首页 > 最新文献

ACS Chemical Biology最新文献

英文 中文
Pharmacologic Induction of ERα SUMOylation Disrupts Its Chromatin Binding.
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-21 DOI: 10.1021/acschembio.4c00606
Lizhen Wang, Ting Han

Estrogen receptor α (ERα)-positive breast cancer patients are typically treated with ERα inhibitors, including selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs). However, the distinct pharmacological properties of various ERα inhibitors remain incompletely understood. In this study, we employed formaldehyde cross-linking followed by ERα immunoprecipitation and mass spectrometry to reveal that fulvestrant, the first FDA-approved SERD, induces the interaction between ERα and SUMO E3 ligases PIAS1 and PIAS2. Biochemical and genomic assays confirmed that fulvestrant induces SUMOylation of ERα, which inhibits ERα's binding to chromatin DNA. In addition, raloxifene (a SERM) and elacestrant (the first FDA-approved oral SERD) were identified as compounds that similarly induce ERα SUMOylation and inhibit its chromatin interaction. Our findings reveal a mechanism by which select ERα inhibitors disrupt ERα function through SUMOylation, offering insights for the development of next-generation ERα-targeted therapies.

{"title":"Pharmacologic Induction of ERα SUMOylation Disrupts Its Chromatin Binding.","authors":"Lizhen Wang, Ting Han","doi":"10.1021/acschembio.4c00606","DOIUrl":"https://doi.org/10.1021/acschembio.4c00606","url":null,"abstract":"<p><p>Estrogen receptor α (ERα)-positive breast cancer patients are typically treated with ERα inhibitors, including selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs). However, the distinct pharmacological properties of various ERα inhibitors remain incompletely understood. In this study, we employed formaldehyde cross-linking followed by ERα immunoprecipitation and mass spectrometry to reveal that fulvestrant, the first FDA-approved SERD, induces the interaction between ERα and SUMO E3 ligases PIAS1 and PIAS2. Biochemical and genomic assays confirmed that fulvestrant induces SUMOylation of ERα, which inhibits ERα's binding to chromatin DNA. In addition, raloxifene (a SERM) and elacestrant (the first FDA-approved oral SERD) were identified as compounds that similarly induce ERα SUMOylation and inhibit its chromatin interaction. Our findings reveal a mechanism by which select ERα inhibitors disrupt ERα function through SUMOylation, offering insights for the development of next-generation ERα-targeted therapies.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142453205","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
Exploring the Biosynthetic Potential of Tistrella Species for Producing Didemnin Antitumor Agents. 探索 Tistrella 菌种生物合成生产地塞米松抗肿瘤药物的潜力。
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-18 Epub Date: 2024-09-23 DOI: 10.1021/acschembio.4c00384
Haili Zhang, Shipeng Huang, Xiaolin Zou, Wenguang Shi, Mengdi Liang, Yang Lin, Min Zheng, Xiaoyu Tang

Didemnins are a class of cyclic depsipeptides derived from sea tunicates that exhibit potent anticancer, antiviral, and immunosuppressive properties. Although certain Tistrella species can produce didemnins, their complete biosynthetic potential remains largely unexplored. In this study, we utilize feature-based molecular networking to analyze the metabolomics of Tistrella mobilis and Tistrella bauzanensis, focusing on the production of didemnin natural products. In addition to didemnin B, we identify nordidemnin B and [hysp2]didemnin B, as well as several minor didemnin analogs. Heterologous expression of the didemnin biosynthetic gene cluster in a Streptomyces host results in the production of only didemnin B and nordidemnin B in limited quantities. Isotope-labeling studies reveal that the substrate promiscuity of the adenylation domains during biosynthesis leads to the accumulation of nordidemnin B and [hysp2]didemnin B. Additionally, precursor-directed biosynthesis is applied to generate eight novel didemnin derivatives by supplementing the culture with structurally related amino acids. Furthermore, we increased the titers of nordidemnin B and [hysp2]didemnin B by supplementing the fermentation medium with l-valine and l-isoleucine, respectively. Finally, both compounds undergo side-chain oxidation to enhance their biological activity, with their anticancer properties found to be as potent as plitidepsin.

