelling, molecular docking, and molecular dynamics (MD) simulations, compounds with inhibitory activity against SARS-CoV-2 can be quickly screened, allowing researchers to focus on the few prioritised candidates. In addition, the ADMET properties of the screened candidate compounds should be further explored to promote the successful discovery of anti-SARS-CoV-2 drugs. In this case, considerable time and economic costs can be saved while minimising the need for extensive animal experiments, in line with the 3R principles. This paper focuses on recent advances in chemometric modelling studies of COVID-19-related inhibitors, highlights current limitations, and outlines potential future directions for development.

10.
J Hazard Mater ; 465: 133410, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38185092

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) represent a common group of environmental pollutants that endanger various aquatic organisms via various pathways. To better prioritize the ecotoxicological hazard of PAHs to aquatic environment, we used 2D descriptors-based quantitative structure-toxicity relationship (QSTR) to assess the toxicity of PAHs toward six aquatic model organisms spanning three trophic levels. According to strict OECD guideline, six easily interpretable, transferable and reproducible 2D-QSTR models were constructed with high robustness and reliability. A mechanistic interpretation unveiled the key structural factors primarily responsible for controlling the aquatic ecotoxicity of PAHs. Furthermore, quantitative read-across and different machine learning approaches were employed to validate and optimize the modelling approach. Importantly, the optimum QSTR models were further applied for predicting the ecotoxicity of hundreds of untested/unknown PAHs gathered from Pesticide Properties Database (PPDB). Especially, we provided a priority list in terms of the toxicity of unknown PAHs to six aquatic species, along with the corresponding mechanistic interpretation. In summary, the models can serve as valuable tools for aquatic risk assessment and prioritization of untested or completely new PAHs chemicals, providing essential guidance for formulating regulatory policies.


Subject(s)
Polycyclic Aromatic Hydrocarbons , Water Pollutants, Chemical , Polycyclic Aromatic Hydrocarbons/toxicity , Reproducibility of Results , Water Pollutants, Chemical/chemistry , Ecotoxicology , Aquatic Organisms , Quantitative Structure-Activity Relationship
11.
Molecules ; 28(23)2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38067461

ABSTRACT

Molecular toxicology is a field that investigates the interactions between chemical or biological molecules and organisms at the molecular level [...].


Subject(s)
Neoplasms , Toxicology , Humans , Neoplasms/genetics , Neoplasms/prevention & control
12.
Biomed Pharmacother ; 167: 115631, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37804814

ABSTRACT

Glioma is extremely difficult to be completely excised by surgery due to its invasive nature. Thus, chemotherapy still is the mainstay in the treatment of glioma after surgery. However, the natural blood-brain barrier (BBB) greatly restricts the penetration of chemotherapeutic agents into the central nervous system. As a front-line anti-glioma agent in clinical, carmustine (BCNU) exerts antitumor effect by inducing DNA damage at the O6 position of guanine. However, the therapeutic effect of BCNU was largely decreased because of the drug resistance mediated by O6-alkylguanine-DNA alkyltransferase (AGT) and insufficient local drug concentrations. To overcome these obstacles, we synthesized a BCNU-loaded hypoxia-responsive nano-micelle with BBB penetrating capacity and AGT inhibitory activity, named as T80-HA-AZO-BG/BCNU NPs. In this nano-system, Tween 80 (T80) serves as a functional coating on the surface of the micelle, promoting transportation across the BBB. Hyaluronic acid (HA) with active tumor-targeting capability was linked with the hydrophobic O6-benzylguanine (BG) analog via a hypoxia-sensitive azo bond. Under hypoxic tumor microenvironment, the azo bond selectively breaks to release O6-BG as AGT inhibitor and BCNU as DNA alkylating agent. The synthesized T80-HA-AZO-BG/BCNU NPs showed good stability, favorable biocompatibility and hypoxia-responsive drug-releasing ability. T80 modification improved the transportation of the micelle across an in vitro BBB model. Moreover, T80-HA-AZO-BG/BCNU NPs exhibited significantly enhanced cytotoxicity against glioma cell lines with high AGT expression compared with traditional combined medication of BCNU plus O6-BG. We expect that the tumor-targeting nano-micelle designed for chloroethylnitrosourea will provide new tools for the development of effective glioma therapy.


