首页 > 最新文献

Cell stem cell最新文献

英文 中文
Modeling blood-brain barrier formation and cerebral cavernous malformations in human PSC-derived organoids 在人造血干细胞衍生的器官组织中模拟血脑屏障的形成和脑海绵畸形
IF 23.9 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-15 DOI: 10.1016/j.stem.2024.04.019
Lan Dao, Zhen You, Lu Lu, Tianyang Xu, Avijite Kumer Sarkar, Hui Zhu, Miao Liu, Riccardo Calandrelli, George Yoshida, Pei Lin, Yifei Miao, Sarah Mierke, Srijan Kalva, Haining Zhu, Mingxia Gu, Sudhakar Vadivelu, Sheng Zhong, L. Frank Huang, Ziyuan Guo

The human blood-brain barrier (hBBB) is a highly specialized structure that regulates passage across blood and central nervous system (CNS) compartments. Despite its critical physiological role, there are no reliable in vitro models that can mimic hBBB development and function. Here, we constructed hBBB assembloids from brain and blood vessel organoids derived from human pluripotent stem cells. We validated the acquisition of blood-brain barrier (BBB)-specific molecular, cellular, transcriptomic, and functional characteristics and uncovered an extensive neuro-vascular crosstalk with a spatial pattern within hBBB assembloids. When we used patient-derived hBBB assembloids to model cerebral cavernous malformations (CCMs), we found that these assembloids recapitulated the cavernoma anatomy and BBB breakdown observed in patients. Upon comparison of phenotypes and transcriptome between patient-derived hBBB assembloids and primary human cavernoma tissues, we uncovered CCM-related molecular and cellular alterations. Taken together, we report hBBB assembloids that mimic the core properties of the hBBB and identify a potentially underlying cause of CCMs.

人体血脑屏障(hBBB)是一种高度特化的结构,可调节血液和中枢神经系统(CNS)之间的通道。尽管血脑屏障具有重要的生理作用,但目前还没有可靠的体外模型可以模拟血脑屏障的发育和功能。在这里,我们利用从人类多能干细胞中提取的脑和血管器官组织构建了hBBB组装体。我们验证了血脑屏障(BBB)特异性分子、细胞、转录组和功能特征的获得,并在hBBB组装体中发现了具有空间模式的广泛神经-血管串联。当我们使用源自患者的 hBBB 组合体来模拟脑海绵畸形(CCMs)时,我们发现这些组合体再现了在患者身上观察到的海绵瘤解剖结构和 BBB 破坏。通过比较患者来源的 hBBB 组合体与原发性人类海绵状瘤组织之间的表型和转录组,我们发现了与海绵状瘤相关的分子和细胞改变。总之,我们报告的 hBBB 组合体模拟了 hBBB 的核心特性,并确定了 CCM 的潜在根本原因。
{"title":"Modeling blood-brain barrier formation and cerebral cavernous malformations in human PSC-derived organoids","authors":"Lan Dao, Zhen You, Lu Lu, Tianyang Xu, Avijite Kumer Sarkar, Hui Zhu, Miao Liu, Riccardo Calandrelli, George Yoshida, Pei Lin, Yifei Miao, Sarah Mierke, Srijan Kalva, Haining Zhu, Mingxia Gu, Sudhakar Vadivelu, Sheng Zhong, L. Frank Huang, Ziyuan Guo","doi":"10.1016/j.stem.2024.04.019","DOIUrl":"https://doi.org/10.1016/j.stem.2024.04.019","url":null,"abstract":"<p>The human blood-brain barrier (hBBB) is a highly specialized structure that regulates passage across blood and central nervous system (CNS) compartments. Despite its critical physiological role, there are no reliable <em>in vitro</em> models that can mimic hBBB development and function. Here, we constructed hBBB assembloids from brain and blood vessel organoids derived from human pluripotent stem cells. We validated the acquisition of blood-brain barrier (BBB)-specific molecular, cellular, transcriptomic, and functional characteristics and uncovered an extensive neuro-vascular crosstalk with a spatial pattern within hBBB assembloids. When we used patient-derived hBBB assembloids to model cerebral cavernous malformations (CCMs), we found that these assembloids recapitulated the cavernoma anatomy and BBB breakdown observed in patients. Upon comparison of phenotypes and transcriptome between patient-derived hBBB assembloids and primary human cavernoma tissues, we uncovered CCM-related molecular and cellular alterations. Taken together, we report hBBB assembloids that mimic the core properties of the hBBB and identify a potentially underlying cause of CCMs.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":null,"pages":null},"PeriodicalIF":23.9,"publicationDate":"2024-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140942880","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
Autophagy counters inflammation-driven glycolytic impairment in aging hematopoietic stem cells 自噬对抗衰老造血干细胞中炎症驱动的糖酵解损伤
IF 23.9 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-15 DOI: 10.1016/j.stem.2024.04.020
Paul V. Dellorusso, Melissa A. Proven, Fernando J. Calero-Nieto, Xiaonan Wang, Carl A. Mitchell, Felix Hartmann, Meelad Amouzgar, Patricia Favaro, Andrew DeVilbiss, James W. Swann, Theodore T. Ho, Zhiyu Zhao, Sean C. Bendall, Sean Morrison, Berthold Göttgens, Emmanuelle Passegué

