Xinyu He, Jinghan Yang, Xue Chen, Jiajia Chen, Haicong Zhao, Juan Liu, Fengpei Du, Peiyi Wang
Bacterial plant diseases, worsened by biofilm-mediated resistance, are increasingly threatening global food security. Numerous attempts have been made to develop agrochemicals that inhibit biofilms, however, their ineffective foliar deposition and difficulty in removing mature biofilms remain major challenges. Herein, multifunctional three-component supramolecular nano-biscuits (NI6R@CB[7]@β-CD) are successfully engineered via ordered self-assembly between two macrocycles [cucurbit[7]uril (CB[7]), β-cyclodextrin (β-CD)] and (R)-2-naphthol-based bis-imidazolium bromide salt (NI6R). This macrocycles-involved bactericidal material combines many advantages. 1) Alleviate the off-target movement of droplets on hydrophobic blade surfaces. 2) Enhance the biofilm-disrupting ability. At a low-dose of 4.44 µg mL-1, the inhibition rate of biofilm formation reached 78.3%. At 35.5 µg mL-1, the potency to remove mature biofilms reached 77.6%. 3) Efficiently hinder bacterial reproduction, swimming, extracellular polysaccharide production, extracellular enzyme secretion, and virulence to plants. These superior characteristics are undoubtedly transmitted to the in vivo control effect. At 200 µg mL-1, this smart material exhibits superior control efficiencies of 49.6%/65.0%/85.4% against three kinds of bacterial diseases (rice leaf blight, citrus canker, and kiwifruit canker), respectively, surpassing the commercial bactericide-thiodiazole-copper-20%SC (33.6%/41.5%/43.2%) and NI6R (40.3%/51.2%/71.2%). Furthermore, NI6R@CB[7]@β-CD is biosafe to non-target organisms. This study is instructive for constructing multifunctional agrochemicals in sustainable crop protection.
{"title":"Fabrication of Multifunctional Three-Component Supramolecular Nano-Biscuits via Two Macrocycles-Involved Self-Assembly for Rice, Citrus and Kiwifruit Protections.","authors":"Xinyu He, Jinghan Yang, Xue Chen, Jiajia Chen, Haicong Zhao, Juan Liu, Fengpei Du, Peiyi Wang","doi":"10.1002/advs.202413826","DOIUrl":"https://doi.org/10.1002/advs.202413826","url":null,"abstract":"<p><p>Bacterial plant diseases, worsened by biofilm-mediated resistance, are increasingly threatening global food security. Numerous attempts have been made to develop agrochemicals that inhibit biofilms, however, their ineffective foliar deposition and difficulty in removing mature biofilms remain major challenges. Herein, multifunctional three-component supramolecular nano-biscuits (NI6R@CB[7]@β-CD) are successfully engineered via ordered self-assembly between two macrocycles [cucurbit[7]uril (CB[7]), β-cyclodextrin (β-CD)] and (R)-2-naphthol-based bis-imidazolium bromide salt (NI6R). This macrocycles-involved bactericidal material combines many advantages. 1) Alleviate the off-target movement of droplets on hydrophobic blade surfaces. 2) Enhance the biofilm-disrupting ability. At a low-dose of 4.44 µg mL<sup>-1</sup>, the inhibition rate of biofilm formation reached 78.3%. At 35.5 µg mL<sup>-1</sup>, the potency to remove mature biofilms reached 77.6%. 3) Efficiently hinder bacterial reproduction, swimming, extracellular polysaccharide production, extracellular enzyme secretion, and virulence to plants. These superior characteristics are undoubtedly transmitted to the in vivo control effect. At 200 µg mL<sup>-1</sup>, this smart material exhibits superior control efficiencies of 49.6%/65.0%/85.4% against three kinds of bacterial diseases (rice leaf blight, citrus canker, and kiwifruit canker), respectively, surpassing the commercial bactericide-thiodiazole-copper-20%SC (33.6%/41.5%/43.2%) and NI6R (40.3%/51.2%/71.2%). Furthermore, NI6R@CB[7]@β-CD is biosafe to non-target organisms. This study is instructive for constructing multifunctional agrochemicals in sustainable crop protection.</p>","PeriodicalId":117,"journal":{"name":"Advanced Science","volume":" ","pages":"e2413826"},"PeriodicalIF":14.3,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031497","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Liyin Wang, Min Gu, Xiaoli Zhang, Tingting Kong, Jun Liao, Dan Zhang, Jingwu Li
