首页 > 最新文献

Biochimica et biophysica acta. Reviews on cancer最新文献

英文 中文
Decoding the role of FOXP3 in esophageal cancer: Underlying mechanisms and therapeutic implications 解码 FOXP3 在食管癌中的作用:基本机制和治疗意义。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-11-01 DOI: 10.1016/j.bbcan.2024.189211
Yuanyuan Wang, Lei Xue
Esophageal cancer is a significant contributor to cancer-related mortality, and its poor prognosis is primarily attributed to the aggressive nature of the tumor and challenges in early detection. Currently, there are no ideal drugs developed for treatment, making it crucial to explore potential biomarkers and molecular targets for esophageal cancer. FOXP3, as a transcription factor and major regulator of regulatory T cells, not only plays a role in promoting or inhibiting tumor development in various types of cancer cells including esophageal cancer cells but also influences the function of Treg cells by regulating the expression of multiple genes. This paper provides an in-depth discussion on the functional properties, regulatory mechanisms, key signaling pathways, as well as the role and potential application of FOXP3 in treating esophageal cancer. Furthermore, it comprehensively analyzes the complex role of this transcription factor within the tumor immune microenvironment with an aim to aid in developing new potential targets for esophageal cancer treatment.
食管癌是导致癌症相关死亡率的一个重要因素,其预后不良的主要原因是肿瘤的侵袭性和早期检测方面的挑战。目前还没有理想的治疗药物,因此探索食管癌的潜在生物标志物和分子靶点至关重要。FOXP3 作为转录因子和调节性 T 细胞的主要调控因子,不仅在促进或抑制包括食管癌细胞在内的各类癌细胞的肿瘤发生发展中发挥作用,还通过调控多种基因的表达影响 Treg 细胞的功能。本文深入探讨了 FOXP3 的功能特性、调控机制、关键信号通路以及在食管癌治疗中的作用和潜在应用。此外,它还全面分析了该转录因子在肿瘤免疫微环境中的复杂作用,旨在帮助开发食管癌治疗的新潜在靶点。
{"title":"Decoding the role of FOXP3 in esophageal cancer: Underlying mechanisms and therapeutic implications","authors":"Yuanyuan Wang,&nbsp;Lei Xue","doi":"10.1016/j.bbcan.2024.189211","DOIUrl":"10.1016/j.bbcan.2024.189211","url":null,"abstract":"<div><div>Esophageal cancer is a significant contributor to cancer-related mortality, and its poor prognosis is primarily attributed to the aggressive nature of the tumor and challenges in early detection. Currently, there are no ideal drugs developed for treatment, making it crucial to explore potential biomarkers and molecular targets for esophageal cancer. FOXP3, as a transcription factor and major regulator of regulatory T cells, not only plays a role in promoting or inhibiting tumor development in various types of cancer cells including esophageal cancer cells but also influences the function of Treg cells by regulating the expression of multiple genes. This paper provides an in-depth discussion on the functional properties, regulatory mechanisms, key signaling pathways, as well as the role and potential application of FOXP3 in treating esophageal cancer. Furthermore, it comprehensively analyzes the complex role of this transcription factor within the tumor immune microenvironment with an aim to aid in developing new potential targets for esophageal cancer treatment.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1879 6","pages":"Article 189211"},"PeriodicalIF":9.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142634326","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
Structure, function, signaling pathways and clinical therapeutics: The translational potential of STAT3 as a target for cancer therapy 结构、功能、信号通路和临床疗法:STAT3 作为癌症治疗靶点的转化潜力。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-11-01 DOI: 10.1016/j.bbcan.2024.189207
Dandan Shi , Jiejing Tao , Shuli Man , Ning Zhang , Long Ma , Lanping Guo , Luqi Huang , Wenyuan Gao
Cancer remains one of the most difficult human diseases to overcome because of its complexity and diversity. Signal transducers and transcriptional activators 3 (STAT3) protein has been found to be overexpressed in a wide range of cancer types. Hyperactivation of STAT3 is particularly associated with low survival in cancer patients. This review summarizes the specific molecular mechanisms of STAT3 in cancer development. STAT3 is activated by extracellular signals in the cytoplasm, interacts with different enzymes in the nucleus, mitochondria or endoplasmic reticulum, and subsequently participates in cancer development. The phosphorylated STAT3 at tyrosine 705 site (YP-STAT3) enters the nucleus and regulates a number of tumor-related biological processes such as angiogenesis, migration invasion, cell proliferation and cancer cell stemness. In contrast, the phosphorylated STAT3 at serine 727 site (SP-STAT3) is found on the mitochondria, affects electron respiration transport chain activity and thereby prevents tumor cell apoptosis. SP-STAT3 also appears on the mitochondria-associated endoplasmic reticulum membrane, influences the flow of Ca2+, and affects tumor progression. In addition, we summarize the direct and indirect inhibitors of STAT3 which are currently undergoing clinical studies. Some of them such as TTI101 and BBI608 have been approved by the FDA for the treatment of certain cancers. All in all, STAT3 plays an important role in cancer progression and becomes a potential target for cancer treatment.
