首页 > 最新文献

ACS Chemical Biology最新文献

英文 中文
Probing the Signal Transduction Mechanism of the Light-Activated Adenylate Cyclase OaPAC Using Unnatural Amino Acid Mutagenesis
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-22 DOI: 10.1021/acschembio.4c0062710.1021/acschembio.4c00627
Samruddhi S. Jewlikar, Jinnette Tolentino Collado, Madeeha I. Ali, Aya Sabbah, YongLe He, James N. Iuliano, Christopher R. Hall, Katrin Adamczyk, Gregory M. Greetham, Andras Lukacs*, Stephen R. Meech* and Peter J. Tonge*, 

OaPAC, the photoactivated adenylyl cyclase from Oscillatoria acuminata, is composed of a blue light using FAD (BLUF) domain fused to an adenylate cyclase (AC) domain. Since both the BLUF and AC domains are part of the same protein, OaPAC is a model for understanding how the ultrafast modulation of the chromophore binding pocket caused by photoexcitation results in the activation of the output domain on the μs-s time scale. In the present work, we use unnatural amino acid mutagenesis to identify specific sites in the protein that are involved in transducing the signal from the FAD binding site to the ATP binding site. To provide insight into site-specific structural dynamics, we replaced W90 which is close to the chromophore pocket, F103 which interacts with W90 across the dimer interface, and F180 in the central core of the AC domain, with the infrared probe azido-Phe (AzPhe). Using ultrafast IR, we show that AzPhe at position 90 responds on multiple time scales following photoexcitation. In contrast, the light minus dark IR spectrum of AzPhe103 shows only a minor perturbation in environment between the dark and light states, while replacement of F180 with AzPhe resulted in a protein with no catalytic activity. We also replaced Y125, which hydrogen bonds with N256 across the dimer interface, with fluoro-Tyr residues. All the fluoro-Tyr substituted proteins retained the light-induced red shift in the flavin absorption spectrum; however, only the 3-FY125 OaPAC retained photoinduced catalytic activity. The loss of activity in 3,5-F2Y125 and 2,3,5-F3Y125 OaPAC, which potentially increase the acidity of the Y125 phenol by more than 1000-fold, suggests that deprotonation of Y125 disrupts the signal transduction pathway from the BLUF to the AC domain.

{"title":"Probing the Signal Transduction Mechanism of the Light-Activated Adenylate Cyclase OaPAC Using Unnatural Amino Acid Mutagenesis","authors":"Samruddhi S. Jewlikar,&nbsp;Jinnette Tolentino Collado,&nbsp;Madeeha I. Ali,&nbsp;Aya Sabbah,&nbsp;YongLe He,&nbsp;James N. Iuliano,&nbsp;Christopher R. Hall,&nbsp;Katrin Adamczyk,&nbsp;Gregory M. Greetham,&nbsp;Andras Lukacs*,&nbsp;Stephen R. Meech* and Peter J. Tonge*,&nbsp;","doi":"10.1021/acschembio.4c0062710.1021/acschembio.4c00627","DOIUrl":"https://doi.org/10.1021/acschembio.4c00627https://doi.org/10.1021/acschembio.4c00627","url":null,"abstract":"<p >OaPAC, the photoactivated adenylyl cyclase from <i>Oscillatoria acuminata</i>, is composed of a blue light using FAD (BLUF) domain fused to an adenylate cyclase (AC) domain. Since both the BLUF and AC domains are part of the same protein, OaPAC is a model for understanding how the ultrafast modulation of the chromophore binding pocket caused by photoexcitation results in the activation of the output domain on the μs-s time scale. In the present work, we use unnatural amino acid mutagenesis to identify specific sites in the protein that are involved in transducing the signal from the FAD binding site to the ATP binding site. To provide insight into site-specific structural dynamics, we replaced W90 which is close to the chromophore pocket, F103 which interacts with W90 across the dimer interface, and F180 in the central core of the AC domain, with the infrared probe azido-Phe (AzPhe). Using ultrafast IR, we show that AzPhe at position 90 responds on multiple time scales following photoexcitation. In contrast, the light minus dark IR spectrum of AzPhe103 shows only a minor perturbation in environment between the dark and light states, while replacement of F180 with AzPhe resulted in a protein with no catalytic activity. We also replaced Y125, which hydrogen bonds with N256 across the dimer interface, with fluoro-Tyr residues. All the fluoro-Tyr substituted proteins retained the light-induced red shift in the flavin absorption spectrum; however, only the 3-FY125 OaPAC retained photoinduced catalytic activity. The loss of activity in 3,5-F<sub>2</sub>Y125 and 2,3,5-F<sub>3</sub>Y125 OaPAC, which potentially increase the acidity of the Y125 phenol by more than 1000-fold, suggests that deprotonation of Y125 disrupts the signal transduction pathway from the BLUF to the AC domain.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":"20 2","pages":"369–377 369–377"},"PeriodicalIF":3.5,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143452724","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Development of an In Situ G Protein-Coupled Receptor Fragment Molecule Screening Approach with High-Resolution Magic Angle Spinning Nuclear Magnetic Resonance
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-21 DOI: 10.1021/acschembio.4c0068610.1021/acschembio.4c00686
Enzo Petracco, Guillaume Ferré, Ivo Kabelka, Flavio Ballante, Jens Carlsson, Emma Mulry, Arka P. Ray, James Collins, Florent Allais and Matthew T. Eddy*, 