地道霉素是一类从海洋单胞藻中提取的环状去肽类化合物,具有很强的抗癌、抗病毒和免疫抑制作用。尽管某些 Tistrella 种类可以产生地得孕肽,但它们的全部生物合成潜力在很大程度上仍未得到开发。在本研究中,我们利用基于特征的分子网络分析了Tistrella mobilis和Tistrella bauzanensis的代谢组学,重点研究了didemnin天然产物的生产。除了大豆苷 B 外,我们还发现了 nordidemnin B 和 [hysp2]didemnin B 以及几种次要的大豆苷类似物。在链霉菌宿主中异源表达大豆苷生物合成基因簇后,只能产生数量有限的大豆苷 B 和北大豆苷 B。同位素标记研究显示,生物合成过程中腺苷酸化结构域的底物杂合性导致了nordidemnin B和[hysp2]didemnin B的积累。此外,我们还通过在发酵培养基中分别添加 l-缬氨酸和 l-异亮氨酸,提高了北冬青素 B 和 [hysp2]didemnin B 的滴度。最后,这两种化合物都经过侧链氧化以增强其生物活性,其抗癌特性与普利替普新一样有效。
{"title":"Exploring the Biosynthetic Potential of <i>Tistrella</i> Species for Producing Didemnin Antitumor Agents.","authors":"Haili Zhang, Shipeng Huang, Xiaolin Zou, Wenguang Shi, Mengdi Liang, Yang Lin, Min Zheng, Xiaoyu Tang","doi":"10.1021/acschembio.4c00384","DOIUrl":"10.1021/acschembio.4c00384","url":null,"abstract":"<p><p>Didemnins are a class of cyclic depsipeptides derived from sea tunicates that exhibit potent anticancer, antiviral, and immunosuppressive properties. Although certain <i>Tistrella</i> species can produce didemnins, their complete biosynthetic potential remains largely unexplored. In this study, we utilize feature-based molecular networking to analyze the metabolomics of <i>Tistrella mobilis</i> and <i>Tistrella bauzanensis</i>, focusing on the production of didemnin natural products. In addition to didemnin B, we identify nordidemnin B and [hysp<sup>2</sup>]didemnin B, as well as several minor didemnin analogs. Heterologous expression of the didemnin biosynthetic gene cluster in a <i>Streptomyces</i> host results in the production of only didemnin B and nordidemnin B in limited quantities. Isotope-labeling studies reveal that the substrate promiscuity of the adenylation domains during biosynthesis leads to the accumulation of nordidemnin B and [hysp<sup>2</sup>]didemnin B. Additionally, precursor-directed biosynthesis is applied to generate eight novel didemnin derivatives by supplementing the culture with structurally related amino acids. Furthermore, we increased the titers of nordidemnin B and [hysp<sup>2</sup>]didemnin B by supplementing the fermentation medium with l-valine and l-isoleucine, respectively. Finally, both compounds undergo side-chain oxidation to enhance their biological activity, with their anticancer properties found to be as potent as plitidepsin.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142277138","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
Leveraging Covalency to Stabilize Ternary Complex Formation For Cell-Cell "Induced Proximity". 利用共价性稳定三元复合物的形成,实现细胞间的 "诱导接近"。
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-18 Epub Date: 2024-09-26 DOI: 10.1021/acschembio.4c00286
Karolina Krygier, Anjalee N Wijetunge, Arthur Srayeddin, Harrison Mccann, Anthony F Rullo
<p><p>Recent advances in the field of translational chemical biology use diverse "proximity-inducing" synthetic modalities to elicit new modes of "event driven" pharmacology. These include mechanisms of targeted protein degradation and immune clearance of pathogenic cells. Heterobifunctional "chimeric" compounds like Proteolysis TArgeting Chimeras (PROTACs) and Antibody Recruiting Molecules (ARMs) leverage these mechanisms, respectively. Both systems function through the formation of reversible "ternary" or higher-order biomolecular complexes. Critical to function are key parameters, such as bifunctional molecule affinity for endogenous proteins, target residence time, and turnover. To probe the mechanism and enhance function, covalent chemical approaches have been developed to kinetically stabilize ternary complexes. These include electrophilic PROTACs and Covalent Immune Recruiters (CIRs), the latter designed to uniquely enforce cell-cell induced proximity. Inducing cell-cell proximity is associated with key challenges arising from a combination of steric and/or mechanical based destabilizing forces on the ternary complex. These factors can attenuate the formation of ternary complexes driven by high affinity bifunctional/proximity inducing molecules. This Account describes initial efforts in our lab to address these challenges using the CIR strategy in antibody recruitment or receptor engineered T cell model systems of cell-cell induced proximity. ARMs form ternary complexes with serum antibodies and surface protein antigens on tumor cells that subsequently engage immune cells via Fc receptors. Binding and clustering of Fc receptors trigger immune cell killing of the tumor cell. We applied the CIR strategy to convert ARMs to covalent chimeras, which "irreversibly" recruit serum antibodies to tumor cells. These covalent chimeras leverage electrophile preorganization and kinetic effective molarity to achieve fast and selective covalent engagement of the target ternary complex protein, e.g., serum antibody. Importantly, covalent engagement can proceed via diverse binding site amino acids beyond cysteine. Covalent chimeras demonstrated striking functional enhancements compared to noncovalent ARM analogs in functional immune assays. We revealed this enhancement was in fact due to the increased kinetic stability <i>and not</i> concentration, of ternary complexes. This finding was recapitulated using analogous CIR modalities that integrate peptidic or carbohydrate binding ligands with Sulfur(VI) Fluoride Exchange (SuFEx) electrophiles to induce cell-cell proximity. Mechanistic studies in a distinct model system that uses T cells engineered with receptors that recognize covalent chimeras or ARMs, revealed covalent receptor engagement uniquely enforces downstream activation signaling. Finally, this Account discusses potential challenges and future directions for adapting and optimizing covalent chimeric/bifunctional molecules for diverse applications in
