Pub Date : 2025-12-19DOI: 10.1007/s10565-025-10131-6
Donovan Drouillard, Maria Poimenidou, Marissa Davies, Donna McAllister, William R Clarke, Samuel T Hwang, Francis C Peterson, Brian F Volkman, Michael B Dwinell
Immune dysregulation by aberrant chemokine production underlies many diseases. Targeting chemokine receptors with small molecule inverse agonists, antagonists, or neutralizing antibodies has proven challenging due to non-specific effects and receptor upregulation. Locked dimers of chemokines, generated via cysteine substitutions to produce constitutively homodimeric molecules, offer a promising alternative for receptor-specific inhibition. This study evaluates the in vivo safety and dosing of an engineered CCL20 locked dimer (CCL20LD), which selectively binds CCR6 without inducing chemotaxis. The antagonist-like properties of CCL20LD make it a potential therapeutic for CCL20-CCR6 driven diseases. Daily 14-day subcutaneous administration of CCL20LD at doses previously shown to be therapeutically effective in preclinical models of psoriasis or psoriatic arthritis did not result in weight loss or immune suppression. CCL20LD administration had little to no effects on the complete blood count with differential, comprehensive metabolic panel, urinalysis, organ weights, or bone marrow progenitors. At single cell resolution, doses near 7.5mg/kg/day modestly disrupted T cell dependent B cell activation. While splenomegaly due to extramedullary hematopoiesis was observed at the highest tested dose, serum cytokine levels were largely unchanged. Combined, these findings indicate that selective targeting of CCR6 with an engineered CCL20 dimer is broadly safe in vivo, exhibiting a wide therapeutic window with minimal adverse or immunomodulatory effects.
{"title":"Biosafety assessment of engineered CCL20 locked dimers in vivo.","authors":"Donovan Drouillard, Maria Poimenidou, Marissa Davies, Donna McAllister, William R Clarke, Samuel T Hwang, Francis C Peterson, Brian F Volkman, Michael B Dwinell","doi":"10.1007/s10565-025-10131-6","DOIUrl":"10.1007/s10565-025-10131-6","url":null,"abstract":"<p><p>Immune dysregulation by aberrant chemokine production underlies many diseases. Targeting chemokine receptors with small molecule inverse agonists, antagonists, or neutralizing antibodies has proven challenging due to non-specific effects and receptor upregulation. Locked dimers of chemokines, generated via cysteine substitutions to produce constitutively homodimeric molecules, offer a promising alternative for receptor-specific inhibition. This study evaluates the in vivo safety and dosing of an engineered CCL20 locked dimer (CCL20LD), which selectively binds CCR6 without inducing chemotaxis. The antagonist-like properties of CCL20LD make it a potential therapeutic for CCL20-CCR6 driven diseases. Daily 14-day subcutaneous administration of CCL20LD at doses previously shown to be therapeutically effective in preclinical models of psoriasis or psoriatic arthritis did not result in weight loss or immune suppression. CCL20LD administration had little to no effects on the complete blood count with differential, comprehensive metabolic panel, urinalysis, organ weights, or bone marrow progenitors. At single cell resolution, doses near 7.5mg/kg/day modestly disrupted T cell dependent B cell activation. While splenomegaly due to extramedullary hematopoiesis was observed at the highest tested dose, serum cytokine levels were largely unchanged. Combined, these findings indicate that selective targeting of CCR6 with an engineered CCL20 dimer is broadly safe in vivo, exhibiting a wide therapeutic window with minimal adverse or immunomodulatory effects.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":" ","pages":"14"},"PeriodicalIF":5.9,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12799736/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793396","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-19DOI: 10.1007/s10565-025-10121-8
Yuanyuan Zhang, Wenjie Zhu, Jiang Zhu, Rui Hu, Yunhuang Yang
Exosomes play a crucial role in the transmission of drug resistance in tumors. However, the mechanism of exosomes-mediated transmission in non-small cell lung cancer (NSCLC) under gefitinib treatment remains limited. In this work, we demonstrated that exosomes derived from HCC827/GR cells (drug-resistant) enhanced the survivability of HCC827 cells (drug-sensitive) under treatment with gefitinib. A total of 157 shared upregulated proteins between exosomes and their parent cells were identified in the comparison of the gefitinib-resistant groups versus the gefitinib-sensitive groups. Notably, 69 of these shared proteins are enzymes, and many of them were enriched in pathways related to fatty acid metabolism. Among these enzymes involved in fatty acid metabolism, ACC1 exhibited the highest fold change in upregulated expression in both drug-resistant groups (exosomes and cells). Moreover, the expression of ACC1 was upregulated in gefitinib-sensitive cells after uptake of exosomes from gefitinib-resistant cells. The role of ACC1 in enhancing the survival of HCC827/GR cells under gefitinib treatment was demonstrated using an inhibitor and siRNA-mediated knockdown. Specifically, the upregulated ACC1 stabilized fatty acid oxidation and reactive oxygen species levels in HCC827/GR cells, thereby maintaining cellular metabolic homeostasis. Collectively, this work reveals the transmission of drug resistance in NSCLC via exosomes that carry the ACC1 protein.