Small molecules are essential for investigating the pharmacology of membrane proteins and remain the most common approach for therapeutically targeting them. However, most experimental small molecule screening methods require ligands containing radiolabels or fluorescent labels and often involve isolating proteins from their cellular environment. Additionally, most conventional screening methods are suited for identifying compounds with moderate to higher affinities (KD < 1 μM) and are less effective at detecting lower affinity compounds, such as weakly binding molecular fragments. To address these limitations, we demonstrated a proof-of-concept application of high-resolution magic angle spinning nuclear magnetic resonance (HRMAS NMR) spectroscopy with small molecules that bind the human A2A adenosine receptor (A2AAR), a class A G protein-coupled receptor. Our approach leverages a streamlined workflow to prepare NMR samples with only milligrams of unpurified cell membranes containing ∼1 μM of A2AAR. Utilizing saturation transfer difference NMR, we identified bound small molecules from spectra recorded within minutes and further derived information on ligand binding poses without the need for detailed structure determination. After establishing optimal criteria for which the HRMAS approach is most sensitive, we leveraged our HRMAS approach to identify and characterize molecular fragments not previously known to be ligands of A2AAR. In molecular docking and simulations, we observed novel binding poses for these fragments, which revealed the potential to grow them into more complex ligands and confirmed HRMAS NMR as a valuable tool for lead compound identification in the context of fragment-based drug discovery.

{"title":"Development of an In Situ G Protein-Coupled Receptor Fragment Molecule Screening Approach with High-Resolution Magic Angle Spinning Nuclear Magnetic Resonance","authors":"Enzo Petracco,&nbsp;Guillaume Ferré,&nbsp;Ivo Kabelka,&nbsp;Flavio Ballante,&nbsp;Jens Carlsson,&nbsp;Emma Mulry,&nbsp;Arka P. Ray,&nbsp;James Collins,&nbsp;Florent Allais and Matthew T. Eddy*,&nbsp;","doi":"10.1021/acschembio.4c0068610.1021/acschembio.4c00686","DOIUrl":"https://doi.org/10.1021/acschembio.4c00686https://doi.org/10.1021/acschembio.4c00686","url":null,"abstract":"<p >Small molecules are essential for investigating the pharmacology of membrane proteins and remain the most common approach for therapeutically targeting them. However, most experimental small molecule screening methods require ligands containing radiolabels or fluorescent labels and often involve isolating proteins from their cellular environment. Additionally, most conventional screening methods are suited for identifying compounds with moderate to higher affinities (<i>K</i><sub>D</sub> &lt; 1 μM) and are less effective at detecting lower affinity compounds, such as weakly binding molecular fragments. To address these limitations, we demonstrated a proof-of-concept application of high-resolution magic angle spinning nuclear magnetic resonance (HRMAS NMR) spectroscopy with small molecules that bind the human A<sub>2A</sub> adenosine receptor (A<sub>2A</sub>AR), a class A G protein-coupled receptor. Our approach leverages a streamlined workflow to prepare NMR samples with only milligrams of unpurified cell membranes containing ∼1 μM of A<sub>2A</sub>AR. Utilizing saturation transfer difference NMR, we identified bound small molecules from spectra recorded within minutes and further derived information on ligand binding poses without the need for detailed structure determination. After establishing optimal criteria for which the HRMAS approach is most sensitive, we leveraged our HRMAS approach to identify and characterize molecular fragments not previously known to be ligands of A<sub>2A</sub>AR. In molecular docking and simulations, we observed novel binding poses for these fragments, which revealed the potential to grow them into more complex ligands and confirmed HRMAS NMR as a valuable tool for lead compound identification in the context of fragment-based drug discovery.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":"20 2","pages":"401–411 401–411"},"PeriodicalIF":3.5,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143452699","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Tools for Intersectional Optical and Chemical Tagging on Cell Surfaces
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-21 DOI: 10.1021/acschembio.4c0075610.1021/acschembio.4c00756
Sarah Innes-Gold, Hanzeng Cheng, Luping Liu* and Adam E. Cohen*, 

We present versatile tools for intersectional optical and chemical tagging of live cells. Photocaged tetrazines serve as “photo-click” adapters between recognition groups on the cell surface and diverse chemical payloads. We describe two new functionalized photocaged tetrazine structures which add a light-gating step to three common cell-targeting chemical methods: HaloTag/chloroalkane labeling, nonspecific primary amine labeling, and antibody labeling. We demonstrate light-gated versions of these three techniques in live cultured cells. We then explore two applications: monitoring tissue flows on the surface of developing zebrafish embryos, and combinatorial multicolor labeling and sorting of optically defined groups of cells. Photoclick adapters add optical control to cell tagging schemes, with modularity in both tag and cell attachment chemistry.

