Pub Date : 2025-10-25DOI: 10.1007/s00204-025-04225-7
Johannes Borchers, Florinda Perugino, Andreas Schlosser, Stephanie Lamer, Leonie Lutz, Lorenzo Pedroni, Luca Dellafiora, Angela Mally
{"title":"Correction: Identification of small GTPases as potential target proteins of the mycotoxin and renal carcinogen ochratoxin A.","authors":"Johannes Borchers, Florinda Perugino, Andreas Schlosser, Stephanie Lamer, Leonie Lutz, Lorenzo Pedroni, Luca Dellafiora, Angela Mally","doi":"10.1007/s00204-025-04225-7","DOIUrl":"https://doi.org/10.1007/s00204-025-04225-7","url":null,"abstract":"","PeriodicalId":8329,"journal":{"name":"Archives of Toxicology","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-10-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145370179","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Fentanyl and its analog valerylfentanyl (CAS: 122882-90-0) represent a major toxicological threat amid the ongoing opioid crisis. Using an integrative in silico approach (STopTox, ProTox 3.0, TEST 5.1.2, VEGA QSAR, Percepta, ADMETlab, admetSAR), we evaluated key toxicity end points relevant to clinical and forensic toxicology. Valerylfentanyl exhibited high acute toxicity with predicted LD50 values as low as 18.0 mg/kg (ProTox) and 150.13 mg/kg (TEST). Both compounds showed strong potential to inhibit the hERG potassium channel (95.7% for valerylfentanyl), indicating a substantial risk of cardiotoxicity. Predicted organ-specific effects were most prominent in the lungs (94%), cardiovascular (89%), and gastrointestinal systems (81%), with moderate impact on kidneys and blood. Skin irritation potential was high (up to 81.6%), while genotoxicity remained low, though valerylfentanyl showed non-negligible mutagenic probability in some models. The structural analysis identified piperidine-related toxicophores responsible for these effects. Our findings confirm that minor structural modifications can significantly alter toxicological profiles. These results underscore the application of integrated in silico approach for rapid, ethical hazard identification of emerging NPS, supporting their critical application in both clinical and forensic toxicology.
{"title":"Toxicity of fentanyl (CAS: 437-38-7) and valerylfentanyl (CAS: 122882-90-0): the first integrative in silico toxicology study with clinical and forensic implications.","authors":"Łukasz Niżnik, Oktawia Fijałkowska, Kamil Jurowski","doi":"10.1007/s00204-025-04214-w","DOIUrl":"https://doi.org/10.1007/s00204-025-04214-w","url":null,"abstract":"<p><p>Fentanyl and its analog valerylfentanyl (CAS: 122882-90-0) represent a major toxicological threat amid the ongoing opioid crisis. Using an integrative in silico approach (STopTox, ProTox 3.0, TEST 5.1.2, VEGA QSAR, Percepta, ADMETlab, admetSAR), we evaluated key toxicity end points relevant to clinical and forensic toxicology. Valerylfentanyl exhibited high acute toxicity with predicted LD<sub>50</sub> values as low as 18.0 mg/kg (ProTox) and 150.13 mg/kg (TEST). Both compounds showed strong potential to inhibit the hERG potassium channel (95.7% for valerylfentanyl), indicating a substantial risk of cardiotoxicity. Predicted organ-specific effects were most prominent in the lungs (94%), cardiovascular (89%), and gastrointestinal systems (81%), with moderate impact on kidneys and blood. Skin irritation potential was high (up to 81.6%), while genotoxicity remained low, though valerylfentanyl showed non-negligible mutagenic probability in some models. The structural analysis identified piperidine-related toxicophores responsible for these effects. Our findings confirm that minor structural modifications can significantly alter toxicological profiles. These results underscore the application of integrated in silico approach for rapid, ethical hazard identification of emerging NPS, supporting their critical application in both clinical and forensic toxicology.</p>","PeriodicalId":8329,"journal":{"name":"Archives of Toxicology","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145318187","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-16DOI: 10.1007/s00204-025-04220-y
Eline Verscheure, Dina Vandervoort, Emily Deruyck, Katrien Poels, Manosij Ghosh, Jeroen Vanoirbeek, Lode Godderis
Antineoplastic agents are hazardous compounds frequently used in cancer treatment. It is already known that the hospital environment poses a risk of occupational exposure to these agents. However, recent years, the rise of outpatient treatment and at-home treatment has introduced an additional risk including also cohabitants of patients. We identified a clear need for highly sensitive monitoring methods to assess exposure to high-risk compounds in a home setting. This study presents two validated methods for quantifying five high-risk antineoplastic agents in urine: one for cyclophosphamide, etoposide, mitomycin C and imatinib, and one for alpha-fluoro-beta-alanine. Liquid-liquid extraction with ethyl acetate was used for extraction of cyclophosphamide, etoposide, mitomycin C and imatinib from urine. Alpha-fluoro-beta-alanine was extracted using solid-phase extraction with Oasis HLB cartridges. All samples were analysed using ultra-performance liquid chromatography coupled to tandem mass spectrometry. During method validation, selectivity, extraction efficiency, matrix effect, process efficiency, linearity, sensitivity, precision and accuracy were established. The lower limits of quantification were determined to be 0.1 ng/mL (cyclophosphamide and mitomycin C), 0.7 ng/mL (etoposide), 1 ng/mL (alpha-fluoro-beta-alanine) and 10 ng/mL (imatinib). The methods were fully validated and are now ready for application in the field.
