Pub Date : 2025-12-11DOI: 10.1038/s44321-025-00353-w
Emmanouela Epeslidou,Julia S Scott,Bim de Klein,Jeremy Tan Cudia,Barbro Melgert,Stefan Prekovic
Human-driven environmental change continues to reshape global patterns of disease, as seen in past pollution-related respiratory crises. Microplastics, persistent synthetic polymer particles, have now emerged as a widespread airborne contaminant with growing relevance for lung health. Continuous inhalation exposure, particularly in indoor environments rich in synthetic fibers, raises concern about their contribution to respiratory disease. Epidemiological and experimental studies increasingly link microplastic exposure to lung cancer, asthma, chronic obstructive pulmonary disease, and pulmonary fibrosis, yet the underlying mechanisms remain poorly defined. This review integrates current evidence on how particle properties influence biological outcomes and outlines how different polymer types, sizes, and aging states affect lung cells through inflammation, oxidative stress, ferroptosis, epithelial-mesenchymal transition, and epigenetic change. Together, these findings suggest that microplastics may act as environmental modifiers that exacerbate disease progression. Recognizing their complex and persistent nature highlights the need for standardized exposure metrics, mechanistic research at realistic doses, and coordinated scientific and regulatory action.
{"title":"Microplastics as environmental modifiers of lung disease.","authors":"Emmanouela Epeslidou,Julia S Scott,Bim de Klein,Jeremy Tan Cudia,Barbro Melgert,Stefan Prekovic","doi":"10.1038/s44321-025-00353-w","DOIUrl":"https://doi.org/10.1038/s44321-025-00353-w","url":null,"abstract":"Human-driven environmental change continues to reshape global patterns of disease, as seen in past pollution-related respiratory crises. Microplastics, persistent synthetic polymer particles, have now emerged as a widespread airborne contaminant with growing relevance for lung health. Continuous inhalation exposure, particularly in indoor environments rich in synthetic fibers, raises concern about their contribution to respiratory disease. Epidemiological and experimental studies increasingly link microplastic exposure to lung cancer, asthma, chronic obstructive pulmonary disease, and pulmonary fibrosis, yet the underlying mechanisms remain poorly defined. This review integrates current evidence on how particle properties influence biological outcomes and outlines how different polymer types, sizes, and aging states affect lung cells through inflammation, oxidative stress, ferroptosis, epithelial-mesenchymal transition, and epigenetic change. Together, these findings suggest that microplastics may act as environmental modifiers that exacerbate disease progression. Recognizing their complex and persistent nature highlights the need for standardized exposure metrics, mechanistic research at realistic doses, and coordinated scientific and regulatory action.","PeriodicalId":11597,"journal":{"name":"EMBO Molecular Medicine","volume":"15 1","pages":""},"PeriodicalIF":11.1,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145728592","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-11DOI: 10.1038/s44321-025-00343-y
Mark E Pepin,Xuemin Gong,Almut Schulze,Johannes Backs
Metabolic adaptations are a functional requirement for the heart to accommodate its broad range of physiologic operating conditions. It is increasingly recognized that persistent and exaggerated metabolic alterations precede adverse cardiac remodeling leading to heart failure. These metabolic shifts are coupled with changes in cardiac gene expression, driven in part by chromatin-modifying enzymes, which have recently been identified as both sensors and transducers of metabolic stress and gene regulatory networks, respectively. This review synthesizes the latest evidence implicating chromatin-modifying enzymes as key regulators of metabolic reprogramming in the heart, providing a framework to understand how metabolic stressors are incorporated as epigenetic modifications that regulate cardiac gene expression. We propose a model of 'metabo-epigenetic circuitry' within which energy metabolic perturbations drive transcriptional and epigenetic changes that ultimately contribute to cardiac dysfunction. Although many nodes in these circuits remain unidentified, this viewpoint opens new avenues for investigating chromatin-modifying enzymes as therapeutic targets to halt the metabolic programs that promote heart failure.