{"title":"Tools for Intersectional Optical and Chemical Tagging on Cell Surfaces","authors":"Sarah Innes-Gold,&nbsp;Hanzeng Cheng,&nbsp;Luping Liu* and Adam E. Cohen*,&nbsp;","doi":"10.1021/acschembio.4c0075610.1021/acschembio.4c00756","DOIUrl":"https://doi.org/10.1021/acschembio.4c00756https://doi.org/10.1021/acschembio.4c00756","url":null,"abstract":"<p >We present versatile tools for intersectional optical and chemical tagging of live cells. Photocaged tetrazines serve as “photo-click” adapters between recognition groups on the cell surface and diverse chemical payloads. We describe two new functionalized photocaged tetrazine structures which add a light-gating step to three common cell-targeting chemical methods: HaloTag/chloroalkane labeling, nonspecific primary amine labeling, and antibody labeling. We demonstrate light-gated versions of these three techniques in live cultured cells. We then explore two applications: monitoring tissue flows on the surface of developing zebrafish embryos, and combinatorial multicolor labeling and sorting of optically defined groups of cells. Photoclick adapters add optical control to cell tagging schemes, with modularity in both tag and cell attachment chemistry.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":"20 2","pages":"455–463 455–463"},"PeriodicalIF":3.5,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143452722","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Understanding the Glycosylation Pathways Involved in the Biosynthesis of the Sulfated Glycan Ligands for Siglecs
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-21 DOI: 10.1021/acschembio.4c0067710.1021/acschembio.4c00677
Jaesoo Jung, Edward N. Schmidt, Hua-Chien Chang, Zeinab Jame-Chenarboo, Jhon R. Enterina, Kelli A. McCord, Taylor E. Gray, Lauren Kageler, Chris D. St. Laurent, Chao Wang, Ryan A. Flynn, Peng Wu, Kay-Hooi Khoo and Matthew S. Macauley*, 

Carbohydrate sulfation plays a pivotal role in modulating the strength of Siglec–glycan interactions. Recently, new aspects of Siglec binding to sulfated cell surface carbohydrates have been discovered, but the class of glycan presenting these sulfated Siglec ligands has not been fully elucidated. In this study, the contribution of different classes of glycans to cis and trans Siglec ligands was investigated within cells expressing the carbohydrate sulfotransferase 1 (CHST1) or CHST2. For some Siglecs, the glycan class mediating binding was clear, such as O-glycans for Siglec-7 and N-glycans for Siglec-2 and Siglec-9. Both N-glycans and mucin-type O-glycans contributed to ligands for Siglec-3, -5, -8, and -15. However, significant levels of Siglec-3 and -8 ligands remained in CHST1-expressing cells lacking complex N-glycans and mucin-type O-glycans. A combination of genetic, pharmacological, and enzymatic treatment strategies ruled out heparan sulfates and glycoRNA as contributors, although Siglec-8 did exhibit some binding to glycolipids. Genetic disruption of O-mannose glycans within CHST1-expressing cells had a small but significant impact on Siglec-3 and -8 binding, demonstrating that this class of glycans can present sulfated Siglec ligands. We also investigated the ability of sulfated cis ligands to mask Siglec-3 and Siglec-7. For Siglec-7, cis ligands were again found to be mucin-type O-glycans. While N-glycans were the major sulfated trans ligands for Siglec-3, disruption of complex mucin-type O-glycans had the largest impact on Siglec-3 masking. Overall, this study enhances our knowledge of the types of sulfated glycans that can serve as Siglec ligands.

{"title":"Understanding the Glycosylation Pathways Involved in the Biosynthesis of the Sulfated Glycan Ligands for Siglecs","authors":"Jaesoo Jung,&nbsp;Edward N. Schmidt,&nbsp;Hua-Chien Chang,&nbsp;Zeinab Jame-Chenarboo,&nbsp;Jhon R. Enterina,&nbsp;Kelli A. McCord,&nbsp;Taylor E. Gray,&nbsp;Lauren Kageler,&nbsp;Chris D. St. Laurent,&nbsp;Chao Wang,&nbsp;Ryan A. Flynn,&nbsp;Peng Wu,&nbsp;Kay-Hooi Khoo and Matthew S. Macauley*,&nbsp;","doi":"10.1021/acschembio.4c0067710.1021/acschembio.4c00677","DOIUrl":"https://doi.org/10.1021/acschembio.4c00677https://doi.org/10.1021/acschembio.4c00677","url":null,"abstract":"<p >Carbohydrate sulfation plays a pivotal role in modulating the strength of Siglec–glycan interactions. Recently, new aspects of Siglec binding to sulfated cell surface carbohydrates have been discovered, but the class of glycan presenting these sulfated Siglec ligands has not been fully elucidated. In this study, the contribution of different classes of glycans to <i>cis</i> and <i>trans</i> Siglec ligands was investigated within cells expressing the carbohydrate sulfotransferase 1 (CHST1) or CHST2. For some Siglecs, the glycan class mediating binding was clear, such as <i>O</i>-glycans for Siglec-7 and <i>N</i>-glycans for Siglec-2 and Siglec-9. Both <i>N</i>-glycans and mucin-type <i>O</i>-glycans contributed to ligands for Siglec-3, -5, -8, and -15. However, significant levels of Siglec-3 and -8 ligands remained in CHST1-expressing cells lacking complex <i>N</i>-glycans and mucin-type <i>O</i>-glycans. A combination of genetic, pharmacological, and enzymatic treatment strategies ruled out heparan sulfates and glycoRNA as contributors, although Siglec-8 did exhibit some binding to glycolipids. Genetic disruption of <i>O</i>-mannose glycans within CHST1-expressing cells had a small but significant impact on Siglec-3 and -8 binding, demonstrating that this class of glycans can present sulfated Siglec ligands. We also investigated the ability of sulfated <i>cis</i> ligands to mask Siglec-3 and Siglec-7. For Siglec-7, <i>cis</i> ligands were again found to be mucin-type <i>O</i>-glycans. While <i>N</i>-glycans were the major sulfated <i>trans</i> ligands for Siglec-3, disruption of complex mucin-type <i>O</i>-glycans had the largest impact on Siglec-3 masking. Overall, this study enhances our knowledge of the types of sulfated glycans that can serve as Siglec ligands.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":"20 2","pages":"386–400 386–400"},"PeriodicalIF":3.5,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143452700","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Breaking the Myth of Enzymatic Azoreduction. 打破酶促偶氮还原的神话。
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-17 Epub Date: 2024-12-21 DOI: 10.1021/acschembio.4c00779
Yu-Ju Peng, Bing Xu, Steven E Rokita