Autophagy is central to the benefits of longevity signaling programs and to hematopoietic stem cell (HSC) response to nutrient stress. With age, a subset of HSCs increases autophagy flux and preserves regenerative capacity, but the signals triggering autophagy and maintaining the functionality of autophagy-activated old HSCs (oHSCs) remain unknown. Here, we demonstrate that autophagy is an adaptive cytoprotective response to chronic inflammation in the aging murine bone marrow (BM) niche. We find that inflammation impairs glucose uptake and suppresses glycolysis in oHSCs through Socs3-mediated inhibition of AKT/FoxO-dependent signaling, with inflammation-mediated autophagy engagement preserving functional quiescence by enabling metabolic adaptation to glycolytic impairment. Moreover, we show that transient autophagy induction via a short-term fasting/refeeding paradigm normalizes glycolytic flux and significantly boosts oHSC regenerative potential. Our results identify inflammation-driven glucose hypometabolism as a key driver of HSC dysfunction with age and establish autophagy as a targetable node to reset oHSC regenerative capacity.

自噬是长寿信号程序和造血干细胞(HSC)应对营养压力的核心。随着年龄的增长,造血干细胞的一个亚群会增加自噬通量并保持再生能力,但触发自噬和维持自噬激活的老年造血干细胞(oHSCs)功能的信号仍然未知。在这里,我们证明自噬是衰老小鼠骨髓(BM)龛对慢性炎症的一种适应性细胞保护反应。我们发现,炎症会损害葡萄糖摄取,并通过 Socs3 介导的 AKT/FoxO 依赖性信号传导抑制 oHSCs 中的糖酵解,而炎症介导的自噬参与可使代谢适应糖酵解损伤,从而维持功能性静止。此外,我们还发现,通过短期禁食/进食模式诱导自噬可使糖酵解通量恢复正常,并显著提高 oHSC 的再生潜力。我们的研究结果表明,炎症驱动的糖代谢低下是造血干细胞随年龄增长而出现功能障碍的一个关键驱动因素,并确定自噬是重置oHSC再生能力的一个目标节点。
{"title":"Autophagy counters inflammation-driven glycolytic impairment in aging hematopoietic stem cells","authors":"Paul V. Dellorusso, Melissa A. Proven, Fernando J. Calero-Nieto, Xiaonan Wang, Carl A. Mitchell, Felix Hartmann, Meelad Amouzgar, Patricia Favaro, Andrew DeVilbiss, James W. Swann, Theodore T. Ho, Zhiyu Zhao, Sean C. Bendall, Sean Morrison, Berthold Göttgens, Emmanuelle Passegué","doi":"10.1016/j.stem.2024.04.020","DOIUrl":"https://doi.org/10.1016/j.stem.2024.04.020","url":null,"abstract":"<p>Autophagy is central to the benefits of longevity signaling programs and to hematopoietic stem cell (HSC) response to nutrient stress. With age, a subset of HSCs increases autophagy flux and preserves regenerative capacity, but the signals triggering autophagy and maintaining the functionality of autophagy-activated old HSCs (oHSCs) remain unknown. Here, we demonstrate that autophagy is an adaptive cytoprotective response to chronic inflammation in the aging murine bone marrow (BM) niche. We find that inflammation impairs glucose uptake and suppresses glycolysis in oHSCs through Socs3-mediated inhibition of AKT/FoxO-dependent signaling, with inflammation-mediated autophagy engagement preserving functional quiescence by enabling metabolic adaptation to glycolytic impairment. Moreover, we show that transient autophagy induction via a short-term fasting/refeeding paradigm normalizes glycolytic flux and significantly boosts oHSC regenerative potential. Our results identify inflammation-driven glucose hypometabolism as a key driver of HSC dysfunction with age and establish autophagy as a targetable node to reset oHSC regenerative capacity.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":null,"pages":null},"PeriodicalIF":23.9,"publicationDate":"2024-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140942948","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
Breast cancer-on-chip for patient-specific efficacy and safety testing of CAR-T cells 用于 CAR-T 细胞特异性疗效和安全性测试的乳腺癌芯片
IF 23.9 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-15 DOI: 10.1016/j.stem.2024.04.018
Tengku Ibrahim Maulana, Claudia Teufel, Madalena Cipriano, Julia Roosz, Lisa Lazarevski, Francijna E. van den Hil, Lukas Scheller, Valeria Orlova, André Koch, Michael Hudecek, Miriam Alb, Peter Loskill

Physiologically relevant human models that recapitulate the challenges of solid tumors and the tumor microenvironment (TME) are highly desired in the chimeric antigen receptor (CAR)-T cell field. We developed a breast cancer-on-chip model with an integrated endothelial barrier that enables the transmigration of perfused immune cells, their infiltration into the tumor, and concomitant monitoring of cytokine release during perfused culture over a period of up to 8 days. Here, we exemplified its use for investigating CAR-T cell efficacy and the ability to control the immune reaction with a pharmacological on/off switch. Additionally, we integrated primary breast cancer organoids to study patient-specific CAR-T cell efficacy. The modular architecture of our tumor-on-chip paves the way for studying the role of other cell types in the TME and thus provides the potential for broad application in bench-to-bedside translation as well as acceleration of the preclinical development of CAR-T cell products.

