首页 > 最新文献

Cancer letters最新文献

英文 中文
MATR3 promotes liver cancer progression by suppressing DHX58–mediated type I interferon response MATR3 通过抑制 DHX58 介导的 I 型干扰素反应促进肝癌进展
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-12 DOI: 10.1016/j.canlet.2024.217231

MATR3 is a nuclear matrix protein implicated in various cancers; however, its specific role in tumor progression remains unclear. The study utilized the TCGA database to reveal that MATR3 expression is upregulated in liver cancer and is correlated with poor prognosis. Functionally, MATR3 promoted liver cancer cell proliferation and metastasis. Comprehensive RNA sequencing analysis showed that MATR3 significantly affected the type I IFN signaling pathway and DHX58 is a downstream target of MATR3. Further experiments showed that MATR3 bound to DHX58 mRNA through its RRM structural domain and recruited YTHDF2, an m6A reader, leading to degradation of DHX58 mRNA and suppression of the type I IFN signaling pathway. The knockout of MATR3 in liver cancer cells triggered a natural immune response that stimulated CD8+ T cells to eliminate liver cancer cells. This study demonstrated that MATR3 downregulates type I IFN signaling in liver cancer cells through m6A modification and inhibits immune cell infiltration within tumors. These findings expand our understanding of the role of MATR3 in liver cancer.

MATR3是一种与多种癌症有关联的核基质蛋白,但它在肿瘤进展中的具体作用仍不清楚。研究利用TCGA数据库发现,MATR3在肝癌中表达上调,并与预后不良相关。在功能上,MATR3能促进肝癌细胞的增殖和转移。全面的RNA测序分析表明,MATR3显著影响I型IFN信号通路,而DHX58是MATR3的下游靶点。进一步的实验表明,MATR3通过其RRM结构域与DHX58 mRNA结合,并招募m6A阅读器YTHDF2,导致DHX58 mRNA降解,抑制I型IFN信号通路。在肝癌细胞中敲除 MATR3 会引发自然免疫反应,刺激 CD8+ T 细胞消灭肝癌细胞。这项研究证明,MATR3 通过 m6A 修饰下调肝癌细胞的 I 型 IFN 信号,并抑制肿瘤内免疫细胞的浸润。这些发现拓展了我们对 MATR3 在肝癌中作用的认识。
{"title":"MATR3 promotes liver cancer progression by suppressing DHX58–mediated type I interferon response","authors":"","doi":"10.1016/j.canlet.2024.217231","DOIUrl":"10.1016/j.canlet.2024.217231","url":null,"abstract":"<div><p>MATR3 is a nuclear matrix protein implicated in various cancers; however, its specific role in tumor progression remains unclear. The study utilized the TCGA database to reveal that MATR3 expression is upregulated in liver cancer and is correlated with poor prognosis. Functionally, MATR3 promoted liver cancer cell proliferation and metastasis. Comprehensive RNA sequencing analysis showed that MATR3 significantly affected the type I IFN signaling pathway and DHX58 is a downstream target of MATR3. Further experiments showed that MATR3 bound to DHX58 mRNA through its RRM structural domain and recruited YTHDF2, an m<sup>6</sup>A reader, leading to degradation of DHX58 mRNA and suppression of the type I IFN signaling pathway. The knockout of MATR3 in liver cancer cells triggered a natural immune response that stimulated CD8<sup>+</sup> T cells to eliminate liver cancer cells. This study demonstrated that MATR3 downregulates type I IFN signaling in liver cancer cells through m<sup>6</sup>A modification and inhibits immune cell infiltration within tumors. These findings expand our understanding of the role of MATR3 in liver cancer.</p></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0304383524006268/pdfft?md5=bf6f0fa94e0c581094108534fb5a7b3f&pid=1-s2.0-S0304383524006268-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142239789","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
Macroautophagy/autophagy promotes resistance to KRASG12D-targeted therapy through glutathione synthesis 大自噬/自噬通过谷胱甘肽合成促进对KRASG12D靶向疗法的耐受性
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-12 DOI: 10.1016/j.canlet.2024.217258

KRASG12D mutation-driven pancreatic ductal adenocarcinoma (PDAC) represents a major challenge in medicine due to late diagnosis and treatment resistance. Here, we report that macroautophagy (hereafter autophagy), a cellular degradation and recycling process, contributes to acquired resistance against novel KRASG12D-targeted therapy. The KRASG12D protein inhibitor MRTX1133 induces autophagy in KRASG12D-mutated PDAC cells by blocking MTOR activity, and increased autophagic flux prevents apoptosis. Mechanistically, autophagy facilitates the generation of glutamic acid, cysteine, and glycine for glutathione synthesis. Increased glutathione levels reduce reactive oxygen species production, which impedes CYCS translocation from mitochondria to the cytosol, ultimately preventing the formation of the APAF1 apoptosome. Consequently, genetic interventions (utilizing ATG5 or BECN1 knockout) or pharmacological inhibition of autophagy (with chloroquine, bafilomycin A1, or spautin-1) enhance the anticancer activity of MRTX1133 in vitro and in various animal models (subcutaneous, patient-derived xenograft, and orthotopic). Moreover, the release of histones by apoptotic cells triggers an adaptive immune response when combining an autophagy inhibitor with MRTX1133 in immunocompetent mice. These findings establish a new strategy to overcome KRASG12D-targeted therapy resistance by inhibiting autophagy-dependent glutathione synthesis.

