首页 > 最新文献

Cell Death & Disease最新文献

英文 中文
Correction: YOD1 protects against MRSA sepsis-induced DIC through Lys33-linked deubiquitination of NLRP3. 更正:YOD1 通过与 NLRP3 的 Lys33 链接的去泛素化来防止 MRSA 败血症诱导的 DIC。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-09-04 DOI: 10.1038/s41419-024-06991-1
Chang Liu, Caihong Fan, Jia Liu, Shiqi Zhang, Huixin Tang, Yashan Liu, Shengzheng Zhang, Qiang Wu, Jiandong Zhang, Zhi Qi, Yanna Shen
{"title":"Correction: YOD1 protects against MRSA sepsis-induced DIC through Lys33-linked deubiquitination of NLRP3.","authors":"Chang Liu, Caihong Fan, Jia Liu, Shiqi Zhang, Huixin Tang, Yashan Liu, Shengzheng Zhang, Qiang Wu, Jiandong Zhang, Zhi Qi, Yanna Shen","doi":"10.1038/s41419-024-06991-1","DOIUrl":"10.1038/s41419-024-06991-1","url":null,"abstract":"","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11374977/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131926","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
QRICH1 suppresses pediatric T-cell acute lymphoblastic leukemia by inhibiting GRP78. QRICH1 通过抑制 GRP78 抑制小儿 T 细胞急性淋巴细胞白血病。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-09-04 DOI: 10.1038/s41419-024-07040-7
Ji'ou Zhao, Meiyun Kang, Huimin Li, Liucheng Rong, Yaping Wang, Yao Xue, Yuqian Yao, Yongjun Fang

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that commonly affects children and adolescents with a poor prognosis. The terminal unfolded protein response (UPR) is an emerging anti-cancer approach, although its role in pediatric T-ALL remains unclear. In our pediatric T-ALL cohort from different centers, a lower QRICH1 expression was found associated with a worse prognosis of pediatric T-ALL. Overexpression of QRICH1 significantly inhibited cell proliferation and stimulated apoptosis of T-ALL both in vitro and in vivo. Upregulation of QRICH1 significantly downregulated 78 KDa glucose-regulated protein (GRP78) and upregulated CHOP, thus activating the terminal UPR. Co-overexpression of GRP78 in T-ALL cells overexpressing QRICH1 partially reverted the inhibited proliferation and stimulated apoptosis. QRICH1 bound to the residues Asp212 and Glu155 of the nucleotide-binding domain (NBD) of GRP78, thereby inhibiting its ATP hydrolysis activity. In addition, QRICH1 was associated with endoplasmic reticulum (ER) stress in T-ALL, and overexpression of QRICH1 reversed drug resistance. Overall, low QRICH1 expression is an independent risk factor for a poor prognosis of pediatric T-ALL. By inhibiting GRP78, QRICH1 suppresses pediatric T-ALL.