嵌合抗原受体(CAR)-T 细胞领域非常需要能再现实体瘤和肿瘤微环境(TME)挑战的生理相关人体模型。我们开发了一种集成了内皮屏障的乳腺癌芯片模型,它能使灌注的免疫细胞迁移、浸润到肿瘤中,并在长达 8 天的灌注培养过程中同时监测细胞因子的释放。在这里,我们举例说明了它在研究 CAR-T 细胞疗效方面的用途,以及通过药理开关控制免疫反应的能力。此外,我们还整合了原发性乳腺癌器官组织来研究患者特异性 CAR-T 细胞的疗效。我们的肿瘤芯片的模块化结构为研究TME中其他细胞类型的作用铺平了道路,从而为从台架到床边的转化提供了广泛的应用潜力,并加速了CAR-T细胞产品的临床前开发。
{"title":"Breast cancer-on-chip for patient-specific efficacy and safety testing of CAR-T cells","authors":"Tengku Ibrahim Maulana, Claudia Teufel, Madalena Cipriano, Julia Roosz, Lisa Lazarevski, Francijna E. van den Hil, Lukas Scheller, Valeria Orlova, André Koch, Michael Hudecek, Miriam Alb, Peter Loskill","doi":"10.1016/j.stem.2024.04.018","DOIUrl":"https://doi.org/10.1016/j.stem.2024.04.018","url":null,"abstract":"<p>Physiologically relevant human models that recapitulate the challenges of solid tumors and the tumor microenvironment (TME) are highly desired in the chimeric antigen receptor (CAR)-T cell field. We developed a breast cancer-on-chip model with an integrated endothelial barrier that enables the transmigration of perfused immune cells, their infiltration into the tumor, and concomitant monitoring of cytokine release during perfused culture over a period of up to 8 days. Here, we exemplified its use for investigating CAR-T cell efficacy and the ability to control the immune reaction with a pharmacological on/off switch. Additionally, we integrated primary breast cancer organoids to study patient-specific CAR-T cell efficacy. The modular architecture of our tumor-on-chip paves the way for studying the role of other cell types in the TME and thus provides the potential for broad application in bench-to-bedside translation as well as acceleration of the preclinical development of CAR-T cell products.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":null,"pages":null},"PeriodicalIF":23.9,"publicationDate":"2024-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140942766","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
Transcription factor dynamics, oscillation, and functions in human enteroendocrine cell differentiation 人类肠内分泌细胞分化过程中转录因子的动态、振荡和功能
IF 23.9 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-10 DOI: 10.1016/j.stem.2024.04.015
Pratik N.P. Singh, Wei Gu, Shariq Madha, Allen W. Lynch, Paloma Cejas, Ruiyang He, Swarnabh Bhattacharya, Miguel Muñoz Gomez, Matthew G. Oser, Myles Brown, Henry W. Long, Clifford A. Meyer, Qiao Zhou, Ramesh A. Shivdasani

Enteroendocrine cells (EECs) secrete serotonin (enterochromaffin [EC] cells) or specific peptide hormones (non-EC cells) that serve vital metabolic functions. The basis for terminal EEC diversity remains obscure. By forcing activity of the transcription factor (TF) NEUROG3 in 2D cultures of human intestinal stem cells, we replicated physiologic EEC differentiation and examined transcriptional and cis-regulatory dynamics that culminate in discrete cell types. Abundant EEC precursors expressed stage-specific genes and TFs. Before expressing pre-terminal NEUROD1, post-mitotic precursors oscillated between transcriptionally distinct ASCL1+ and HES6hi cell states. Loss of either factor accelerated EEC differentiation substantially and disrupted EEC individuality; ASCL1 or NEUROD1 deficiency had opposing consequences on EC and non-EC cell features. These TFs mainly bind cis-elements that are accessible in undifferentiated stem cells, and they tailor subsequent expression of TF combinations that underlie discrete EEC identities. Thus, early TF oscillations retard EEC maturation to enable accurate diversity within a medically important cell lineage.