转化化学生物学领域的最新进展是利用各种 "近似诱导 "合成模式来激发新的 "事件驱动 "药理学模式。其中包括靶向降解蛋白质和免疫清除致病细胞的机制。蛋白水解嵌合体(PROTACs)和抗体招募分子(ARMs)等异种功能 "嵌合 "化合物分别利用了这些机制。这两种系统都通过形成可逆的 "三元 "或高阶生物分子复合物来发挥作用。双功能分子对内源性蛋白质的亲和力、目标停留时间和周转率等关键参数对功能至关重要。为了探究机理和增强功能,人们开发了共价化学方法来对三元复合物进行动力学稳定。这些方法包括亲电性 PROTAC 和共价免疫招募剂(CIR),后者旨在独特地加强细胞间的诱导接近。诱导细胞接近与三元复合物上的立体和/或机械不稳定力的组合所带来的关键挑战有关。这些因素会削弱由高亲和力双功能/亲近性诱导分子驱动的三元复合物的形成。本报告介绍了我们实验室在抗体招募或细胞-细胞诱导接近的受体工程 T 细胞模型系统中使用 CIR 策略应对这些挑战的初步努力。ARM 与肿瘤细胞上的血清抗体和表面蛋白抗原形成三元复合物,随后通过 Fc 受体与免疫细胞结合。Fc 受体的结合和聚集会引发免疫细胞对肿瘤细胞的杀伤。我们采用 CIR 策略将 ARM 转化为共价嵌合体,"不可逆地 "将血清抗体吸附到肿瘤细胞上。这些共价嵌合体利用亲电子预组织和动力学有效摩尔度实现目标三元复合蛋白(如血清抗体)的快速和选择性共价啮合。重要的是,共价啮合可通过半胱氨酸以外的多种结合位点氨基酸进行。与非共价 ARM 类似物相比,共价嵌合体在功能免疫测定中表现出惊人的功能增强。我们发现,这种增强实际上是由于三元复合物的动力学稳定性而非浓度增加所致。我们使用类似的 CIR 模式再现了这一发现,这种模式将肽或碳水化合物结合配体与氟(VI)硫交换(SuFEx)亲电体整合在一起,以诱导细胞-细胞接近。在一个独特的模型系统中进行的机理研究显示,共价受体参与能独特地加强下游激活信号的传递。最后,本报告讨论了将共价嵌合体/双功能分子应用于细胞-细胞诱导接近的各种领域并对其进行优化的潜在挑战和未来方向。
{"title":"Leveraging Covalency to Stabilize Ternary Complex Formation For Cell-Cell \"Induced Proximity\".","authors":"Karolina Krygier, Anjalee N Wijetunge, Arthur Srayeddin, Harrison Mccann, Anthony F Rullo","doi":"10.1021/acschembio.4c00286","DOIUrl":"10.1021/acschembio.4c00286","url":null,"abstract":"&lt;p&gt;&lt;p&gt;Recent advances in the field of translational chemical biology use diverse \"proximity-inducing\" synthetic modalities to elicit new modes of \"event driven\" pharmacology. These include mechanisms of targeted protein degradation and immune clearance of pathogenic cells. Heterobifunctional \"chimeric\" compounds like Proteolysis TArgeting Chimeras (PROTACs) and Antibody Recruiting Molecules (ARMs) leverage these mechanisms, respectively. Both systems function through the formation of reversible \"ternary\" or higher-order biomolecular complexes. Critical to function are key parameters, such as bifunctional molecule affinity for endogenous proteins, target residence time, and turnover. To probe the mechanism and enhance function, covalent chemical approaches have been developed to kinetically stabilize ternary complexes. These include electrophilic PROTACs and Covalent Immune Recruiters (CIRs), the latter designed to uniquely enforce cell-cell induced proximity. Inducing cell-cell proximity is associated with key challenges arising from a combination of steric and/or mechanical based destabilizing forces on the ternary complex. These factors can attenuate the formation of ternary complexes driven by high affinity bifunctional/proximity inducing molecules. This Account describes initial efforts in our lab to address these challenges using the CIR strategy in antibody recruitment or receptor engineered T cell model systems of cell-cell induced proximity. ARMs form ternary complexes with serum antibodies and surface protein antigens on tumor cells that subsequently engage immune cells via Fc receptors. Binding and clustering of Fc receptors trigger immune cell killing of the tumor cell. We applied the CIR strategy to convert ARMs to covalent chimeras, which \"irreversibly\" recruit serum antibodies to tumor cells. These covalent chimeras leverage electrophile preorganization and kinetic effective molarity to achieve fast and selective covalent engagement of the target ternary complex protein, e.g., serum antibody. Importantly, covalent engagement can proceed via diverse binding site amino acids beyond cysteine. Covalent chimeras demonstrated striking functional enhancements compared to noncovalent ARM analogs in functional immune assays. We revealed this enhancement was in fact due to the increased kinetic stability &lt;i&gt;and not&lt;/i&gt; concentration, of ternary complexes. This finding was recapitulated using analogous CIR modalities that integrate peptidic or carbohydrate binding ligands with Sulfur(VI) Fluoride Exchange (SuFEx) electrophiles to induce cell-cell proximity. Mechanistic studies in a distinct model system that uses T cells engineered with receptors that recognize covalent chimeras or ARMs, revealed covalent receptor engagement uniquely enforces downstream activation signaling. Finally, this Account discusses potential challenges and future directions for adapting and optimizing covalent chimeric/bifunctional molecules for diverse applications in","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337252","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
Next Generation SICLOPPS Screening for the Identification of Inhibitors of the HIF-1α/HIF-1β Protein-Protein Interaction. 用于鉴定 HIF-1α/HIF-1β 蛋白-蛋白相互作用抑制剂的新一代 SICLOPPS 筛选。
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-18 Epub Date: 2024-09-23 DOI: 10.1021/acschembio.4c00494
Alexander McDermott, Leonie M Windeln, Jacob S D Valentine, Leonardo Baldassarre, Andrew D Foster, Ali Tavassoli

