TOXICOLOGY AND APPLIED PHARMACOLOGY

High-density lipoprotein cholesterol protects against delayed encephalopathy after acute carbon monoxide poisoning
Mu Y, Gao N, Wang Y, Wang S and Pang L
High-density lipoprotein cholesterol (HDL-C) plays a crucial role in neurological disorders. In this study, we aimed to elucidate the role of HDL-C in delayed encephalopathy after acute carbon monoxide poisoning (DEACMP), which presents with both neurological and psychiatric symptoms. Two-sample Mendelian randomization was employed on 201 lipid summary statistics to investigate potential causality. Data from the FinnGen database of 306,787 individuals were used. Mendelian randomization analysis results were screened using Bayesian model averaging. The results were validated in a multicenter cohort of 1368 patients, and the role of the antioxidant properties of high-density lipoprotein in DEACMP was examined. Mendelian randomization analysis identified six high-density lipoprotein-related variants significantly associated with DEACMP, with the cholesterol to total lipids ratio in medium high-density lipoprotein showing the strongest effect (marginal inclusion probability = 0.51, p = 1.00 × 10, false discovery rate = 6.00 × 10). Clinical validation confirmed HDL-C as an independent protective factor. Patients without DEACMP had higher high-density lipoprotein oxidant index values (1.23 [interquartile range: 1.02-1.36]) than those who developed DEACMP (0.84 [interquartile range: 0.66-0.90]); the high-density lipoprotein oxidant index declined significantly in postmenopausal women (p = 0.023). These findings demonstrate that HDL-C mitigates the risk of DEACMP through its antioxidant capacity. The integration of genetic evidence, clinical validation, and functional assays provides robust support for HDL-C as a predictive biomarker of neural recovery after carbon monoxide poisoning.
Integrated assessment of the effects of PFOA exposure on hepatic transcriptome and lipid profiles in mice expressing human PPARα
Nielsen G, Reed E, Lara B, Sherr DH, Heiger-Bernays WJ, Hyötyläinen T, Webster TF and Schlezinger JJ
Per- and polyfluoroalkyl substances (PFAS) are a family of persistent chemicals that continue to be released pervasively into the environment, leading to widespread human exposure. Emerging epidemiological evidence shows adverse effects on liver lipids; however, past toxicological studies have been limited by a focus on peroxisome proliferator activated receptor α (PPARα) driven effects on triglycerides in rodent systems. Here, we use a more agonostic approach incorporating lipidomics and transcriptomics to test the hypothesis that activation of human PPARα by perfluorooctanoic acid (PFOA), disrupts liver lipid homeostasis, broadly, similar to that seen in human liver diseases. Female and male mice expressing human PPARα or that were PPARα null were fed a What We Eat In America diet and exposed to PFOA via drinking water for 6 weeks. Serum PFOA concentrations averaged 48 ± 9 μg/mL. PFOA changed the expression of ∼2000 hepatic genes with changes in expression of a larger number of genes in hPPARα versus PPARα null mice. In this occupational level PFOA exposure scenario, less than 60 % of transcriptional changes induced by PFOA depended on hPPARα expression. CAR was another major molecular initiating event, with other transcription factors pathways more likely to be modulated downstream of hPPARα activation. In hPPARα mice of both sexes, PFOA increased total liver lipids. In addition to triacylglycerides, lipid classes strongly altered by PFOA exposure predominantly belong to the phosphatidylcholine and sphingolipid classes. PFOA significantly decreased sphingomyelin abundance and increased ceramide abundance regardless of genotype, which coincided with an increase in expression of SMase, the enzyme that converts sphingomyelin to ceramide. These results highlight the ability of PFOA to modulate liver lipids beyond triacylglycerides in both an hPPARα-dependent and -independent manner.
