首页 > 最新文献

ACS Chemical Biology最新文献

英文 中文
Antimicrobial Lasso Peptide Cloacaenodin Utilizes a Unique TonB-Dependent Transporter to Access Susceptible Bacteria 抗菌拉索肽 Cloacaenodin 利用独特的 TonB 依赖性转运体进入易感细菌。
IF 4 2区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-03-25 DOI: 10.1021/acschembio.4c00009
Drew V. Carson, Reecan J. Juarez, Truc Do, Zhongyue J. Yang and A. James Link*, 

The development of new antimicrobial agents effective against Gram-negative bacteria remains a major challenge in drug discovery. The lasso peptide cloacaenodin has potent antimicrobial activity against multiple strains in the Enterobacter genus, one of the ESKAPE pathogens. Here, we show that cloacaenodin uses a previously uncharacterized TonB-dependent transporter, which we name CloU, to cross the outer membrane (OM) of susceptible bacteria. Inner membrane transport is mediated by the protein SbmA. CloU is distinct from the known OM transporters (FhuA and PupB) utilized by other antimicrobial lasso peptides and thus offers important insight into the spectrum of activity of cloacaenodin. Using knowledge of the transport pathway to predict other cloacaenodin-susceptible strains, we demonstrate the activity of cloacaenodin against clinical isolates of Enterobacter and of a Kluyvera strain. Further, we use molecular dynamics simulations and mutagenesis of CloU to explain the variation in cloacaenodin susceptibility observed across different strains of Enterobacter. This work expands the currently limited understanding of lasso peptide uptake and advances the potential of cloacaenodin as an antibiotic.

开发能有效对抗革兰氏阴性菌的新型抗菌剂仍然是药物研发领域的一大挑战。拉索肽 cloacaenodin 对 ESKAPE 致病菌之一的肠杆菌属的多个菌株具有强效抗菌活性。在这里,我们发现,氯卡诺定利用一种以前未定性的依赖于 TonB 的转运体(我们将其命名为 CloU)穿过易感细菌的外膜(OM)。内膜转运由蛋白质 SbmA 介导。CloU 有别于其他抗菌拉索肽所利用的已知外膜转运体(FhuA 和 PupB),因此为了解氯雷诺定的活性范围提供了重要信息。利用对转运途径的了解来预测其他对氯雷他定敏感的菌株,我们证明了氯雷他定对临床分离的肠杆菌和克鲁伊韦拉菌株的活性。此外,我们还利用分子动力学模拟和 CloU 诱变来解释不同肠杆菌菌株对氯雷他定的敏感性差异。这项工作拓展了目前对拉索肽摄取的有限了解,并提高了氯氰碘柳胺作为抗生素的潜力。
{"title":"Antimicrobial Lasso Peptide Cloacaenodin Utilizes a Unique TonB-Dependent Transporter to Access Susceptible Bacteria","authors":"Drew V. Carson,&nbsp;Reecan J. Juarez,&nbsp;Truc Do,&nbsp;Zhongyue J. Yang and A. James Link*,&nbsp;","doi":"10.1021/acschembio.4c00009","DOIUrl":"10.1021/acschembio.4c00009","url":null,"abstract":"<p >The development of new antimicrobial agents effective against Gram-negative bacteria remains a major challenge in drug discovery. The lasso peptide cloacaenodin has potent antimicrobial activity against multiple strains in the <i>Enterobacter</i> genus, one of the ESKAPE pathogens. Here, we show that cloacaenodin uses a previously uncharacterized TonB-dependent transporter, which we name CloU, to cross the outer membrane (OM) of susceptible bacteria. Inner membrane transport is mediated by the protein SbmA. CloU is distinct from the known OM transporters (FhuA and PupB) utilized by other antimicrobial lasso peptides and thus offers important insight into the spectrum of activity of cloacaenodin. Using knowledge of the transport pathway to predict other cloacaenodin-susceptible strains, we demonstrate the activity of cloacaenodin against clinical isolates of <i>Enterobacter</i> and of a <i>Kluyvera</i> strain. Further, we use molecular dynamics simulations and mutagenesis of CloU to explain the variation in cloacaenodin susceptibility observed across different strains of <i>Enterobacter</i>. This work expands the currently limited understanding of lasso peptide uptake and advances the potential of cloacaenodin as an antibiotic.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-03-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140287584","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
DNA-Encoded Library Technology─A Catalyst for Covalent Ligand Discovery DNA 编码文库技术--共价配体发现的催化剂。
IF 4 2区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-03-25 DOI: 10.1021/acschembio.3c00803
Paige Dickson*, 

The identification of novel covalent ligands for therapeutic purposes has long depended on serendipity, with dedicated hit finding techniques emerging only in the early 2000s. Advances in chemoproteomics have enabled robust characterization of putative drugs to derisk the unique liabilities associated with covalent hit molecules, leading to a renewed interest in this targeting modality. DNA-encoded library (DEL) technology has similarly emerged over the past two decades as a highly efficient method to identify new chemical equity toward protein targets of interest. A number of commercial and academic groups have reported methods in covalent DEL synthesis and hit identification; however, it is evident that there is still much to be done to fully realize the power of this technology for covalent ligand discovery. This perspective will explore the current approaches in covalent DEL technology and reflect on the next steps to advance this field.

