Aude Sylvain , Natacha Stoehr , Fupeng Ma , Artiom Cernijenko , Martin Schröder , Maryam Khoshouei , Melanie Vogelsanger , Michel Schoenboerner , Ashley Burke , Pasupuleti Rao , Jonathan M. Solomon , Joshiawa Paulk , Lei Xu , Janet Dawson , Damien Begue , Peggy Lefeuvre , Erik Ahrne , Andreas Hofmann , Callum J. Dickson , Philip Arabin , Zuni I. Bassi
{"title":"A cereblon-based glue degrader of NEK7 regulates NLRP3 inflammasome in a context-dependent manner","authors":"Aude Sylvain , Natacha Stoehr , Fupeng Ma , Artiom Cernijenko , Martin Schröder , Maryam Khoshouei , Melanie Vogelsanger , Michel Schoenboerner , Ashley Burke , Pasupuleti Rao , Jonathan M. Solomon , Joshiawa Paulk , Lei Xu , Janet Dawson , Damien Begue , Peggy Lefeuvre , Erik Ahrne , Andreas Hofmann , Callum J. Dickson , Philip Arabin , Zuni I. Bassi","doi":"10.1016/j.chembiol.2025.06.005","DOIUrl":"10.1016/j.chembiol.2025.06.005","url":null,"abstract":"<div><div>Aberrant NLRP3 (NACHT-, leucine-rich repeat [LRR]- and pyrin domain [PYD]- containing protein 3) inflammasome activation is linked to many inflammatory diseases, driving the search for therapeutics inhibiting this pathway. NEK7 is proposed to mediate NLRP3 inflammasome assembly and activation by bridging adjacent NLRP3 subunits. Hence, reduction of NEK7 protein may block NLRP3 activation. We identified NK7-902, a potent and selective cereblon (CRBN) glue degrader of NEK7. NK7-902 degraded NEK7 in human immune cells and whole blood. However, full NEK7 degradation completely blocked NLRP3-dependent interleukin-1β (IL-1β) release <em>in vitro</em> only in certain donors and experimental conditions. Unlike most CRBN glue degraders, NK7-902 effectively degraded NEK7 in murine cells and inhibited IL-1β release in mouse <em>in vivo</em>. By contrast, oral administration of NK7-902 in cynomolgus monkey caused long-lasting NEK7 degradation but only transiently blocked IL-1β in blood. These findings suggest NEK7 contributes to but is not absolutely required for NLRP3 activation in monkeys and humans.</div></div>","PeriodicalId":265,"journal":{"name":"Cell Chemical Biology","volume":"32 7","pages":"Pages 955-968.e13"},"PeriodicalIF":6.6,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144586803","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}
{"title":"Silencing stress: Structural insights into ISR termination by the SIFI ubiquitin ligase","authors":"Zeba Rizvi , Gabriel C. Lander","doi":"10.1016/j.chembiol.2025.06.007","DOIUrl":"10.1016/j.chembiol.2025.06.007","url":null,"abstract":"<div><div>The E3 ligase complex SIFI silences the integrated stress response (ISR) by targeting stress-induced proteins for degradation. In the May 6<sup>th</sup> issue of <em>Nature</em>, Yang et al.<span><span><sup>1</sup></span></span> revealed how this megadalton complex recognizes diverse substrates and coordinates ubiquitin chain formation. Their insights into the ISR shutdown mechanism suggest new avenues for modulating stress responses in neurodegenerative disease.</div></div>","PeriodicalId":265,"journal":{"name":"Cell Chemical Biology","volume":"32 7","pages":"Pages 905-907"},"PeriodicalIF":6.6,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144645526","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}
Flore Nardella , Tiantian Jiang , Lushun Wang , Monica J. Bohmer , Subhoja Chakraborty , John Okombo , Jaeson Calla , Tatiane Macedo Silva , Samuel Pazicky , Jianwei Che , Jin Jeon , Evie Vincent , Nonlawat Boonyalai , Rachael Coyle , Mairi J. Buchanan , Samuel Schaefer , Daisy Chen , Amaan Khan , Emily Mayville , Mariana Laureano De Souza , Debopam Chakrabarti