Split-intein circular ligation of proteins and peptides (SICLOPPS) is a method for generating intracellular libraries of cyclic peptides that has yielded several first-in-class inhibitors. Here, we detail a revised high-content, high-throughput SICLOPPS screening protocol that utilizes next-generation sequencing, biopanning, and computational tools to identify hits against a given protein-protein interaction. We used this platform for the identification of inhibitors of the HIF-1α/HIF-1β protein-protein interaction. The revised platform resulted in a significantly higher positive hit rate than that previously reported for SICLOPPS screens, and the identified cyclic peptides were more active in vitro and in cells than our previously reported inhibitors. The platform detailed here may be used for the identification of inhibitors of a wide range of other targets.

蛋白质和多肽的分离-引物环状连接(SICLOPPS)是一种生成细胞内环状多肽库的方法,它已经产生了几种一流的抑制剂。在这里,我们详细介绍了一种经过修订的高含量、高通量 SICLOPPS 筛选方案,该方案利用新一代测序、生物扫描和计算工具来鉴定针对特定蛋白质-蛋白质相互作用的命中物。我们利用这一平台鉴定了 HIF-1α/HIF-1β 蛋白-蛋白相互作用的抑制剂。与之前报道的 SICLOPPS 筛选相比,修订后的平台的阳性命中率明显更高,而且与之前报道的抑制剂相比,鉴定出的环肽在体外和细胞中更有活性。本文详述的平台可用于鉴定其他多种靶标的抑制剂。
{"title":"Next Generation SICLOPPS Screening for the Identification of Inhibitors of the HIF-1α/HIF-1β Protein-Protein Interaction.","authors":"Alexander McDermott, Leonie M Windeln, Jacob S D Valentine, Leonardo Baldassarre, Andrew D Foster, Ali Tavassoli","doi":"10.1021/acschembio.4c00494","DOIUrl":"10.1021/acschembio.4c00494","url":null,"abstract":"<p><p>Split-intein circular ligation of proteins and peptides (SICLOPPS) is a method for generating intracellular libraries of cyclic peptides that has yielded several first-in-class inhibitors. Here, we detail a revised high-content, high-throughput SICLOPPS screening protocol that utilizes next-generation sequencing, biopanning, and computational tools to identify hits against a given protein-protein interaction. We used this platform for the identification of inhibitors of the HIF-1α/HIF-1β protein-protein interaction. The revised platform resulted in a significantly higher positive hit rate than that previously reported for SICLOPPS screens, and the identified cyclic peptides were more active in vitro and in cells than our previously reported inhibitors. The platform detailed here may be used for the identification of inhibitors of a wide range of other targets.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142306509","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
Enhancing the Anticancer Activity of Attenuated Listeria monocytogenes by Cell Wall Functionalization with "Clickable" Doxorubicin. 通过细胞壁功能化与 "可点击 "多柔比星增强减毒李斯特菌的抗癌活性
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-18 Epub Date: 2024-09-24 DOI: 10.1021/acschembio.4c00250
Irene Lepori, Marta Roncetti, Marianna Vitiello, Elisabetta Barresi, Raffaella De Paolo, Paolo Maria Tentori, Caterina Baldanzi, Melissa Santi, Monica Evangelista, Giovanni Signore, Lorena Tedeschi, Claudia Gravekamp, Francesco Cardarelli, Sabrina Taliani, Federico Da Settimo, M Sloan Siegrist, Laura Poliseno

Among bacteria used as anticancer vaccines, attenuated Listeria monocytogenes (Lmat) stands out, because it spreads from one infected cancer cell to the next, induces a strong adaptive immune response, and is suitable for repeated injection cycles. Here, we use click chemistry to functionalize the Lmat cell wall and turn the bacterium into an "intelligent carrier" of the chemotherapeutic drug doxorubicin. Doxorubicin-loaded Lmat retains most of its biological properties and, compared to the control fluorophore-functionalized bacteria, shows enhanced cytotoxicity against melanoma cells both in vitro and in a xenograft model in zebrafish. Our results show that drugs can be covalently loaded on the Lmat cell wall and pave the way to the development of new two-in-one therapeutic approaches combining immunotherapy with chemotherapy.

