Ditrioxzin synergizes with 2-deoxy-d-glucose to induce redox-dependent metabolic crisis in gastric cancer through dual targeting of peroxiredoxin 3 and glycolysis
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.
Exploring the role of metformin in high fluoride ingestion-induced bone lesions
This study was to investigate the potential mechanisms of metformin administration against skeletal fluorosis by mitigating bone turnover. Rats were treated with fluoride via intragastric gavage to develop a model of skeletal fluorosis, and half of them were concurrently treated with metformin for 12 weeks. The cells involved in the process of bone turnover, including osteoclasts, osteoblast, and osteocyte were exposed to varying concentrations of fluoride with or without metformin. Results showed that excessive fluoride treatment increased levels of bone turnover markers in the serum and the expression of bone turnover-related factors in the femur. Additionally, fluoride treatment damaged the trabecular microstructure and mechanical function in long bone. Concurrent treatment of metformin reduced the serum levels of bone turnover markers and protein expression of bone turnover-related factors in the femurs of fluorotic rats. Furthermore, metformin cotreatment restored the trabecular microstructure of and mechanical function of the long bones of fluorotic rats. In vitro studies demonstrated that low-dose fluoride stimulated osteoclastic viability and upregulated osteoclastic differentiation proteins, but metformin inhibited the stimulatory effect of fluoride on them. Though fluoride exposure increased the apoptosis rate of osteoclasts and osteoblasts, and metformin aggravated fluoride-induced osteoclastic apoptosis but alleviated osteoblastic apoptosis. Additional, metformin stimulated SOST expression and inhibited RANKL expression in osteocytes exposed to fluoride. This is consistent with the KEGG enriched pathway in metformin-treated osteocytes, such as the osteoclast differentiation and Wnt signaling pathways. These results suggested that metformin counteracted high bone turnover that occurred in skeletal fluorosis by inhibiting osteoclastogenesis and regulating osteocytes to delay osteogenesis.
Aurocyanide, an active metabolite of auranofin, exerts cytoprotective effects via ROS-mediated Nrf2 activation in HepG2 cells
Auranofin, an FDA-approved oral gold(I) compound for rheumatoid arthritis, is being repurposed due to its established clinical safety and emerging therapeutic potential. Given its extensive in vivo metabolism, identifying active metabolites and characterizing their biological activities are critical for understanding its therapeutic mechanisms. Here, we evaluated the cytoprotective effects of seven potential auranofin metabolites against tert-butylhydroperoxide-induced oxidative injury in HepG2 cells. Among them, only aurocyanide markedly improved cell viability, as demonstrated by MTT and live-cell imaging assays. This study is the first to elucidate the molecular mechanisms underlying the cytoprotective effects of aurocyanide, with a focus on Nrf2 signaling. Aurocyanide suppressed tert-butylhydroperoxide-induced intracellular and mitochondrial ROS production and significantly increased antioxidant responsive element (ARE)-luciferase activity, comparable to that of sulforaphane. It also promoted nuclear accumulation of Nrf2, leading to upregulation of Nrf2-regulated antioxidant enzymes (GCL, HO-1, NQO1). Induction of Nrf2 target genes was confirmed in both HepG2 cells and primary mouse hepatocytes. Aurocyanide elevated intracellular GSH levels and alleviated tert-butylhydroperoxide-induced GSH depletion. Importantly, the cytoprotective effect of aurocyanide was significantly reduced in Nrf2-knockdown cells, highlighting the essential role of Nrf2. Pretreatment with N-acetyl-L-cysteine (NAC) abolished aurocyanide-induced Nrf2 activation and HO-1 upregulation, indicating the involvement of ROS in Nrf2/ARE pathway activation. Aurocyanide induced mild ROS production, which was reduced by NAC, and NAC also diminished the protective effects of aurocyanide against oxidative injury. In conclusion, aurocyanide, an active metabolite of auranofin, exerts antioxidant and hepatoprotective effects via ROS-mediated activation of the Nrf2 signaling pathway, broadening our understanding of its pharmacological actions in liver pathology.