由于其复杂性和多样性,癌症仍然是最难攻克的人类疾病之一。信号转导和转录激活因子 3(STAT3)蛋白被发现在多种癌症类型中过度表达。STAT3 的过度激活尤其与癌症患者的低生存率有关。本综述总结了 STAT3 在癌症发展中的具体分子机制。STAT3 在细胞质中被细胞外信号激活,与细胞核、线粒体或内质网中的不同酶相互作用,随后参与癌症的发生发展。酪氨酸 705 位点磷酸化的 STAT3(YP-STAT3)进入细胞核,调节一系列与肿瘤相关的生物过程,如血管生成、迁移侵袭、细胞增殖和癌细胞干性。相反,丝氨酸 727 位点磷酸化的 STAT3(SP-STAT3)存在于线粒体上,影响电子呼吸运输链的活性,从而阻止肿瘤细胞凋亡。SP-STAT3 还出现在线粒体相关的内质网膜上,影响 Ca2+ 的流动,并影响肿瘤的进展。此外,我们还总结了目前正在进行临床研究的 STAT3 直接和间接抑制剂。其中一些抑制剂,如 TTI101 和 BBI608,已被美国食品及药物管理局批准用于治疗某些癌症。总之,STAT3 在癌症进展中扮演着重要角色,并成为癌症治疗的潜在靶点。
{"title":"Structure, function, signaling pathways and clinical therapeutics: The translational potential of STAT3 as a target for cancer therapy","authors":"Dandan Shi ,&nbsp;Jiejing Tao ,&nbsp;Shuli Man ,&nbsp;Ning Zhang ,&nbsp;Long Ma ,&nbsp;Lanping Guo ,&nbsp;Luqi Huang ,&nbsp;Wenyuan Gao","doi":"10.1016/j.bbcan.2024.189207","DOIUrl":"10.1016/j.bbcan.2024.189207","url":null,"abstract":"<div><div>Cancer remains one of the most difficult human diseases to overcome because of its complexity and diversity. Signal transducers and transcriptional activators 3 (STAT3) protein has been found to be overexpressed in a wide range of cancer types. Hyperactivation of STAT3 is particularly associated with low survival in cancer patients. This review summarizes the specific molecular mechanisms of STAT3 in cancer development. STAT3 is activated by extracellular signals in the cytoplasm, interacts with different enzymes in the nucleus, mitochondria or endoplasmic reticulum, and subsequently participates in cancer development. The phosphorylated STAT3 at tyrosine 705 site (YP-STAT3) enters the nucleus and regulates a number of tumor-related biological processes such as angiogenesis, migration invasion, cell proliferation and cancer cell stemness. In contrast, the phosphorylated STAT3 at serine 727 site (SP-STAT3) is found on the mitochondria, affects electron respiration transport chain activity and thereby prevents tumor cell apoptosis. SP-STAT3 also appears on the mitochondria-associated endoplasmic reticulum membrane, influences the flow of Ca<sup>2+</sup>, and affects tumor progression. In addition, we summarize the direct and indirect inhibitors of STAT3 which are currently undergoing clinical studies. Some of them such as TTI101 and BBI608 have been approved by the FDA for the treatment of certain cancers. All in all, STAT3 plays an important role in cancer progression and becomes a potential target for cancer treatment.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1879 6","pages":"Article 189207"},"PeriodicalIF":9.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142585121","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
Roles of K(H)SRP in modulating gene transcription throughout cancer progression: Insights from cellular studies to clinical perspectives K(H)SRP 在整个癌症进展过程中调节基因转录的作用:从细胞研究到临床观点的启示。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-11-01 DOI: 10.1016/j.bbcan.2024.189202
Yi-Chieh Yang , Kuo-Hao Ho , Kuo-Tai Hua , Ming-Hsien Chien
The KH-type splicing regulatory protein (KHSRP), also known as KSRP, is an RNA-binding protein that regulates gene expressions through various mechanisms, including messenger (m)RNA degradation, micro (mi)RNA maturation, and transcriptional activity. KSRP has been implicated in a wide range of physiological and pathological processes, with emerging evidence highlighting its role in modulating diverse aspects of cancer behaviors. In this review, we provide a comprehensive overview of KSRP's clinical relevance and its multifaceted regulatory mechanisms in cancer. Our extensive pan-cancer analysis uncovers associations of KSRP with clinical outcomes and identifies cell cycle progression as a key signaling pathway correlated with KSRP expression. Furthermore, we identify miR-17-5p as critical miRNAs positively correlated with KSRP, and it is associated with poor survival in various cancers. Collectively, this review offers new insights into the potential of KSRP as a target for therapeutic strategies in cancer treatment.