在用作抗癌疫苗的细菌中,减毒李斯特菌(Lmat)脱颖而出,因为它能从一个受感染的癌细胞扩散到下一个癌细胞,诱导强烈的适应性免疫反应,而且适合反复注射。在这里,我们利用点击化学将 Lmat 细胞壁功能化,使这种细菌成为化疗药物多柔比星的 "智能载体"。负载多柔比星的 Lmat 保留了其大部分生物特性,与对照组荧光团功能化细菌相比,它在体外和斑马鱼异种移植模型中对黑色素瘤细胞的细胞毒性都有所增强。我们的研究结果表明,药物可以共价载入 Lmat 细胞壁,为开发结合免疫疗法和化疗的二合一新疗法铺平了道路。
{"title":"Enhancing the Anticancer Activity of Attenuated <i>Listeria monocytogenes</i> by Cell Wall Functionalization with \"Clickable\" Doxorubicin.","authors":"Irene Lepori, Marta Roncetti, Marianna Vitiello, Elisabetta Barresi, Raffaella De Paolo, Paolo Maria Tentori, Caterina Baldanzi, Melissa Santi, Monica Evangelista, Giovanni Signore, Lorena Tedeschi, Claudia Gravekamp, Francesco Cardarelli, Sabrina Taliani, Federico Da Settimo, M Sloan Siegrist, Laura Poliseno","doi":"10.1021/acschembio.4c00250","DOIUrl":"10.1021/acschembio.4c00250","url":null,"abstract":"<p><p>Among bacteria used as anticancer vaccines, attenuated <i>Listeria monocytogenes</i> (Lm<sup>at</sup>) stands out, because it spreads from one infected cancer cell to the next, induces a strong adaptive immune response, and is suitable for repeated injection cycles. Here, we use click chemistry to functionalize the Lm<sup>at</sup> cell wall and turn the bacterium into an \"intelligent carrier\" of the chemotherapeutic drug doxorubicin. Doxorubicin-loaded Lm<sup>at</sup> retains most of its biological properties and, compared to the control fluorophore-functionalized bacteria, shows enhanced cytotoxicity against melanoma cells both in vitro and in a xenograft model in zebrafish. Our results show that drugs can be covalently loaded on the Lm<sup>at</sup> cell wall and pave the way to the development of new two-in-one therapeutic approaches combining immunotherapy with chemotherapy.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337251","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
Glucose Transporter-Targeting Chimeras Enabling Tumor-Selective Degradation of Secreted and Membrane Proteins. 葡萄糖转运体靶向嵌合体能使肿瘤选择性地降解分泌蛋白和膜蛋白。
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-18 Epub Date: 2024-10-07 DOI: 10.1021/acschembio.4c00584
Chengyu Yun, Na Li, Yishu Zhang, Tong Fang, Jing Ma, Zhenting Zheng, Subing Zhou, Xiaoqing Cai

Tumor-selective degradation of target proteins has the potential to offer superior therapeutic benefits with maximized therapeutic windows and minimized off-target effects. However, the development of effective lysosome-targeted degradation platforms for achieving selective protein degradation in tumors remains a substantial challenge. Cancer cells depend on certain solute carrier (SLC) transporters to acquire extracellular nutrients to sustain their metabolism and growth. This current study exploits facilitative glucose transporters (GLUTs), a group of SLC transporters widely overexpressed in numerous types of cancer, to drive the endocytosis and lysosomal degradation of target proteins in tumor cells. GLUT-targeting chimeras (GTACs) were generated by conjugating multiple glucose ligands to an antibody specific for the target protein. We demonstrate that the constructed GTACs can induce the internalization and lysosomal degradation of the extracellular and membrane proteins streptavidin, tumor necrosis factor-alpha (TNF-α), and human epidermal growth factor receptor 2 (HER2). Compared with the parent antibody, the GTAC exhibited higher potency in inhibiting the growth of tumor cells in vitro and enhanced tumor-targeting capacity in a tumor-bearing mouse model. Thus, the GTAC platform represents a novel degradation strategy that harnesses an SLC transporter for tumor-selective depletion of secreted and membrane proteins of interest.

靶蛋白的肿瘤选择性降解有可能提供更优越的治疗效果,使治疗窗口最大化,脱靶效应最小化。然而,开发有效的溶酶体靶向降解平台以实现肿瘤中蛋白质的选择性降解仍然是一个巨大的挑战。癌细胞依赖某些溶质载体(SLC)转运体获取细胞外营养物质,以维持新陈代谢和生长。葡萄糖转运体(GLUTs)是一组在多种癌症中广泛过度表达的 SLC 转运体,本研究利用它来驱动肿瘤细胞中靶蛋白的内吞和溶酶体降解。通过将多种葡萄糖配体与靶蛋白的特异性抗体连接,产生了GLUT靶向嵌合体(GTACs)。我们证明所构建的 GTACs 能诱导细胞外蛋白和膜蛋白链霉亲和素、肿瘤坏死因子-α(TNF-α)和人表皮生长因子受体 2(HER2)的内化和溶酶体降解。与母体抗体相比,GTAC 在体外抑制肿瘤细胞生长的效力更高,在肿瘤小鼠模型中的肿瘤靶向能力更强。因此,GTAC 平台代表了一种新型降解策略,它利用 SLC 转运体对肿瘤分泌蛋白和相关膜蛋白进行选择性清除。
{"title":"Glucose Transporter-Targeting Chimeras Enabling Tumor-Selective Degradation of Secreted and Membrane Proteins.","authors":"Chengyu Yun, Na Li, Yishu Zhang, Tong Fang, Jing Ma, Zhenting Zheng, Subing Zhou, Xiaoqing Cai","doi":"10.1021/acschembio.4c00584","DOIUrl":"10.1021/acschembio.4c00584","url":null,"abstract":"<p><p>Tumor-selective degradation of target proteins has the potential to offer superior therapeutic benefits with maximized therapeutic windows and minimized off-target effects. However, the development of effective lysosome-targeted degradation platforms for achieving selective protein degradation in tumors remains a substantial challenge. Cancer cells depend on certain solute carrier (SLC) transporters to acquire extracellular nutrients to sustain their metabolism and growth. This current study exploits facilitative glucose transporters (GLUTs), a group of SLC transporters widely overexpressed in numerous types of cancer, to drive the endocytosis and lysosomal degradation of target proteins in tumor cells. GLUT-targeting chimeras (GTACs) were generated by conjugating multiple glucose ligands to an antibody specific for the target protein. We demonstrate that the constructed GTACs can induce the internalization and lysosomal degradation of the extracellular and membrane proteins streptavidin, tumor necrosis factor-alpha (TNF-α), and human epidermal growth factor receptor 2 (HER2). Compared with the parent antibody, the GTAC exhibited higher potency in inhibiting the growth of tumor cells in vitro and enhanced tumor-targeting capacity in a tumor-bearing mouse model. Thus, the GTAC platform represents a novel degradation strategy that harnesses an SLC transporter for tumor-selective depletion of secreted and membrane proteins of interest.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142386348","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
Identification and Benchmarking of Myokinasib-II as a Selective and Potent Chemical Probe for Exploring MLCK1 Inhibition. 鉴定 Myokinasib-II 并将其作为探索 MLCK1 抑制作用的选择性强效化学探针的基准
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-18 Epub Date: 2024-09-20 DOI: 10.1021/acschembio.4c00336
Gautam Kumar, Prema Kumari Agarwala, Aswin T Srivatsav, Ashok Ravula, G Ashmitha, Sreenath Balakrishnan, Shobhna Kapoor, Rishikesh Narayan