长期以来,用于治疗目的的新型共价配体的鉴定一直依赖于偶然性,而专门的命中发现技术直到本世纪初才出现。化学蛋白质组学的进步使人们能够对可能的药物进行强有力的特征描述,从而发现与共价配体分子相关的独特缺陷,从而重新激发了人们对这种靶向方式的兴趣。DNA 编码文库(DEL)技术在过去二十年中同样崭露头角,成为一种高效的方法,用于鉴定针对感兴趣的蛋白质靶点的新化学权益。一些商业和学术团体已经报道了共价 DEL 合成和命中识别方法;然而,要充分发挥这项技术在共价配体发现方面的威力,显然还有许多工作要做。本视角将探讨共价 DEL 技术的现有方法,并思考推动这一领域发展的下一步措施。
{"title":"DNA-Encoded Library Technology─A Catalyst for Covalent Ligand Discovery","authors":"Paige Dickson*,&nbsp;","doi":"10.1021/acschembio.3c00803","DOIUrl":"10.1021/acschembio.3c00803","url":null,"abstract":"<p >The identification of novel covalent ligands for therapeutic purposes has long depended on serendipity, with dedicated hit finding techniques emerging only in the early 2000s. Advances in chemoproteomics have enabled robust characterization of putative drugs to derisk the unique liabilities associated with covalent hit molecules, leading to a renewed interest in this targeting modality. DNA-encoded library (DEL) technology has similarly emerged over the past two decades as a highly efficient method to identify new chemical equity toward protein targets of interest. A number of commercial and academic groups have reported methods in covalent DEL synthesis and hit identification; however, it is evident that there is still much to be done to fully realize the power of this technology for covalent ligand discovery. This perspective will explore the current approaches in covalent DEL technology and reflect on the next steps to advance this field.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-03-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140287585","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 of a Gut Bacterial Metabolic Pathway that Drives α-Synuclein Aggregation 发现驱动α-突触核蛋白聚集的肠道细菌代谢途径
IF 4 2区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-03-22 DOI: 10.1021/acschembio.4c00095
Lizett Ortiz de Ora, Julia M. Balsamo, Kylie S. Uyeda and Elizabeth N. Bess*, 

Parkinson’s disease (PD) etiology is associated with aggregation and accumulation of α-synuclein (α-syn) proteins in midbrain dopaminergic neurons. Emerging evidence suggests that in certain subtypes of PD, α-syn aggregates originate in the gut and subsequently spread to the brain. However, mechanisms that instigate α-syn aggregation in the gut have remained elusive. In the brain, the aggregation of α-syn is induced by oxidized dopamine. Such a mechanism has not been explored in the context of the gastrointestinal tract, a niche harboring 46% of the body’s dopamine reservoirs. Here, we report that Enterobacteriaceae, a bacterial family prevalent in human gut microbiotas, induce α-syn aggregation. More specifically, our in vitro data indicate that respiration of nitrate by Escherichia coli K-12, which results in production of nitrite that mediates oxidation of Fe2+ to Fe3+, creates an oxidizing redox potential. These oxidizing conditions enabled the formation of dopamine-derived quinones and α-syn aggregates. Exposing nitrite, but not nitrate, to enteroendocrine STC-1 cells induced aggregation of α-syn that is natively expressed in these cells, which line the intestinal tract. Taken together, our findings indicate that bacterial nitrate reduction may be critical for initiating intestinal α-syn aggregation.

帕金森病(PD)的病因与中脑多巴胺能神经元中α-突触核蛋白(α-syn)蛋白的聚集和堆积有关。新的证据表明,在某些亚型的帕金森病中,α-syn 聚合体起源于肠道,随后扩散到大脑。然而,促使α-syn在肠道内聚集的机制仍然难以捉摸。在大脑中,氧化多巴胺会诱导α-syn聚集。这种机制尚未在胃肠道中得到探索,而胃肠道是人体 46% 的多巴胺储存库。在这里,我们报告了肠杆菌科细菌--一种普遍存在于人体肠道微生物群中的细菌--会诱导α-syn聚集。更具体地说,我们的体外数据表明,大肠杆菌 K-12 对硝酸盐的呼吸作用会产生亚硝酸盐,从而介导 Fe2+ 氧化为 Fe3+,形成氧化还原电位。这些氧化条件使多巴胺衍生的醌类物质和 α-syn 聚集体得以形成。将亚硝酸盐(而非硝酸盐)暴露于肠道内分泌 STC-1 细胞可诱导 α-syn 的聚集,α-syn 在这些细胞中原生表达。综上所述,我们的研究结果表明,细菌的硝酸盐还原可能是引发肠道α-syn聚集的关键。
{"title":"Discovery of a Gut Bacterial Metabolic Pathway that Drives α-Synuclein Aggregation","authors":"Lizett Ortiz de Ora,&nbsp;Julia M. Balsamo,&nbsp;Kylie S. Uyeda and Elizabeth N. Bess*,&nbsp;","doi":"10.1021/acschembio.4c00095","DOIUrl":"10.1021/acschembio.4c00095","url":null,"abstract":"<p >Parkinson’s disease (PD) etiology is associated with aggregation and accumulation of α-synuclein (α-syn) proteins in midbrain dopaminergic neurons. Emerging evidence suggests that in certain subtypes of PD, α-syn aggregates originate in the gut and subsequently spread to the brain. However, mechanisms that instigate α-syn aggregation in the gut have remained elusive. In the brain, the aggregation of α-syn is induced by oxidized dopamine. Such a mechanism has not been explored in the context of the gastrointestinal tract, a niche harboring 46% of the body’s dopamine reservoirs. Here, we report that <i>Enterobacteriaceae</i>, a bacterial family prevalent in human gut microbiotas, induce α-syn aggregation. More specifically, our <i>in vitro</i> data indicate that respiration of nitrate by <i>Escherichia coli</i> K-12, which results in production of nitrite that mediates oxidation of Fe<sup>2+</sup> to Fe<sup>3+</sup>, creates an oxidizing redox potential. These oxidizing conditions enabled the formation of dopamine-derived quinones and α-syn aggregates. Exposing nitrite, but not nitrate, to enteroendocrine STC-1 cells induced aggregation of α-syn that is natively expressed in these cells, which line the intestinal tract. Taken together, our findings indicate that bacterial nitrate reduction may be critical for initiating intestinal α-syn aggregation.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-03-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/epdf/10.1021/acschembio.4c00095","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140183065","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
Characterization of Chemical Interactions between Clinical Drugs and the Oral Bacterium, Corynebacterium matruchotii, via Bioactivity-HiTES 通过 Bioactivity-HiTES 表征临床药物与口腔杆菌 Corynebacterium matruchotii 之间的化学相互作用。
IF 4 2区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-03-21 DOI: 10.1021/acschembio.3c00798
Da Yeong Lee, Jonghwan Kim, Gyu Sung Lee, Sehwan Park, Jeongwon Song, Bum Soo Lee, Seoung Rak Lee, Ki Hyun Kim and Chung Sub Kim*, 