KH 型剪接调控蛋白(KHSRP),又称 KSRP,是一种 RNA 结合蛋白,通过各种机制调控基因表达,包括信使 (m)RNA 降解、微 (mi)RNA 成熟和转录活性。KSRP 与多种生理和病理过程都有关联,新出现的证据强调了它在调节癌症行为的不同方面所起的作用。在这篇综述中,我们全面概述了 KSRP 的临床相关性及其在癌症中的多方面调控机制。我们广泛的泛癌症分析发现了 KSRP 与临床结果的关联,并确定细胞周期进展是与 KSRP 表达相关的关键信号通路。此外,我们还发现 miR-17-5p 是与 KSRP 呈正相关的关键 miRNA,它与各种癌症的不良生存率相关。总之,本综述为 KSRP 作为癌症治疗策略靶点的潜力提供了新的见解。
{"title":"Roles of K(H)SRP in modulating gene transcription throughout cancer progression: Insights from cellular studies to clinical perspectives","authors":"Yi-Chieh Yang ,&nbsp;Kuo-Hao Ho ,&nbsp;Kuo-Tai Hua ,&nbsp;Ming-Hsien Chien","doi":"10.1016/j.bbcan.2024.189202","DOIUrl":"10.1016/j.bbcan.2024.189202","url":null,"abstract":"<div><div>The KH-type splicing regulatory protein (KHSRP), also known as KSRP, is an RNA-binding protein that regulates gene expressions through various mechanisms, including messenger (m)RNA degradation, micro (mi)RNA maturation, and transcriptional activity. KSRP has been implicated in a wide range of physiological and pathological processes, with emerging evidence highlighting its role in modulating diverse aspects of cancer behaviors. In this review, we provide a comprehensive overview of KSRP's clinical relevance and its multifaceted regulatory mechanisms in cancer. Our extensive pan-cancer analysis uncovers associations of KSRP with clinical outcomes and identifies cell cycle progression as a key signaling pathway correlated with KSRP expression. Furthermore, we identify miR-17-5p as critical miRNAs positively correlated with KSRP, and it is associated with poor survival in various cancers. Collectively, this review offers new insights into the potential of KSRP as a target for therapeutic strategies in cancer treatment.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1879 6","pages":"Article 189202"},"PeriodicalIF":9.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142514567","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 curious case of type I interferon signaling in cancer 干扰素-1 信号在癌症中的奇异作用。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-11-01 DOI: 10.1016/j.bbcan.2024.189204
Abu Sufiyan Chhipa , Valentina Boscaro , Margherita Gallicchio , Snehal Patel
Cytokines are the crucial signaling proteins that mediate the crosstalks between the cells of tumor microenvironment (TME). Interferon-1 (IFN-1) are the important cytokines that are widely known for their tumor suppressive roles comprising of cancer cell intrinsic and extrinsic mechanisms. Despite having known antitumor effects, IFN-1 are also reported to have tumor promoting functions under varying circumstances. This dichotomy in the functions of IFN-1 is largely attributed to the acute and chronic activation of IFN-1 signaling in TME. The chronic activation of IFN-1 signaling in tumor cells results in altered stimulation of downstream pathways that result in the expression of tumor promoting proteins, while the acute IFN-1 signaling activation maintains its tumor inhibiting functions. In the present review, we have discussed the anti- and pro-tumor actions of IFN-1 signaling under acute and chronic IFN-1 signaling activation. We have also discussed the downstream changes in signaling components that result in tumor supportive functions of a constitutive IFN-1 signaling. We have further discussed the possible strategies to overcome the detrimental effects of chronic IFN-1 pathway activation and to successfully employ IFN-1 for their beneficial anti-tumor effects.