Glioblastoma multiforme (GBM) is a highly aggressive and malignant brain tumor originating from glial cells, characterized by high recurrence rates and poor patient prognosis. The heterogeneity and complex biology of GBM, coupled with the protective nature of the blood-brain barrier (BBB), significantly limit the efficacy of traditional therapies. The rapid development of nanoenzyme technology presents a promising therapeutic paradigm for the rational and targeted treatment of GBM. In this review, the underlying mechanisms of GBM pathogenesis are comprehensively discussed, emphasizing the impact of the BBB on treatment strategies. Recent advances in nanoenzyme-based approaches for GBM therapy are explored, highlighting how these nanoenzymes enhance various treatment modalities through their multifunctional capabilities and potential for precise drug delivery. Finally, the challenges and therapeutic prospects of translating nanoenzymes from laboratory research to clinical application, including issues of stability, targeting efficiency, safety, and regulatory hurdles are critically analyzed. By providing a thorough understanding of both the opportunities and obstacles associated with nanoenzyme-based therapies, future research directions are aimed to be informed and contribute to the development of more effective treatments for GBM.
{"title":"Recent Advances in Nanoenzymes Based Therapies for Glioblastoma: Overcoming Barriers and Enhancing Targeted Treatment.","authors":"Liyin Wang, Min Gu, Xiaoli Zhang, Tingting Kong, Jun Liao, Dan Zhang, Jingwu Li","doi":"10.1002/advs.202413367","DOIUrl":"https://doi.org/10.1002/advs.202413367","url":null,"abstract":"<p><p>Glioblastoma multiforme (GBM) is a highly aggressive and malignant brain tumor originating from glial cells, characterized by high recurrence rates and poor patient prognosis. The heterogeneity and complex biology of GBM, coupled with the protective nature of the blood-brain barrier (BBB), significantly limit the efficacy of traditional therapies. The rapid development of nanoenzyme technology presents a promising therapeutic paradigm for the rational and targeted treatment of GBM. In this review, the underlying mechanisms of GBM pathogenesis are comprehensively discussed, emphasizing the impact of the BBB on treatment strategies. Recent advances in nanoenzyme-based approaches for GBM therapy are explored, highlighting how these nanoenzymes enhance various treatment modalities through their multifunctional capabilities and potential for precise drug delivery. Finally, the challenges and therapeutic prospects of translating nanoenzymes from laboratory research to clinical application, including issues of stability, targeting efficiency, safety, and regulatory hurdles are critically analyzed. By providing a thorough understanding of both the opportunities and obstacles associated with nanoenzyme-based therapies, future research directions are aimed to be informed and contribute to the development of more effective treatments for GBM.</p>","PeriodicalId":117,"journal":{"name":"Advanced Science","volume":" ","pages":"e2413367"},"PeriodicalIF":14.3,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031541","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xizhi Yu, Junjie Qian, Limin Ding, Caixu Pan, Xi Liu, Qinchuan Wu, Shuai Wang, Jianpeng Liu, Mingge Shang, Rong Su, Danjing Guo, Haiyang Xie, Shengyong Yin, Lin Zhou, Shusen Zheng
Tumor-associated macrophages (TAMs) are commonly considered accomplices in tumorigenesis and tumor development. However, the precise mechanism by which tumor cells prompt TAMs to aid in evading immune surveillance remains to be further investigated. Here, it is elucidated that tumor-secreted galectin-1 (Gal1) conferred immunosuppressive properties to TAMs. Specifically, patient specimens and a public database is first used to analyze the clinical relevance of Gal1 in hepatocellular carcinoma (HCC). Then, it is demonstrated that TAMs functioned as a critical mediator in the Gal1-induced progression of HCC and the establishment of an immunosuppressive tumor microenvironment. Furthermore, RNA-sequencing determined that Gal1 promoted the upregulation of chemokine (C-C motif) ligand 20 (CCL20) in TAMs via activating the PI3K/AKT/NF-κB pathway. Employing an anti-CCL20 neutralizing antibody and Foxp3DTR mice, it is demonstrated that CCR6+Foxp3+ regulatory T cells (Tregs) recruited by Gal1-induced TAMs contributed to reduced infiltration and dysfunctional state of CD8+ T cells, subsequently facilitating tumor progression. Targeting Gal1 dampened the secretion of CCL20 and inhibits the recruitment of Tregs, thereby activating anti-tumor immunity and ameliorating anti-PD-1 resistance. Together, this findings revealed that Gal1-induced TAMs recruited Tregs through the CCL20-CCR6 axis. Inhibition of Gal1 improves the effectiveness of anti-PD1 therapy, shedding important new light on the combination immunotherapy of HCC.
