Pub Date : 2026-01-10DOI: 10.1016/j.celrep.2025.116810
Katarzyna Groborz, Marcin Poręba, Marcin Drąg, Guy Salvesen
Quantifying enzymatic activity at the single-cell level remains challenging. Building on previous development of metal-tagged activity-based probes (time-of-flight [TOF]-probes) compatible with mass cytometry, we adapt this technology to quantify individual proteases in immune cells. We optimize and validate TOF-probes targeting catalytically active cysteine and serine proteases in two primary immune populations: neutrophils and monocyte-derived macrophages. Using this platform, we observe striking shifts in cathepsin activity during monocyte-to-macrophage differentiation, along with a functional rewiring from apoptotic to pyroptotic protease programs. In neutrophils, unexpectedly, we detect high levels of active serine proteases even in the resting state, highlighting their potential role as pre-armed effector cells poised for rapid inflammatory responses. These findings lay the foundation for using TOF-probes to quantify active proteases in situ at the single-cell level and provide new insights into protease activation states across immune cells.
{"title":"TOF-probe-based mass cytometry reveals individual protease activity as an important driver of immune cell differentiation and function.","authors":"Katarzyna Groborz, Marcin Poręba, Marcin Drąg, Guy Salvesen","doi":"10.1016/j.celrep.2025.116810","DOIUrl":"https://doi.org/10.1016/j.celrep.2025.116810","url":null,"abstract":"<p><p>Quantifying enzymatic activity at the single-cell level remains challenging. Building on previous development of metal-tagged activity-based probes (time-of-flight [TOF]-probes) compatible with mass cytometry, we adapt this technology to quantify individual proteases in immune cells. We optimize and validate TOF-probes targeting catalytically active cysteine and serine proteases in two primary immune populations: neutrophils and monocyte-derived macrophages. Using this platform, we observe striking shifts in cathepsin activity during monocyte-to-macrophage differentiation, along with a functional rewiring from apoptotic to pyroptotic protease programs. In neutrophils, unexpectedly, we detect high levels of active serine proteases even in the resting state, highlighting their potential role as pre-armed effector cells poised for rapid inflammatory responses. These findings lay the foundation for using TOF-probes to quantify active proteases in situ at the single-cell level and provide new insights into protease activation states across immune cells.</p>","PeriodicalId":9798,"journal":{"name":"Cell reports","volume":"45 1","pages":"116810"},"PeriodicalIF":6.9,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951146","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
While high-fiber diets (HfiDs) promote weight loss, their long-term efficacy is limited by rapid weight regain upon returning to high-fat diets (HFDs). Using C57BL/6J mice in diet-switching paradigms, we characterized tissue-specific responses to HfiD-to-HFD transitions through single-nucleus and spatial transcriptomics. HfiD pre-feeding enhanced mesenteric white adipose tissue progenitor/adipocyte sensitivity to subsequent HFD exposure. In the intestine, HfiD prevented HFD-induced immune-enterocyte expansion in the duodenum and reversed the enterocyte-to-goblet cell shift in the colon while maintaining persistent epigenetic reprogramming. Although HfiD-induced microbiome changes were largely reversed by HFD, we identified sexually dimorphic remodeling of adipose cell populations during diet transitions. Our findings demonstrate that prior HfiD feeding fundamentally reprograms adipose and intestinal responses to subsequent HFD challenge, providing mechanistic insights into dietary intervention outcomes. This work establishes a spatiotemporal resource for understanding tissue plasticity during dietary changes, offering new perspectives for obesity management strategies.