Capsaicin alleviates DEHP-induced testicular dysfunction by suppressing oxidative stress in mice
Di(2-ethylhexyl) phthalate (DEHP) is a widespread environmental endocrine disruptor known to impair testicular function. Capsaicin, the bioactive compound in chili peppers, has not been thoroughly explored for its protective effects against DEHP-induced testicular damage. In this study, male C57BL/6 mice were divided into control, DEHP-exposed (200 mg/kg/day orally for 6 weeks), and DEHP-exposed with varying doses of capsaicin (5, 10, and 20 mg/kg/day orally for 6 weeks). Leydig and Sertoli cells were cultured in vitro with DEHP or capsaicin (0, 25, 50, and 100 μM). Chronic DEHP exposure impaired testicular development, leading to morphological abnormalities and reduced sperm quality, which were largely restored by capsaicin treatment. Both in vivo and in vitro assays revealed that capsaicin modulated the BAX/BCL2 ratio and inhibited testicular apoptosis. Additionally, capsaicin restored DEHP-induced suppression of testosterone biosynthesis in Leydig cells and maintained the integrity of the blood-testis barrier in Sertoli cells. Mechanistically, these protective effects were likely due to the antioxidant properties of capsaicin. In conclusion, our findings suggest that capsaicin may serve as a promising therapeutic agent for mitigating DEHP-induced testicular dysfunction, offering valuable insights for potential clinical applications.
Matrine triggers cardiotoxicity via apoptosis induced by reduction of GSH synthesis through the ATF4/CTH pathway
Matrine (MT), an alkaloid extracted from Sophora flavescens, is widely used in traditional Chinese medicine. However, its clinical application is limited by its toxic effects. In this study, we explored the function of the activating transcription factor 4 /cystathionine γ-lyase (ATF4/CTH) pathway in MT-induced cardiac injury. Our results indicated that MT significantly decreased cysteine (Cys) and glutathione (GSH) levels, leading to mitochondrial dysfunction and apoptosis characterised by ROS accumulation, mPTP opening, ΔΨm disruption, and caspase-9/3 activation. However, caspase-3 inhibition and GSH supplementation alleviated these toxic effects. NAC similarly protected against MT-induced apoptosis by providing Cys as a substrate for GSH synthesis, whereas BSO abolished this protection by inhibiting GSH biosynthesis. Additionally, MT suppressed both ATF4 expression and its transcriptional activity, leading to reduced mRNA and protein levels of the downstream target gene CTH, and consequently diminished CTH-dependent Cys production. Overexpression of either ATF4 or CTH attenuated MT-induced apoptosis by restoring intracellular Cys and GSH levels. CTH knock-down completely abrogated the protective effects of ATF4 overexpression, whereas CTH overexpression retained its efficacy, even in the absence of ATF4. These results indicated that MT-induced suppression of the ATF4/CTH pathway causes GSH deficiency, leading to mitochondria-dependent apoptosis and cardiotoxicity.
Zingiberensis new saponin reverses sorafenib resistance by targeting lncRNA TCONS-00026762/AKR1C1 and modulating autophagy and ferroptosis in hepatocellular carcinoma
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.
mTOR mediates airway epithelial E-cadherin disruption in toluene diisocyanate-induced asthma
E-cadherin is a critical adheren junctional protein for maintaining airway epithelial integrity. Downregulation of E-cadherin is commonly seen in asthma. Mammalian target of rapamycin (mTOR), a central regulator of metabolism, is implicated in asthma pathogenesis. This study was aimed to elucidate the role of mTOR signaling pathway on airway epithelial E-cadherin dysfunction in toluene diisocyanate (TDI)-induced asthma. Male BALB/c mice and in vitro cultured airway epithelial cell line BEAS-2B were exposed to TDI for modeling, and treated with rapamycin, an inhibitor of the mTOR signaling. We observed increased phosphorylation of mTOR and its downstream molecule p70s6k in TDI-exposed mice and cultured epithelia, indicating activation of mTOR signaling. In vivo, treatment with rapamycin dramatically alleviated TDI-induced airway hyperreactivity, decreased airway neutrophilia and eosinophilia, and suppressed the release of IL-4, IL-5 and IL-17 in the bronchoalveolar lavage fluid (BALF), suggesting a central role for mTOR in the development of TDI-induced asthma. Moreover, the TDI-induced downregulated E-cadherin expression in the lung was also significantly recovered by rapamycin, accompanied by less production of soluble E-cadherin (sE-cadherin), which is a marker of E-cadherin disruption and epithelial injury. Similar results were observed in cultured airway epithelial cells. Taken together, our data demonstrated that mTOR mediates airway epithelial E-cadherin disruption in TDI-induced asthma.