{"title":"Galectin-1-Induced Tumor Associated Macrophages Repress Antitumor Immunity in Hepatocellular Carcinoma Through Recruitment of Tregs.","authors":"Xizhi Yu, Junjie Qian, Limin Ding, Caixu Pan, Xi Liu, Qinchuan Wu, Shuai Wang, Jianpeng Liu, Mingge Shang, Rong Su, Danjing Guo, Haiyang Xie, Shengyong Yin, Lin Zhou, Shusen Zheng","doi":"10.1002/advs.202408788","DOIUrl":"https://doi.org/10.1002/advs.202408788","url":null,"abstract":"<p><p>Tumor-associated macrophages (TAMs) are commonly considered accomplices in tumorigenesis and tumor development. However, the precise mechanism by which tumor cells prompt TAMs to aid in evading immune surveillance remains to be further investigated. Here, it is elucidated that tumor-secreted galectin-1 (Gal1) conferred immunosuppressive properties to TAMs. Specifically, patient specimens and a public database is first used to analyze the clinical relevance of Gal1 in hepatocellular carcinoma (HCC). Then, it is demonstrated that TAMs functioned as a critical mediator in the Gal1-induced progression of HCC and the establishment of an immunosuppressive tumor microenvironment. Furthermore, RNA-sequencing determined that Gal1 promoted the upregulation of chemokine (C-C motif) ligand 20 (CCL20) in TAMs via activating the PI3K/AKT/NF-κB pathway. Employing an anti-CCL20 neutralizing antibody and Foxp3DTR mice, it is demonstrated that CCR6<sup>+</sup>Foxp3<sup>+</sup> regulatory T cells (Tregs) recruited by Gal1-induced TAMs contributed to reduced infiltration and dysfunctional state of CD8<sup>+</sup> T cells, subsequently facilitating tumor progression. Targeting Gal1 dampened the secretion of CCL20 and inhibits the recruitment of Tregs, thereby activating anti-tumor immunity and ameliorating anti-PD-1 resistance. Together, this findings revealed that Gal1-induced TAMs recruited Tregs through the CCL20-CCR6 axis. Inhibition of Gal1 improves the effectiveness of anti-PD1 therapy, shedding important new light on the combination immunotherapy of HCC.</p>","PeriodicalId":117,"journal":{"name":"Advanced Science","volume":" ","pages":"e2408788"},"PeriodicalIF":14.3,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031501","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yameng Fan, Xin Wang, Guyue Bo, Xun Xu, Khay Wai See, Bernt Johannessen, Wei Kong Pang
Rechargeable batteries are central to modern energy storage systems, from portable electronics to electric vehicles. The cathode material, a critical component, largely dictates a battery's energy density, capacity, and overall performance. This review focuses on the application of operando X-ray absorption spectroscopy (XAS) to study cathode materials in Li-ion, Na-ion, Li-S, and Na-S batteries. Operando XAS provides real-time insights into the local electronic structure, oxidation states, and coordination environments, which are crucial for understanding complex electrochemical processes, such as redox reactions, phase transitions, and structural degradation. The review highlights the strengths of hard and soft XAS techniques in probing transition metal (TM) and anionic redox processes, particularly in layered oxide cathodes, where reversible oxygen redox and TM behavior are pivotal. The role of operando XAS is also explored in analyzing conversion-type S-based cathodes, where it helps unravel the intricate reaction mechanisms. Furthermore, the review addresses the challenges of in situ cell design for operando XAS, especially under ultrahigh vacuum conditions for soft XAS. By discussing recent advancements and future directions, this review underscores the critical role of operando XAS in driving innovation and improving the design and performance of next-generation battery technologies.
