首页 > 最新文献

International Journal of Biological Sciences最新文献

英文 中文
Piezo1-driven mechanotransduction regulates mitochondrial biogenesis by AMPK/SIRT1-mediated PGC-1α deacetylation to ameliorate bone loss in disuse osteoporosis. 压电驱动的机械转导通过AMPK/ sirt1介导的PGC-1α去乙酰化调节线粒体生物发生,以改善废用性骨质疏松症的骨质流失。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 DOI: 10.7150/ijbs.124043
Jianpeng Chen, Dengying Wu, Chengbin Huang, Zijian Yan, Jiahao Wang, Siteng Li, Xuankuai Chen, Yanbin Zhu, Yingze Zhang

Disuse osteoporosis (DOP), a skeletal disorder triggered by insufficient mechanical loading, manifests as progressive bone mass deterioration and microarchitectural weakening. Piezo1, a key mechanosensitive ion channel expressed in bone cells, is implicated in maintaining skeletal homeostasis. Using a murine hindlimb unloading (HLU) model simulating microgravity-induced bone loss, we observed significant downregulation of Piezo1 expression in bone tissue and isolated bone marrow-derived mesenchymal stem cells (BMSCs). Systemic administration of the Piezo1 agonist Yoda1 attenuated HLU-induced osteopenia and improved bone formation capacity. Mechanistic studies in BMSCs demonstrated that Piezo1 activation promoted mitochondrial biogenesis. This effect required AMPK/SIRT1 signaling-dependent deacetylation of PGC-1α, leading to enhanced mitochondrial function, improved osteogenic differentiation, and reduced apoptosis. Critically, pharmacologic inhibition of SIRT1 abolished the osteoprotective effects of Yoda1 in vivo. These findings establish that mechanical unloading impairs Piezo1-mediated mechanotransduction in BMSCs, contributing to disrupted skeletal homeostasis, which can be mitigated by exogenous Piezo1 activation. Our results define a mechanism where Piezo1 integrates mechanical signals into the AMPK/SIRT1/PGC-1α signaling cascade to regulate mechanoadaptive bone formation, highlighting Piezo1 activation as a potential mechanism-based therapeutic strategy for disuse osteoporosis.

废用性骨质疏松症(DOP)是一种由机械负荷不足引发的骨骼疾病,表现为进行性骨量恶化和微结构弱化。Piezo1是骨细胞中表达的一个关键的机械敏感离子通道,与维持骨骼稳态有关。通过模拟微重力诱导骨丢失的小鼠后肢卸载(HLU)模型,我们观察到骨组织和分离的骨髓间充质干细胞(BMSCs)中Piezo1表达的显著下调。全身给药Piezo1激动剂Yoda1可减轻hlu诱导的骨质减少并改善骨形成能力。骨髓间充质干细胞的机制研究表明,Piezo1的激活促进了线粒体的生物发生。这种作用需要AMPK/SIRT1信号依赖的PGC-1α去乙酰化,从而增强线粒体功能,改善成骨分化,减少细胞凋亡。重要的是,SIRT1的药理学抑制在体内消除了Yoda1的骨保护作用。这些发现表明,机械卸载会损害骨髓间充质干细胞中Piezo1介导的机械转导,导致骨骼稳态被破坏,这可以通过外源性Piezo1激活来缓解。我们的研究结果确定了一种机制,其中Piezo1将机械信号整合到AMPK/SIRT1/PGC-1α信号级联中,以调节机械适应性骨形成,突出了Piezo1激活作为一种潜在的基于机制的治疗策略。
{"title":"Piezo1-driven mechanotransduction regulates mitochondrial biogenesis by AMPK/SIRT1-mediated PGC-1α deacetylation to ameliorate bone loss in disuse osteoporosis.","authors":"Jianpeng Chen, Dengying Wu, Chengbin Huang, Zijian Yan, Jiahao Wang, Siteng Li, Xuankuai Chen, Yanbin Zhu, Yingze Zhang","doi":"10.7150/ijbs.124043","DOIUrl":"10.7150/ijbs.124043","url":null,"abstract":"<p><p>Disuse osteoporosis (DOP), a skeletal disorder triggered by insufficient mechanical loading, manifests as progressive bone mass deterioration and microarchitectural weakening. Piezo1, a key mechanosensitive ion channel expressed in bone cells, is implicated in maintaining skeletal homeostasis. Using a murine hindlimb unloading (HLU) model simulating microgravity-induced bone loss, we observed significant downregulation of Piezo1 expression in bone tissue and isolated bone marrow-derived mesenchymal stem cells (BMSCs). Systemic administration of the Piezo1 agonist Yoda1 attenuated HLU-induced osteopenia and improved bone formation capacity. Mechanistic studies in BMSCs demonstrated that Piezo1 activation promoted mitochondrial biogenesis. This effect required AMPK/SIRT1 signaling-dependent deacetylation of PGC-1α, leading to enhanced mitochondrial function, improved osteogenic differentiation, and reduced apoptosis. Critically, pharmacologic inhibition of SIRT1 abolished the osteoprotective effects of Yoda1 in vivo. These findings establish that mechanical unloading impairs Piezo1-mediated mechanotransduction in BMSCs, contributing to disrupted skeletal homeostasis, which can be mitigated by exogenous Piezo1 activation. Our results define a mechanism where Piezo1 integrates mechanical signals into the AMPK/SIRT1/PGC-1α signaling cascade to regulate mechanoadaptive bone formation, highlighting Piezo1 activation as a potential mechanism-based therapeutic strategy for disuse osteoporosis.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"22 1","pages":"308-326"},"PeriodicalIF":10.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12681831/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145707282","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pan-cancer bone metastasis atlas at single-cell resolution identifies a distinct tumor-associated macrophage subset for mediating Denosumab-induced immunosensitization in lung cancer bone metastasis. 单细胞分辨率的泛癌骨转移图谱确定了一个独特的肿瘤相关巨噬细胞亚群,可介导denosumab诱导的肺癌骨转移免疫致敏。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 DOI: 10.7150/ijbs.119777
Xianglin Hu, Nan Du, Yansha Song, Ke Lang, Wanning Tong, Qingrong Ye, Xuesi Liu, Haoyu Zheng, Mo Cheng, Yingzheng Ji, Haibo Wu, Minghe Zhang, Xinhong He, Yan Zhang, Xiaomeng Li, Yao Zhu, Kun Li, Weiluo Cai, Wangjun Yan, Wending Huang