Integrative network toxicology and experimental validation reveal novel molecular mediators of omeprazole-induced nephrotoxicity
Omeprazole, a widely used proton pump inhibitor, has been increasingly linked to adverse renal outcomes such as acute kidney injury (AKI) and chronic kidney disease (CKD), yet its nephrotoxic mechanisms remain unclear. In this study, we utilized an integrative network toxicology approach, combined with molecular docking and in vitro validation, to elucidate potential mechanisms underlying omeprazole-induced renal toxicity. By intersecting omeprazole-related genes with those implicated in AKI and CKD, we identified 73 overlapping targets enriched in the PI3K-Akt signaling pathway, EGFR inhibitor resistance, and xenobiotic response. A protein-protein interaction network and topological analysis revealed six hub targets (CASP3, CCND1, EGFR, MMP9, PARP1, PPARG), all showing strong binding affinities to omeprazole in molecular docking simulations. Functional validation in HK-2 cells demonstrated that omeprazole reduced cell viability, induced morphological damage, and increased apoptosis in both acute (300 μM, 24 h) and chronic (20 μM, 7 days) exposure models. qPCR analysis confirmed significant upregulation of CASP3, EGFR, MMP9, PARP1, and PPARG, with CCND1 exhibiting model-dependent expression changes. These findings provide new insights into the molecular basis of omeprazole-associated nephrotoxicity and suggest potential biomarkers and therapeutic targets for mitigating renal injury.
Nomilin ameliorates perfluorooctanoic acid (PFOA)-induced impairment of zebrafish (Danio rerio) ocular development and visual function through PIK3CA activation
Perfluorooctanoic acid (PFOA), a pervasive environmental contaminant, is implicated in ocular diseases through prenatal/embryonic exposure. This study investigated the protective effects of nomilin, a citrus-derived bioactive compound with therapeutic properties, against PFOA-induced ocular and visual impairments in zebrafish. Integrating network toxicology and molecular docking, we identified seven shared targets (including PIK3CA and mTOR) linking PFOA, nomilin, and ocular diseases. Experimental results demonstrated that PFOA exposure suppressed the PIK3CA/AKT/mTOR/pax6 axis, significantly downregulated (by approximately 0.23- to 0.65-fold) ocular development genes (rx1, vsx1, rpgra, lhx4), and induced structural defects (reduced eye size, lens diameter and retinal layer thickness) and visual dysfunction. Nomilin treatment dose-dependently reversed these effects by activating the PIK3CA/AKT/mTOR/pax6 axis, restoring the expression of ocular developmental related genes by 1.45- to 2.85-fold compared to the PFOA-exposed group, improving ocular morphology, and enhancing visual response behaviors. Furthermore, nomilin attenuated PFOA-induced apoptosis. These findings reveal that nomilin mitigates PFOA-mediated ocular toxicity via PIK3CA activation, offering novel therapeutic insights for environmental pollutant-related ocular disorders.
Integrated assessment of the effects of PFOA exposure on hepatic transcriptome and lipid profiles in mice expressing human PPARα
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.
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
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.
Drug screening reveals the mechanism of toyocamycin-induced apoptosis in triple-negative breast cancer organoids
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
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.
High-density lipoprotein cholesterol protects against delayed encephalopathy after acute carbon monoxide poisoning
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.
Artesunate attenuates pulmonary fibrosis by suppressing fibroblast senescence through inhibition of the STAT3/p53 signaling pathway
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and lethal lung disease with elusive pathogenesis. Accumulating evidence implicates cellular senescence as a key mechanism in IPF. Current therapies (pirfenidone/nintedanib) reduce acute exacerbation risk and delay lung function decline in mild-to-moderate IPF, but adverse effects often limit their use. The JAK/STAT pathway is critically involved in the pathogenesis of IPF. While artesunate (Art) alleviates pulmonary fibrosis in rats through anti-inflammatory actions, its effects on fibrosis via STAT3 inhibition-particularly in murine models and human tissues-remain underexplored.