Flavin-dependent azoreductases have been applied to a wide range of tasks from decolorizing numerous azo dyes to releasing azo-conjugated prodrugs. A general narrative reiterated in much of the literature suggests that this enzyme promotes sequential reduction of both the azo-containing substrate and its corresponding hydrazo product to release the aryl amine components while consuming two equivalents of NAD(P)H. Indeed, such aryl amines can be formed by incubation of certain azo compounds with azoreductases, but the nature of the substrates capable of this apparent azo bond lysis remained unknown. We have now prepared a set of azobenzene derivatives and characterized their turnover and products after treatment with azoreductase from Escherichia coli to discover the structural basis regulating aryl amine formation. Without resonance donation by aryl substituents, reduction ceases at the hydrazo product. This indicates that azoreductases do not act on the hydrazo bond. Instead, aryl amine formation depends on a spontaneous hydrazo bond lysis that is promoted by resonance stabilization and subsequent reduction of the quinone-like intermediate by azoreductase. Experimental and computational approaches confirm the substituent dependence of this process. With knowledge of this requirement, full release of aryl amines from azo-conjugates can now be designed and applied with confidence.

黄素依赖性偶氮还原酶已广泛应用于各种任务,从脱色大量偶氮染料到释放偶氮偶联前药。在许多文献中重申的一般叙述表明,该酶促进含偶氮的底物及其相应的肼产物的顺序还原,以释放芳胺成分,同时消耗两等量的NAD(P)H。事实上,这种芳基胺可以通过某些偶氮化合物与偶氮还原酶的孵育形成,但是能够这种明显的偶氮键裂解的底物的性质仍然未知。我们现在制备了一组偶氮苯衍生物,并对其在大肠杆菌中的偶氮还原酶处理后的转化和产物进行了表征,以发现调节芳胺形成的结构基础。如果没有芳基取代基的共振给予,则在偶联产物上停止还原。这表明偶氮还原酶不作用于偶氮键。相反,芳基胺的形成依赖于自发的腙键裂解,这是由共振稳定和随后的偶氮还原酶对类醌中间体的还原所促进的。实验和计算方法证实了这一过程的取代基依赖性。有了这方面的知识,从偶氮偶联物完全释放芳基胺现在可以设计和应用的信心。
{"title":"Breaking the Myth of Enzymatic Azoreduction.","authors":"Yu-Ju Peng, Bing Xu, Steven E Rokita","doi":"10.1021/acschembio.4c00779","DOIUrl":"10.1021/acschembio.4c00779","url":null,"abstract":"<p><p>Flavin-dependent azoreductases have been applied to a wide range of tasks from decolorizing numerous azo dyes to releasing azo-conjugated prodrugs. A general narrative reiterated in much of the literature suggests that this enzyme promotes sequential reduction of both the azo-containing substrate and its corresponding hydrazo product to release the aryl amine components while consuming two equivalents of NAD(P)H. Indeed, such aryl amines can be formed by incubation of certain azo compounds with azoreductases, but the nature of the substrates capable of this apparent azo bond lysis remained unknown. We have now prepared a set of azobenzene derivatives and characterized their turnover and products after treatment with azoreductase from <i>Escherichia coli</i> to discover the structural basis regulating aryl amine formation. Without resonance donation by aryl substituents, reduction ceases at the hydrazo product. This indicates that azoreductases do not act on the hydrazo bond. Instead, aryl amine formation depends on a spontaneous hydrazo bond lysis that is promoted by resonance stabilization and subsequent reduction of the quinone-like intermediate by azoreductase. Experimental and computational approaches confirm the substituent dependence of this process. With knowledge of this requirement, full release of aryl amines from azo-conjugates can now be designed and applied with confidence.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":"229-237"},"PeriodicalIF":3.5,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142870570","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Relationship between Substrate Structure and Selectivity of Ketoreduction in Multimodular Polyketide Synthases: A Comparative Study of A-Type Ketoreductases from Late Modules Using Complex Precursor Analogues. 多模聚酮合成酶中底物结构与酮还原选择性的关系:采用复杂前体类似物的后期模A型酮还原酶的比较研究。
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-17 Epub Date: 2025-01-08 DOI: 10.1021/acschembio.4c00669
Lisa N K T Nguyen, Sebastian Derra, Frank Hahn