KRASG12D突变驱动的胰腺导管腺癌(PDAC)因诊断晚和耐药而成为医学界的一大挑战。在这里,我们报告了大自噬(以下简称自噬)这一细胞降解和回收过程导致了对新型 KRASG12D 靶向疗法的获得性耐药性。KRASG12D蛋白抑制剂MRTX1133通过阻断MTOR活性诱导KRASG12D突变的PDAC细胞自噬,自噬通量的增加可防止细胞凋亡。从机制上讲,自噬可促进谷氨酸、半胱氨酸和甘氨酸的生成,用于谷胱甘肽的合成。谷胱甘肽水平的提高会减少活性氧的产生,从而阻碍 CYCS 从线粒体转运到细胞质,最终阻止 APAF1 细胞凋亡小体的形成。因此,基因干预(利用 ATG5 或 BECN1 基因敲除)或药物抑制自噬(使用氯喹、巴磷霉素 A1 或 spautin-1)可增强 MRTX1133 在体外和各种动物模型(皮下、患者来源异种移植和正位)中的抗癌活性。此外,将自噬抑制剂与 MRTX1133 在免疫功能正常的小鼠中结合使用时,凋亡细胞释放的组蛋白会引发适应性免疫反应。这些发现确立了一种新策略,通过抑制依赖于自噬的谷胱甘肽合成来克服 KRASG12D 靶向治疗的耐药性。
{"title":"Macroautophagy/autophagy promotes resistance to KRASG12D-targeted therapy through glutathione synthesis","authors":"","doi":"10.1016/j.canlet.2024.217258","DOIUrl":"10.1016/j.canlet.2024.217258","url":null,"abstract":"<div><p><em>KRAS</em><sup><em>G12D</em></sup> mutation-driven pancreatic ductal adenocarcinoma (PDAC) represents a major challenge in medicine due to late diagnosis and treatment resistance. Here, we report that macroautophagy (hereafter autophagy), a cellular degradation and recycling process, contributes to acquired resistance against novel KRAS<sup>G12D</sup>-targeted therapy. The KRAS<sup>G12D</sup> protein inhibitor MRTX1133 induces autophagy in <em>KRAS</em><sup><em>G12D</em></sup>-mutated PDAC cells by blocking MTOR activity, and increased autophagic flux prevents apoptosis. Mechanistically, autophagy facilitates the generation of glutamic acid, cysteine, and glycine for glutathione synthesis. Increased glutathione levels reduce reactive oxygen species production, which impedes CYCS translocation from mitochondria to the cytosol, ultimately preventing the formation of the APAF1 apoptosome. Consequently, genetic interventions (utilizing <em>ATG5</em> or <em>BECN1</em> knockout) or pharmacological inhibition of autophagy (with chloroquine, bafilomycin A<sub>1</sub>, or spautin-1) enhance the anticancer activity of MRTX1133 <em>in vitro</em> and in various animal models (subcutaneous, patient-derived xenograft, and orthotopic). Moreover, the release of histones by apoptotic cells triggers an adaptive immune response when combining an autophagy inhibitor with MRTX1133 in immunocompetent mice. These findings establish a new strategy to overcome KRAS<sup>G12D</sup>-targeted therapy resistance by inhibiting autophagy-dependent glutathione synthesis.</p></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0304383524006530/pdfft?md5=04097899eb61102cd2f3f2ea0ed89db5&pid=1-s2.0-S0304383524006530-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142239786","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 tumor-promotional molecular axis CircMAPKBP1/miR-17-3p/TGFβ2 activates autophagy pathway to drive tongue squamous cell carcinoma cisplatin chemoresistance 促进肿瘤生长的分子轴CircMAPKBP1/miR-17-3p/TGFβ2激活自噬通路,驱动舌鳞状细胞癌的顺铂化疗抗性
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-12 DOI: 10.1016/j.canlet.2024.217230
Platinum-based chemotherapy is the first-line treatment for tongue squamous cell carcinoma (TSCC), but most patients rapidly develop resistance. Circular RNAs (circRNAs) are a class of critical regulators in the pathogenesis of several tumors, but their role in cisplatin resistance in TSCC has not been fully elucidated. Here we found that circMAPKBP1 was enriched in cisplatin resistant TSCC cells and was closely associated with enhanced autophagic activity. Functionally, silencing circMAPKBP1 significantly restored the chemosensitivity of cisplatin-resistant TSCC cells both in vitro and in vivo by suppressing autophagy. Mechanistically, circMAPKBP1 enhanced cisplatin sensitivity through the miR-17-3p/TGFβ2 axis by activating autophagy pathway. Data from clinical studies revealed that high expression of circMAPKBP1 and TGFβ2 was closely linked to a poor outcome in TSCC patients. We thus concluded that circMAPKBP1 is a tumor promoting factor and confers cisplatin sensitivity by activating the miR-17-3p/TGFβ2 axis-mediated autophagy. We propose that circMAPKBP1 may be a potential therapeutic target for TSCC.
