TOXICOLOGY AND APPLIED PHARMACOLOGY

On the relationship between hERG inhibition and the magnitude of QTc prolongation: An in vitro to clinical translational analysis
Harmer AR and Rolf MG
Assessing the magnitude of QTc prolongation is crucial in drug development due to its association with Torsades de Pointes. Inhibition of the hERG channel, pivotal in cardiac repolarization, is a key factor in evaluating this risk. In this study, the relationship between hERG inhibition and QTc prolongation magnitude was investigated, with the aim to derive simple guidance on the required hERG margin to avoid a large (>20 ms) QTc prolongation.
Exploring the potential use of Caenorhabditis elegans as an animal model for evaluating chemical-induced intestinal dysfunction
Zhang XP, Ma X, Liu JL and Liu AL
Evaluating intestinal toxicity is crucial for identifying and preventing the harmful effects of environmental chemicals. Owing to the limitations of existing models in evaluating intestinal toxicity, the development of alternative models is urgently needed. This study explored the potential use of the nematode Caenorhabditis elegans as a model animal for assessing chemical-induced intestinal dysfunction. Changes in intestinal permeability and nutrient absorption in C. elegans individuals exposed to four intestine-disrupting chemicals (sodium dodecyl sulfate (SDS), dextran sulfate sodium (DSS), lipopolysaccharide (LPS) and ethanol) were examined using dye stain assays, an enzymatic photometric assay, and fluorescent probe uptake assays. Additionally, epigallocatechin-3-gallate (EGCG), an intestine-protecting phytochemical, was chosen to prevent ethanol-induced intestinal damage. The results indicated that SDS, DSS, LPS, and ethanol compromised the intestinal barrier in C. elegans. SDS had no effect on glucose absorption, but LPS, DSS, and ethanol inhibited or tended to inhibit glucose absorption. SDS, DSS, LPS, and ethanol reduced fatty acid absorption. LPS increased peptide absorption at a low dose but decreased it at a high dose; SDS, DSS, and ethanol attenuated peptide absorption. EGCG protected against the disruption of the intestinal barrier that was induced by ethanol treatment. These results suggest that C. elegans is a suitable surrogate model animal for evaluating chemical-induced intestinal dysfunction. These findings also provide new insights into the effects of SDS, DSS, LPS, and ethanol on intestinal function and highlight the potential of EGCG as a natural dietary intervention to protect individuals who use excess alcohol from intestinal injury.
Valifenalate-induced non-adverse thyroid changes via adaptive induction of uridine 5'-diphospho-glucuronosyltransferase (UGT) in the liver of dogs and rats but not humans
Walter C, Baze A, Grant C, Richert L and Bomann W
Some rat and dog toxicology studies with the fungicide valifenalate showed minimal, non-adverse thyroid changes, mostly above the maximum tolerated dose, and concomitantly with liver effects. This publication describes their mode of action (MOA), combining in vivo and new approach methodologies (NAMs), in a weight of evidence approach. Data demonstrate a MOA of liver enzyme induction via nuclear receptor CAR/PXR activation, increased thyroxine (T4) metabolism and elevated thyroid stimulating hormone (TSH) level, leading to thyroid follicular cell hypertrophy and increased thyroid weight. Non-human relevance of the MOA was demonstrated in in vitro cross species assays in rat, dog and human hepatocytes. Increased gene expression and activity of cytochrome P450s (CYPs) and uridine 5'-diphospho-glucuronosyltransferases (UGTs) were observed in rat and dog hepatocytes exposed to valifenalate, with increased T4 clearance and/or T4 glucuronidation/T4-UGT activity. Therefore, a causal relationship between increased liver enzyme induction and thyroid effects in dogs and rats is concluded. Rat hepatocytes were most sensitive, while valifenalate did not increase T4-UGT activity above 2-fold of vehicle control or T4 glucuronidation and T4 clearance in human hepatocytes. Consequently, valifenalate exposure in humans is unlikely to lead to decreased T4 levels, and subsequent thyroid and developmental neurotoxicity effects. Alternative human-relevant thyroid MOAs were excluded, i.e. inhibition of deiodinases (DIO), thyroperoxidase (TPO) or the sodium iodide symporter (NIS). Due to known species differences in thyroid homeostasis between humans and laboratory animals and, importantly, based on the presented data, this liver enzyme mediated MOA is considered not relevant for human hazard assessment.