In the field of natural product research, the rediscovery of already-known compounds is one of the significant issues hindering new drug development. Recently, an innovative approach called bioactivity-HiTES has been developed to overcome this limitation, and several new bioactive metabolites have been successfully characterized by this method. In this study, we applied bioactivity-HiTES to Corynebacterium matruchotii, the human oral bacterium, with 3120 clinical drugs as potential elicitors. As a result, we identified two cryptic metabolites, methylindole-3-acetate (MIAA) and indole-3-acetic acid (IAA), elicited by imidafenacin, a urinary antispasmodic drug approved by the Japanese Pharmaceuticals and Medical Devices Agency (PMDA). MIAA showed weak antibacterial activity against a pulmonary disease-causing Mycobacterium conceptionense with an IC50 value of 185.7 μM. Unexpectedly, we also found that C. matruchotii metabolized fludarabine phosphate, a USFDA-approved anticancer drug, to 2-fluoroadenine which displayed moderate antibacterial activity against both Bacillus subtilis and Escherichia coli, with IC50 values of 8.9 and 20.1 μM, respectively. Finally, acelarin, a prodrug of the anticancer drug gemcitabine, was found to exhibit unreported antibacterial activity against B. subtilis with an IC50 value of 33.6 μM through the bioactivity-HiTES method as well. These results indicate that bioactivity-HiTES can also be applied to discover biotransformed products in addition to finding cryptic metabolites in microbes.

在天然产物研究领域,重新发现已知化合物是阻碍新药开发的重要问题之一。最近,一种被称为生物活性-HiTES 的创新方法被开发出来以克服这一限制,并通过这种方法成功鉴定了几种新的生物活性代谢物。在本研究中,我们将生物活性-HiTES 应用于人类口腔细菌 Corynebacterium matruchotii,并将 3120 种临床药物作为潜在的诱导剂。结果,我们发现了两种隐性代谢物,即甲基吲哚-3-乙酸酯(MIAA)和吲哚-3-乙酸(IAA),它们是由日本药品和医疗器械管理局(PMDA)批准的一种解痉药物咪达那新激发的。MIAA 对肺部致病分枝杆菌的抗菌活性较弱,IC50 值为 185.7 μM。意想不到的是,我们还发现 C. matruchotii 还能将美国食品与药物管理局批准的抗癌药物磷酸氟达拉滨代谢为 2-氟腺嘌呤,后者对枯草杆菌和大肠杆菌具有中等抗菌活性,IC50 值分别为 8.9 和 20.1 μM。最后,通过生物活性-HiTES 方法,还发现抗癌药物吉西他滨的原药 acelarin 对枯草杆菌具有未报道的抗菌活性,IC50 值为 33.6 μM。这些结果表明,生物活性-HiTES 除了用于发现微生物中的隐性代谢产物外,还可用于发现生物转化产物。
{"title":"Characterization of Chemical Interactions between Clinical Drugs and the Oral Bacterium, Corynebacterium matruchotii, via Bioactivity-HiTES","authors":"Da Yeong Lee,&nbsp;Jonghwan Kim,&nbsp;Gyu Sung Lee,&nbsp;Sehwan Park,&nbsp;Jeongwon Song,&nbsp;Bum Soo Lee,&nbsp;Seoung Rak Lee,&nbsp;Ki Hyun Kim and Chung Sub Kim*,&nbsp;","doi":"10.1021/acschembio.3c00798","DOIUrl":"10.1021/acschembio.3c00798","url":null,"abstract":"<p >In the field of natural product research, the rediscovery of already-known compounds is one of the significant issues hindering new drug development. Recently, an innovative approach called bioactivity-HiTES has been developed to overcome this limitation, and several new bioactive metabolites have been successfully characterized by this method. In this study, we applied bioactivity-HiTES to <i>Corynebacterium matruchotii</i>, the human oral bacterium, with 3120 clinical drugs as potential elicitors. As a result, we identified two cryptic metabolites, methylindole-3-acetate (MIAA) and indole-3-acetic acid (IAA), elicited by imidafenacin, a urinary antispasmodic drug approved by the Japanese Pharmaceuticals and Medical Devices Agency (PMDA). MIAA showed weak antibacterial activity against a pulmonary disease-causing <i>Mycobacterium conceptionense</i> with an IC<sub>50</sub> value of 185.7 μM. Unexpectedly, we also found that <i>C. matruchotii</i> metabolized fludarabine phosphate, a USFDA-approved anticancer drug, to 2-fluoroadenine which displayed moderate antibacterial activity against both <i>Bacillus subtilis</i> and <i>Escherichia coli</i>, with IC<sub>50</sub> values of 8.9 and 20.1 μM, respectively. Finally, acelarin, a prodrug of the anticancer drug gemcitabine, was found to exhibit unreported antibacterial activity against <i>B. subtilis</i> with an IC<sub>50</sub> value of 33.6 μM through the bioactivity-HiTES method as well. These results indicate that bioactivity-HiTES can also be applied to discover biotransformed products in addition to finding cryptic metabolites in microbes.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140183063","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
Integrated Proteomics Characterization of NLRP3 Inflammasome Inhibitor MCC950 in Monocytic Cell Line Confirms Direct MCC950 Engagement with Endogenous NLRP3 单核细胞系中 NLRP3 炎症小体抑制剂 MCC950 的综合蛋白质组学特征证实 MCC950 直接参与内源性 NLRP3。
IF 4 2区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-03-21 DOI: 10.1021/acschembio.3c00777
Heng Zhao, Praveen Kumar, Tiago Jose Paschoal Sobreira, Mackenzie Smith, Steven Novick, Anders Johansson, Anna Luchniak, Andrew Zhang, Kevin J Woollard, Niklas Larsson* and Aarti Kawatkar*, 