肠内分泌细胞(EECs)分泌血清素(肠粘膜细胞[EC])或特定的肽类激素(非肠粘膜细胞),发挥着重要的新陈代谢功能。终端 EEC 多样性的基础仍然模糊不清。通过强迫转录因子(TF)NEUROG3在人类肠干细胞二维培养物中的活性,我们复制了EEC的生理性分化,并研究了最终形成不同细胞类型的转录和顺式调控动态。丰富的EEC前体表达了阶段特异性基因和TFs。在表达前端 NEUROD1 之前,有丝分裂后的前体在转录不同的 ASCL1+ 和 HES6hi 细胞状态之间摇摆。ASCL1或NEUROD1的缺失对EC和非EC细胞特征的影响截然相反。这些TF主要与未分化干细胞中可接触到的顺式元件结合,它们可调整TF组合的后续表达,而这些TF组合是EEC离散特性的基础。因此,早期TF振荡会延缓EEC的成熟,从而使这一医学上重要的细胞系具有准确的多样性。
{"title":"Transcription factor dynamics, oscillation, and functions in human enteroendocrine cell differentiation","authors":"Pratik N.P. Singh, Wei Gu, Shariq Madha, Allen W. Lynch, Paloma Cejas, Ruiyang He, Swarnabh Bhattacharya, Miguel Muñoz Gomez, Matthew G. Oser, Myles Brown, Henry W. Long, Clifford A. Meyer, Qiao Zhou, Ramesh A. Shivdasani","doi":"10.1016/j.stem.2024.04.015","DOIUrl":"https://doi.org/10.1016/j.stem.2024.04.015","url":null,"abstract":"<p>Enteroendocrine cells (EECs) secrete serotonin (enterochromaffin [EC] cells) or specific peptide hormones (non-EC cells) that serve vital metabolic functions. The basis for terminal EEC diversity remains obscure. By forcing activity of the transcription factor (TF) NEUROG3 in 2D cultures of human intestinal stem cells, we replicated physiologic EEC differentiation and examined transcriptional and <em>cis</em>-regulatory dynamics that culminate in discrete cell types. Abundant EEC precursors expressed stage-specific genes and TFs. Before expressing pre-terminal NEUROD1, post-mitotic precursors oscillated between transcriptionally distinct <em>ASCL1</em><sup><em>+</em></sup> and <em>HES6</em><sup><em>hi</em></sup> cell states. Loss of either factor accelerated EEC differentiation substantially and disrupted EEC individuality; ASCL1 or NEUROD1 deficiency had opposing consequences on EC and non-EC cell features. These TFs mainly bind <em>cis</em>-elements that are accessible in undifferentiated stem cells, and they tailor subsequent expression of TF combinations that underlie discrete EEC identities. Thus, early TF oscillations retard EEC maturation to enable accurate diversity within a medically important cell lineage.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":null,"pages":null},"PeriodicalIF":23.9,"publicationDate":"2024-05-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140903336","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
Metabolic regulator ERRγ governs gastric stem cell differentiation into acid-secreting parietal cells 代谢调节因子ERRγ控制胃干细胞向分泌胃酸的顶叶细胞分化
IF 23.9 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-10 DOI: 10.1016/j.stem.2024.04.016
Mahliyah Adkins-Threats, Sumimasa Arimura, Yang-Zhe Huang, Margarita Divenko, Sarah To, Heather Mao, Yongji Zeng, Jenie Y. Hwang, Joseph R. Burclaff, Shilpa Jain, Jason C. Mills

Parietal cells (PCs) produce gastric acid to kill pathogens and aid digestion. Dysregulated PC census is common in disease, yet how PCs differentiate is unclear. Here, we identify the PC progenitors arising from isthmal stem cells, using mouse models and human gastric cells, and show that they preferentially express cell-metabolism regulator and orphan nuclear receptor Estrogen-related receptor gamma (Esrrg, encoding ERRγ). Esrrg expression facilitated the tracking of stepwise molecular, cellular, and ultrastructural stages of PC differentiation. EsrrgP2ACreERT2 lineage tracing revealed that Esrrg expression commits progenitors to differentiate into mature PCs. scRNA-seq indicated the earliest Esrrg+ PC progenitors preferentially express SMAD4 and SP1 transcriptional targets and the GTPases regulating acid-secretion signal transduction. As progenitors matured, ERRγ-dependent metabolic transcripts predominated. Organoid and mouse studies validated the requirement of ERRγ for PC differentiation. Our work chronicles stem cell differentiation along a single lineage in vivo and suggests ERRγ as a therapeutic target for PC-related disorders.