Drug screening reveals the mechanism of toyocamycin-induced apoptosis in triple-negative breast cancer organoids
Ma H, Lu L, Tu Y, Chang X, Jiang H, Yang T, Wang S, Wang J, Xue J, Chen Y and Luo G
Triple-negative breast cancer (TNBC) is characterized by high aggressiveness and molecular heterogeneity, limiting therapeutic efficacy and drug resistance, necessitating reliable preclinical models and novel therapeutic agents. This study utilized tumor tissues derived from breast cancer patients of various molecular subtypes, with a particular focus on TNBC, to construct patient-derived organoid models (PDOs). These models effectively recapitulate the in vivo characteristics of tumors and provide a cost-effective platform for high-throughput drug screening. The study employed a label-free in vitro drug screening system based on bright-field imaging, which continuously monitors changes in organoid area and brightness to assess the drug responses of 505 compounds. This approach avoids the interference associated with traditional cell viability assay reagents. Screening of the natural compound library using this system revealed that Toyocamycin effectively inhibits the growth of two TNBC organoid models, exhibiting significant dose-dependency. Further mechanistic studies demonstrated that Toyocamycin induces apoptosis in TNBC organoids by activating the p38 MAPK signaling pathway, specifically manifested by the upregulation of key genes such as TNFR, MAP3K7, MAP2K3, and DDIT3. It initially triggers cytotoxicity to suppress proliferation and subsequently induces sustained apoptosis. This process can be reversed by the p38 inhibitor Adezmapimod, further confirming that its apoptosis-inducing effect is dependent on the p38 MAPK pathway. This study not only validates the reliability of patient-derived organoids in personalized drug screening but also uncovers the potential therapeutic value of Toyocamycin for TNBC, providing a novel model and theoretical foundation for the precision treatment of TNBC.
Nanocapsule-based vitamin B12 as a novel strategy against vancomycin-induced nephrotoxicity: Targeting oxidative stress, ER stress, inflammation, and fibrosis in rats
El-Emam MMA, Kilany MS, Ibrahim M, Qelliny MR, Mostafa M, El Sayed MM, Khamis T, Agaga RAA, Eltaweel AM and Mansour MF
Vancomycin (VCM) is an essential glycopeptide antibiotic employed for treating methicillin-resistant Staphylococcus infections. However, its clinical use is limited by nephrotoxicity. Vitamin B12 (Vit B12) possesses antioxidant, anti-inflammatory, and anti-fibrotic properties that may protect against nephrotoxicity. However, Vitamin B12 bioavailability is inherently low. Therefore, nanotechnology-based approaches have been employed to overcome these limitations. Our research examined the formulation and preclinical assessment of Vitamin B12 nanocapsules (Vit B12 NC) against VCM-induced oxidative and, endoplasmic reticulum (ER) stress, inflammation, and fibrosis in rats. Nephrotoxicity was induced by administering VCM, followed by treatment with oral Vit B12 NC. Renal function, oxidative and ER stress markers, inflammatory cytokines, fibrosis markers, and histopathological changes in kidney tissue were evaluated. Vit B12 NC treatment reduced serum creatinine, uric acid, and urea levels, raised antioxidant enzyme activities (catalase and total antioxidant capacity), and decreased malondialdehyde (MDA) levels. It also downregulated ER stress markers, including inositol-requiring enzyme 1 (IRE1), TNF receptor-associated factor 2 (TRAF2), c-Jun N-terminal kinase (JNK), and C/EBP homologous protein (CHOP). Inflammatory mediators such as Toll-like receptor 4 (TLR4), interleukin-17 (IL-17), and interleukin-18 (IL-18) were also repressed. Also, it reduced renal fibrosis as indicated by decreased expression of VIM, miR-382-5p, and miR-92a-3p. Furthermore, it reversed the histopathological alterations in renal tissues. These findings suggest that Vit B12 NC exhibits promising nephroprotective potential against VCM-induced nephrotoxicity.
Corrigendum to "Arsenic exposure affects Pdgfrα stromal cells in the ileum of the small intestine" [Toxicology and Applied Pharmacology Volume 505, December 2025, 117582]
Ventrello SW, Lea KA and Bain LJ
Ditrioxzin synergizes with 2-deoxy-d-glucose to induce redox-dependent metabolic crisis in gastric cancer through dual targeting of peroxiredoxin 3 and glycolysis
Su N, Pan L, Shi X, Liu Y, Fan X, Liu S and Ma YC
Emerging evidence demonstrates that dual inhibition of glycolysis and mitochondrial function represents a potent anticancer strategy. Here, we report that Ditrioxzin (DTO), a synthetic ent-kaurane diterpenoid analog, selectively disrupts mitochondrial redox homeostasis by targeting peroxiredoxin 3 (Prx3) to induce hydrogen peroxide (HO) accumulation, thereby depolarizing mitochondrial membrane potential (MMP) and impairing oxidative phosphorylation (OXPHOS) in gastric cancer cells. DTO synergized with the glycolytic inhibitor 2-deoxy-d-glucose (2-DG) to deplete ATP through dual metabolic blockade. In vitro studies revealed that DTO exerted selective cytotoxicity against gastric cancer cells (IC 3.82-6.10 μM) but spared normal gastric epithelial cells (GES-1). Mechanistically, DTO directly bound Prx3, elevating HO levels (>3-fold at 8 μM), oxidizing mitochondrial peroxiredoxins, and triggering redox-dependent mitochondrial dysfunction. Combined DTO/2-DG treatment promoted ATP depletion and apoptosis (69.6 % vs 24.1 % DTO alone) via ROS-dependent pathways, an effect abrogated by N-acetylcysteine. In vivo, DTO (10 mg/kg) and 2-DG (500 mg/kg) synergistically suppressed tumor growth (66 %, P < 0.001) in xenograft models without body weight loss or histopathological changes in kidney/heart. Our findings establish DTO as a novel Prx3-targeted agent that synergizes with 2-DG to induce metabolic crisis, providing a high-safety-profile therapeutic strategy for gastric cancer.