{"title":"Operando Synchrotron X-Ray Absorption Spectroscopy: A Key Tool for Cathode Material Studies in Next-Generation Batteries.","authors":"Yameng Fan, Xin Wang, Guyue Bo, Xun Xu, Khay Wai See, Bernt Johannessen, Wei Kong Pang","doi":"10.1002/advs.202414480","DOIUrl":"https://doi.org/10.1002/advs.202414480","url":null,"abstract":"<p><p>Rechargeable batteries are central to modern energy storage systems, from portable electronics to electric vehicles. The cathode material, a critical component, largely dictates a battery's energy density, capacity, and overall performance. This review focuses on the application of operando X-ray absorption spectroscopy (XAS) to study cathode materials in Li-ion, Na-ion, Li-S, and Na-S batteries. Operando XAS provides real-time insights into the local electronic structure, oxidation states, and coordination environments, which are crucial for understanding complex electrochemical processes, such as redox reactions, phase transitions, and structural degradation. The review highlights the strengths of hard and soft XAS techniques in probing transition metal (TM) and anionic redox processes, particularly in layered oxide cathodes, where reversible oxygen redox and TM behavior are pivotal. The role of operando XAS is also explored in analyzing conversion-type S-based cathodes, where it helps unravel the intricate reaction mechanisms. Furthermore, the review addresses the challenges of in situ cell design for operando XAS, especially under ultrahigh vacuum conditions for soft XAS. By discussing recent advancements and future directions, this review underscores the critical role of operando XAS in driving innovation and improving the design and performance of next-generation battery technologies.</p>","PeriodicalId":117,"journal":{"name":"Advanced Science","volume":" ","pages":"e2414480"},"PeriodicalIF":14.3,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031526","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yan Liang, Xiang-Yu Xiong, Guo-Wang Lin, Xiaomeng Bai, Fugui Li, Josephine Mun-Yee Ko, Yun-He Zhou, An-Yi Xu, Shu-Qiang Liu, Shuai He, Pan-Pan Wei, Qiu-Yan Chen, Lin-Quan Tang, Vivien Ya-Fan Wang, Hai-Qiang Mai, Chun-Ling Luo, Yanni Zeng, Maria Li Lung, Mingfang Ji, Jin-Xin Bei
Nasopharyngeal carcinoma (NPC) is an Asia-prevalent malignancy, yet its genetic underpinnings remain incompletely understood. Here, a transcriptome-wide association study (TWAS) is conducted on NPC, leveraging gene expression prediction models based on epithelial tissues and genome-wide association study (GWAS) summary statistics from 1577 NPC cases and 6359 controls of southern Chinese descent. The TWAS identifies VAMP8 on chromosome 2p11.2 as a novel susceptibility gene for NPC. Further fine-mapping analyses pinpoint rs1058588, located within VAMP8, as a causal variant through eQTL colocalization, and GWAS analyses across multiple cohorts, achieving GWAS significance (OR = 1.18, P = 3.09 × 10-10). Functional assays demonstrate that VAMP8 exerts a tumorigenic role in NPC, enhancing cell proliferation, migration, and tumor growth. Mechanically, it is uncovered that rs1058588 modulates VAMP8 expression by altering its binding affinity to miR-185. Furthermore, the results show that VAMP8 interacts with DHX9 to facilitate the nuclear recruitment of p65, activating the NF-κB pathway. Collectively, the findings shed light on the genetic predisposition to NPC and underscore the critical role of the functional axis involving miR-185, VAMP8, DHX9, and the NF-κB pathway in NPC pathogenesis.
