首页 > 最新文献

Signal Transduction and Targeted Therapy最新文献

英文 中文
Pancreatic cancer: molecular pathogenesis and emerging therapeutic strategies 胰腺癌:分子发病机制和新兴治疗策略
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-03 DOI: 10.1038/s41392-025-02499-y
Enrique Rozengurt, Guido Eibl
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease for which there is no effective treatment. A deep understanding of the mechanisms underlying the molecular pathogenesis, signaling pathways and risk factors leading to PDAC is of paramount importance for identifying novel targets, prognostic markers, preventive strategies, and signature markers for use in specific and personalized therapeutic procedures. Activating somatic mutations in the KRAS oncogene play a critical role in PDAC initiation and maintenance. Here, we highlight the complex interplay between KRAS signaling, the transcriptional coactivator YES1-associated protein (YAP) and Src family kinases (SFKs) in the pathogenesis of PDAC and drug sensitivity. We subsequently focused on diet-induced obesity, which has been correlated with an increased risk for developing PDAC in humans and mice and more severe clinical outcomes. Accumulating evidence also indicates that neural signals regulate critical functions of cancer cells, including their proliferation and dissemination, and that chronic stress promotes PDAC through the sympathetic nervous system via β-adrenergic receptors expressed by PDAC cells and other cells in the tumor microenvironment. Obesogenic mediators and stress neurotransmitters stimulate protein kinases, including PKA and PKD, which converge on CREB/ATF1 phosphorylation in PDAC cells. Since stress and obesity cooperate to promote the progression of PDAC, novel combinatorial strategies to prevent this devastating disease could be developed, repositioning FDA-approved drugs that are extensively used to treat cardiovascular and metabolic disorders and diseases. Finally, we review new advances in the treatment of PDAC, focusing on the discovery of novel drugs that directly inhibit KRAS and YAP function.
胰腺导管腺癌(PDAC)是一种侵袭性疾病,目前尚无有效治疗方法。深入了解导致PDAC的分子发病机制、信号通路和风险因素对于确定新的靶点、预后标志物、预防策略和用于特定和个性化治疗程序的标志性标志物至关重要。激活KRAS癌基因的体细胞突变在PDAC的启动和维持中起着关键作用。在这里,我们强调了KRAS信号,转录辅助激活因子yes1相关蛋白(YAP)和Src家族激酶(SFKs)在PDAC发病机制和药物敏感性中的复杂相互作用。我们随后将重点放在饮食引起的肥胖上,这与人类和小鼠发生PDAC的风险增加以及更严重的临床结果相关。越来越多的证据还表明,神经信号调节癌细胞的关键功能,包括癌细胞的增殖和传播,慢性应激通过交感神经系统通过PDAC细胞和肿瘤微环境中其他细胞表达的β-肾上腺素能受体促进PDAC。致肥介质和应激神经递质刺激蛋白激酶,包括PKA和PKD,它们聚集在PDAC细胞中CREB/ATF1磷酸化。由于压力和肥胖共同促进PDAC的进展,可以开发新的组合策略来预防这种毁灭性疾病,重新定位fda批准的广泛用于治疗心血管和代谢紊乱和疾病的药物。最后,我们回顾了PDAC治疗的新进展,重点介绍了直接抑制KRAS和YAP功能的新药物的发现。
{"title":"Pancreatic cancer: molecular pathogenesis and emerging therapeutic strategies","authors":"Enrique Rozengurt, Guido Eibl","doi":"10.1038/s41392-025-02499-y","DOIUrl":"https://doi.org/10.1038/s41392-025-02499-y","url":null,"abstract":"Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease for which there is no effective treatment. A deep understanding of the mechanisms underlying the molecular pathogenesis, signaling pathways and risk factors leading to PDAC is of paramount importance for identifying novel targets, prognostic markers, preventive strategies, and signature markers for use in specific and personalized therapeutic procedures. Activating somatic mutations in the <jats:italic>KRAS</jats:italic> oncogene play a critical role in PDAC initiation and maintenance. Here, we highlight the complex interplay between KRAS signaling, the transcriptional coactivator YES1-associated protein (YAP) and Src family kinases (SFKs) in the pathogenesis of PDAC and drug sensitivity. We subsequently focused on diet-induced obesity, which has been correlated with an increased risk for developing PDAC in humans and mice and more severe clinical outcomes. Accumulating evidence also indicates that neural signals regulate critical functions of cancer cells, including their proliferation and dissemination, and that chronic stress promotes PDAC through the sympathetic nervous system via β-adrenergic receptors expressed by PDAC cells and other cells in the tumor microenvironment. Obesogenic mediators and stress neurotransmitters stimulate protein kinases, including PKA and PKD, which converge on CREB/ATF1 phosphorylation in PDAC cells. Since stress and obesity cooperate to promote the progression of PDAC, novel combinatorial strategies to prevent this devastating disease could be developed, repositioning FDA-approved drugs that are extensively used to treat cardiovascular and metabolic disorders and diseases. Finally, we review new advances in the treatment of PDAC, focusing on the discovery of novel drugs that directly inhibit KRAS and YAP function.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"44 1","pages":"6"},"PeriodicalIF":39.3,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145893970","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cell death: targeting ferroptosis in cancer 细胞死亡:针对癌症中的铁下垂
