Chamila Gunathilake, Ibrahim Soliman, Dhruba Panthi, Peter Tandler, Omar Fatani, Noman Alias Ghulamullah, Dinesh Marasinghe, Mohamed Farhath, Terrence Madhujith, Kirt Conrad, Yanhai Du and Mietek Jaroniec
The transformation from combustion-based to renewable energy technologies is of paramount importance due to the rapid depletion of fossil fuels and the dramatic increase in atmospheric CO2 levels resulting from growing global energy demands. To achieve the Paris Agreement's long-term goal of carbon neutrality by 2050, the full implementation of clean and sustainable energy sources is essential. Consequently, there is an urgent demand for zero or low-carbon fuels with high energy density that can produce electricity and heat, power vehicles, and support global trade. This review presents the global motivation to reduce carbon dioxide by utilizing hydrogen technology, which is key to meeting future energy demands. It discusses the basic properties of hydrogen and its application in both prototype and large-scale efficient technologies. Hydrogen is a clean fuel and a versatile energy carrier; when used in fuel cells or combustion devices, the final product is water vapor. Hydrogen gas production methods are reviewed across renewable and non-renewable sources, with reaction processes categorized as green, blue, grey, black, pink, and turquoise, depending on the reaction pathway and CO2 emissions management. This review covers the applications of hydrogen technology in petroleum refining, chemical and metrological production, hydrogen fuel cell electric vehicles (HFCEVs), backup power generation, and its use in transportation, space, and aeronautics. It assesses physical and material-based hydrogen storage methods, evaluating their feasibility, performance, and safety, and comparing HFCEVs with battery and gasoline vehicles from environmental and economic perspectives. Finally, the prospects and challenges associated with hydrogen production, handling, storage, transportation, and safety are also discussed.
{"title":"A comprehensive review on hydrogen production, storage, and applications","authors":"Chamila Gunathilake, Ibrahim Soliman, Dhruba Panthi, Peter Tandler, Omar Fatani, Noman Alias Ghulamullah, Dinesh Marasinghe, Mohamed Farhath, Terrence Madhujith, Kirt Conrad, Yanhai Du and Mietek Jaroniec","doi":"10.1039/D3CS00731F","DOIUrl":"10.1039/D3CS00731F","url":null,"abstract":"<p >The transformation from combustion-based to renewable energy technologies is of paramount importance due to the rapid depletion of fossil fuels and the dramatic increase in atmospheric CO<small><sub>2</sub></small> levels resulting from growing global energy demands. To achieve the Paris Agreement's long-term goal of carbon neutrality by 2050, the full implementation of clean and sustainable energy sources is essential. Consequently, there is an urgent demand for zero or low-carbon fuels with high energy density that can produce electricity and heat, power vehicles, and support global trade. This review presents the global motivation to reduce carbon dioxide by utilizing hydrogen technology, which is key to meeting future energy demands. It discusses the basic properties of hydrogen and its application in both prototype and large-scale efficient technologies. Hydrogen is a clean fuel and a versatile energy carrier; when used in fuel cells or combustion devices, the final product is water vapor. Hydrogen gas production methods are reviewed across renewable and non-renewable sources, with reaction processes categorized as green, blue, grey, black, pink, and turquoise, depending on the reaction pathway and CO<small><sub>2</sub></small> emissions management. This review covers the applications of hydrogen technology in petroleum refining, chemical and metrological production, hydrogen fuel cell electric vehicles (HFCEVs), backup power generation, and its use in transportation, space, and aeronautics. It assesses physical and material-based hydrogen storage methods, evaluating their feasibility, performance, and safety, and comparing HFCEVs with battery and gasoline vehicles from environmental and economic perspectives. Finally, the prospects and challenges associated with hydrogen production, handling, storage, transportation, and safety are also discussed.</p>","PeriodicalId":40,"journal":{"name":"Inorganic Chemistry","volume":" 22","pages":" 10900-10969"},"PeriodicalIF":40.4,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142448189","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