{"title":"Method validation for quantification of five antineoplastic agents in urine using UPLC-ESI-MS/MS.","authors":"Eline Verscheure, Dina Vandervoort, Emily Deruyck, Katrien Poels, Manosij Ghosh, Jeroen Vanoirbeek, Lode Godderis","doi":"10.1007/s00204-025-04220-y","DOIUrl":"https://doi.org/10.1007/s00204-025-04220-y","url":null,"abstract":"<p><p>Antineoplastic agents are hazardous compounds frequently used in cancer treatment. It is already known that the hospital environment poses a risk of occupational exposure to these agents. However, recent years, the rise of outpatient treatment and at-home treatment has introduced an additional risk including also cohabitants of patients. We identified a clear need for highly sensitive monitoring methods to assess exposure to high-risk compounds in a home setting. This study presents two validated methods for quantifying five high-risk antineoplastic agents in urine: one for cyclophosphamide, etoposide, mitomycin C and imatinib, and one for alpha-fluoro-beta-alanine. Liquid-liquid extraction with ethyl acetate was used for extraction of cyclophosphamide, etoposide, mitomycin C and imatinib from urine. Alpha-fluoro-beta-alanine was extracted using solid-phase extraction with Oasis HLB cartridges. All samples were analysed using ultra-performance liquid chromatography coupled to tandem mass spectrometry. During method validation, selectivity, extraction efficiency, matrix effect, process efficiency, linearity, sensitivity, precision and accuracy were established. The lower limits of quantification were determined to be 0.1 ng/mL (cyclophosphamide and mitomycin C), 0.7 ng/mL (etoposide), 1 ng/mL (alpha-fluoro-beta-alanine) and 10 ng/mL (imatinib). The methods were fully validated and are now ready for application in the field.</p>","PeriodicalId":8329,"journal":{"name":"Archives of Toxicology","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145298387","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-10DOI: 10.1007/s00204-025-04218-6
Luis Kenedy Alves Rocha Filho, Graziella Iara Silva, Mayara Salgado Silva
The increasing use of bee products in the food, cosmetic, and pharmaceutical industries has intensified interest in their therapeutic properties. However, the safety of these natural compounds warrants careful consideration, particularly within the context of pharmacovigilance. This study aimed to conduct an integrative review of the scientific literature and official pharmacovigilance databases (Notivisa, EudraVigilance, FDA/MedWatch, TGA, among others) to identify adverse effects and toxicological risks associated with the use of honey, apitoxin (bee venom), propolis, royal jelly, bee pollen, and beeswax. The findings indicated that although widely used, these products are not free from risk. Reported cases included infant botulism and grayanotoxin poisoning linked to honey; anaphylaxis and fatalities following therapeutic use of bee venom; contact dermatitis and systemic reactions induced by propolis; severe asthma attacks after royal jelly consumption; adulteration of pollen-based supplements with illicit pharmacological substances; and allergic reactions to beeswax in cosmetic products. Allergic responses were the most prevalent type of adverse event, particularly among atopic individuals. These findings highlight the need for appropriate labeling, post-marketing surveillance, and educational campaigns to ensure the safe use of these products. It is concluded that despite their recognized benefits, apitherapeutic products must be used with caution, and the strengthening of pharmacovigilance systems is essential to safeguard public health.