{"title":"Exosomal proteomics reveals fatty acid metabolism linked to gefitinib resistance in non-small cell lung cancer.","authors":"Yuanyuan Zhang, Wenjie Zhu, Jiang Zhu, Rui Hu, Yunhuang Yang","doi":"10.1007/s10565-025-10121-8","DOIUrl":"10.1007/s10565-025-10121-8","url":null,"abstract":"<p><p>Exosomes play a crucial role in the transmission of drug resistance in tumors. However, the mechanism of exosomes-mediated transmission in non-small cell lung cancer (NSCLC) under gefitinib treatment remains limited. In this work, we demonstrated that exosomes derived from HCC827/GR cells (drug-resistant) enhanced the survivability of HCC827 cells (drug-sensitive) under treatment with gefitinib. A total of 157 shared upregulated proteins between exosomes and their parent cells were identified in the comparison of the gefitinib-resistant groups versus the gefitinib-sensitive groups. Notably, 69 of these shared proteins are enzymes, and many of them were enriched in pathways related to fatty acid metabolism. Among these enzymes involved in fatty acid metabolism, ACC1 exhibited the highest fold change in upregulated expression in both drug-resistant groups (exosomes and cells). Moreover, the expression of ACC1 was upregulated in gefitinib-sensitive cells after uptake of exosomes from gefitinib-resistant cells. The role of ACC1 in enhancing the survival of HCC827/GR cells under gefitinib treatment was demonstrated using an inhibitor and siRNA-mediated knockdown. Specifically, the upregulated ACC1 stabilized fatty acid oxidation and reactive oxygen species levels in HCC827/GR cells, thereby maintaining cellular metabolic homeostasis. Collectively, this work reveals the transmission of drug resistance in NSCLC via exosomes that carry the ACC1 protein.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"42 1","pages":"3"},"PeriodicalIF":5.9,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12717232/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793025","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
COVID-19 has caused millions of deaths worldwide since 2019. Vaccination has reduced both transmission and disease severity. However, emerging viral variants have weakened vaccine effectiveness, highlighting the need for new antiviral therapies. This study examines how the SARS-CoV-2-Spike protein (SARS-2-S) induces the VSIR-ISX signaling pathway, leading to metabolic disturbances that may worsen disease progression. Using RNA sequencing, we found that SARS-2-S expression in pulmonary cells activates genes involved in tryptophan and arachidonic acid (AA) metabolism, altering bioactive mediators like kynurenine and prostanoids, which are crucial for inflammation and immune responses. Mechanistically, the ACE2-MYD88 pathway, activated by SARS-2-S, enhances the VSIR-ISX axis through NF-κB signaling, driving these metabolic disruptions. Chromatin immunoprecipitation and genome sequencing revealed that ISX, activated via VSIR-MAPK signaling, upregulates enzymes involved in AA metabolism by binding directly to their gene promoters. Notably, disrupting the VSIR-ISX axis using shRNA interference or NF-κB inhibitors effectively mitigated these metabolic disturbances. Our findings suggest that the VSIR-ISX pathway could be a promising therapeutic target for treating COVID-19 by addressing virus-induced metabolic disruptions.