{"title":"Metabo-epigenetic circuits of heart failure: chromatin-modifying enzymes as determinants of metabolic plasticity.","authors":"Mark E Pepin,Xuemin Gong,Almut Schulze,Johannes Backs","doi":"10.1038/s44321-025-00343-y","DOIUrl":"https://doi.org/10.1038/s44321-025-00343-y","url":null,"abstract":"Metabolic adaptations are a functional requirement for the heart to accommodate its broad range of physiologic operating conditions. It is increasingly recognized that persistent and exaggerated metabolic alterations precede adverse cardiac remodeling leading to heart failure. These metabolic shifts are coupled with changes in cardiac gene expression, driven in part by chromatin-modifying enzymes, which have recently been identified as both sensors and transducers of metabolic stress and gene regulatory networks, respectively. This review synthesizes the latest evidence implicating chromatin-modifying enzymes as key regulators of metabolic reprogramming in the heart, providing a framework to understand how metabolic stressors are incorporated as epigenetic modifications that regulate cardiac gene expression. We propose a model of 'metabo-epigenetic circuitry' within which energy metabolic perturbations drive transcriptional and epigenetic changes that ultimately contribute to cardiac dysfunction. Although many nodes in these circuits remain unidentified, this viewpoint opens new avenues for investigating chromatin-modifying enzymes as therapeutic targets to halt the metabolic programs that promote heart failure.","PeriodicalId":11597,"journal":{"name":"EMBO Molecular Medicine","volume":"152 1","pages":""},"PeriodicalIF":11.1,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145728547","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-11DOI: 10.1038/s44321-025-00345-w
Coraline Heron, Theo Lemarcis, Océane Laguerre, Bénjamin Bourgeois, Corentin Thuilliez, Chloé Valentin, Anais Dumesnil, Manon Valet, David Godefroy, Damien Schapman, Gaetan Riou, Sophie Candon, Céline Derambure, Alma Zernecke, Caroline Berard, Hélène Dauchel, Virginie Tardif, Ebba Brakenhielm
Cardiac lymphatic alterations and insufficient lymphatic drainage have been found in cardiovascular diseases (CVDs). To unravel the mechanisms underlying lymphatic dysfunction, we applied single-cell (sc) analyses in murine heart failure (HF) models. Transaortic constriction (TAC) in C57BL/6J and BALB/c mice modeled chronic pressure -overload-induced cardiac hypertrophy and HF, respectively. Cardiac lymphatic (LEC) and blood vascular endothelial cells (BEC) were analyzed by scRNAseq, and targets validated by immunohistochemistry and human LEC cultures. While LEC profiles were comparable between strains in healthy mice, we found expansion of lymphatic capillaries and loss of valves post-TAC only in BALB/c. Differentially expressed gene (DEG) analysis revealed a reduction post-TAC only in BALB/c of lymphatic junctional components. Conversely, LEC expression of immune cell cross-talk mediators was mostly preserved post-TAC. Interestingly, around 35% of DEGs identified in cardiac LECs post-TAC were similarly altered in interleukin (IL)1β-stimulated human LECs. In conclusion, loss of lymphatic valves and dysregulated lymphatic barrier may underlie poor drainage capacity during pressure-overload-induced HF, despite potent lymphangiogenesis and preserved LEC immune attraction. Our work provides tractable targets to restore lymphatic health in CVDs.