T 细胞急性淋巴细胞白血病(T-ALL)是一种侵袭性血液恶性肿瘤,常见于预后不良的儿童和青少年。末端未折叠蛋白反应(UPR)是一种新兴的抗癌方法,但它在小儿 T-ALL 中的作用仍不明确。在我们来自不同中心的小儿 T-ALL 队列中,发现 QRICH1 表达较低与小儿 T-ALL 预后较差有关。在体外和体内,QRICH1的过表达都能显著抑制T-ALL的细胞增殖并刺激细胞凋亡。QRICH1的上调能明显下调78 KDa葡萄糖调节蛋白(GRP78)并上调CHOP,从而激活末端UPR。在过表达 QRICH1 的 T-ALL 细胞中联合表达 GRP78 可部分逆转增殖抑制和凋亡刺激。QRICH1 与 GRP78 核苷酸结合域(NBD)的 Asp212 和 Glu155 残基结合,从而抑制了其 ATP 水解活性。此外,QRICH1与T-ALL的内质网(ER)应激有关,过表达QRICH1可逆转耐药性。总体而言,QRICH1的低表达是导致小儿T-ALL预后不良的一个独立危险因素。通过抑制GRP78,QRICH1可抑制小儿T-ALL。
{"title":"QRICH1 suppresses pediatric T-cell acute lymphoblastic leukemia by inhibiting GRP78.","authors":"Ji'ou Zhao, Meiyun Kang, Huimin Li, Liucheng Rong, Yaping Wang, Yao Xue, Yuqian Yao, Yongjun Fang","doi":"10.1038/s41419-024-07040-7","DOIUrl":"10.1038/s41419-024-07040-7","url":null,"abstract":"<p><p>T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that commonly affects children and adolescents with a poor prognosis. The terminal unfolded protein response (UPR) is an emerging anti-cancer approach, although its role in pediatric T-ALL remains unclear. In our pediatric T-ALL cohort from different centers, a lower QRICH1 expression was found associated with a worse prognosis of pediatric T-ALL. Overexpression of QRICH1 significantly inhibited cell proliferation and stimulated apoptosis of T-ALL both in vitro and in vivo. Upregulation of QRICH1 significantly downregulated 78 KDa glucose-regulated protein (GRP78) and upregulated CHOP, thus activating the terminal UPR. Co-overexpression of GRP78 in T-ALL cells overexpressing QRICH1 partially reverted the inhibited proliferation and stimulated apoptosis. QRICH1 bound to the residues Asp212 and Glu155 of the nucleotide-binding domain (NBD) of GRP78, thereby inhibiting its ATP hydrolysis activity. In addition, QRICH1 was associated with endoplasmic reticulum (ER) stress in T-ALL, and overexpression of QRICH1 reversed drug resistance. Overall, low QRICH1 expression is an independent risk factor for a poor prognosis of pediatric T-ALL. By inhibiting GRP78, QRICH1 suppresses pediatric T-ALL.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11371816/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142124954","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Author Correction: Chemotherapy-induced pyroptosis is mediated by BAK/BAX-caspase-3-GSDME pathway and inhibited by 2-bromopalmitate. 作者更正:化疗诱导的热蛋白沉积由 BAK/BAX-caspase-3-GSDME 通路介导,并受 2-溴棕榈酸酯的抑制。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-09-04 DOI: 10.1038/s41419-024-07031-8
Lei Hu, Meng Chen, Xueran Chen, Chenggang Zhao, Zhiyou Fang, Hongzhi Wang, Haiming Dai
{"title":"Author Correction: Chemotherapy-induced pyroptosis is mediated by BAK/BAX-caspase-3-GSDME pathway and inhibited by 2-bromopalmitate.","authors":"Lei Hu, Meng Chen, Xueran Chen, Chenggang Zhao, Zhiyou Fang, Hongzhi Wang, Haiming Dai","doi":"10.1038/s41419-024-07031-8","DOIUrl":"10.1038/s41419-024-07031-8","url":null,"abstract":"","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11375077/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131923","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Author Correction: Ischemia-induced cleavage of OPA1 at S1 site aggravates mitochondrial fragmentation and reperfusion injury in neurons. 作者更正:缺血诱导的 OPA1 在 S1 位点的裂解会加重神经元的线粒体破碎和再灌注损伤。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-09-04 DOI: 10.1038/s41419-024-07032-7
Xiang Li, Haiying Li, Zhongmou Xu, Cheng Ma, Tianyi Wang, Wanchun You, Zhengquan Yu, Haitao Shen, Gang Chen
{"title":"Author Correction: Ischemia-induced cleavage of OPA1 at S1 site aggravates mitochondrial fragmentation and reperfusion injury in neurons.","authors":"Xiang Li, Haiying Li, Zhongmou Xu, Cheng Ma, Tianyi Wang, Wanchun You, Zhengquan Yu, Haitao Shen, Gang Chen","doi":"10.1038/s41419-024-07032-7","DOIUrl":"10.1038/s41419-024-07032-7","url":null,"abstract":"","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11375096/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131924","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
L3MBTL1, a polycomb protein, promotes Osimertinib acquired resistance through epigenetic regulation of DNA damage response in lung adenocarcinoma. L3MBTL1是一种多聚酶蛋白,它通过对肺腺癌DNA损伤反应的表观遗传调控促进奥希替尼获得性耐药性的产生。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-09-04 DOI: 10.1038/s41419-024-06796-2
Zihe Zhang, Yongwen Li, Ruifeng Shi, Chaoyi Jia, Songlin Xu, Guangsheng Zhu, Peijun Cao, Hua Huang, Xuanguang Li, Hongbing Zhang, Minghui Liu, Chen Chen, Hongyu Liu, Chunsheng Kang, Jun Chen

Osimertinib is a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (EGFR-TKI) approved for patients with EGFR T790M resistance mutations as first- or second-line treatment of EGFR-positive patients. Resistance to Osimertinib will inevitably develop, and the underlying mechanisms are largely unknown. In this study, we discovered that acquired resistance to Osimertinib is associated with abnormal DNA damage response (DDR) in lung adenocarcinoma cells. We discovered that the polycomb protein Lethal(3) Malignant Brain Tumor-Like Protein 1 (L3MBTL1) regulates chromatin structure, thereby contributing to DDR and Osimertinib resistance. EGFR oncogene inhibition reduced L3MBTL1 ubiquitination while stabilizing its expression in Osimertinib-resistant cells. L3MBTL1 reduction and treatment with Osimertinib significantly inhibited DDR and proliferation of Osimertinib-resistant lung cancer cells in vitro and in vivo. L3MBTL1 binds throughout the genome and plays an important role in EGFR-TKI resistance. It also competes with 53BP1 for H4K20Me2 and inhibits the development of drug resistance in Osimertinib-resistant lung cancer cells in vitro and in vivo. Our findings suggest that L3MBTL1 inhibition is a novel approach to overcoming EGFR-TKI-acquired resistance.