{"title":"Prior high fiber intake impinges on the cellular responses of mesenteric adipose and intestinal tissues to subsequent high fat feeding.","authors":"Zhi Peng, Houyu Zhang, Yifei Ding, Zhen Liu, Meng Xie","doi":"10.1016/j.celrep.2025.116801","DOIUrl":"https://doi.org/10.1016/j.celrep.2025.116801","url":null,"abstract":"<p><p>While high-fiber diets (HfiDs) promote weight loss, their long-term efficacy is limited by rapid weight regain upon returning to high-fat diets (HFDs). Using C57BL/6J mice in diet-switching paradigms, we characterized tissue-specific responses to HfiD-to-HFD transitions through single-nucleus and spatial transcriptomics. HfiD pre-feeding enhanced mesenteric white adipose tissue progenitor/adipocyte sensitivity to subsequent HFD exposure. In the intestine, HfiD prevented HFD-induced immune-enterocyte expansion in the duodenum and reversed the enterocyte-to-goblet cell shift in the colon while maintaining persistent epigenetic reprogramming. Although HfiD-induced microbiome changes were largely reversed by HFD, we identified sexually dimorphic remodeling of adipose cell populations during diet transitions. Our findings demonstrate that prior HfiD feeding fundamentally reprograms adipose and intestinal responses to subsequent HFD challenge, providing mechanistic insights into dietary intervention outcomes. This work establishes a spatiotemporal resource for understanding tissue plasticity during dietary changes, offering new perspectives for obesity management strategies.</p>","PeriodicalId":9798,"journal":{"name":"Cell reports","volume":"45 1","pages":"116801"},"PeriodicalIF":6.9,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145950924","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-10DOI: 10.1016/j.celrep.2025.116808
Jie Fu, Xin Zong, Hong Zhang, Luoyi Zhu, Tao Gong, Yuanzhi Cheng, Fengqin Wang, Zeqing Lu, Caiqiao Zhang, Mingliang Jin, Yizhen Wang
NLRP3 inflammasome activation requires both transcriptional priming and complex assembly, but how RNA m6A methylation coordinates these steps remains unclear. Here, we show that m6A levels increase during macrophage NLRP3 inflammasome activation and that METTL3 loss suppresses this activation. Myeloid-specific Mettl3 knockout mice display reduced inflammation and improved metabolic outcomes in lipopolysaccharide (LPS)-induced sepsis, monosodium urate (MSU)-induced arthritis, and diet-induced obesity. Integrated chromatin-associated RNA sequencing (chrRNA-seq), kethoxal-assisted single-stranded DNA sequencing (KAS-seq), and chrRNA-methylated RNA immunoprecipitation (MeRIP)-seq analyses show that METTL3 installs m6A co-transcriptionally on nascent Jak1, Nlrp3, and Il1β RNAs and that METTL3 regulates dynamic transcription and chromatin accessibility while selectively maintaining Nlrp3/Il1β transcription. YTHDF1-driven translation of Jak1 activates the JAK1-STAT3-C/EBPβ axis to initiate Nlrp3/Il1β transcription, and m6A-YTHDF1 translation of Nlrp3/Il1β amplifies protein output, forming a coupled transcriptional-translational circuit. Pharmacologic STAT3 inhibition and METTL3 catalytic rescue validate this pathway and identify METTL3-mediated m6A as a therapeutic target for inflammasome-driven diseases.
NLRP3炎性小体激活需要转录启动和复合物组装,但RNA m6A甲基化如何协调这些步骤尚不清楚。本研究表明,在巨噬细胞NLRP3炎性体激活过程中,m6A水平升高,而METTL3缺失抑制了这种激活。髓系特异性Mettl3基因敲除小鼠在脂多糖(LPS)诱导的败血症、尿酸钠(MSU)诱导的关节炎和饮食诱导的肥胖中显示出炎症减少和代谢结果改善。综合染色质相关RNA测序(chrRNA-seq)、酮醇辅助单链DNA测序(ka -seq)和chrrna甲基化RNA免疫沉淀(MeRIP)-seq分析表明,METTL3将m6A共转录安装在新生的Jak1、Nlrp3和Il1β RNA上,并且METTL3调节动态转录和染色质可及性,同时选择性地维持Nlrp3/Il1β转录。ythdf1驱动的Jak1翻译激活Jak1 - stat3 - c /EBPβ轴启动Nlrp3/Il1β转录,m6A-YTHDF1翻译Nlrp3/Il1β放大蛋白输出,形成一个耦合的转录-翻译回路。药理学上STAT3抑制和METTL3催化挽救验证了这一途径,并确定了METTL3介导的m6A作为炎症小体驱动疾病的治疗靶点。
{"title":"METTL3-mediated m<sup>6</sup>A on nascent RNA coordinates translational and transcriptional programs to activate the NLRP3 inflammasome in macrophages.","authors":"Jie Fu, Xin Zong, Hong Zhang, Luoyi Zhu, Tao Gong, Yuanzhi Cheng, Fengqin Wang, Zeqing Lu, Caiqiao Zhang, Mingliang Jin, Yizhen Wang","doi":"10.1016/j.celrep.2025.116808","DOIUrl":"https://doi.org/10.1016/j.celrep.2025.116808","url":null,"abstract":"<p><p>NLRP3 inflammasome activation requires both transcriptional priming and complex assembly, but how RNA m<sup>6</sup>A methylation coordinates these steps remains unclear. Here, we show that m<sup>6</sup>A levels increase during macrophage NLRP3 inflammasome activation and that METTL3 loss suppresses this activation. Myeloid-specific Mettl3 knockout mice display reduced inflammation and improved metabolic outcomes in lipopolysaccharide (LPS)-induced sepsis, monosodium urate (MSU)-induced arthritis, and diet-induced obesity. Integrated chromatin-associated RNA sequencing (chrRNA-seq), kethoxal-assisted single-stranded DNA sequencing (KAS-seq), and chrRNA-methylated RNA immunoprecipitation (MeRIP)-seq analyses show that METTL3 installs m<sup>6</sup>A co-transcriptionally on nascent Jak1, Nlrp3, and Il1β RNAs and that METTL3 regulates dynamic transcription and chromatin accessibility while selectively maintaining Nlrp3/Il1β transcription. YTHDF1-driven translation of Jak1 activates the JAK1-STAT3-C/EBPβ axis to initiate Nlrp3/Il1β transcription, and m<sup>6</sup>A-YTHDF1 translation of Nlrp3/Il1β amplifies protein output, forming a coupled transcriptional-translational circuit. Pharmacologic STAT3 inhibition and METTL3 catalytic rescue validate this pathway and identify METTL3-mediated m<sup>6</sup>A as a therapeutic target for inflammasome-driven diseases.