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-03 DOI: 10.1038/s41392-025-02534-y
Hayley M. Sabol, Lorenzo Galluzzi, Lucia Borriello
{"title":"Cell death: targeting ferroptosis in cancer","authors":"Hayley M. Sabol, Lorenzo Galluzzi, Lucia Borriello","doi":"10.1038/s41392-025-02534-y","DOIUrl":"https://doi.org/10.1038/s41392-025-02534-y","url":null,"abstract":"","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"359 1","pages":"5"},"PeriodicalIF":39.3,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145893971","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Remodeling the tumor dormancy ecosystem to prevent recurrence and metastasis 重塑肿瘤休眠生态系统,防止复发和转移
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-03 DOI: 10.1038/s41392-025-02328-2
Yu Liang, Wo-Ming Chen, Youming Zhang, Lei Li
Dormant tumor cells, major contributors to tumor recurrence and metastasis, are characterized by cell cycle arrest and reactivation potential. Tumor dormancy arises from the dynamic interplay between intrinsic tumor properties and extrinsic factors within the tumor ecosystem. This ecosystem operates at two distinct levels: the tumor microenvironment (TME) and the systemic macroenvironment (SME). Within the dormant TME, tumor cells engage in complex interactions with surrounding stromal cells, extracellular matrix components, and the vasculature, which are mediated through growth factors, cytokines, and metabolic byproducts. At the systemic level, the SME modulates tumor dormancy via inflammatory responses, metabolic homeostasis, hormonal regulation, and neural signaling. The TME and SME collectively maintain tumor dormancy through their bidirectional crosstalk. Disruption of this delicate ecological equilibrium can trigger tumor reactivation and metastatic progression. Consequently, effective therapeutic strategies should simultaneously target both TME remodeling and SME modulation. In this review, we provide a comprehensive analysis of the coordinated roles of the TME and SME in regulating tumor cell dormancy and reactivation while summarizing potential therapeutic approaches and clinical trials aimed at either eliminating dormant tumor cells or sustaining dormancy. Consequently, we propose a novel two-dimensional combined treatment strategy that concurrently addresses both the TME and SME to prevent tumor recurrence and metastasis.
休眠肿瘤细胞具有细胞周期阻滞和再激活潜能,是肿瘤复发和转移的主要因素。肿瘤休眠是肿瘤内在特性与肿瘤生态系统外部因素动态相互作用的结果。这个生态系统在两个不同的层面上运作:肿瘤微环境(TME)和系统宏观环境(SME)。在休眠TME中,肿瘤细胞通过生长因子、细胞因子和代谢副产物介导与周围基质细胞、细胞外基质成分和脉管系统进行复杂的相互作用。在系统水平上,SME通过炎症反应、代谢稳态、激素调节和神经信号传导调节肿瘤休眠。TME和SME通过它们的双向串扰共同维持肿瘤休眠。这种微妙的生态平衡的破坏可以触发肿瘤的再激活和转移进展。因此,有效的治疗策略应该同时针对TME重塑和SME调节。在这篇综述中,我们全面分析了TME和SME在调节肿瘤细胞休眠和再激活中的协同作用,同时总结了旨在消除休眠肿瘤细胞或维持休眠的潜在治疗方法和临床试验。因此,我们提出了一种新的二维联合治疗策略,同时解决TME和SME,以防止肿瘤复发和转移。
{"title":"Remodeling the tumor dormancy ecosystem to prevent recurrence and metastasis","authors":"Yu Liang, Wo-Ming Chen, Youming Zhang, Lei Li","doi":"10.1038/s41392-025-02328-2","DOIUrl":"https://doi.org/10.1038/s41392-025-02328-2","url":null,"abstract":"Dormant tumor cells, major contributors to tumor recurrence and metastasis, are characterized by cell cycle arrest and reactivation potential. Tumor dormancy arises from the dynamic interplay between intrinsic tumor properties and extrinsic factors within the tumor ecosystem. This ecosystem operates at two distinct levels: the tumor microenvironment (TME) and the systemic macroenvironment (SME). Within the dormant TME, tumor cells engage in complex interactions with surrounding stromal cells, extracellular matrix components, and the vasculature, which are mediated through growth factors, cytokines, and metabolic byproducts. At the systemic level, the SME modulates tumor dormancy via inflammatory responses, metabolic homeostasis, hormonal regulation, and neural signaling. The TME and SME collectively maintain tumor dormancy through their bidirectional crosstalk. Disruption of this delicate ecological equilibrium can trigger tumor reactivation and metastatic progression. Consequently, effective therapeutic strategies should simultaneously target both TME remodeling and SME modulation. In this review, we provide a comprehensive analysis of the coordinated roles of the TME and SME in regulating tumor cell dormancy and reactivation while summarizing potential therapeutic approaches and clinical trials aimed at either eliminating dormant tumor cells or sustaining dormancy. Consequently, we propose a novel two-dimensional combined treatment strategy that concurrently addresses both the TME and SME to prevent tumor recurrence and metastasis.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"88 1","pages":"1"},"PeriodicalIF":39.3,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145895593","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Signaling pathways and targeted interventions for precancers 癌症前期的信号通路和靶向干预