DEHP induces hepatic fibrosis through STAT3-SLC7A11-ROS axis
Liang S, Xu Z, You X, Yuan T, He J, Mao L, Jin A, Zhou X, Yi B, Li M, Tu Q and Xu B
Di-(2-ethylhexyl) phthalate (DEHP) is a pervasive environmental contaminant commonly found in daily life, and numerous studies have linked it to the progression of liver diseases and organ fibrosis. However, the precise molecular mechanisms by which DEHP induces liver fibrosis remain incompletely understood. This study aimed to investigate the effects of DEHP on liver fibrosis and its underlying mechanisms. We observed that 100 μM DEHP and 6 days of exposure significantly induced activated human hepatic stellate cells (HSCs) and upregulated the fibrosis marker α-smooth muscle actin (α-SMA). By integrating DEHP and liver fibrosis-related target information from various databases, followed by an assessment of the protein-protein interactome (PPI) network and corresponding functional pathway mapping, we predicted that DEHP-induced liver fibrosis is closely associated with the accumulation of reactive oxygen species (ROS). Molecular docking experiments revealed that DEHP spontaneously binds to STAT3, with GLU-638 identified as a critical amino acid residue for this interaction. Further functional experiments confirmed that DEHP promotes ROS accumulation by downregulating SLC7A11 expression, a process mediated by STAT3. In summary, DEHP facilitates intracellular ROS accumulation by mediating the STAT3-SLC7A11-ROS signaling axis, thereby triggering the formation of liver fibrosis. This research provides novel molecular targets and therapeutic strategies for the future prevention and treatment of liver fibrosis.
Maternal ozone exposure and differential cardiometabolic outcomes in aging male and female offspring
Yoo B, Dye JA, Hazari MS, Farraj AK, Stewart EJ, Thompson LC, Nguyen HH, Snow SJ, Fiamingo M, Fisher AA, O'Shaughnessy KL and Miller CN
Fetal development is a vulnerable life stage for exposure to environmental stressors. Gestational exposure to ozone (O) has been shown to compromise fetal growth, predisposing offspring to increased risk of pulmonary and metabolic dysfunction later in life. However, the cardiovascular consequences of maternal O exposure on offspring health remain uncharacterized. Herein, pregnant Long-Evans rats were exposed to 0.8 ppm O for 4 h each day on gestation day (GD) 5 and 6. Following weaning, male and female offspring were monitored for cardiometabolic health for up to ∼5 months of age. Although there were little-to-no changes in most metabolic endpoints (e.g., growth, food intake, body composition, or glucose tolerance), offspring from O exposed dams had an altered respiratory exchange ratio at ∼4 months old that differed by sex. Furthermore, female offspring had adipocyte hyperplasia in the retroperitoneal depot, effects that were not evident in male offspring. Males from O-exposed dams had altered cardiac structure and function, including left ventricular wall thickening and increased ejection fraction and fractional shortening. Females from O-exposed dams, on the other hand, had decreased myocardial performance attributed to shortened aortic ejection. RNAseq on hearts from GD 21 fetuses revealed sexually dimorphic effects of maternal O exposure on cardiac gene expression, consistent with altered structure and function that was present in adulthood. Collectively, these findings demonstrate that peri-implantation O exposure increases the risk of multiple adverse effects, including cardiac dysfunction, in adulthood.