PROteolysis TArgeting Chimeras (PROTACs) have been considered the next blockbuster therapies. However, due to their inherent limitations, the efficacy of PROTACs is frequently impaired by limited tissue penetration and particularly insufficient cellular internalization into their action sites. Herein, based on the ultra-pH-sensitive and enzyme-sensitive nanotechnology, a type of polymer PROTAC conjugated and pH/cathepsin B sequential responsive nanoparticles (PSRNs) are deliberately designed, following the construction of the PROTAC for Cyclin-dependent kinase 4 and 6 (CDK4/6). Colorectal cancer (CRC) which hardly responds to many treatments even immune checkpoint blockades was selected as the tumor model in this study. As a result, PSRNs were found to maintain nanostructure (40 nm) in circulation and efficiently accumulated in tumors via enhanced permeation and retention effect. Then, they were dissociated into unimers (<10 nm) in response to an acidic tumor microenvironment, facilitating tumor penetration and cellular internalization. Eventually, the CDK4/6 degrading PROTACs were released intracellularly following the cleavage of cathepsin B. Importantly, PSRNs led to the enhanced degradation of target protein in vitro and in vivo. The degradation of CDK4/6 also augmented the efficacy of immune checkpoint blockades, through the upregulation of programmed cell death-ligand 1 (PD-L1) expression in cancer cells and the suppression of regulatory T cells cell proliferation in tumor microenvironment. By combination with α-PD-1, an enhanced anti-tumor outcome is well achieved in CT26 tumor model. Overall, our study verifies the significance of precise intracellular delivery of PROTACs and introduces a promising therapeutic strategy for the targeted combination treatment of CRC.
{"title":"Sequential responsive nano-PROTACs for precise intracellular delivery and enhanced degradation efficacy in colorectal cancer therapy","authors":"Liuqing Yang, Ye Yang, Jing Zhang, Minghui Li, Long Yang, Xing Wang, Meifang Chen, Hua Zhang, Bing He, Xueqing Wang, Wenbing Dai, Yiguang Wang, Qiang Zhang","doi":"10.1038/s41392-024-01983-1","DOIUrl":"https://doi.org/10.1038/s41392-024-01983-1","url":null,"abstract":"<p>PROteolysis TArgeting Chimeras (PROTACs) have been considered the next blockbuster therapies. However, due to their inherent limitations, the efficacy of PROTACs is frequently impaired by limited tissue penetration and particularly insufficient cellular internalization into their action sites. Herein, based on the ultra-pH-sensitive and enzyme-sensitive nanotechnology, a type of polymer PROTAC conjugated and pH/cathepsin B sequential responsive nanoparticles (PSRNs) are deliberately designed, following the construction of the PROTAC for Cyclin-dependent kinase 4 and 6 (CDK4/6). Colorectal cancer (CRC) which hardly responds to many treatments even immune checkpoint blockades was selected as the tumor model in this study. As a result, PSRNs were found to maintain nanostructure (40 nm) in circulation and efficiently accumulated in tumors via enhanced permeation and retention effect. Then, they were dissociated into unimers (<10 nm) in response to an acidic tumor microenvironment, facilitating tumor penetration and cellular internalization. Eventually, the CDK4/6 degrading PROTACs were released intracellularly following the cleavage of cathepsin B. Importantly, PSRNs led to the enhanced degradation of target protein in vitro and in vivo. The degradation of CDK4/6 also augmented the efficacy of immune checkpoint blockades, through the upregulation of programmed cell death-ligand 1 (PD-L1) expression in cancer cells and the suppression of regulatory T cells cell proliferation in tumor microenvironment. By combination with α-PD-1, an enhanced anti-tumor outcome is well achieved in CT26 tumor model. Overall, our study verifies the significance of precise intracellular delivery of PROTACs and introduces a promising therapeutic strategy for the targeted combination treatment of CRC.</p>","PeriodicalId":40,"journal":{"name":"Inorganic Chemistry","volume":"78 1","pages":""},"PeriodicalIF":39.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142448209","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-18DOI: 10.1038/s41392-024-01979-x
Bo Wu, Bo Zhang, Bowen Li, Haoqi Wu, Meixi Jiang
Immunotherapy has made significant strides in cancer treatment, particularly through immune checkpoint blockade (ICB), which has shown notable clinical benefits across various tumor types. Despite the transformative impact of ICB treatment in cancer therapy, only a minority of patients exhibit a positive response to it. In patients with solid tumors, those who respond well to ICB treatment typically demonstrate an active immune profile referred to as the “hot” (immune-inflamed) phenotype. On the other hand, non-responsive patients may exhibit a distinct “cold” (immune-desert) phenotype, differing from the features of “hot” tumors. Additionally, there is a more nuanced “excluded” immune phenotype, positioned between the “cold” and “hot” categories, known as the immune “excluded” type. Effective differentiation between “cold” and “hot” tumors, and understanding tumor intrinsic factors, immune characteristics, TME, and external factors are critical for predicting tumor response and treatment results. It is widely accepted that ICB therapy exerts a more profound effect on “hot” tumors, with limited efficacy against “cold” or “altered” tumors, necessitating combinations with other therapeutic modalities to enhance immune cell infiltration into tumor tissue and convert “cold” or “altered” tumors into “hot” ones. Therefore, aligning with the traits of “cold” and “hot” tumors, this review systematically delineates the respective immune characteristics, influencing factors, and extensively discusses varied treatment approaches and drug targets based on “cold” and “hot” tumors to assess clinical efficacy.