Sildenafil citrate induces prostatic hyperplasia in BPH model rats and aged rats
Huang S, Huang D, Su X, Yang R, Shao C, Jiang J, Li J and Wu J
Erectile dysfunction (ED), a prevalent disease among middle-aged and elderly males, significantly impacts both patient and partner quality of life. Phosphodiesterase type 5 inhibitor (PDE5i) represents an effective therapeutic method for ED. Given their widespread global utilization, concerns arise regarding potential reproduction-related problems arising from clinical use. During the extensive development of PDE5i, we speculated that the potential of these inhibitors to variably induce prostatic hyperplasia, but this field remains unexplored. In order to verify the male reproductive toxicity of PDE5i, sildenafil citrate at doses of 5, 10 and 20 mg/kg was administered in BPH model rats and aged rats. Anatomical and pathological analyses indicate a compelling association between sildenafil citrate administration and the promotion of prostatic hyperplasia in both BPH model rats and aged rats. Serum analysis showed that serum prostate-binding protein (PBP) exhibited a non-significant but increasing trend following administration of sildenafil citrate to BPH model rats. Furthermore, significant increase in serum levels of E2 and T, as well as T in dorsal lobe prostate tissue of aged rats, were observed compared to the model control group. These results confirm the hypothesis that sildenafil citrate has reproductive toxicity in males.
Cepharanthine inhibits the proliferation of glioblastoma cells by blocking the autophagy-lysosomal pathway
Dong X, Zhu W and Wang N
Cepharanthine (CEP) is a Stephania cepharantha-derived bioactive alkaloid that can inhibit the progression of numerous tumors. However, the effects and specific mechanisms of CEP performance in glioblastoma (GBM) remain unclear. Thus, this study focused on exploring the effects of CEP on GBM and clarifying the underlying mechanisms. U251 and U87 cells were selected to estimate the anti-GBM effects of CEP, and the possible targets of CEP were analyzed using RNA sequencing (RNA-seq). Validation experiments based on RNA-seq data were performed to clarify the key pathway by which CEP mediates GBM cells response. Results showed that CEP administration successfully inhibited the proliferation and induced the cell cycle arrest and apoptosis of the GBM cells. RNA-seq analysis after CEP administration identified 386 differentially expressed genes, which were highly enriched in the autophagy-lysosomal pathway. Subsequent findings demonstrated that CEP exhibited the potential to curb GBM progression by causing lysosomal and autophagic dysfunction. Taken together, our results indicate that CEP is a potential drug candidate for GBM intervention.
Network toxicology and cell experiments reveal the mechanism of DEHP-induced diabetic nephropathy via Wnt signaling pathway
Hu XX, Yin YC, Xu P, Wei M and Zhang W
Di(2-ethylhexyl) phthalate (DEHP), a widely recognized endocrine disruptor, has been linked to the pathogenesis of diabetic nephropathy (DN) through its interference with hormonal and metabolic homeostasis. This study integrates network toxicology with cell-based assays to elucidate the molecular mechanisms of DEHP-induced DN, seeking to identify novel targets for toxicity assessment and therapeutic intervention. Through comprehensive screening across multiple toxicology and disease-related databases, six core genes (CTNNB1, EGFR, TNF, CCND1, BCL2, CASP3) were identified as shared mediators of DEHP exposure and DN. These genes are predominantly associated with the Wnt signaling pathway, a pivotal regulator of podocyte function, including cellular adhesion, differentiation, apoptosis, and inflammatory response. Mouse glomerular podocytes (MPC-5) exposed to graded concentrations of DEHP, with or without the Wnt pathway inhibitor XAV-939, displayed significant DEHP-induced disruptions: reduced cell adhesion, proliferation, and differentiation; increased autophagy, apoptosis, and migratory activity; elevated inflammatory mediator release; and pronounced activation of the Wnt signaling pathway, evidenced by upregulation of β-catenin, EGFR, TNF, CCND1, BCL2 and downregulation of CASP3. DEHP exposure further altered transcriptional activity and chromatin structure at key loci (CTNNB1, EGFR, and TNF). XAV-939 effectively mitigated these effects, underscoring the Wnt pathway's central role in DN progression under DEHP influence. These findings highlight the complex multi-target, multi-pathway interactions of DEHP in DN pathophysiology, offering deeper mechanistic insights and potential targets for therapeutic intervention against DEHP-induced nephrotoxicity.