细胞因子是介导肿瘤微环境(TME)细胞之间相互联系的重要信号蛋白。干扰素-1(IFN-1)是一种重要的细胞因子,其抑制肿瘤的作用已广为人知,包括癌细胞的内在和外在机制。尽管 IFN-1 具有已知的抗肿瘤作用,但也有报道称其在不同情况下具有促进肿瘤生长的功能。IFN-1 功能的这种两极分化主要归因于 IFN-1 信号在 TME 中的急性和慢性激活。肿瘤细胞中 IFN-1 信号的慢性激活会导致下游通路的刺激改变,从而导致肿瘤促进蛋白的表达,而急性 IFN-1 信号激活则会保持其肿瘤抑制功能。在本综述中,我们讨论了在急性和慢性 IFN-1 信号激活情况下 IFN-1 信号的抗肿瘤和促肿瘤作用。我们还讨论了信号成分的下游变化导致组成型 IFN-1 信号的肿瘤支持功能。我们还进一步讨论了克服慢性 IFN-1 通路激活的不利影响并成功利用 IFN-1 发挥有益抗肿瘤作用的可能策略。
{"title":"The curious case of type I interferon signaling in cancer","authors":"Abu Sufiyan Chhipa ,&nbsp;Valentina Boscaro ,&nbsp;Margherita Gallicchio ,&nbsp;Snehal Patel","doi":"10.1016/j.bbcan.2024.189204","DOIUrl":"10.1016/j.bbcan.2024.189204","url":null,"abstract":"<div><div>Cytokines are the crucial signaling proteins that mediate the crosstalks between the cells of tumor microenvironment (TME). Interferon-1 (IFN-1) are the important cytokines that are widely known for their tumor suppressive roles comprising of cancer cell intrinsic and extrinsic mechanisms. Despite having known antitumor effects, IFN-1 are also reported to have tumor promoting functions under varying circumstances. This dichotomy in the functions of IFN-1 is largely attributed to the acute and chronic activation of IFN-1 signaling in TME. The chronic activation of IFN-1 signaling in tumor cells results in altered stimulation of downstream pathways that result in the expression of tumor promoting proteins, while the acute IFN-1 signaling activation maintains its tumor inhibiting functions. In the present review, we have discussed the anti- and pro-tumor actions of IFN-1 signaling under acute and chronic IFN-1 signaling activation. We have also discussed the downstream changes in signaling components that result in tumor supportive functions of a constitutive IFN-1 signaling. We have further discussed the possible strategies to overcome the detrimental effects of chronic IFN-1 pathway activation and to successfully employ IFN-1 for their beneficial anti-tumor effects.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1879 6","pages":"Article 189204"},"PeriodicalIF":9.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549517","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 emerging role of extracellular vesicles and particles in prostate cancer diagnosis, and risk stratification 细胞外囊泡和颗粒在前列腺癌诊断和风险分层中的新作用。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-11-01 DOI: 10.1016/j.bbcan.2024.189210
Qi Wang , Bairen Pang , Joseph Bucci , Junhui Jiang , Yong Li
Current approaches for prostate cancer (PCa) diagnosis and risk stratification require greater accuracy. Extracellular vesicles and particles (EVPs) containing diverse cargos from parent cells are released into the extracellular microenvironment and play a critical role in intercellular communication. Accumulating evidence demonstrates that EVPs are emerging as a promising focus for the exploration of cancer biomarkers and therapeutic targets. However, the precise categorisation and nomenclature of EVP subpopulations remains challenging due to their compositional complexity, inherent heterogeneity in molecular composition, and structure. The recent identification of two novel non-vesicular extracellular particle subtypes, exomeres and supermeres, has altered our understanding of the distinct subpopulations of EVPs and their roles in biological and physiological processes. Here, we discuss recent advances in the field of EVPs, describe characteristics of EVP subpopulations, focus on the application and potential of EVPs in PCa diagnosis and risk stratification by liquid biopsy, and highlight the major challenges and prospects of EVP research in PCa area.
目前的前列腺癌(PCa)诊断和风险分层方法需要更高的准确性。细胞外囊泡和颗粒(EVPs)含有来自母细胞的各种载体,它们被释放到细胞外微环境中,在细胞间通信中发挥着至关重要的作用。越来越多的证据表明,EVPs 正在成为探索癌症生物标志物和治疗靶点的一个有前途的重点。然而,EVP 亚群的精确分类和命名仍具有挑战性,这是因为它们的组成复杂,分子组成和结构存在固有的异质性。最近发现的两种新型非囊状细胞外颗粒亚型--exomeres 和 supermeres--改变了我们对不同 EVP 亚群及其在生物和生理过程中作用的认识。在此,我们将讨论EVP领域的最新进展,描述EVP亚群的特征,重点介绍EVP在PCa诊断和液体活检风险分层中的应用和潜力,并强调PCa领域EVP研究的主要挑战和前景。
{"title":"The emerging role of extracellular vesicles and particles in prostate cancer diagnosis, and risk stratification","authors":"Qi Wang ,&nbsp;Bairen Pang ,&nbsp;Joseph Bucci ,&nbsp;Junhui Jiang ,&nbsp;Yong Li","doi":"10.1016/j.bbcan.2024.189210","DOIUrl":"10.1016/j.bbcan.2024.189210","url":null,"abstract":"<div><div>Current approaches for prostate cancer (PCa) diagnosis and risk stratification require greater accuracy. Extracellular vesicles and particles (EVPs) containing diverse cargos from parent cells are released into the extracellular microenvironment and play a critical role in intercellular communication. Accumulating evidence demonstrates that EVPs are emerging as a promising focus for the exploration of cancer biomarkers and therapeutic targets. However, the precise categorisation and nomenclature of EVP subpopulations remains challenging due to their compositional complexity, inherent heterogeneity in molecular composition, and structure. The recent identification of two novel non-vesicular extracellular particle subtypes, exomeres and supermeres, has altered our understanding of the distinct subpopulations of EVPs and their roles in biological and physiological processes. Here, we discuss recent advances in the field of EVPs, describe characteristics of EVP subpopulations, focus on the application and potential of EVPs in PCa diagnosis and risk stratification by liquid biopsy, and highlight the major challenges and prospects of EVP research in PCa area.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1879 6","pages":"Article 189210"},"PeriodicalIF":9.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142607222","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
Decoding secret role of mesenchymal stem cells in regulating cancer stem cells and drug resistance 解密间充质干细胞在调节癌症干细胞和耐药性方面的秘密作用。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-11-01 DOI: 10.1016/j.bbcan.2024.189205
Sameer Kumar Panda , Nirmal Robinson , Vincenzo Desiderio
Drug resistance caused by the efflux of chemotherapeutic drugs is one of the most challenging obstacles to successful cancer therapy. Several efflux transporters have been identified since the discovery of the P-gp/ABCB1 transporter in 1976. Over the last four decades, researchers have focused on developing efflux transporter inhibitors to overcome drug resistance. However, even with the third-generation inhibitors available, we are still far from effectively inhibiting the efflux transporters. Additionally, Cancer stem cells (CSCs) pose another significant challenge, contributing to cancer recurrence even after successful treatment. The ability of CSCs to enter dormancy and evade detection makes them almost invulnerable to chemotherapeutic drug treatment. In this review, we discuss how Mesenchymal stem cells (MSCs), one of the key components of the Tumor Microenvironment (TME), regulate both the CSCs and efflux transporters. We propose a new approach focusing on MSCs, which can be crucial to successfully address CSCs and efflux transporters.