Inhibition of the NLRP3 inflammasome is a promising strategy for the development of new treatments for inflammatory diseases. MCC950 is a potent and selective small-molecule inhibitor of the NLRP3 pathway and has been validated in numerous species and disease models. Although the capacity of MCC950 to block NLRP3 signaling is well-established, it is still critical to identify the mechanism of action and molecular targets of MCC950 to inform and derisk drug development. Quantitative proteomics performed in disease-relevant systems provides a powerful method to study both direct and indirect pharmacological responses to small molecules to elucidate the mechanism of action and confirm target engagement. A comprehensive target deconvolution campaign requires the use of complementary chemical biology techniques. Here we applied two orthogonal chemical biology techniques: compressed Cellular Thermal Shift Assay (CETSA) and photoaffinity labeling chemoproteomics, performed under biologically relevant conditions with LPS-primed THP-1 cells, thereby deconvoluting, for the first time, the molecular targets of MCC950 using chemical biology techniques. In-cell chemoproteomics with inlysate CETSA confirmed the suspected mechanism as the disruption of inflammasome formation via NLRP3. Further cCETSA (c indicates compressed) in live cells mapped the stabilization of NLRP3 inflammasome pathway proteins, highlighting modulation of the targeted pathway. This is the first evidence of direct MCC950 engagement with endogenous NLRP3 in a human macrophage cellular system using discovery proteomics chemical biology techniques, providing critical information for inflammasome studies.

抑制 NLRP3 炎性体是开发治疗炎症性疾病新方法的一种有前途的策略。MCC950 是一种强效、选择性的 NLRP3 通路小分子抑制剂,已在多个物种和疾病模型中得到验证。虽然 MCC950 阻断 NLRP3 信号传导的能力已得到证实,但确定 MCC950 的作用机制和分子靶点对于药物开发的信息和风险评估仍然至关重要。在疾病相关系统中进行的定量蛋白质组学研究提供了一种强大的方法来研究小分子的直接和间接药理反应,以阐明作用机制并确认靶点参与。全面的靶点解旋活动需要使用互补的化学生物学技术。在这里,我们应用了两种正交的化学生物学技术:压缩细胞热转移分析法(CETSA)和光亲和标记化学蛋白质组学,在生物相关条件下用 LPS 激发的 THP-1 细胞进行,从而首次利用化学生物学技术解构了 MCC950 的分子靶点。使用析出液 CETSA 进行细胞内化学蛋白质组学研究,证实了通过 NLRP3 破坏炎性体形成的可疑机制。在活细胞中进一步使用 cCETSA(c 表示压缩)绘制了 NLRP3 炎症小体通路蛋白的稳定图,突显了对目标通路的调节。这是利用发现蛋白质组学化学生物学技术在人类巨噬细胞系统中首次发现 MCC950 与内源性 NLRP3 直接接触的证据,为炎性体研究提供了关键信息。
{"title":"Integrated Proteomics Characterization of NLRP3 Inflammasome Inhibitor MCC950 in Monocytic Cell Line Confirms Direct MCC950 Engagement with Endogenous NLRP3","authors":"Heng Zhao,&nbsp;Praveen Kumar,&nbsp;Tiago Jose Paschoal Sobreira,&nbsp;Mackenzie Smith,&nbsp;Steven Novick,&nbsp;Anders Johansson,&nbsp;Anna Luchniak,&nbsp;Andrew Zhang,&nbsp;Kevin J Woollard,&nbsp;Niklas Larsson* and Aarti Kawatkar*,&nbsp;","doi":"10.1021/acschembio.3c00777","DOIUrl":"10.1021/acschembio.3c00777","url":null,"abstract":"<p >Inhibition of the NLRP3 inflammasome is a promising strategy for the development of new treatments for inflammatory diseases. MCC950 is a potent and selective small-molecule inhibitor of the NLRP3 pathway and has been validated in numerous species and disease models. Although the capacity of MCC950 to block NLRP3 signaling is well-established, it is still critical to identify the mechanism of action and molecular targets of MCC950 to inform and derisk drug development. Quantitative proteomics performed in disease-relevant systems provides a powerful method to study both direct and indirect pharmacological responses to small molecules to elucidate the mechanism of action and confirm target engagement. A comprehensive target deconvolution campaign requires the use of complementary chemical biology techniques. Here we applied two orthogonal chemical biology techniques: compressed Cellular Thermal Shift Assay (CETSA) and photoaffinity labeling chemoproteomics, performed under biologically relevant conditions with LPS-primed THP-1 cells, thereby deconvoluting, for the first time, the molecular targets of MCC950 using chemical biology techniques. In-cell chemoproteomics with inlysate CETSA confirmed the suspected mechanism as the disruption of inflammasome formation via NLRP3. Further cCETSA (c indicates compressed) in live cells mapped the stabilization of NLRP3 inflammasome pathway proteins, highlighting modulation of the targeted pathway. This is the first evidence of direct MCC950 engagement with endogenous NLRP3 in a human macrophage cellular system using discovery proteomics chemical biology techniques, providing critical information for inflammasome studies.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140173524","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 of a Covalent Inhibitor Selectively Targeting the Autophosphorylation Site of c-Src Kinase 发现一种选择性靶向 c-Src 激酶自身磷酸化位点的共价抑制剂
IF 4 2区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-03-21 DOI: 10.1021/acschembio.4c00048
Huimin Zhang, Dounan Xu, Hongchan Huang, Hao Jiang, Linghao Hu, Liping Liu, Ge Sun, Jing Gao, Yuanqing Li, Cuicui Xia, Shijie Chen, Hu Zhou, Xiangqian Kong*, Mingliang Wang* and Cheng Luo*, 