{"title":"Plasmodium falciparum protein kinase 6 and hemozoin formation are inhibited by a type II human kinase inhibitor exhibiting antimalarial activity","authors":"Flore Nardella , Tiantian Jiang , Lushun Wang , Monica J. Bohmer , Subhoja Chakraborty , John Okombo , Jaeson Calla , Tatiane Macedo Silva , Samuel Pazicky , Jianwei Che , Jin Jeon , Evie Vincent , Nonlawat Boonyalai , Rachael Coyle , Mairi J. Buchanan , Samuel Schaefer , Daisy Chen , Amaan Khan , Emily Mayville , Mariana Laureano De Souza , Debopam Chakrabarti","doi":"10.1016/j.chembiol.2025.06.003","DOIUrl":"10.1016/j.chembiol.2025.06.003","url":null,"abstract":"<div><div>Kinase inhibitors are potent therapeutics, but most essential <em>Plasmodium</em> kinases remain unexploited as antimalarial targets. We identified compound <strong>12</strong>, a type II kinase inhibitor based on aminopyridine and 2,6-benzimidazole scaffolds, as a lead compound with nanomolar potency, fast action, and <em>in vivo</em> activity in the <em>Plasmodium berghei</em> rodent malaria model. Three-hybrid luciferase fragment complementation, enzymatic studies, and cellular thermal shift assays implicated <em>Plasmodium</em> protein kinase 6 (PfPK6) as the target. However, conditional knockdown of PfPK6 did not alter <strong>12</strong> potency, suggesting complex mechanisms of action. <em>In vitro</em> selection for compound <strong>12</strong> resistance revealed mutations in three transporters: multidrug-resistance protein 1, chloroquine resistance transporter and V-type ATPase, indicating a digestive vacuole site of action. Compound <strong>12</strong> inhibited β-hematin and hemozoin formation while increasing free heme levels, suggesting antimalarial activity via blockade of heme detoxification. Our studies repurpose a safe human kinase inhibitor as a potent, fast-acting antimalarial with established <em>in vivo</em> efficacy.</div></div>","PeriodicalId":265,"journal":{"name":"Cell Chemical Biology","volume":"32 7","pages":"Pages 926-941.e23"},"PeriodicalIF":6.6,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144568957","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}
Maoyuan Linghu , Xianyu Luo , Xinru Zhou , Didi Liu , Qian Huang , Yi Ru , Yingli Luo , Yinchu Ma , Yi Huang
{"title":"Covalent inhibition of ACSL4 alleviates ferroptosis-induced acute liver injury","authors":"Maoyuan Linghu , Xianyu Luo , Xinru Zhou , Didi Liu , Qian Huang , Yi Ru , Yingli Luo , Yinchu Ma , Yi Huang","doi":"10.1016/j.chembiol.2025.06.004","DOIUrl":"10.1016/j.chembiol.2025.06.004","url":null,"abstract":"<div><div>Ferroptosis, a form of regulated cell death, is characterized by iron-dependent phospholipid peroxidation and is closely linked to various liver diseases. Although covalent inhibitors have gained attention for their high potency and prolonged effects, no specific covalent inhibitor for ferroptosis exists. Here, we identify Rociletinib (ROC) as a potent inhibitor of ferroptosis through virtual screening and mechanistic studies. Our results demonstrate that ROC covalently binds to cysteine 170 of ACSL4, inhibiting its enzymatic activity and thereby suppressing lipid peroxidation and ferroptosis. ROC effectively mitigates ferroptosis-mediated acute liver injury in mouse models. These findings establish ROC as the targeted covalent inhibitor directly targeting ACSL4, offering a promising therapeutic strategy for ferroptosis-related diseases.</div></div>","PeriodicalId":265,"journal":{"name":"Cell Chemical Biology","volume":"32 7","pages":"Pages 942-954.e5"},"PeriodicalIF":6.6,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144578077","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}
Nil Casajuana-Martin , Eli F. McDonald , M. Madan Babu
{"title":"Flush with insight: Decoding GPCR crosstalk in the spinal defecation center","authors":"Nil Casajuana-Martin , Eli F. McDonald , M. Madan Babu","doi":"10.1016/j.chembiol.2025.05.011","DOIUrl":"10.1016/j.chembiol.2025.05.011","url":null,"abstract":"<div><div>G protein-coupled receptors (GPCRs) are increasingly recognized as part of interconnected, cell-context-specific signaling networks. In the June 5<sup>th</sup> issue of <em>Molecular Cell</em>, Dehkhoda et al.<span><span><sup>1</sup></span></span> demonstrate that the constitutive activity of the ghrelin receptor drives dopamine D2 receptor-dependent calcium mobilization, highlighting the complexity of GPCR signaling and opening new avenues for therapeutic development.</div></div>","PeriodicalId":265,"journal":{"name":"Cell Chemical Biology","volume":"32 6","pages":"Pages 786-788"},"PeriodicalIF":6.6,"publicationDate":"2025-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144313950","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}