Lung cancer (LC), prostate cancer (PC), and breast cancer (BC) are the three most prevalent cancers that lead to bone metastasis (BoM). In this study, we conducted an integrated analysis of single-cell transcriptomic data from the primary tumors and BoM across PC, LC, and BC. We discover a novel subtype of tumor-associated macrophages (TAMs) that are positive both for matrix metalloproteinase 19 (MMP19) and receptor activator of nuclear factor-κB (RANK) expression (MMP19+ RANK+ TAMs). MMP19+ RANK+ TAMs demonstrate an increased level of M2 polarization and act as a critical driving factor for LC-BoM. MMP19+ RANK+ TAMs are organized in a ring-like arrangement surrounding the tumor nests, constructing a barrier structure that impedes the infiltration of CD8+ T cells into the tumor core in LC-BoM. RANKL inhibitor Denosumab has been shown to effectively reduce the level of M2 polarization, decrease the population of MMP19+ RANK+ TAMs, and disrupt their barrier structure. Denosumab facilitates the infiltration of CD8+ T cells into the interior of LC-BoM tissues. Based on this mechanism, we observed in both clinical cohorts and preclinical models that RANKL inhibitor can enhance the efficacy of immunotherapy in treating LC-BoM.

肺癌(LC)、前列腺癌(PC)和乳腺癌(BC)是导致骨转移(BoM)的三种最常见的癌症。在这项研究中,我们对PC、LC和BC的原发肿瘤和BoM的单细胞转录组数据进行了综合分析。我们发现了一种新的肿瘤相关巨噬细胞(tam)亚型,其基质金属蛋白酶19 (MMP19)和核因子κ b受体激活因子(RANK)表达(MMP19+ RANK+ tam)均呈阳性。MMP19+ RANK+ tam表现出M2极化水平的增加,并作为LC-BoM的关键驱动因素。在LC-BoM中,MMP19+ RANK+ tam在肿瘤巢周围呈环状排列,形成屏障结构,阻止CD8+ T细胞浸润到肿瘤核心。RANKL抑制剂Denosumab已被证明可以有效降低M2极化水平,减少MMP19+ RANK+ tam的数量,并破坏其屏障结构。Denosumab促进CD8+ T细胞向LC-BoM组织内部浸润。基于这一机制,我们在临床队列和临床前模型中观察到RANKL抑制剂可以增强免疫疗法治疗LC-BoM的疗效。
{"title":"Pan-cancer bone metastasis atlas at single-cell resolution identifies a distinct tumor-associated macrophage subset for mediating Denosumab-induced immunosensitization in lung cancer bone metastasis.","authors":"Xianglin Hu, Nan Du, Yansha Song, Ke Lang, Wanning Tong, Qingrong Ye, Xuesi Liu, Haoyu Zheng, Mo Cheng, Yingzheng Ji, Haibo Wu, Minghe Zhang, Xinhong He, Yan Zhang, Xiaomeng Li, Yao Zhu, Kun Li, Weiluo Cai, Wangjun Yan, Wending Huang","doi":"10.7150/ijbs.119777","DOIUrl":"10.7150/ijbs.119777","url":null,"abstract":"<p><p>Lung cancer (LC), prostate cancer (PC), and breast cancer (BC) are the three most prevalent cancers that lead to bone metastasis (BoM). In this study, we conducted an integrated analysis of single-cell transcriptomic data from the primary tumors and BoM across PC, LC, and BC. We discover a novel subtype of tumor-associated macrophages (TAMs) that are positive both for matrix metalloproteinase 19 (MMP19) and receptor activator of nuclear factor-κB (RANK) expression (MMP19<sup>+</sup> RANK<sup>+</sup> TAMs). MMP19<sup>+</sup> RANK<sup>+</sup> TAMs demonstrate an increased level of M2 polarization and act as a critical driving factor for LC-BoM. MMP19<sup>+</sup> RANK<sup>+</sup> TAMs are organized in a ring-like arrangement surrounding the tumor nests, constructing a barrier structure that impedes the infiltration of CD8<sup>+</sup> T cells into the tumor core in LC-BoM. RANKL inhibitor Denosumab has been shown to effectively reduce the level of M2 polarization, decrease the population of MMP19<sup>+</sup> RANK<sup>+</sup> TAMs, and disrupt their barrier structure. Denosumab facilitates the infiltration of CD8<sup>+</sup> T cells into the interior of LC-BoM tissues. Based on this mechanism, we observed in both clinical cohorts and preclinical models that RANKL inhibitor can enhance the efficacy of immunotherapy in treating LC-BoM.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"22 1","pages":"365-386"},"PeriodicalIF":10.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12681936/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145707308","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Dual Phosphorylation of STAT1 at Y701/S727 by TNFα Drives AIM2-Mediated PANoptosis of Renal Tubular Epithelial Cells and Fibrotic Progression in Renal Allografts. TNFα对Y701/S727位点STAT1的双重磷酸化驱动aim2介导的肾小管上皮细胞泛凋亡和同种异体肾移植的纤维化进展
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 DOI: 10.7150/ijbs.123441
Qianguang Han, Jiawen Liu, Jianjian Zhang, Qinghuan Shen, Junqi Zhang, Shuang Fei, Hao Chen, Li Sun, Zhengkai Huang, Zhijian Han, Jun Tao, Min Gu, Xiaobing Ju, Ruoyun Tan

Renal allograft interstitial fibrosis, a key pathological feature of chronic renal allograft dysfunction (CAD), is a critical determinant of long-term graft survival. However, its underlying molecular mechanisms remain incompletely understood. This study uncovers the central role of programmed cell death, particularly the novel PANoptosis modality, in the progression of CAD. PANoptosis integrates features of pyroptosis, apoptosis, and necroptosis, but does not fit within the confines of any single pathway, with its mechanisms previously undefined. By analyzing cell death patterns in CAD tissues through single-cell sequencing and validating findings via in vivo and in vitro experiments, this work demonstrates that in the context of chronic inflammation, tumor necrosis factor-alpha (TNF-α) modulates signal transducer and activator of transcription 1 (STAT1) through dual phosphorylation. This process directly induces tyrosine 701 phosphorylation and activates serine 727 phosphorylation via the p38 MAPK pathway. Phosphorylated STAT1 subsequently upregulates the PANoptosome sensor absent in melanoma 2 (AIM2), driving PANoptosis in renal tubular epithelial cells. This mechanism further exacerbates interstitial fibrosis by promoting the paracrine secretion of interleukin-6 and transforming growth factor-beta, which induces epithelial-mesenchymal transition (EMT) in adjacent tubular cells. These findings represent the first demonstration of the TNF-α/STAT1/AIM2 axis in triggering PANoptosis and its downstream EMT-fibrosis cascade, offering novel therapeutic targets for CAD intervention.