Nonreceptor tyrosine kinase c-Src plays a crucial role in cell signaling and contributes to tumor progression. However, the development of selective c-Src inhibitors turns out to be challenging. In our previous study, we performed posttranslational modification-inspired drug design (PTMI-DD) to provide a plausible way for designing selective kinase inhibitors. In this study, after identifying a unique pocket comprising a less conserved cysteine and an autophosphorylation site in c-Src as well as a promiscuous covalent inhibitor, chemical optimization was performed to obtain (R)-LW-Srci-8 with nearly 75-fold improved potency (IC50 = 35.83 ± 7.21 nM). Crystallographic studies revealed the critical C–F···C═O interactions that may contribute to tight binding. The kinact and Ki values validated the improved binding affinity and decreased warhead reactivity of (R)-LW-Srci-8 for c-Src. Notably, in vitro tyrosine kinase profiling and cellular activity-based protein profiling (ABPP) cooperatively indicated a specific inhibition of c-Src by (R)-LW-Srci-8. Intriguingly, (R)-LW-Srci-8 preferentially binds to inactive c-Src with unphosphorylated Y419 both in vitro and in cells, subsequently disrupting the autophosphorylation. Collectively, our study demonstrated the feasibility of developing selective kinase inhibitors by cotargeting a nucleophilic residue and a posttranslational modification site and providing a chemical probe for c-Src functional studies.

非受体酪氨酸激酶 c-Src 在细胞信号传导过程中发挥着至关重要的作用,并对肿瘤的发展起到推波助澜的作用。然而,开发选择性 c-Src 抑制剂是一项挑战。在之前的研究中,我们进行了翻译后修饰启发药物设计(PTMI-DD),为设计选择性激酶抑制剂提供了一种可行的方法。在本研究中,在确定了一个由 c-Src 中一个不太保守的半胱氨酸和一个自磷酸化位点组成的独特口袋以及一种杂合共价抑制剂后,我们进行了化学优化,得到了 (R)-LW-Srci-8,其效力提高了近 75 倍(IC50 = 35.83 ± 7.21 nM)。晶体学研究揭示了可能导致紧密结合的关键 C-F-C═O 相互作用。Kinact和Ki值验证了(R)-LW-Srci-8与c-Src结合亲和力的提高和弹头反应性的降低。值得注意的是,体外酪氨酸激酶分析和基于细胞活性的蛋白质分析(ABPP)共同表明,(R)-LW-Srci-8 对 c-Src 有特异性抑制作用。耐人寻味的是,(R)-LW-Srci-8 在体外和细胞中都优先与未磷酸化 Y419 的非活性 c-Src 结合,随后破坏其自身磷酸化。总之,我们的研究证明了通过共靶向亲核残基和翻译后修饰位点开发选择性激酶抑制剂的可行性,并为 c-Src 功能研究提供了化学探针。
{"title":"Discovery of a Covalent Inhibitor Selectively Targeting the Autophosphorylation Site of c-Src Kinase","authors":"Huimin Zhang,&nbsp;Dounan Xu,&nbsp;Hongchan Huang,&nbsp;Hao Jiang,&nbsp;Linghao Hu,&nbsp;Liping Liu,&nbsp;Ge Sun,&nbsp;Jing Gao,&nbsp;Yuanqing Li,&nbsp;Cuicui Xia,&nbsp;Shijie Chen,&nbsp;Hu Zhou,&nbsp;Xiangqian Kong*,&nbsp;Mingliang Wang* and Cheng Luo*,&nbsp;","doi":"10.1021/acschembio.4c00048","DOIUrl":"10.1021/acschembio.4c00048","url":null,"abstract":"<p >Nonreceptor tyrosine kinase c-Src plays a crucial role in cell signaling and contributes to tumor progression. However, the development of selective c-Src inhibitors turns out to be challenging. In our previous study, we performed posttranslational modification-inspired drug design (PTMI-DD) to provide a plausible way for designing selective kinase inhibitors. In this study, after identifying a unique pocket comprising a less conserved cysteine and an autophosphorylation site in c-Src as well as a promiscuous covalent inhibitor, chemical optimization was performed to obtain <b>(</b><i><b>R</b></i><b>)-LW-Srci-8</b> with nearly 75-fold improved potency (IC<sub>50</sub> = 35.83 ± 7.21 nM). Crystallographic studies revealed the critical C–F···C═O interactions that may contribute to tight binding. The <i>k</i><sub>inact</sub> and <i>K<sub>i</sub></i> values validated the improved binding affinity and decreased warhead reactivity of <b>(</b><i><b>R</b></i><b>)-LW-Srci-8</b> for c-Src. Notably, in vitro tyrosine kinase profiling and cellular activity-based protein profiling (ABPP) cooperatively indicated a specific inhibition of c-Src by <b>(</b><i><b>R</b></i><b>)-LW-Srci-8</b>. Intriguingly, <b>(</b><i><b>R</b></i><b>)-LW-Srci-8</b> preferentially binds to inactive c-Src with unphosphorylated Y419 both in vitro and in cells, subsequently disrupting the autophosphorylation. Collectively, our study demonstrated the feasibility of developing selective kinase inhibitors by cotargeting a nucleophilic residue and a posttranslational modification site and providing a chemical probe for c-Src functional studies.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140183064","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
Lipid-Restricted Culture Media Reveal Unexpected Cancer Cell Sensitivities 限脂培养基揭示了意想不到的癌细胞敏感性
IF 4 2区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-03-20 DOI: 10.1021/acschembio.3c00699
Ralston B. Goldfarb, Matthew J. Atala Pleshinger, David F. Yan and Drew J. Adams*, 