{"title":"Electrochemical sensor toolkit for simultaneous glutamate detection at edge of cleft and peri-soma","authors":"Jie Liu , Yuandong Liu , Dongmin Yin , Yang Tian","doi":"10.1016/j.chembiol.2025.05.002","DOIUrl":"10.1016/j.chembiol.2025.05.002","url":null,"abstract":"<div><div>Simultaneously monitoring glutamate (Glu) dynamic at edge of synaptic cleft and peri-soma is crucial for understanding Glu-related pathology. Here, we created an electrochemical Glu sensors toolkit with spatial resolution of ∼60 nm, combining biologically engineered Glu binding protein for specifically capturing Glu together with chemically designed ferrocene groups for signal labeling. Modulation conjugation approach between GluR and ferrocene significantly improved sensitivity up to 32-folds. More importantly, protein engineering of residue mutation and linker peptides flexibility expanded linear range from 10 μM to 6 mM, accelerated on/off times down to 35/40 ms. This toolkit realized real-time quantifying of Glu both at edge of cleft and peri-soma, we discovered that Glu was almost released through SLC7A11 channels in calyx of held synapse upon oxygen-glucose-deprivation, while Glu was mainly released through hemichannels upon β-amyloid<sub>42</sub> stimulation. Our work provided a methodology for investigating Glu release and reuptake and offered insights for Glu related pathology.</div></div>","PeriodicalId":265,"journal":{"name":"Cell Chemical Biology","volume":"32 6","pages":"Pages 885-898.e11"},"PeriodicalIF":6.6,"publicationDate":"2025-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144202419","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}
Marta Baro , Hojin Lee , Vanessa Kelley , Rongliang Lou , Chatchai Phoomak , Katerina Politi , Caroline J. Zeiss , Michael Van Zandt , Joseph N. Contessa
{"title":"Redundancy of the OST catalytic subunit facilitates therapeutic targeting of N-glycosylation","authors":"Marta Baro , Hojin Lee , Vanessa Kelley , Rongliang Lou , Chatchai Phoomak , Katerina Politi , Caroline J. Zeiss , Michael Van Zandt , Joseph N. Contessa","doi":"10.1016/j.chembiol.2025.05.005","DOIUrl":"10.1016/j.chembiol.2025.05.005","url":null,"abstract":"<div><div>Protein asparagine (N)-glycosylation, which promotes the folding and trafficking of cell surface receptors, has not traditionally been viewed as a viable target in oncology due to the essential and non-redundant enzymatic activities required for glycan synthesis and transfer. However, in mammals, an exception is the presence of the oligosaccharyltransferase (OST) catalytic subunit paralogs, STT3A and STT3B. In this study, we investigate the biological activity of OST inhibitors and develop a strategy for selectively inhibiting N-glycosylation that is optimized for its downstream effects on the EGFR glycoprotein. Small molecules with improved pharmacokinetic properties and selective preferences for STT3A or STT3B were synthesized, characterized <em>in vitro</em>, and advanced to <em>in vivo</em> testing. The lead compound from this series, NGI-189, induces tumor regression or growth delay in patient-derived and TKI-resistant EGFR-mutant lung cancer xenografts without causing toxicity. Collectively, these findings suggest that bioavailable OST inhibitors can be developed as therapeutic agents for oncology.</div></div>","PeriodicalId":265,"journal":{"name":"Cell Chemical Biology","volume":"32 6","pages":"Pages 839-853.e6"},"PeriodicalIF":6.6,"publicationDate":"2025-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144237922","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}
Yuchen Zhang (章雨辰) , Hongpeng Li (李鸿鹏) , Yi Hao (郝熠) , Jiaqi Chen (陈家祺) , Xing Chen (陈兴) , Hang Yin (尹航)