{"title":"Integrative Transcriptome-Wide Association Study With Expression Quantitative Trait Loci Colocalization Identifies a Causal VAMP8 Variant for Nasopharyngeal Carcinoma Susceptibility.","authors":"Yan Liang, Xiang-Yu Xiong, Guo-Wang Lin, Xiaomeng Bai, Fugui Li, Josephine Mun-Yee Ko, Yun-He Zhou, An-Yi Xu, Shu-Qiang Liu, Shuai He, Pan-Pan Wei, Qiu-Yan Chen, Lin-Quan Tang, Vivien Ya-Fan Wang, Hai-Qiang Mai, Chun-Ling Luo, Yanni Zeng, Maria Li Lung, Mingfang Ji, Jin-Xin Bei","doi":"10.1002/advs.202412580","DOIUrl":"https://doi.org/10.1002/advs.202412580","url":null,"abstract":"<p><p>Nasopharyngeal carcinoma (NPC) is an Asia-prevalent malignancy, yet its genetic underpinnings remain incompletely understood. Here, a transcriptome-wide association study (TWAS) is conducted on NPC, leveraging gene expression prediction models based on epithelial tissues and genome-wide association study (GWAS) summary statistics from 1577 NPC cases and 6359 controls of southern Chinese descent. The TWAS identifies VAMP8 on chromosome 2p11.2 as a novel susceptibility gene for NPC. Further fine-mapping analyses pinpoint rs1058588, located within VAMP8, as a causal variant through eQTL colocalization, and GWAS analyses across multiple cohorts, achieving GWAS significance (OR = 1.18, P = 3.09 × 10<sup>-10</sup>). Functional assays demonstrate that VAMP8 exerts a tumorigenic role in NPC, enhancing cell proliferation, migration, and tumor growth. Mechanically, it is uncovered that rs1058588 modulates VAMP8 expression by altering its binding affinity to miR-185. Furthermore, the results show that VAMP8 interacts with DHX9 to facilitate the nuclear recruitment of p65, activating the NF-κB pathway. Collectively, the findings shed light on the genetic predisposition to NPC and underscore the critical role of the functional axis involving miR-185, VAMP8, DHX9, and the NF-κB pathway in NPC pathogenesis.</p>","PeriodicalId":117,"journal":{"name":"Advanced Science","volume":" ","pages":"e2412580"},"PeriodicalIF":14.3,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031521","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Baichuan Xu, Xianpeng Ye, Kangfu Sun, Liang Chen, Zhaoyang Wen, Qigang Lan, Jun Chen, Mo Chen, Mingqiang Shen, Song Wang, Yang Xu, Xi Zhang, Jinghong Zhao, Junping Wang, Shilei Chen
Platelets play crucial roles in multiple pathophysiological processes after energy-dependent activation. It is puzzling how such a small cellular debris has abundant energy supply. In this study, it is shown that insulin-regulated aminopeptidase (IRAP), a type II transmembrane protein, is a key regulator for platelet activation by promoting energy regeneration during septic thrombosis. Through interaction with certain endosome membrane proteins, IRAP can not only promote granule release, but also facilitate lysosomal degradation of theoretically discarded ribosomes in an mTORC1- and S-acylation-dependent manner in activated platelets. Plentiful amino acids obtained from IRAP-mediated ribophagy are recruited to aerobic glycolysis and then promote energy metabolism reprogramming, thereby producing abundant energy for platelet life extension and prolonged activation. Consequently, targeted blocking IRAP can dramatically alleviate platelet hyperactivation and septic thrombosis.
{"title":"IRAP Drives Ribosomal Degradation to Refuel Energy for Platelet Activation during Septic Thrombosis.","authors":"Baichuan Xu, Xianpeng Ye, Kangfu Sun, Liang Chen, Zhaoyang Wen, Qigang Lan, Jun Chen, Mo Chen, Mingqiang Shen, Song Wang, Yang Xu, Xi Zhang, Jinghong Zhao, Junping Wang, Shilei Chen","doi":"10.1002/advs.202411914","DOIUrl":"https://doi.org/10.1002/advs.202411914","url":null,"abstract":"<p><p>Platelets play crucial roles in multiple pathophysiological processes after energy-dependent activation. It is puzzling how such a small cellular debris has abundant energy supply. In this study, it is shown that insulin-regulated aminopeptidase (IRAP), a type II transmembrane protein, is a key regulator for platelet activation by promoting energy regeneration during septic thrombosis. Through interaction with certain endosome membrane proteins, IRAP can not only promote granule release, but also facilitate lysosomal degradation of theoretically discarded ribosomes in an mTORC1- and S-acylation-dependent manner in activated platelets. Plentiful amino acids obtained from IRAP-mediated ribophagy are recruited to aerobic glycolysis and then promote energy metabolism reprogramming, thereby producing abundant energy for platelet life extension and prolonged activation. Consequently, targeted blocking IRAP can dramatically alleviate platelet hyperactivation and septic thrombosis.</p>","PeriodicalId":117,"journal":{"name":"Advanced Science","volume":" ","pages":"e2411914"},"PeriodicalIF":14.3,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031522","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gyeong Hoon Yi, Jiwoo Choi, Hyeongyun Song, Olivia Miano, Jaewoong Choi, Kihoon Bang, Byungju Lee, Seok Su Sohn, David Buttler, Anna Hiszpanski, Sang Soo Han, Donghun Kim