顶叶细胞(PC)产生胃酸,以杀死病原体并帮助消化。PC普查失调在疾病中很常见,但PC如何分化尚不清楚。在这里,我们利用小鼠模型和人类胃细胞确定了由峡部干细胞产生的PC祖细胞,并表明它们优先表达细胞代谢调节因子和孤儿核受体雌激素相关受体γ(Esrrg,编码ERRγ)。Esrrg的表达有助于追踪PC分化的分子、细胞和超微结构阶段。scRNA-seq表明最早的Esrrg+ PC祖细胞优先表达SMAD4和SP1转录靶标以及调节酸分泌信号转导的GTP酶。随着祖细胞的成熟,依赖ERRγ的代谢转录本占主导地位。类器官和小鼠研究验证了ERRγ对PC分化的要求。我们的工作记录了干细胞在体内单系分化的过程,并建议将ERRγ作为PC相关疾病的治疗靶点。
{"title":"Metabolic regulator ERRγ governs gastric stem cell differentiation into acid-secreting parietal cells","authors":"Mahliyah Adkins-Threats, Sumimasa Arimura, Yang-Zhe Huang, Margarita Divenko, Sarah To, Heather Mao, Yongji Zeng, Jenie Y. Hwang, Joseph R. Burclaff, Shilpa Jain, Jason C. Mills","doi":"10.1016/j.stem.2024.04.016","DOIUrl":"https://doi.org/10.1016/j.stem.2024.04.016","url":null,"abstract":"<p>Parietal cells (PCs) produce gastric acid to kill pathogens and aid digestion. Dysregulated PC census is common in disease, yet how PCs differentiate is unclear. Here, we identify the PC progenitors arising from isthmal stem cells, using mouse models and human gastric cells, and show that they preferentially express cell-metabolism regulator and orphan nuclear receptor Estrogen-related receptor gamma (<em>Esrrg</em>, encoding ERRγ). <em>Esrrg</em> expression facilitated the tracking of stepwise molecular, cellular, and ultrastructural stages of PC differentiation. <em>Esrrg</em><sup><em>P2ACreERT2</em></sup> lineage tracing revealed that <em>Esrrg</em> expression commits progenitors to differentiate into mature PCs. scRNA-seq indicated the earliest <em>Esrrg</em>+ PC progenitors preferentially express SMAD4 and SP1 transcriptional targets and the GTPases regulating acid-secretion signal transduction. As progenitors matured, ERRγ-dependent metabolic transcripts predominated. Organoid and mouse studies validated the requirement of ERRγ for PC differentiation. Our work chronicles stem cell differentiation along a single lineage <em>in vivo</em> and suggests ERRγ as a therapeutic target for PC-related disorders.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":null,"pages":null},"PeriodicalIF":23.9,"publicationDate":"2024-05-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140903341","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
Activating innate immune responses repolarizes hPSC-derived CAR macrophages to improve anti-tumor activity 激活先天性免疫反应可使 hPSC 衍生的 CAR 巨噬细胞重新极化,从而提高抗肿瘤活性
IF 23.9 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-08 DOI: 10.1016/j.stem.2024.04.012
Jun Shen, Shuzhen Lyu, Yingxi Xu, Shuo Zhang, Li Li, Jinze Li, Junli Mou, Leling Xie, Kejing Tang, Wei Wen, Xuemei Peng, Ying Yang, Yu Shi, Xinjie Li, Min Wang, Xin Li, Jianxiang Wang, Tao Cheng

Generation of chimeric antigen receptor macrophages (CAR-Ms) from human pluripotent stem cells (hPSCs) offers new prospects for cancer immunotherapy but is currently challenged by low differentiation efficiency and limited function. Here, we develop a highly efficient monolayer-based system that can produce around 6,000 macrophages from a single hPSC within 3 weeks. Based on CAR structure screening, we generate hPSC-CAR-Ms with stable CAR expression and potent tumoricidal activity in vitro. To overcome the loss of tumoricidal activity of hPSC-CAR-Ms in vivo, we use interferon-γ and monophosphoryl lipid A to activate an innate immune response that repolarizes the hPSC-CAR-Ms to tumoricidal macrophages. Moreover, through combined activation of T cells by hPSC-CAR-Ms, we demonstrate that activating a collaborative innate-adaptive immune response can further enhance the anti-tumor effect of hPSC-CAR-Ms in vivo. Collectively, our study provides feasible methodologies that significantly improve the production and function of hPSC-CAR-Ms to support their translation into clinical applications.

从人类多能干细胞(hPSC)中生成嵌合抗原受体巨噬细胞(CAR-Ms)为癌症免疫疗法提供了新的前景,但目前面临着分化效率低和功能有限的挑战。在这里,我们开发了一种基于单层的高效系统,可在 3 周内从单个 hPSC 中产生约 6000 个巨噬细胞。基于 CAR 结构筛选,我们生成了具有稳定 CAR 表达和强大体外杀瘤活性的 hPSC-CAR-Ms。为了克服 hPSC-CAR-Ms 在体内失去杀瘤活性的问题,我们使用干扰素-γ 和单磷脂 A 激活先天性免疫反应,使 hPSC-CAR-Ms 重新极化为具有杀瘤活性的巨噬细胞。此外,通过 hPSC-CAR-Ms 对 T 细胞的联合激活,我们证明了激活协作性先天适应性免疫反应可进一步增强 hPSC-CAR-Ms 在体内的抗肿瘤效果。总之,我们的研究提供了可行的方法,可显著改善 hPSC-CAR-Ms 的生产和功能,支持其转化为临床应用。
{"title":"Activating innate immune responses repolarizes hPSC-derived CAR macrophages to improve anti-tumor activity","authors":"Jun Shen, Shuzhen Lyu, Yingxi Xu, Shuo Zhang, Li Li, Jinze Li, Junli Mou, Leling Xie, Kejing Tang, Wei Wen, Xuemei Peng, Ying Yang, Yu Shi, Xinjie Li, Min Wang, Xin Li, Jianxiang Wang, Tao Cheng","doi":"10.1016/j.stem.2024.04.012","DOIUrl":"https://doi.org/10.1016/j.stem.2024.04.012","url":null,"abstract":"<p>Generation of chimeric antigen receptor macrophages (CAR-Ms) from human pluripotent stem cells (hPSCs) offers new prospects for cancer immunotherapy but is currently challenged by low differentiation efficiency and limited function. Here, we develop a highly efficient monolayer-based system that can produce around 6,000 macrophages from a single hPSC within 3 weeks. Based on CAR structure screening, we generate hPSC-CAR-Ms with stable CAR expression and potent tumoricidal activity <em>in vitro</em>. To overcome the loss of tumoricidal activity of hPSC-CAR-Ms <em>in vivo</em>, we use interferon-γ and monophosphoryl lipid A to activate an innate immune response that repolarizes the hPSC-CAR-Ms to tumoricidal macrophages. Moreover, through combined activation of T cells by hPSC-CAR-Ms, we demonstrate that activating a collaborative innate-adaptive immune response can further enhance the anti-tumor effect of hPSC-CAR-Ms <em>in vivo</em>. Collectively, our study provides feasible methodologies that significantly improve the production and function of hPSC-CAR-Ms to support their translation into clinical applications.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":null,"pages":null},"PeriodicalIF":23.9,"publicationDate":"2024-05-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140890135","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
ARID1B controls transcriptional programs of axon projection in an organoid model of the human corpus callosum ARID1B控制人类胼胝体器官模型中轴突投射的转录程序
IF 23.9 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-07 DOI: 10.1016/j.stem.2024.04.014
Catarina Martins-Costa, Andrea Wiegers, Vincent A. Pham, Jaydeep Sidhaye, Balint Doleschall, Maria Novatchkova, Thomas Lendl, Marielle Piber, Angela Peer, Paul Möseneder, Marlene Stuempflen, Siu Yu A. Chow, Rainer Seidl, Daniela Prayer, Romana Höftberger, Gregor Kasprian, Yoshiho Ikeuchi, Nina S. Corsini, Jürgen A. Knoblich