</p>","PeriodicalId":9798,"journal":{"name":"Cell reports","volume":"45 1","pages":"116808"},"PeriodicalIF":6.9,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951417","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ruminants thrive in diverse ecosystems by leveraging their rumen microbiome to ferment fibrous plants. However, the spatial biogeography of rumen microbiome and the genetic diversity of the ventral rumen epithelium remain unknown. Here, we present a multi-omics study in roe deer, sika deer, and sheep, integrating region-resolved microbiome and metabolome across 11 ruminal sacs, as well as single-cell RNA sequencing (scRNA-seq), assay for transposase-accessible chromatin using sequencing (ATAC-seq), and bulk RNA sequencing (RNA-seq) of ventral epithelium. We reveal species-specific microbial compositions and metabolic capacities contributing to differences in short-chain fatty acid and vitamin B production. We uncover functional divergence, genomic specialization, and metabolic changes across the microbiome of distinct ruminal sacs. Single-cell profiling reveals changes of immune responses and structural remodeling of the ruminal ventral epithelium. We demonstrate that vitamin B12 promotes epithelial growth and we identify genes enhancing stem cell differentiation. Our results highlight variation in microbial ecology and epithelial architecture among three ruminant species, offering insights to improve livestock productivity.
{"title":"Rumen microbiome biogeography and ventral epithelial architecture in three ruminant species.","authors":"Jianan Sang, Songze Li, Chao Xu, Xiangyu Pan, Yuhang Zhu, Yongxiang Li, Cuiliu Ma, Yunxi Zhang, Sibo Chen, Qiang Qiu, Huazhe Si, Zan Huang, Jun Wang, Jinzhen Jiao, Zhipeng Li","doi":"10.1016/j.celrep.2025.116779","DOIUrl":"https://doi.org/10.1016/j.celrep.2025.116779","url":null,"abstract":"<p><p>Ruminants thrive in diverse ecosystems by leveraging their rumen microbiome to ferment fibrous plants. However, the spatial biogeography of rumen microbiome and the genetic diversity of the ventral rumen epithelium remain unknown. Here, we present a multi-omics study in roe deer, sika deer, and sheep, integrating region-resolved microbiome and metabolome across 11 ruminal sacs, as well as single-cell RNA sequencing (scRNA-seq), assay for transposase-accessible chromatin using sequencing (ATAC-seq), and bulk RNA sequencing (RNA-seq) of ventral epithelium. We reveal species-specific microbial compositions and metabolic capacities contributing to differences in short-chain fatty acid and vitamin B production. We uncover functional divergence, genomic specialization, and metabolic changes across the microbiome of distinct ruminal sacs. Single-cell profiling reveals changes of immune responses and structural remodeling of the ruminal ventral epithelium. We demonstrate that vitamin B<sub>12</sub> promotes epithelial growth and we identify genes enhancing stem cell differentiation. Our results highlight variation in microbial ecology and epithelial architecture among three ruminant species, offering insights to improve livestock productivity.</p>","PeriodicalId":9798,"journal":{"name":"Cell reports","volume":"45 1","pages":"116779"},"PeriodicalIF":6.9,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951159","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Interferon regulatory factor 3 (IRF3) functions as a key transcription factor in the innate antiviral immune response, which depends on its nuclear localization. However, its function in the cytoplasm during non-infection states remains largely unknown. In this study, we found that resting cytoplasmic IRF3 interacts with hypoxia-inducible factor (HIF)-1α and HIF-2α, two master regulators of hypoxia signaling. This interaction retains HIF-α in the cytoplasm under hypoxic conditions, preventing it from exerting its transcription factor function and attenuating hypoxia signaling. Disruption of IRF3 in both mice and zebrafish resulted in increased expression of hypoxia response genes and enhanced tolerance to hypoxia. These findings suggest that, in the absence of viral infection, cytoplasmic IRF3 modulates hypoxia signaling by retaining HIF-α in the cytoplasm under hypoxic conditions.