{"title":"Spike protein-induced VSIR-ISX signaling disrupts metabolic homeostasis and promotes COVID-19-related immune dysfunction.","authors":"Li-Ting Wang, Shen-Nien Wang, Shyh-Shin Chiou, Chee-Yin Chai, Shih-Hsien Hsu","doi":"10.1007/s10565-025-10119-2","DOIUrl":"10.1007/s10565-025-10119-2","url":null,"abstract":"<p><p>COVID-19 has caused millions of deaths worldwide since 2019. Vaccination has reduced both transmission and disease severity. However, emerging viral variants have weakened vaccine effectiveness, highlighting the need for new antiviral therapies. This study examines how the SARS-CoV-2-Spike protein (SARS-2-S) induces the VSIR-ISX signaling pathway, leading to metabolic disturbances that may worsen disease progression. Using RNA sequencing, we found that SARS-2-S expression in pulmonary cells activates genes involved in tryptophan and arachidonic acid (AA) metabolism, altering bioactive mediators like kynurenine and prostanoids, which are crucial for inflammation and immune responses. Mechanistically, the ACE2-MYD88 pathway, activated by SARS-2-S, enhances the VSIR-ISX axis through NF-κB signaling, driving these metabolic disruptions. Chromatin immunoprecipitation and genome sequencing revealed that ISX, activated via VSIR-MAPK signaling, upregulates enzymes involved in AA metabolism by binding directly to their gene promoters. Notably, disrupting the VSIR-ISX axis using shRNA interference or NF-κB inhibitors effectively mitigated these metabolic disturbances. Our findings suggest that the VSIR-ISX pathway could be a promising therapeutic target for treating COVID-19 by addressing virus-induced metabolic disruptions.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"42 1","pages":"2"},"PeriodicalIF":5.9,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12717150/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793212","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Paclitaxel (PTX), a commonly utilized chemotherapy drug, is linked to peripheral neuropathy, which limits dosing and significantly affects patients' quality of life. C-terminal binding protein 1 (CtBP1) is a transcriptional coregulator that participates in epigenetic gene regulation, but its role in PTX-induced neuropathic pain remains unclear. In this study, the role of CtBP1 in PTX-induced neuropathic pain is examined, with a focus on its epigenetic regulation in the dorsal root ganglia (DRGs). PTX administration markedly increased CtBP1 protein levels in DRG neurons, which coincided with the development and continuation of mechanical allodynia and thermal hyperalgesia in rat models. Our findings also revealed that CtBP1 interacts with the histone demethylase LSD1-a regulator of H3K9me2-at ErbB2 promoter sites in DRG neurons. PTX treatment increased CtBP1 protein levels, which subsequently induced LSD1 expression and decreased H3K9me2 protein levels at the ErbB2 promoter, indicating epigenetic activation of ErbB2 signaling in DRG neurons implicated in neuropathic pain. Reducing either CtBP1 or LSD1 expression reversed ErbB2 upregulation and attenuated PTX-induced pain sensitivity. These results suggest that the CtBP1-LSD1 complex epigenetically increases ErbB2 expression in DRG neurons, contributing to PTX-induced neuropathy. Targeting the CtBP1-LSD1 pathway could represent a promising therapeutic strategy for the treatment of chemotherapy-induced neuropathic pain.