Mutations in ARID1B, a member of the mSWI/SNF complex, cause severe neurodevelopmental phenotypes with elusive mechanisms in humans. The most common structural abnormality in the brain of ARID1B patients is agenesis of the corpus callosum (ACC), characterized by the absence of an interhemispheric white matter tract that connects distant cortical regions. Here, we find that neurons expressing SATB2, a determinant of callosal projection neuron (CPN) identity, show impaired maturation in ARID1B+/− neural organoids. Molecularly, a reduction in chromatin accessibility of genomic regions targeted by TCF-like, NFI-like, and ARID-like transcription factors drives the differential expression of genes required for corpus callosum (CC) development. Through an in vitro model of the CC tract, we demonstrate that this transcriptional dysregulation impairs the formation of long-range axonal projections, causing structural underconnectivity. Our study uncovers new functions of the mSWI/SNF during human corticogenesis, identifying cell-autonomous axonogenesis defects in SATB2+ neurons as a cause of ACC in ARID1B patients.

ARID1B是mSWI/SNF复合体的一个成员,它的突变会导致人类严重的神经发育表型,其机制难以捉摸。ARID1B 患者大脑中最常见的结构异常是胼胝体(ACC)缺失,其特征是大脑半球间没有连接远处皮质区域的白质束。在这里,我们发现表达 SATB2(胼胝体投射神经元(CPN)特性的决定因素)的神经元在 ARID1B+/- 神经器官组织中的成熟度受损。从分子角度看,TCF 样、NFI 样和 ARID 样转录因子靶标基因组区域染色质可及性的降低驱动了胼胝体(CC)发育所需基因的差异表达。通过一个体外的 CC 道模型,我们证明了这种转录失调会损害长距离轴突投射的形成,从而导致结构上的连接不足。我们的研究揭示了 mSWI/SNF 在人类皮质发育过程中的新功能,确定了 SATB2+ 神经元的细胞自主轴突生长缺陷是 ARID1B 患者 ACC 的病因之一。
{"title":"ARID1B controls transcriptional programs of axon projection in an organoid model of the human corpus callosum","authors":"Catarina Martins-Costa, Andrea Wiegers, Vincent A. Pham, Jaydeep Sidhaye, Balint Doleschall, Maria Novatchkova, Thomas Lendl, Marielle Piber, Angela Peer, Paul Möseneder, Marlene Stuempflen, Siu Yu A. Chow, Rainer Seidl, Daniela Prayer, Romana Höftberger, Gregor Kasprian, Yoshiho Ikeuchi, Nina S. Corsini, Jürgen A. Knoblich","doi":"10.1016/j.stem.2024.04.014","DOIUrl":"https://doi.org/10.1016/j.stem.2024.04.014","url":null,"abstract":"<p>Mutations in <em>ARID1B</em>, a member of the mSWI/SNF complex, cause severe neurodevelopmental phenotypes with elusive mechanisms in humans. The most common structural abnormality in the brain of ARID1B patients is agenesis of the corpus callosum (ACC), characterized by the absence of an interhemispheric white matter tract that connects distant cortical regions. Here, we find that neurons expressing SATB2, a determinant of callosal projection neuron (CPN) identity, show impaired maturation in <em>ARID1B</em><sup><em>+/−</em></sup> neural organoids. Molecularly, a reduction in chromatin accessibility of genomic regions targeted by TCF-like, NFI-like, and ARID-like transcription factors drives the differential expression of genes required for corpus callosum (CC) development. Through an <em>in vitro</em> model of the CC tract, we demonstrate that this transcriptional dysregulation impairs the formation of long-range axonal projections, causing structural underconnectivity. Our study uncovers new functions of the mSWI/SNF during human corticogenesis, identifying cell-autonomous axonogenesis defects in SATB2<sup>+</sup> neurons as a cause of ACC in ARID1B patients.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":null,"pages":null},"PeriodicalIF":23.9,"publicationDate":"2024-05-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140846070","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
Vascular architecture regulates mesenchymal stromal cell heterogeneity via P53-PDGF signaling in the mouse incisor 血管结构通过 P53-PDGF 信号调节小鼠门齿间充质基质细胞的异质性
IF 23.9 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-03 DOI: 10.1016/j.stem.2024.04.011
Tingwei Guo, Fei Pei, Mingyi Zhang, Takahiko Yamada, Jifan Feng, Junjun Jing, Thach-Vu Ho, Yang Chai