{"title":"IRF3 attenuates hypoxia signaling by retaining HIF-α in the cytoplasm.","authors":"Hongyan Deng, Shuke Jia, Chunchun Zhu, Jiale Hua, Zixuan Wang, Xueyi Sun, Wen Liu, Liyun Shi, Wenhua Li, Jian-Fang Gui, Xing Liu, Wuhan Xiao","doi":"10.1016/j.celrep.2025.116815","DOIUrl":"https://doi.org/10.1016/j.celrep.2025.116815","url":null,"abstract":"<p><p>Interferon regulatory factor 3 (IRF3) functions as a key transcription factor in the innate antiviral immune response, which depends on its nuclear localization. However, its function in the cytoplasm during non-infection states remains largely unknown. In this study, we found that resting cytoplasmic IRF3 interacts with hypoxia-inducible factor (HIF)-1α and HIF-2α, two master regulators of hypoxia signaling. This interaction retains HIF-α in the cytoplasm under hypoxic conditions, preventing it from exerting its transcription factor function and attenuating hypoxia signaling. Disruption of IRF3 in both mice and zebrafish resulted in increased expression of hypoxia response genes and enhanced tolerance to hypoxia. These findings suggest that, in the absence of viral infection, cytoplasmic IRF3 modulates hypoxia signaling by retaining HIF-α in the cytoplasm under hypoxic conditions.</p>","PeriodicalId":9798,"journal":{"name":"Cell reports","volume":"45 1","pages":"116815"},"PeriodicalIF":6.9,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951420","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-09DOI: 10.1016/j.celrep.2025.116802
Derek H Janssens, Christine A Codomo, Dominik J Otto, Lev Silberstein, Kami Ahmad, Steve Henikoff
Promoter-proximal pausing of RNA polymerase II (Pol II) primes genes for rapid activation, yet how Pol II dynamics are temporally organized in adult stem cells to enable fast and flexible responses to environmental cues remain unknown. To address this, we developed sciCUT&Tag2in1 for joint profiling of Pol II and histone modifications in single cells. By profiling over 200,000 CD34+ hematopoietic stem cells (HSCs) and progenitors, we identify a Pol II regulatory cascade that directs the response to granulocyte colony-stimulating factor (G-CSF)-induced inflammatory stress. HSCs are activated by elevated Pol II occupancy and reduced Polycomb repression of immune response genes. Lineage commitment proceeds through sequential modes of Pol II activation, beginning with rapid pause-and-release genes, followed by slower initiate-and-release of Polycomb-repressed targets. sciCUT&Tag2in1 defines the temporal logic of how adult stem cells use paused Pol II to enable flexible lineage decisions, providing a powerful tool for studying the intersection of development, inflammation, and disease.