{"title":"Molecular determinants of cardiac lymphatic dysfunction in a chronic pressure-overload model.","authors":"Coraline Heron, Theo Lemarcis, Océane Laguerre, Bénjamin Bourgeois, Corentin Thuilliez, Chloé Valentin, Anais Dumesnil, Manon Valet, David Godefroy, Damien Schapman, Gaetan Riou, Sophie Candon, Céline Derambure, Alma Zernecke, Caroline Berard, Hélène Dauchel, Virginie Tardif, Ebba Brakenhielm","doi":"10.1038/s44321-025-00345-w","DOIUrl":"https://doi.org/10.1038/s44321-025-00345-w","url":null,"abstract":"<p><p>Cardiac lymphatic alterations and insufficient lymphatic drainage have been found in cardiovascular diseases (CVDs). To unravel the mechanisms underlying lymphatic dysfunction, we applied single-cell (sc) analyses in murine heart failure (HF) models. Transaortic constriction (TAC) in C57BL/6J and BALB/c mice modeled chronic pressure -overload-induced cardiac hypertrophy and HF, respectively. Cardiac lymphatic (LEC) and blood vascular endothelial cells (BEC) were analyzed by scRNAseq, and targets validated by immunohistochemistry and human LEC cultures. While LEC profiles were comparable between strains in healthy mice, we found expansion of lymphatic capillaries and loss of valves post-TAC only in BALB/c. Differentially expressed gene (DEG) analysis revealed a reduction post-TAC only in BALB/c of lymphatic junctional components. Conversely, LEC expression of immune cell cross-talk mediators was mostly preserved post-TAC. Interestingly, around 35% of DEGs identified in cardiac LECs post-TAC were similarly altered in interleukin (IL)1β-stimulated human LECs. In conclusion, loss of lymphatic valves and dysregulated lymphatic barrier may underlie poor drainage capacity during pressure-overload-induced HF, despite potent lymphangiogenesis and preserved LEC immune attraction. Our work provides tractable targets to restore lymphatic health in CVDs.</p>","PeriodicalId":11597,"journal":{"name":"EMBO Molecular Medicine","volume":" ","pages":""},"PeriodicalIF":8.3,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145741572","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-09DOI: 10.1038/s44321-025-00350-z
Lennart Roos,Aida Garrido-Charles,Niels Albrecht,Anna Vavakou,Alexey Alekseev,Martina Bleyer,Anupriya Thirumalai,Artur Mittring,Theocharis Alvanos,Antoine T Huet,Ernst Bamberg,Kathrin Kusch,Bettina J Wolf,Tobias Moser,Thomas Mager
Optogenetics allows versatile control of excitable cell networks, which advances basic science research and drives the development of future medical applications. Fast-closing channelrhodopsins (ChRs) are required for high temporal fidelity of neurostimulation, but their short channel open times require sufficient plasma membrane expression and high light intensity, challenging clinical translation. Here, we addressed the need of high-rate neurostimulation by engineering optimized blue-light-sensitive ChR variants. In particular, we report on the ChR2 variant f-ChR2 TC enabling high frequency stimulation at low light requirements, due to its good plasma membrane targeted expression and balanced closing kinetics. Upon Adeno-associated virus (AAV) mediated f-ChR2 TC expression in spiral ganglion neurons of the inner ear in mice, f-ChR2 TC accordingly enabled optogenetic stimulation of the auditory nerve with sizeable responses beyond 300 Hz and low pulse energy thresholds. Translating the approach to the larger cochlea of gerbils, we tested the utility of f-ChR2 TC for evaluating multichannel optical cochlear implants with blue light emitting diodes and found light-efficient stimulation of the auditory pathway by single LEDs at rates ≥100 Hz.
{"title":"Channelrhodopsin variants for high-rate optogenetic neurostimulation at low light intensities.","authors":"Lennart Roos,Aida Garrido-Charles,Niels Albrecht,Anna Vavakou,Alexey Alekseev,Martina Bleyer,Anupriya Thirumalai,Artur Mittring,Theocharis Alvanos,Antoine T Huet,Ernst Bamberg,Kathrin Kusch,Bettina J Wolf,Tobias Moser,Thomas Mager","doi":"10.1038/s44321-025-00350-z","DOIUrl":"https://doi.org/10.1038/s44321-025-00350-z","url":null,"abstract":"Optogenetics allows versatile control of excitable cell networks, which advances basic science research and drives the development of future medical applications. Fast-closing channelrhodopsins (ChRs) are required for high temporal fidelity of neurostimulation, but their short channel open times require sufficient plasma membrane expression and high light intensity, challenging clinical translation. Here, we addressed the need of high-rate neurostimulation by engineering optimized blue-light-sensitive ChR variants. In particular, we report on the ChR2 variant f-ChR2 TC enabling high frequency stimulation at low light requirements, due to its good plasma membrane targeted expression and balanced closing kinetics. Upon Adeno-associated virus (AAV) mediated f-ChR2 TC expression in spiral ganglion neurons of the inner ear in mice, f-ChR2 TC accordingly enabled optogenetic stimulation of the auditory nerve with sizeable responses beyond 300 Hz and low pulse energy thresholds. Translating the approach to the larger cochlea of gerbils, we tested the utility of f-ChR2 TC for evaluating multichannel optical cochlear implants with blue light emitting diodes and found light-efficient stimulation of the auditory pathway by single LEDs at rates ≥100 Hz.","PeriodicalId":11597,"journal":{"name":"EMBO Molecular Medicine","volume":"12 1","pages":""},"PeriodicalIF":11.1,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145710901","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-09DOI: 10.1038/s44321-025-00357-6