铂类化疗是舌鳞状细胞癌(TSCC)的一线治疗方法,但大多数患者会迅速产生耐药性。环状 RNA(circRNA)是多种肿瘤发病机制中的一类关键调控因子,但它们在 TSCC 的顺铂耐药性中的作用尚未完全阐明。在这里,我们发现circMAPKBP1在顺铂耐药的TSCC细胞中富集,并与自噬活性增强密切相关。从功能上讲,沉默circMAPKBP1可通过抑制自噬作用显著恢复顺铂耐药TSCC细胞在体外和体内的化疗敏感性。从机理上讲,circMAPKBP1通过激活自噬通路,通过miR-17-3p/TGFβ2轴增强顺铂敏感性。临床研究数据显示,circMAPKBP1和TGFβ2的高表达与TSCC患者的不良预后密切相关。因此我们认为,circMAPKBP1是一种肿瘤促进因子,通过激活miR-17-3p/TGFβ2轴介导的自噬而赋予顺铂敏感性。我们建议将 circMAPKBP1 作为 TSCC 的潜在治疗靶点。
{"title":"A tumor-promotional molecular axis CircMAPKBP1/miR-17-3p/TGFβ2 activates autophagy pathway to drive tongue squamous cell carcinoma cisplatin chemoresistance","authors":"","doi":"10.1016/j.canlet.2024.217230","DOIUrl":"10.1016/j.canlet.2024.217230","url":null,"abstract":"<div><div>Platinum-based chemotherapy is the first-line treatment for tongue squamous cell carcinoma (TSCC), but most patients rapidly develop resistance. Circular RNAs (circRNAs) are a class of critical regulators in the pathogenesis of several tumors, but their role in cisplatin resistance in TSCC has not been fully elucidated. Here we found that circMAPKBP1 was enriched in cisplatin resistant TSCC cells and was closely associated with enhanced autophagic activity. Functionally, silencing circMAPKBP1 significantly restored the chemosensitivity of cisplatin-resistant TSCC cells both in vitro and in vivo by suppressing autophagy. Mechanistically, circMAPKBP1 enhanced cisplatin sensitivity through the miR-17-3p/TGFβ2 axis by activating autophagy pathway. Data from clinical studies revealed that high expression of circMAPKBP1 and TGFβ2 was closely linked to a poor outcome in TSCC patients. We thus concluded that circMAPKBP1 is a tumor promoting factor and confers cisplatin sensitivity by activating the miR-17-3p/TGFβ2 axis-mediated autophagy. We propose that circMAPKBP1 may be a potential therapeutic target for TSCC.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142280531","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
The deubiquitinase OTUD5 stabilizes SLC7A11 to promote progression and reduce paclitaxel sensitivity in triple-negative breast cancer 去泛素化酶OTUD5能稳定SLC7A11,从而促进三阴性乳腺癌的进展并降低紫杉醇的敏感性。
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-12 DOI: 10.1016/j.canlet.2024.217232
Ferroptosis is a newly defined form of programmed cell death characterized by iron-dependent lipid peroxide accumulation and is associated with the progression of cancer. Solute carrier family 7 member 11 (SLC7A11), a key component of cystine/glutamate antiporter, has been characterized as a critical regulator of ferroptosis. Although many studies have established the transcriptional regulation of SLC7A11, it remains largely unknown how the stability of SLC7A11 is regulated in cancers, especially in triple-negative breast cancer (TNBC). Here we demonstrated that ovarian tumor domain-containing protein 5 (OTUD5), which deubiquitinated and stabilized SLC7A11, played a key role in TNBC progression and paclitaxel chemosensitivity through modulating ferroptosis. The clinical data analysis showed OTUD5 was higher expressed in TNBC, which positively correlated with SLC7A11 level. Mechanistically, OTUD5 interacted with SLC7A11 and cleaved K48-linked polyubiquitin chains from SLC7A11 to enhance the stability of SLC7A11. Taken together, these findings uncover a functional and mechanistic role of OTUD5 in TNBC progression and paclitaxel sensitivity, indicating OTUD5 could be a potential target for TNBC treatment.