{"title":"CtBP1-LSD1 complex drives ErbB2 activation via H3K9me2 demethylation in DRGs during paclitaxel-induced neuropathic pain.","authors":"Hsien-Yu Peng, Cheng-Yuan Lai, Gin-Den Chen, Yun-Chih Peng, Chou-Ming Yeh, Tzer-Bin Lin, Hsueh-Hsiao Wang, Jen-Kun Cheng, Po-Sheng Yang, Chia-Man Chou, Dylan Chou, Kuan-Hung Lin, Siao-Tong Nie, Ming-Chun Hsieh","doi":"10.1007/s10565-025-10122-7","DOIUrl":"10.1007/s10565-025-10122-7","url":null,"abstract":"<p><p>Paclitaxel (PTX), a commonly utilized chemotherapy drug, is linked to peripheral neuropathy, which limits dosing and significantly affects patients' quality of life. C-terminal binding protein 1 (CtBP1) is a transcriptional coregulator that participates in epigenetic gene regulation, but its role in PTX-induced neuropathic pain remains unclear. In this study, the role of CtBP1 in PTX-induced neuropathic pain is examined, with a focus on its epigenetic regulation in the dorsal root ganglia (DRGs). PTX administration markedly increased CtBP1 protein levels in DRG neurons, which coincided with the development and continuation of mechanical allodynia and thermal hyperalgesia in rat models. Our findings also revealed that CtBP1 interacts with the histone demethylase LSD1-a regulator of H3K9me2-at ErbB2 promoter sites in DRG neurons. PTX treatment increased CtBP1 protein levels, which subsequently induced LSD1 expression and decreased H3K9me2 protein levels at the ErbB2 promoter, indicating epigenetic activation of ErbB2 signaling in DRG neurons implicated in neuropathic pain. Reducing either CtBP1 or LSD1 expression reversed ErbB2 upregulation and attenuated PTX-induced pain sensitivity. These results suggest that the CtBP1-LSD1 complex epigenetically increases ErbB2 expression in DRG neurons, contributing to PTX-induced neuropathy. Targeting the CtBP1-LSD1 pathway could represent a promising therapeutic strategy for the treatment of chemotherapy-induced neuropathic pain.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"42 1","pages":"1"},"PeriodicalIF":5.9,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12714790/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145773602","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-13DOI: 10.1007/s10565-025-10105-8
Yan Hu, Chuntao Liu
Asthma, a chronic inflammatory disease, has a high disability rate, which greatly increases the disease burden. T cells are pivotal in the pathogenesis of asthma, and Treg cells, due to their role in maintaining immune system balance, represent a promising avenue for therapeutic intervention. Initial weighted correlation network analysis (WGCNA) analysis of asthma-related datasets indicates that N-glycosylation plays a critical role in asthma development. The establishment of an OVA-sensitized asthma model, along with the isolation of naive CD4+ T cells and subsequent in vitro induction of Treg cell differentiation, further underscores the significance of N-glycosylation in the Treg cell differentiation of asthma-related Treg cells. Employing immunofluorescence, flow cytometry, and Western blot techniques revealed that SIRT3-SUMO is instrumental in regulating N-glycosylation-mediated Treg cells development. Mechanistically, overexpression and deSUMOylation of SIRT3 enhance the expression levels of CPT1 and VLCAD to promote fatty acid oxidation (FAO), thereby increasing intracellular acetyl-CoA concentrations. Acetyl-CoA subsequently facilitates the synthesis of N-glycosylation substrates via the hexosamine biosynthetic pathway (HBP), promoting Treg cell differentiation. Ultimately, our in vivo experiments demonstrate that SIRT3-SUMO modulates asthma progression by influencing Treg cells differentiation; thus, augmenting Treg cells populations can inhibit Th2-type and non-Th2-type asthmatic developments. These findings elucidate mechanisms underlying Treg cell differentiation and provide theoretical foundations for targeted therapies aimed at enhancing Treg cells for asthma management.