Ketoreductases (KRs) are domains in the reductive loops of type I polyketide synthases (PKSs) and are responsible for the majority of stereocenters in reduced polyketides. Although the highly stereoselective reduction of ACP-bound β-ketothioester intermediates by KRs is crucial for the overall functioning of PKSs, the substrate-dependent stereoselectivity of KRs is a factor that is not yet fully understood, especially for KR domains in late PKS modules that act on biosynthetic precursors with complex polyketidic moieties. We present studies on the three KR domains FosKR7, PlmKR6, and EryKR6 from the biosynthetic pathways of fostriecin, phoslactomycin, and erythromycin by in vitro assays using close surrogates of the octaketidic FosKR7 biosynthetic precursor, complex derivatives and a diketide in the form of their biomimetic N-acetylcysteamine thioesters. Supported by molecular modeling, specific interactions of the studied KR domains with the extended polyketide moieties of their natural precursors were identified and correlated to the differences in stereoselectivity observed in the in vitro assays. These results reinforce the importance of the substrate-dependent stereoselectivity of KR domains in PKSs and suggest more detailed experimental and structural studies with isolated KRs and full PKS modules that could ultimately lead to improved results in PKS engineering.

酮还原酶(Ketoreductases, KRs)是I型聚酮合酶(pks)的还原环中的结构域,在还原的聚酮中起着大多数立体中心的作用。虽然KRs对acp结合的β-酮硫酯中间体的高度立体选择性还原对PKS的整体功能至关重要,但KRs依赖于底物的立体选择性是一个尚未完全了解的因素,特别是对于作用于具有复杂多酮基团的生物合成前体的PKS模块晚期的KR结构域。我们利用八摄性FosKR7生物合成前体的近代物、复合衍生物和以其仿生n -乙酰半胱胺硫酯形式存在的二酮,在体外研究了FosKR7、光霉素和红霉素生物合成途径中的三个KR结构域FosKR7、PlmKR6和EryKR6。在分子模型的支持下,研究的KR结构域与其天然前体的延伸聚酮部分的特定相互作用被确定,并与体外实验中观察到的立体选择性差异相关。这些结果强化了PKS中KR结构域依赖底物立体选择性的重要性,并建议对分离的KRs和完整的PKS模块进行更详细的实验和结构研究,最终可以改善PKS工程的结果。
{"title":"The Relationship between Substrate Structure and Selectivity of Ketoreduction in Multimodular Polyketide Synthases: A Comparative Study of A-Type Ketoreductases from Late Modules Using Complex Precursor Analogues.","authors":"Lisa N K T Nguyen, Sebastian Derra, Frank Hahn","doi":"10.1021/acschembio.4c00669","DOIUrl":"10.1021/acschembio.4c00669","url":null,"abstract":"<p><p>Ketoreductases (KRs) are domains in the reductive loops of type I polyketide synthases (PKSs) and are responsible for the majority of stereocenters in reduced polyketides. Although the highly stereoselective reduction of ACP-bound β-ketothioester intermediates by KRs is crucial for the overall functioning of PKSs, the substrate-dependent stereoselectivity of KRs is a factor that is not yet fully understood, especially for KR domains in late PKS modules that act on biosynthetic precursors with complex polyketidic moieties. We present studies on the three KR domains FosKR7, PlmKR6, and EryKR6 from the biosynthetic pathways of fostriecin, phoslactomycin, and erythromycin by in vitro assays using close surrogates of the octaketidic FosKR7 biosynthetic precursor, complex derivatives and a diketide in the form of their biomimetic <i>N</i>-acetylcysteamine thioesters. Supported by molecular modeling, specific interactions of the studied KR domains with the extended polyketide moieties of their natural precursors were identified and correlated to the differences in stereoselectivity observed in the in vitro assays. These results reinforce the importance of the substrate-dependent stereoselectivity of KR domains in PKSs and suggest more detailed experimental and structural studies with isolated KRs and full PKS modules that could ultimately lead to improved results in PKS engineering.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":"186-196"},"PeriodicalIF":3.5,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142941420","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Functional Characterization of Pathway Inhibitors for the Ubiquitin-Proteasome System (UPS) as Tool Compounds for CRBN and VHL-Mediated Targeted Protein Degradation. 泛素-蛋白酶体系统(UPS)作为CRBN和vhl介导的靶向蛋白质降解工具化合物的途径抑制剂的功能表征。
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-17 Epub Date: 2025-01-03 DOI: 10.1021/acschembio.4c00450
Martin P Schwalm, Amelie Menge, Lewis Elson, Francesco A Greco, Matthew B Robers, Susanne Müller, Stefan Knapp