Ginsenoside Rg3 attenuates the stemness of breast cancer stem cells by activating the hippo signaling pathway
Deng Z, Ou M, Shi Y, Li G and Lv L
Ginsenoside Rg3 (Rg3), a bioactive compound from ginseng, is gaining attention for its potential in targeting cancer stem cells in cancer therapy. The therapeutic effect of Rg3 on breast cancer stem cells (BCSCs) has not been systematically explored using a suitable approach. Our study leverages a multi-faceted strategy, including network pharmacology, molecular docking, and in vitro experiments validation, to explore the effect of Rg3 against BCSCs. We identified 38 common targets of Rg3 and BCSCs through public databases mining. The analysis of protein-protein interaction network revealed Myc, Stat3, Bcl2, Cdh1, Egf, Il6, Egfr, Nfkb1, Sox2 and Sirt1 as the top 10 potential targets. Molecular docking further validated Rg3 has robust binding potential with these targets. Utilizing the BCSC-enriched MCF-7 and MDA-MB-231 mammosphere model, in vitro experiments substantiated Rg3's ability to induce apoptosis, suppress proliferation, and inhibit mammospheres formation of BCSCs. Rg3 also decreased the ALDH and CD44/CD24 subpopulations and downregulated the expression of cancer stem cell markers such as c-MYC, ALDH1A1, NANOG in BCSCs. After Rg3 treatment, most of the top 10 genes in BCSC-enriched MCF-7 mammospheres showed a significant reduction in expression, with Cdh1 (E-cadherin) being the most markedly downregulated. The E-cadherin/catenin complex acts as an upstream regulator of the Hippo signaling pathway, which is crucial for BCSC function and is among the top 20 enriched pathways identified by KEGG analysis. Mechanistically, Rg3 attenuates the stemness of BCSCs by activating the Hippo signaling pathway. This study provides a comprehensive evaluation of Rg3 as a promising therapeutic agent against BCSCs.
Protein arginine methyltransferase 5 confers the resistance of triple-negative breast cancer to nanoparticle albumin-bound paclitaxel by enhancing autophagy through the dimethylation of ULK1
Kong J, Dong Y, Li M, Fan J and Wang T
Chemotherapy remains the major strategy for treating triple-negative breast cancer (TNBC); however, frequently acquired chemoresistance greatly limits the treatment outcomes. Protein arginine methyltransferase 5 (PRMT5), which modulates arginine methylation, is important in chemoresistance acquisition across various cancers. The function of PRMT5 in the development of chemoresistance in TNBC is still not well understood. This work focused on defining PRMT5's function in contributing to the chemoresistance in TNBC and demonstrating the possible mechanisms involved. Two TNBC cell lines resistant to nanoparticle albumin-bound paclitaxel (Nab-PTX), designated MDA-MB-231/R and MDA-MB-468/R, were developed. The expression of PRMT5 was markedly elevated in the cytoplasm of Nab-PTX-resistant cells accompanied with enhanced autophagy. The depletion of PRMT5 rendered these cells sensitive to Nab-PTX-evoked cytotoxicity. The autophagic flux was upregulated in Nab-PTX-resistant cells, which was markedly repressed by PRMT5 depletion. The dimethylation of ULK1 was markedly elevated in Nab-PTX-resistant cells, which was decreased by silencing PRMT5. Re-expression of PRMT5 in PRMT5-depleted cells restored the dimethylation and activation of ULK1 as well as the autophagic flux, while the catalytically-dead PRMT5 (R368A) mutant showed no significant effects. The depletion of PRMT5 rendered the subcutaneous tumors formed by Nab-PTX-resistant TNBC cells sensitive to Nab-PTX. The findings of this work illustrate that PRMT5 confers chemoresistance of TNBC by enhancing autophagy through dimethylation and the activation of ULK1, revealing a novel mechanism for understanding the acquisition of chemoresistance in TNBC. Targeting PRMT5 could be a viable approach for overcoming chemoresistance in the treatment of TNBC.