同种异体肾间质纤维化是慢性同种异体肾功能障碍(CAD)的一个关键病理特征,是移植物长期存活的关键决定因素。然而,其潜在的分子机制仍不完全清楚。这项研究揭示了程序性细胞死亡的核心作用,特别是新的PANoptosis模式,在冠心病的进展。PANoptosis融合了焦亡、凋亡和坏死的特征,但不属于任何单一途径的范围,其机制以前未明确。通过单细胞测序分析CAD组织中的细胞死亡模式,并通过体内和体外实验验证研究结果,本研究表明,在慢性炎症的背景下,肿瘤坏死因子-α (TNF-α)通过双磷酸化调节信号换能器和转录激活因子1 (STAT1)。该过程直接诱导酪氨酸701磷酸化,并通过p38 MAPK途径激活丝氨酸727磷酸化。磷酸化的STAT1随后上调黑色素瘤2 (AIM2)中缺失的PANoptosome传感器,导致肾小管上皮细胞PANoptosis。该机制通过促进旁分泌白介素-6和转化生长因子- β,从而诱导邻近小管细胞上皮-间质转化(EMT),进一步加剧间质纤维化。这些发现首次证明了TNF-α/STAT1/AIM2轴触发PANoptosis及其下游emt -纤维化级联,为CAD干预提供了新的治疗靶点。
{"title":"Dual Phosphorylation of STAT1 at Y701/S727 by TNFα Drives AIM2-Mediated PANoptosis of Renal Tubular Epithelial Cells and Fibrotic Progression in Renal Allografts.","authors":"Qianguang Han, Jiawen Liu, Jianjian Zhang, Qinghuan Shen, Junqi Zhang, Shuang Fei, Hao Chen, Li Sun, Zhengkai Huang, Zhijian Han, Jun Tao, Min Gu, Xiaobing Ju, Ruoyun Tan","doi":"10.7150/ijbs.123441","DOIUrl":"10.7150/ijbs.123441","url":null,"abstract":"<p><p>Renal allograft interstitial fibrosis, a key pathological feature of chronic renal allograft dysfunction (CAD), is a critical determinant of long-term graft survival. However, its underlying molecular mechanisms remain incompletely understood. This study uncovers the central role of programmed cell death, particularly the novel PANoptosis modality, in the progression of CAD. PANoptosis integrates features of pyroptosis, apoptosis, and necroptosis, but does not fit within the confines of any single pathway, with its mechanisms previously undefined. By analyzing cell death patterns in CAD tissues through single-cell sequencing and validating findings via <i>in vivo</i> and <i>in vitro</i> experiments, this work demonstrates that in the context of chronic inflammation, tumor necrosis factor-alpha (TNF-α) modulates signal transducer and activator of transcription 1 (STAT1) through dual phosphorylation. This process directly induces tyrosine 701 phosphorylation and activates serine 727 phosphorylation via the p38 MAPK pathway. Phosphorylated STAT1 subsequently upregulates the PANoptosome sensor absent in melanoma 2 (AIM2), driving PANoptosis in renal tubular epithelial cells. This mechanism further exacerbates interstitial fibrosis by promoting the paracrine secretion of interleukin-6 and transforming growth factor-beta, which induces epithelial-mesenchymal transition (EMT) in adjacent tubular cells. These findings represent the first demonstration of the TNF-α/STAT1/AIM2 axis in triggering PANoptosis and its downstream EMT-fibrosis cascade, offering novel therapeutic targets for CAD intervention.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"22 2","pages":"582-600"},"PeriodicalIF":10.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12780947/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951909","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Role of Hedgehog Signaling in Non-small Cell Lung Cancer: Targeting Tumor Invasion, Therapy Resistance and Novel Therapeutic Strategies. 刺猬信号在非小细胞肺癌中的作用:靶向肿瘤侵袭,治疗抵抗和新的治疗策略。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 DOI: 10.7150/ijbs.123287
Yu Kang, Hongmei Zheng, Qiuyuan Wen, Songqing Fan

Although essential for normal development and tissue homeostasis, aberrant activation of the Hedgehog (Hh) signaling pathway is implicated in non-small cell lung cancer (NSCLC) progression and treatment resistance. This review details the contribution of Hh signaling to NSCLC, focusing on its promotion of tumor invasion and therapeutic resistance, and establishes a rationale for disrupting this pathway to improve treatment efficacy. Malignant phenotypes in NSCLC are driven by dysregulated Hh pathway activity, often via autocrine or paracrine loops. We specifically assess how Hh pathway activation enables tumor invasion, metastasis, and the development of drug resistance. The review elucidates key resistance mechanisms against diverse therapies-encompassing chemotherapy, targeted therapy and immunotherapy-with a focus on epithelial-mesenchymal transition (EMT), cancer stem cell maintenance, and multidrug resistance (MDR). Therefore, combining Hh pathway inhibitors with standard therapies represents a promising approach for managing treatment-resistant NSCLC.