{"title":"Pharmacovigilance and toxicological risks associated with apitherapeutic products: a systematic overview.","authors":"Luis Kenedy Alves Rocha Filho, Graziella Iara Silva, Mayara Salgado Silva","doi":"10.1007/s00204-025-04218-6","DOIUrl":"https://doi.org/10.1007/s00204-025-04218-6","url":null,"abstract":"<p><p>The increasing use of bee products in the food, cosmetic, and pharmaceutical industries has intensified interest in their therapeutic properties. However, the safety of these natural compounds warrants careful consideration, particularly within the context of pharmacovigilance. This study aimed to conduct an integrative review of the scientific literature and official pharmacovigilance databases (Notivisa, EudraVigilance, FDA/MedWatch, TGA, among others) to identify adverse effects and toxicological risks associated with the use of honey, apitoxin (bee venom), propolis, royal jelly, bee pollen, and beeswax. The findings indicated that although widely used, these products are not free from risk. Reported cases included infant botulism and grayanotoxin poisoning linked to honey; anaphylaxis and fatalities following therapeutic use of bee venom; contact dermatitis and systemic reactions induced by propolis; severe asthma attacks after royal jelly consumption; adulteration of pollen-based supplements with illicit pharmacological substances; and allergic reactions to beeswax in cosmetic products. Allergic responses were the most prevalent type of adverse event, particularly among atopic individuals. These findings highlight the need for appropriate labeling, post-marketing surveillance, and educational campaigns to ensure the safe use of these products. It is concluded that despite their recognized benefits, apitherapeutic products must be used with caution, and the strengthening of pharmacovigilance systems is essential to safeguard public health.</p>","PeriodicalId":8329,"journal":{"name":"Archives of Toxicology","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145273545","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-08DOI: 10.1007/s00204-025-04215-9
Germaine Aalderink, Hugo Brouwer, Jingxuan Wang, Aafke W F Janssen, Meike van der Zande, Coen Govers, Tamara Hoppenbrouwers, Hans Bouwmeester, Mathias Busch
The intestinal epithelium forms a selective barrier between the intestinal lumen and the subepithelial layer. Intestinal epithelium plays a critical role in initiating inflammatory tissue responses in vivo, which remains challenging to emulate in vitro. Caco-2 cells are commonly used models of the intestinal epithelium, but lack crucial receptors and pathways associated with pro-inflammatory reactions. Human-induced pluripotent stem cell (iPSC)-based in vitro models are assumed to provide a system that better emulates in vivo responses. This study evaluated the inflammatory response of iPSC-derived intestinal epithelial cells (IEC) and Caco-2-derived intestinal epithelial cells to the microbial toxins lipopolysaccharide (LPS) and nigericin. Here, iPSCs were differentiated towards enterocyte, goblet- and Paneth-like cells without using three-dimensional culture techniques. The formed monolayer barriers were exposed to a combination of 0-100 µM nigericin and 100 ng/mL LPS on either the apical or basolateral side. The treatment-induced expression of cytokine genes and cytokine secretion were compared between the iPSC-derived cell model and differentiated Caco-2 cell layers. Nigericin exposure in combination with LPS significantly reduced transepithelial electrical resistance in the iPSC-derived model, and resulted in a tenfold increased secretion of the pro-inflammatory cytokines interleukin (IL)-6, IL-8, and tumor necrosis factor-alpha compared to the negative control. A similar increase was observed for the mRNA expression of these cytokines. No significant effect on TEER, cytokine secretion, or mRNA expression was observed in the Caco-2 model. Overall, this study shows that iPSC-IECs are a more sensitive model compared to Caco-2 to emulate inflammatory perturbations of the human intestinal epithelium.
{"title":"Pro-inflammatory response of human iPSC-derived intestinal epithelial monolayers towards microbial toxins LPS and nigericin.","authors":"Germaine Aalderink, Hugo Brouwer, Jingxuan Wang, Aafke W F Janssen, Meike van der Zande, Coen Govers, Tamara Hoppenbrouwers, Hans Bouwmeester, Mathias Busch","doi":"10.1007/s00204-025-04215-9","DOIUrl":"https://doi.org/10.1007/s00204-025-04215-9","url":null,"abstract":"<p><p>The intestinal epithelium forms a selective barrier between the intestinal lumen and the subepithelial layer. Intestinal epithelium plays a critical role in initiating inflammatory tissue responses in vivo, which remains challenging to emulate in vitro. Caco-2 cells are commonly used models of the intestinal epithelium, but lack crucial receptors and pathways associated with pro-inflammatory reactions. Human-induced pluripotent stem cell (iPSC)-based in vitro models are assumed to provide a system that better emulates in vivo responses. This study evaluated the inflammatory response of iPSC-derived intestinal epithelial cells (IEC) and Caco-2-derived intestinal epithelial cells to the microbial toxins lipopolysaccharide (LPS) and nigericin. Here, iPSCs were differentiated towards enterocyte, goblet- and Paneth-like cells without using three-dimensional culture techniques. The formed monolayer barriers were exposed to a combination of 0-100 µM nigericin and 100 ng/mL LPS on either the apical or basolateral side. The treatment-induced expression of cytokine genes and cytokine secretion were compared between the iPSC-derived cell model and differentiated Caco-2 cell layers. Nigericin exposure in combination with LPS significantly reduced transepithelial electrical resistance in the iPSC-derived model, and resulted in a tenfold increased secretion of the pro-inflammatory cytokines interleukin (IL)-6, IL-8, and tumor necrosis factor-alpha compared to the negative control. A similar increase was observed for the mRNA expression of these cytokines. No significant effect on TEER, cytokine secretion, or mRNA expression was observed in the Caco-2 model. Overall, this study shows that iPSC-IECs are a more sensitive model compared to Caco-2 to emulate inflammatory perturbations of the human intestinal epithelium.</p>","PeriodicalId":8329,"journal":{"name":"Archives of Toxicology","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145249477","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-07DOI: 10.1007/s00204-025-04204-y