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-01 DOI: 10.1038/s41392-025-02342-4
Jin Yang, Shimeng Wang, Xin Li, Hongdan Xu, Tongxu Sun, Tao Hu, Jingjing Luo, Hongmei Zhou
Precancers, defined as normal-appearing or morphologically altered tissues with a risk of oncogenesis, exhibit various detectable manifestations across anatomical sites, including epithelial dysplasia, metaplasia, hyperplasia, and stromal fibrosis. Considering the prevailing assumption that most cancers arise from precancers, early intervention at the precancerous stage has immense potential to reduce cancer-related morbidity and mortality. However, the complex signaling networks governing precancer initiation and progression remain elusive, hampering the development of effective targeted interventions. This review synthesizes three critical dimensions of precancer biology: historical foundations tracing the conceptual evolution of precancer research over the past century; mechanisms underlying the multistep progression of precancer biology, encompassing epithelial and macro/microenvironmental remodeling; and signaling networks cataloging dysregulated pathways and their therapeutic potential. Over 10 signaling pathways, including the transforming growth factor-β (TGF-β), p53, Wnt, phosphatidylinositol 3-kinase (PI3K), and mitogen-activated protein kinase (MAPK) pathways, drive multistep malignant transformation. We further synthesize emerging evidence supporting microenvironmental dominance, proposing the novel “soil degeneration” hypothesis. This paradigm shift underscores the necessity for dual-window intervention in which early-phase microenvironmental normalization prevents the establishment of precancerous lesions and advanced-phase treatment concurrently addresses epithelial malignancy and stromal degeneration. This review bridges foundational molecular discoveries with translational clinical potential and advocates for precision intervention frameworks that extend from biomarker-guided risk assessment to synergistic remodeling of the precancer microenvironment, thereby redefining precancer intervention in the molecularly targeted era.
癌前病变被定义为外观正常或形态学改变的组织,具有致癌风险,在解剖部位表现出各种可检测的表现,包括上皮发育不良、化生、增生和间质纤维化。考虑到大多数癌症起源于癌前病变的普遍假设,在癌前病变阶段进行早期干预具有降低癌症相关发病率和死亡率的巨大潜力。然而,控制癌前病变起始和进展的复杂信号网络仍然难以捉摸,阻碍了有效靶向干预措施的发展。这篇综述综合了癌前生物学的三个关键方面:追溯过去一个世纪癌前研究的概念演变的历史基础;包括上皮和宏观/微环境重塑在内的癌前生物学多步骤进展的机制;信号网络编目失调通路及其治疗潜力。转化生长因子-β (TGF-β)、p53、Wnt、磷脂酰肌醇3-激酶(PI3K)、丝裂原活化蛋白激酶(MAPK)等10多种信号通路驱动多步骤恶性转化。我们进一步综合了支持微环境优势的新证据,提出了新的“土壤退化”假说。这种模式的转变强调了双窗口干预的必要性,在双窗口干预中,早期微环境正常化可以防止癌前病变的建立,而晚期治疗可以同时解决上皮恶性肿瘤和间质变性。本综述将基础分子发现与转化临床潜力联系起来,并倡导从生物标志物引导的风险评估到癌前微环境的协同重塑的精确干预框架,从而重新定义分子靶向时代的癌前干预。
{"title":"Signaling pathways and targeted interventions for precancers","authors":"Jin Yang, Shimeng Wang, Xin Li, Hongdan Xu, Tongxu Sun, Tao Hu, Jingjing Luo, Hongmei Zhou","doi":"10.1038/s41392-025-02342-4","DOIUrl":"https://doi.org/10.1038/s41392-025-02342-4","url":null,"abstract":"Precancers, defined as normal-appearing or morphologically altered tissues with a risk of oncogenesis, exhibit various detectable manifestations across anatomical sites, including epithelial dysplasia, metaplasia, hyperplasia, and stromal fibrosis. Considering the prevailing assumption that most cancers arise from precancers, early intervention at the precancerous stage has immense potential to reduce cancer-related morbidity and mortality. However, the complex signaling networks governing precancer initiation and progression remain elusive, hampering the development of effective targeted interventions. This review synthesizes three critical dimensions of precancer biology: historical foundations tracing the conceptual evolution of precancer research over the past century; mechanisms underlying the multistep progression of precancer biology, encompassing epithelial and macro/microenvironmental remodeling; and signaling networks cataloging dysregulated pathways and their therapeutic potential. Over 10 signaling pathways, including