Efficiently extracting data from tables in the scientific literature is pivotal for building large-scale databases. However, the tables reported in materials science papers exist in highly diverse forms; thus, rule-based extractions are an ineffective approach. To overcome this challenge, the study presents MaTableGPT, which is a GPT-based table data extractor from the materials science literature. MaTableGPT features key strategies of table data representation and table splitting for better GPT comprehension and filtering hallucinated information through follow-up questions. When applied to a vast volume of water splitting catalysis literature, MaTableGPT achieves an extraction accuracy (total F1 score) of up to 96.8%. Through comprehensive evaluations of the GPT usage cost, labeling cost, and extraction accuracy for the learning methods of zero-shot, few-shot, and fine-tuning, the study presents a Pareto-front mapping where the few-shot learning method is found to be the most balanced solution owing to both its high extraction accuracy (total F1 score >95%) and low cost (GPT usage cost of 5.97 US dollars and labeling cost of 10 I/O paired examples). The statistical analyses conducted on the database generated by MaTableGPT revealed valuable insights into the distribution of the overpotential and elemental utilization across the reported catalysts in the water splitting literature.
{"title":"MaTableGPT: GPT-Based Table Data Extractor from Materials Science Literature.","authors":"Gyeong Hoon Yi, Jiwoo Choi, Hyeongyun Song, Olivia Miano, Jaewoong Choi, Kihoon Bang, Byungju Lee, Seok Su Sohn, David Buttler, Anna Hiszpanski, Sang Soo Han, Donghun Kim","doi":"10.1002/advs.202408221","DOIUrl":"https://doi.org/10.1002/advs.202408221","url":null,"abstract":"<p><p>Efficiently extracting data from tables in the scientific literature is pivotal for building large-scale databases. However, the tables reported in materials science papers exist in highly diverse forms; thus, rule-based extractions are an ineffective approach. To overcome this challenge, the study presents MaTableGPT, which is a GPT-based table data extractor from the materials science literature. MaTableGPT features key strategies of table data representation and table splitting for better GPT comprehension and filtering hallucinated information through follow-up questions. When applied to a vast volume of water splitting catalysis literature, MaTableGPT achieves an extraction accuracy (total F1 score) of up to 96.8%. Through comprehensive evaluations of the GPT usage cost, labeling cost, and extraction accuracy for the learning methods of zero-shot, few-shot, and fine-tuning, the study presents a Pareto-front mapping where the few-shot learning method is found to be the most balanced solution owing to both its high extraction accuracy (total F1 score >95%) and low cost (GPT usage cost of 5.97 US dollars and labeling cost of 10 I/O paired examples). The statistical analyses conducted on the database generated by MaTableGPT revealed valuable insights into the distribution of the overpotential and elemental utilization across the reported catalysts in the water splitting literature.</p>","PeriodicalId":117,"journal":{"name":"Advanced Science","volume":" ","pages":"e2408221"},"PeriodicalIF":14.3,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031524","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Tong-Yu Rui, He-Zhou Huang, Kai Zheng, Hong-Wei Fan, Juan Zhang, Zi-Yuan Guo, Heng-Ye Man, Nadezhda Brazhe, Alexey Semyanov, You-Ming Lu, Dan Liu, Ling-Qiang Zhu
Dietary high salt intake is increasingly recognized as a risk factor for cognitive decline and dementia, including Alzheimer's disease (AD). Recent studies have identified a population of disease-associated astrocytes (DAA)-like astrocytes closely linked to amyloid deposition and tau pathology in an AD mouse model. However, the presence and role of these astrocytes in high-salt diet (HSD) models remain unexplored. In this study, it is demonstrated that HSD significantly induces enhanced reactivity of DAA-like astrocytes in the hippocampal CA3 region of mice, with this reactivity being critically dependent on neuronal tau pathology. Neuronal tau pathology activates adenosine A1R signaling, exacerbating tau pathology by inhibiting the Cers1 pathway, which sustains astrocyte reactivity. Additionally, neurons burdened with tau pathology promote astrocyte reactivity via releasing Proteins Associated with Promoting DAA-like Astrocyte Reactivity (PAPD), with Lcn2 playing a pivotal role. Knockout of Lcn2 or its receptor 24p3R significantly mitigates HSD-induced DAA reactivity and neuroinflammation. These findings suggest a vicious cycle between tau pathology and A1R signaling, driving DAA-like astrocyte reactivity. Targeting the Tau-A1R axis may provide a novel therapeutic strategy for reducing HSD-induced neuroinflammation and cognitive deficits.