New insights into the mechanisms underlying 5-hydroxymethylfurfural-induced suppression of testosterone biosynthesis in vivo and in vitro
Orta Yilmaz B and Aydin Y
5-hydroxymethylfurfural (HMF), produced by the Maillard reaction, can indicate thermal processes in food. Previous research has examined the cytotoxic, genotoxic, mutagenic, and carcinogenic characteristics of HMF and its derivatives in different organs. Nevertheless, there is currently no available evidence about the impact of HMF on male reproductive toxicity. In this study, the effects of HMF on testosterone biosynthesis in both mouse testis and TM3 Leydig cells were investigated. HMF was administered to mice at doses of 30 and 300 mg/kg/day for 21 days and to Leydig cells at concentrations of 0.1, 1, and 10 mM for 24 h. The mechanism of action of HMF on testosterone biosynthesis in both mouse testis and Leydig cells was revealed by measuring the amount of testosterone, 3',5'-cyclic adenosine monophosphate (cAMP) levels, and the expression level of some important genes in the steroidogenic pathway. In addition, its effects on general testis were examined through histopathological evaluations. Upon examination of the results, it was observed that HMF had a significant impact on reducing testosterone and cAMP levels. Furthermore, HMF inhibited the expression of steroidogenic genes, including steroidogenic acute regulatory protein, cholesterol side-chain cleavage enzyme, 3β-hydroxy dehydrogenase, and 17β-hydroxy dehydrogenase, as well as transcription factors, such as steroidogenic factor-1, GATA binding protein-4, and nerve growth factor IB. HMF-administrated groups had germinal epithelium degradation, vacuolization, and disorders in the interstitial area. Consequently, it has been proven for the first time that HMF can damage the male reproductive system by detrimentally impacting the production of testosterone.
ROS responsive nanozyme loaded with STING silencing for the treatment of sepsis-induced acute lung injury
Zhang YJ, Chen LY, Lin F, Zhang X, Xiang HF and Rao Q
Acute lung injury (ALI) is a common complication of sepsis and a leading cause of mortality in septic patients. Studies indicate that STING may play a crucial role in the pathogenesis of sepsis-induced ALI by interacting with the PARP-1/NLRP3 pathway. Therefore, targeting STING inhibition has potential as a novel therapeutic strategy for ALI. However, effective inhibition remains challenging due to the widespread expression of STING across various tissues. In this study, we developed a nanozyme-based drug delivery system, DSPE-TK-mPEG-MnO@siSTING (abbreviated as DTmM@siSTING), using DSPE-TK-mPEG-MnO as the carrier, and characterized it via scanning electron microscopy, dynamic light scattering, nanoparticle size analysis, and gel electrophoresis. To evaluate the therapeutic effects of DTmM@siSTING, an in vitro ALI cell model and an in vivo ALI mouse model were established, assessing the nanozyme's impact on ROS levels, inflammatory responses, and the PARP-1/NLRP3 pathway in sepsis-induced ALI. Results demonstrated that DTmM@siSTING exhibited good physiological stability. In vitro, DTmM@siSTING significantly reduced ROS levels, myeloperoxidase activity, and expression of inflammatory cytokines, while also inhibiting PARP-1/NLRP3 pathway activation. In vivo experiments further revealed that DTmM@siSTING effectively delivered siSTING to the lungs, mitigating sepsis-induced ALI and associated inflammatory responses. Additionally, DTmM@siSTING displayed excellent biocompatibility. In summary, our findings suggest that DTmM@siSTING significantly enhances the therapeutic efficacy of siSTING, alleviating ALI by inhibiting ROS production, inflammatory responses, and activation of the PARP-1/NLRP3 pathway. This novel approach presents a promising therapeutic avenue for sepsis-induced ALI.
Inhibition of CDGSH iron‑sulfur domain 2 exhibits tumor-suppressing effects on diffuse large B-cell lymphoma (DLBCL) by inducing ferroptosis through the regulation of the NRF2/SLC7A11/GPX4 pathway
Zhang J, Wang T, Zhang H, Li N, Liu B and Fang L
CDGSH iron‑sulfur domain 2 (CISD2) is recognized as a ferroptosis-related gene that has potential as a target for cancer treatment. However, it is still uncertain whether targeting CISD2 can modulate ferroptosis in diffuse large B-cell lymphoma (DLBCL) cells and exhibit cancer-suppressing effects. The present study thoroughly investigated the role of CISD2 in DLBCL. CISD2 was found to be overexpressed in DLBCL, and its inhibition resulted in substantial growth inhibition in DLBCL cells. The growth inhibition effect resulting from CISD2 silencing could be reversed by a ferroptosis inhibitor, whereas inhibitors of apoptosis and necrosis did not yield the same reversal. CISD2-silenced DLBCL cells exhibited increased sensitivity to growth inhibition induced by ferroptosis suppressors. The inhibition of CISD2 induced ferroptotic cell death in DLBCL cells, which was supported by the overproduction of lipid peroxides, depletion of glutathione, accumulation of iron, and increased presence of shrunken mitochondria. Further investigation revealed reduced levels of NRF2, GPX4, and SLC7A11 in CISD2-silenced DLBCL cells. The overexpression of NRF2 significantly reduced the occurrence of ferroptotic cell death in DLBCL cells in which CISD2 was silenced. Furthermore, CISD2 inhibition exhibited tumor-suppressing effects in vivo associated with the induction of ferroptotic cell death in xenografts. These findings suggest that CISD2inhibition has tumor-suppressing effects on DLBCL by promoting ferroptotic cell death via the NRF2/SLC7A11/GPX4 pathway. Therefore, CISD2 holds promise as a viable candidate target for treating DLBCL.