奥希替尼是第三代表皮生长因子受体(EGFR)酪氨酸激酶抑制剂(EGFR-TKI),已被批准用于EGFR T790M耐药突变患者的一线或二线治疗。奥希替尼的耐药性不可避免地会产生,而其潜在机制在很大程度上是未知的。在这项研究中,我们发现奥希替尼的获得性耐药性与肺腺癌细胞的DNA损伤反应(DDR)异常有关。我们发现多聚核蛋白致命(3)恶性脑肿瘤样蛋白1(L3MBTL1)调控染色质结构,从而导致DDR和奥希替尼耐药。表皮生长因子受体(EGFR)癌基因抑制减少了L3MBTL1的泛素化,同时稳定了其在奥希替尼耐药细胞中的表达。减少 L3MBTL1 并用奥希替尼治疗可显著抑制体外和体内奥希替尼耐药肺癌细胞的 DDR 和增殖。L3MBTL1与整个基因组结合,在表皮生长因子受体-TKI耐药中发挥重要作用。它还与 53BP1 竞争 H4K20Me2,并抑制奥希替尼耐药肺癌细胞在体外和体内的耐药性发展。我们的研究结果表明,L3MBTL1抑制剂是克服表皮生长因子受体-TKI获得性耐药性的一种新方法。
{"title":"L3MBTL1, a polycomb protein, promotes Osimertinib acquired resistance through epigenetic regulation of DNA damage response in lung adenocarcinoma.","authors":"Zihe Zhang, Yongwen Li, Ruifeng Shi, Chaoyi Jia, Songlin Xu, Guangsheng Zhu, Peijun Cao, Hua Huang, Xuanguang Li, Hongbing Zhang, Minghui Liu, Chen Chen, Hongyu Liu, Chunsheng Kang, Jun Chen","doi":"10.1038/s41419-024-06796-2","DOIUrl":"10.1038/s41419-024-06796-2","url":null,"abstract":"<p><p>Osimertinib is a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (EGFR-TKI) approved for patients with EGFR T790M resistance mutations as first- or second-line treatment of EGFR-positive patients. Resistance to Osimertinib will inevitably develop, and the underlying mechanisms are largely unknown. In this study, we discovered that acquired resistance to Osimertinib is associated with abnormal DNA damage response (DDR) in lung adenocarcinoma cells. We discovered that the polycomb protein Lethal(3) Malignant Brain Tumor-Like Protein 1 (L3MBTL1) regulates chromatin structure, thereby contributing to DDR and Osimertinib resistance. EGFR oncogene inhibition reduced L3MBTL1 ubiquitination while stabilizing its expression in Osimertinib-resistant cells. L3MBTL1 reduction and treatment with Osimertinib significantly inhibited DDR and proliferation of Osimertinib-resistant lung cancer cells in vitro and in vivo. L3MBTL1 binds throughout the genome and plays an important role in EGFR-TKI resistance. It also competes with 53BP1 for H4K20Me2 and inhibits the development of drug resistance in Osimertinib-resistant lung cancer cells in vitro and in vivo. Our findings suggest that L3MBTL1 inhibition is a novel approach to overcoming EGFR-TKI-acquired resistance.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11374981/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131927","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
HIF1A-AS2 promotes the metabolic reprogramming and progression of colorectal cancer via miR-141-3p/FOXC1 axis. HIF1A-AS2通过miR-141-3p/FOXC1轴促进代谢重编程和结直肠癌的进展。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-09-03 DOI: 10.1038/s41419-024-06958-2
Xinyang Zhong, Yaxian Wang, Xuefeng He, Xinxin He, Zijuan Hu, Huixia Huang, Jiayu Chen, Keji Chen, Ping Wei, Senlin Zhao, Yilin Wang, Hong Zhang, Bo Feng, Dawei Li

lncRNA can regulate tumorigenesis development and distant metastasis of colorectal cancer (CRC). However, the detailed molecular mechanisms are still largely unknown. Using RNA-sequencing data, RT-qPCR, and FISH assay, we found that HIF1A-AS2 was upregulated in CRC tissues and associated with poor prognosis. Functional experiments were performed to determine the roles of HIF1A-AS2 in tumor progression and we found that HIF1A-AS2 can promote the proliferation, metastasis, and aerobic glycolysis of CRC cells. Mechanistically, HIF1A-AS2 can promote FOXC1 expression by sponging miR-141-3p. SP1 can transcriptionally activate HIF1A-AS2. Further, HIF1A-AS2 can be packaged into exosomes and promote the malignant phenotype of recipient tumor cells. Taken together, we discovered that SP1-induced HIF1A-AS2 can promote the metabolic reprogramming and progression of CRC via miR-141-3p/FOXC1 axis. HIF1A-AS2 is a promising diagnostic marker and treatment target in CRC.