尽管对正常发育和组织稳态至关重要,但Hedgehog (Hh)信号通路的异常激活与非小细胞肺癌(NSCLC)的进展和治疗耐药性有关。本文详细介绍了Hh信号在NSCLC中的作用,重点关注其促进肿瘤侵袭和治疗耐药性,并建立了破坏该通路以提高治疗疗效的理论基础。NSCLC的恶性表型是由Hh通路活性失调驱动的,通常通过自分泌或旁分泌回路。我们特别评估Hh通路激活如何使肿瘤侵袭、转移和耐药性的发展。这篇综述阐述了针对多种治疗(包括化疗、靶向治疗和免疫治疗)的关键耐药机制,重点是上皮-间质转化(EMT)、癌症干细胞维持和多药耐药(MDR)。因此,Hh通路抑制剂与标准疗法相结合是治疗耐药NSCLC的一种有希望的方法。
{"title":"The Role of Hedgehog Signaling in Non-small Cell Lung Cancer: Targeting Tumor Invasion, Therapy Resistance and Novel Therapeutic Strategies.","authors":"Yu Kang, Hongmei Zheng, Qiuyuan Wen, Songqing Fan","doi":"10.7150/ijbs.123287","DOIUrl":"10.7150/ijbs.123287","url":null,"abstract":"<p><p>Although essential for normal development and tissue homeostasis, aberrant activation of the Hedgehog (Hh) signaling pathway is implicated in non-small cell lung cancer (NSCLC) progression and treatment resistance. This review details the contribution of Hh signaling to NSCLC, focusing on its promotion of tumor invasion and therapeutic resistance, and establishes a rationale for disrupting this pathway to improve treatment efficacy. Malignant phenotypes in NSCLC are driven by dysregulated Hh pathway activity, often via autocrine or paracrine loops. We specifically assess how Hh pathway activation enables tumor invasion, metastasis, and the development of drug resistance. The review elucidates key resistance mechanisms against diverse therapies-encompassing chemotherapy, targeted therapy and immunotherapy-with a focus on epithelial-mesenchymal transition (EMT), cancer stem cell maintenance, and multidrug resistance (MDR). Therefore, combining Hh pathway inhibitors with standard therapies represents a promising approach for managing treatment-resistant NSCLC.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"22 2","pages":"701-712"},"PeriodicalIF":10.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12781077/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951119","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GIPC1 Restrains the Progression and Chemoresistance of Colorectal Cancer by Regulating TTC7B/mTOR/NF-κB Axis. GIPC1通过调控TTC7B/mTOR/NF-κB轴抑制结直肠癌的进展和化疗耐药
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 DOI: 10.7150/ijbs.119064
Dongxue Gan, Cheng Yang, Xiangjing Shen, Jingjie Shi, Ronglin Wang, Liaoliao Zhu, Hong Li, Jing Luo, Ting Zhao, Junqiang Li, Yang Song, Haichuan Su

Colorectal cancer (CRC) remains a leading cause of cancer-related mortality, highlighting the need for a deeper understanding of its molecular mechanisms to drive the development of novel therapeutic approaches. In this study, the findings indicated a significant reduction in PDZ Domain Containing Family Member 1 (GIPC1) expression in CRC tissues, which correlated with poor prognosis in patients with CRC at pathological stages T1 and T2. GIPC1 acted as a tumor suppressor gene that inhibited CRC cell proliferation, colony formation, migration, and invasion. Additionally, it enhanced CRC cell sensitivity to first-line chemotherapies such as 5-fluorouracil (5-FU), oxaliplatin (OXA), and irinotecan (CPT-11). Mechanistically, GIPC1 reduced the ubiquitination level of tetratricopeptide repeat domain 7B (TTC7B) by downregulating the E3 ubiquitin ligase TRIM21, thereby stabilizing TTC7B's expression and inhibiting the downstream mTOR/NF-κB signaling cascade. Moreover, in vivo studies confirmed the inhibitory role of GIPC1 in CRC growth and found that GIPC1-loaded lipid nanoparticles (GIPC1-LNPs) combined with 5-FU treatment had a more significant antitumor effect. In conclusion, this study reveals the GIPC1/TRIM21/TTC7B/mTOR/NF-κB tumor-suppressive axis in CRC and highlights the potential of GIPC1 for early diagnosis and overcoming chemoresistance in CRC patients.

结直肠癌(CRC)仍然是癌症相关死亡的主要原因,强调需要更深入地了解其分子机制,以推动新治疗方法的发展。本研究发现,在CRC组织中,PDZ Domain Containing Family Member 1 (GIPC1)表达显著降低,与病理期T1和T2的CRC患者预后不良相关。GIPC1作为肿瘤抑制基因,抑制结直肠癌细胞增殖、集落形成、迁移和侵袭。此外,它增强了CRC细胞对一线化疗的敏感性,如5-氟尿嘧啶(5-FU)、奥沙利铂(OXA)和伊立替康(CPT-11)。机制上,GIPC1通过下调E3泛素连接酶TRIM21,降低四肽重复结构域7B (TTC7B)的泛素化水平,从而稳定TTC7B的表达,抑制下游mTOR/NF-κB信号级联。此外,体内研究证实了GIPC1对结直肠癌生长的抑制作用,并发现负载GIPC1的脂质纳米颗粒(GIPC1- lnps)联合5-FU治疗具有更显著的抗肿瘤作用。总之,本研究揭示了GIPC1/TRIM21/TTC7B/mTOR/NF-κB肿瘤抑制轴在结直肠癌中的作用,并强调了GIPC1在结直肠癌患者早期诊断和克服化疗耐药方面的潜力。
{"title":"GIPC1 Restrains the Progression and Chemoresistance of Colorectal Cancer by Regulating TTC7B/mTOR/NF-κB Axis.","authors":"Dongxue Gan, Cheng Yang, Xiangjing Shen, Jingjie Shi, Ronglin Wang, Liaoliao Zhu, Hong Li, Jing Luo, Ting Zhao, Junqiang Li, Yang Song, Haichuan Su","doi":"10.7150/ijbs.119064","DOIUrl":"10.7150/ijbs.119064","url":null,"abstract":"<p><p>Colorectal cancer (CRC) remains a leading cause of cancer-related mortality, highlighting the need for a deeper understanding of its molecular mechanisms to drive the development of novel therapeutic approaches. In this study, the findings indicated a significant reduction in PDZ Domain Containing Family Member 1 (GIPC1) expression in CRC tissues, which correlated with poor prognosis in patients with CRC at pathological stages T1 and T2. GIPC1 acted as a tumor suppressor gene that inhibited CRC cell proliferation, colony formation, migration, and invasion. Additionally, it enhanced CRC cell sensitivity to first-line chemotherapies such as 5-fluorouracil (5-FU), oxaliplatin (OXA), and irinotecan (CPT-11). Mechanistically, GIPC1 reduced the ubiquitination level of tetratricopeptide repeat domain 7B (TTC7B) by downregulating the E3 ubiquitin ligase TRIM21, thereby stabilizing TTC7B's expression and inhibiting the downstream mTOR/NF-κB signaling cascade. Moreover, <i>in vivo</i> studies confirmed the inhibitory role of GIPC1 in CRC growth and found that GIPC1-loaded lipid nanoparticles (GIPC1-LNPs) combined with 5-FU treatment had a more significant antitumor effect. In conclusion, this study reveals the GIPC1/TRIM21/TTC7B/mTOR/NF-κB tumor-suppressive axis in CRC and highlights the potential of GIPC1 for early diagnosis and overcoming chemoresistance in CRC patients.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"22 2","pages":"786-801"},"PeriodicalIF":10.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12781073/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951822","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Co-targeting MRPS7-23 synergistically enhances cisplatin efficacy to suppress nasopharyngeal carcinoma growth and metastasis. 共靶向MRPS7-23可协同增强顺铂抑制鼻咽癌生长转移的疗效。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 DOI: 10.7150/ijbs.115523
Zhangqi Cao, Can Pan, Zeyu Liu, Qi Quan, Mengping Li, Yu Huang, Chuwen Liang, Yuwen Chen, Teng Fan, Ping Chen, Fu Kai, Shuangli Zhu, Sijia Li, Xin Su, Fang Wang, Liwu Fu, Bei Zhang