{"title":"Cold and hot tumors: from molecular mechanisms to targeted therapy","authors":"Bo Wu, Bo Zhang, Bowen Li, Haoqi Wu, Meixi Jiang","doi":"10.1038/s41392-024-01979-x","DOIUrl":"https://doi.org/10.1038/s41392-024-01979-x","url":null,"abstract":"<p>Immunotherapy has made significant strides in cancer treatment, particularly through immune checkpoint blockade (ICB), which has shown notable clinical benefits across various tumor types. Despite the transformative impact of ICB treatment in cancer therapy, only a minority of patients exhibit a positive response to it. In patients with solid tumors, those who respond well to ICB treatment typically demonstrate an active immune profile referred to as the “hot” (immune-inflamed) phenotype. On the other hand, non-responsive patients may exhibit a distinct “cold” (immune-desert) phenotype, differing from the features of “hot” tumors. Additionally, there is a more nuanced “excluded” immune phenotype, positioned between the “cold” and “hot” categories, known as the immune “excluded” type. Effective differentiation between “cold” and “hot” tumors, and understanding tumor intrinsic factors, immune characteristics, TME, and external factors are critical for predicting tumor response and treatment results. It is widely accepted that ICB therapy exerts a more profound effect on “hot” tumors, with limited efficacy against “cold” or “altered” tumors, necessitating combinations with other therapeutic modalities to enhance immune cell infiltration into tumor tissue and convert “cold” or “altered” tumors into “hot” ones. Therefore, aligning with the traits of “cold” and “hot” tumors, this review systematically delineates the respective immune characteristics, influencing factors, and extensively discusses varied treatment approaches and drug targets based on “cold” and “hot” tumors to assess clinical efficacy.</p>","PeriodicalId":40,"journal":{"name":"Inorganic Chemistry","volume":"30 1","pages":""},"PeriodicalIF":39.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142448208","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-18DOI: 10.1038/s41392-024-02000-1
Maike Hofmann, Robert Thimme, Wolfgang W. Schamel
{"title":"PD-1 and LAG-3: synergistic fostering of T cell exhaustion","authors":"Maike Hofmann, Robert Thimme, Wolfgang W. Schamel","doi":"10.1038/s41392-024-02000-1","DOIUrl":"https://doi.org/10.1038/s41392-024-02000-1","url":null,"abstract":"","PeriodicalId":40,"journal":{"name":"Inorganic Chemistry","volume":"29 1","pages":""},"PeriodicalIF":39.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142448206","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Saman Hosseinkhani, Mojdeh Amandadi, Parisa Ghanavatian, Fateme Zarein, Farangis Ataei, Maryam Nikkhah, Peter Vandenabeele
Regulated cell death is a fate of cells in (patho)physiological conditions during which extrinsic or intrinsic signals or redox equilibrium pathways following infection, cellular stress or injury are coupled to cell death modalities like apoptosis, necroptosis, pyroptosis or ferroptosis. An immediate survival response to cellular stress is often induction of autophagy, a process that deals with removal of aggregated proteins and damaged organelles by a lysosomal recycling process. These cellular processes and their regulation are crucial in several human diseases. Exploiting high-throughput assays which discriminate distinct cell death modalities and autophagy are critical to identify potential therapeutic agents that modulate these cellular responses. In the past few years, luciferase-based assays have been widely developed for assessing regulated cell death and autophagy pathways due to their simplicity, sensitivity, known chemistry, different spectral properties and high-throughput potential. Here, we review basic principles of bioluminescent reactions from a mechanistic perspective, along with their implication in vitro and in vivo for probing cell death and autophagy pathways. These include applying luciferase-, luciferin-, and ATP-based biosensors for investigating regulated cell death modalities. We discuss multiplex bioluminescence platforms which simultaneously distinguish between the various cell death phenomena and cellular stress recovery processes such as autophagy. We also highlight the recent technological achievements of bioluminescent tools for the prediction of drug effectiveness in pathways associated with regulated cell death.