Unraveling the intrinsic and photodynamic effects of aluminum chloride phthalocyanine on bioenergetics and oxidative state in rat liver mitochondria
Klosowski EM, de Souza BTL, Nanami LF, Bizerra PFV, Mito MS, Esquissato GNM, Constantin RP, Joia BM, Menezes PVMDC, Caetano W, Pereira PCS, Gonçalves RS, Garcia FP, Bidoia DL, Nakamura TU, Nakamura CV, Ishii-Iwamoto EL, Dos Santos WD, Ferrarese-Filho O, Marchiosi R and Constantin RP
Previous research has revealed that mitochondria are an important target for photodynamic therapy (PDT), which might be employed as a therapeutic approach for several malignancies, including hepatocellular carcinoma (HCC). In this study, we investigated both intrinsic toxicity and photodynamic effects of the photosensitizer (PS) aluminum chloride phthalocyanine (AlClPc) on mitochondrial functions. Several aspects of mitochondrial bioenergetics, structure, and oxidative state were investigated in the isolated mitochondria obtained from rat liver by differential centrifugation. Additionally, experiments were conducted to demonstrate the intrinsic and photodynamic effects of AlClPc on the viability of HepG2 cells. AlClPc interacted with mitochondria regardless of photostimulation; however, at the maximum utilized concentration (40 μM), photostimulation reduced its interaction with mitochondria. Although AlClPc hindered catalase (CAT) and glutathione reductase (GR) activities intrinsically, it had no discernable capacity to generate oxidative stress or impact bioenergetics in mitochondria without photostimulation, as one would anticipate from an ideal PS. When exposed to light, however, AlClPc had a substantially unfavorable influence on mitochondrial function, strengthening its intrinsic inhibitory action on CAT, producing oxidative stress, and jeopardizing mitochondrial bioenergetics. In terms of oxidative stress parameters, AlClPc induced lipid peroxidation and decreased the level of reduced glutathione (GSH) in mitochondria. Regarding bioenergetics, AlClPc promoted oxidative phosphorylation uncoupling and photodynamic inactivation of complex I, complex II, and the FF-ATP synthase complex, lowering mitochondrial ATP production. Lastly, AlClPc exhibited a concentration-dependent decrease in the viability of HepG2 cells, regardless of the presence or absence of photostimulation. While the harmful photodynamic effects of AlClPc on mitochondrial bioenergetics hold promise for treating HCC and other malignancies, the inherent toxic impacts on HepG2 cells underscore the need for caution in its application for this purpose.
Mechanistic role for mTORC1 signaling in profibrotic toxicity of low-dose cadmium
Lee CM, Lee HY, Jarrell ZR, Smith RM, Jones DP and Go YM
Cadmium (Cd) is a toxic environmental metal that is naturally present in foods and drinking water. Cd is of increasing concern to human health due to its association with age-related diseases and long biologic half-life. Previous studies show that low-dose Cd exposure via drinking water induces mechanistic target of rapamycin complex 1 (mTORC1) signaling in mice; however, the role of mTORC1 pathway in Cd-induced pro-fibrotic responses has not been established. In the present study, we used human lung fibroblasts to examine whether inhibiting the mTORC1 pathway prevents lung fibrosis signaling induced by low-dose Cd exposure. Results show that rapamycin, a pharmacological inhibitor of mTORC1, inhibited Cd-dependent phosphorylation of ribosomal protein S6, a downstream marker of mTORC1 activation. Rapamycin also decreased Cd-dependent increases in pro-fibrotic markers, α-smooth muscle actin, collagen 1α1 and fibronectin. Cd activated mitochondrial spare respiratory capacity in association with increased cell proliferation. Rapamycin decreased these responses, showing that mTORC1 signaling supports mitochondrial energy supply for cell proliferation, an important step in fibroblast trans-differentiation into myofibroblasts. Collectively, these results establish a key mechanistic role for mTORC1 activation in environmental Cd-dependent lung fibrosis.