{"title":"SIRT3-SUMO regulated Treg cell differentiation and asthma development by mediating N-glycosylation through the FAO pathway.","authors":"Yan Hu, Chuntao Liu","doi":"10.1007/s10565-025-10105-8","DOIUrl":"10.1007/s10565-025-10105-8","url":null,"abstract":"<p><p>Asthma, a chronic inflammatory disease, has a high disability rate, which greatly increases the disease burden. T cells are pivotal in the pathogenesis of asthma, and Treg cells, due to their role in maintaining immune system balance, represent a promising avenue for therapeutic intervention. Initial weighted correlation network analysis (WGCNA) analysis of asthma-related datasets indicates that N-glycosylation plays a critical role in asthma development. The establishment of an OVA-sensitized asthma model, along with the isolation of naive CD4<sup>+</sup> T cells and subsequent in vitro induction of Treg cell differentiation, further underscores the significance of N-glycosylation in the Treg cell differentiation of asthma-related Treg cells. Employing immunofluorescence, flow cytometry, and Western blot techniques revealed that SIRT3-SUMO is instrumental in regulating N-glycosylation-mediated Treg cells development. Mechanistically, overexpression and deSUMOylation of SIRT3 enhance the expression levels of CPT1 and VLCAD to promote fatty acid oxidation (FAO), thereby increasing intracellular acetyl-CoA concentrations. Acetyl-CoA subsequently facilitates the synthesis of N-glycosylation substrates via the hexosamine biosynthetic pathway (HBP), promoting Treg cell differentiation. Ultimately, our in vivo experiments demonstrate that SIRT3-SUMO modulates asthma progression by influencing Treg cells differentiation; thus, augmenting Treg cells populations can inhibit Th2-type and non-Th2-type asthmatic developments. These findings elucidate mechanisms underlying Treg cell differentiation and provide theoretical foundations for targeted therapies aimed at enhancing Treg cells for asthma management.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"164"},"PeriodicalIF":5.9,"publicationDate":"2025-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12700947/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145741345","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-11DOI: 10.1007/s10565-025-10101-y
Francesca Zuppini, Lucia Renzullo, Francesca Tornatore, Pietro Poggio, Mara Brancaccio
Cells respond to a variety of environmental stressors, including oxidative stress, nutrient deprivation, hypoxia and pathogenic invasion, which challenge cellular homeostasis and trigger adaptive responses. One of the first and most conserved effects is the activation of the heat shock response, which induces the transcription of heat shock proteins (HSPs), molecular chaperones involved in protein folding, assembly and turnover. Beyond their canonical role in maintaining proteostasis, HSPs also exert housekeeping functions, including endocytosis, a process essential for molecule internalization, nutrient uptake, receptor recycling, membrane turnover and cell migration. In this review, we explore the emerging roles of chaperone proteins in endocytic trafficking, with a particular focus on HSP90, HSP70 and small HSPs. We also highlight open questions like their attitude to act in cooperation or competition, and their propensity to form dynamics complexes. In addition, we discuss evidence suggesting that the involvement of these chaperones renders the endocytic process sensitive to stress, speculating on the role of HSPs in endocytosis as an integral component of the cellular stress response. Although some of the molecular mechanisms are still unclear, the available data reveal promising and interesting directions for further research.
{"title":"Heat shock proteins at the crossroads of endosomal trafficking pathways.","authors":"Francesca Zuppini, Lucia Renzullo, Francesca Tornatore, Pietro Poggio, Mara Brancaccio","doi":"10.1007/s10565-025-10101-y","DOIUrl":"10.1007/s10565-025-10101-y","url":null,"abstract":"<p><p>Cells respond to a variety of environmental stressors, including oxidative stress, nutrient deprivation, hypoxia and pathogenic invasion, which challenge cellular homeostasis and trigger adaptive responses. One of the first and most conserved effects is the activation of the heat shock response, which induces the transcription of heat shock proteins (HSPs), molecular chaperones involved in protein folding, assembly and turnover. Beyond their canonical role in maintaining proteostasis, HSPs also exert housekeeping functions, including endocytosis, a process essential for molecule internalization, nutrient uptake, receptor recycling, membrane turnover and cell migration. In this review, we explore the emerging roles of chaperone proteins in endocytic trafficking, with a particular focus on HSP90, HSP70 and small HSPs. We also highlight open questions like their attitude to act in cooperation or competition, and their propensity to form dynamics complexes. In addition, we discuss evidence suggesting that the involvement of these chaperones renders the endocytic process sensitive to stress, speculating on the role of HSPs in endocytosis as an integral component of the cellular stress response. Although some of the molecular mechanisms are still unclear, the available data reveal promising and interesting directions for further research.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"162"},"PeriodicalIF":5.9,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12696064/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145721118","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-11DOI: 10.1007/s10565-025-10112-9
Yuxiao Cai, Hongyu Zhang, Xinying Wang, Mengtong Liu, Rui Jin, Tianyu Zeng, Jie Ju
ANoptosis represents a unique form of programmed cell death that amalgamates the core characteristics of pyroptosis, apoptosis, and necroptosis, yet it constitutes a distinct pathway beyond the scope of any single one of them. During pathogen infection, PANoptosis is regulated by multi-protein complexes called PANoptosomes, which sense specific stimuli, including viral, bacterial, or fungal components, ultimately resulting in PANoptosis. This process contributes to pathogen clearance as well as excessive inflammatory response. Additionally, in some diseases such as renal damage, myocardial disease, nervous system diseases, and various cancers, PANoptosis influences disease occurrence, progression, and treatment outcomes. Targeting PANoptosis represents a promising way to enhance immune responses and achieve disease treatment in systemic disorder. This review synthesizes the current state of knowledge regarding the molecular mechanisms underlying PANoptosis in pathogen infection and systemic diseases, highlighting its role in pathological processes.