While cisplatin-based chemoradiotherapy regimens (gemcitabine-cisplatin [GP] and docetaxel-cisplatin-5-fluorouracil [TPF]) remain standard treatments for advanced nasopharyngeal carcinoma (NPC), 30-40% of patients exhibit intrinsic chemoresistance, resulting in therapeutic failure. The molecular underpinnings of this resistance are poorly characterized. Through integrative multi-omics profiling, we identified Mitochondrial Ribosomal Protein S7 (MRPS7) and Mitochondrial Ribosomal Protein S23 (MRPS23) as novel drivers of cisplatin resistance in NPC. Mechanistically, integrated single-cell RNA-seq (scRNA-seq) analysis, mass spectrometry, and functional studies revealed that MRPS7 and MRPS23 stabilized β-catenin by inhibiting its ubiquitination, thereby promoting β-catenin-mediated cancer stemness and epithelial-mesenchymal transition (EMT) to establish cisplatin resistance in NPC. Additionally, we identified Ubiquitin Specific Peptidase 10 (USP10) as a critical upstream regulator that protects MRPS7/23 from proteasomal degradation and sustaining their oncogenic activity. Notably, Spautin-1, a potent USP10 inhibitor, demonstrates synergistic therapeutic activity with cisplatin in diminished tumor growth and metastasis in NPC mice. This research established the USP10-MRPS7/MRPS23-β-catenin axis as a promising precision medicine strategy to combat metastatic dissemination and reverse cisplatin chemoresistance in advanced NPC, which offers a promising opportunity to develop cisplatin sensitizers for the clinical translation of NPC therapies.

虽然以顺铂为基础的放化疗方案(吉西他滨-顺铂[GP]和多西他赛-顺铂-5-氟尿嘧啶[TPF])仍然是晚期鼻咽癌(NPC)的标准治疗方案,但30-40%的患者表现出内在的化疗耐药,导致治疗失败。这种耐药性的分子基础尚未得到很好的表征。通过综合多组学分析,我们发现线粒体核糖体蛋白S7 (MRPS7)和线粒体核糖体蛋白S23 (MRPS23)是鼻咽癌顺铂耐药的新驱动因素。机制上,综合单细胞RNA-seq (scRNA-seq)分析、质谱分析和功能研究表明,MRPS7和MRPS23通过抑制β-catenin的泛素化来稳定β-catenin,从而促进β-catenin介导的肿瘤干细胞和上皮-间质转化(EMT),从而在鼻咽癌中建立顺铂耐药。此外,我们发现泛素特异性肽酶10 (USP10)是一个关键的上游调节因子,可以保护MRPS7/23免受蛋白酶体降解并维持其致癌活性。值得注意的是,Spautin-1,一种有效的USP10抑制剂,在NPC小鼠中显示出与顺铂协同治疗抑制肿瘤生长和转移的活性。本研究确立了USP10-MRPS7/MRPS23-β-catenin轴作为打击晚期鼻咽癌转移传播和逆转顺铂化疗耐药的有前景的精准医学策略,这为开发用于鼻咽癌治疗临床转化的顺铂增敏剂提供了有希望的机会。
{"title":"Co-targeting MRPS7-23 synergistically enhances cisplatin efficacy to suppress nasopharyngeal carcinoma growth and metastasis.","authors":"Zhangqi Cao, Can Pan, Zeyu Liu, Qi Quan, Mengping Li, Yu Huang, Chuwen Liang, Yuwen Chen, Teng Fan, Ping Chen, Fu Kai, Shuangli Zhu, Sijia Li, Xin Su, Fang Wang, Liwu Fu, Bei Zhang","doi":"10.7150/ijbs.115523","DOIUrl":"10.7150/ijbs.115523","url":null,"abstract":"<p><p>While cisplatin-based chemoradiotherapy regimens (gemcitabine-cisplatin [GP] and docetaxel-cisplatin-5-fluorouracil [TPF]) remain standard treatments for advanced nasopharyngeal carcinoma (NPC), 30-40% of patients exhibit intrinsic chemoresistance, resulting in therapeutic failure. The molecular underpinnings of this resistance are poorly characterized. Through integrative multi-omics profiling, we identified Mitochondrial Ribosomal Protein S7 (MRPS7) and Mitochondrial Ribosomal Protein S23 (MRPS23) as novel drivers of cisplatin resistance in NPC. Mechanistically, integrated single-cell RNA-seq (scRNA-seq) analysis, mass spectrometry, and functional studies revealed that MRPS7 and MRPS23 stabilized β-catenin by inhibiting its ubiquitination, thereby promoting β-catenin-mediated cancer stemness and epithelial-mesenchymal transition (EMT) to establish cisplatin resistance in NPC. Additionally, we identified Ubiquitin Specific Peptidase 10 (USP10) as a critical upstream regulator that protects MRPS7/23 from proteasomal degradation and sustaining their oncogenic activity. Notably, Spautin-1, a potent USP10 inhibitor, demonstrates synergistic therapeutic activity with cisplatin in diminished tumor growth and metastasis in NPC mice. This research established the USP10-MRPS7/MRPS23-β-catenin axis as a promising precision medicine strategy to combat metastatic dissemination and reverse cisplatin chemoresistance in advanced NPC, which offers a promising opportunity to develop cisplatin sensitizers for the clinical translation of NPC therapies.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"22 2","pages":"970-994"},"PeriodicalIF":10.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12781175/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951878","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
RNA Methylation in Cancer Metabolism: from Mechanisms to Therapeutic Opportunities. RNA甲基化在癌症代谢中的作用:从机制到治疗机会。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 DOI: 10.7150/ijbs.124177
Zeyu Wu, Yuncan Xing, Shiwen Mei, Tixian Xiao, Fangze Wei, Qian Liu

One of the most important changes in the transformation of normal cells into tumor cells is metabolism. In order to satisfy the more active proliferation, migration and metastasis of cancer cells, abnormal changes occur in various pathways and molecules involved in metabolism, which eventually lead to metabolic reprogramming of tumor cells. This process involves the uptake of nutrients and changes in major metabolic forms. As an important part of post-transcriptional epigenetics, RNA methylation modifications can regulate RNA processing and metabolism, while dynamically and reversibly influencing the expression of specific molecules, thereby ultimately affecting diverse biological processes and cellular phenotypes. In this review, various types of RNA methylation modifications involved in cancer are summarized. Subsequently, we systematically elucidate the mechanism of RNA modification for metabolic reprogramming in cancer, including glucose, lipid, amino acid and mitochondrial metabolism. Most importantly, we discuss in depth the clinical significance of RNA modification in metabolic targeted therapy and immunotherapy from mechanism to therapeutic application.