Annette Zschiesche, Jeremy Carlier, Jörg Pietsch, Martin Scheu, Jasmin Seibt, Francesco P Busardò, Volker Auwärter, Laura M Huppertz
The rapid emergence of synthetic cannabinoid receptor agonists (SCRAs) poses challenges for drug testing, particularly when analyzing urine samples due to the rapid metabolization of the parent compounds. In early 2023, two novel SCRAs were reported to the European Union Drugs Agency (EUDA): ADMB-3TMS-PrINACA and Cumyl-3TMS-PrINACA, which are both indazole SCRAs featuring a trimethylsilyl propyl moiety connected to the tertiary indazole nitrogen. Peaks corresponding to metabolites of ADMB-BINACA (also known as ADB-BUTINACA) and Cumyl-4CN-BINACA observed with retention time shifts in a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for detecting SCRAs were later identified as metabolites of ADMB- and Cumyl-3TMS-PrINACA. Pooled human liver microsome (pHLMs, 25 µmol/L) and pooled human hepatocyte (PHH, 20 µmol/L) assays were performed to generate metabolites. Additionally, human urine samples were analyzed by reversed phase liquid chromatography-quadrupole-time-of-flight-mass spectrometry (LC-QToF-MS), assisted by GLORYx and BioTransformer 3.0 for in silico metabolite prediction. Gas chromatography-mass spectrometry (GC-MS) was used to identify substances in seized materials. In total, 34 metabolites for ADMB-3TMS-PrINACA and 38 for Cumyl-3TMS-PrINACA were tentatively identified. Major biotransformations included side chain monohydroxylation (specific markers) and TMS-group cleavage, likely initiated by oxidative Si-demethylation followed by further hydroxylation resulting in an N-3-OH-propyl metabolite and further oxidation to the respective N-propionic acid. Most of these biomarkers were detected in the blood, urine, and stomach content of a deceased poly-drug user exposed to ADMB-3TMS-PrINACA. Overall, Cumyl-3TMS-PrINACA was more prevalent than ADMB-3TMS-PrINACA in Germany according to routine urine testing. This work provides the first investigation of the metabolic fate and suggests biomarkers for these new SCRAs.
{"title":"Synthetic cannabinoid receptor agonists containing silicon: exploring the metabolic pathways of ADMB- and Cumyl-3TMS-PrINACA in human urine specimens and post mortem material compared to in vitro and in silico data.","authors":"Annette Zschiesche, Jeremy Carlier, Jörg Pietsch, Martin Scheu, Jasmin Seibt, Francesco P Busardò, Volker Auwärter, Laura M Huppertz","doi":"10.1007/s00204-025-04204-y","DOIUrl":"https://doi.org/10.1007/s00204-025-04204-y","url":null,"abstract":"<p><p>The rapid emergence of synthetic cannabinoid receptor agonists (SCRAs) poses challenges for drug testing, particularly when analyzing urine samples due to the rapid metabolization of the parent compounds. In early 2023, two novel SCRAs were reported to the European Union Drugs Agency (EUDA): ADMB-3TMS-PrINACA and Cumyl-3TMS-PrINACA, which are both indazole SCRAs featuring a trimethylsilyl propyl moiety connected to the tertiary indazole nitrogen. Peaks corresponding to metabolites of ADMB-BINACA (also known as ADB-BUTINACA) and Cumyl-4CN-BINACA observed with retention time shifts in a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for detecting SCRAs were later identified as metabolites of ADMB- and Cumyl-3TMS-PrINACA. Pooled human liver microsome (pHLMs, 25 µmol/L) and pooled human hepatocyte (PHH, 20 µmol/L) assays were performed to generate metabolites. Additionally, human urine samples were analyzed by reversed phase liquid chromatography-quadrupole-time-of-flight-mass spectrometry (LC-QToF-MS), assisted by GLORYx and BioTransformer 3.0 for in silico metabolite prediction. Gas chromatography-mass spectrometry (GC-MS) was used to identify substances in seized materials. In total, 34 metabolites for ADMB-3TMS-PrINACA and 38 for Cumyl-3TMS-PrINACA were tentatively identified. Major biotransformations included side chain monohydroxylation (specific markers) and TMS-group cleavage, likely initiated by oxidative Si-demethylation followed by further hydroxylation resulting in an N-3-OH-propyl metabolite and further oxidation to the respective N-propionic acid. Most of these biomarkers were detected in the blood, urine, and stomach content of a deceased poly-drug user exposed to ADMB-3TMS-PrINACA. Overall, Cumyl-3TMS-PrINACA was more prevalent than ADMB-3TMS-PrINACA in Germany according to routine urine testing. This work provides the first investigation of the metabolic fate and suggests biomarkers for these new SCRAs.</p>","PeriodicalId":8329,"journal":{"name":"Archives of Toxicology","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145237753","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-06DOI: 10.1007/s00204-025-04216-8
Rajan K Tripathy, Khushi Goyal, Prakash Y Khandave, Janek Bzdrenga, Xavier Brazzolotto, Florian Nachon, Abhay H Pande