Cancer cell culture models frequently rely on fetal bovine serum as a source of protein and lipid factors that support cell survival and proliferation; however, serum-containing media imperfectly mimic the in vivo cancer environment. Recent studies suggest that typical serum-containing cell culture conditions can mask cancer dependencies, for example, on cholesterol biosynthesis enzymes, that exist in vivo and emerge when cells are cultured in media that provide more realistic levels of lipids. Here, we describe a high-throughput screen that identified fenretinide and ivermectin as small molecules whose cytotoxicity is greatly enhanced in lipid-restricted media formulations. The mechanism of action studies indicates that ivermectin-induced cell death involves oxidative stress, while fenretinide likely targets delta 4-desaturase, sphingolipid 1, a lipid desaturase necessary for ceramide synthesis, to induce cell death. Notably, both fenretinide and ivermectin have previously demonstrated in vivo anticancer efficacy despite their low cytotoxicity under typical cell culture conditions. These studies suggest ceramide synthesis as a targetable vulnerability of cancer cells cultured under lipid-restricted conditions and reveal a general screening strategy for identifying additional cancer dependencies masked by the superabundance of medium lipids.

癌症细胞培养模型通常依赖胎牛血清作为支持细胞存活和增殖的蛋白质和脂质因子的来源;然而,含血清培养基并不能完全模拟体内癌症环境。最近的研究表明,典型的含血清细胞培养条件可能会掩盖癌症依赖性,例如对胆固醇生物合成酶的依赖性。在这里,我们介绍了一种高通量筛选方法,筛选出了芬瑞替尼和伊维菌素这两种小分子,它们在脂质限制培养基配方中的细胞毒性大大增强。作用机制研究表明,伊维菌素诱导的细胞死亡涉及氧化应激,而芬雷肽则可能以δ 4-去饱和酶、鞘脂 1(一种合成神经酰胺所必需的脂质去饱和酶)为靶标诱导细胞死亡。值得注意的是,尽管芬瑞替尼和伊维菌素在典型的细胞培养条件下细胞毒性较低,但这两种药物以前都显示出体内抗癌功效。这些研究表明,神经酰胺合成是在脂质受限条件下培养的癌细胞的一个可靶向的弱点,并揭示了一种通用筛选策略,可用于识别被介质脂质的超丰度所掩盖的其他癌症依赖性。
{"title":"Lipid-Restricted Culture Media Reveal Unexpected Cancer Cell Sensitivities","authors":"Ralston B. Goldfarb,&nbsp;Matthew J. Atala Pleshinger,&nbsp;David F. Yan and Drew J. Adams*,&nbsp;","doi":"10.1021/acschembio.3c00699","DOIUrl":"10.1021/acschembio.3c00699","url":null,"abstract":"<p >Cancer cell culture models frequently rely on fetal bovine serum as a source of protein and lipid factors that support cell survival and proliferation; however, serum-containing media imperfectly mimic the in vivo cancer environment. Recent studies suggest that typical serum-containing cell culture conditions can mask cancer dependencies, for example, on cholesterol biosynthesis enzymes, that exist in vivo and emerge when cells are cultured in media that provide more realistic levels of lipids. Here, we describe a high-throughput screen that identified fenretinide and ivermectin as small molecules whose cytotoxicity is greatly enhanced in lipid-restricted media formulations. The mechanism of action studies indicates that ivermectin-induced cell death involves oxidative stress, while fenretinide likely targets delta 4-desaturase, sphingolipid 1, a lipid desaturase necessary for ceramide synthesis, to induce cell death. Notably, both fenretinide and ivermectin have previously demonstrated in vivo anticancer efficacy despite their low cytotoxicity under typical cell culture conditions. These studies suggest ceramide synthesis as a targetable vulnerability of cancer cells cultured under lipid-restricted conditions and reveal a general screening strategy for identifying additional cancer dependencies masked by the superabundance of medium lipids.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-03-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140173525","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
Geometric Antibody Engineering Reveals the Spatial Factor on the Efficacy of Bispecific T Cell Engagers 几何抗体工程揭示了影响双特异性 T 细胞参与因子疗效的空间因素
IF 4 2区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-03-16 DOI: 10.1021/acschembio.3c00728
Yu Zhang, Zhe Yang, Dilizhatai Saimi, Xiaowen Shen, Junqing Ye, Bingke Yu, Noah Pefaur, Justin M. Scheer, Andrew E. Nixon* and Zhixing Chen*, 

Bispecific antibodies (BsAbs) represent an emerging class of biologics that can recognize two different antigens or epitopes. T-cell engagers (TcEs) bind two targets in trans on the cell surface of the effector and target cell to induce proximal immune effects, opening exciting windows for immunotherapies. To date, the engineering of BsAbs has been mainly focused on tuning the molecular weight and valency. However, the effects of spatial factors on the biological functions of BsAbs have been less explored due to the lack of biochemical methods to precisely manipulate protein geometry. Here, we studied the geometric effects of the TcEs. First, by genetically inserting rigidly designed ankyrin repeat proteins into TcEs, we revealed that the efficacy progressively decreased as the spacer distance of the two binding domains increased. Then, we constructed 26 pairs of TcEs with the same size but varying orientations using click chemistry-mediated conjugation at different mutation sites. We found that linear ligation sites play a minor role in modulating cell-killing efficacy. Next, we rendered the TcEs’ advanced topology by cyclization chemistry using the SpyTag/SpyCatcher pair or sortase ligation approaches. Cyclized TcEs were generally more potent than their linear counterparts. Particularly, sortase A cyclized TcEs, bearing a minimal tagging motif, exhibited better cell-killing efficacy in vitro and improved stability both in vitro and in vivo compared to the linear TcE. This work combines modern bioconjugation chemistry and protein engineering tools for antibody engineering, shedding light on the elusive spatial factors of BsAbs functionality.