the transforming growth factor-β (TGF-β), p53, Wnt, phosphatidylinositol 3-kinase (PI3K), and mitogen-activated protein kinase (MAPK) pathways, drive multistep malignant transformation. We further synthesize emerging evidence supporting microenvironmental dominance, proposing the novel “soil degeneration” hypothesis. This paradigm shift underscores the necessity for dual-window intervention in which early-phase microenvironmental normalization prevents the establishment of precancerous lesions and advanced-phase treatment concurrently addresses epithelial malignancy and stromal degeneration. This review bridges foundational molecular discoveries with translational clinical potential and advocates for precision intervention frameworks that extend from biomarker-guided risk assessment to synergistic remodeling of the precancer microenvironment, thereby redefining precancer intervention in the molecularly targeted era.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"15 1","pages":""},"PeriodicalIF":39.3,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145903751","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
When vaccines reset tumors: SARS-CoV-2 mRNA shots create a transient checkpoint-sensitive state. 当疫苗重置肿瘤时:SARS-CoV-2 mRNA注射会产生短暂的检查点敏感状态。
IF 52.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-31 DOI: 10.1038/s41392-025-02521-3
Hao Chi, Michele Carbone, Youping Deng
{"title":"When vaccines reset tumors: SARS-CoV-2 mRNA shots create a transient checkpoint-sensitive state.","authors":"Hao Chi, Michele Carbone, Youping Deng","doi":"10.1038/s41392-025-02521-3","DOIUrl":"10.1038/s41392-025-02521-3","url":null,"abstract":"","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"10 1","pages":"423"},"PeriodicalIF":52.7,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12753749/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145864731","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
mTOR signaling networks: mechanistic insights and translational frontiers in disease therapeutics. mTOR信号网络:疾病治疗的机制见解和翻译前沿。
IF 52.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-30 DOI: 10.1038/s41392-025-02493-4
Hanxiao Zhang, Xia Xiao, Zhenrui Pan, Svetlana Dokudovskaya

The mammalian target of rapamycin (mTOR) pathway is a central regulator of cellular growth, metabolism, and homeostasis, integrating a wide array of intracellular and extracellular cues, including nutrient availability, growth factors, and cellular stress, to coordinate anabolic and catabolic processes such as protein, lipid, and nucleotide synthesis; autophagy; and proteasomal degradation. The dysregulation of this signaling hub has broad implications for health and disease. To commemorate the 50th anniversary of the discovery of rapamycin, we provide a comprehensive synthesis of five decades of mTOR research. This review traces the historical trajectory from the early characterization of the biological effects of rapamycin to the elucidation of its molecular target and downstream pathways. We integrate fundamental and emerging insights into the roles of mTOR across nearly all domains of cell biology and development, with a particular focus on the expanding landscape of therapeutic interventions targeting this pathway. Special emphasis is placed on the crosstalk between mTOR signaling and mitochondrial regulation, highlighting the mechanisms by which these two metabolic hubs co-regulate cellular adaptation, survival, and disease progression. The dynamic interplay between mTOR and mitochondrial networks governs key aspects of bioenergetics, redox balance, and cell fate decisions and is increasingly implicated in pathophysiological contexts ranging from cancer and aging to neurodegenerative and immune disorders.

哺乳动物雷帕霉素靶点(mTOR)通路是细胞生长、代谢和稳态的中心调节因子,整合了广泛的细胞内和细胞外信号,包括营养可利用性、生长因子和细胞应激,以协调合成代谢和分解代谢过程,如蛋白质、脂质和核苷酸合成;自噬;和蛋白酶体降解。这个信号中枢的失调对健康和疾病有着广泛的影响。为了纪念雷帕霉素发现50周年,我们提供了一份50年来mTOR研究的综合报告。本文回顾了从雷帕霉素生物学效应的早期表征到其分子靶点和下游途径的阐明的历史轨迹。我们将基本的和新兴的见解整合到几乎所有细胞生物学和发育领域的mTOR的作用,特别关注针对这一途径的治疗干预的扩展景观。特别强调mTOR信号和线粒体调节之间的串扰,强调这两个代谢中心共同调节细胞适应、生存和疾病进展的机制。mTOR和线粒体网络之间的动态相互作用控制着生物能量学、氧化还原平衡和细胞命运决定的关键方面,并且越来越多地涉及从癌症、衰老到神经退行性疾病和免疫疾病的病理生理背景。