化疗药物外流导致的耐药性是成功治疗癌症的最大障碍之一。自 1976 年发现 P-gp/ABCB1 转运体以来,已经发现了多种外流转运体。过去四十年来,研究人员一直致力于开发外排转运体抑制剂,以克服耐药性。然而,即使有了第三代抑制剂,我们仍远不能有效抑制外排转运体。此外,癌症干细胞(CSCs)构成了另一个重大挑战,即使在成功治疗后也会导致癌症复发。癌干细胞能够进入休眠状态并逃避检测,这使得它们几乎不受化疗药物治疗的影响。在这篇综述中,我们将讨论间充质干细胞(MSCs)作为肿瘤微环境(TME)的关键组成部分之一,是如何调控CSCs和外排转运体的。我们提出了一种以间叶干细胞为重点的新方法,这对成功解决干细胞和外排转运体问题至关重要。
{"title":"Decoding secret role of mesenchymal stem cells in regulating cancer stem cells and drug resistance","authors":"Sameer Kumar Panda ,&nbsp;Nirmal Robinson ,&nbsp;Vincenzo Desiderio","doi":"10.1016/j.bbcan.2024.189205","DOIUrl":"10.1016/j.bbcan.2024.189205","url":null,"abstract":"<div><div>Drug resistance caused by the efflux of chemotherapeutic drugs is one of the most challenging obstacles to successful cancer therapy. Several efflux transporters have been identified since the discovery of the P-gp/ABCB1 transporter in 1976. Over the last four decades, researchers have focused on developing efflux transporter inhibitors to overcome drug resistance. However, even with the third-generation inhibitors available, we are still far from effectively inhibiting the efflux transporters. Additionally, Cancer stem cells (CSCs) pose another significant challenge, contributing to cancer recurrence even after successful treatment. The ability of CSCs to enter dormancy and evade detection makes them almost invulnerable to chemotherapeutic drug treatment. In this review, we discuss how Mesenchymal stem cells (MSCs), one of the key components of the Tumor Microenvironment (TME), regulate both the CSCs and efflux transporters. We propose a new approach focusing on MSCs, which can be crucial to successfully address CSCs and efflux transporters.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1879 6","pages":"Article 189205"},"PeriodicalIF":9.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142559725","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
Mechanisms of cancer cachexia and targeted therapeutic strategies 癌症恶病质的机理和靶向治疗策略。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-11-01 DOI: 10.1016/j.bbcan.2024.189208
Long Li , Zhi-Qiang Ling
Tumor cachexia is a multifactorial syndrome characterized by systemic dysfunction, including anorexia and severe weight loss that is resistant to standard nutritional interventions. It is estimated that approximately 20 % of cancer patients succumb to cachexia in the later stages of their disease. Thus, understanding its pathogenesis is vital for improving therapeutic outcomes. Recent research has focused on the imbalance between energy intake and expenditure in cachexia. Clinically, cachexia presents with anorexia, adipose tissue atrophy, and skeletal muscle wasting, each driven by distinct mechanisms. Anorexia arises primarily from tumor-secreted factors and cancer-induced hormonal disruptions that impair hypothalamic regulation of appetite. Adipose tissue atrophy is largely attributed to enhanced lipolysis, driven by increased activity of enzymes such as adipose triglyceride lipase and hormone-sensitive lipase, coupled with decreased lipoprotein lipase activity. The browning of white adipose tissue, facilitated by uncoupling protein 1, further accelerates fat breakdown by increasing energy expenditure. Skeletal muscle atrophy, a hallmark of cachexia, results from dysregulated protein turnover via the ubiquitin-proteasome and autophagy-lysosomal pathways, as well as mitochondrial dysfunction. Additionally, chemotherapy can exacerbate cachexia. This review examines the molecular mechanisms underlying cancer cachexia and discusses current therapeutic strategies, aiming to inform future research and improve treatment approaches.