正常细胞向肿瘤细胞转化过程中最重要的变化之一是代谢。为了满足癌细胞更加活跃的增殖、迁移和转移,参与代谢的各种途径和分子发生异常变化,最终导致肿瘤细胞的代谢重编程。这一过程涉及营养物质的吸收和主要代谢形式的变化。RNA甲基化修饰作为转录后表观遗传学的重要组成部分,可以调控RNA加工和代谢,同时动态可逆地影响特定分子的表达,从而最终影响多种生物过程和细胞表型。本文综述了与癌症相关的各种类型的RNA甲基化修饰。随后,我们系统地阐明了RNA修饰在癌症中代谢重编程的机制,包括葡萄糖、脂质、氨基酸和线粒体代谢。最重要的是,我们从机制到治疗应用,深入讨论了RNA修饰在代谢靶向治疗和免疫治疗中的临床意义。
{"title":"RNA Methylation in Cancer Metabolism: from Mechanisms to Therapeutic Opportunities.","authors":"Zeyu Wu, Yuncan Xing, Shiwen Mei, Tixian Xiao, Fangze Wei, Qian Liu","doi":"10.7150/ijbs.124177","DOIUrl":"10.7150/ijbs.124177","url":null,"abstract":"<p><p>One of the most important changes in the transformation of normal cells into tumor cells is metabolism. In order to satisfy the more active proliferation, migration and metastasis of cancer cells, abnormal changes occur in various pathways and molecules involved in metabolism, which eventually lead to metabolic reprogramming of tumor cells. This process involves the uptake of nutrients and changes in major metabolic forms. As an important part of post-transcriptional epigenetics, RNA methylation modifications can regulate RNA processing and metabolism, while dynamically and reversibly influencing the expression of specific molecules, thereby ultimately affecting diverse biological processes and cellular phenotypes. In this review, various types of RNA methylation modifications involved in cancer are summarized. Subsequently, we systematically elucidate the mechanism of RNA modification for metabolic reprogramming in cancer, including glucose, lipid, amino acid and mitochondrial metabolism. Most importantly, we discuss in depth the clinical significance of RNA modification in metabolic targeted therapy and immunotherapy from mechanism to therapeutic application.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"22 2","pages":"920-950"},"PeriodicalIF":10.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12781177/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951879","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IGF2BP2 Drives Thyroid Cancer Dedifferentiation Through m6A-Dependent STAT1 mRNA Destabilization. IGF2BP2通过m6a依赖性STAT1 mRNA不稳定驱动甲状腺癌去分化。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 DOI: 10.7150/ijbs.121503
Rui Chen, Yi-Xun Li, Wei-Lin Lu, Ke-Fei Wu, Yu-Xin Wang, Zi-Wen Wang, Yi-Han Li, Hai-Yan Yang, Xu Zhang, Liang Shi, Dong Zhou, Ying Wang, Qiang Ding

Thyroid cancer is the most common endocrine malignancy globally. While papillary thyroid carcinoma (PTC) typically exhibits favorable prognosis, a subset undergoes dedifferentiation into anaplastic thyroid carcinoma (ATC), an aggressive, treatment-refractory subtype with near-universal lethality. However, the molecular driver of this process remains elusive. In this study, we find that IGF2BP2 is upregulated in ATC and correlates with adverse prognosis. Pseudotime trajectory analysis tracks progressively escalating IGF2BP2 expression throughout dedifferentiation. Functionally, IGF2BP2 promotes proliferation, suppresses thyroid differentiation genes (TSHR, SLC26A4, SLC5A5, TPO, PAX8, FOXE1, and NKX2.1), and enhances cancer stemness. Mechanistically, integrated multi-omics analysis (RNA-seq, RIP-seq, and MeRIP-seq) reveals that IGF2BP2 binds m6A-modified STAT1 mRNA, accelerating its decay. STAT1 directly activates transcription of thyroid differentiation genes. Rescue experiments confirms that STAT1 mediates IGF2BP2-driven dedifferentiation. The IGF2BP2-m6A-STAT1 complex is a master regulator of thyroid cancer dedifferentiation, establishing a novel therapeutic target for redifferentiation therapy in advanced thyroid cancer.