Deciphering the functional relevance of every protein is crucial to developing a better (patho)physiological understanding of human biology. The discovery and use of quality chemical probes propel exciting developments for developing drugs in therapeutic areas with unmet clinical needs. Myosin light-chain kinase (MLCK) serves as a possible therapeutic target in a plethora of diseases, including inflammatory diseases, cancer, etc. Recent years have seen a substantial increase in interest in exploring MLCK biology. However, there is only one widely used MLCK modulator, namely, ML-7, that too with a narrow working concentration window and high toxicity profile leading to limited insights. Herein, we report the identification of a potent and highly selective chemical probe, Myokinasib-II, from the synthesis and structure-activity relationship studies of a focused indotropane-based compound collection. Notably, it is structurally distinct from ML-7 and hence meets the need for an alternative inhibitor to study MLCK biology as per the recommended best practices. Moreover, our extensive benchmarking studies demonstrate that Myokinasib-II displays better potency, better selectivity profile, and no nonspecific interference in relevant assays as compared to other known MLCK inhibitors.

破译每种蛋白质的功能相关性对于更好地了解人类生物学(病理)至关重要。优质化学探针的发现和使用推动了在临床需求未得到满足的治疗领域开发药物的激动人心的发展。肌球蛋白轻链激酶(MLCK)是炎症性疾病、癌症等多种疾病的可能治疗靶点。近年来,人们对探索肌球蛋白轻链激酶生物学的兴趣大大增加。然而,目前只有一种被广泛使用的 MLCK 调节剂,即 ML-7,它的工作浓度窗口很窄,毒性也很高,导致人们对它的了解有限。在此,我们报告了通过对基于吲哚丙烷的化合物集进行合成和结构-活性关系研究,发现了一种强效、高选择性的化学探针 Myokinasib-II。值得注意的是,它在结构上不同于 ML-7,因此满足了研究 MLCK 生物学所需的替代抑制剂,符合推荐的最佳实践。此外,我们进行的大量基准研究表明,与其他已知的 MLCK 抑制剂相比,Myokinasib-II 具有更强的效力和更好的选择性,并且在相关试验中不会产生非特异性干扰。
{"title":"Identification and Benchmarking of Myokinasib-II as a Selective and Potent Chemical Probe for Exploring MLCK1 Inhibition.","authors":"Gautam Kumar, Prema Kumari Agarwala, Aswin T Srivatsav, Ashok Ravula, G Ashmitha, Sreenath Balakrishnan, Shobhna Kapoor, Rishikesh Narayan","doi":"10.1021/acschembio.4c00336","DOIUrl":"10.1021/acschembio.4c00336","url":null,"abstract":"<p><p>Deciphering the functional relevance of every protein is crucial to developing a better (patho)physiological understanding of human biology. The discovery and use of quality chemical probes propel exciting developments for developing drugs in therapeutic areas with unmet clinical needs. Myosin light-chain kinase (MLCK) serves as a possible therapeutic target in a plethora of diseases, including inflammatory diseases, cancer, etc. Recent years have seen a substantial increase in interest in exploring MLCK biology. However, there is only one widely used MLCK modulator, namely, ML-7, that too with a narrow working concentration window and high toxicity profile leading to limited insights. Herein, we report the identification of a potent and highly selective chemical probe, Myokinasib-II, from the synthesis and structure-activity relationship studies of a focused indotropane-based compound collection. Notably, it is structurally distinct from ML-7 and hence meets the need for an alternative inhibitor to study MLCK biology as per the recommended best practices. Moreover, our extensive benchmarking studies demonstrate that Myokinasib-II displays better potency, better selectivity profile, and no nonspecific interference in relevant assays as compared to other known MLCK inhibitors.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142277139","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
Discovery, Characterization, and Structure of a Cell Active PAD2 Inhibitor Acting through a Novel Allosteric Mechanism. 通过新型异构机制发挥作用的细胞活性 PAD2 抑制剂的发现、特征描述和结构。
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-18 Epub Date: 2024-09-24 DOI: 10.1021/acschembio.4c00397
Laura J Byrnes, Won Young Choi, Paul Balbo, Mary Ellen Banker, Jeanne Chang, Shi Chen, Xuemin Cheng, Yang Cong, Jeff Culp, Hongxia Di, Matt Griffor, Justin Hall, Xiaoyun Meng, Barry Morgan, James J Mousseau, Jennifer Nicki, Thomas O'Connell, Simeon Ramsey, Alex Shaginian, Suman Shanker, John Trujillo, Jinqiao Wan, Fabien Vincent, Stephen W Wright, Felix Vajdos