Suresh Chava,Suresh Bugide,Parmanand Malvi,Kelly D DeMarco,Boyang Ma,Chaitanya N Parikh,Marcus Ruscetti,Allan Zajac,Guoping Cai,Romi Gupta,Narendra Wajapeyee
Natural Killer (NK) cells play a critical role in regulating tumor growth, but our understanding of the mechanisms underlying their anti-tumor activity remains limited. We identified the histone methyltransferase EHMT2 as a key suppressor of NK cell-mediated cytotoxicity. EHMT2 inhibition in cancer cells enhanced NK cell-mediated elimination of diverse cancers, including uveal melanoma, breast cancer, and pancreatic cancer. EHMT2 loss increased AZGP1 and decreased TGF-β1 levels, resulting in the autocrine elevation of NKG2D ligands MICB and ULBP3, chemokines in cancer cells, and the paracrine stimulation of NK cell function. In a syngeneic pancreatic cancer model, EHMT2 inhibition suppressed tumors in an NK cell-dependent manner, as NK cell depletion restored tumor growth. This effect persisted and remained dependent on NK cells in Rag2 knockout mice (lacking T and B cells), but not in NSG mice (lacking T-, B- and NK-cells). Furthermore, EHMT2 and TGF-β1 inhibitors suppressed tumors in immunocompetent, but not in immunodeficient mice. These findings establish EHMT2 as a suppressor of NK cell-mediated anti-tumor immunity and a promising therapeutic target.
{"title":"Loss of EHMT2 enhances NK cell-driven anti-tumor immunity through TGF-β1 suppression.","authors":"Suresh Chava,Suresh Bugide,Parmanand Malvi,Kelly D DeMarco,Boyang Ma,Chaitanya N Parikh,Marcus Ruscetti,Allan Zajac,Guoping Cai,Romi Gupta,Narendra Wajapeyee","doi":"10.1038/s44321-025-00357-6","DOIUrl":"https://doi.org/10.1038/s44321-025-00357-6","url":null,"abstract":"Natural Killer (NK) cells play a critical role in regulating tumor growth, but our understanding of the mechanisms underlying their anti-tumor activity remains limited. We identified the histone methyltransferase EHMT2 as a key suppressor of NK cell-mediated cytotoxicity. EHMT2 inhibition in cancer cells enhanced NK cell-mediated elimination of diverse cancers, including uveal melanoma, breast cancer, and pancreatic cancer. EHMT2 loss increased AZGP1 and decreased TGF-β1 levels, resulting in the autocrine elevation of NKG2D ligands MICB and ULBP3, chemokines in cancer cells, and the paracrine stimulation of NK cell function. In a syngeneic pancreatic cancer model, EHMT2 inhibition suppressed tumors in an NK cell-dependent manner, as NK cell depletion restored tumor growth. This effect persisted and remained dependent on NK cells in Rag2 knockout mice (lacking T and B cells), but not in NSG mice (lacking T-, B- and NK-cells). Furthermore, EHMT2 and TGF-β1 inhibitors suppressed tumors in immunocompetent, but not in immunodeficient mice. These findings establish EHMT2 as a suppressor of NK cell-mediated anti-tumor immunity and a promising therapeutic target.","PeriodicalId":11597,"journal":{"name":"EMBO Molecular Medicine","volume":"6 1","pages":""},"PeriodicalIF":11.1,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145710900","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Relapse in melanoma after targeted or immune therapy necessitates the rapid identification of effective alternatives. To address this gap, we investigated whether the timely generation of preclinical models for functional drug testing could reveal additional therapeutic options. Our study focused on: (i) the feasibility of generating in vivo and in vitro models from melanoma lymph node (LN)-derived disseminated cancer cells (DCCs) before relapse, (ii) the implementation of preclinical models to identify therapeutic alternatives, and (iii) the ability to detect patients who could benefit from early functional in vitro drug testing. Successful model generation was significantly associated with DCC quantity, LN origin, and mortality risk. All patient-derived xenograft models were available before patient death and, in 82% of cases, before relapse. Proof-of-concept in vitro drug screening using 315 anti-cancer drugs identified additional candidates, and coculture of DCCs and LN cells revealed specific T-cell activation and responses to immunotherapy. Our data establish a process for selecting melanoma patients at high risk of progression, enabling the timely generation of patient-derived models to support functionally guided treatment decisions at relapse.