{"title":"mTOR signaling networks: mechanistic insights and translational frontiers in disease therapeutics.","authors":"Hanxiao Zhang, Xia Xiao, Zhenrui Pan, Svetlana Dokudovskaya","doi":"10.1038/s41392-025-02493-4","DOIUrl":"10.1038/s41392-025-02493-4","url":null,"abstract":"<p><p>The mammalian target of rapamycin (mTOR) pathway is a central regulator of cellular growth, metabolism, and homeostasis, integrating a wide array of intracellular and extracellular cues, including nutrient availability, growth factors, and cellular stress, to coordinate anabolic and catabolic processes such as protein, lipid, and nucleotide synthesis; autophagy; and proteasomal degradation. The dysregulation of this signaling hub has broad implications for health and disease. To commemorate the 50th anniversary of the discovery of rapamycin, we provide a comprehensive synthesis of five decades of mTOR research. This review traces the historical trajectory from the early characterization of the biological effects of rapamycin to the elucidation of its molecular target and downstream pathways. We integrate fundamental and emerging insights into the roles of mTOR across nearly all domains of cell biology and development, with a particular focus on the expanding landscape of therapeutic interventions targeting this pathway. Special emphasis is placed on the crosstalk between mTOR signaling and mitochondrial regulation, highlighting the mechanisms by which these two metabolic hubs co-regulate cellular adaptation, survival, and disease progression. The dynamic interplay between mTOR and mitochondrial networks governs key aspects of bioenergetics, redox balance, and cell fate decisions and is increasingly implicated in pathophysiological contexts ranging from cancer and aging to neurodegenerative and immune disorders.</p>","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"10 1","pages":"428"},"PeriodicalIF":52.7,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12753737/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145864343","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Low density lipoprotein receptor-related protein 8: a critical receptor for tick-borne encephalitis virus entry. 低密度脂蛋白受体相关蛋白8:蜱传脑炎病毒进入的关键受体
IF 52.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-30 DOI: 10.1038/s41392-025-02509-z
Pengtao Jiao, Lin Du, Yan Zeng
{"title":"Low density lipoprotein receptor-related protein 8: a critical receptor for tick-borne encephalitis virus entry.","authors":"Pengtao Jiao, Lin Du, Yan Zeng","doi":"10.1038/s41392-025-02509-z","DOIUrl":"10.1038/s41392-025-02509-z","url":null,"abstract":"","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"10 1","pages":"422"},"PeriodicalIF":52.7,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12749373/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145857569","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A novel antituberculosis agent exhibits potent clinical efficacy and good safety profile: an open-label, randomized-controlled, multicenter, phase 2a trial. 一种新型抗结核药物显示出强大的临床疗效和良好的安全性:一项开放标签、随机对照、多中心、2a期试验。
IF 52.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-30 DOI: 10.1038/s41392-025-02517-z
Naihui Chu, Wenjuan Nie, Juan Du, Manni Wang, Liping Ma, Qingfeng Wang, Jun Wang, Xiaomeng Hu, Jin Wu, Yu Lu, Mailing Huang, Yuquan Wei, Zhenling Wang, Jian-Qing He, Zhenyu Ding, Xiawei Wei

Tuberculosis (TB) is a contagious disease that threatens human health worldwide. Combination chemotherapy is usually recommended for this disease. Recently, 2 nitroimidazole-based agents, namely, delamanid and pretomanid, have been approved by regulatory agencies. JDB0131 is a novel, structurally optimized third-generation nitroimidazole antituberculosis agent that incorporates the advantages of earlier compounds. This multicenter, prospective, randomized phase 2a trial was conducted to evaluate its efficacy and safety in patients with tuberculosis (NCT06224036). In total, 52 patients with newly diagnosed TB were recruited. JDB0131 was tested in a dose escalation manner (cohort 1: 100 mg bid, cohort 2: 200 mg qd, and cohort 3: 200 mg bid). For comparison, delamanid (100 mg bid) and classic fixed-dose combination (FDC) regimens were included as controls. The primary endpoint was logarithmic changes in the number of colony formation units (CFUs) in the solid media culture of sputum TB (log10 CFU). The early bactericidal activity (EBA) of JDB0131 was better than that of delamanid. During the time interval between days 0 and 14, JDB0131 at a dose of 200 mg bid (cohort 3) showed superior efficacy over delamanid. At the end of drug intervention (day 14), JDB0131 (all 3 dose levels) achieved superior time to positivity (TTP) over delamanid. Ninety-one adverse events (AEs), including no serious AEs, were attributed to JDB0131 in 30 patients. This trial identified a promising new drug for the increasing TB burden worldwide.