Peptidyl arginine deiminases (PADs) are important enzymes in many diseases, especially those involving inflammation and autoimmunity. Despite many years of effort, developing isoform-specific inhibitors has been a challenge. We describe herein the discovery of a potent, noncovalent PAD2 inhibitor, with selectivity over PAD3 and PAD4, from a DNA-encoded library. The biochemical and biophysical characterization of this inhibitor and two noninhibitory binders indicated a novel, Ca2+ competitive mechanism of inhibition. This was confirmed via X-ray crystallographic analysis. Finally, we demonstrate that this inhibitor selectively inhibits PAD2 in a cellular context.

肽基精氨酸脱氨酶(PADs)是许多疾病中的重要酶类,尤其是涉及炎症和自身免疫的疾病。尽管经过多年的努力,开发同工酶特异性抑制剂仍是一项挑战。我们在本文中介绍了从 DNA 编码文库中发现的一种强效、非共价的 PAD2 抑制剂,它对 PAD3 和 PAD4 具有选择性。这种抑制剂和两种非抑制性结合剂的生物化学和生物物理特性分析表明了一种新型的 Ca2+ 竞争性抑制机制。这一点通过 X 射线晶体学分析得到了证实。最后,我们证明了这种抑制剂能在细胞环境中选择性地抑制 PAD2。
{"title":"Discovery, Characterization, and Structure of a Cell Active PAD2 Inhibitor Acting through a Novel Allosteric Mechanism.","authors":"Laura J Byrnes, Won Young Choi, Paul Balbo, Mary Ellen Banker, Jeanne Chang, Shi Chen, Xuemin Cheng, Yang Cong, Jeff Culp, Hongxia Di, Matt Griffor, Justin Hall, Xiaoyun Meng, Barry Morgan, James J Mousseau, Jennifer Nicki, Thomas O'Connell, Simeon Ramsey, Alex Shaginian, Suman Shanker, John Trujillo, Jinqiao Wan, Fabien Vincent, Stephen W Wright, Felix Vajdos","doi":"10.1021/acschembio.4c00397","DOIUrl":"10.1021/acschembio.4c00397","url":null,"abstract":"<p><p>Peptidyl arginine deiminases (PADs) are important enzymes in many diseases, especially those involving inflammation and autoimmunity. Despite many years of effort, developing isoform-specific inhibitors has been a challenge. We describe herein the discovery of a potent, noncovalent PAD2 inhibitor, with selectivity over PAD3 and PAD4, from a DNA-encoded library. The biochemical and biophysical characterization of this inhibitor and two noninhibitory binders indicated a novel, Ca<sup>2+</sup> competitive mechanism of inhibition. This was confirmed via X-ray crystallographic analysis. Finally, we demonstrate that this inhibitor selectively inhibits PAD2 in a cellular context.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337250","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
Structural Insights and Reaction Profile of a New Unspecific Peroxygenase from Marasmius wettsteinii Produced in a Tandem-Yeast Expression System. 在串联酵母表达系统中从 Marasmius wettsteinii 中产生的一种新的非特异性过氧化氢酶的结构见解和反应特征
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-18 Epub Date: 2024-10-05 DOI: 10.1021/acschembio.4c00504
Israel Sánchez-Moreno, Angela Fernandez-Garcia, Ivan Mateljak, Patricia Gomez de Santos, Martin Hofrichter, Harald Kellner, Julia Sanz-Aparicio, Miguel Alcalde

Fungal unspecific peroxygenases (UPOs) are gaining momentum in synthetic chemistry. Of special interest is the UPO from Marasmius rotula (MroUPO), which shows an exclusive repertoire of oxyfunctionalizations, including the terminal hydroxylation of alkanes, the α-oxidation of fatty acids and the C-C cleavage of corticosteroids. However, the lack of heterologous expression systems to perform directed evolution has impeded its engineering for practical applications. Here, we introduce a close ortholog of MroUPO, a UPO gene from Marasmius wettsteinii (MweUPO-1), that has a similar reaction profile to MroUPO and for which we have set up a directed evolution platform based on tandem-yeast expression. Recombinant MweUPO-1 was produced at high titers in the bioreactor (0.7 g/L) and characterized at the biochemical and atomic levels. The conjunction of soaking crystallographic experiments at a resolution up to 1.6 Å together with the analysis of reaction patterns sheds light on the substrate preferences of this promiscuous biocatalyst.