甲状腺癌是全球最常见的内分泌恶性肿瘤。虽然甲状腺乳头状癌(PTC)通常表现出良好的预后,但一个亚群会经历去分化为间变性甲状腺癌(ATC),这是一种侵袭性的、治疗难治的亚型,几乎具有普遍的致死率。然而,这一过程的分子驱动仍然难以捉摸。本研究中,我们发现IGF2BP2在ATC中表达上调,并与不良预后相关。伪时间轨迹分析追踪IGF2BP2在整个去分化过程中逐渐升高的表达。在功能上,IGF2BP2促进细胞增殖,抑制甲状腺分化基因(TSHR、SLC26A4、SLC5A5、TPO、PAX8、FOXE1、NKX2.1),增强肿瘤的发生。机制上,综合多组学分析(RNA-seq, RIP-seq和MeRIP-seq)显示IGF2BP2结合m6a修饰的STAT1 mRNA,加速其衰变。STAT1直接激活甲状腺分化基因的转录。援救实验证实STAT1介导igf2bp2驱动的去分化。IGF2BP2-m6A-STAT1复合物是甲状腺癌去分化的主要调控因子,为晚期甲状腺癌的再分化治疗建立了新的治疗靶点。
{"title":"IGF2BP2 Drives Thyroid Cancer Dedifferentiation Through m6A-Dependent STAT1 mRNA Destabilization.","authors":"Rui Chen, Yi-Xun Li, Wei-Lin Lu, Ke-Fei Wu, Yu-Xin Wang, Zi-Wen Wang, Yi-Han Li, Hai-Yan Yang, Xu Zhang, Liang Shi, Dong Zhou, Ying Wang, Qiang Ding","doi":"10.7150/ijbs.121503","DOIUrl":"10.7150/ijbs.121503","url":null,"abstract":"<p><p>Thyroid cancer is the most common endocrine malignancy globally. While papillary thyroid carcinoma (PTC) typically exhibits favorable prognosis, a subset undergoes dedifferentiation into anaplastic thyroid carcinoma (ATC), an aggressive, treatment-refractory subtype with near-universal lethality. However, the molecular driver of this process remains elusive. In this study, we find that IGF2BP2 is upregulated in ATC and correlates with adverse prognosis. Pseudotime trajectory analysis tracks progressively escalating IGF2BP2 expression throughout dedifferentiation. Functionally, IGF2BP2 promotes proliferation, suppresses thyroid differentiation genes (<i>TSHR</i>, <i>SLC26A4</i>, <i>SLC5A5</i>, <i>TPO</i>, <i>PAX8</i>, <i>FOXE1</i>, and <i>NKX2.1</i>), and enhances cancer stemness. Mechanistically, integrated multi-omics analysis (RNA-seq, RIP-seq, and MeRIP-seq) reveals that IGF2BP2 binds m6A-modified <i>STAT1</i> mRNA, accelerating its decay. STAT1 directly activates transcription of thyroid differentiation genes. Rescue experiments confirms that STAT1 mediates IGF2BP2-driven dedifferentiation. The IGF2BP2-m6A-STAT1 complex is a master regulator of thyroid cancer dedifferentiation, establishing a novel therapeutic target for redifferentiation therapy in advanced thyroid cancer.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"22 2","pages":"622-640"},"PeriodicalIF":10.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12780945/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951887","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Crosstalk between arachidonic acid metabolism and glycolysis drives integrated metabolic-inflammatory reprogramming in macrophages. 花生四烯酸代谢和糖酵解之间的串扰驱动巨噬细胞的综合代谢-炎症重编程。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 DOI: 10.7150/ijbs.116671
Yuanyuan Cheng, Ni Fan, Xiuying Zhang, Wei Zhao, Baoping Xie, Jia Zhao, Marlene Rong, Xuechen Li, Hung-Fat Tse, Jianhui Rong

Arachidonic acid (AA)-derived lipid mediators play pivotal roles in inflammation and its resolution. While glycolysis is a key metabolic pathway determining macrophage polarization, the crosstalk between specific AA metabolites and glycolytic reprogramming remains poorly understood. In this study, we explore whether certain AA metabolites modulate macrophage function through covalent protein modification, with therapeutic implications for myocardial ischemia-reperfusion injury. Unlike conventional specialized pro-resolving mediators (SPMs) that primarily act via receptors, here we identify an endogenous electrophilic AA metabolite, 15-keto-prostaglandin F2α (15KPF), that covalently modifies pyruvate kinase M2 (PKM2) at Cys49. Such interaction enhanced PKM2 tetramerization, suppressed the PKM2/HIF-1α/STAT3 axis, redirected energy metabolism from glycolysis to mitochondrial respiration, and promoted pro-resolving M2 macrophage polarization. Mutated PKM2(C49S) failed to inhibit STAT3 signaling and blocked the effect of 15KPF on M1 to M2 phenotype switch. Moreover, 15KPF reduced infarct size and preserved myocardial integrity in in vivo model. Taken together, covalent 15-keto-PGF2α-PKM2 conjugation represents a self-regulatory mechanism linking AA metabolism to glycolysis to drive macrophage metabolic-inflammatory reprogramming. This pathway positions 15KPF as a promising therapeutic candidate for inflammatory and metabolic diseases, including ischemia-reperfusion injury, and distinguishes it from synthetic allosteric PKM2 activators such as TEPP-46.

花生四烯酸(AA)衍生的脂质介质在炎症及其消退中起关键作用。虽然糖酵解是决定巨噬细胞极化的关键代谢途径,但特定AA代谢物与糖酵解重编程之间的串扰仍然知之甚少。在这项研究中,我们探讨了某些AA代谢物是否通过共价蛋白修饰来调节巨噬细胞的功能,从而对心肌缺血再灌注损伤具有治疗意义。与传统的主要通过受体起作用的特殊促分解介质(SPMs)不同,本研究发现了一种内源性亲电性AA代谢物15-酮前列腺素F2α (15KPF),它在Cys49位点共价修饰丙酮酸激酶M2 (PKM2)。这种相互作用增强PKM2四聚化,抑制PKM2/HIF-1α/STAT3轴,将能量代谢从糖酵解重定向到线粒体呼吸,并促进促分解M2巨噬细胞极化。突变的PKM2(C49S)未能抑制STAT3信号传导,并阻断了15KPF对M1到M2表型转换的作用。此外,在体内模型中,15KPF可减小梗死面积并保持心肌完整性。综上所述,共价15-酮- pgf2 α- pkm2偶联代表了一种将AA代谢与糖酵解联系起来的自我调节机制,从而驱动巨噬细胞代谢-炎症重编程。该途径将15KPF定位为炎症和代谢性疾病(包括缺血-再灌注损伤)的有希望的治疗候选者,并将其与合成的变张力PKM2激活剂(如TEPP-46)区分开来。
{"title":"Crosstalk between arachidonic acid metabolism and glycolysis drives integrated metabolic-inflammatory reprogramming in macrophages.","authors":"Yuanyuan Cheng, Ni Fan, Xiuying Zhang, Wei Zhao, Baoping Xie, Jia Zhao, Marlene Rong, Xuechen Li, Hung-Fat Tse, Jianhui Rong","doi":"10.7150/ijbs.116671","DOIUrl":"10.7150/ijbs.116671","url":null,"abstract":"<p><p>Arachidonic acid (AA)-derived lipid mediators play pivotal roles in inflammation and its resolution. While glycolysis is a key metabolic pathway determining macrophage polarization, the crosstalk between specific AA metabolites and glycolytic reprogramming remains poorly understood. In this study, we explore whether certain AA metabolites modulate macrophage function through covalent protein modification, with therapeutic implications for myocardial ischemia-reperfusion injury. Unlike conventional specialized pro-resolving mediators (SPMs) that primarily act via receptors, here we identify an endogenous electrophilic AA metabolite, 15-keto-prostaglandin F2α (15KPF), that covalently modifies pyruvate kinase M2 (PKM2) at Cys49. Such interaction enhanced <i>PKM2</i> tetramerization, suppressed the <i>PKM2/HIF-1α/STAT3</i> axis, redirected energy metabolism from glycolysis to mitochondrial respiration, and promoted pro-resolving M2 macrophage polarization. Mutated <i>PKM2</i>(C49S) failed to inhibit STAT3 signaling and blocked the effect of 15KPF on M1 to M2 phenotype switch. Moreover, 15KPF reduced infarct size and preserved myocardial integrity in <i>in vivo</i> model. Taken together, covalent 15-keto-PGF2α-<i>PKM2</i> conjugation represents a self-regulatory mechanism linking AA metabolism to glycolysis to drive macrophage metabolic-inflammatory reprogramming. This pathway positions 15KPF as a promising therapeutic candidate for inflammatory and metabolic diseases, including ischemia-reperfusion injury, and distinguishes it from synthetic allosteric PKM2 activators such as TEPP-46.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"22 2","pages":"771-785"},"PeriodicalIF":10.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12781075/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951911","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Schisandrin B Targets PXR to Enhance Bile Acid Metabolism and Alleviate ANIT-Induced Cholestatic Liver Injury via Dual Pathways. 五味子素B通过双途径靶向PXR促进胆汁酸代谢,减轻抗炎药诱导的胆汁淤积性肝损伤。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 DOI: 10.7150/ijbs.121475
Ying Zhang, Huan Lan, Xuechun Yu, Lin Zhuo, Bixin Zhao, Fang Liu, Lin An, Fan Zhang, Zhongqiu Liu, Caiyan Wang