The potential amelioration of oxaliplatin-induced peripheral neuropathy in Sprague Dawley rats by sodium butyrate via targeting neuro-immuno-inflammatory axis
Oxaliplatin is widely used as one of the first-line treatments for colorectal carcinoma. Oxaliplatin-induced peripheral neuropathy (OIPN) is the most common dose-limiting adverse effect leading to discontinuation of treatment. This work aimed to investigate the potential ameliorative effect of sodium Butyrate (NaB) on OIPN via assessment of neurotrophic factors and the immune-inflammatory axis, either before induction (PRE) or WITH the induction of OIPN in Sprague Dawley rats. In this study, OIPN was induced by oxaliplatin in a dose of 2.4 mg/kg once daily for 5 consecutive days/ week for 3 weeks by i.p. injection. Behavioral tests were performed, and the sciatic nerves were dissected and processed for further assessment of biochemical parameters and the histopathological picture. NaB has a marked neurotrophic, anti-inflammatory and immunomodulatory effect represented by a significant decrease in mechanical and cold allodynia scores and tissue levels of TNF-α and a significant increase in tissue levels of nerve growth factor (NGF) and IL-10 with a significant improvement in the histopathological changes by H&E and Toluidine blue as well as a significant increase in the IHC expression of FOXP-3 and PPAR-γ. NaB-PRE OIPN showed more improvement in the neurotrophic, anti-inflammatory, and immunomodulatory properties than NaB WITH OIPN. These elaborated data indicated that NaB is a potential neuroprotective candidate for OIPN treatment due to its immunomodulatory, anti-inflammatory, and neurotrophic effects, particularly when used before the initiation of chemotherapeutic cycles to improve the clinical aspects of OIPN.
Nitazoxanide reverses pulmonary vascular remodeling in pulmonary hypertension by targeting the IMPA1-RAGE signaling axis
Pulmonary hypertension (PH) is a life-threatening disorder characterized by excessive proliferation and migration of pulmonary artery smooth muscle cells (PASMCs), leading to pulmonary vascular remodeling, elevated pulmonary vascular resistance (PVR), and increased pulmonary artery pressure (PAP). These pathological changes ultimately induce right ventricular hypertrophy, right heart failure, and death. Current therapeutic approaches inadequately address the remodeling aspect of PH. Thus, novel therapeutic strategies targeting PASMCs proliferation and vascular remodeling are critically needed. Nitazoxanide, an FDA-approved antiparasitic agent with favorable safety and bioavailability, significantly reduced PAP and alleviated pulmonary vascular remodeling in experimental models of PH, including the SU5416/hypoxia and monocrotaline rat models. Utilizing drug affinity responsive target stability (DARTS), cellular thermal shift assay (CETSA), co-immunoprecipitation and Western blot analysis, we identified inositol monophosphatase 1 (IMPA1) as a novel direct molecular target of nitazoxanide. Mechanistically, nitazoxanide treatment inhibited the IMPA1-RAGE interaction, thereby suppressing downstream activation of the PI3K/Akt/mTOR signaling cascade and attenuating the enhanced glycolysis characteristic of PASMCs in PH. Collectively, our findings highlight nitazoxanide as a promising therapeutic candidate for pulmonary vascular remodeling and pulmonary hypertension.
Endometriosis lesion growth inhibition and immune modulation function of thymol in vitro and in vivo
Thymol, a phenolic monoterpene derivative found in the essential oil of Thymus vulgaris L., Lamiaceae, is currently under investigation for potential applications in pharmaceuticals, functional foods, and cosmetics. Despite ongoing research in these areas, the potential therapeutic effects of thymol on endometriosis remain unconfirmed. Endometriosis, characterized by the growth of endometrial tissue outside the uterus. Available treatment options, including medications and surgery, have limitations and are no curative. To address these limitations, our study aimed to explore the therapeutic potential of thymol in endometriosis, through simultaneous in vitro and in vivo investigations. Our results indicate that thymol induces antioxidant activity and regulate intracellular calcium ions. Further thymol administration reduced the size of endometriosis lesions in animal models and modulated immune responses, resulting in alterations in spleen populations of CD4 T and CD8 T cells and macrophages, as well as changes in the expression of cytokine-related genes. Overall, our findings suggest that thymol holds promise as a therapeutic agent for endometriosis.