铁变性是一种新定义的程序性细胞死亡形式,其特点是铁依赖性过氧化脂质积累,与癌症的进展有关。胱氨酸/谷氨酸反转运体的关键成分溶质运载体家族 7 成员 11(SLC7A11)已被定性为铁突变的关键调控因子。尽管许多研究已经确定了 SLC7A11 的转录调控,但人们仍然不知道 SLC7A11 的稳定性在癌症中是如何调控的,尤其是在三阴性乳腺癌(TNBC)中。在这里,我们证明了卵巢肿瘤含域蛋白5(OTUD5)能去泛素化并稳定SLC7A11,通过调节铁突变在TNBC进展和紫杉醇化疗敏感性中发挥关键作用。临床数据分析显示,OTUD5在TNBC中表达较高,与SLC7A11水平呈正相关。从机理上讲,OTUD5与SLC7A11相互作用,并从SLC7A11上裂解K48连接的多泛素链,从而增强SLC7A11的稳定性。综上所述,这些发现揭示了OTUD5在TNBC进展和紫杉醇敏感性中的功能和机制作用,表明OTUD5可能是TNBC治疗的潜在靶点。
{"title":"The deubiquitinase OTUD5 stabilizes SLC7A11 to promote progression and reduce paclitaxel sensitivity in triple-negative breast cancer","authors":"","doi":"10.1016/j.canlet.2024.217232","DOIUrl":"10.1016/j.canlet.2024.217232","url":null,"abstract":"<div><div>Ferroptosis is a newly defined form of programmed cell death characterized by iron-dependent lipid peroxide accumulation and is associated with the progression of cancer. Solute carrier family 7 member 11 (SLC7A11), a key component of cystine/glutamate antiporter, has been characterized as a critical regulator of ferroptosis. Although many studies have established the transcriptional regulation of SLC7A11, it remains largely unknown how the stability of SLC7A11 is regulated in cancers, especially in triple-negative breast cancer (TNBC). Here we demonstrated that ovarian tumor domain-containing protein 5 (OTUD5), which deubiquitinated and stabilized SLC7A11, played a key role in TNBC progression and paclitaxel chemosensitivity through modulating ferroptosis. The clinical data analysis showed OTUD5 was higher expressed in TNBC, which positively correlated with SLC7A11 level. Mechanistically, OTUD5 interacted with SLC7A11 and cleaved K48-linked polyubiquitin chains from SLC7A11 to enhance the stability of SLC7A11. Taken together, these findings uncover a functional and mechanistic role of OTUD5 in TNBC progression and paclitaxel sensitivity, indicating OTUD5 could be a potential target for TNBC treatment.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142280547","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
Rewired glutamate metabolism diminishes cytostatic action of L-asparaginase 谷氨酸的新陈代谢会削弱 L-天冬酰胺酶的细胞抑制作用。
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-11 DOI: 10.1016/j.canlet.2024.217242
Tumor cells often adapt to amino acid deprivation through metabolic rewiring, compensating for the loss with alternative amino acids/substrates. We have described such a scenario in leukemic cells treated with L-asparaginase (ASNase). Clinical effect of ASNase is based on nutrient stress achieved by its dual enzymatic action which leads to depletion of asparagine and glutamine and is accompanied with elevated aspartate and glutamate concentrations in serum of acute lymphoblastic leukemia patients. We showed that in these limited conditions glutamate uptake compensates for the loss of glutamine availability. Extracellular glutamate flux detection confirms its integration into the TCA cycle and its participation in nucleotide and glutathione synthesis. Importantly, it is glutamate-driven de novo synthesis of glutathione which is the essential metabolic pathway necessary for glutamate's pro-survival effect. In vivo findings support this effect by showing that inhibition of glutamate transporters enhances the therapeutic effect of ASNase. In summary, ASNase induces elevated extracellular glutamate levels under nutrient stress, which leads to a rewiring of intracellular glutamate metabolism and has a negative impact on ASNase treatment.