Valproic acid-induced oxidative stress: Systematic review, meta-analysis and network pharmacology highlights disruption in antioxidant pathways in rodents
Asghar MA, Tang S, Wan B, Chen Y, Zhang X and Zhao Q
Valproic acid (VPA) is a widely used antiepileptic drug, but its effects on oxidative stress in rodent models have not been systematically reviewed. This meta-analysis aimed to evaluate the impact of VPA on oxidative stress markers in rodents and explore underlying mechanisms through network pharmacology. A systematic search of PubMed, Web of Science, and PsycINFO (2010-2024) was conducted, following PRISMA and CAMARADES guidelines. Forty-two studies involving 639 rodents were included. Meta-analysis and meta-regression were performed using SPSS and R, and network pharmacology identified key pathways. From 1802 studies, 42 met the criteria, involving 639 rodents. VPA treatment was associated with a significant increase in malondialdehyde (MDA) levels (SMD = 30.45, 95 % CI: 17.64-43.25, P < 0.001) and a decrease in clinically relevant biomarkers, such as superoxide dismutase (SOD) (SMD = -13.22, 95 % CI: -19.39--7.04, P < 0.001), glutathione (GSH) (SMD = -16.97, 95 % CI: -28.13--5.82, P < 0.001), catalase (CAT) (SMD = -9.24, 95 % CI: -13.85--4.62, P < 0.001), glutathione S-transferases (GST) (SMD = -8.82, 95 % CI: -17.40--0.24, P = 0.040), and glutathione peroxidase (GPx) (SMD = -36.05, 95 % CI: -60.72--11.37, P < 0.001). Meta-regression analysis suggested that dosing periods and doses significantly impacted oxidative stress markers. Network pharmacology analysis identified 33 key targets and significant pathways, including MAPK signaling, Toll-like receptor signaling, and TNF signaling. VPA induces oxidative stress in rodent models by increasing MDA and reducing antioxidants, suggesting potential oxidative stress-related side effects in patients.
Ferritinophagy is involved in hexavalent chromium-induced ferroptosis in Sertoli cells
Zhuge R, Zhang L, Xue Q, Wang R, Xu J, Wang C, Meng C, Lu R, Yin F and Guo L
Hexavalent chromium [Cr(VI)] has significant adverse effects on the environment and human health, particularly on the male reproductive system. Previously, we observed ferroptosis and autophagy in rat testicular injury induced by Cr(VI). In the present study, we focused on the association between ferroptosis and autophagy in mouse Sertoli cells (TM4) exposed to concentrations of 2.5 μМ, 5 μМ, and 10 μМ Cr(VI). Cr(VI) exposure altered mitochondrial ultrastructure; increased intracellular iron, malondialdehyde, and reactive oxygen species (ROS) levels; decreased glutathione content; increased TfR1 protein expression; and decreased GPX4, FPN1, and SLC7A11 protein expression, ultimately resulting in ferroptosis. Additionally, we observed ferritinophagy, increased expression of BECLIN1, LC3B, and NCOA4, and decreased expression of FTH1 and P62. Inhibition of autophagy and ferritinophagy via 3-MA and small interfering RNA (siRNA)-mediated silencing of NCOA4 ameliorated changes in ferritinophagy- and ferroptosis-associated protein expression, and reduced ROS levels. Rats exposed to Cr(VI) exhibited atrophy of testicular seminiferous tubules, a reduction in germ and Sertoli cells, and the occurrence of ferritinophagy and ferroptosis in cells of the rat testes. These results indicate that ferroptosis, triggered by NCOA4-mediated ferritinophagy, is one of the mechanisms that contribute to Cr(VI)-induced damage in Sertoli cells.