In Silico, in vitro, and in vivo studies of a 2-substituted quinazolin-4(3H)-one in T-cell acute lymphoblastic leukemia
Antoniolli G, Franchi Junior GC, Lima K, de Mello Lopes R, Barbosa EG, Machado-Neto JA, Lima CSP, Rodrigues T and Coelho F
This study reports the synthesis, physicochemical characterization, and preliminary pharmacological evaluation of a novel 2-substituted quinazolin-4(3H)-one, Qona11. The compound was synthesized from 2-aminobenzamide and 1H-benzo[d]imidazole-2-carbaldehyde in dimethyl sulfoxide with a 55 % yield, in a catalyst-free, atom-efficient process that adheres to Green Chemistry principles. Structural confirmation was achieved through IR (1667 cm, carbonyl), H NMR (13.50 and 12.50 ppm, NH protons), and C NMR (161.17 ppm, carbonyl carbon). In silico analysis suggested Qona11 possesses favorable oral bioavailability, high intestinal absorption, limited CYP450 inhibition, and predicted blood-brain barrier permeability. Toxicity predictions highlighted potential hepatotoxicity, neurotoxicity, and respiratory toxicity, while no significant risks for cardiotoxicity, carcinogenicity, immunotoxicity, or cytotoxicity were found. Comparative analysis with idelalisib revealed similar toxicity profiles to Qona11, distinct from vincristine. Biological evaluation in acute leukemia models demonstrated concentration- and time-dependent cytotoxicity, with Jurkat T-ALL cells being more sensitive (IC 2.3 μM) than NB4 APL cells (IC 12.7 μM). Flow cytometry confirmed apoptosis induction in Jurkat cells via mitochondrial permeabilization and caspase 3 activation. In vivo studies in NOD/SCID mice bearing Jurkat xenografts showed that Qona11 (100 mg.kg) was well tolerated with no systemic toxicity, although it did not inhibit leukemia cell proliferation in immune-independent models. Overall, Qona11 exhibits promising anticancer activity and low systemic toxicity, warranting further preclinical investigation in solid tumor models and combination therapies.
Rupestonic acid targets ENO1 to exert antitumor activity and synergizes with paclitaxel in hepatocellular carcinoma
Mulati S, Maimaitiming M, Ma J, Zhang M, Jiang R, Aibai S and Tuerxun X
Rupestonic acid, a sesquiterpene, has protective properties against liver damage, inflammation, and tumor formation. Despite these known effects, its specific role and mechanism of action in combating hepatocellular carcinoma (HCC) remain insufficiently understood. This study aimed to investigate the anti-HCC effects of rupestonic acid and to identify its potential molecular targets. We employed cell counting kit-8 (CCK-8), colony formation, and flow cytometry assays to assess its impact on cell viability, proliferation, and apoptosis in HCC cell lines. Additionally, target fishing, cellular thermal shift assays (CETSA), ribonucleic acid interference, and Western blot (WB) were employed to identify rupestonic acid's protein targets in HCC cells. Our results showed that rupestonic acid significantly inhibited HCC cell proliferation, induced G0/G1 phase cell cycle arrest, and promoted apoptosis through the mitochondrial pathway. Target engagement studies employing an alkyne-rupestonic acid probe combined with mass spectrometry identified enolase 1 (ENO1) as a direct binding target, with CETSA confirming its destabilization. Furthermore, rupestonic acid inhibited the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/Forkhead box protein O (FOXO) signaling pathway, and rupestonic acid demonstrated a synergistic cytotoxic effect with paclitaxel (PTX). These findings suggest that rupestonic acid is a promising candidate for HCC treatment. They also underscore the potential of rupestonic acid in the design and development of lead compounds for HCC treatment and identify ENO1 as a viable therapeutic target.