lncRNA 可调控结直肠癌(CRC)的肿瘤发生、发展和远处转移。然而,其详细的分子机制仍不为人知。利用 RNA 序列数据、RT-qPCR 和 FISH 检测,我们发现 HIF1A-AS2 在 CRC 组织中上调,并与不良预后相关。为了确定 HIF1A-AS2 在肿瘤进展中的作用,我们进行了功能实验,结果发现 HIF1A-AS2 可以促进 CRC 细胞的增殖、转移和有氧糖酵解。从机制上讲,HIF1A-AS2可通过海绵状miR-141-3p促进FOXC1的表达。SP1 可以转录激活 HIF1A-AS2。此外,HIF1A-AS2 还能被包装成外泌体,促进受体肿瘤细胞的恶性表型。综上所述,我们发现SP1诱导的HIF1A-AS2可通过miR-141-3p/FOXC1轴促进代谢重编程和CRC的进展。HIF1A-AS2 是一种很有前景的 CRC 诊断标志物和治疗靶点。
{"title":"HIF1A-AS2 promotes the metabolic reprogramming and progression of colorectal cancer via miR-141-3p/FOXC1 axis.","authors":"Xinyang Zhong, Yaxian Wang, Xuefeng He, Xinxin He, Zijuan Hu, Huixia Huang, Jiayu Chen, Keji Chen, Ping Wei, Senlin Zhao, Yilin Wang, Hong Zhang, Bo Feng, Dawei Li","doi":"10.1038/s41419-024-06958-2","DOIUrl":"10.1038/s41419-024-06958-2","url":null,"abstract":"<p><p>lncRNA can regulate tumorigenesis development and distant metastasis of colorectal cancer (CRC). However, the detailed molecular mechanisms are still largely unknown. Using RNA-sequencing data, RT-qPCR, and FISH assay, we found that HIF1A-AS2 was upregulated in CRC tissues and associated with poor prognosis. Functional experiments were performed to determine the roles of HIF1A-AS2 in tumor progression and we found that HIF1A-AS2 can promote the proliferation, metastasis, and aerobic glycolysis of CRC cells. Mechanistically, HIF1A-AS2 can promote FOXC1 expression by sponging miR-141-3p. SP1 can transcriptionally activate HIF1A-AS2. Further, HIF1A-AS2 can be packaged into exosomes and promote the malignant phenotype of recipient tumor cells. Taken together, we discovered that SP1-induced HIF1A-AS2 can promote the metabolic reprogramming and progression of CRC via miR-141-3p/FOXC1 axis. HIF1A-AS2 is a promising diagnostic marker and treatment target in CRC.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11372083/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142124951","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
PILRB potentiates the PI3K/AKT signaling pathway and reprograms cholesterol metabolism to drive gastric tumorigenesis and metastasis. PILRB 强化了 PI3K/AKT 信号通路,并重新规划了胆固醇代谢,从而推动了胃肿瘤的发生和转移。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-09-03 DOI: 10.1038/s41419-024-07026-5
Xing Wang, Yuanyuan Liu, Qiuyan Zhao, Xin Wang, Xinyi Chen, Li Hou, Shaodan Tian, Zi-Mei Peng, Xiao-Jian Han, Tao Wang, Zhen Zhang, Fang-Fang Tou, Shan Huang, Jun Rao, Lixiao Chen, Zhi Zheng

Paired immunoglobin-like type 2 receptor beta (PILRB) mainly plays a crucial role in regulating innate immunity, but whether PILRB is involved in cancer is poorly understood. Here, we report that PILRB potentiates the PI3K/AKT pathway to drive gastric tumorigenesis by binding and stabilizing IRS4, which could hyperactivate the PI3K/AKT pathway. Firstly, the levels of PILRB are upregulated in human gastric cancer (GC) specimens and associated with poor prognosis in patients with GC. In addition, our data show that PILRB promotes cell proliferation, colony formation, cell migration and invasion in GC cells in vitro and in vivo. Mechanistically, PILRB recruits the deubiquitination enzymes OTUB1 to IRS4 and relieves K48-linked ubiquitination of IRS4, protecting IRS4 protein from proteasomal-mediated degradation and subsequent activation of the PI3K/AKT pathway. Importantly, the levels of PILRB are positively correlated with IRS4 in GC specimens. Meanwhile, we also found that PILRB reprogrammed cholesterol metabolism by altering ABCA1 and SCARB1 expression levels, and PILRB-expression confers GC cell resistance to statin treatment. Taken together, our findings illustrate that the oncogenic role of PILRB in gastric tumorigenesis, providing new insights into the regulation of PI3K/AKT signaling in GC and establishing PILRB as a biomarker for simvastatin therapy resistance in GC.