Subject(s)
Carmustine , Glioma , Humans , Carmustine/pharmacology , Carmustine/therapeutic use , Micelles , Blood-Brain Barrier , Glioma/drug therapy , Hypoxia/drug therapy , Tumor Microenvironment
13.
Pharmaceutics ; 15(8)2023 Aug 21.
Article in English | MEDLINE | ID: mdl-37631385

ABSTRACT

O6-methylguanine-DNA methyltransferase (MGMT) constitutes an important cellular mechanism for repairing potentially cytotoxic DNA damage induced by guanine O6-alkylating agents and can render cells highly resistant to certain cancer chemotherapeutic drugs. A wide variety of potential MGMT inactivators have been designed and synthesized for the purpose of overcoming MGMT-mediated tumor resistance. We determined the inactivation potency of these compounds against human recombinant MGMT using [3H]-methylated-DNA-based MGMT inactivation assays and calculated the IC50 values. Using the results of 370 compounds, we performed quantitative structure-activity relationship (QSAR) modeling to identify the correlation between the chemical structure and MGMT-inactivating ability. Modeling was based on subdividing the sorted pIC50 values or on chemical structures or was random. A total of nine molecular descriptors were presented in the model equation, in which the mechanistic interpretation indicated that the status of nitrogen atoms, aliphatic primary amino groups, the presence of O-S at topological distance 3, the presence of Al-O-Ar/Ar-O-Ar/R..O..R/R-O-C=X, the ionization potential and hydrogen bond donors are the main factors responsible for inactivation ability. The final model was of high internal robustness, goodness of fit and prediction ability (R2pr = 0.7474, Q2Fn = 0.7375-0.7437, CCCpr = 0.8530). After the best splitting model was decided, we established the full model based on the entire set of compounds using the same descriptor combination. We also used a similarity-based read-across technique to further improve the external predictive ability of the model (R2pr = 0.7528, Q2Fn = 0.7387-0.7449, CCCpr = 0.8560). The prediction quality of 66 true external compounds was checked using the "Prediction Reliability Indicator" tool. In summary, we defined key structural features associated with MGMT inactivation, thus allowing for the design of MGMT inactivators that might improve clinical outcomes in cancer treatment.

14.
J Nanobiotechnology ; 21(1): 291, 2023 Aug 23.
Article in English | MEDLINE | ID: mdl-37612719

ABSTRACT

Carmustine (BCNU), a vital type of chloroethylnitrosourea (CENU), inhibits tumor cells growth by inducing DNA damage at O6 position of guanine and eventually forming dG-dC interstrand cross-links (ICLs). However, the clinical application of BCNU is hindered to some extent by the absence of tumor selectivity, poor stability and O6-alkylguanine-DNA alkyltransferase (AGT) mediated drug resistance. In recent years, tumor microenvironment has been widely utilized for advanced drug delivery. In the light of the features of tumor microenvironment, we constructed a multifunctional hypoxia/esterase-degradable nanomicelle with AGT inhibitory activity named HACB NPs for tumor-targeting BCNU delivery and tumor sensitization. HACB NPs was self-assembled from hyaluronic acid azobenzene AGT inhibitor conjugates, in which O6-BG analog acted as an AGT inhibitor, azobenzene acted as a hypoxia-responsive linker and carboxylate ester bond acted as both an esterase-sensitive switch and a connector with hyaluronic acid (HA). The obtained HACB NPs possessed good stability, favorable biosafety and hypoxia/esterase-responsive drug-releasing ability. BCNU-loaded HACB/BCNU NPs exhibited superior cytotoxicity and apoptosis-inducing ability toward the human uterine cervix carcinoma HeLa cells compared with traditional combined medication of BCNU plus O6-BG. In vivo studies further demonstrated that after a selective accumulation in the tumor site, the micelles could respond to hypoxic tumor tissue for rapid drug release to an effective therapeutic dosage. Thus, this multifunctional stimulus-responsive nanocarrier could be a new promising strategy to enhance the anticancer efficacy and reduce the side effects of BCNU and other CENUs.


Subject(s)
Carcinoma , Carmustine , Female , Humans , Carmustine/pharmacology , HeLa Cells , Hyaluronic Acid , Tumor Microenvironment
15.
Int J Biol Macromol ; 246: 125657, 2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37399878