调节性细胞死亡是细胞在(病理)生理条件下的一种命运,在感染、细胞应激或损伤后,外在或内在信号或氧化还原平衡途径与细胞凋亡、坏死、热凋亡或铁凋亡等细胞死亡模式相耦合。对细胞应激的直接生存反应通常是诱导自噬,这是一个通过溶酶体回收过程清除聚集蛋白质和受损细胞器的过程。这些细胞过程及其调控对多种人类疾病至关重要。利用高通量检测方法来区分不同的细胞死亡模式和自噬,对于确定调节这些细胞反应的潜在治疗药物至关重要。在过去几年中,基于荧光素酶的检测方法因其简便、灵敏、已知化学成分、不同光谱特性和高通量潜力,已被广泛用于评估受调控的细胞死亡和自噬途径。在此,我们从机理的角度回顾了生物发光反应的基本原理,以及它们在体外和体内探测细胞死亡和自噬途径的意义。其中包括应用基于荧光素酶、荧光素和 ATP 的生物传感器来研究受调控的细胞死亡模式。我们讨论了同时区分各种细胞死亡现象和细胞应激恢复过程(如自噬)的多重生物发光平台。我们还重点介绍了生物发光工具的最新技术成果,这些工具可用于预测与调节性细胞死亡相关的通路中药物的有效性。
{"title":"Harnessing luciferase chemistry in regulated cell death modalities and autophagy: overview and perspectives.","authors":"Saman Hosseinkhani, Mojdeh Amandadi, Parisa Ghanavatian, Fateme Zarein, Farangis Ataei, Maryam Nikkhah, Peter Vandenabeele","doi":"10.1039/d3cs00743j","DOIUrl":"https://doi.org/10.1039/d3cs00743j","url":null,"abstract":"<p><p>Regulated cell death is a fate of cells in (patho)physiological conditions during which extrinsic or intrinsic signals or redox equilibrium pathways following infection, cellular stress or injury are coupled to cell death modalities like apoptosis, necroptosis, pyroptosis or ferroptosis. An immediate survival response to cellular stress is often induction of autophagy, a process that deals with removal of aggregated proteins and damaged organelles by a lysosomal recycling process. These cellular processes and their regulation are crucial in several human diseases. Exploiting high-throughput assays which discriminate distinct cell death modalities and autophagy are critical to identify potential therapeutic agents that modulate these cellular responses. In the past few years, luciferase-based assays have been widely developed for assessing regulated cell death and autophagy pathways due to their simplicity, sensitivity, known chemistry, different spectral properties and high-throughput potential. Here, we review basic principles of bioluminescent reactions from a mechanistic perspective, along with their implication <i>in vitro</i> and <i>in vivo</i> for probing cell death and autophagy pathways. These include applying luciferase-, luciferin-, and ATP-based biosensors for investigating regulated cell death modalities. We discuss multiplex bioluminescence platforms which simultaneously distinguish between the various cell death phenomena and cellular stress recovery processes such as autophagy. We also highlight the recent technological achievements of bioluminescent tools for the prediction of drug effectiveness in pathways associated with regulated cell death.</p>","PeriodicalId":40,"journal":{"name":"Inorganic Chemistry","volume":" ","pages":""},"PeriodicalIF":40.4,"publicationDate":"2024-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142453607","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lilyanna Armstrong, Sarah L Chang, Nia Clements, Zoheb Hirani, Lauren B Kimberly, Keturah Odoi-Adams, Paolo Suating, Hailey F Taylor, Sara A Trauth, Adam R Urbach
The development of methodology for attaching ligand binding sites to proteins of interest has accelerated biomedical science. Such protein tags have widespread applications as well as properties that significantly limit their utility. This review describes the mechanisms and applications of supramolecular systems comprising the synthetic receptors cucurbit[7]uril (Q7) or cucurbit[8]uril (Q8) and their polypeptide ligands. Molecular recognition of peptides and proteins occurs at sites of 1-3 amino acids with high selectivity and affinity via several distinct mechanisms, which are supported by extensive thermodynamic and structural studies in aqueous media. The commercial availability, low cost, high stability, and biocompatibility of these synthetic receptors has led to the development of myriad applications. This comprehensive review compiles the molecular recognition studies and the resulting applications with the goals of providing a valuable resource to the community and inspiring the next generation of innovation.