Small molecule degraders such as PROteolysis TArgeting Chimeras (PROTACs) and molecular glues are new modalities for drug development and important tools for target validation. When appropriately optimized, both modalities lead to proteasomal degradation of the protein of interest (POI). Due to the complexity of the induced multistep degradation process, controls for degrader evaluation are critical and commonly used in the literature. However, comparative studies and evaluations of cellular potencies of these control compounds have not been published so far. Here, we investigated a diverse set of ubiquitin pathway inhibitors and evaluated their potency and utility within the CRBN and VHL-mediated degradation pathway. We used the HiBiT system to measure the level of target rescue after treatment with the control compounds. In addition, the cell health was assessed using a multiplexed high-content assay. These assays allowed us to determine nontoxic effective concentrations for control experiments and to perform rescue experiments in the absence of cellular toxicity.

蛋白水解靶向嵌合体(PROteolysis TArgeting Chimeras, PROTACs)和分子胶等小分子降解物是药物开发的新模式,也是靶标验证的重要工具。当适当优化时,这两种方式都会导致感兴趣蛋白(POI)的蛋白酶体降解。由于诱导的多步降解过程的复杂性,对降解器评价的控制是至关重要的,并且在文献中被广泛使用。然而,到目前为止,这些对照化合物的细胞效力的比较研究和评价尚未发表。在这里,我们研究了多种泛素途径抑制剂,并评估了它们在CRBN和vhl介导的降解途径中的效力和效用。我们使用HiBiT系统测量对照化合物处理后的靶救水平。此外,使用多重高含量测定法评估细胞健康状况。这些试验使我们能够确定对照实验的无毒有效浓度,并在没有细胞毒性的情况下进行拯救实验。
{"title":"Functional Characterization of Pathway Inhibitors for the Ubiquitin-Proteasome System (UPS) as Tool Compounds for CRBN and VHL-Mediated Targeted Protein Degradation.","authors":"Martin P Schwalm, Amelie Menge, Lewis Elson, Francesco A Greco, Matthew B Robers, Susanne Müller, Stefan Knapp","doi":"10.1021/acschembio.4c00450","DOIUrl":"10.1021/acschembio.4c00450","url":null,"abstract":"<p><p>Small molecule degraders such as PROteolysis TArgeting Chimeras (PROTACs) and molecular glues are new modalities for drug development and important tools for target validation. When appropriately optimized, both modalities lead to proteasomal degradation of the protein of interest (POI). Due to the complexity of the induced multistep degradation process, controls for degrader evaluation are critical and commonly used in the literature. However, comparative studies and evaluations of cellular potencies of these control compounds have not been published so far. Here, we investigated a diverse set of ubiquitin pathway inhibitors and evaluated their potency and utility within the CRBN and VHL-mediated degradation pathway. We used the HiBiT system to measure the level of target rescue after treatment with the control compounds. In addition, the cell health was assessed using a multiplexed high-content assay. These assays allowed us to determine nontoxic effective concentrations for control experiments and to perform rescue experiments in the absence of cellular toxicity.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":"94-104"},"PeriodicalIF":3.5,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11745162/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142925797","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Emergence of Oligonucleotide Building Blocks in the Multispecific Proximity-Inducing Drug Toolbox of Destruction. 多特异性接近诱导药物破坏工具箱中寡核苷酸构建块的出现。
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-17 Epub Date: 2024-12-20 DOI: 10.1021/acschembio.4c00311
Kevin Xiao Tong Zhou, Katherine E Bujold

Oligonucleotides are a rapidly emerging class of therapeutics. Their most well-known examples are informational drugs that modify gene expression by binding mRNA. Despite inducing proximity between biological machinery and mRNA when applied to modulating gene expression, oligonucleotides are not typically labeled as "proximity-inducing" in literature. Yet, they have recently been explored as building blocks for multispecific proximity-inducing drugs (MPIDs). MPIDs are unique because they can direct endogenous biological machinery to destroy targeted molecules and cells, in contrast to traditional drugs that inhibit only their functions. The unique mechanism of action of MPIDs has enabled the targeting of previously "undruggable" molecular entities that cannot be effectively inhibited. However, the development of MPIDs must ensure that these molecules will selectively direct a potent, destruction-based mechanism of action toward intended targets over healthy tissues to avoid causing life-threatening toxicities. Oligonucleotides have emerged as promising building blocks for the design of MPIDs because they are sequence-controlled molecules that can be rationally designed to program multispecific binding interactions. In this Review, we examine the emergence of oligonucleotide-containing MPIDs in the proximity induction space, which has been dominated by antibody and small molecule MPID modalities. Moreover, examples of oligonucleotides developed as MPID candidates in immunotherapy and protein degradation are discussed to demonstrate the utility of oligonucleotides in expanding the scope and selectivity of the MPID toolbox. Finally, we discuss the utility of programming "AND" gates into oligonucleotide scaffolds to encode conditional responses that have the potential to be incorporated into MPIDs, which can further enhance their selectivity, thus increasing the scope of this drug category.