{"title":"Tau Pathology Drives Disease-Associated Astrocyte Reactivity in Salt-Induced Neurodegeneration.","authors":"Tong-Yu Rui, He-Zhou Huang, Kai Zheng, Hong-Wei Fan, Juan Zhang, Zi-Yuan Guo, Heng-Ye Man, Nadezhda Brazhe, Alexey Semyanov, You-Ming Lu, Dan Liu, Ling-Qiang Zhu","doi":"10.1002/advs.202410799","DOIUrl":"https://doi.org/10.1002/advs.202410799","url":null,"abstract":"<p><p>Dietary high salt intake is increasingly recognized as a risk factor for cognitive decline and dementia, including Alzheimer's disease (AD). Recent studies have identified a population of disease-associated astrocytes (DAA)-like astrocytes closely linked to amyloid deposition and tau pathology in an AD mouse model. However, the presence and role of these astrocytes in high-salt diet (HSD) models remain unexplored. In this study, it is demonstrated that HSD significantly induces enhanced reactivity of DAA-like astrocytes in the hippocampal CA3 region of mice, with this reactivity being critically dependent on neuronal tau pathology. Neuronal tau pathology activates adenosine A1R signaling, exacerbating tau pathology by inhibiting the Cers1 pathway, which sustains astrocyte reactivity. Additionally, neurons burdened with tau pathology promote astrocyte reactivity via releasing Proteins Associated with Promoting DAA-like Astrocyte Reactivity (PAPD), with Lcn2 playing a pivotal role. Knockout of Lcn2 or its receptor 24p3R significantly mitigates HSD-induced DAA reactivity and neuroinflammation. These findings suggest a vicious cycle between tau pathology and A1R signaling, driving DAA-like astrocyte reactivity. Targeting the Tau-A1R axis may provide a novel therapeutic strategy for reducing HSD-induced neuroinflammation and cognitive deficits.</p>","PeriodicalId":117,"journal":{"name":"Advanced Science","volume":" ","pages":"e2410799"},"PeriodicalIF":14.3,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031557","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yisong Yao, Zihui Zhao, He Li, Yongfeng Zhao, H P Zhang, Masaki Sano
The past decade witnessed a surge in discoveries where biological systems, such as bacteria or living cells, inherently portray active polar or nematic behavior: they prefer to align with each other and form local order during migration. Although the underlying mechanisms remain unclear, utilizing their physical properties to achieve controllable cell-layer transport will be of fundamental importance. In this study, the ratchet effect is harnessed to control the collective motion of neural progenitor cells (NPCs) in vitro. NPCs travel back-and-forth and do not specify head or tail, and therefore regarded as nematics alike liquid crystals. Ratchet and splay-shaped confinements are crafted to modulate collective cell dynamics in dense environments, while jamming is not explicitly spotted. The adaptation of an agent-based simulation further revealed how the ratchet's asymmetry and active forces from nematic order synergistically reinforce the directional cell flow. These findings provide insights into topotaxis in cell populations when restricted to crowded 2D ratchets and the mechanisms that regulate collective behavior of the cells.