成对免疫球蛋白样2型受体β(PILRB)主要在调节先天性免疫中发挥关键作用,但PILRB是否参与癌症却鲜为人知。在这里,我们报道了PILRB通过结合和稳定IRS4,使PI3K/AKT通路过度激活,从而增强PI3K/AKT通路的作用,驱动胃肿瘤发生。首先,PILRB水平在人类胃癌(GC)标本中上调,并与GC患者的不良预后相关。此外,我们的数据显示,PILRB 可促进 GC 细胞在体外和体内的增殖、集落形成、细胞迁移和侵袭。从机理上讲,PILRB 将去泛素化酶 OTUB1 募集到 IRS4 上,解除了 IRS4 与 K48 链接的泛素化,保护 IRS4 蛋白免受蛋白酶体介导的降解,从而激活 PI3K/AKT 通路。重要的是,在 GC 标本中,PILRB 的水平与 IRS4 呈正相关。同时,我们还发现 PILRB 通过改变 ABCA1 和 SCARB1 的表达水平重编程胆固醇代谢,并且 PILRB 的表达赋予 GC 细胞对他汀类药物治疗的抵抗力。综上所述,我们的研究结果表明了 PILRB 在胃肿瘤发生中的致癌作用,为 PI3K/AKT 信号在 GC 中的调控提供了新的见解,并将 PILRB 确立为辛伐他汀治疗 GC 耐药性的生物标志物。
{"title":"PILRB potentiates the PI3K/AKT signaling pathway and reprograms cholesterol metabolism to drive gastric tumorigenesis and metastasis.","authors":"Xing Wang, Yuanyuan Liu, Qiuyan Zhao, Xin Wang, Xinyi Chen, Li Hou, Shaodan Tian, Zi-Mei Peng, Xiao-Jian Han, Tao Wang, Zhen Zhang, Fang-Fang Tou, Shan Huang, Jun Rao, Lixiao Chen, Zhi Zheng","doi":"10.1038/s41419-024-07026-5","DOIUrl":"10.1038/s41419-024-07026-5","url":null,"abstract":"<p><p>Paired immunoglobin-like type 2 receptor beta (PILRB) mainly plays a crucial role in regulating innate immunity, but whether PILRB is involved in cancer is poorly understood. Here, we report that PILRB potentiates the PI3K/AKT pathway to drive gastric tumorigenesis by binding and stabilizing IRS4, which could hyperactivate the PI3K/AKT pathway. Firstly, the levels of PILRB are upregulated in human gastric cancer (GC) specimens and associated with poor prognosis in patients with GC. In addition, our data show that PILRB promotes cell proliferation, colony formation, cell migration and invasion in GC cells in vitro and in vivo. Mechanistically, PILRB recruits the deubiquitination enzymes OTUB1 to IRS4 and relieves K48-linked ubiquitination of IRS4, protecting IRS4 protein from proteasomal-mediated degradation and subsequent activation of the PI3K/AKT pathway. Importantly, the levels of PILRB are positively correlated with IRS4 in GC specimens. Meanwhile, we also found that PILRB reprogrammed cholesterol metabolism by altering ABCA1 and SCARB1 expression levels, and PILRB-expression confers GC cell resistance to statin treatment. Taken together, our findings illustrate that the oncogenic role of PILRB in gastric tumorigenesis, providing new insights into the regulation of PI3K/AKT signaling in GC and establishing PILRB as a biomarker for simvastatin therapy resistance in GC.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11372125/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142124952","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
PIM1 kinase promotes EMT-associated osimertinib resistance via regulating GSK3β signaling pathway in EGFR-mutant non-small cell lung cancer. PIM1 激酶通过调节 GSK3β 信号通路促进 EGFR 突变非小细胞肺癌中与 EMT 相关的奥希替尼耐药性。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-09-03 DOI: 10.1038/s41419-024-07039-0
Jing Zhou, Xinyue Wang, Zhaona Li, Fan Wang, Lianjing Cao, Xiuqiong Chen, Dingzhi Huang, Richeng Jiang

Acquired resistance is inevitable in the treatment of non-small cell lung cancer (NSCLC) with osimertinib, and one of the primary mechanisms responsible for this resistance is the epithelial-mesenchymal transition (EMT). We identify upregulation of the proviral integration site for Moloney murine leukemia virus 1 (PIM1) and functional inactivation of glycogen synthase kinase 3β (GSK3β) as drivers of EMT-associated osimertinib resistance. Upregulation of PIM1 promotes the growth, invasion, and resistance of osimertinib-resistant cells and is significantly correlated with EMT molecules expression. Functionally, PIM1 suppresses the ubiquitin-proteasome degradation of snail family transcriptional repressor 1 (SNAIL) and snail family transcriptional repressor 2 (SLUG) by deactivating GSK3β through phosphorylation. The stability and accumulation of SNAIL and SLUG facilitate EMT and encourage osimertinib resistance. Furthermore, treatment with PIM1 inhibitors prevents EMT progression and re-sensitizes osimertinib-resistant NSCLC cells to osimertinib. PIM1/GSK3β signaling is activated in clinical samples of osimertinib-resistant NSCLC, and dual epidermal growth factor receptor (EGFR)/PIM1 blockade synergistically reverse osimertinib-resistant NSCLC in vivo. These data identify PIM1 as a driver of EMT-associated osimertinib-resistant NSCLC cells and predict that PIM1 inhibitors and osimertinib combination therapy will provide clinical benefit in patients with EGFR-mutant NSCLC.