{"title":"Emerging role of PANoptosis in pathogen infection and systemic diseases.","authors":"Yuxiao Cai, Hongyu Zhang, Xinying Wang, Mengtong Liu, Rui Jin, Tianyu Zeng, Jie Ju","doi":"10.1007/s10565-025-10112-9","DOIUrl":"10.1007/s10565-025-10112-9","url":null,"abstract":"<p><p>ANoptosis represents a unique form of programmed cell death that amalgamates the core characteristics of pyroptosis, apoptosis, and necroptosis, yet it constitutes a distinct pathway beyond the scope of any single one of them. During pathogen infection, PANoptosis is regulated by multi-protein complexes called PANoptosomes, which sense specific stimuli, including viral, bacterial, or fungal components, ultimately resulting in PANoptosis. This process contributes to pathogen clearance as well as excessive inflammatory response. Additionally, in some diseases such as renal damage, myocardial disease, nervous system diseases, and various cancers, PANoptosis influences disease occurrence, progression, and treatment outcomes. Targeting PANoptosis represents a promising way to enhance immune responses and achieve disease treatment in systemic disorder. This review synthesizes the current state of knowledge regarding the molecular mechanisms underlying PANoptosis in pathogen infection and systemic diseases, highlighting its role in pathological processes.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"163"},"PeriodicalIF":5.9,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698769/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145721164","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-11DOI: 10.1007/s10565-025-10117-4
Shuoqi Li, Tiezhu Chen, Xiongjie Shen, Wanying Su, Xiaosheng Li
Traditional Chinese medicine is gaining prominence in lumbar disc herniation (LDH) management, but the mechanisms of its active compounds and their molecular targets remain largely unclear. Herein, we aim to elucidate the therapeutic mechanism of Gentisic acid by investigating its role in regulating S100A9 in LDH. Clinical analysis reveals that S100A9 expression and inflammatory levels correlat positively with LDH severity. S100A9 is found to promote M1 macrophage polarization and impair dorsal root ganglion (DRG) neuronal activity. Mechanistically, Gentisic acid binds to MAPK14, downregulates S100A9 via MAPK14, and then suppresses M1 polarization, enhances neuronal autophagic flux, and improves neuronal viability through the S100A9/Rac1/2 pathway. In vivo experiments demonstrate that Gentisic acid ameliorates disc injury, improves neurological function, and alleviates pain in a rat LDH model, with efficacy comparable to celecoxib. These results suggest that Gentisic acid could alleviate LDH symptoms by modulating macrophage polarization and autophagy through the MAPK14/S100A9/Rac1/2 axis, offering a promising therapeutic strategy for LDH.