Ghrelin-disrupting activity of arsenic and its relation to cardiometabolic diseases
Rossi KN, Abedin F, Mohanto NC, Ahmed B, Mia S, Uddin N, Rimi RK, Goni O, Sarder SJ, Hossain S, Islam M, Jubayar AM, Islam MS, Hossain S, Hoque MA, Sumi D, Saud ZA, Himeno S and Hossain K
Arsenic (As) exposure is linked to a special type of obesity without increasing body mass index. This obesity is accompanied by the reduction of skeletal muscle mass and elevation of insulin resistance (IR). Obesity and IR are the key risk factors for cardiometabolic diseases (CMDs). Ghrelin, a small peptide hormone, is linked to CMDs through multiple mechanisms. However, the ghrelin-disrupting activity of As and its implication in the promotion of CMDs has not yet been documented. Therefore, this study was designed to explore the association of As exposure with serum ghrelin levels and its relation to the risk of CMDs, particularly obesity, skeletal muscle mass reduction, and IR in the participants (n=421) selected from low- and high-As exposure rural areas in Bangladesh. The participants in high-exposure areas had a significantly lower median interquartile range of serum ghrelin levels than those in low-exposure area. Serum ghrelin levels of the participants were decreased with increasing concentrations of As in drinking water, hair, and nails. Ghrelin levels were inversely linked to obesity measures related to As exposure, including waist circumference, triceps skinfold thickness, and serum leptin levels. Decreased ghrelin levels were associated with the reduction of muscle mass measures, serum creatinine levels, and lean body mass. Ghrelin levels were decreased with increasing levels of insulin and IR as assessed by HOMA-IR. Furthermore, As-related HOMA-IR was significantly mediated by lowering the ghrelin levels. These findings collectively indicated that the ghrelin-disrupting activity of As might be involved in the pathophysiology of As-promoted CMDs.
Exposure to a PFAS mixture alters cholesterol lipoprotein subfractions and induces a foam cell-like aortic macrophage expression profile in hyperlipidemic LDLr mice
Roth K, Yang Z, Agarwal M, Gurdziel K and Petriello MC
Per- and polyfluoroalkyl substances (PFAS) have been associated with elevated cholesterol, a clinically-relevant risk factor for atherosclerosis. Macrophages are key mediators of atherosclerosis progression through their polarization to various subsets including inflammatory macrophages and foam cells. However, studies examining impacts of PFAS on macrophages in the context of atherosclerosis are lacking. Here, we investigate the impact of PFAS mixtures on cholesterol subfractions and transcriptional profiling of aortic macrophages during early atherosclerosis. Male low density lipoprotein receptor (Ldlr) deficient mice were fed an atherogenic diet and exposed via their drinking water to a mixture of 5 PFAS (i.e., PFOA, PFOS, PFNA, PFHxS, and GenX), each at a concentration of 2 mg/L, for 7 weeks. Circulating cholesterol subfractions and subclasses were analyzed, and aortic macrophages were isolated using immuno-magnetic beads for RNA-sequencing. Total circulating cholesterol was significantly elevated by 10 % following PFAS exposure which was predominately due to a 25 % increase in intermediate-density lipoprotein (IDL). The densest subfraction of low-density lipoprotein, LDL7, also increased by 206 %. RNA sequencing of aortic macrophages revealed PFAS downregulated 389 and upregulated 593 genes; many related to lipid metabolism and foam cell development. Specifically, expression of inflammatory mediators chemokine (C-X-C motif) ligand 2 (Cxcl2) and chemokine (C-X-C motif) ligand 17 (Cxcl17) were significantly increased due to PFAS (2.4 log2 fold change and 10.4 log2 FC respectively) and levels of lipid metabolism and transport genes fatty acid binding protein 4 (Fabp4) and fatty acid synthase (Fasn) were similarly increased (3 log2 FC and 5.2 log2 FC respectively). This work provides additional mechanistic information related to PFAS-mediated acceleration of atherosclerosis.
Andrographolide-induced PANoptosis underlies its multiple organ toxicity in mice
Lu N, Cai YW, Cai QH, Liang XW, Sun N, Chan OK, Shi ZJ, Hu B, He XH, Zha QB and Ouyang DY
Andrographolide (Andro), the major bioactive component of Andrographis paniculata, exhibits potent anti-inflammatory properties but has raised safety concerns due to reported organ toxicity. This study aimed to investigate the mechanisms underlying Andro's in vitro and in vivo toxicity. In mice, single dose (≤100 mg/kg) Andro administration showed no acute toxicity, with no overt histopathological organ injury. But repeated administration of the same dose of Andro triggered damage in lung, liver, uterus, and kidney, characterized by pulmonary alveolar disruption, renal tubular edema, and elevated serum aspartate aminotransferase (AST)/alanine aminotransferase (ALT). Concurrent with systemic injury, PANoptosis was induced by Andro in these organs, as evidenced by the activation of caspase-1/-8/-3 (apoptosis), gasdermin D/E (GSDMD/E, pyroptosis), and MLKL (necroptosis), indicating the correlation between PANoptosis and organ toxicity. In vitro, Andro caused lytic cell death in macrophages and other cells in a time- and dose-dependent manner. During this process, Andro induced rapid activation of caspase-8, followed by caspase-1/-3 and GSDME cleavage and phosphorylation of MLKL (p-MLKL), indicative of the activation of the PANoptosis signaling pathway. Consistent with this, Andro induced lytic cell death was markedly attenuated by caspase-1 inhibitor VX-765, pan-caspase inhibitors (IDN-6556, Z-VAD-FMK) and GSDMD/E inhibitor (disulfiram). In addition, RIPK1 inhibition (by Nec-1) partially reduced cell death, confirming RIPK1-dependent necroptosis as a minor contributor. In conclusion, our data establish PANoptosis as an important mechanism of Andro-induced organ injury, providing a mechanistic framework for Andro's dichotomous bioactivity, informing evidence-based dosing strategies to maximize therapeutic efficacy while mitigating toxicity risks in clinical practice.