A novel series of toxic nerve agents called Novichok agents (A-agents and their binary form) was developed in the Soviet Union during the 70s-90s under the FOLIANT program. These agents, including A-230, A-232, and A-234, are structurally distinct from earlier G- and V-series agents and pose significant challenges due to their high environmental persistence, poor aqueous degradation, and rapid irreversible inhibition of acetylcholinesterase (AChE). Current medical countermeasures, such as atropine and oxime reactivators, show limited efficacy against A-agents, particularly due to the low reactivity of A-agent-AChE conjugates. Surrogate-based studies have provided partial insights into the reactivation and decontamination strategies, but they do not fully replicate the behavior of actual A-agents. Developing efficient reactivators against A-agents appears challenging. Emerging skin decontamination strategies, including Reactive Skin Decontamination Lotion and metal-organic framework catalysts, show some success. In this context, enzymatic biocatalysts such as engineered variants of paraoxonase (PON1) and phosphotriesterase (PTE) are valuable antidotes, although their catalytic efficiencies against A-agents remain suboptimal. The development of broad-spectrum bioscavengers with prolonged circulatory half-life, like butyrylcholinesterase, or other recombinant enzyme candidates, enhanced through fusion protein engineering and mutagenesis, represents a promising avenue. However, significant knowledge gaps persist due to limited availability and high-risk handling of these agents. Advancing countermeasures requires continued integration of computational modeling, biochemical engineering, and surrogate validation strategies to overcome these biochemical and therapeutic challenges.
{"title":"Challenges and advances in the development of antidotes against A-series nerve agents.","authors":"Rajan K Tripathy, Khushi Goyal, Prakash Y Khandave, Janek Bzdrenga, Xavier Brazzolotto, Florian Nachon, Abhay H Pande","doi":"10.1007/s00204-025-04216-8","DOIUrl":"https://doi.org/10.1007/s00204-025-04216-8","url":null,"abstract":"<p><p>A novel series of toxic nerve agents called Novichok agents (A-agents and their binary form) was developed in the Soviet Union during the 70s-90s under the FOLIANT program. These agents, including A-230, A-232, and A-234, are structurally distinct from earlier G- and V-series agents and pose significant challenges due to their high environmental persistence, poor aqueous degradation, and rapid irreversible inhibition of acetylcholinesterase (AChE). Current medical countermeasures, such as atropine and oxime reactivators, show limited efficacy against A-agents, particularly due to the low reactivity of A-agent-AChE conjugates. Surrogate-based studies have provided partial insights into the reactivation and decontamination strategies, but they do not fully replicate the behavior of actual A-agents. Developing efficient reactivators against A-agents appears challenging. Emerging skin decontamination strategies, including Reactive Skin Decontamination Lotion and metal-organic framework catalysts, show some success. In this context, enzymatic biocatalysts such as engineered variants of paraoxonase (PON1) and phosphotriesterase (PTE) are valuable antidotes, although their catalytic efficiencies against A-agents remain suboptimal. The development of broad-spectrum bioscavengers with prolonged circulatory half-life, like butyrylcholinesterase, or other recombinant enzyme candidates, enhanced through fusion protein engineering and mutagenesis, represents a promising avenue. However, significant knowledge gaps persist due to limited availability and high-risk handling of these agents. Advancing countermeasures requires continued integration of computational modeling, biochemical engineering, and surrogate validation strategies to overcome these biochemical and therapeutic challenges.</p>","PeriodicalId":8329,"journal":{"name":"Archives of Toxicology","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145231101","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-04DOI: 10.1007/s00204-025-04210-0
Yoon-Seok Seo, Kwang-Hoon Park, Jung-Min Park, Jae-Hyeong Kim, Seong-Jin Choi, Min-Seok Kim, Kyuhong Lee, Moo-Yeol Lee
Smoking is a well-established risk factor for cardiovascular diseases, yet direct evidence linking cigarette smoke (CS) exposure to neointimal formation remains limited. To address this gap, we investigated the effects of CS exposure on neointimal formation using an injury-induced arterial mouse model. Neointimal formation was induced in the femoral artery via mechanical injury with a guidewire, and mice were exposed to CS generated from 3R4F reference cigarettes at a total particulate matter concentration of 600 µg/L for 2 h daily. CS exposure commenced three days prior to injury induction and continued until euthanasia on days 7 or 14 post-injury. CS exposure significantly exacerbated neointimal formation; however, in the absence of injury, it did not induce structural alterations in the femoral artery. In vitro, cigarette smoke extract (CSE) at 0.1%-corresponding to approximately 50 ng/mL nicotine, a clinically relevant concentration in smokers-enhanced the proliferation of aortic smooth muscle cells, a critical process in neointimal development. However, CSE exhibited minimal effects on other cellular processes involved in neointimal formation, including phenotype switching, adhesion, migration, and extracellular matrix deposition. Mechanistically, CSE exposure increased Akt and FOXO3a phosphorylation, leading to a downregulation of p27 and an upregulation of CDK2 and cyclin E, ultimately promoting Rb phosphorylation and cell cycle progression. In conclusion, although CS alone does not appear sufficient to initiate neointimal formation, it significantly exacerbates or accelerates its progression in a primed vascular environment. The promotion of vascular smooth muscle cell proliferation via cell cycle progression may underlie this effect.