{"title":"Molecular recognition of peptides and proteins by cucurbit[<i>n</i>]urils: systems and applications.","authors":"Lilyanna Armstrong, Sarah L Chang, Nia Clements, Zoheb Hirani, Lauren B Kimberly, Keturah Odoi-Adams, Paolo Suating, Hailey F Taylor, Sara A Trauth, Adam R Urbach","doi":"10.1039/d4cs00569d","DOIUrl":"https://doi.org/10.1039/d4cs00569d","url":null,"abstract":"<p><p>The development of methodology for attaching ligand binding sites to proteins of interest has accelerated biomedical science. Such protein tags have widespread applications as well as properties that significantly limit their utility. This review describes the mechanisms and applications of supramolecular systems comprising the synthetic receptors cucurbit[7]uril (Q7) or cucurbit[8]uril (Q8) and their polypeptide ligands. Molecular recognition of peptides and proteins occurs at sites of 1-3 amino acids with high selectivity and affinity <i>via</i> several distinct mechanisms, which are supported by extensive thermodynamic and structural studies in aqueous media. The commercial availability, low cost, high stability, and biocompatibility of these synthetic receptors has led to the development of myriad applications. This comprehensive review compiles the molecular recognition studies and the resulting applications with the goals of providing a valuable resource to the community and inspiring the next generation of innovation.</p>","PeriodicalId":40,"journal":{"name":"Inorganic Chemistry","volume":" ","pages":""},"PeriodicalIF":40.4,"publicationDate":"2024-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484504/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142453608","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-16DOI: 10.1038/s41392-024-01977-z
Lin Xie, Fei Xue, Cheng Cheng, Wenhai Sui, Jie Zhang, Linlin Meng, Yue Lu, Wenjing Xiong, Peili Bu, Feng Xu, Xiao Yu, Bo Xi, Lin Zhong, Jianmin Yang, Cheng Zhang, Yun Zhang
The pathogenesis of doxorubicin-induced cardiomyopathy remains unclear. This study was carried out to test our hypothesis that ADAM17 aggravates cardiomyocyte apoptosis induced by doxorubicin and inhibition of ADAM17 may ameliorate doxorubicin-induced cardiomyopathy. C57BL/6J mice were intraperitoneally injected with a cumulative dose of doxorubicin to induce cardiomyopathy. Cardiomyocyte-specific ADAM17-knockout (A17α-MHCKO) and ADAM17-overexpressing (AAV9-oeA17) mice were generated. In addition, RNA sequencing of the heart tissues in different mouse groups and in vitro experiments in neonatal rat cardiomyocytes (NRCMs) receiving different treatment were performed. Mouse tumor models were constructed in A17fl/fl and A17α-MHCKO mice. In addition, cardiomyocyte-specific TRAF3-knockdown and TRAF3-overexpressing mice were generated. ADAM17 expression and activity were markedly upregulated in doxorubicin-treated mouse hearts and NRCMs. A17α-MHCKO mice showed less cardiomyocyte apoptosis induced by doxorubicin than A17fl/fl mice, and cardiomyocyte ADAM17 deficiency did not affect the anti-tumor effect of doxorubicin. In contrast, AAV9-oeA17 mice exhibited markedly aggravated cardiomyocyte apoptosis relative to AAV9-oeNC mice after doxorubicin treatment. Mechanistically, doxorubicin enhanced the expression of transcription factor C/EBPβ, leading to increased expression and activity of ADAM17 in cardiomyocyte, which enhanced TNF-α shedding and upregulated the expression of TRAF3. Increased TRAF3 promoted TAK1 autophosphorylation, resulting in activated MAPKs pathway and cardiomyocyte apoptosis. ADAM17 acted as a positive regulator of cardiomyocyte apoptosis and cardiac remodeling and dysfunction induced by doxorubicin by upregulating TRAF3/TAK1/MAPKs signaling. Thus, targeting ADAM17/TRAF3/TAK1/MAPKs signaling holds a promising potential for treating doxorubicin-induced cardiotoxicity.