寡核苷酸是一种新兴的治疗药物。最著名的例子是通过结合mRNA来改变基因表达的信息药物。尽管应用于调节基因表达时诱导生物机制和mRNA之间的接近性,但寡核苷酸在文献中通常不被标记为“接近性诱导”。然而,它们最近被探索作为多特异性邻近诱导药物(MPIDs)的基础。MPIDs是独一无二的,因为它们可以指导内源性生物机制破坏目标分子和细胞,而传统药物只能抑制它们的功能。MPIDs独特的作用机制使其能够靶向以前无法有效抑制的“不可药物”分子实体。然而,MPIDs的开发必须确保这些分子能够选择性地引导一种有效的、基于破坏的作用机制,针对健康组织的预期目标,以避免引起危及生命的毒性。寡核苷酸已成为MPIDs设计的重要组成部分,因为它们是序列控制的分子,可以合理地设计用于编程多特异性结合相互作用。在这篇综述中,我们研究了在近距离诱导空间中出现的含有寡核苷酸的MPID,该空间一直由抗体和小分子MPID模式主导。此外,本文还讨论了在免疫治疗和蛋白质降解中作为MPID候选物的寡核苷酸的例子,以证明寡核苷酸在扩大MPID工具箱的范围和选择性方面的效用。最后,我们讨论了将“AND”门编程到寡核苷酸支架中的效用,以编码有可能被纳入MPIDs的条件反应,这可以进一步提高它们的选择性,从而增加该药物类别的范围。
{"title":"The Emergence of Oligonucleotide Building Blocks in the Multispecific Proximity-Inducing Drug Toolbox of Destruction.","authors":"Kevin Xiao Tong Zhou, Katherine E Bujold","doi":"10.1021/acschembio.4c00311","DOIUrl":"10.1021/acschembio.4c00311","url":null,"abstract":"<p><p>Oligonucleotides are a rapidly emerging class of therapeutics. Their most well-known examples are informational drugs that modify gene expression by binding mRNA. Despite inducing proximity between biological machinery and mRNA when applied to modulating gene expression, oligonucleotides are not typically labeled as \"proximity-inducing\" in literature. Yet, they have recently been explored as building blocks for multispecific proximity-inducing drugs (MPIDs). MPIDs are unique because they can direct endogenous biological machinery to destroy targeted molecules and cells, in contrast to traditional drugs that inhibit only their functions. The unique mechanism of action of MPIDs has enabled the targeting of previously \"undruggable\" molecular entities that cannot be effectively inhibited. However, the development of MPIDs must ensure that these molecules will selectively direct a potent, destruction-based mechanism of action toward intended targets over healthy tissues to avoid causing life-threatening toxicities. Oligonucleotides have emerged as promising building blocks for the design of MPIDs because they are sequence-controlled molecules that can be rationally designed to program multispecific binding interactions. In this Review, we examine the emergence of oligonucleotide-containing MPIDs in the proximity induction space, which has been dominated by antibody and small molecule MPID modalities. Moreover, examples of oligonucleotides developed as MPID candidates in immunotherapy and protein degradation are discussed to demonstrate the utility of oligonucleotides in expanding the scope and selectivity of the MPID toolbox. Finally, we discuss the utility of programming \"AND\" gates into oligonucleotide scaffolds to encode conditional responses that have the potential to be incorporated into MPIDs, which can further enhance their selectivity, thus increasing the scope of this drug category.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":"3-18"},"PeriodicalIF":3.5,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142862500","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PEGylated ATP-Independent Luciferins for Noninvasive High-Sensitivity High-Speed Bioluminescence Imaging. 聚乙二醇化atp非依赖性荧光素用于无创高灵敏度高速生物发光成像。
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-17 Epub Date: 2024-12-23 DOI: 10.1021/acschembio.4c00601
Xiaodong Tian, Yiyu Zhang, Hui-Wang Ai

Bioluminescence imaging (BLI) is a powerful, noninvasive imaging method for animal studies. NanoLuc luciferase and its derivatives are attractive bioluminescent reporters recognized for their efficient photon production and ATP independence. However, utilizing them for animal imaging poses notable challenges. Low substrate solubility has been a prominent problem, limiting in vivo brightness, while the susceptibility of luciferins to auto-oxidation by molecular oxygen in air increases handling complexity and poses an obstacle to obtaining consistent results. To address these issues, we developed a range of caged PEGylated luciferins with increased auto-oxidation resistance and water solubility of up to 25 mM, resulting in substantial in vivo bioluminescence increases in mouse models. This advancement has created the brightest and most sensitive luciferase-luciferin combination, enabling high-speed video-rate imaging of freely moving mice with brain-expressed luciferase. These innovative substrates offer new possibilities for investigating a wide range of biological processes and are poised to become invaluable resources for chemical, biological, and biomedical fields.