肿瘤恶病质是一种多因素综合征,其特点是全身功能失调,包括厌食和体重严重下降,对标准营养干预措施产生抗药性。据估计,约有 20% 的癌症患者在疾病后期会因恶病质而死亡。因此,了解其发病机制对于改善治疗效果至关重要。近期的研究重点是恶病质中能量摄入和消耗的失衡。在临床上,恶病质表现为厌食、脂肪组织萎缩和骨骼肌萎缩,每种情况都由不同的机制驱动。厌食主要是由于肿瘤分泌的因子和癌症诱发的激素紊乱损害了下丘脑对食欲的调节。脂肪组织萎缩主要是由于脂肪甘油三酯脂肪酶和激素敏感脂肪酶等酶的活性增强,同时脂蛋白脂肪酶的活性降低,导致脂肪分解作用增强。解偶联蛋白 1 促进白色脂肪组织褐变,通过增加能量消耗进一步加速脂肪分解。骨骼肌萎缩是恶病质的一个特征,它是通过泛素-蛋白酶体和自噬-溶酶体途径以及线粒体功能障碍导致蛋白质周转失调的结果。此外,化疗也会加剧恶病质。这篇综述探讨了癌症恶病质的分子机制,并讨论了当前的治疗策略,旨在为未来的研究提供信息并改进治疗方法。
{"title":"Mechanisms of cancer cachexia and targeted therapeutic strategies","authors":"Long Li ,&nbsp;Zhi-Qiang Ling","doi":"10.1016/j.bbcan.2024.189208","DOIUrl":"10.1016/j.bbcan.2024.189208","url":null,"abstract":"<div><div>Tumor cachexia is a multifactorial syndrome characterized by systemic dysfunction, including anorexia and severe weight loss that is resistant to standard nutritional interventions. It is estimated that approximately 20 % of cancer patients succumb to cachexia in the later stages of their disease. Thus, understanding its pathogenesis is vital for improving therapeutic outcomes. Recent research has focused on the imbalance between energy intake and expenditure in cachexia. Clinically, cachexia presents with anorexia, adipose tissue atrophy, and skeletal muscle wasting, each driven by distinct mechanisms. Anorexia arises primarily from tumor-secreted factors and cancer-induced hormonal disruptions that impair hypothalamic regulation of appetite. Adipose tissue atrophy is largely attributed to enhanced lipolysis, driven by increased activity of enzymes such as adipose triglyceride lipase and hormone-sensitive lipase, coupled with decreased lipoprotein lipase activity. The browning of white adipose tissue, facilitated by uncoupling protein 1, further accelerates fat breakdown by increasing energy expenditure. Skeletal muscle atrophy, a hallmark of cachexia, results from dysregulated protein turnover via the ubiquitin-proteasome and autophagy-lysosomal pathways, as well as mitochondrial dysfunction. Additionally, chemotherapy can exacerbate cachexia. This review examines the molecular mechanisms underlying cancer cachexia and discusses current therapeutic strategies, aiming to inform future research and improve treatment approaches.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1879 6","pages":"Article 189208"},"PeriodicalIF":9.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142634257","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
An updated outlook on autophagy mechanism and how it supports acute myeloid leukemia maintenance 自噬机制及其如何支持急性髓性白血病维持的最新展望。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-11-01 DOI: 10.1016/j.bbcan.2024.189214
Brunno Gilberto Santos de Macedo , Manuela Albuquerque de Melo , Diego Antonio Pereira-Martins , João Agostinho Machado-Neto , Fabiola Traina
The gradual acquisition of genetic and epigenetic disturbances bestows malignant traits upon hematopoietic stem cells, subverting them into a founder and reservoir cell for de novo acute myeloid leukemia (AML) known as leukemic stem cells (LSC). Beyond its molecular heterogeneity, AML is also characterized by rewiring biological processes to support its onset and maintenance.
LSC were observed to inherently and actively trigger mitochondrial turnover through selective autophagic removal such that impairing the process led to cell differentiation at the expense of its stemness.
This review provides a current take on autophagy regulation mechanisms according to the current molecular characterization of the process; describes autophagy as a drug resistance mechanism, and a pivotal mechanism whereby LSC harmonize their strong reliance on mitochondrial respiration to obtain energy, and their necessity for lower internal oxidative stress to avoid exhaustion. Therefore, targeting autophagy presents a promising strategy to promote long-term remissions in AML.