{"title":"Micrometastasis-derived models enable drug testing for early-stage, high-risk melanoma patients.","authors":"Kathrin Weidele,Christian Werno,Steffi Treitschke,Catherine Botteron,Martin Hoffmann,Sebastian Scheitler,Lukas Wöhrl,Zbigniew Czyz,Giancarlo Feliciello,Florian Weber,Adithi Ravikumar Varadarajan,Jens Warfsmann,Silvia Materna-Reichelt,Marie Katzer,Laura Schreieder,Parvaneh Mohammadi,Hedayatollah Hosseini,Kamran Honarnejad,Sebastian Haferkamp,Melanie Werner-Klein,Christoph A Klein","doi":"10.1038/s44321-025-00339-8","DOIUrl":"https://doi.org/10.1038/s44321-025-00339-8","url":null,"abstract":"Relapse in melanoma after targeted or immune therapy necessitates the rapid identification of effective alternatives. To address this gap, we investigated whether the timely generation of preclinical models for functional drug testing could reveal additional therapeutic options. Our study focused on: (i) the feasibility of generating in vivo and in vitro models from melanoma lymph node (LN)-derived disseminated cancer cells (DCCs) before relapse, (ii) the implementation of preclinical models to identify therapeutic alternatives, and (iii) the ability to detect patients who could benefit from early functional in vitro drug testing. Successful model generation was significantly associated with DCC quantity, LN origin, and mortality risk. All patient-derived xenograft models were available before patient death and, in 82% of cases, before relapse. Proof-of-concept in vitro drug screening using 315 anti-cancer drugs identified additional candidates, and coculture of DCCs and LN cells revealed specific T-cell activation and responses to immunotherapy. Our data establish a process for selecting melanoma patients at high risk of progression, enabling the timely generation of patient-derived models to support functionally guided treatment decisions at relapse.","PeriodicalId":11597,"journal":{"name":"EMBO Molecular Medicine","volume":"7 1","pages":""},"PeriodicalIF":11.1,"publicationDate":"2025-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145680609","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Circulating blood proteomics enables minimally invasive biomarker discovery. Nanoparticle-based circulating plasma proteomics studies have reported varying number of proteins (ca 2000-7000), but it remains unclear whether a higher protein number is more informative. Here, we first develop OmniProt-a silica-nanoparticle workflow optimized through a systematic evaluation of nanoparticle types and protein corona formation parameters. Next, we present an Astral spectral library for 10,109 protein groups. Using the Astral with 60 sample-per-day throughput, OmniProt identifies ca 3000 to 6000 protein groups from human plasma. Platelet/erythrocyte/coagulation-related contamination artificially inflates protein identifications and compromises quantification accuracy in nanoparticle-enriched samples. Through controlled contamination experiments, we identified biomarkers for platelet/erythrocyte/coagulation-related contamination in nanoparticle-based plasma proteomics. We developed open-access software Baize for contamination assessment. We validated the pipeline in 193 patients with CT-indistinct benign nodules or early-stage lung cancers, flagging five contaminated samples. This study reveals that contamination alters protein identification/quantification in nanoparticle-based plasma proteomics and presents Baize software to evaluate it.