真菌非特异性过氧酶(UPOs)在合成化学领域的应用日益广泛。其中特别引人关注的是来自轮叶真菌(Marasmius rotula)的过氧化物酶(MroUPO),它具有独特的氧官能化功能,包括烷烃的末端羟基化、脂肪酸的α-氧化和皮质类固醇的 C-C 裂解。然而,由于缺乏进行定向进化的异源表达系统,阻碍了它在实际应用中的工程化。在这里,我们介绍了 MroUPO 的近源直向同源物,即来自 Marasmius wettsteinii 的 UPO 基因(MweUPO-1),它与 MroUPO 具有相似的反应谱,我们为其建立了一个基于串联酵母表达的定向进化平台。重组 MweUPO-1 在生物反应器中以高滴度(0.7 克/升)生产,并在生化和原子水平上进行了表征。分辨率高达 1.6 Å 的浸泡晶体学实验与反应模式分析相结合,揭示了这种杂交生物催化剂的底物偏好。
{"title":"Structural Insights and Reaction Profile of a New Unspecific Peroxygenase from <i>Marasmius wettsteinii</i> Produced in a Tandem-Yeast Expression System.","authors":"Israel Sánchez-Moreno, Angela Fernandez-Garcia, Ivan Mateljak, Patricia Gomez de Santos, Martin Hofrichter, Harald Kellner, Julia Sanz-Aparicio, Miguel Alcalde","doi":"10.1021/acschembio.4c00504","DOIUrl":"10.1021/acschembio.4c00504","url":null,"abstract":"<p><p>Fungal unspecific peroxygenases (UPOs) are gaining momentum in synthetic chemistry. Of special interest is the UPO from <i>Marasmius rotula</i> (<i>Mro</i>UPO), which shows an exclusive repertoire of oxyfunctionalizations, including the terminal hydroxylation of alkanes, the α-oxidation of fatty acids and the C-C cleavage of corticosteroids. However, the lack of heterologous expression systems to perform directed evolution has impeded its engineering for practical applications. Here, we introduce a close ortholog of <i>Mro</i>UPO, a UPO gene from <i>Marasmius wettsteinii</i> (<i>Mwe</i>UPO-1), that has a similar reaction profile to <i>Mro</i>UPO and for which we have set up a directed evolution platform based on tandem-yeast expression. Recombinant <i>Mwe</i>UPO-1 was produced at high titers in the bioreactor (0.7 g/L) and characterized at the biochemical and atomic levels. The conjunction of soaking crystallographic experiments at a resolution up to 1.6 Å together with the analysis of reaction patterns sheds light on the substrate preferences of this promiscuous biocatalyst.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142378784","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
What Is the Crystallographic Resolution of Structural Models of Proteins Generated with AlphaFold2? 用 AlphaFold2 生成的蛋白质结构模型的晶体分辨率是多少?
IF 3.5 2区 生物学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-18 Epub Date: 2024-09-24 DOI: 10.1021/acschembio.4c00376
Oliviero Carugo

Recent advancements in AI-driven computational modeling, especially AlphaFold2, have revolutionized the prediction of biological macromolecule structures. AlphaFold2 enabled accurate predictions of structural domains and complex arrangements. However, computational models lack a clear metric for accuracy. This study explores whether computational models can match the crystallographic resolution of crystal structures. By comparing distances between atoms in models and crystal structures using t tests, it was found that AlphaFold2 models are comparable to high-resolution crystal structures (1.1 to 1.5 Å). While these models exhibit exceptional quality, their accuracy is lower than the crystal structure with resolutions better than 1 Å.

人工智能计算建模的最新进展,尤其是 AlphaFold2,彻底改变了生物大分子结构的预测。AlphaFold2 能够准确预测结构域和复杂排列。然而,计算模型缺乏明确的准确性衡量标准。本研究探讨了计算模型能否与晶体结构的晶体学分辨率相匹配。通过使用 t 检验比较模型和晶体结构中原子间的距离,发现 AlphaFold2 模型可与高分辨率晶体结构(1.1 至 1.5 Å)相媲美。虽然这些模型显示出卓越的质量,但其精确度低于分辨率高于 1 Å 的晶体结构。
{"title":"What Is the Crystallographic Resolution of Structural Models of Proteins Generated with AlphaFold2?","authors":"Oliviero Carugo","doi":"10.1021/acschembio.4c00376","DOIUrl":"10.1021/acschembio.4c00376","url":null,"abstract":"<p><p>Recent advancements in AI-driven computational modeling, especially AlphaFold2, have revolutionized the prediction of biological macromolecule structures. AlphaFold2 enabled accurate predictions of structural domains and complex arrangements. However, computational models lack a clear metric for accuracy. This study explores whether computational models can match the crystallographic resolution of crystal structures. By comparing distances between atoms in models and crystal structures using <i>t</i> tests, it was found that AlphaFold2 models are comparable to high-resolution crystal structures (1.1 to 1.5 Å). While these models exhibit exceptional quality, their accuracy is lower than the crystal structure with resolutions better than 1 Å.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337254","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
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