{"title":"Sequential RNA polymerase II activation drives human hematopoiesis.","authors":"Derek H Janssens, Christine A Codomo, Dominik J Otto, Lev Silberstein, Kami Ahmad, Steve Henikoff","doi":"10.1016/j.celrep.2025.116802","DOIUrl":"https://doi.org/10.1016/j.celrep.2025.116802","url":null,"abstract":"<p><p>Promoter-proximal pausing of RNA polymerase II (Pol II) primes genes for rapid activation, yet how Pol II dynamics are temporally organized in adult stem cells to enable fast and flexible responses to environmental cues remain unknown. To address this, we developed sciCUT&Tag2in1 for joint profiling of Pol II and histone modifications in single cells. By profiling over 200,000 CD34<sup>+</sup> hematopoietic stem cells (HSCs) and progenitors, we identify a Pol II regulatory cascade that directs the response to granulocyte colony-stimulating factor (G-CSF)-induced inflammatory stress. HSCs are activated by elevated Pol II occupancy and reduced Polycomb repression of immune response genes. Lineage commitment proceeds through sequential modes of Pol II activation, beginning with rapid pause-and-release genes, followed by slower initiate-and-release of Polycomb-repressed targets. sciCUT&Tag2in1 defines the temporal logic of how adult stem cells use paused Pol II to enable flexible lineage decisions, providing a powerful tool for studying the intersection of development, inflammation, and disease.</p>","PeriodicalId":9798,"journal":{"name":"Cell reports","volume":"45 1","pages":"116802"},"PeriodicalIF":6.9,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951157","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-09DOI: 10.1016/j.celrep.2025.116828
Conor W Templeton, Laimonis A Laimins
The levels of N6-methyladenosine (m6A)-modified RNAs are reduced within cells with high-risk human papillomaviruses (HPVs) in comparison to normal cells. These reduced m6A levels stabilize RNA:DNA hybrids (R-loops), whose regulation is critical for viral replication. HPV E6 induces the degradation of the m6A methyltransferases, Mettl3 and Mettl14, in an E6AP-dependent manner to reduce m6A levels, while the RNA helicase senataxin (SETX) counteracts this by activating the expression of m6A regulators. Maintenance of the residual m6A levels in undifferentiated cells is also critical for viral replication. A subset of early viral transcripts is m6A modified, and the inhibition of Mettl3 impairs viral gene expression. Upon differentiation, the levels of SETX and m6A are increased by over 8-fold, which is necessary for the productive replication of HPVs. These studies identify a role for SETX and E6 in regulating m6A deposition to control HPV pathogenesis in a differentiation-dependent manner.
{"title":"N(6)-methyladenosine modification of RNA is regulated by senataxin and E6 to control HPV replication.","authors":"Conor W Templeton, Laimonis A Laimins","doi":"10.1016/j.celrep.2025.116828","DOIUrl":"https://doi.org/10.1016/j.celrep.2025.116828","url":null,"abstract":"<p><p>The levels of N6-methyladenosine (m6A)-modified RNAs are reduced within cells with high-risk human papillomaviruses (HPVs) in comparison to normal cells. These reduced m6A levels stabilize RNA:DNA hybrids (R-loops), whose regulation is critical for viral replication. HPV E6 induces the degradation of the m6A methyltransferases, Mettl3 and Mettl14, in an E6AP-dependent manner to reduce m6A levels, while the RNA helicase senataxin (SETX) counteracts this by activating the expression of m6A regulators. Maintenance of the residual m6A levels in undifferentiated cells is also critical for viral replication. A subset of early viral transcripts is m6A modified, and the inhibition of Mettl3 impairs viral gene expression. Upon differentiation, the levels of SETX and m6A are increased by over 8-fold, which is necessary for the productive replication of HPVs. These studies identify a role for SETX and E6 in regulating m6A deposition to control HPV pathogenesis in a differentiation-dependent manner.</p>","PeriodicalId":9798,"journal":{"name":"Cell reports","volume":"45 1","pages":"116828"},"PeriodicalIF":6.9,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145948350","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-08DOI: 10.1016/j.celrep.2025.116784
Yurui Ma, Evon Poon, Chenchen Jin, Barbara Martins da Costa, Yuewei Xu, Sadiya Quazi, Nikolaos Zourdoumis, Chiharu Wickremesinghe, Louis Chesler, Hector C Keun, Anke Nijhuis
Precise control of pre-mRNA splicing is vital for transcriptome integrity, and its disruption is an emerging cancer vulnerability. Here, we use indisulam to degrade RBM39 and show that the clinical ALK inhibitor alectinib can be repurposed to inhibit SRPK1. Co-treatment of indisulam and alectinib inhibited cell proliferation, induced apoptosis, and caused cell-cycle arrest in multiple cancer cell lines, including MYCN-amplified high-risk neuroblastoma. RNA sequencing revealed enhanced splicing defects preferentially in DNA repair-related genes following combination treatment, leading to R-loop accumulation and increased DNA damage. In the Th-MYCN/ALKF1174L neuroblastoma mouse model, combination therapy induced complete tumor regression and significantly improved survival rates compared with monotherapies. These findings demonstrate that combining indisulam and alectinib is a promising approach to treating aggressive malignancies such as high-risk neuroblastoma, exploiting the untapped polypharmacology of alectinib as an RNA splicing inhibitor and supporting the therapeutic value of co-targeting interdependent splicing factors for synergistic benefit.