Cholestatic liver injury (CLI) is a rapid progressive liver disorder characterized by the accumulation of bile acids (BA). Although pregnane X receptor (PXR) is a critical regulator of BA metabolism, the synergistic mechanisms of natural compounds targeting these pathways remain unclear. In this study, we demonstrated a positive correlation between BA accumulation and disease severity in clinical samples. Further, we identified Schisandrin B (Sin B), a lignan from Schisandra chinensis, as a potent hepatoprotective agent in α-naphthyl isothiocyanate (ANIT)- induced CLI. We demonstrated that Sin B treatment reduced BA levels and inflammation in ANIT-induced WRL68 cells, liver lobule chips, and mice. Notably, Sin B activated PXR, increased the levels of UDP-glucuronosyltransferase 1A1 (UGT1A1), CYP3A4 (in humans) / CYP3A11 (in mice) and MRPs, and enhanced TFEB transcriptional activity and autophagic flux in vivo and in vitro. Knockout of hepatic Pxr or Tfeb blocked these effects of Sin B. Mechanistic investigation revealed that Sin B is directly binds to PXR at residues S106, G144, and W299, inducing conformational changes in the ligand-binding domain (LBD) was verified through target fishing, molecular dynamics (MD) simulations, drug affinity responsive target stability assay, isothermal titration calorimetry and surface plasmon resonance. Our findings provide structural and functional insights into the dual-pathway mechanism of Sin B and support its therapeutic potential for CLI.

胆汁淤积性肝损伤(CLI)是一种以胆汁酸(BA)积累为特征的快速进行性肝脏疾病。虽然妊娠X受体(PXR)是BA代谢的关键调节因子,但天然化合物靶向这些途径的协同机制尚不清楚。在本研究中,我们证明了临床样本中BA积累与疾病严重程度呈正相关。此外,我们发现五味子木脂素五味子素B (sinb)在α-萘异硫氰酸酯(ANIT)诱导的CLI中具有有效的肝保护剂作用。我们证明,在anit诱导的WRL68细胞、肝小叶芯片和小鼠中,Sin B处理降低了BA水平和炎症。值得注意的是,Sin B激活了PXR,增加了udp -葡萄糖醛酸糖基转移酶1A1 (UGT1A1)、CYP3A4(人)/ CYP3A11(小鼠)和MRPs的水平,增强了体内和体外TFEB的转录活性和自噬通量。敲除肝脏Pxr或Tfeb阻断了Sin B的这些作用。机制研究表明,Sin B直接与Pxr残基S106, G144和W299结合,诱导配体结合域(LBD)的构象变化,通过靶捕集,分子动力学(MD)模拟,药物亲和力响应靶稳定性测定,等温滴定量热法和表面等离子体共振验证。我们的研究结果为Sin B的双通路机制提供了结构和功能方面的见解,并支持其治疗CLI的潜力。
{"title":"Schisandrin B Targets PXR to Enhance Bile Acid Metabolism and Alleviate ANIT-Induced Cholestatic Liver Injury via Dual Pathways.","authors":"Ying Zhang, Huan Lan, Xuechun Yu, Lin Zhuo, Bixin Zhao, Fang Liu, Lin An, Fan Zhang, Zhongqiu Liu, Caiyan Wang","doi":"10.7150/ijbs.121475","DOIUrl":"10.7150/ijbs.121475","url":null,"abstract":"<p><p>Cholestatic liver injury (CLI) is a rapid progressive liver disorder characterized by the accumulation of bile acids (BA). Although pregnane X receptor (PXR) is a critical regulator of BA metabolism, the synergistic mechanisms of natural compounds targeting these pathways remain unclear. In this study, we demonstrated a positive correlation between BA accumulation and disease severity in clinical samples. Further, we identified Schisandrin B (Sin B), a lignan from <i>Schisandra chinensis</i>, as a potent hepatoprotective agent in α-naphthyl isothiocyanate (ANIT)- induced CLI. We demonstrated that Sin B treatment reduced BA levels and inflammation in ANIT-induced WRL68 cells, liver lobule chips, and mice. Notably, Sin B activated PXR, increased the levels of UDP-glucuronosyltransferase 1A1 (UGT1A1), CYP3A4 (in humans) / CYP3A11 (in mice) and MRPs, and enhanced TFEB transcriptional activity and autophagic flux <i>in vivo</i> and <i>in vitro</i>. Knockout of hepatic <i>Pxr</i> or <i>Tfeb</i> blocked these effects of Sin B. Mechanistic investigation revealed that Sin B is directly binds to PXR at residues S106, G144, and W299, inducing conformational changes in the ligand-binding domain (LBD) was verified through target fishing, molecular dynamics (MD) simulations, drug affinity responsive target stability assay, isothermal titration calorimetry and surface plasmon resonance. Our findings provide structural and functional insights into the dual-pathway mechanism of Sin B and support its therapeutic potential for CLI.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"22 1","pages":"387-409"},"PeriodicalIF":10.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12681940/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145707640","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
International Journal of Biological Sciences
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1