{"title":"Systematic evaluation of blood contamination in nanoparticle-based plasma proteomics.","authors":"Huanhuan Gao, Yuecheng Zhan, Yuanqi Liu, Zhiyi Zhu, Yuxiu Zheng, Liqin Qian, Zhangzhi Xue, Honghan Cheng, Zongxiang Nie, Weigang Ge, Senlin Ruan, Jiaxu Liu, Jikai Zhang, Yingying Sun, Lei Zhou, Dongyue Xun, Yingrui Wang, Heyun Xu, Huiwen Miao, Yi Zhu, Tiannan Guo","doi":"10.1038/s44321-025-00346-9","DOIUrl":"https://doi.org/10.1038/s44321-025-00346-9","url":null,"abstract":"<p><p>Circulating blood proteomics enables minimally invasive biomarker discovery. Nanoparticle-based circulating plasma proteomics studies have reported varying number of proteins (ca 2000-7000), but it remains unclear whether a higher protein number is more informative. Here, we first develop OmniProt-a silica-nanoparticle workflow optimized through a systematic evaluation of nanoparticle types and protein corona formation parameters. Next, we present an Astral spectral library for 10,109 protein groups. Using the Astral with 60 sample-per-day throughput, OmniProt identifies ca 3000 to 6000 protein groups from human plasma. Platelet/erythrocyte/coagulation-related contamination artificially inflates protein identifications and compromises quantification accuracy in nanoparticle-enriched samples. Through controlled contamination experiments, we identified biomarkers for platelet/erythrocyte/coagulation-related contamination in nanoparticle-based plasma proteomics. We developed open-access software Baize for contamination assessment. We validated the pipeline in 193 patients with CT-indistinct benign nodules or early-stage lung cancers, flagging five contaminated samples. This study reveals that contamination alters protein identification/quantification in nanoparticle-based plasma proteomics and presents Baize software to evaluate it.</p>","PeriodicalId":11597,"journal":{"name":"EMBO Molecular Medicine","volume":" ","pages":""},"PeriodicalIF":8.3,"publicationDate":"2025-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145687179","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01DOI: 10.1038/s44321-025-00347-8
Salma Srour, Francesca K Brown, James W Sheffield, Mohamed ElGhazaly, Daniel O'Connor, Malick M Gibani, Thomas C Darton, Andrew J Pollard, Mark O Collins, Daniel Humphreys
Salmonella Typhi secretes typhoid toxin that activates cellular DNA damage responses (DDR) during acute typhoid fever. Human infection challenge studies revealed that the toxin suppresses bacteraemia via unknown mechanisms. Using quantitative proteomic analysis on the plasma of bacteraemic participants, we demonstrate that wild-type toxigenic Salmonella induced secretion of lysozyme (LYZ) and apolipoprotein C3 (APOC3). Recombinant typhoid toxin or Salmonella infection recapitulated LYZ and APOC3 secretion in cultured cells, which involved ATM/ATR-dependent DDRs and confirmed observations in typhoid fever. LYZ caused spheroplast formation, inhibited the Salmonella type 3 secretion system, and intracellular infections. LYZ expression was regulated by p53 in a cell type-specific manner and driven by mitochondrial oxidative stress that caused nuclear DDRs and p53-mediated senescence responses. Addition of LYZ inhibited oxidative DNA damage and resulting senescence responses caused by typhoid toxin. Our findings may indicate that toxin-induced DDRs elicit antimicrobial responses, which suppress Salmonella bacteraemia during typhoid fever.