肿瘤细胞通常会通过新陈代谢重新布线来适应氨基酸的缺失,用替代氨基酸/底物来补偿氨基酸的缺失。我们在用 L-天冬酰胺酶(ASNase)处理的白血病细胞中描述了这种情况。ASNase 的临床效果基于其双重酶促作用所产生的营养压力,这种作用导致天冬酰胺和谷氨酰胺的消耗,并伴随着急性淋巴细胞白血病患者血清中天冬氨酸和谷氨酸浓度的升高。我们的研究表明,在这些有限的条件下,谷氨酸的吸收可以补偿谷氨酰胺的损失。细胞外谷氨酸通量检测证实,谷氨酸融入了 TCA 循环,并参与了核苷酸和谷胱甘肽的合成。重要的是,谷氨酸驱动的谷胱甘肽从头合成是谷氨酸促进生存效应所必需的重要代谢途径。体内研究结果表明,抑制谷氨酸转运体可增强 ASNase 的治疗效果,从而证实了这一作用。总之,ASNase 会在营养压力下诱导细胞外谷氨酸水平升高,从而导致细胞内谷氨酸代谢的重构,并对 ASNase 治疗产生负面影响。
{"title":"Rewired glutamate metabolism diminishes cytostatic action of L-asparaginase","authors":"","doi":"10.1016/j.canlet.2024.217242","DOIUrl":"10.1016/j.canlet.2024.217242","url":null,"abstract":"<div><div>Tumor cells often adapt to amino acid deprivation through metabolic rewiring, compensating for the loss with alternative amino acids/substrates. We have described such a scenario in leukemic cells treated with L-asparaginase (ASNase). Clinical effect of ASNase is based on nutrient stress achieved by its dual enzymatic action which leads to depletion of asparagine and glutamine and is accompanied with elevated aspartate and glutamate concentrations in serum of acute lymphoblastic leukemia patients. We showed that in these limited conditions glutamate uptake compensates for the loss of glutamine availability. Extracellular glutamate flux detection confirms its integration into the TCA cycle and its participation in nucleotide and glutathione synthesis. Importantly, it is glutamate-driven <em>de novo</em> synthesis of glutathione which is the essential metabolic pathway necessary for glutamate's pro-survival effect. <em>In vivo</em> findings support this effect by showing that inhibition of glutamate transporters enhances the therapeutic effect of ASNase. In summary, ASNase induces elevated extracellular glutamate levels under nutrient stress, which leads to a rewiring of intracellular glutamate metabolism and has a negative impact on ASNase treatment.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142280545","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
WTAP weakens oxaliplatin chemosensitivity of colorectal cancer by preventing PANoptosis WTAP 通过阻止泛凋亡削弱奥沙利铂对结直肠癌的化疗敏感性
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-11 DOI: 10.1016/j.canlet.2024.217254

As the most abundant post-transcriptional modification in eukaryotes, N6-methyladenosine (m6A) plays a crucial role in cancer cell proliferation, invasion and chemoresistance. However, its specific effects on chemosensitivity to oxaliplatin-based regimens and the impact of these drugs on m6A methylation levels in colorectal cancer (CRC) remain largely unexplored. In this study, we demonstrated that the m6A methyltransferase Wilms tumor 1-associating protein (WTAP) weakens oxaliplatin chemosensitivity in HCT116 and DLD1 cells. Mechanistically, oxaliplatin treatment upregulated WTAP expression, preventing multiple forms of cell death simultaneously, a process known as PANoptosis, by decreasing intracellular oxidative stress through maintaining the expression of nuclear factor erythroid-2-related factor 2 (NRF2), a major antioxidant response element, in an m6A-dependent manner. In addition, high WTAP expression in CRC patients is associated with a poor prognosis and reduced benefit from standard chemotherapy by clinical data analysis of The Cancer Genome Atlas (TCGA) database and patient cohort study. These findings suggest that targeting WTAP-NRF2-PANoptosis axis could enhance the antitumor efficacy of oxaliplatin-based chemotherapy in CRC treatment.

作为真核生物中最丰富的转录后修饰,N6-甲基腺苷(m6A)在癌细胞增殖、侵袭和化疗抗性中起着至关重要的作用。然而,它对以奥沙利铂为基础的化疗方案的化疗敏感性的具体影响以及这些药物对结直肠癌(CRC)中 m6A 甲基化水平的影响在很大程度上仍未得到探讨。在这项研究中,我们证实了 m6A 甲基转移酶 Wilms tumor 1-associating protein(WTAP)会削弱 HCT116 和 DLD1 细胞对奥沙利铂的化疗敏感性。从机理上讲,奥沙利铂处理可上调 WTAP 的表达,通过维持主要抗氧化反应元件核因子红细胞-2 相关因子 2(NRF2)的表达,以 m6A 依赖性方式降低细胞内氧化应激,从而防止多种形式的细胞同时死亡,这一过程被称为 PANoptosis。此外,通过对癌症基因组图谱(TCGA)数据库和患者队列研究的临床数据分析,WTAP在CRC患者中的高表达与预后不良和标准化疗获益减少有关。这些研究结果表明,靶向 WTAP-NRF2-PANoptosis 轴可提高奥沙利铂化疗在 CRC 治疗中的抗肿瘤疗效。