{"title":"EGR2 O-GlcNAcylation orchestrates the development of protumoral macrophages to limit CD8+ T cell antitumor responses","authors":"Yuchen Zhang (章雨辰) , Hongpeng Li (李鸿鹏) , Yi Hao (郝熠) , Jiaqi Chen (陈家祺) , Xing Chen (陈兴) , Hang Yin (尹航)","doi":"10.1016/j.chembiol.2025.05.007","DOIUrl":"10.1016/j.chembiol.2025.05.007","url":null,"abstract":"<div><div>Tumor associated macrophages (TAMs) exhibit a high capacity to take up glucose. However, how metabolic cues derived from glucose rewire TAMs remains unclear. Here, we report that glucose metabolism-driven protein O-GlcNAcylation increases in TAMs and shapes the differentiation and protumoral function of TAMs. Deficiency of O-GlcNAc transferase (OGT) in TAMs restricted tumor growth by reducing the proportion of C1QC<sup>+</sup> F4/80<sup>+</sup> TREM2<sup>+</sup> MerTK<sup>+</sup> TAMs as well as <em>Trem2 e</em>xpression, which in turn preserved the cytotoxic function of effector CD8<sup>+</sup> T cells while exhibiting reduced features of exhaustion. Mechanistically, O-GlcNAc targeted the macrophage-specific transcription factor EGR2 to promote its transcriptional activity. Transcriptional profiling revealed that OGT increased EGR2-related motifs accessibility in TAMs. O-GlcNAcylation of EGR2 at serine 299 enhanced its binding to myeloid cell differentiation-associated genes, including <em>Trem2</em>, thus facilitating the protumoral function of TAMs in GM-CSF-sufficient tumor. Overall, our work defines a tumor-specific reprogramming of protumoral TAMs via O-GlcNAc-modified EGR2 transcriptional regulation.</div></div>","PeriodicalId":265,"journal":{"name":"Cell Chemical Biology","volume":"32 6","pages":"Pages 809-825.e7"},"PeriodicalIF":6.6,"publicationDate":"2025-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144252587","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}
Yongliang Liu , Hui Hua , Yalan Cao , Minjing Li , Hongying Zhang , Shan Du , Jieya Liu , Ting Luo , Yangfu Jiang
{"title":"Mechanism by which the molecular glue-like verteporfin induces IRE1α dimerization and activation to synergize with AKT inhibition in breast cancer","authors":"Yongliang Liu , Hui Hua , Yalan Cao , Minjing Li , Hongying Zhang , Shan Du , Jieya Liu , Ting Luo , Yangfu Jiang","doi":"10.1016/j.chembiol.2025.05.004","DOIUrl":"10.1016/j.chembiol.2025.05.004","url":null,"abstract":"<div><div>Inositol-requiring enzyme 1α (IRE1α) signaling is one of three arms of the unfolded protein response, playing a vital role in maintaining endoplasmic reticulum homeostasis. Pharmacological modulation of this pathway offers potential therapeutic strategies for various diseases. Molecular glues may regulate protein stability and activity by inducing protein-protein interaction. Here, we find that verteporfin functions as a molecular glue, promoting IRE1α dimerization and activation. Specifically, verteporfin binds to IRE1α, facilitating its dimerization, which relies on the His692 residue. This activation of IRE1α triggers XBP1 splicing and miR-153-mediated downregulation of PTEN, along with AKT phosphorylation. Additionally, we identify the pro-metastasis gene <em>BACH1</em> as a novel target of miR-153, which is downregulated by IRE1α and verteporfin. While verteporfin inhibits breast cancer cell viability and invasion, its combination with an AKT inhibitor synergistically suppresses breast cancer progression. Our findings establish a mechanistic link between IRE1α and PI3K/AKT signaling, highlighting a possibility for therapeutic intervention.</div></div>","PeriodicalId":265,"journal":{"name":"Cell Chemical Biology","volume":"32 6","pages":"Pages 854-871.e6"},"PeriodicalIF":6.6,"publicationDate":"2025-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144211168","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}
{"title":"Two sides of a co(i)ndensate","authors":"Emre Pekbilir , Dorothee Dormann","doi":"10.1016/j.chembiol.2025.05.010","DOIUrl":"10.1016/j.chembiol.2025.05.010","url":null,"abstract":"<div><div>In the June 5<sup>th</sup> issue of <em>Molecular Cell</em>, Das et al.<span><span><sup>1</sup></span></span> quantified the stability of condensates and fibrils formed from the prion-like low complexity region of hnRNPA1. They demonstrate that condensate interiors function as sinks and suppress fibril growth by slowing protein efflux, illuminating the interplay between condensation and fibril formation.</div></div>","PeriodicalId":265,"journal":{"name":"Cell Chemical Biology","volume":"32 6","pages":"Pages 783-785"},"PeriodicalIF":6.6,"publicationDate":"2025-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144314297","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}