{"title":"Exploiting the polypharmacology of alectinib for synergistic RNA splicing disruption with RBM39 degraders.","authors":"Yurui Ma, Evon Poon, Chenchen Jin, Barbara Martins da Costa, Yuewei Xu, Sadiya Quazi, Nikolaos Zourdoumis, Chiharu Wickremesinghe, Louis Chesler, Hector C Keun, Anke Nijhuis","doi":"10.1016/j.celrep.2025.116784","DOIUrl":"https://doi.org/10.1016/j.celrep.2025.116784","url":null,"abstract":"<p><p>Precise control of pre-mRNA splicing is vital for transcriptome integrity, and its disruption is an emerging cancer vulnerability. Here, we use indisulam to degrade RBM39 and show that the clinical ALK inhibitor alectinib can be repurposed to inhibit SRPK1. Co-treatment of indisulam and alectinib inhibited cell proliferation, induced apoptosis, and caused cell-cycle arrest in multiple cancer cell lines, including MYCN-amplified high-risk neuroblastoma. RNA sequencing revealed enhanced splicing defects preferentially in DNA repair-related genes following combination treatment, leading to R-loop accumulation and increased DNA damage. In the Th-MYCN/ALK<sup>F1174L</sup> neuroblastoma mouse model, combination therapy induced complete tumor regression and significantly improved survival rates compared with monotherapies. These findings demonstrate that combining indisulam and alectinib is a promising approach to treating aggressive malignancies such as high-risk neuroblastoma, exploiting the untapped polypharmacology of alectinib as an RNA splicing inhibitor and supporting the therapeutic value of co-targeting interdependent splicing factors for synergistic benefit.</p>","PeriodicalId":9798,"journal":{"name":"Cell reports","volume":"45 1","pages":"116784"},"PeriodicalIF":6.9,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145948361","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-07DOI: 10.1016/j.celrep.2025.116803
Tingting Li, Hyeonwi Son, Vipin Arora, John Shannonhouse, Sinu Kumari, Dennis Chang, Michael Caterina, Yu Shin Kim, Man-Kyo Chung
The identities of primary afferents transducing mechanical allodynia following nerve injury remain unclear. We genetically label brushing-activated (BA) trigeminal ganglia (TG) neurons using FosCreER mice with trigeminal nerve injury (TNI). BA TG neurons are largely medium-sized. Many express neurofilament200 and Ntrk3, markers for low-threshold mechanoreceptors, with lower co-localization with nociceptor markers such as Calca or Trpv1. Chemogenetic inhibition of BA TG neurons reduces mechanical allodynia, whereas their chemogenetic activation increases spontaneous face wiping after TNI. Brushing-induced conditional knockdown (bcKD) of Piezo2 from BA TG afferents reduces punctate and dynamic mechanical allodynia. In vivo TG GCaMP Ca2+ imaging shows that Piezo2 bcKD reduces not only hypersensitivity to low-force mechanical stimulation, mostly among medium-sized neurons, but also, unexpectedly, TNI-induced spontaneous activity. Therefore, Fos is useful for genetic labeling and manipulation of BA TG neurons. Furthermore, innocuous mechanical stimuli activate multiple TG afferent subtypes after TNI, possibly accounting for the complexity of resulting painful symptoms.