结核病是一种在世界范围内威胁人类健康的传染性疾病。这种疾病通常建议联合化疗。最近,两种基于硝基咪唑的药物,即delamanid和pretomanid,已获得监管机构的批准。JDB0131是一种新型的,结构优化的第三代硝基咪唑抗结核药,结合了早期化合物的优点。这项多中心、前瞻性、随机2a期试验旨在评估其在结核病患者中的有效性和安全性(NCT06224036)。总共招募了52名新诊断的结核病患者。JDB0131以剂量递增方式进行试验(队列1:每日100 mg,队列2:每日200 mg,队列3:每日200 mg)。为了进行比较,将delamanid (100mg bid)和经典的固定剂量联合(FDC)方案作为对照。主要终点是痰结核固体培养基中菌落形成单位(CFU)数量的对数变化(log10 CFU)。JDB0131的早期杀菌活性(EBA)优于delamanid。在第0天至第14天的时间间隔内,JDB0131剂量为200mg / bid(队列3)的疗效优于delamanid。在药物干预结束时(第14天),JDB0131(所有3种剂量水平)比delamanid获得了更长的阳性时间(TTP)。30例患者中有91例不良事件(ae)归因于JDB0131,其中无严重ae。该试验确定了一种有希望的新药,可用于治疗全球日益增加的结核病负担。
{"title":"A novel antituberculosis agent exhibits potent clinical efficacy and good safety profile: an open-label, randomized-controlled, multicenter, phase 2a trial.","authors":"Naihui Chu, Wenjuan Nie, Juan Du, Manni Wang, Liping Ma, Qingfeng Wang, Jun Wang, Xiaomeng Hu, Jin Wu, Yu Lu, Mailing Huang, Yuquan Wei, Zhenling Wang, Jian-Qing He, Zhenyu Ding, Xiawei Wei","doi":"10.1038/s41392-025-02517-z","DOIUrl":"10.1038/s41392-025-02517-z","url":null,"abstract":"<p><p>Tuberculosis (TB) is a contagious disease that threatens human health worldwide. Combination chemotherapy is usually recommended for this disease. Recently, 2 nitroimidazole-based agents, namely, delamanid and pretomanid, have been approved by regulatory agencies. JDB0131 is a novel, structurally optimized third-generation nitroimidazole antituberculosis agent that incorporates the advantages of earlier compounds. This multicenter, prospective, randomized phase 2a trial was conducted to evaluate its efficacy and safety in patients with tuberculosis (NCT06224036). In total, 52 patients with newly diagnosed TB were recruited. JDB0131 was tested in a dose escalation manner (cohort 1: 100 mg bid, cohort 2: 200 mg qd, and cohort 3: 200 mg bid). For comparison, delamanid (100 mg bid) and classic fixed-dose combination (FDC) regimens were included as controls. The primary endpoint was logarithmic changes in the number of colony formation units (CFUs) in the solid media culture of sputum TB (log10 CFU). The early bactericidal activity (EBA) of JDB0131 was better than that of delamanid. During the time interval between days 0 and 14, JDB0131 at a dose of 200 mg bid (cohort 3) showed superior efficacy over delamanid. At the end of drug intervention (day 14), JDB0131 (all 3 dose levels) achieved superior time to positivity (TTP) over delamanid. Ninety-one adverse events (AEs), including no serious AEs, were attributed to JDB0131 in 30 patients. This trial identified a promising new drug for the increasing TB burden worldwide.</p>","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"10 1","pages":"427"},"PeriodicalIF":52.7,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12748780/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145857645","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Dysadherin/YAP axis fuels stem plasticity and immune escape in liver cancer. 粘附异常/YAP轴促进肝癌干细胞可塑性和免疫逃逸。
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-29 DOI: 10.1038/s41392-025-02520-4
Tae-Young Jang,So-El Jeon,Hyeon-Ji Yun,Choong-Jae Lee,Da-Ye Lim,Sang Hoon Lee,Dajun Lee,Seungwon Lee,Jungmin Choi,Hyung-Sik Kim,Jeong-Seok Nam
Hepatocellular carcinoma (HCC) is an aggressive malignancy that is often refractory to chemotherapy and immune checkpoint inhibitors. This therapeutic resistance is driven in part by the persistence of cancer stem-like cells (CSCs) and the development of an immune-cold tumor microenvironment. However, the upstream regulators that coordinate these malignant features remain poorly defined. In this study, we identified dysadherin as a novel upstream activator of YAP that promotes both CSC plasticity and immune evasion through the FAK/YAP/TEAD2 signaling axis. Using single-cell transcriptomic analysis, in vitro assays, and multiple in vivo models including a humanized immune mouse system, we showed that dysadherin enhances the expression of pluripotency genes, such as OCT4 and upregulates PD-L1. These changes support stem-like tumor behavior and contribute to T-cell exclusion, fostering an immunosuppressive niche. Notably, genetic knockdown or peptide-based pharmacologic inhibition of dysadherin effectively restored antitumor immune activation, suppressed metastasis and improved therapeutic responsiveness. Our findings reveal a mechanistic link between dysadherin-mediated cell adhesion signaling and the transcriptional regulation of both stemness and immune escape. Collectively, these findings establish the dysadherin/YAP axis as a key driver of HCC progression and resistance, and highlight it as a compelling therapeutic target that could overcome treatment failure in advanced liver cancer.