{"title":"Inhalation exposure to cigarette smoke promotes neointimal formation in mouse model of arterial injury.","authors":"Yoon-Seok Seo, Kwang-Hoon Park, Jung-Min Park, Jae-Hyeong Kim, Seong-Jin Choi, Min-Seok Kim, Kyuhong Lee, Moo-Yeol Lee","doi":"10.1007/s00204-025-04210-0","DOIUrl":"https://doi.org/10.1007/s00204-025-04210-0","url":null,"abstract":"<p><p>Smoking is a well-established risk factor for cardiovascular diseases, yet direct evidence linking cigarette smoke (CS) exposure to neointimal formation remains limited. To address this gap, we investigated the effects of CS exposure on neointimal formation using an injury-induced arterial mouse model. Neointimal formation was induced in the femoral artery via mechanical injury with a guidewire, and mice were exposed to CS generated from 3R4F reference cigarettes at a total particulate matter concentration of 600 µg/L for 2 h daily. CS exposure commenced three days prior to injury induction and continued until euthanasia on days 7 or 14 post-injury. CS exposure significantly exacerbated neointimal formation; however, in the absence of injury, it did not induce structural alterations in the femoral artery. In vitro, cigarette smoke extract (CSE) at 0.1%-corresponding to approximately 50 ng/mL nicotine, a clinically relevant concentration in smokers-enhanced the proliferation of aortic smooth muscle cells, a critical process in neointimal development. However, CSE exhibited minimal effects on other cellular processes involved in neointimal formation, including phenotype switching, adhesion, migration, and extracellular matrix deposition. Mechanistically, CSE exposure increased Akt and FOXO3a phosphorylation, leading to a downregulation of p27 and an upregulation of CDK2 and cyclin E, ultimately promoting Rb phosphorylation and cell cycle progression. In conclusion, although CS alone does not appear sufficient to initiate neointimal formation, it significantly exacerbates or accelerates its progression in a primed vascular environment. The promotion of vascular smooth muscle cell proliferation via cell cycle progression may underlie this effect.</p>","PeriodicalId":8329,"journal":{"name":"Archives of Toxicology","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145224617","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-04DOI: 10.1007/s00204-025-04217-7
Rui Filipe Malheiro, Ana Catarina Costa, Helena Carmo, Félix Carvalho, João Pedro Silva
Synthetic cannabinoids (SCs) have been increasingly associated with neurodevelopmental impairment; however, the underlying mechanisms remain poorly understood. In particular, the impact of SCs on mitochondria during neurodifferentiation remains largely unexplored, despite the central role of these organelles in this process. Building upon our previous findings that THJ-2201, a widely used SC, enhances neurite outgrowth in NG108-15 neuroblastoma-glioma cells at biologically relevant concentrations (1 pM-1 μM), we investigated whether this SC influences mitochondrial function, morphology, and dynamics during neurodifferentiation. THJ-2201 exposure caused a 30-40% reduction in intracellular ATP levels in a CB1-dependent manner, along with a 20-30% decrease in TMRE retention during NG108-15 neurodifferentiation. Cells treated with 1 μM THJ-2201 failed to sustain the expected increase in VDAC levels (an indirect marker of mitochondrial mass) during regular differentiation. Concurrently, THJ-2201 elevated PGC-1α levels, a key regulator of mitochondrial biogenesis, by disrupting its translocation to the nucleus. Expression of both fusion (Opa1, Mfn1, and Mfn2) and fission (Drp1 and Fis1) markers exhibited a less pronounced increase between 24 and 72 h in THJ-2201-treated cells. Mitochondrial morphology exhibited alterations in mean area, perimeter, branching, and circularity in the soma after 72 h exposure. Additionally, THJ-2201 reduced mitochondrial mobility in neurites without affecting their average speed or run length and led to a mitochondrial accumulation within neurites, as indicated by decreased Miro1 expression. Overall, these findings suggest that THJ-2201-induced mitochondrial remodelling and redistribution may transiently enhance local energy supply for neurite outgrowth, but at the expense of somatic mitochondrial function, resulting in an overall bioenergetic imbalance.