circSHOC1-SLC25A3 promotes 2-naphthylamine-induced DNA damage in bronchial epithelial cells via activation of oxidative stress
Smoking, a pivotal environmental risk factor, drives diseases including lung cancer through genetic and epigenetic alterations. While 2-Naphthylamine (2-NA), a tobacco-derived carcinogen, is established as a bladder carcinogen via DNA damage, its role in lung carcinogenesis remains mechanistically uncharacterized despite epidemiological associations. This study identifies bronchial epithelium as a direct target of 2-NA, demonstrating dose-dependent DNA damage in 16HBE cells (peak at 250 μM, 12 h; P < 0.05), accompanied by S-phase arrest, apoptosis, reduced proliferation, and reactive oxygen species (ROS) generation. 2-NA exposure upregulated the expression of circSHOC1, which is a circular RNA derived from the SHOC1 gene, in a dose-dependent manner. Functional assays revealed that circSHOC1 overexpression exacerbated 2-NA-induced DNA damage by enhancing ROS production and 8-hydroxy-2'-deoxyguanosine (8-OHdG) accumulation, whereas knockdown attenuated these effects. Mechanistically, circSHOC1 interacted with mitochondrial protein SLC25A3 (Solute Carrier Family 25 Member 3), a key oxidative stress regulator; SLC25A3 knockdown mitigated DNA damage (P < 0.01), and co-transfection experiments confirmed SLC25A3 as a critical mediator of circSHOC1-driven genotoxicity. Collectively, this work provides the first experimental evidence that 2-NA induces bronchial epithelial DNA damage via a circSHOC1-SLC25A3-ROS axis, supporting a novel mechanism for tobacco-associated lung carcinogenesis and highlighting 2-NA as a potential pulmonary carcinogen.
Paeoniflorin attenuates ASA-induced enteropathy in simulated high-altitude hypoxia in rats by regulating intestinal inflammation, intestinal barrier, and intestinal flora
This study aimed to investigate whether high-altitude hypoxia exacerbates non-steroidal anti-inflammatory drug (NSAID)-induced small intestinal injury and to evaluate the protective effects of Paeoniflorin (PF).
Activated autophagy drives the adaptive response to LPS-evoked lung pyroptosis in adolescent mice
The disease of lipopolysaccharide (LPS)-induced acute lung injury (ALI) is prevalent among clinical respiratory patients. The LPS adaptive response is a phenomenon whereby prior exposure to a low dose of LPS results in insensitivity to a subsequent high-dose challenge. However, the LPS adaptive response and its mechanism in mice with LPS-induced ALI remains limited. In this study, CD-1 male mice received a low-dose LPS (0.1 mg/kg) pretreatment for 24 h before high-dose LPS (5 mg/kg) challenge. Pretreatment with low-dose LPS mitigates the effects of high-dose LPS-induced lung pyroptosis and ALI. Low-dose LPS pretreatment also blocked high-dose LPS-induced activation of NLRC4 inflammasome in the lung. Interestingly, low-dose LPS pretreatment further increased the level of autophagy-related proteins in the lung with high-dose LPS treatment, suggesting that autophagy was further activated after low-dose LPS pretreatment. It is also important to note that 3-methyladenine is a specific inhibitor of autophagy, blocks the adaptive response to LPS-evoked pyroptosis and ALI. Inversely, rapamycin, an autophagy inducer, promotes adaptive response to LPS-evoked pyroptosis and ALI. Mechanistically, activated autophagy drives the adaptive response to LPS-evoked lung pyroptosis in mice via promoting p62-dependent degradation of NLRC4. Collectively, our results indicate that low-dose LPS pretreatment activates autophagy to degrade NLRC4 by p62 dependent manner, thereby protecting against pyroptosis and ALI induced by high-dose of LPS.