Targeting sonic hedgehog (shh) signaling pathways by the concentration-dependent topical resveratrol for protection from cyclophosphamide-induced alopecia in a mouse model
El-Din AA, Tahoon DM, Abd Elmaaboud MA, Abd Elmoniem FF and Abdin AA
Chemotherapy-induced alopecia (CIA) is a common and inevitable side effect of systemic cancer treatment. There is an urgent need for novel therapies for cancer patients suffering from hair loss to improve their quality of life. This study aimed to investigate the potential protective effect of concentration-dependent topical resveratrol on hair follicles via targeting sonic hedgehog (Shh) signaling and its related downstream regulatory (Sirt-1), proliferative (Ki-67), and apoptotic status (caspase-3 and Bcl-2) pathways in cyclophosphamide-induced alopecia in female C57BL/6 mice model.
Duloxetine-induced inhibition of voltage-gated K 3.1 (Kv3.1) channels and underlying electrophysiological mechanisms
An JR, Jeong J, Kim HR, Lee S, Sultana A, Das R, Woo J, Choi SW, Bae YM, Lim Y, Park H, Seo MS and Park WS
Duloxetine is a serotonin-norepinephrine reuptake inhibitor that has been widely used to treat major depression; however, it has also been associated with severe neuropsychiatric side effects, including hallucinations, confusion, and suicide attempts. Nevertheless, the electrophysiological mechanisms underlying these adverse effects remain poorly understood. In this study, we investigated the effect of duloxetine on cloned neuronal rat voltage-gated K (Kv) channel subunit Kv3.1, stably expressed in Chinese hamster ovary (CHO) cells. Duloxetine inhibited the Kv3.1 current in a concentration-dependent manner with a half-maximal inhibitory concentration (IC) of 2.04 ± 0.27 μM (approximately 5-fold higher than the peak therapeutic plasma concentration of 0.4 μM) and a Hill coefficient of 0.94 ± 0.08. This inhibitory effect was associated with accelerated current inactivation. The association and dissociation rate constants for duloxetine were 43.43 ± 4.57 μM·s and 122.12 ± 68.2 s, respectively. In addition, duloxetine shifted the voltage dependence of Kv3.1 steady-state inactivation toward a more negative direction and led to use-dependent inhibition upon repetitive stimulation (1 and 2 Hz). Duloxetine also slowed recovery from inactivation. Docking analysis predicted that duloxetine binds to the central pore and interface between the voltage-sensing and pore domains on Kv3.1 channel, supporting the inhibitory mechanisms of duloxetine. Furthermore, duloxetine inhibited Kv3.1-mediated currents in SH-SY5Y human neuroblastoma cells. Taken together, our results indicate that duloxetine inhibits Kv3.1 expressed in CHO cells in concentration-, time-, and use (open and inactivated states)-dependent manners, independently of its anti-depressive effects.