肝细胞癌(HCC)是一种侵袭性恶性肿瘤,通常对化疗和免疫检查点抑制剂难以治疗。这种治疗耐药性部分是由癌症干细胞样细胞(CSCs)的持续存在和免疫冷肿瘤微环境的发展所驱动的。然而,协调这些恶性特征的上游调节因子仍然不明确。在这项研究中,我们发现粘附异常蛋白是一种新的YAP上游激活剂,通过FAK/YAP/TEAD2信号轴促进CSC可塑性和免疫逃避。通过单细胞转录组学分析、体外实验以及包括人源化免疫小鼠系统在内的多种体内模型,研究人员发现,粘附障碍增强了OCT4等多能性基因的表达,并上调了PD-L1的表达。这些变化支持干细胞样肿瘤行为,促进t细胞排斥,培养免疫抑制生态位。值得注意的是,基因敲除或基于肽的药物抑制异常粘附蛋白有效地恢复抗肿瘤免疫激活,抑制转移并提高治疗反应性。我们的研究结果揭示了粘附障碍介导的细胞粘附信号与干细胞和免疫逃逸的转录调节之间的机制联系。总的来说,这些发现确定了粘附不良/YAP轴是HCC进展和耐药的关键驱动因素,并强调了它是一个令人信服的治疗靶点,可以克服晚期肝癌的治疗失败。
{"title":"Dysadherin/YAP axis fuels stem plasticity and immune escape in liver cancer.","authors":"Tae-Young Jang,So-El Jeon,Hyeon-Ji Yun,Choong-Jae Lee,Da-Ye Lim,Sang Hoon Lee,Dajun Lee,Seungwon Lee,Jungmin Choi,Hyung-Sik Kim,Jeong-Seok Nam","doi":"10.1038/s41392-025-02520-4","DOIUrl":"https://doi.org/10.1038/s41392-025-02520-4","url":null,"abstract":"Hepatocellular carcinoma (HCC) is an aggressive malignancy that is often refractory to chemotherapy and immune checkpoint inhibitors. This therapeutic resistance is driven in part by the persistence of cancer stem-like cells (CSCs) and the development of an immune-cold tumor microenvironment. However, the upstream regulators that coordinate these malignant features remain poorly defined. In this study, we identified dysadherin as a novel upstream activator of YAP that promotes both CSC plasticity and immune evasion through the FAK/YAP/TEAD2 signaling axis. Using single-cell transcriptomic analysis, in vitro assays, and multiple in vivo models including a humanized immune mouse system, we showed that dysadherin enhances the expression of pluripotency genes, such as OCT4 and upregulates PD-L1. These changes support stem-like tumor behavior and contribute to T-cell exclusion, fostering an immunosuppressive niche. Notably, genetic knockdown or peptide-based pharmacologic inhibition of dysadherin effectively restored antitumor immune activation, suppressed metastasis and improved therapeutic responsiveness. Our findings reveal a mechanistic link between dysadherin-mediated cell adhesion signaling and the transcriptional regulation of both stemness and immune escape. Collectively, these findings establish the dysadherin/YAP axis as a key driver of HCC progression and resistance, and highlight it as a compelling therapeutic target that could overcome treatment failure in advanced liver cancer.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"20 1","pages":"421"},"PeriodicalIF":39.3,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145847255","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Inhibin beta A drives colorectal cancer progression through macrophage M2 polarization and mitochondria-dependent ferroptosis suppression. 抑制素β A通过巨噬细胞M2极化和线粒体依赖性铁下垂抑制驱动结直肠癌的进展。
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-26 DOI: 10.1038/s41392-025-02518-y
Wentao Li,Lin Liang,Siyi Liu,Jingqiong Tang,Shuangyan Ou,Zhijun Yuan,Yanhong Zhou,Xia Yuan
Colorectal cancer (CRC) is a prevalent malignant tumor, and its pathogenesis has not yet been fully elucidated. The tumor microenvironment (TME) and ferroptosis in cancer cells are key drivers of tumor progression and metastasis. This research revealed that elevated INHBA expression in CRC tissues correlates with unfavorable clinical outcomes. In vitro and in vivo studies demonstrated that elevated INHBA enhances CRC cellular growth, migration, and invasion, whereas INHBA knockdown inhibits these malignant biological behaviors. Further investigation revealed that INHBA drives malignancy by reprogramming tumor-associated macrophages (TAMs) toward the M2 phenotype in the TME and by inhibiting mitochondrial-dependent ferroptosis in CRC cells. Mechanistically, INHBA upregulates SLC25A10 to activate the succinate/SUCNR1 axis, thus facilitating M2-like TAM polarization. It also activates the mitochondrial glutathione (mtGSH)/glutathione peroxidase 4 (GPX4) pathway to suppress mitochondria-dependent ferroptosis in CRC cells. Additionally, INHBA acts as a scaffold protein to inhibit TRIM21-mediated ubiquitination and degradation of SLC25A10, thereby stabilizing the SLC25A10 protein. In summary, INHBA drives tumor progression by remodeling the immune microenvironment and antagonizing ferroptosis in CRC cells, providing a theoretical basis for developing INHBA-targeted inhibitors or combined immunoferroptosis therapeutic strategies. Mechanisms of INHBA in colorectal cancer In colorectal cancer, INHBA is upregulated. Acting as a scaffold protein, INHBA inhibits the K48-linked ubiquitination and degradation of the mitochondrial protein SLC25A10, mediated by the E3 ubiquitin ligase TRIM21. This inhibition leads to the upregulation of SLC25A10 expression. The upregulated SLC25A10 facilitates the transport of succinate from the mitochondrial matrix to the cytoplasm and further secretes it outside the tumor cells. The secreted succinate binds to SUCNR1 on macrophages, activating the succinate/SUCNR1 axis, which in turn promotes the M2 polarization of tumor-associated macrophages (TAMs). Meanwhile, SLC25A10, as one of the key mitochondrial glutathione (mtGSH) transporters embedded in the mitochondrial inner membrane, promotes the transport of glutathione (GSH) synthesized in the cytoplasm into the mitochondria. This process activates the mitochondrial GSH-GPX4 axis, thereby inhibiting mitochondrial ferroptosis. Through these two mechanisms, INHBA ultimately promotes the malignant progression of colorectal cancer.