The synergistic anti-Warburg efficacy of temozolomide, metformin and epigallocatechin gallate in glioblastoma
Kuduvalli SS, Senthilathiban DP, Biswas I, Antony JS, Subramani M and Anitha TS
An important hallmark of glioblastoma aggressiveness is its altered metabolism of glucose. This metabolic shift wherein the tumor cells employ aerobic glycolysis regardless of oxygen availability via reprogramming of mitochondrial oxidative phosphorylation is known as the Warburg effect. Previous literatures have linked this metabolic reprograming to tumor progression and glioblastoma cell proliferation making it a key target for targeted drug therapy. Based on this lacuna, the current study aimed to explore the therapeutic efficacy of the triple-drug combination of temozolomide, metformin and epigallocatechin gallate in attenuating Warburg effect and glucose uptake in glioblastoma both in vitro and in vivo. Our results showed that the triple-drug combination had significantly reduced glucose uptake and reversed the Warburg effect in glioblastoma cells and in the glioma-induced xenograft rat model. Thus, the triple-drug combination would serve as an effective therapeutic regime to hamper glioblastoma progression via altering glucose metabolism and improving the overall prognosis in patient setting.
Study on the mechanism of brain injury caused by acute diquat poisoning based on metabolomics
Wang C, Hu H, Liu J, Rong X, Zhang J and Du Y
Brain injury following acute diquat poisoning has become increasingly common in moderate to severe cases, with unclear pathogenesis and high mortality. To investigate this, we conducted metabolomics on brain tissue from poisoned rats, combined with clinical biochemical and pathological analyses. In the high-dose group, 24 metabolites showed significant differences compared to the control group: 18 were upregulated, including cytosine, sedoheptulose-7-phosphate, indole, 3-dehydroshikimate, etc.; 6 were downregulated, including 6-phosphogluconic acid, 3-hydroxybenzoic acid, dAMP, etc. In the low-dose group, 10 metabolites showed significant differences: 4 were upregulated, including pentamidine, γ-tocotrienol, benzoylecgonine, etc.; and 6 were downregulated, including dAMP, glutathione, 3-hydroxybenzoic acid, etc. Enrichment analysis identified two key pathways-phenylalanine, tyrosine, and tryptophan biosynthesis, and the pentose phosphate pathway-as involved in brain injury. ROC analysis of six differential metabolites showed that sedoheptulose-7-phosphate, (2R)-2-hydroxy-3-(phosphonatooxy)propanoate, and 3-hydroxybenzoic acid had AUC values above 0.8. These findings suggest that these three metabolites demonstrate strong diagnostic potential for brain injury induced by diquat poisoning. Correlation analysis linked these biomarkers to clinical indicators such as neutrophil count and the eutrophil to lymphocyte ratio, supporting their relevance. This study provides insights into the mechanisms and biomarkers of diquat-induced brain injury, offering a foundation for future treatment and rapid detection.
Hepatotoxicity of N-nitrosodin-propylamine in larval zebrafish by upregulating the Wnt pathway
Wang Y, Huang S, Wang D, Wu J, Liu F, Liao X, Shi X, Xiao J, Zhang S and Lu H
N-nitrosodin-propylamine is an organic compound mainly used in organic synthesis. As a typical pollutant, the accidental release of N-nitrosodin-propylamine may cause environmental pollution, especially water environment pollution. In the present study, we used the zebrafish model for the first time to evaluate the developmental toxicity of this drug in the liver. Zebrafish larvae fertilized at 72hpf showed a range of toxic responses after 72hpf exposure to the drug. These include increased mortality, delayed absorption of yolk sac nutrients, shorter body length, abnormal liver morphology, gene disruption, and altered expression of various indicators with increasing dose. Studies on the mechanism of toxicity showed that N-nitrosodin-propylamine exposure increased the level of oxidative stress, increased the level of apoptosis in hepatocytes, and up-regulated the transcriptional expression level of Wnt signaling pathway genes. Astaxanthin and IWR-1 can effectively save the liver toxicity in zebrafish caused by N-nitrosodin-propylamine. Our study showed that the drug exposure induced hepatotoxicity in zebrafish larvae through the up-regulation of Wnt signaling pathway, oxidative stress and apoptosis.