{"title":"WTAP weakens oxaliplatin chemosensitivity of colorectal cancer by preventing PANoptosis","authors":"","doi":"10.1016/j.canlet.2024.217254","DOIUrl":"10.1016/j.canlet.2024.217254","url":null,"abstract":"<div><p>As the most abundant post-transcriptional modification in eukaryotes, <em>N</em><sup>6</sup>-methyladenosine (m<sup>6</sup>A) plays a crucial role in cancer cell proliferation, invasion and chemoresistance. However, its specific effects on chemosensitivity to oxaliplatin-based regimens and the impact of these drugs on m<sup>6</sup>A methylation levels in colorectal cancer (CRC) remain largely unexplored. In this study, we demonstrated that the m<sup>6</sup>A methyltransferase Wilms tumor 1-associating protein (WTAP) weakens oxaliplatin chemosensitivity in HCT116 and DLD1 cells. Mechanistically, oxaliplatin treatment upregulated WTAP expression, preventing multiple forms of cell death simultaneously, a process known as PANoptosis, by decreasing intracellular oxidative stress through maintaining the expression of nuclear factor erythroid-2-related factor 2 (NRF2), a major antioxidant response element, in an m<sup>6</sup>A-dependent manner. In addition, high WTAP expression in CRC patients is associated with a poor prognosis and reduced benefit from standard chemotherapy by clinical data analysis of The Cancer Genome Atlas (TCGA) database and patient cohort study. These findings suggest that targeting WTAP-NRF2-PANoptosis axis could enhance the antitumor efficacy of oxaliplatin-based chemotherapy in CRC treatment.</p></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0304383524006499/pdfft?md5=c5658d6ca90dcca3ad730f64b9fd96f8&pid=1-s2.0-S0304383524006499-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142271838","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
Letter to the editor on ‘A deep neural network predictor to predict the sensitivity of neoadjuvant chemoradiotherapy in locally advanced rectal cancer’ 致编辑的信,主题为 "预测局部晚期直肠癌新辅助化疗敏感性的深度神经网络预测器
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-11 DOI: 10.1016/j.canlet.2024.217212
{"title":"Letter to the editor on ‘A deep neural network predictor to predict the sensitivity of neoadjuvant chemoradiotherapy in locally advanced rectal cancer’","authors":"","doi":"10.1016/j.canlet.2024.217212","DOIUrl":"10.1016/j.canlet.2024.217212","url":null,"abstract":"","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142232334","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
Bcl-xL is translocated to the nucleus via CtBP2 to epigenetically promote metastasis Bcl-xL通过CtBP2转位到细胞核,从表观遗传学上促进转移
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-10 DOI: 10.1016/j.canlet.2024.217240

Nuclear Bcl-xL is found to promote cancer metastasis independently of its mitochondria-based anti-apoptotic activity. How Bcl-xL is translocated into the nucleus and how nuclear Bcl-xL regulates histone H3 trimethyl Lys4 (H3K4me3) modification have yet to be understood. Here, we report that C-terminal Binding Protein 2 (CtBP2) binds to Bcl-xL via its N-terminus and translocates Bcl-xL into the nucleus. Knockdown of CtBP2 by shRNA decreases the nuclear portion of Bcl-xL and reverses Bcl-xL-induced invasion and metastasis in mouse models. Furthermore, knockout of CtBP2 not only reduces the nuclear portion of Bcl-xL but also suppresses Bcl-xL transcription. The binding between Bcl-xL and CtBP2 is required for their interaction with MLL1, a histone H3K4 methyltransferase. Pharmacologic inhibition of the MLL1 enzymatic activity reverses Bcl-xL-induced H3K4me3 and TGFβ mRNA upregulation, as well as invasion. Moreover, the cleavage under targets and release using nuclease (CUT&RUN) assay coupled with next-generation sequencing reveals that H3K4me3 modifications are particularly enriched in the promotor regions of genes encoding TGFβ and its signaling pathway members in cancer cells overexpressing Bcl-xL. Altogether, the metastatic function of Bcl-xL is mediated by its interaction with CtBP2 and MLL1 and this study offers new therapeutic strategies to treat Bcl-xL-overexpressing cancer.

研究发现,核Bcl-xL可促进癌症转移,而与其线粒体抗凋亡活性无关。Bcl-xL如何转运到细胞核以及核Bcl-xL如何调节组蛋白H3三甲基Lys4(H3K4me3)的修饰尚不清楚。在这里,我们报告了C-末端结合蛋白2(CtBP2)通过其N-末端与Bcl-xL结合,并将Bcl-xL转运到细胞核中。在小鼠模型中,通过 shRNA 敲除 CtBP2 可减少 Bcl-xL 的核部分,并逆转 Bcl-xL 诱导的侵袭和转移。此外,敲除 CtBP2 不仅会减少 Bcl-xL 的核部分,还会抑制 Bcl-xL 的转录。Bcl-xL 和 CtBP2 之间的结合需要它们与组蛋白 H3K4 甲基转移酶 MLL1 的相互作用。药物抑制 MLL1 酶的活性可逆转 Bcl-xL 诱导的 H3K4me3 和 TGFβ mRNA 上调以及侵袭。此外,靶标下的裂解和核酸酶释放(CUT&RUN)检测结合新一代测序发现,在过表达 Bcl-xL 的癌细胞中,H3K4me3 修饰在编码 TGFβ 及其信号通路成员的基因启动子区域特别富集。总之,Bcl-xL的转移功能是由其与CtBP2和MLL1的相互作用介导的,这项研究为治疗过表达Bcl-xL的癌症提供了新的治疗策略。