生物发光成像(BLI)是一种强大的、无创的动物研究成像方法。纳米荧光素酶及其衍生物因其高效的光子产生和ATP独立性而被认为是有吸引力的生物发光报告者。然而,利用它们进行动物成像带来了显著的挑战。底物溶解度低一直是一个突出的问题,限制了荧光素在体内的亮度,而荧光素对空气中分子氧的自氧化的敏感性增加了处理的复杂性,并对获得一致的结果构成了障碍。为了解决这些问题,我们开发了一系列笼化聚乙二醇化荧光素,这些荧光素具有增强的自抗氧化性和高达25 mM的水溶性,在小鼠模型中导致大量的体内生物发光增加。这一进步创造了最明亮、最敏感的荧光素酶-荧光素组合,使大脑表达的荧光素酶能够对自由运动的小鼠进行高速视频成像。这些创新的基质为研究广泛的生物过程提供了新的可能性,并有望成为化学、生物和生物医学领域的宝贵资源。
{"title":"PEGylated ATP-Independent Luciferins for Noninvasive High-Sensitivity High-Speed Bioluminescence Imaging.","authors":"Xiaodong Tian, Yiyu Zhang, Hui-Wang Ai","doi":"10.1021/acschembio.4c00601","DOIUrl":"10.1021/acschembio.4c00601","url":null,"abstract":"<p><p>Bioluminescence imaging (BLI) is a powerful, noninvasive imaging method for animal studies. NanoLuc luciferase and its derivatives are attractive bioluminescent reporters recognized for their efficient photon production and ATP independence. However, utilizing them for animal imaging poses notable challenges. Low substrate solubility has been a prominent problem, limiting <i>in vivo</i> brightness, while the susceptibility of luciferins to auto-oxidation by molecular oxygen in air increases handling complexity and poses an obstacle to obtaining consistent results. To address these issues, we developed a range of caged PEGylated luciferins with increased auto-oxidation resistance and water solubility of up to 25 mM, resulting in substantial <i>in vivo</i> bioluminescence increases in mouse models. This advancement has created the brightest and most sensitive luciferase-luciferin combination, enabling high-speed video-rate imaging of freely moving mice with brain-expressed luciferase. These innovative substrates offer new possibilities for investigating a wide range of biological processes and are poised to become invaluable resources for chemical, biological, and biomedical fields.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":"128-136"},"PeriodicalIF":3.5,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11744661/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142875325","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Click Chemistry Methodology: The Novel Paintbrush of Drug Design. 点击化学方法论:药物设计的新画笔。
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-01-17 Epub Date: 2024-12-27 DOI: 10.1021/acschembio.4c00608
Ioana Oprea, Terry K Smith

Click chemistry is an immensely powerful technique for the synthesis of reliable and efficient covalent linkages. When undertaken in living cells, the concept is thereby coined bioorthogonal chemistry. Used in conjunction with the photo-cross-linking methodology, it serves as a sound strategy in the exploration of biological processes and beyond. Its broad scope has led to widespread use in many disciplines; however, this Review focuses on the use of click and bioorthogonal chemistry within medicinal chemistry, specifically with regards to drug development applications, namely, the use of DNA-encoded libraries as a novel technique for lead compound discovery, as well as the synthesis of antisense oligonucleotides and protein-drug conjugates. This Review aims to provide a critical perspective and a future outlook of this methodology, such as potential widespread use in cancer therapy and personalized medicine.

点击化学是一种非常强大的技术,用于合成可靠和有效的共价键。当在活细胞中进行时,因此创造了生物正交化学的概念。与光交联方法结合使用,它可以作为探索生物过程及其他方面的良好策略。它的广泛范围已导致在许多学科的广泛使用;然而,本文将重点介绍click和生物正交化学在药物化学中的应用,特别是在药物开发应用方面,即使用dna编码文库作为先导化合物发现的新技术,以及反义寡核苷酸和蛋白质-药物偶联物的合成。这篇综述的目的是提供一个批判性的观点和未来的展望,如该方法在癌症治疗和个性化医疗中的潜在广泛应用。
{"title":"Click Chemistry Methodology: The Novel Paintbrush of Drug Design.","authors":"Ioana Oprea, Terry K Smith","doi":"10.1021/acschembio.4c00608","DOIUrl":"10.1021/acschembio.4c00608","url":null,"abstract":"<p><p>Click chemistry is an immensely powerful technique for the synthesis of reliable and efficient covalent linkages. When undertaken in living cells, the concept is thereby coined bioorthogonal chemistry. Used in conjunction with the photo-cross-linking methodology, it serves as a sound strategy in the exploration of biological processes and beyond. Its broad scope has led to widespread use in many disciplines; however, this Review focuses on the use of click and bioorthogonal chemistry within medicinal chemistry, specifically with regards to drug development applications, namely, the use of DNA-encoded libraries as a novel technique for lead compound discovery, as well as the synthesis of antisense oligonucleotides and protein-drug conjugates. This Review aims to provide a critical perspective and a future outlook of this methodology, such as potential widespread use in cancer therapy and personalized medicine.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":" ","pages":"19-32"},"PeriodicalIF":3.5,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11744672/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142890579","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
ACS Chemical Biology
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