造血干细胞逐渐获得遗传和表观遗传学干扰,赋予其恶性特征,使其成为新发急性髓性白血病(AML)的创始细胞和储库细胞,即白血病干细胞(LSC)。除了分子异质性外,急性髓细胞白血病还具有重构生物过程以支持其发病和维持的特点。据观察,白血病干细胞通过选择性自噬清除,固有地、主动地触发线粒体周转,因此损害这一过程会导致细胞分化,牺牲细胞的干性。本综述根据目前自噬过程的分子特征,对自噬调控机制进行了分析;将自噬描述为一种耐药机制,也是一种关键机制,LSC 可借此协调其对线粒体呼吸获取能量的强烈依赖和降低内部氧化应激以避免衰竭的必要性。因此,以自噬为靶点是促进急性髓细胞性白血病长期缓解的一种有前景的策略。
{"title":"An updated outlook on autophagy mechanism and how it supports acute myeloid leukemia maintenance","authors":"Brunno Gilberto Santos de Macedo ,&nbsp;Manuela Albuquerque de Melo ,&nbsp;Diego Antonio Pereira-Martins ,&nbsp;João Agostinho Machado-Neto ,&nbsp;Fabiola Traina","doi":"10.1016/j.bbcan.2024.189214","DOIUrl":"10.1016/j.bbcan.2024.189214","url":null,"abstract":"<div><div>The gradual acquisition of genetic and epigenetic disturbances bestows malignant traits upon hematopoietic stem cells, subverting them into a founder and reservoir cell for <em>de novo</em> acute myeloid leukemia (AML) known as leukemic stem cells (LSC). Beyond its molecular heterogeneity, AML is also characterized by rewiring biological processes to support its onset and maintenance.</div><div>LSC were observed to inherently and actively trigger mitochondrial turnover through selective autophagic removal such that impairing the process led to cell differentiation at the expense of its stemness.</div><div>This review provides a current take on autophagy regulation mechanisms according to the current molecular characterization of the process; describes autophagy as a drug resistance mechanism, and a pivotal mechanism whereby LSC harmonize their strong reliance on mitochondrial respiration to obtain energy, and their necessity for lower internal oxidative stress to avoid exhaustion. Therefore, targeting autophagy presents a promising strategy to promote long-term remissions in AML.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1879 6","pages":"Article 189214"},"PeriodicalIF":9.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142634291","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
Advances of SS18-SSX fusion gene in synovial sarcoma: Emerging novel functions and therapeutic potentials 滑膜肉瘤中 SS18-SSX 融合基因的研究进展:新功能和治疗潜力。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-11-01 DOI: 10.1016/j.bbcan.2024.189215
Chongmin Ren , Jia Liu , Francis J. Hornicek , Bin Yue , Zhenfeng Duan
Synovial sarcoma is a rare type of soft tissue sarcoma that primarily affects adolescents and young adults, featured by aggressive behavior and a high potential for metastasis. Genetically, synovial sarcoma is defined by the fusion oncogene SS18-SSX arising from the translocation of t(X;18)(p11;q11). SS18-SSX fusion gene is the major driver of the oncogenic event in synovial sarcoma. SS18-SSX fusion protein, while not containing any DNA-binding motifs, binds to the SWI/SNF (BAF) complex, a major epigenetic regulator, leading to the disruption of gene expression which results in tumor initiation and progression. Emerging studies on the molecular mechanisms of SS18-SSX associated signaling pathway hold promise for developments in diagnosis and treatments. Advanced diagnostic methods facilitate early and precise detection of the tumor, enabling disease monitoring and prognostic improvements. Treatment of synovial sarcoma typically comprises local surgery, radiotherapy and chemotherapy, while novel managements such as immunotherapy, targeted therapies and epigenetic modifiers are explored. This review focuses on the recent studies of SS18-SSX fusion gene, epigenetic landscape, signaling pathways, diagnostic techniques, and relevant therapeutic advances, aiming to inhibit the oncogenic processes and improve outcomes for patients with synovial sarcoma.