{"title":"Cardiomyocyte-specific knockout of ADAM17 alleviates doxorubicin-induced cardiomyopathy via inhibiting TNFα–TRAF3–TAK1–MAPK axis","authors":"Lin Xie, Fei Xue, Cheng Cheng, Wenhai Sui, Jie Zhang, Linlin Meng, Yue Lu, Wenjing Xiong, Peili Bu, Feng Xu, Xiao Yu, Bo Xi, Lin Zhong, Jianmin Yang, Cheng Zhang, Yun Zhang","doi":"10.1038/s41392-024-01977-z","DOIUrl":"https://doi.org/10.1038/s41392-024-01977-z","url":null,"abstract":"<p>The pathogenesis of doxorubicin-induced cardiomyopathy remains unclear. This study was carried out to test our hypothesis that ADAM17 aggravates cardiomyocyte apoptosis induced by doxorubicin and inhibition of ADAM17 may ameliorate doxorubicin-induced cardiomyopathy. C57BL/6J mice were intraperitoneally injected with a cumulative dose of doxorubicin to induce cardiomyopathy. Cardiomyocyte-specific ADAM17-knockout (A17<sup>α-MHCKO</sup>) and ADAM17-overexpressing (AAV9-oeA17) mice were generated. In addition, RNA sequencing of the heart tissues in different mouse groups and in vitro experiments in neonatal rat cardiomyocytes (NRCMs) receiving different treatment were performed. Mouse tumor models were constructed in A17<sup>fl/fl</sup> and A17<sup>α-MHCKO</sup> mice. In addition, cardiomyocyte-specific TRAF3-knockdown and TRAF3-overexpressing mice were generated. ADAM17 expression and activity were markedly upregulated in doxorubicin-treated mouse hearts and NRCMs. A17<sup>α-MHCKO</sup> mice showed less cardiomyocyte apoptosis induced by doxorubicin than A17<sup>fl/fl</sup> mice, and cardiomyocyte ADAM17 deficiency did not affect the anti-tumor effect of doxorubicin. In contrast, AAV9-oeA17 mice exhibited markedly aggravated cardiomyocyte apoptosis relative to AAV9-oeNC mice after doxorubicin treatment. Mechanistically, doxorubicin enhanced the expression of transcription factor C/EBPβ, leading to increased expression and activity of ADAM17 in cardiomyocyte, which enhanced TNF-α shedding and upregulated the expression of TRAF3. Increased TRAF3 promoted TAK1 autophosphorylation, resulting in activated MAPKs pathway and cardiomyocyte apoptosis. ADAM17 acted as a positive regulator of cardiomyocyte apoptosis and cardiac remodeling and dysfunction induced by doxorubicin by upregulating TRAF3/TAK1/MAPKs signaling. Thus, targeting ADAM17/TRAF3/TAK1/MAPKs signaling holds a promising potential for treating doxorubicin-induced cardiotoxicity.</p>","PeriodicalId":40,"journal":{"name":"Inorganic Chemistry","volume":"10 1","pages":""},"PeriodicalIF":39.3,"publicationDate":"2024-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142439244","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Denan Kong, Chunli Zhu, Chunyu Zhao, Jijian Liu, Ping Wang, Xiangwei Huang, Shoujun Zheng, Dezhi Zheng, Ruibin Liu and Jiadong Zhou