{"title":"Bcl-xL is translocated to the nucleus via CtBP2 to epigenetically promote metastasis","authors":"","doi":"10.1016/j.canlet.2024.217240","DOIUrl":"10.1016/j.canlet.2024.217240","url":null,"abstract":"<div><p>Nuclear Bcl-xL is found to promote cancer metastasis independently of its mitochondria-based anti-apoptotic activity. How Bcl-xL is translocated into the nucleus and how nuclear Bcl-xL regulates histone H3 trimethyl Lys4 (H3K4me3) modification have yet to be understood. Here, we report that C-terminal Binding Protein 2 (CtBP2) binds to Bcl-xL via its N-terminus and translocates Bcl-xL into the nucleus. Knockdown of CtBP2 by shRNA decreases the nuclear portion of Bcl-xL and reverses Bcl-xL-induced invasion and metastasis in mouse models. Furthermore, knockout of CtBP2 not only reduces the nuclear portion of Bcl-xL but also suppresses Bcl-xL transcription. The binding between Bcl-xL and CtBP2 is required for their interaction with MLL1, a histone H3K4 methyltransferase. Pharmacologic inhibition of the MLL1 enzymatic activity reverses Bcl-xL-induced H3K4me3 and TGFβ mRNA upregulation, as well as invasion. Moreover, the cleavage under targets and release using nuclease (CUT&amp;RUN) assay coupled with next-generation sequencing reveals that H3K4me3 modifications are particularly enriched in the promotor regions of genes encoding TGFβ and its signaling pathway members in cancer cells overexpressing Bcl-xL. Altogether, the metastatic function of Bcl-xL is mediated by its interaction with CtBP2 and MLL1 and this study offers new therapeutic strategies to treat Bcl-xL-overexpressing cancer.</p></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142232333","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
Immune checkpoint inhibitors rechallenge in non-small cell lung cancer: Current evidence and future directions 免疫检查点抑制剂在非小细胞肺癌中的再挑战:当前证据和未来方向
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-10 DOI: 10.1016/j.canlet.2024.217241

Immunotherapy, remarkably immune checkpoint inhibitors (ICIs), has significantly altered the treatment landscape for non-small cell lung cancer (NSCLC). Despite their success, the discontinuation of ICIs therapy may occur due to factors such as prior treatment completion, disease progression during ICIs treatment, or immune-related adverse events (irAEs). As numerous studies highlight the dynamic nature of immune responses and the sustained benefits of ICIs, ICIs rechallenge has become an attractive and feasible option. However, the decision-making process for ICIs rechallenge in clinical settings is complicated by numerous uncertainties. This review systematically analyses existing clinical research evidence, classifying ICIs rechallenge into distinct clinical scenarios, exploring methods to overcome ICIs resistance in rechallenge instances, and identifying biomarkers to select patients likely to benefit from rechallenge. By integrating recent studies and new technologies, we offer crucial recommendations for future clinical trial design and provide a practical guideline to maximize the therapeutic benefits of immunotherapy for NSCLC patients.

免疫疗法,尤其是免疫检查点抑制剂(ICIs),极大地改变了非小细胞肺癌(NSCLC)的治疗格局。尽管免疫检查点抑制剂取得了成功,但由于之前的治疗已结束、治疗期间疾病进展或免疫相关不良事件(irAEs)等因素,ICIs 治疗可能会中断。由于大量研究强调了免疫反应的动态性质和 ICIs 的持续疗效,ICIs 重新挑战已成为一种有吸引力的可行选择。然而,在临床环境中,ICIs 重试的决策过程因众多不确定因素而变得复杂。本综述系统分析了现有的临床研究证据,将 ICIs 重试分为不同的临床情况,探讨了在重试情况下克服 ICIs 耐药性的方法,并确定了生物标志物来选择可能从重试中获益的患者。通过整合最新研究和新技术,我们为未来的临床试验设计提供了重要建议,并为最大限度地发挥免疫疗法对NSCLC患者的治疗效果提供了实用指南。
{"title":"Immune checkpoint inhibitors rechallenge in non-small cell lung cancer: Current evidence and future directions","authors":"","doi":"10.1016/j.canlet.2024.217241","DOIUrl":"10.1016/j.canlet.2024.217241","url":null,"abstract":"<div><p>Immunotherapy, remarkably immune checkpoint inhibitors (ICIs), has significantly altered the treatment landscape for non-small cell lung cancer (NSCLC). Despite their success, the discontinuation of ICIs therapy may occur due to factors such as prior treatment completion, disease progression during ICIs treatment, or immune-related adverse events (irAEs). As numerous studies highlight the dynamic nature of immune responses and the sustained benefits of ICIs, ICIs rechallenge has become an attractive and feasible option. However, the decision-making process for ICIs rechallenge in clinical settings is complicated by numerous uncertainties. This review systematically analyses existing clinical research evidence, classifying ICIs rechallenge into distinct clinical scenarios, exploring methods to overcome ICIs resistance in rechallenge instances, and identifying biomarkers to select patients likely to benefit from rechallenge. By integrating recent studies and new technologies, we offer crucial recommendations for future clinical trial design and provide a practical guideline to maximize the therapeutic benefits of immunotherapy for NSCLC patients.</p></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0304383524006360/pdfft?md5=53c7e40945c977bb341fe976ce574e11&pid=1-s2.0-S0304383524006360-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142230168","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