{"title":"Active Nematics Reinforce the Ratchet Flow in Dense Environments Without Jamming.","authors":"Yisong Yao, Zihui Zhao, He Li, Yongfeng Zhao, H P Zhang, Masaki Sano","doi":"10.1002/advs.202412750","DOIUrl":"https://doi.org/10.1002/advs.202412750","url":null,"abstract":"<p><p>The past decade witnessed a surge in discoveries where biological systems, such as bacteria or living cells, inherently portray active polar or nematic behavior: they prefer to align with each other and form local order during migration. Although the underlying mechanisms remain unclear, utilizing their physical properties to achieve controllable cell-layer transport will be of fundamental importance. In this study, the ratchet effect is harnessed to control the collective motion of neural progenitor cells (NPCs) in vitro. NPCs travel back-and-forth and do not specify head or tail, and therefore regarded as nematics alike liquid crystals. Ratchet and splay-shaped confinements are crafted to modulate collective cell dynamics in dense environments, while jamming is not explicitly spotted. The adaptation of an agent-based simulation further revealed how the ratchet's asymmetry and active forces from nematic order synergistically reinforce the directional cell flow. These findings provide insights into topotaxis in cell populations when restricted to crowded 2D ratchets and the mechanisms that regulate collective behavior of the cells.</p>","PeriodicalId":117,"journal":{"name":"Advanced Science","volume":" ","pages":"e2412750"},"PeriodicalIF":14.3,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143021367","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pedro Vicente, Lara R Inocêncio, Asier Ullate-Agote, Ana F Louro, João Jacinto, Beatriz Gamelas, Olalla Iglesias-García, Patxi San Martin-Uriz, Paula Aguirre-Ruiz, Gonzalo R Ríos-Muñoz, María Eugenia Fernández-Santos, Alain van Mil, Joost P G Sluijter, Felipe Prósper, Manuel M Mazo Vega, Paula M Alves, Margarida Serra
Generation of upscaled quantities of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM), for therapeutic or testing applications, is both expensive and time-consuming. Herein, a scalable bioprocess for hiPSC-CM expansion in stirred-tank bioreactors (STB) is developed. By combining the continuous activation of the Wnt pathway, through perfusion of CHIR99021, within a mild hypoxia environment, the expansion of hiPSC-CM as aggregates is maximized, reaching 4 billion of pure hiPSC-CM in 2L STB. In particular, the importance of i) controlling the dissolved oxygen at 10% O2 to reduce reactive oxygen species production and upregulate genes involved in cell proliferation, resulting in higher expansion rates (tenfold) compared to normoxic conditions, and ii) maintaining constant power input per volume as a scale-up criteria is demonstrated. After expansion, hiPSC-CM further mature in culture, revealing more mature transcriptional signatures, higher sarcomere alignment and improved calcium handling. This new bioprocess opens the door to time- and cost-effective generation of hiPSC-CM.
{"title":"Billion-Scale Expansion of Functional hiPSC-Derived Cardiomyocytes in Bioreactors Through Oxygen Control and Continuous Wnt Activation.","authors":"Pedro Vicente, Lara R Inocêncio, Asier Ullate-Agote, Ana F Louro, João Jacinto, Beatriz Gamelas, Olalla Iglesias-García, Patxi San Martin-Uriz, Paula Aguirre-Ruiz, Gonzalo R Ríos-Muñoz, María Eugenia Fernández-Santos, Alain van Mil, Joost P G Sluijter, Felipe Prósper, Manuel M Mazo Vega, Paula M Alves, Margarida Serra","doi":"10.1002/advs.202410510","DOIUrl":"https://doi.org/10.1002/advs.202410510","url":null,"abstract":"<p><p>Generation of upscaled quantities of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM), for therapeutic or testing applications, is both expensive and time-consuming. Herein, a scalable bioprocess for hiPSC-CM expansion in stirred-tank bioreactors (STB) is developed. By combining the continuous activation of the Wnt pathway, through perfusion of CHIR99021, within a mild hypoxia environment, the expansion of hiPSC-CM as aggregates is maximized, reaching 4 billion of pure hiPSC-CM in 2L STB. In particular, the importance of i) controlling the dissolved oxygen at 10% O<sub>2</sub> to reduce reactive oxygen species production and upregulate genes involved in cell proliferation, resulting in higher expansion rates (tenfold) compared to normoxic conditions, and ii) maintaining constant power input per volume as a scale-up criteria is demonstrated. After expansion, hiPSC-CM further mature in culture, revealing more mature transcriptional signatures, higher sarcomere alignment and improved calcium handling. This new bioprocess opens the door to time- and cost-effective generation of hiPSC-CM.</p>","PeriodicalId":117,"journal":{"name":"Advanced Science","volume":" ","pages":"e2410510"},"PeriodicalIF":14.3,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143021371","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}