{"title":"Typhoid toxin of Salmonella Typhi elicits host antimicrobial response during acute typhoid fever.","authors":"Salma Srour, Francesca K Brown, James W Sheffield, Mohamed ElGhazaly, Daniel O'Connor, Malick M Gibani, Thomas C Darton, Andrew J Pollard, Mark O Collins, Daniel Humphreys","doi":"10.1038/s44321-025-00347-8","DOIUrl":"https://doi.org/10.1038/s44321-025-00347-8","url":null,"abstract":"<p><p>Salmonella Typhi secretes typhoid toxin that activates cellular DNA damage responses (DDR) during acute typhoid fever. Human infection challenge studies revealed that the toxin suppresses bacteraemia via unknown mechanisms. Using quantitative proteomic analysis on the plasma of bacteraemic participants, we demonstrate that wild-type toxigenic Salmonella induced secretion of lysozyme (LYZ) and apolipoprotein C3 (APOC3). Recombinant typhoid toxin or Salmonella infection recapitulated LYZ and APOC3 secretion in cultured cells, which involved ATM/ATR-dependent DDRs and confirmed observations in typhoid fever. LYZ caused spheroplast formation, inhibited the Salmonella type 3 secretion system, and intracellular infections. LYZ expression was regulated by p53 in a cell type-specific manner and driven by mitochondrial oxidative stress that caused nuclear DDRs and p53-mediated senescence responses. Addition of LYZ inhibited oxidative DNA damage and resulting senescence responses caused by typhoid toxin. Our findings may indicate that toxin-induced DDRs elicit antimicrobial responses, which suppress Salmonella bacteraemia during typhoid fever.</p>","PeriodicalId":11597,"journal":{"name":"EMBO Molecular Medicine","volume":" ","pages":""},"PeriodicalIF":8.3,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145653920","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-11-04DOI: 10.1038/s44321-025-00330-3
Christos Karampelias, Kaiyuan Yang, Falk J Farkas, Michael Sterr, Mireia Molina van Den Bosch, Simone Renner, Janina Fuß, Christine von Toerne, Sören Franzenburg, Tatsuya Kin, Eckhard Wolf, Elisabeth Kemter, Heiko Lickert
Primary human pancreatic ductal organoids (HPDO) have emerged as a model to study pancreas biology and model disease like pancreatitis and pancreatic cancer. Yet, donor material availability, genetic variability and a lack of extensive benchmarking to healthy and disease pancreas limits the range of applications. To address this gap, we established porcine pancreatic ductal organoids (PPDO) as a system from a reliable, genetically defined and easily obtainable source to model pancreatic ductal/progenitor biology. We benchmarked PPDO to HPDO and primary porcine pancreas using single-cell RNA sequencing (scRNA-Seq). We observed no overt phenotypic differences in PPDO derived from distinct developmental stages using extensive proteomics profiling, with a WNT/basal cell signaling enriched population characterizing PPDO. PPDO exhibited differentiation potential towards mature ductal cells and limited potential towards endocrine lineages. We used PPDO as a chemical screening platform to assess the safety of FDA-approved drugs and showed conserved toxicity of statins and α-adrenergic receptor inhibitors between PPDO and HPDO cultures. Overall, our results highlight the PPDO as a model for mammalian duct/progenitor applications.
{"title":"Benchmarking porcine pancreatic ductal organoids for drug screening applications.","authors":"Christos Karampelias, Kaiyuan Yang, Falk J Farkas, Michael Sterr, Mireia Molina van Den Bosch, Simone Renner, Janina Fuß, Christine von Toerne, Sören Franzenburg, Tatsuya Kin, Eckhard Wolf, Elisabeth Kemter, Heiko Lickert","doi":"10.1038/s44321-025-00330-3","DOIUrl":"10.1038/s44321-025-00330-3","url":null,"abstract":"<p><p>Primary human pancreatic ductal organoids (HPDO) have emerged as a model to study pancreas biology and model disease like pancreatitis and pancreatic cancer. Yet, donor material availability, genetic variability and a lack of extensive benchmarking to healthy and disease pancreas limits the range of applications. To address this gap, we established porcine pancreatic ductal organoids (PPDO) as a system from a reliable, genetically defined and easily obtainable source to model pancreatic ductal/progenitor biology. We benchmarked PPDO to HPDO and primary porcine pancreas using single-cell RNA sequencing (scRNA-Seq). We observed no overt phenotypic differences in PPDO derived from distinct developmental stages using extensive proteomics profiling, with a WNT/basal cell signaling enriched population characterizing PPDO. PPDO exhibited differentiation potential towards mature ductal cells and limited potential towards endocrine lineages. We used PPDO as a chemical screening platform to assess the safety of FDA-approved drugs and showed conserved toxicity of statins and α-adrenergic receptor inhibitors between PPDO and HPDO cultures. Overall, our results highlight the PPDO as a model for mammalian duct/progenitor applications.</p>","PeriodicalId":11597,"journal":{"name":"EMBO Molecular Medicine","volume":" ","pages":"3657-3688"},"PeriodicalIF":8.3,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12686538/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145444474","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}