{"title":"Gentisic acid ameliorates lumbar disc herniation by regulating M1/M2 Polarization via the MAPK14/S100A9/Rac1/2 pathway.","authors":"Shuoqi Li, Tiezhu Chen, Xiongjie Shen, Wanying Su, Xiaosheng Li","doi":"10.1007/s10565-025-10117-4","DOIUrl":"10.1007/s10565-025-10117-4","url":null,"abstract":"<p><p>Traditional Chinese medicine is gaining prominence in lumbar disc herniation (LDH) management, but the mechanisms of its active compounds and their molecular targets remain largely unclear. Herein, we aim to elucidate the therapeutic mechanism of Gentisic acid by investigating its role in regulating S100A9 in LDH. Clinical analysis reveals that S100A9 expression and inflammatory levels correlat positively with LDH severity. S100A9 is found to promote M1 macrophage polarization and impair dorsal root ganglion (DRG) neuronal activity. Mechanistically, Gentisic acid binds to MAPK14, downregulates S100A9 via MAPK14, and then suppresses M1 polarization, enhances neuronal autophagic flux, and improves neuronal viability through the S100A9/Rac1/2 pathway. In vivo experiments demonstrate that Gentisic acid ameliorates disc injury, improves neurological function, and alleviates pain in a rat LDH model, with efficacy comparable to celecoxib. These results suggest that Gentisic acid could alleviate LDH symptoms by modulating macrophage polarization and autophagy through the MAPK14/S100A9/Rac1/2 axis, offering a promising therapeutic strategy for LDH.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":"41 1","pages":"161"},"PeriodicalIF":5.9,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12696013/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145721132","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-10DOI: 10.1007/s10565-025-10128-1
Yushu Huang, He He, Xueqing Xia, Lifang Jiang, Qifeng Peng, Qingfeng Du, Yanting You
This study aims to explore biomarkers linked to the progression from non-alcoholic fatty liver disease (NAFLD) to hepatocellular carcinoma (HCC) and their therapeutic potential. Using bioinformatics, we identified key differentially expressed genes from various databases, focusing on genes related to NAFLD, non-alcoholic steatohepatitis, liver cirrhosis, and HCC. Among the upregulated genes, baculoviral IAP repeat containing 5 (BIRC5), cyclin B1 (CCNB1), cyclin-dependent kinase 1 (CDK1), and DNA topoisomerase II alpha (TOP2A) were found to be significant. In vivo and in vitro models of NAFLD and clinical HCC samples validated BIRC5 as a critical regulator in the disease progression. Functional assays revealed that knocking down BIRC5 alleviated fatty acid-induced liver damage and mitochondrial dysfunction in NAFLD models, while also inhibiting HCC cells proliferation and migration, further leading to mitochondrial dysfunction. YM155 (a specific BIRC5 inhibitor) also confirmed the previous experimental results. Then we performed experiments using BIRC5 overexpression plasmid. BIRC5 overexpression exacerbated hepatic steatosis and mitochondrial function in free fatty acid (FFA) -induced AML12 hepatocytes, and enhanced HCC cells proliferation, migration, and invasion. These findings highlight BIRC5 as a pivotal driver in the NAFLD-HCC transition, mediating metabolic dysfunction and malignant transformation. This study proposes BIRC5 as a therapeutic target and diagnostic biomarker, offering perspectives for HCC diagnosis and treatment of HCC. These results underscore the importance of BIRC5 in halting NAFLD-HCC progression and provide valuable insights for future clinical applications.
{"title":"From bioinformatics to clinical translation: BIRC5 as a pivotal diagnostic biomarker and therapeutic target for NAFLD-driven HCC.","authors":"Yushu Huang, He He, Xueqing Xia, Lifang Jiang, Qifeng Peng, Qingfeng Du, Yanting You","doi":"10.1007/s10565-025-10128-1","DOIUrl":"https://doi.org/10.1007/s10565-025-10128-1","url":null,"abstract":"<p><p>This study aims to explore biomarkers linked to the progression from non-alcoholic fatty liver disease (NAFLD) to hepatocellular carcinoma (HCC) and their therapeutic potential. Using bioinformatics, we identified key differentially expressed genes from various databases, focusing on genes related to NAFLD, non-alcoholic steatohepatitis, liver cirrhosis, and HCC. Among the upregulated genes, baculoviral IAP repeat containing 5 (BIRC5), cyclin B1 (CCNB1), cyclin-dependent kinase 1 (CDK1), and DNA topoisomerase II alpha (TOP2A) were found to be significant. In vivo and in vitro models of NAFLD and clinical HCC samples validated BIRC5 as a critical regulator in the disease progression. Functional assays revealed that knocking down BIRC5 alleviated fatty acid-induced liver damage and mitochondrial dysfunction in NAFLD models, while also inhibiting HCC cells proliferation and