Gastrodin alleviates alcohol-induced developmental and neurotoxic effects in zebrafish larvae by suppressing ferroptosis via regulating the Nrf2/GPX4 signaling pathway
Li R, Yang W, Zheng L, Yan X, Liu C, Zhang Y and Li J
Prenatal alcohol exposure is a leading cause of developmental abnormalities and neurobehavioral deficits, collectively known as fetal alcohol spectrum disorder (FASD). The underlying molecular mechanisms, however, are not fully elucidated, hindering the development of effective therapeutic strategies. Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a key pathological process in various diseases. Gastrodin (GAS), the primary bioactive component of Gastrodia elata, has demonstrated potent antioxidant and neuroprotective properties. This study aimed to investigate the protective effects of GAS against alcohol-induced developmental and neurotoxic damage and to elucidate the underlying molecular mechanisms. Using a zebrafish larval model, we found that exposure to 200 mM alcohol from 2 to 24 h post-fertilization (hpf) induced significant developmental toxicity, including a decreased hatching rate, body length and eye diameter, and increased morphological malformations in larvae. Alcohol-exposed larvae also exhibited severe neurobehavioral deficits, characterized by a reduction in movement distance and average velocity in dark conditions. Mechanistically, alcohol exposure triggered ferroptosis, evidenced by an increase in intracellular Fe, malondialdehyde (MDA), and reactive oxygen species (ROS) levels, alongside a decrease in the levels of glutathione (GSH) and reduced glutathione peroxidase 4 (GPX4) and the nuclear factor erythroid 2-related factor 2 (Nrf2) activities. Co-treatment with GAS (200 mg/L) significantly ameliorated these alcohol-induced developmental and neurobehavioral defects. GAS administration effectively suppressed the hallmarks of ferroptosis by restoring the ROS level and altering the expression of genes related to oxidative stress. In addition, GAS suppressed alcohol-induced cell apoptosis, downregulated caspase3b, bax, caspase8, and upregulated bcl2 in mRNA levels. Molecular analysis revealed that GAS exerts its anti-ferroptotic effect by activating Nrf2/GPX4 signaling pathway, which was suppressed by alcohol. Our findings indicate that ferroptosis plays a key role in alcohol-induced developmental neurotoxicity, and GAS provides protection by activating the Nrf2/GPX4 axis. This suggests that GAS could be a potential therapeutic option for reducing the negative effects of prenatal alcohol exposure.
Evaluation of 2-isopropyl-N-2,3-trimethylbutyramide by a comprehensive toxicity study using gpt delta rats
Mitsumoto T, Ishii Y, Takimoto N, Takasu S, Namiki M, Toyoda T and Ogawa K
2-Isopropyl-N-2,3-trimethylbutyramide (ITB) is a food-flavoring agent classified as an aliphatic amide. In 2016, the Joint FAO/WHO Expert Committee on Food Additives evaluated ITB and concluded that additional data on toxicity and in vivo genotoxicity are required for its safety evaluation. In this study, we comprehensively investigated ITB toxicity using reporter gene transgenic animals. Male F344 gpt delta rats were administered ITB by oral gavage at doses of 0, 5, 50, or 500 mg/kg/day for 13 weeks. Neurological symptoms were observed in the early phase of treatment at doses ≥50 mg/kg. Periportal hepatocellular vacuolation was observed histopathologically at doses ≥50 mg/kg, along with increased liver weight and serum alanine aminotransferase levels. Kidney weight increased and serum chloride levels decreased at doses ≥5 mg/kg, indicating that ITB exerted potential nephrotoxic effects at lower doses. Accordingly, the lowest observed adverse effect level in the present study was at 5 mg/kg/day. No significant changes in gpt and red/gam mutant frequencies were detected in the liver or kidney, demonstrating a lack of ITB genotoxicity. Immunohistochemical analysis of GST-P-positive foci also suggested that ITB showed no hepatocarcinogenic potential. Overall, our findings demonstrate that ITB induces hepatic and renal toxicity but shows no evidence of in vivo genotoxicity or hepatocarcinogenic potential, providing essential information for safety assessment.