Mesenchymal stem cells (MSCs) reside in niches to maintain tissue homeostasis and contribute to repair and regeneration. Although the physiological functions of blood and lymphatic vasculature are well studied, their regulation of MSCs as niche components remains largely unknown. Using adult mouse incisors as a model, we uncover the role of Trp53 in regulating vascular composition through THBS2 to maintain mesenchymal tissue homeostasis. Loss of Trp53 in GLI1+ progeny increases arteries and decreases other vessel types. Platelet-derived growth factors from arteries deposit in the MSC region and interact with PDGFRA and PDGFRB. Significantly, PDGFRA+ and PDGFRB+ cells differentially contribute to defined cell lineages in the adult mouse incisor. Collectively, our results highlight Trp53’s importance in regulating the vascular niche for MSCs. They also shed light on how different arterial cells provide unique cues to regulate MSC subpopulations and maintain their heterogeneity. Furthermore, they provide mechanistic insight into MSC-vasculature crosstalk.

间充质干细胞(MSCs)驻留在龛位中,以维持组织稳态并促进修复和再生。虽然血液和淋巴管的生理功能已被深入研究,但它们对作为龛位成分的间充质干细胞的调控作用在很大程度上仍不为人所知。我们以成年小鼠门齿为模型,揭示了Trp53通过THBS2调节血管组成以维持间充质组织稳态的作用。GLI1+后代中的Trp53缺失会增加动脉,减少其他类型的血管。来自动脉的血小板衍生生长因子沉积在间充质组织区域,并与 PDGFRA 和 PDGFRB 相互作用。值得注意的是,PDGFRA+ 和 PDGFRB+ 细胞对成年小鼠门齿中确定的细胞系有不同的贡献。总之,我们的研究结果凸显了 Trp53 在调节间充质干细胞血管龛中的重要性。这些结果还揭示了不同的动脉细胞如何提供独特的线索来调节间充质干细胞亚群并保持其异质性。此外,它们还为间充质干细胞与血管的相互影响提供了机理上的启示。
{"title":"Vascular architecture regulates mesenchymal stromal cell heterogeneity via P53-PDGF signaling in the mouse incisor","authors":"Tingwei Guo, Fei Pei, Mingyi Zhang, Takahiko Yamada, Jifan Feng, Junjun Jing, Thach-Vu Ho, Yang Chai","doi":"10.1016/j.stem.2024.04.011","DOIUrl":"https://doi.org/10.1016/j.stem.2024.04.011","url":null,"abstract":"<p>Mesenchymal stem cells (MSCs) reside in niches to maintain tissue homeostasis and contribute to repair and regeneration. Although the physiological functions of blood and lymphatic vasculature are well studied, their regulation of MSCs as niche components remains largely unknown. Using adult mouse incisors as a model, we uncover the role of <em>Trp53</em> in regulating vascular composition through THBS2 to maintain mesenchymal tissue homeostasis. Loss of <em>Trp53</em> in GLI1+ progeny increases arteries and decreases other vessel types. Platelet-derived growth factors from arteries deposit in the MSC region and interact with PDGFRA and PDGFRB. Significantly, PDGFRA+ and PDGFRB+ cells differentially contribute to defined cell lineages in the adult mouse incisor. Collectively, our results highlight <em>Trp53</em>’s importance in regulating the vascular niche for MSCs. They also shed light on how different arterial cells provide unique cues to regulate MSC subpopulations and maintain their heterogeneity. Furthermore, they provide mechanistic insight into MSC-vasculature crosstalk.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":null,"pages":null},"PeriodicalIF":23.9,"publicationDate":"2024-05-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140826427","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
Addressing challenges for repairing adult spinal cord with insights from neonates 从新生儿身上汲取灵感,应对修复成人脊髓的挑战
IF 23.9 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-02 DOI: 10.1016/j.stem.2024.04.008
Mickey E. Abraham, Joel Martin, Joseph D. Ciacci

Stem cell therapy has emerged as a promising area of scientific investigation, sparking considerable interest, especially in spinal cord injury (SCI). Sun et al.1 discover that the extracellular matrix (ECM) from the neonatal spinal cord transmits biochemical signals to endogenous axons, thus promoting axonal regeneration.