migration, further leading to mitochondrial dysfunction. YM155 (a specific BIRC5 inhibitor) also confirmed the previous experimental results. Then we performed experiments using BIRC5 overexpression plasmid. BIRC5 overexpression exacerbated hepatic steatosis and mitochondrial function in free fatty acid (FFA) -induced AML12 hepatocytes, and enhanced HCC cells proliferation, migration, and invasion. These findings highlight BIRC5 as a pivotal driver in the NAFLD-HCC transition, mediating metabolic dysfunction and malignant transformation. This study proposes BIRC5 as a therapeutic target and diagnostic biomarker, offering perspectives for HCC diagnosis and treatment of HCC. These results underscore the importance of BIRC5 in halting NAFLD-HCC progression and provide valuable insights for future clinical applications.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":" ","pages":""},"PeriodicalIF":5.9,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145721112","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}
The potential adverse effects of zinc oxide nanoparticles (ZnONPs) on human reproductive health may arise from their increasing industrial and commercial applications. However, their effects on preimplantation embryonic development and the related molecular mechanisms are still not well understood. Here, we demonstrate that ZnONPs exposure exhibit toxicity to a critical developmental period in mice. We observed that sustained exposure to ZnONPs in vitro resulted in embryonic development arrest at the 2-cell stage. To identify the susceptible stage, we controlled experiments to treat embryos with ZnONPs in the different processes of early embryonic development and determined that ZnONPs mainly to affect 2-cell stage embryos. According to the RNA-seq and EU (5-ethynyl uridine) analysis, the transcriptional activity of minor ZGA genes increased in the late 2-cell embryos following ZnONPs exposure. Subsequently, we employed multi-omics assays, including CUT&Tag and ATAC-seq. We found that ZnONPs exposure led to increased enrichment of H3K27ac (Histone H3 acetylated lysine 27) in late 2-cell embryos and enhanced chromatin accessibility, which led to abnormal upregulation of minor zygotic genome activation (ZGA) genes. In addition, the direct occupancy of ZnONPs at H3K27ac modification sites was verified through pulldown and immunoprecipitation. In conclusion, our findings demonstrate that ZnONPs exposure disrupting minor ZGA by interfering with H3K27ac erasure on the embryonic genome and ultimately impairing the developmental potential of embryos.
{"title":"Exposure to zinc oxide nanoparticles inhibits preimplantation embryonic development by disrupting zygotic genome activation.","authors":"Xuemei Liu, Chenshi Li, Sicheng Zhang, Xiaoping Zhang, Ling Zhu, Bei Liu, Yanhua Zhao, Jiayu Huang, Jingyu Li, Shimeng Guo","doi":"10.1007/s10565-025-10127-2","DOIUrl":"10.1007/s10565-025-10127-2","url":null,"abstract":"<p><p>The potential adverse effects of zinc oxide nanoparticles (ZnONPs) on human reproductive health may arise from their increasing industrial and commercial applications. However, their effects on preimplantation embryonic development and the related molecular mechanisms are still not well understood. Here, we demonstrate that ZnONPs exposure exhibit toxicity to a critical developmental period in mice. We observed that sustained exposure to ZnONPs in vitro resulted in embryonic development arrest at the 2-cell stage. To identify the susceptible stage, we controlled experiments to treat embryos with ZnONPs in the different processes of early embryonic development and determined that ZnONPs mainly to affect 2-cell stage embryos. According to the RNA-seq and EU (5-ethynyl uridine) analysis, the transcriptional activity of minor ZGA genes increased in the late 2-cell embryos following ZnONPs exposure. Subsequently, we employed multi-omics assays, including CUT&Tag and ATAC-seq. We found that ZnONPs exposure led to increased enrichment of H3K27ac (Histone H3 acetylated lysine 27) in late 2-cell embryos and enhanced chromatin accessibility, which led to abnormal upregulation of minor zygotic genome activation (ZGA) genes. In addition, the direct occupancy of ZnONPs at H3K27ac modification sites was verified through pulldown and immunoprecipitation. In conclusion, our findings demonstrate that ZnONPs exposure disrupting minor ZGA by interfering with H3K27ac erasure on the embryonic genome and ultimately impairing the developmental potential of embryos.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":" ","pages":"13"},"PeriodicalIF":5.9,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12795883/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145721138","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}