Corrigendum to "Elucidating the distinctive regulatory effects and mechanisms of active compounds in Salvia miltiorrhiza Bunge via network pharmacology: Unveiling their roles in the modulation of platelet activation and thrombus formation" [Toxicology and Applied Pharmacology volume 484, March 2024, 116871]
Zhang Y, Xin G, Zhou Q, Yu X, Feng L, Wen A, Zhang K, Wen T, Zhou X, Wu Q, He H and Huang W
Mechanistic insights into trimetazidine's protection against bladder ischemia-reperfusion injury via mirR-211/CHOP modulation and SIRT1/AMPK/PGC1α-mediated mitochondrial biogenesis
Alrashdi S, Mohamed SK, Elbaz M and El-Sayed EK
Bladder ischemia, frequently associated with vascular insufficiency, contributes to lower urinary tract symptoms via oxidative stress, inflammation, endoplasmic reticulum (ER) stress, mitochondrial defect, and apoptosis. Ischemia-reperfusion (I/R) injury exacerbates these effects by generating excessive reactive oxygen species. Trimetazidine (TMZ), an anti-ischemic agent, has shown protective effects in several I/R models; however, its role in bladder injury remains insufficiently characterized. This study investigated the protective effect of TMZ against bladder I/R injury in rats, focusing on oxidative stress, inflammation, ER stress, mitochondrial biogenesis, microRNA regulation, and apoptosis. Forty rats were allocated into four groups: sham control, I/R, and two TMZ-pretreated groups (10 or 20 mg/kg/day, p.o., for 14 days) prior to I/R induction. Controls received Tween 80 vehicle. Bladder tissues were collected for biochemical, molecular, and histopathological analyses. TMZ showed protection by lowering MDA (∼43.5-60.8 %) and enhancing GSH (∼2-2.6 fold) and SOD activity (∼2-3.2 fold). ER stress was attenuated, with reduced p-PERK (∼29.4-63 %) and CHOP (∼29.1-60 %), alongside upregulation of mirR-211 (∼1.4-1.9 fold). TMZ restored mitochondrial biogenesis through increased SIRT1 (∼1.9-2.4 fold), PGC1α (∼2.1-4.3 fold), p-AMPK (∼3-6.3 fold), and ATP (∼2-2.8 fold). It also downregulated pro-apoptotic (Bax, Caspase 3) and pro-inflammatory (TNF-α, IL-1β) mediators. Histopathology revealed marked preservation of bladder architecture, particularly at 20 mg/kg. TMZ exerts strong antioxidant, anti-inflammatory, anti-apoptotic, and cytoprotective effects in bladder I/R injury via modulation of oxidative stress, ER stress, mitochondrial pathways, and the mirR-211/CHOP axis. These findings suggest that TMZ may represent a promising therapeutic candidate for ischemia-associated bladder dysfunction, providing a mechanistic basis for future translational and clinical investigation.
miR-207 mitigates silica-induced pulmonary fibrosis by suppressing fibroblast-to-Myofibroblast transition via multi-target modulation of the TGF-β1/SMADs signaling pathways in mice
Du SL, Cui YY, Zhou X, Gong WL, Zhang ZQ and Han GZ
TGF-β1/SMADs signaling pathway plays a vital role in development of silicosis, with SMADs serving as the core transducers. Accordingly, any fluctuation in SMAD abundance can decisively steer the disease trajectory. Our previous research revealed miR-207 suppresses the progression of silicosis fibrosis by targeting Smad3. Further bioinformatic analysis suggested that miR-207 could also bind to the sequences of genes of Smad2 and Smad7, raising the possibility that miR-207 functions as a coordinated rheostat of multiple SMADs. However, the specific regulatory mechanisms of miR-207 in silicosis remains unexplored. In this study, a mouse model of silicosis was established by administering a silica suspension (20 μg/μL, 80 μL) via nasal drip daily for 16 days. On day 17, the silica-dusted mice were transfected with either miR-207 mimics or inhibitors. Lungs samples were harvested on day 45 for histological assessment of injury. Then, the expression levels of miR-207, Smad2, and Smad7 were determined using RT-qPCR, and the levels of SMAD2 and 7, Collagen I and III, and indicators of fibroblast-to-myofibroblast transdifferentiation (FMT) (α-SMA, FAP-1, and Vimentin) were determined using Western blot. The results showed that miR-207 coordinately downregulated SMAD2 and upregulated SMAD7 at both the mRNA and protein levels in silica-exposed mice, with concomitant reductions in FMT indicators (α-SMA, FAP-1 and Vimentin) and collagen levels. Therefore, we concluded that miR-207 suppresses silicosis progression in mice by inhibiting FMT via modulation of the TGF-β1/SMADs signaling pathway by targeting SMADs.
Zingiberensis new saponin reverses sorafenib resistance by targeting lncRNA TCONS-00026762/AKR1C1 and modulating autophagy and ferroptosis in hepatocellular carcinoma
Luo L, Zhou P and He K
Zingiberensis new saponin (ZnS) extracted from Dioscorea zingiberensis has antitumor activity. Our previous study found that Zns could exert anti- hepatocellular carcinoma (HCC) effects by regulating lncRNA TCONS-00026762. In addition, lncRNA TCONS-00026762 may act synergistically with AKR1C1. However, the relationship between them and their specific molecular mechanism underlying the anti-HCC effects of Zns has not been elucidated. This study aimed to investigate the role of TCONS-00026762/ AKR1C1 axis and ZnS in HCC cells from the perspective of autophagy, ferroptosis, and sorafenib resistance.