Hepatic stellate cells promote hepatocellular carcinoma development by regulating histone lactylation: Novel insights from single-cell RNA sequencing and spatial transcriptomics analyses 肝星状细胞通过调节组蛋白乳酰化促进肝细胞癌的发展单细胞 RNA 测序和空间转录组学分析的新发现
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-10 DOI: 10.1016/j.canlet.2024.217243

This study evaluated the cellular heterogeneity and molecular mechanisms of hepatocellular carcinoma (HCC). Single cell RNA sequencing (scRNA-seq), transcriptomic data, histone lactylation-related genes were collected from public databases. Cell-cell interaction, trajectory, pathway, and spatial transcriptome analyses were executed. Differential expression and survival analyses were conducted. Western blot, Real-time reverse transcription PCR (qRT-PCR), and Cell Counting Kit 8 (CCK8) assay were used to detect the expression of αSMA, AKR1B10 and its target genes, and verify the roles of AKR1B10 in HCC cells. Hepatic stellate cell (HSC) subgroups strongly interacted with tumor cell subgroups, and their spatial distribution was heterogeneous. Two candidate prognostic genes (AKR1B10 and RMRP) were obtained. LONP1, NPIPB3, and ZSWIM6 were determined as AKR1B10 targets. Besides, the expression levels of AKR1B10 and αSMA were significantly increased in LX-2 + HepG2 and LX-2 + HuH7 groups compared to those in LX-2 group, respectively. sh-AKR1B10 significantly inhibited the HCC cell proliferation and change the expression of AKR1B10 target genes, Bcl-2, Bax, Pan Kla, and H3K18la at protein levels. Our findings unveil the pivotal role of HSCs in HCC pathogenesis through regulating histone lactylation.

本研究评估了肝细胞癌(HCC)的细胞异质性和分子机制。研究人员从公共数据库中收集了单细胞 RNA 测序(scRNA-seq)、转录组数据和组蛋白乳化相关基因。进行了细胞-细胞相互作用、轨迹、通路和空间转录组分析。进行了差异表达和存活分析。利用 Western 印迹、实时逆转录 PCR(qRT-PCR)和细胞计数试剂盒 8(CCK8)检测αSMA、AKR1B10 及其靶基因的表达,并验证 AKR1B10 在 HCC 细胞中的作用。肝星状细胞(HSC)亚群与肿瘤细胞亚群之间存在着强烈的相互作用,而且它们的空间分布具有异质性。结果发现了两个候选预后基因(AKR1B10 和 RMRP)。LONP1、NPIPB3和ZSWIM6被确定为AKR1B10的靶点。此外,与LX-2组相比,AKR1B10和αSMA在LX-2 + HepG2组和LX-2 + HuH7组的表达水平均显著升高。 sh-AKR1B10能显著抑制HCC细胞的增殖,并在蛋白水平上改变AKR1B10靶基因Bcl-2、Bax、Pan Kla和H3K18la的表达。我们的研究结果揭示了造血干细胞通过调控组蛋白乳酰化在HCC发病机制中的关键作用。
{"title":"Hepatic stellate cells promote hepatocellular carcinoma development by regulating histone lactylation: Novel insights from single-cell RNA sequencing and spatial transcriptomics analyses","authors":"","doi":"10.1016/j.canlet.2024.217243","DOIUrl":"10.1016/j.canlet.2024.217243","url":null,"abstract":"<div><p>This study evaluated the cellular heterogeneity and molecular mechanisms of hepatocellular carcinoma (HCC). Single cell RNA sequencing (scRNA-seq), transcriptomic data, histone lactylation-related genes were collected from public databases. Cell-cell interaction, trajectory, pathway, and spatial transcriptome analyses were executed. Differential expression and survival analyses were conducted. Western blot, Real-time reverse transcription PCR (qRT-PCR), and Cell Counting Kit 8 (CCK8) assay were used to detect the expression of <em>αSMA</em>, <em>AKR1B10</em> and its target genes, and verify the roles of <em>AKR1B10</em> in HCC cells. Hepatic stellate cell (HSC) subgroups strongly interacted with tumor cell subgroups, and their spatial distribution was heterogeneous. Two candidate prognostic genes (<em>AKR1B10</em> and <em>RMRP</em>) were obtained. <em>LONP1</em>, <em>NPIPB3</em>, and <em>ZSWIM6</em> were determined as <em>AKR1B10</em> targets. Besides, the expression levels of AKR1B10 and αSMA were significantly increased in LX-2 + HepG2 and LX-2 + HuH7 groups compared to those in LX-2 group, respectively. sh-<em>AKR1B10</em> significantly inhibited the HCC cell proliferation and change the expression of <em>AKR1B10</em> target genes, Bcl-2, Bax, Pan Kla, and H3K18la at protein levels. Our findings unveil the pivotal role of HSCs in HCC pathogenesis through regulating histone lactylation.</p></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":null,"pages":null},"PeriodicalIF":9.1,"publicationDate":"2024-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142230037","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
Cancer letters
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