Lonicerin protects pancreatic acinar cells from caerulein-induced apoptosis, inflammation, and ferroptosis by activating the SIRT1/GPX4 signaling pathway
Li D, Li C, Jiang S, Wang T, Zhang C, Zhu Z, Zhang G and Fang B
Acute pancreatitis (AP) is a familiar emergency of digestive system characterized by pancreatic inflammation. Lonicerin (LCR) has been reported to exert anti-inflammatory and immunomodulatory characteristics in several inflammatory diseases. Nevertheless, its role and mechanism involved in AP are still unknown. This study was designed to explore the protective effect and potential mechanism of LCR in AP. In this study, LCR and ferrostatin-1 alleviated, but erastin aggravated caerulein (CAE) exposure-induced cytotoxicity and reduction of cell viability in AR42J cells. LCR exhibited a protective role in CAE-treated AR42J cells, as evidenced by alleviation of apoptosis, inflammation, and ferroptosis. Mechanistically, LCR decreased the phosphorylation level of nuclear factor-kappa B p65 and increased the levels of silent information regulator 1 (SIRT1) and glutathione peroxidase 4 (GPX4) in CAE-treated AR42J cells. Furthermore, functional rescue experiments manifested that knockdown of SIRT1 partially negated the inhibitory action of LCR against CAE-induced apoptosis, inflammation, and ferroptosis in AR42J cells. Overall, LCR mitigates apoptosis, inflammation, and ferroptosis in CAE-exposed AR42J cells, which is related to the activation of the SIRT1/GPX4 signaling pathway.
Synergistic polytherapy for the broad-spectrum treatment of chemically-induced seizures in rats
Cornelissen AS, van den Berg RM, Klaassen SD, de Koning JC, Langenberg JP, de Lange ECM and Joosen MJA
Chemically-induced seizures, as a result of exposure to a neurotoxic compound, present a serious health concern. Compounds can elicit seizure activity through disruption of neuronal signaling by neurotransmitters, either by mimicking, modulating or antagonizing their action at the receptor or interfering with their metabolism. Benzodiazepines, such as diazepam and midazolam, and barbiturates are the mainstay of treatment of seizures. However, chemically-induced seizures are often persistent, requiring repeated treatment and increased doses of anticonvulsants, which in turn may lead to severe adverse effects such as respiratory depression. Here, we investigated the potential of rational polytherapy consisting of the benzodiazepine midazolam and the selective α-adrenergic agonist dexmedetomidine as an improved, generically applicable anticonvulsant treatment regimen. Therapeutic efficacy was evaluated against two experimental paradigm compounds that induce persistent seizures in rats, the rodenticide TETS and the nerve agent soman. Following exposure, both TETS and soman elicited profound seizure activity and convulsions, associated with substantial mortality. Treatment with midazolam or dexmedetomidine alone provided no or limited suppression of seizure activity and improvement of survival at 4 h. Polytherapy consisting of midazolam and dexmedetomidine showed excellent anticonvulsant efficacy. Even at low doses, polytherapy showed a profound effect that lasted for the duration of the experiment. Analysis of the dose-response relationships confirmed presence of synergy. Administration of polytherapy in non-exposed animals did not indicate aggravation of adverse effects on respiration or heart rate. Even though more research is needed for the translation to clinical use, polytherapy consisting of midazolam and dexmedetomidine shows promise for the broad-spectrum treatment of (chemically-induced) seizures in emergency situations.
Acute ammonia stress affects the immune response, oxidative stress, ammonia transport and detoxication in the hepatopancreas of freshwater mollusk Solenaia oleivora
Zhang T, Xu D, Zhou Y, Ma X and Wen H
Ammonia is a common and major pollutant in aquatic systems. Excessive ammonia has toxic effects on hepatopancreas in aquatic animals. In this study, we investigated the toxic effects of acute ammonia (concentration: 10 mg/L; test duration: 48 h) stress on the hepatopancreas of a freshwater mollusk, Solenaia oleivora. Transcriptome analysis identified 3355 differentially expressed genes (DEGs), including 1432 up-regulated and 1923 down-regulated genes. Many DEGs were associated with immune and stress responses, including heat shock proteins, pattern recognition receptors, and lysozyme. In addition, some DEGs were related to ammonia transport and detoxification, such as aquaporins, Kchannel, V-ATPase, cytochrome p450, glutathione transferase, and glutamine synthetase. Physiological analysis showed that ammonia stress increased the activities of antioxidant enzymes (superoxide dismutase and catalase) and non-specific immune enzymes (acid phosphatase) and the levels of liver injury markers (malonaldehyde, aspartate aminotransferase, and alanine transaminase). TdT-mediated dUTP nick-end labeling assay revealed that ammonia stress induced apoptosis in the hepatopancreas. These results indicated the toxic effects of ammonia on hepatopancreas on the immune response, oxidative stress, ammonia transport and detoxification of S. oleivora. Our findings will accumulate data on the toxic effects of ammonia on the hepatopancreas of aquatic animals.