双特异性抗体(BsAbs)是一类新兴的生物制剂,可以识别两种不同的抗原或表位。双特异性抗体(BsAbs)是一类新兴的生物制剂,能识别两种不同的抗原或表位。T 细胞吞噬因子(TcEs)能反式结合效应细胞和靶细胞表面的两个靶点,诱导近端免疫效应,为免疫疗法打开了令人兴奋的窗口。迄今为止,BsAbs 的工程设计主要集中在分子量和效价的调整上。然而,由于缺乏精确操纵蛋白质几何结构的生化方法,人们对空间因素对 BsAbs 生物功能的影响探索较少。在这里,我们研究了 TcEs 的几何效应。首先,通过在 TcEs 中插入刚性设计的 ankyrin 重复蛋白,我们发现随着两个结合域的间隔距离的增加,药效逐渐降低。然后,我们在不同的突变位点上利用点击化学介导的连接技术构建了 26 对大小相同但方向不同的 TcEs。我们发现,线性连接位点在调节细胞杀伤效力方面作用较小。接下来,我们利用 SpyTag/SpyCatcher 对或分类酶连接方法,通过环化化学作用呈现了 TcEs 的高级拓扑结构。环化 TcEs 一般比线性 TcEs 更有效。特别是,与线性 TcE 相比,带有最小标记基团的分选酶 A 环化 TcE 在体外具有更好的细胞杀伤效力,在体外和体内的稳定性也有所提高。这项研究将现代生物结合化学和蛋白质工程工具结合到抗体工程中,揭示了 BsAbs 功能中难以捉摸的空间因素。
{"title":"Geometric Antibody Engineering Reveals the Spatial Factor on the Efficacy of Bispecific T Cell Engagers","authors":"Yu Zhang,&nbsp;Zhe Yang,&nbsp;Dilizhatai Saimi,&nbsp;Xiaowen Shen,&nbsp;Junqing Ye,&nbsp;Bingke Yu,&nbsp;Noah Pefaur,&nbsp;Justin M. Scheer,&nbsp;Andrew E. Nixon* and Zhixing Chen*,&nbsp;","doi":"10.1021/acschembio.3c00728","DOIUrl":"10.1021/acschembio.3c00728","url":null,"abstract":"<p >Bispecific antibodies (BsAbs) represent an emerging class of biologics that can recognize two different antigens or epitopes. T-cell engagers (TcEs) bind two targets in trans on the cell surface of the effector and target cell to induce proximal immune effects, opening exciting windows for immunotherapies. To date, the engineering of BsAbs has been mainly focused on tuning the molecular weight and valency. However, the effects of spatial factors on the biological functions of BsAbs have been less explored due to the lack of biochemical methods to precisely manipulate protein geometry. Here, we studied the geometric effects of the TcEs. First, by genetically inserting rigidly designed ankyrin repeat proteins into TcEs, we revealed that the efficacy progressively decreased as the spacer distance of the two binding domains increased. Then, we constructed 26 pairs of TcEs with the same size but varying orientations using click chemistry-mediated conjugation at different mutation sites. We found that linear ligation sites play a minor role in modulating cell-killing efficacy. Next, we rendered the TcEs’ advanced topology by cyclization chemistry using the SpyTag/SpyCatcher pair or sortase ligation approaches. Cyclized TcEs were generally more potent than their linear counterparts. Particularly, sortase A cyclized TcEs, bearing a minimal tagging motif, exhibited better cell-killing efficacy in vitro and improved stability both in vitro and in vivo compared to the linear TcE. This work combines modern bioconjugation chemistry and protein engineering tools for antibody engineering, shedding light on the elusive spatial factors of BsAbs functionality.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-03-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140139593","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
Click-Capable Phenanthriplatin Derivatives as Tools to Study Pt(II)-Induced Nucleolar Stress 可单击的菲三铂衍生物作为研究铂(II)诱导的核极应激的工具
IF 4 2区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-03-14 DOI: 10.1021/acschembio.3c00607
Paul D. O’Dowd, Andres S. Guerrero, Katelyn R. Alley, Hannah C. Pigg, Fiona O’Neill, Justine Meiller, Chloe Hobbs, Daniel A. Rodrigues, Brendan Twamley, Finbarr O’Sullivan, Victoria J. DeRose and Darren M. Griffith*, 

It is well established that oxaliplatin, one of the three Pt(II) anticancer drugs approved worldwide, and phenanthriplatin, an important preclinical monofunctional Pt(II) anticancer drug, possess a different mode of action from that of cisplatin and carboplatin, namely, the induction of nucleolar stress. The exact mechanisms that lead to Pt-induced nucleolar stress are, however, still poorly understood. As such, studies aimed at better understanding the biological targets of both oxaliplatin and phenanthriplatin are urgently needed to expand our understanding of Pt-induced nucleolar stress and guide the future design of Pt chemotherapeutics. One approach that has seen great success in the past is the use of Pt-click complexes to study the biological targets of Pt drugs. Herein, we report the synthesis and characterization of the first examples of click-capable phenanthriplatin complexes. Furthermore, through monitoring the relocalization of nucleolar proteins, RNA transcription levels, and DNA damage repair biomarker γH2AX, and by investigating their in vitro cytotoxicity, we show that these complexes successfully mimic the cellular responses observed for phenanthriplatin treatment in the same experiments. The click-capable phenanthriplatin derivatives described here expand the existing library of Pt-click complexes. Significantly they are suitable for studying nucleolar stress mechanisms and further elucidating the biological targets of Pt complexes.