ABSTRACT

Carmustine (BCNU) is a typical chemotherapy used for treatment of cerebroma and other solid tumors, which exerts antitumor effect by inducing DNA damage at O6 position of guanine. However, the clinical application of BCNU was extremely limited due to the drug resistance mainly mediated by O6-alkylguanine-DNA alkyltransferase (AGT) and absence of tumor-targeting ability. To overcome these limitations, we developed a hypoxia-responsive nanomicelle with AGT inhibitory activity, which was successfully loaded with BCNU. In this nano-system, hyaluronic acid (HA) acts as an active tumor-targeting ligand to bind the overexpressing CD44 receptors on the surface of tumor cells. An azo bond selectively breaks in hypoxic tumor microenvironment to release O6-benzylguanine (BG) as AGT inhibitor and BCNU as DNA alkylating agent. The obtained HA-AZO-BG NPs with shell core structure had an average particle size of 176.98 ± 11.19 nm and exhibited good stability. Meanwhile, HA-AZO-BG NPs possessed a hypoxia-responsive drug release profile. After immobilizing BCNU into HA-AZO-BG NPs, the obtained HA-AZO-BG/BCNU NPs exhibited obvious hypoxia-selectivity and superior cytotoxicity in T98G, A549, MCF-7 and SMMC-7721 cells with IC50 at 189.0, 183.2, 90.1 and 100.1 µm, respectively, under hypoxic condition. Near-infrared imaging in HeLa tumor xenograft models showed that HA-AZO-BG/DiR NPs could effectively accumulate in tumor site at 4 h of post-injection, suggesting its good tumor-targetability. In addition, in vivo anti-tumor efficacy and toxicity evaluation indicated that HA-AZO-BG/BCNU NPs was more effective and less harmful compared to the other groups. After treatment, the tumor weight of HA-AZO-BG/BCNU NPs group was 58.46 % and 63.33 % of the control group and BCNU group, respectively. Overall, HA-AZO-BG/BCNU NPs was expected to be a promising candidate for targeted delivery of BCNU and elimination of chemoresistance.


Subject(s)
Antineoplastic Agents, Alkylating , Carmustine , Humans , Carmustine/pharmacology , Micelles , Tumor Cells, Cultured , Carrier Proteins , Hypoxia , Hyaluronan Receptors
16.
Biochem Pharmacol ; 215: 115726, 2023 09.
Article in English | MEDLINE | ID: mdl-37524206

ABSTRACT

Guanine O6-alkylating agents are widely used as first-line chemotherapeutic drugs due to their ability to induce cytotoxic DNA damage. However, a major hurdle in their effectiveness is the emergence of chemoresistance, largely attributed to the DNA repair pathway mediated by O6-methylguanine-DNA methyltransferase (MGMT). MGMT plays an important role in removing the alkyl groups from lethal O6-alkylguanine (O6-AlkylG) adducts formed by chemotherapeutic alkylating agents. By doing so, MGMT enables tumor cells to evade apoptosis and develop drug resistance toward DNA alkylating agents. Although covalent inhibitors of MGMT, such as O6-benzylguanine (O6-BG) and O6-(4-bromothenyl)guanine (O6-4-BTG or lomeguatrib), have been explored in clinical settings, their utility is limited due to severe delayed hematological toxicity observed in most patients when combined with alkylating agents. Therefore, there is an urgent need to identify new targets and unravel the underlying molecular mechanisms and to develop alternative therapeutic strategies that can overcome MGMT-mediated tumor resistance. In this context, the regulation of MGMT expression via interfering the specific cell signaling pathways (e.g., Wnt/ß-catenin, NF-κB, Hedgehog, PI3K/AKT/mTOR, JAK/STAT) emerges as a promising strategy for overcoming tumor resistance, and ultimately enhancing the efficacy of DNA alkylating agents in chemotherapy.


Subject(s)
Neoplasms , O(6)-Methylguanine-DNA Methyltransferase , Humans , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Neoplasms/metabolism , Alkylating Agents/therapeutic use , Signal Transduction , DNA , DNA Modification Methylases/metabolism , DNA Modification Methylases/therapeutic use , Tumor Suppressor Proteins/metabolism , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/therapeutic use
17.
Sci Total Environ ; 876: 162736, 2023 Jun 10.
Article in English | MEDLINE | ID: mdl-36907405

ABSTRACT

Fused/non-fused polycyclic aromatic hydrocarbons (FNFPAHs) have a variety of toxic effects on ecosystems and human body, but the acquisition of their toxicity data is greatly limited by the limited resources available. Here, we followed the EU REACH regulation and used Pimephales promelas as a model organism to investigate the quantitative structure-activity relationship (QSAR) between the FNFPAHs and their toxicity for the aquatic environment for the first time. We developed a single QSAR model (SM1) containing five simple and interpretable 2D molecular descriptors, which met the validation of OECD QSAR-related principles, and analyzed their mechanistic relationships with toxicity in detail. The model had good degree of fitting and robustness, and had better external prediction performance (MAEtest = 0.4219) than ECOSAR model (MAEtest = 0.5614). To further enhance its prediction accuracy, the three qualified single models (SMs) were used for constructing consensus models (CMs), the best one CM2 (MAEtest = 0.3954) had a significantly higher prediction accuracy for test compounds than SM1, and also outperformed the T.E.S.T. consensus model (MAEtest = 0.4233). Subsequently, the toxicity of 252 true external FNFPAHs from Pesticide Properties Database (PPDB) was predicted by SM1, the prediction results showed that 94.84 % compounds were reliably predicted within the model's application domain (AD). We also applied the best CM2 to predict the untested 252 FNFPAHs. Furthermore, we provided a mechanistic analysis and explanation for pesticides ranked as top 10 most toxic FNFPAHs. In summary, all developed QSAR and consensus models can be used as efficient tools for predicting the acute toxicity of unknown FNFPAHs to Pimephales promelas, thus being important for the risk assessment and regulation of FNFPAHs contamination in aquatic environment.