奥希替尼治疗非小细胞肺癌(NSCLC)不可避免地会产生获得性耐药性,而导致这种耐药性的主要机制之一是上皮-间质转化(EMT)。我们发现莫罗尼小鼠白血病病毒1(PIM1)前病毒整合位点的上调和糖原合酶激酶3β(GSK3β)的功能性失活是EMT相关奥希替尼耐药的驱动因素。PIM1的上调促进了奥希替尼耐药细胞的生长、侵袭和耐药性,并与EMT分子的表达显著相关。在功能上,PIM1通过磷酸化使GSK3β失活,从而抑制蜗牛家族转录抑制因子1(SNAIL)和蜗牛家族转录抑制因子2(SLUG)的泛素蛋白酶体降解。SNAIL和SLUG的稳定和积累促进了EMT,并促使奥希替尼产生耐药性。此外,用PIM1抑制剂治疗可防止EMT进展,并使奥希替尼耐药的NSCLC细胞对奥希替尼重新敏感。在奥希替尼耐药的 NSCLC 临床样本中,PIM1/GSK3β 信号被激活,表皮生长因子受体(EGFR)/PIM1 双阻断在体内可协同逆转奥希替尼耐药的 NSCLC。这些数据确定了 PIM1 是与 EMT 相关的奥西美替尼耐药 NSCLC 细胞的驱动因素,并预测 PIM1 抑制剂和奥西美替尼联合疗法将为表皮生长因子受体突变 NSCLC 患者带来临床获益。
{"title":"PIM1 kinase promotes EMT-associated osimertinib resistance via regulating GSK3β signaling pathway in EGFR-mutant non-small cell lung cancer.","authors":"Jing Zhou, Xinyue Wang, Zhaona Li, Fan Wang, Lianjing Cao, Xiuqiong Chen, Dingzhi Huang, Richeng Jiang","doi":"10.1038/s41419-024-07039-0","DOIUrl":"10.1038/s41419-024-07039-0","url":null,"abstract":"<p><p>Acquired resistance is inevitable in the treatment of non-small cell lung cancer (NSCLC) with osimertinib, and one of the primary mechanisms responsible for this resistance is the epithelial-mesenchymal transition (EMT). We identify upregulation of the proviral integration site for Moloney murine leukemia virus 1 (PIM1) and functional inactivation of glycogen synthase kinase 3β (GSK3β) as drivers of EMT-associated osimertinib resistance. Upregulation of PIM1 promotes the growth, invasion, and resistance of osimertinib-resistant cells and is significantly correlated with EMT molecules expression. Functionally, PIM1 suppresses the ubiquitin-proteasome degradation of snail family transcriptional repressor 1 (SNAIL) and snail family transcriptional repressor 2 (SLUG) by deactivating GSK3β through phosphorylation. The stability and accumulation of SNAIL and SLUG facilitate EMT and encourage osimertinib resistance. Furthermore, treatment with PIM1 inhibitors prevents EMT progression and re-sensitizes osimertinib-resistant NSCLC cells to osimertinib. PIM1/GSK3β signaling is activated in clinical samples of osimertinib-resistant NSCLC, and dual epidermal growth factor receptor (EGFR)/PIM1 blockade synergistically reverse osimertinib-resistant NSCLC in vivo. These data identify PIM1 as a driver of EMT-associated osimertinib-resistant NSCLC cells and predict that PIM1 inhibitors and osimertinib combination therapy will provide clinical benefit in patients with EGFR-mutant NSCLC.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11372188/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142124953","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Targeting POLRMT by IMT1 inhibits colorectal cancer cell growth. 通过 IMT1 靶向 POLRMT 可抑制结直肠癌细胞生长。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-09-03 DOI: 10.1038/s41419-024-07023-8
Hao Wang, Yuxin Liu, Xing-Sheng Lu, Yongyou Wu, Wen Gu, Guojian Yin

This study investigates the potential anti-colorectal cancer (CRC) activity of IMT1, a novel specific inhibitor of mitochondrial RNA polymerase (POLRMT). Single-cell RNA sequencing data reveal that POLRMT is overexpressed in CRC cells. Additionally, elevated POLRMT expression was observed in local CRC tissues and cells, while its expression remained relatively low in colon epithelial tissues and cells. IMT1 significantly inhibited colony formation, cell viability, proliferation, cell cycle progression, and migration in both primary and immortalized CRC cells. Furthermore, IMT1 induced apoptosis and cell death in CRC cells. The inhibition of POLRMT by IMT1 disrupted mitochondrial functions in CRC cells, leading to mitochondrial depolarization, oxidative damage, and decreased ATP levels. Using targeted shRNA to silence POLRMT closely mirrored the effects of IMT1, showing robust anti-CRC cell activity. Crucially, the efficacy of IMT1 was diminished in CRC cells with silenced POLRMT. Contrarily, boosting POLRMT expression externally by a lentiviral construct promoted the proliferation and migration of CRC cells. Importantly, treatment with IMT1 or silencing POLRMT in primary colon cancer cells decreased the phosphorylation of Akt1-S6K1, whereas overexpression of POLRMT had the opposite effect. In nude mice, orally administering IMT1 potently restrained primary colon cancer xenograft growth. IMT1 suppressed POLRMT activity, disrupted mitochondrial function, hindered Akt-mTOR activation, and prompted apoptosis within the xenograft tissues. In addition, IMT1 administration suppressed lung metastasis of primary colon cancer cells in nude mice. These combined results highlight the robust anti-CRC activity of IMT1 by specifically targeting POLRMT.