干细胞疗法已成为一个前景广阔的科学研究领域,尤其在脊髓损伤(SCI)方面引起了广泛关注。Sun 等人1 发现,新生儿脊髓细胞外基质(ECM)可向内源性轴突传递生化信号,从而促进轴突再生。
{"title":"Addressing challenges for repairing adult spinal cord with insights from neonates","authors":"Mickey E. Abraham, Joel Martin, Joseph D. Ciacci","doi":"10.1016/j.stem.2024.04.008","DOIUrl":"https://doi.org/10.1016/j.stem.2024.04.008","url":null,"abstract":"<p>Stem cell therapy has emerged as a promising area of scientific investigation, sparking considerable interest, especially in spinal cord injury (SCI). Sun et al.<span><sup>1</sup></span> discover that the extracellular matrix (ECM) from the neonatal spinal cord transmits biochemical signals to endogenous axons, thus promoting axonal regeneration.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":null,"pages":null},"PeriodicalIF":23.9,"publicationDate":"2024-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140819709","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
Lumen expansion is initially driven by apical actin polymerization followed by osmotic pressure in a human epiblast model 在人类外胚层模型中,管腔扩张最初由顶端肌动蛋白聚合驱动,随后由渗透压驱动
IF 23.9 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-02 DOI: 10.1016/j.stem.2024.03.016
Dhiraj Indana, Andrei Zakharov, Youngbin Lim, Alexander R. Dunn, Nidhi Bhutani, Vivek B. Shenoy, Ovijit Chaudhuri

Post-implantation, the pluripotent epiblast in a human embryo forms a central lumen, paving the way for gastrulation. Osmotic pressure gradients are considered the drivers of lumen expansion across development, but their role in human epiblasts is unknown. Here, we study lumenogenesis in a pluripotent-stem-cell-based epiblast model using engineered hydrogels. We find that leaky junctions prevent osmotic pressure gradients in early epiblasts and, instead, forces from apical actin polymerization drive lumen expansion. Once the lumen reaches a radius of ∼12 μm, tight junctions mature, and osmotic pressure gradients develop to drive further growth. Computational modeling indicates that apical actin polymerization into a stiff network mediates initial lumen expansion and predicts a transition to pressure-driven growth in larger epiblasts to avoid buckling. Human epiblasts show transcriptional signatures consistent with these mechanisms. Thus, actin polymerization drives lumen expansion in the human epiblast and may serve as a general mechanism of early lumenogenesis.

胚胎植入后,人类胚胎的多能上胚层会形成一个中央管腔,为胃形成铺平道路。渗透压梯度被认为是整个发育过程中管腔扩张的驱动力,但它们在人类上胚层中的作用尚不清楚。在这里,我们使用工程水凝胶研究了基于多能干细胞的上胚层模型中的管腔形成。我们发现,渗漏连接阻止了早期上胚泡中的渗透压梯度,相反,顶端肌动蛋白聚合的力量推动了管腔的扩张。一旦管腔半径达到 12 μm,紧密连接就会成熟,渗透压梯度就会形成,从而推动管腔进一步生长。计算模型表明,顶端肌动蛋白聚合成一个坚硬的网络介导了最初的管腔扩张,并预测较大的外胚层会过渡到压力驱动的生长,以避免弯曲。人类外胚层显示出与这些机制一致的转录特征。因此,肌动蛋白聚合推动了人类上胚层的管腔扩张,并可能成为早期管腔形成的一般机制。
{"title":"Lumen expansion is initially driven by apical actin polymerization followed by osmotic pressure in a human epiblast model","authors":"Dhiraj Indana, Andrei Zakharov, Youngbin Lim, Alexander R. Dunn, Nidhi Bhutani, Vivek B. Shenoy, Ovijit Chaudhuri","doi":"10.1016/j.stem.2024.03.016","DOIUrl":"https://doi.org/10.1016/j.stem.2024.03.016","url":null,"abstract":"<p>Post-implantation, the pluripotent epiblast in a human embryo forms a central lumen, paving the way for gastrulation. Osmotic pressure gradients are considered the drivers of lumen expansion across development, but their role in human epiblasts is unknown. Here, we study lumenogenesis in a pluripotent-stem-cell-based epiblast model using engineered hydrogels. We find that leaky junctions prevent osmotic pressure gradients in early epiblasts and, instead, forces from apical actin polymerization drive lumen expansion. Once the lumen reaches a radius of ∼12 μm, tight junctions mature, and osmotic pressure gradients develop to drive further growth. Computational modeling indicates that apical actin polymerization into a stiff network mediates initial lumen expansion and predicts a transition to pressure-driven growth in larger epiblasts to avoid buckling. Human epiblasts show transcriptional signatures consistent with these mechanisms. Thus, actin polymerization drives lumen expansion in the human epiblast and may serve as a general mechanism of early lumenogenesis.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":null,"pages":null},"PeriodicalIF":23.9,"publicationDate":"2024-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140819887","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
Cell stem cell
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