众所周知,奥沙利铂(全球已批准的三种铂(II)抗癌药物之一)和菲铂(一种重要的临床前单功能铂(II)抗癌药物)具有与顺铂和卡铂不同的作用模式,即诱导核极应激。然而,人们对铂诱导核极应激的确切机制仍知之甚少。因此,迫切需要开展研究,以更好地了解奥沙利铂和菲铂的生物靶点,从而扩大我们对铂诱导的核极应激的了解,并指导未来铂类化疗药物的设计。过去取得巨大成功的一种方法是利用铂簇复合物来研究铂类药物的生物靶点。在此,我们报告了首例具有点击能力的菲铂复合物的合成和表征。此外,通过监测核极蛋白的重新定位、RNA转录水平和DNA损伤修复生物标志物γH2AX,以及研究它们的体外细胞毒性,我们表明这些复合物成功地模拟了在相同实验中观察到的菲铂处理的细胞反应。本文描述的具有点击能力的菲铂衍生物扩展了现有的铂点击复合物库。重要的是,它们适用于研究核极应激机制和进一步阐明铂复合物的生物靶标。
{"title":"Click-Capable Phenanthriplatin Derivatives as Tools to Study Pt(II)-Induced Nucleolar Stress","authors":"Paul D. O’Dowd,&nbsp;Andres S. Guerrero,&nbsp;Katelyn R. Alley,&nbsp;Hannah C. Pigg,&nbsp;Fiona O’Neill,&nbsp;Justine Meiller,&nbsp;Chloe Hobbs,&nbsp;Daniel A. Rodrigues,&nbsp;Brendan Twamley,&nbsp;Finbarr O’Sullivan,&nbsp;Victoria J. DeRose and Darren M. Griffith*,&nbsp;","doi":"10.1021/acschembio.3c00607","DOIUrl":"10.1021/acschembio.3c00607","url":null,"abstract":"<p >It is well established that oxaliplatin, one of the three Pt(II) anticancer drugs approved worldwide, and phenanthriplatin, an important preclinical monofunctional Pt(II) anticancer drug, possess a different mode of action from that of cisplatin and carboplatin, namely, the induction of nucleolar stress. The exact mechanisms that lead to Pt-induced nucleolar stress are, however, still poorly understood. As such, studies aimed at better understanding the biological targets of both oxaliplatin and phenanthriplatin are urgently needed to expand our understanding of Pt-induced nucleolar stress and guide the future design of Pt chemotherapeutics. One approach that has seen great success in the past is the use of Pt-click complexes to study the biological targets of Pt drugs. Herein, we report the synthesis and characterization of the first examples of click-capable phenanthriplatin complexes. Furthermore, through monitoring the relocalization of nucleolar proteins, RNA transcription levels, and DNA damage repair biomarker γH2AX, and by investigating their <i>in vitro</i> cytotoxicity, we show that these complexes successfully mimic the cellular responses observed for phenanthriplatin treatment in the same experiments. The click-capable phenanthriplatin derivatives described here expand the existing library of Pt-click complexes. Significantly they are suitable for studying nucleolar stress mechanisms and further elucidating the biological targets of Pt complexes.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/epdf/10.1021/acschembio.3c00607","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140118104","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
Roles of Nucleic Acids in Protein Folding, Aggregation, and Disease 核酸在蛋白质折叠、聚集和疾病中的作用。
IF 4 2区 生物学 Q2 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-03-13 DOI: 10.1021/acschembio.3c00695
Theodore J. Litberg,  and , Scott Horowitz*, 

The role of nucleic acids in protein folding and aggregation is an area of continued research, with relevance to understanding both basic biological processes and disease. In this review, we provide an overview of the trajectory of research on both nucleic acids as chaperones and their roles in several protein misfolding diseases. We highlight key questions that remain on the biophysical and biochemical specifics of how nucleic acids have large effects on multiple proteins’ folding and aggregation behavior and how this pertains to multiple protein misfolding diseases.

核酸在蛋白质折叠和聚集中的作用是一个持续研究的领域,与了解基本生物过程和疾病都有关系。在这篇综述中,我们概述了核酸作为伴侣的研究轨迹及其在几种蛋白质折叠错误疾病中的作用。我们强调了在核酸如何对多种蛋白质的折叠和聚集行为产生巨大影响的生物物理和生物化学细节方面仍然存在的关键问题,以及这与多种蛋白质折叠错误疾病的关系。
{"title":"Roles of Nucleic Acids in Protein Folding, Aggregation, and Disease","authors":"Theodore J. Litberg,&nbsp; and ,&nbsp;Scott Horowitz*,&nbsp;","doi":"10.1021/acschembio.3c00695","DOIUrl":"10.1021/acschembio.3c00695","url":null,"abstract":"<p >The role of nucleic acids in protein folding and aggregation is an area of continued research, with relevance to understanding both basic biological processes and disease. In this review, we provide an overview of the trajectory of research on both nucleic acids as chaperones and their roles in several protein misfolding diseases. We highlight key questions that remain on the biophysical and biochemical specifics of how nucleic acids have large effects on multiple proteins’ folding and aggregation behavior and how this pertains to multiple protein misfolding diseases.</p>","PeriodicalId":11,"journal":{"name":"ACS Chemical Biology","volume":null,"pages":null},"PeriodicalIF":4.0,"publicationDate":"2024-03-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140108374","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
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