{"title":"Genetic identification of mouse trigeminal afferents responsible for mechanical allodynia.","authors":"Tingting Li, Hyeonwi Son, Vipin Arora, John Shannonhouse, Sinu Kumari, Dennis Chang, Michael Caterina, Yu Shin Kim, Man-Kyo Chung","doi":"10.1016/j.celrep.2025.116803","DOIUrl":"https://doi.org/10.1016/j.celrep.2025.116803","url":null,"abstract":"<p><p>The identities of primary afferents transducing mechanical allodynia following nerve injury remain unclear. We genetically label brushing-activated (BA) trigeminal ganglia (TG) neurons using Fos<sup>CreER</sup> mice with trigeminal nerve injury (TNI). BA TG neurons are largely medium-sized. Many express neurofilament200 and Ntrk3, markers for low-threshold mechanoreceptors, with lower co-localization with nociceptor markers such as Calca or Trpv1. Chemogenetic inhibition of BA TG neurons reduces mechanical allodynia, whereas their chemogenetic activation increases spontaneous face wiping after TNI. Brushing-induced conditional knockdown (bcKD) of Piezo2 from BA TG afferents reduces punctate and dynamic mechanical allodynia. In vivo TG GCaMP Ca<sup>2+</sup> imaging shows that Piezo2 bcKD reduces not only hypersensitivity to low-force mechanical stimulation, mostly among medium-sized neurons, but also, unexpectedly, TNI-induced spontaneous activity. Therefore, Fos is useful for genetic labeling and manipulation of BA TG neurons. Furthermore, innocuous mechanical stimuli activate multiple TG afferent subtypes after TNI, possibly accounting for the complexity of resulting painful symptoms.</p>","PeriodicalId":9798,"journal":{"name":"Cell reports","volume":"45 1","pages":"116803"},"PeriodicalIF":6.9,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145942641","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-07DOI: 10.1016/j.celrep.2025.116830
Christine M Joyce, Julien Bacal, Soham P Chowdhury, Andrew N Brown, Amy K Wang, Carmen Cruz, Kameron Bains, Zachary N Rodriguez, Nathan J McCormick, Yaara Tzadikario, Katherine U Tavasoli, Brooke M Gardner, Chris D Richardson
The FANCD2-FANCI heterodimer contributes to DNA repair at interstrand crosslinks and sites of replication stress. This complex has been physically and mechanistically linked to double-strand break (DSB) repair, but its role in that process remains undefined. Here, we show that the FANCD2-FANCI heterodimer dynamically interacts with open chromatin regions, including transient DSB-induced open chromatin, where it can be stabilized through co-activation by the DNA repair kinase ATM and the Fanconi anemia core ubiquitin ligase. The loaded FANCD2-FANCI heterodimer stabilizes open chromatin and promotes resection and loading of RPA through increased association of BRCA1 and BLM. Chromatin-loaded FANCD2-FANCI has a second, distinct function promoting a G2 cell cycle arrest that is dependent on the ATR-CHK1-WEE1 axis. Our results support a two-step genome surveillance model in which FANCD2-FANCI monitors open chromatin sites and is stably loaded to coordinate DNA repair activities in response to signaling from a DNA repair kinase.
{"title":"The FANCD2-FANCI heterodimer coordinates chromatin openness and cell cycle progression throughout DNA double-strand break repair.","authors":"Christine M Joyce, Julien Bacal, Soham P Chowdhury, Andrew N Brown, Amy K Wang, Carmen Cruz, Kameron Bains, Zachary N Rodriguez, Nathan J McCormick, Yaara Tzadikario, Katherine U Tavasoli, Brooke M Gardner, Chris D Richardson","doi":"10.1016/j.celrep.2025.116830","DOIUrl":"https://doi.org/10.1016/j.celrep.2025.116830","url":null,"abstract":"<p><p>The FANCD2-FANCI heterodimer contributes to DNA repair at interstrand crosslinks and sites of replication stress. This complex has been physically and mechanistically linked to double-strand break (DSB) repair, but its role in that process remains undefined. Here, we show that the FANCD2-FANCI heterodimer dynamically interacts with open chromatin regions, including transient DSB-induced open chromatin, where it can be stabilized through co-activation by the DNA repair kinase ATM and the Fanconi anemia core ubiquitin ligase. The loaded FANCD2-FANCI heterodimer stabilizes open chromatin and promotes resection and loading of RPA through increased association of BRCA1 and BLM. Chromatin-loaded FANCD2-FANCI has a second, distinct function promoting a G2 cell cycle arrest that is dependent on the ATR-CHK1-WEE1 axis. Our results support a two-step genome surveillance model in which FANCD2-FANCI monitors open chromatin sites and is stably loaded to coordinate DNA repair activities in response to signaling from a DNA repair kinase.</p>","PeriodicalId":9798,"journal":{"name":"Cell reports","volume":"45 1","pages":"116830"},"PeriodicalIF":6.9,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932513","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}