本研究探讨了线粒体 RNA 聚合酶(POLRMT)的新型特异性抑制剂 IMT1 的潜在抗结直肠癌(CRC)活性。单细胞 RNA 测序数据显示,POLRMT 在 CRC 细胞中过度表达。此外,在局部 CRC 组织和细胞中观察到 POLRMT 表达升高,而在结肠上皮组织和细胞中其表达仍相对较低。IMT1 能明显抑制原代和永生化 CRC 细胞的集落形成、细胞活力、增殖、细胞周期进展和迁移。此外,IMT1 还能诱导 CRC 细胞凋亡和细胞死亡。IMT1 对 POLRMT 的抑制破坏了 CRC 细胞的线粒体功能,导致线粒体去极化、氧化损伤和 ATP 水平下降。使用靶向 shRNA 沉默 POLRMT 与 IMT1 的效果密切相关,显示出强大的抗 CRC 细胞活性。重要的是,在 POLRMT 被沉默的 CRC 细胞中,IMT1 的功效减弱了。相反,通过慢病毒构建体从外部增强 POLRMT 的表达能促进 CRC 细胞的增殖和迁移。重要的是,在原发性结肠癌细胞中使用 IMT1 或沉默 POLRMT 会降低 Akt1-S6K1 的磷酸化,而过表达 POLRMT 则会产生相反的效果。在裸鼠体内,口服 IMT1 能有效抑制原发性结肠癌异种移植的生长。IMT1 抑制了 POLRMT 的活性,破坏了线粒体功能,阻碍了 Akt-mTOR 的激活,并促使异种组织内的细胞凋亡。此外,IMT1 还能抑制原发性结肠癌细胞在裸鼠体内的肺转移。这些综合结果凸显了 IMT1 通过特异性靶向 POLRMT 而发挥的强大抗结肠癌活性。
{"title":"Targeting POLRMT by IMT1 inhibits colorectal cancer cell growth.","authors":"Hao Wang, Yuxin Liu, Xing-Sheng Lu, Yongyou Wu, Wen Gu, Guojian Yin","doi":"10.1038/s41419-024-07023-8","DOIUrl":"10.1038/s41419-024-07023-8","url":null,"abstract":"<p><p>This study investigates the potential anti-colorectal cancer (CRC) activity of IMT1, a novel specific inhibitor of mitochondrial RNA polymerase (POLRMT). Single-cell RNA sequencing data reveal that POLRMT is overexpressed in CRC cells. Additionally, elevated POLRMT expression was observed in local CRC tissues and cells, while its expression remained relatively low in colon epithelial tissues and cells. IMT1 significantly inhibited colony formation, cell viability, proliferation, cell cycle progression, and migration in both primary and immortalized CRC cells. Furthermore, IMT1 induced apoptosis and cell death in CRC cells. The inhibition of POLRMT by IMT1 disrupted mitochondrial functions in CRC cells, leading to mitochondrial depolarization, oxidative damage, and decreased ATP levels. Using targeted shRNA to silence POLRMT closely mirrored the effects of IMT1, showing robust anti-CRC cell activity. Crucially, the efficacy of IMT1 was diminished in CRC cells with silenced POLRMT. Contrarily, boosting POLRMT expression externally by a lentiviral construct promoted the proliferation and migration of CRC cells. Importantly, treatment with IMT1 or silencing POLRMT in primary colon cancer cells decreased the phosphorylation of Akt1-S6K1, whereas overexpression of POLRMT had the opposite effect. In nude mice, orally administering IMT1 potently restrained primary colon cancer xenograft growth. IMT1 suppressed POLRMT activity, disrupted mitochondrial function, hindered Akt-mTOR activation, and prompted apoptosis within the xenograft tissues. In addition, IMT1 administration suppressed lung metastasis of primary colon cancer cells in nude mice. These combined results highlight the robust anti-CRC activity of IMT1 by specifically targeting POLRMT.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11372113/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142124955","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Mesenchymal stromal cells restrain the Th17 cell response via L-amino-acid oxidase within lymph nodes 间充质基质细胞通过淋巴结内的 L-氨基酸氧化酶抑制 Th17 细胞反应
IF 9 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-09-02 DOI: 10.1038/s41419-024-07024-7
Qi Ni, Le Zhen, Zhu Zeng, Jingwen Yang, Yukai Wang, Huanke Xu, Qixiang Zhang, Yongcheng Zhu, Yu Tao, Jing Wang, Qing Liu, Kezheng Yi, Yang Chen, Qian Chen, Guangji Wang, Fang Zhou, Yunlong Shan