{"title":"The synthetic cannabinoid THJ-2201 modulates mitochondrial activity and enhances mitochondrial recruitment to newly-forming neurites during neurodifferentiation of NG108-15 cells.","authors":"Rui Filipe Malheiro, Ana Catarina Costa, Helena Carmo, Félix Carvalho, João Pedro Silva","doi":"10.1007/s00204-025-04217-7","DOIUrl":"https://doi.org/10.1007/s00204-025-04217-7","url":null,"abstract":"<p><p>Synthetic cannabinoids (SCs) have been increasingly associated with neurodevelopmental impairment; however, the underlying mechanisms remain poorly understood. In particular, the impact of SCs on mitochondria during neurodifferentiation remains largely unexplored, despite the central role of these organelles in this process. Building upon our previous findings that THJ-2201, a widely used SC, enhances neurite outgrowth in NG108-15 neuroblastoma-glioma cells at biologically relevant concentrations (1 pM-1 μM), we investigated whether this SC influences mitochondrial function, morphology, and dynamics during neurodifferentiation. THJ-2201 exposure caused a 30-40% reduction in intracellular ATP levels in a CB1-dependent manner, along with a 20-30% decrease in TMRE retention during NG108-15 neurodifferentiation. Cells treated with 1 μM THJ-2201 failed to sustain the expected increase in VDAC levels (an indirect marker of mitochondrial mass) during regular differentiation. Concurrently, THJ-2201 elevated PGC-1α levels, a key regulator of mitochondrial biogenesis, by disrupting its translocation to the nucleus. Expression of both fusion (Opa1, Mfn1, and Mfn2) and fission (Drp1 and Fis1) markers exhibited a less pronounced increase between 24 and 72 h in THJ-2201-treated cells. Mitochondrial morphology exhibited alterations in mean area, perimeter, branching, and circularity in the soma after 72 h exposure. Additionally, THJ-2201 reduced mitochondrial mobility in neurites without affecting their average speed or run length and led to a mitochondrial accumulation within neurites, as indicated by decreased Miro1 expression. Overall, these findings suggest that THJ-2201-induced mitochondrial remodelling and redistribution may transiently enhance local energy supply for neurite outgrowth, but at the expense of somatic mitochondrial function, resulting in an overall bioenergetic imbalance.</p>","PeriodicalId":8329,"journal":{"name":"Archives of Toxicology","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145224582","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-03DOI: 10.1007/s00204-025-04134-9
Michael L Dourson, Laura C Green, Edmund A C Crouch, Harvey J Clewell, Thomas Colnot, Tony Cox, Wolfgang Dekant, Linda D Dell, James A Deyo, Bernard K Gadagbui, Helmut Greim, Mahesh Rachamalla Gupta, Tamara House-Knight, Ashish Jachak, Vijayavel Kannappan, Travis R Kline, Therese Manning, Ravi Naidu, Paul Nathanail, Chijioke Onyema, Frank Pagone, Andrew Pawlisz, Tiago Severo Peixe, Katie Richardson, Anurag Sharma, James S Smith, Nitin Verma, Andrea Wojtyniak, Jackie Wright
Many government agencies and expert groups have estimated a safe dose (aka a "reference dose," [RfD]) for perfluorooctane sulfonate (PFOS). Notably, these agencies have derived safe doses that vary over at least 600-fold range. The range is larger still if one includes the U.S. Environmental Protection Agency (USEPA) current science-policy position under the Safe Drinking Water Act, which is that the only safe dose of PFOS is zero. This wide range in safe dose-estimates is surprising, since PFOS is a relatively well-studied, and ubiquitous, chemical. The Steering Committee of the Alliance for Risk Assessment (ARA) called for health-scientists interested in attempting to understand and, if possible, narrow this range of estimates. An advisory committee of eight scientists from four countries was selected from nominations received, and a subsequent invitation to scientists internationally led to the formation of three teams comprised of 24 scientists from nine countries. Each team independently reviewed toxicologic and epidemiologic data, and developed PFOS safe dose-estimates. All three teams concluded that currently available epidemiologic data could not form a reliable basis for PFOS safe dose-assessments. In contrast, results of bioassays of PFOS in laboratory monkeys and rats did provide usable bases from which serum-concentration-based "points of departure" were derived. After applying several, necessarily imprecise, uncertainty factors, the three groups derived PFOS safe dose-estimates that ranged, narrowly, from 20 to 100 nanograms (ng) of PFOS/kg body weight/day. In contrast, USEPA's current (United States Environmental Protection Agency (USEPA) (2024) Human health toxicity assessment for perfluorooctane sulfonic acid (PFOS) and Related Salts. EPA Document No. 815R24007.) estimate of the safe dose is 0.1 ng of PFOS/kg-day.