滑膜肉瘤是一种罕见的软组织肉瘤,主要累及青少年和年轻人,具有侵袭性和高度转移性。从遗传学角度看,滑膜肉瘤是由 t(X;18)(p11;q11)易位产生的融合癌基因 SS18-SSX 确定的。SS18-SSX 融合基因是滑膜肉瘤致癌事件的主要驱动因素。SS18-SSX 融合蛋白虽然不含任何 DNA 结合基团,却能与 SWI/SNF (BAF)复合物(一种主要的表观遗传调节因子)结合,导致基因表达紊乱,从而导致肿瘤的发生和发展。有关 SS18-SSX 信号通路分子机制的新研究为诊断和治疗带来了希望。先进的诊断方法有助于早期精确检测肿瘤,从而实现疾病监测和改善预后。滑膜肉瘤的治疗通常包括局部手术、放疗和化疗,同时也在探索免疫疗法、靶向疗法和表观遗传修饰剂等新型治疗方法。本综述重点介绍 SS18-SSX 融合基因、表观遗传格局、信号通路、诊断技术和相关治疗进展的最新研究,旨在抑制致癌过程,改善滑膜肉瘤患者的预后。
{"title":"Advances of SS18-SSX fusion gene in synovial sarcoma: Emerging novel functions and therapeutic potentials","authors":"Chongmin Ren ,&nbsp;Jia Liu ,&nbsp;Francis J. Hornicek ,&nbsp;Bin Yue ,&nbsp;Zhenfeng Duan","doi":"10.1016/j.bbcan.2024.189215","DOIUrl":"10.1016/j.bbcan.2024.189215","url":null,"abstract":"<div><div>Synovial sarcoma is a rare type of soft tissue sarcoma that primarily affects adolescents and young adults, featured by aggressive behavior and a high potential for metastasis. Genetically, synovial sarcoma is defined by the fusion oncogene SS18-SSX arising from the translocation of t(X;18)(p11;q11). SS18-SSX fusion gene is the major driver of the oncogenic event in synovial sarcoma. SS18-SSX fusion protein, while not containing any DNA-binding motifs, binds to the SWI/SNF (BAF) complex, a major epigenetic regulator, leading to the disruption of gene expression which results in tumor initiation and progression. Emerging studies on the molecular mechanisms of SS18-SSX associated signaling pathway hold promise for developments in diagnosis and treatments. Advanced diagnostic methods facilitate early and precise detection of the tumor, enabling disease monitoring and prognostic improvements. Treatment of synovial sarcoma typically comprises local surgery, radiotherapy and chemotherapy, while novel managements such as immunotherapy, targeted therapies and epigenetic modifiers are explored. This review focuses on the recent studies of SS18-SSX fusion gene, epigenetic landscape, signaling pathways, diagnostic techniques, and relevant therapeutic advances, aiming to inhibit the oncogenic processes and improve outcomes for patients with synovial sarcoma.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1879 6","pages":"Article 189215"},"PeriodicalIF":9.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142634275","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
Developing pioneering pharmacological strategies with CRISPR/Cas9 library screening to overcome cancer drug resistance 利用 CRISPR/Cas9 文库筛选技术开发开创性药理学策略,克服癌症抗药性。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-11-01 DOI: 10.1016/j.bbcan.2024.189212
Yu He , Huan Li , Xueming Ju , Bo Gong
Cancer drug resistance is a major obstacle to the effectiveness of chemoradiotherapy, targeted therapy, and immunotherapy. CRISPR/Cas9 library screening has emerged as a powerful genetic screening tool with significant potential to address this challenge. This review provides an overview of the development, methodologies, and applications of CRISPR/Cas9 library screening in the study of cancer drug resistance. We explore its role in elucidating resistance mechanisms, identifying novel anticancer targets, and optimizing treatment strategies. The use of in vivo single-cell CRISPR screens is also highlighted for their capacity to reveal T-cell regulatory networks in cancer immunotherapy. Challenges in clinical translation are discussed, including off-target effects, complexities in data interpretation, and model selection. Despite these obstacles, continuous technological advancements indicate a promising future for CRISPR/Cas9 library screening in overcoming cancer drug resistance.
癌症耐药性是化放疗、靶向治疗和免疫治疗有效性的主要障碍。CRISPR/Cas9 文库筛选已成为一种强大的基因筛选工具,具有应对这一挑战的巨大潜力。本综述概述了 CRISPR/Cas9 文库筛选在癌症耐药性研究中的发展、方法和应用。我们探讨了它在阐明抗药性机制、确定新型抗癌靶点和优化治疗策略方面的作用。此外,我们还强调了体内单细胞 CRISPR 筛选在揭示癌症免疫疗法中的 T 细胞调控网络方面的作用。报告还讨论了临床转化过程中面临的挑战,包括脱靶效应、数据解读的复杂性和模型选择。尽管存在这些障碍,但技术的不断进步预示着 CRISPR/Cas9 文库筛选在克服癌症耐药性方面大有可为。
{"title":"Developing pioneering pharmacological strategies with CRISPR/Cas9 library screening to overcome cancer drug resistance","authors":"Yu He ,&nbsp;Huan Li ,&nbsp;Xueming Ju ,&nbsp;Bo Gong","doi":"10.1016/j.bbcan.2024.189212","DOIUrl":"10.1016/j.bbcan.2024.189212","url":null,"abstract":"<div><div>Cancer drug resistance is a major obstacle to the effectiveness of chemoradiotherapy, targeted therapy, and immunotherapy. CRISPR/Cas9 library screening has emerged as a powerful genetic screening tool with significant potential to address this challenge. This review provides an overview of the development, methodologies, and applications of CRISPR/Cas9 library screening in the study of cancer drug resistance. We explore its role in elucidating resistance mechanisms, identifying novel anticancer targets, and optimizing treatment strategies. The use of <em>in vivo</em> single-cell CRISPR screens is also highlighted for their capacity to reveal T-cell regulatory networks in cancer immunotherapy. Challenges in clinical translation are discussed, including off-target effects, complexities in data interpretation, and model selection. Despite these obstacles, continuous technological advancements indicate a promising future for CRISPR/Cas9 library screening in overcoming cancer drug resistance.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1879 6","pages":"Article 189212"},"PeriodicalIF":9.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142634331","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
Biochimica et biophysica acta. Reviews on cancer
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