Two-dimensional (2D) semiconductors have attracted considerable attention for their potential in extending Moore's law and advancing next-generation electronic devices. Notably, the discovery and development of 2D ferromagnetic semiconductors (FMSs) open exciting opportunities in manipulating both charge and spin, enabling the exploration of exotic properties and the design of innovative spintronic devices. In this review, we aim to offer a comprehensive summary of emerging 2D FMSs, covering their atomic structures, physical properties, preparation methods, growth mechanisms, magnetism modulation techniques, and potential applications. We begin with a brief introduction of the atomic structures and magnetic properties of novel 2D FMSs. Next, we delve into the latest advancements in the exotic physical properties of 2D FMSs. Following that, we summarize the growth methods, associated growth mechanisms, magnetism modulation techniques and spintronic applications of 2D FMSs. Finally, we offer insights into the challenges and potential applications of 2D FMSs, which may inspire further research in developing high-density, non-volatile storage devices based on 2D FMSs.
{"title":"Emerging two-dimensional ferromagnetic semiconductors","authors":"Denan Kong, Chunli Zhu, Chunyu Zhao, Jijian Liu, Ping Wang, Xiangwei Huang, Shoujun Zheng, Dezhi Zheng, Ruibin Liu and Jiadong Zhou","doi":"10.1039/D4CS00378K","DOIUrl":"10.1039/D4CS00378K","url":null,"abstract":"<p >Two-dimensional (2D) semiconductors have attracted considerable attention for their potential in extending Moore's law and advancing next-generation electronic devices. Notably, the discovery and development of 2D ferromagnetic semiconductors (FMSs) open exciting opportunities in manipulating both charge and spin, enabling the exploration of exotic properties and the design of innovative spintronic devices. In this review, we aim to offer a comprehensive summary of emerging 2D FMSs, covering their atomic structures, physical properties, preparation methods, growth mechanisms, magnetism modulation techniques, and potential applications. We begin with a brief introduction of the atomic structures and magnetic properties of novel 2D FMSs. Next, we delve into the latest advancements in the exotic physical properties of 2D FMSs. Following that, we summarize the growth methods, associated growth mechanisms, magnetism modulation techniques and spintronic applications of 2D FMSs. Finally, we offer insights into the challenges and potential applications of 2D FMSs, which may inspire further research in developing high-density, non-volatile storage devices based on 2D FMSs.</p>","PeriodicalId":40,"journal":{"name":"Inorganic Chemistry","volume":" 22","pages":" 11228-11250"},"PeriodicalIF":40.4,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142436075","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
In a recent article published in Cell by Savage et al., the authors developed a computational workflow for integrating multi-omics data from readily available public online databases to provide novel insights into the proteogenomic landscape of several types of cancers and reveal new druggable targets for drug development or repurposing.1
{"title":"Integrate and conquer: pan-cancer proteogenomics uncovers cancer vulnerabilities and therapeutic opportunities","authors":"Debomita Chakraborty, Rossana Romero, Krishnaraj Rajalingam","doi":"10.1038/s41392-024-02009-6","DOIUrl":"https://doi.org/10.1038/s41392-024-02009-6","url":null,"abstract":"<p>In a recent article published in <i>Cell</i> by Savage et al., the authors developed a computational workflow for integrating multi-omics data from readily available public online databases to provide novel insights into the proteogenomic landscape of several types of cancers and reveal new druggable targets for drug development or repurposing.<sup>1</sup></p>","PeriodicalId":40,"journal":{"name":"Inorganic Chemistry","volume":"229 1","pages":""},"PeriodicalIF":39.3,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142435968","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}