许多政府机构和专家组估计了全氟辛烷磺酸(PFOS)的安全剂量(即“参考剂量”[RfD])。值得注意的是,这些机构得出的安全剂量至少相差600倍。如果考虑到美国环境保护署(USEPA)目前在《安全饮用水法》(Safe Drinking Water Act)下的科学政策立场,即全氟辛烷磺酸的唯一安全剂量为零,那么这个范围就更大了。如此大范围的安全剂量估计令人惊讶,因为全氟辛烷磺酸是一种研究相对充分、普遍存在的化学物质。风险评估联盟指导委员会呼吁有兴趣的卫生科学家尝试理解并在可能的情况下缩小这一估计范围。从收到的提名中选出了一个由4个国家的8名科学家组成的咨询委员会,随后向国际科学家发出邀请,形成了由9个国家的24名科学家组成的3个小组。每个小组独立审查毒理学和流行病学数据,并制定全氟辛烷磺酸安全剂量估计。所有三个小组的结论是,目前现有的流行病学数据不能构成全氟辛烷磺酸安全剂量评估的可靠基础。相比之下,实验室猴子和大鼠的全氟辛烷磺酸生物测定结果确实提供了可用的基础,从中得出基于血清浓度的“出发点”。在应用了几个必然不精确的不确定因素后,三个小组得出了全氟辛烷磺酸的安全剂量估计,范围从20到100毫微克/公斤体重/天。相比之下,美国环保署目前(美国环境保护局(USEPA)(2024)对全氟辛烷磺酸(PFOS)和相关盐的人体健康毒性评估。EPA文件编号815R24007)的安全剂量估计为0.1 ng /kg-day。
{"title":"Estimating safe doses of perfluorooctane sulfonate (PFOS): an international collaboration.","authors":"Michael L Dourson, Laura C Green, Edmund A C Crouch, Harvey J Clewell, Thomas Colnot, Tony Cox, Wolfgang Dekant, Linda D Dell, James A Deyo, Bernard K Gadagbui, Helmut Greim, Mahesh Rachamalla Gupta, Tamara House-Knight, Ashish Jachak, Vijayavel Kannappan, Travis R Kline, Therese Manning, Ravi Naidu, Paul Nathanail, Chijioke Onyema, Frank Pagone, Andrew Pawlisz, Tiago Severo Peixe, Katie Richardson, Anurag Sharma, James S Smith, Nitin Verma, Andrea Wojtyniak, Jackie Wright","doi":"10.1007/s00204-025-04134-9","DOIUrl":"https://doi.org/10.1007/s00204-025-04134-9","url":null,"abstract":"<p><p>Many government agencies and expert groups have estimated a safe dose (aka a \"reference dose,\" [RfD]) for perfluorooctane sulfonate (PFOS). Notably, these agencies have derived safe doses that vary over at least 600-fold range. The range is larger still if one includes the U.S. Environmental Protection Agency (USEPA) current science-policy position under the Safe Drinking Water Act, which is that the only safe dose of PFOS is zero. This wide range in safe dose-estimates is surprising, since PFOS is a relatively well-studied, and ubiquitous, chemical. The Steering Committee of the Alliance for Risk Assessment (ARA) called for health-scientists interested in attempting to understand and, if possible, narrow this range of estimates. An advisory committee of eight scientists from four countries was selected from nominations received, and a subsequent invitation to scientists internationally led to the formation of three teams comprised of 24 scientists from nine countries. Each team independently reviewed toxicologic and epidemiologic data, and developed PFOS safe dose-estimates. All three teams concluded that currently available epidemiologic data could not form a reliable basis for PFOS safe dose-assessments. In contrast, results of bioassays of PFOS in laboratory monkeys and rats did provide usable bases from which serum-concentration-based \"points of departure\" were derived. After applying several, necessarily imprecise, uncertainty factors, the three groups derived PFOS safe dose-estimates that ranged, narrowly, from 20 to 100 nanograms (ng) of PFOS/kg body weight/day. In contrast, USEPA's current (United States Environmental Protection Agency (USEPA) (2024) Human health toxicity assessment for perfluorooctane sulfonic acid (PFOS) and Related Salts. EPA Document No. 815R24007.) estimate of the safe dose is 0.1 ng of PFOS/kg-day.</p>","PeriodicalId":8329,"journal":{"name":"Archives of Toxicology","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145224874","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}