Subject(s)
Cyprinidae , Quantitative Structure-Activity Relationship , Animals , Humans , Consensus , Ecosystem , Ecotoxicology
18.
Org Lett ; 25(14): 2420-2425, 2023 Apr 14.
Article in English | MEDLINE | ID: mdl-37001013

ABSTRACT

A simple, efficient method has been developed for the CoIII-H-catalyzed aerobic C-C bond cleavage of tertiary allylic alcohols to access ketones. This novel approach presents excellent chemoselectivity, good functional group compatibility, and high yields. This reaction occurs through a HAT-initiated peroxide intermediate, and an adjacent glycol-type diradical fragmentation process is recommended.

19.
Food Sci Nutr ; 11(2): 1013-1023, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36789073

ABSTRACT

In this study, dry-cured Spanish mackerel (Scomberomorus niphonius, DCSM) was prepared via three different methods (hot-air drying, cold-air drying, and sun drying). The content of 4-hydroxy-2-hexenal (HHE) and 4-hydroxy-2-nonenal (HNE) derived from lipid oxidation in whole processes was investigated by HPLC-MS/MS. The changes in fatty acid composition were detected by GC-MS, and the degree of lipid oxidation was evaluated by the levels of acid values (AV), peroxide values (POV), and thiobarbituric acid-reactive substances (TBARS). The results showed that the drying process significantly accelerated lipid oxidation in DCSM. The contents of HHE and HNE were significantly increased after processing. The content of HHE was higher by 18.44-, 13.45-, and 16.32-folds compared with that of HNE after three different processes, respectively. The HHE and HNE contents fluctuated upward during the hot-air and cold-air drying process. However, the contents of HHE and HNE increased time-dependent during the sun drying process, with the highest values of 86.33 ± 10.54 and 5.29 ± 0.54 mg/kg fish among the three different processes. Besides, there was a significant positive correlation between HHE contents and n-3 fatty acids content in hot-air drying and sun drying processes (Pearson's r = .991/.996), and HNE occurrence was closely related to n-6 fatty acid content in sun drying process (Pearson's r = .989). Regression analysis indicated that the content of HHE and TOTOXTBA values in DCSM showed good linear relationships (R 2 value = .907), which suggested that the content of HHE could be used to estimate the oxidative deterioration of dry-cured fish products.

20.
J Hazard Mater ; 448: 130912, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36758436

ABSTRACT

Direct, convenient, and sensitive monitoring of the residues of multiple drugs in complex environments is important but remains a challenge. Here, we report a surface-enhanced Raman scattering (SERS)-based multiplexed lateral flow immunoassay (LFA) that supports the simultaneous and sensitive detection of commonly used drugs kanamycin, ractopamine, clenbuterol, and chloramphenicol in unprocessed complex samples through the dual signal amplification strategy of numerous efficient hotspots and magnetic enrichment. Multilayered magnetic-core dual-shell nanoparticles (MDAu@Ag) with controllable subtle nanogaps were fabricated via the polyethyleneimine-mediated layer-by-layer (LBL) assembly of two layers of Au@Ag satellites onto superparamagnetic Fe3O4 cores and conjugated with specific antibodies as multifunctional tags in the LFA system for rapid capture, separation, and quantitative analysis. Two Raman reporters were embedded in internal nanogaps and modified on the surface of MDAu@Ag for the simultaneous and ultrasensitive detection of four targets on two test lines, which greatly simplified the fabrication and signal reading of SERS-LFA. The proposed assay can rapidly detect multiple drug residues in 35 min with detection limits down to pg/mL level. Moreover, the MDAu@Ag-based SERS-LFA demonstrated better stability, higher throughput, and superior sensitivity (at least 400 times) than traditional colloidal gold immunochromatography, showing its great potential in the field of point-of-care testing.


Subject(s)
Metal Nanoparticles , Nanoparticles , Veterinary Drugs , Antibodies , Immunoassay , Spectrum Analysis, Raman/methods , Magnetic Phenomena , Metal Nanoparticles/chemistry , Limit of Detection
SELECTION OF CITATIONS
SEARCH DETAIL
...