Mesenchymal stromal/stem cells (MSC) have emerged as a promising therapeutic avenue for treating autoimmune diseases, eliciting considerable interest and discussion regarding their underlying mechanisms. This study revealed the distinctive ability of human umbilical cord MSC to aggregate within the lymph nodes of mice afflicted with autoimmune diseases, but this phenomenon was not observed in healthy mice. The specific distribution is driven by the heightened expression of the CCL21-CCR7 axis in mice with autoimmune diseases, facilitating the targeted homing of MSC to the lymph nodes. Within the lymph nodes, MSC exhibit a remarkable capacity to modulate Th17 cell function, exerting a pronounced anti-inflammatory effect. Transplanted MSC stimulates the secretion of L-amino-acid oxidase (LAAO), a response triggered by elevated levels of tumor necrosis factor-α (TNF-α) in mice with autoimmune diseases through the NF-κB pathway. The presence of LAAO is indispensable for the efficacy of MSC, as it significantly contributes to the inhibition of Th17 cells. Furthermore, LAAO-derived indole-3-pyruvic acid (I3P) serves as a potent suppressor of Th17 cells by activating the aryl hydrocarbon receptor (AHR) pathway. These findings advance our understanding of the global immunomodulatory effects exerted by MSC, providing valuable information for optimizing therapeutic outcomes.

间充质基质/干细胞(MSC)已成为治疗自身免疫性疾病的一种很有前景的治疗方法,引起了人们对其潜在机制的极大兴趣和讨论。这项研究揭示了人脐带间充质干细胞在患有自身免疫性疾病的小鼠淋巴结内聚集的独特能力,但在健康小鼠身上却没有观察到这种现象。自身免疫性疾病小鼠体内CCL21-CCR7轴的高表达促进了间充质干细胞向淋巴结的定向归巢,从而推动了间充质干细胞的特异性分布。在淋巴结内,间充质干细胞具有调节 Th17 细胞功能的显著能力,可发挥明显的抗炎作用。移植的间充质干细胞能刺激L-氨基酸氧化酶(LAAO)的分泌,这种反应是患有自身免疫性疾病的小鼠体内肿瘤坏死因子-α(TNF-α)水平升高时通过NF-κB途径引发的。LAAO 的存在对间叶干细胞的疗效不可或缺,因为它能显著抑制 Th17 细胞。此外,LAAO衍生的吲哚-3-丙酮酸(I3P)通过激活芳基烃受体(AHR)通路,可有效抑制Th17细胞。这些发现加深了我们对间叶干细胞的整体免疫调节作用的理解,为优化治疗效果提供了宝贵的信息。
{"title":"Mesenchymal stromal cells restrain the Th17 cell response via L-amino-acid oxidase within lymph nodes","authors":"Qi Ni, Le Zhen, Zhu Zeng, Jingwen Yang, Yukai Wang, Huanke Xu, Qixiang Zhang, Yongcheng Zhu, Yu Tao, Jing Wang, Qing Liu, Kezheng Yi, Yang Chen, Qian Chen, Guangji Wang, Fang Zhou, Yunlong Shan","doi":"10.1038/s41419-024-07024-7","DOIUrl":"https://doi.org/10.1038/s41419-024-07024-7","url":null,"abstract":"<p>Mesenchymal stromal/stem cells (MSC) have emerged as a promising therapeutic avenue for treating autoimmune diseases, eliciting considerable interest and discussion regarding their underlying mechanisms. This study revealed the distinctive ability of human umbilical cord MSC to aggregate within the lymph nodes of mice afflicted with autoimmune diseases, but this phenomenon was not observed in healthy mice. The specific distribution is driven by the heightened expression of the CCL21-CCR7 axis in mice with autoimmune diseases, facilitating the targeted homing of MSC to the lymph nodes. Within the lymph nodes, MSC exhibit a remarkable capacity to modulate Th17 cell function, exerting a pronounced anti-inflammatory effect. Transplanted MSC stimulates the secretion of L-amino-acid oxidase (LAAO), a response triggered by elevated levels of tumor necrosis factor-α (TNF-α) in mice with autoimmune diseases through the NF-κB pathway. The presence of LAAO is indispensable for the efficacy of MSC, as it significantly contributes to the inhibition of Th17 cells. Furthermore, LAAO-derived indole-3-pyruvic acid (I3P) serves as a potent suppressor of Th17 cells by activating the aryl hydrocarbon receptor (AHR) pathway. These findings advance our understanding of the global immunomodulatory effects exerted by MSC, providing valuable information for optimizing therapeutic outcomes.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":9.0,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142218500","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cell Death & Disease
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:481959085
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