结直肠癌(Colorectal cancer, CRC)是一种常见的恶性肿瘤,其发病机制尚未完全阐明。肿瘤微环境(tumor microenvironment, TME)和铁下垂是肿瘤进展和转移的关键驱动因素。本研究表明,在结直肠癌组织中INHBA表达升高与不良临床结果相关。体外和体内研究表明,升高的INHBA可促进结直肠癌细胞的生长、迁移和侵袭,而INHBA基因敲低可抑制这些恶性生物学行为。进一步的研究表明,INHBA通过将肿瘤相关巨噬细胞(tam)重编程为TME中的M2表型和抑制CRC细胞中线粒体依赖性铁凋亡来驱动恶性肿瘤。在机制上,INHBA上调SLC25A10激活琥珀酸/SUCNR1轴,从而促进m2样TAM极化。它还激活线粒体谷胱甘肽(mtGSH)/谷胱甘肽过氧化物酶4 (GPX4)途径,抑制CRC细胞线粒体依赖性铁凋亡。此外,INHBA作为支架蛋白抑制trim21介导的SLC25A10的泛素化和降解,从而稳定SLC25A10蛋白。综上所述,INHBA通过重塑免疫微环境和拮抗CRC细胞中的铁下沉来驱动肿瘤进展,为开发以INHBA为靶点的抑制剂或联合免疫铁下沉治疗策略提供了理论基础。INHBA在结直肠癌中的作用机制在结直肠癌中,INHBA上调。作为支架蛋白,INHBA抑制由E3泛素连接酶TRIM21介导的k48连接的线粒体蛋白SLC25A10的泛素化和降解。这种抑制导致SLC25A10表达上调。上调的SLC25A10促进琥珀酸盐从线粒体基质转运到细胞质,并进一步将其分泌到肿瘤细胞外。分泌的琥珀酸盐与巨噬细胞上的SUCNR1结合,激活琥珀酸/SUCNR1轴,进而促进肿瘤相关巨噬细胞(tam)的M2极化。同时,SLC25A10作为嵌入线粒体内膜的关键线粒体谷胱甘肽(mtGSH)转运蛋白之一,促进细胞质中合成的谷胱甘肽(GSH)转运至线粒体。这一过程激活线粒体GSH-GPX4轴,从而抑制线粒体铁下垂。通过这两种机制,INHBA最终促进结直肠癌的恶性进展。
{"title":"Inhibin beta A drives colorectal cancer progression through macrophage M2 polarization and mitochondria-dependent ferroptosis suppression.","authors":"Wentao Li,Lin Liang,Siyi Liu,Jingqiong Tang,Shuangyan Ou,Zhijun Yuan,Yanhong Zhou,Xia Yuan","doi":"10.1038/s41392-025-02518-y","DOIUrl":"https://doi.org/10.1038/s41392-025-02518-y","url":null,"abstract":"Colorectal cancer (CRC) is a prevalent malignant tumor, and its pathogenesis has not yet been fully elucidated. The tumor microenvironment (TME) and ferroptosis in cancer cells are key drivers of tumor progression and metastasis. This research revealed that elevated INHBA expression in CRC tissues correlates with unfavorable clinical outcomes. In vitro and in vivo studies demonstrated that elevated INHBA enhances CRC cellular growth, migration, and invasion, whereas INHBA knockdown inhibits these malignant biological behaviors. Further investigation revealed that INHBA drives malignancy by reprogramming tumor-associated macrophages (TAMs) toward the M2 phenotype in the TME and by inhibiting mitochondrial-dependent ferroptosis in CRC cells. Mechanistically, INHBA upregulates SLC25A10 to activate the succinate/SUCNR1 axis, thus facilitating M2-like TAM polarization. It also activates the mitochondrial glutathione (mtGSH)/glutathione peroxidase 4 (GPX4) pathway to suppress mitochondria-dependent ferroptosis in CRC cells. Additionally, INHBA acts as a scaffold protein to inhibit TRIM21-mediated ubiquitination and degradation of SLC25A10, thereby stabilizing the SLC25A10 protein. In summary, INHBA drives tumor progression by remodeling the immune microenvironment and antagonizing ferroptosis in CRC cells, providing a theoretical basis for developing INHBA-targeted inhibitors or combined immunoferroptosis therapeutic strategies. Mechanisms of INHBA in colorectal cancer In colorectal cancer, INHBA is upregulated. Acting as a scaffold protein, INHBA inhibits the K48-linked ubiquitination and degradation of the mitochondrial protein SLC25A10, mediated by the E3 ubiquitin ligase TRIM21. This inhibition leads to the upregulation of SLC25A10 expression. The upregulated SLC25A10 facilitates the transport of succinate from the mitochondrial matrix to the cytoplasm and further secretes it outside the tumor cells. The secreted succinate binds to SUCNR1 on macrophages, activating the succinate/SUCNR1 axis, which in turn promotes the M2 polarization of tumor-associated macrophages (TAMs). Meanwhile, SLC25A10, as one of the key mitochondrial glutathione (mtGSH) transporters embedded in the mitochondrial inner membrane, promotes the transport of glutathione (GSH) synthesized in the cytoplasm into the mitochondria. This process activates the mitochondrial GSH-GPX4 axis, thereby inhibiting mitochondrial ferroptosis. Through these two mechanisms, INHBA ultimately promotes the malignant progression of colorectal cancer.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"107 1","pages":"420"},"PeriodicalIF":39.3,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145830238","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Signal Transduction and Targeted Therapy
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