首页 > 最新文献

Analytical Chemistry最新文献

英文 中文
Ferrous Tungstate Nanomaterials with Excellent Enzyme-Mimicking Activity to Enhance Lateral Flow Immunoassay Sensitivity.
IF 6.7 1区 化学 Q1 CHEMISTRY, ANALYTICAL Pub Date : 2025-02-11 Epub Date: 2025-01-30 DOI: 10.1021/acs.analchem.4c04761
Xianqing Tang, Hua Zhang, Jinghuang Chen, Donghui Wu, Yu Wang, Jian Sun, Zhenxin Wang, Xiurong Yang

Lateral flow immunochromatography (LFIA) with gold nanoparticles (AuNPs) is widely used in the biomedical field as a rapid and simple in vitro detection technique. However, the conventional AuNP-LFIA has limitations in sensitivity and detection range. In this study, nonprecious metal iron-based bimetallic FeWO4 nanomaterials with convenient and excellent enzyme-mimetic catalytic activities were synthesized by a one-pot hydrothermal method. Here, FeWO4 nanomaterials were combined with C-reactive protein (CRP) detection antibodies to form a novel signal-enhancing probe for the analysis of CRP. The probe further achieves significant signal amplification by catalyzing the oxidation reaction of 3-amino-9-ethylcarbazole in the LFIA assay, thereby improving the sensitivity and accuracy of the assay. The application of FeWO4-based LFIA improved the limit of detection of CRP to 19.38 ng/mL after catalytic amplification, which is approximately 30-fold lower than that of the conventional AuNP-LFIA method. In addition, the method demonstrated good stability and reproducibility, providing a promising and prospective strategy for the early diagnosis of inflammatory diseases.

{"title":"Ferrous Tungstate Nanomaterials with Excellent Enzyme-Mimicking Activity to Enhance Lateral Flow Immunoassay Sensitivity.","authors":"Xianqing Tang, Hua Zhang, Jinghuang Chen, Donghui Wu, Yu Wang, Jian Sun, Zhenxin Wang, Xiurong Yang","doi":"10.1021/acs.analchem.4c04761","DOIUrl":"10.1021/acs.analchem.4c04761","url":null,"abstract":"<p><p>Lateral flow immunochromatography (LFIA) with gold nanoparticles (AuNPs) is widely used in the biomedical field as a rapid and simple in vitro detection technique. However, the conventional AuNP-LFIA has limitations in sensitivity and detection range. In this study, nonprecious metal iron-based bimetallic FeWO<sub>4</sub> nanomaterials with convenient and excellent enzyme-mimetic catalytic activities were synthesized by a one-pot hydrothermal method. Here, FeWO<sub>4</sub> nanomaterials were combined with C-reactive protein (CRP) detection antibodies to form a novel signal-enhancing probe for the analysis of CRP. The probe further achieves significant signal amplification by catalyzing the oxidation reaction of 3-amino-9-ethylcarbazole in the LFIA assay, thereby improving the sensitivity and accuracy of the assay. The application of FeWO<sub>4</sub>-based LFIA improved the limit of detection of CRP to 19.38 ng/mL after catalytic amplification, which is approximately 30-fold lower than that of the conventional AuNP-LFIA method. In addition, the method demonstrated good stability and reproducibility, providing a promising and prospective strategy for the early diagnosis of inflammatory diseases.</p>","PeriodicalId":27,"journal":{"name":"Analytical Chemistry","volume":" ","pages":"2714-2723"},"PeriodicalIF":6.7,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143062305","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Consolidated Microscale Interferon-γ Release Assay with Tip Optofluidic Immunoassay for Dynamic Parallel Diagnosis of Tuberculosis Infection.
IF 6.7 1区 化学 Q1 CHEMISTRY, ANALYTICAL Pub Date : 2025-02-11 Epub Date: 2025-01-31 DOI: 10.1021/acs.analchem.4c05390
Binmao Zhang, Yuzhong Xu, Zhen Huang, Ruihan Li, Tianen Zhu, Shangyan Liang, Hao Huang, Siyan Zhong, Hui Yang, Xudong Fan, Xiaotian Tan, Yujuan Chai

Interferon-γ release assay (IGRA) is one of the most important diagnostic tools for tuberculosis (TB) infection. Despite its high accuracy, conventional IGRA has several drawbacks, including complicated procedures, large blood volume requirements, lengthy incubation times, and difficulties in parallel testing. Efforts have been made to develop miniaturized and highly sensitive biosensors for interferon-γ or to evaluate the specific immune response through microfluidic platforms. However, the need for sophisticated consumables and equipment, as well as the partial experimental design, has limited the application of these advanced techniques in TB diagnosis and disease control. Here, we report the development of a tip optofluidic immunoassay (TOI)-based consolidated microscale IGRA (CM-IGRA) for the dynamic and parallel evaluation of TB infection, refining both the blood incubation and interferon-γ quantification processes. The TOI system comprises 12 microfluidic immuno-reactors and a portable chemiluminescent imaging station, capable of quantifying interferon-γ with high sensitivity (8.00 pg/mL in plasma) and a wide detection range (∼104). The results generated with CM-IGRA achieved 98.39% agreement with the standard IGRA while reducing blood sample consumption to 50 μL per assay (20-fold reduction) and significantly shortening the incubation time from 20 to 10 h. This diagnostic method simplifies operations and improves efficiency for the parallel assays required in IGRA, providing a promising solution for TB screening in patients for whom current methods are inconvenient, such as children and older adults.

{"title":"Consolidated Microscale Interferon-γ Release Assay with Tip Optofluidic Immunoassay for Dynamic Parallel Diagnosis of Tuberculosis Infection.","authors":"Binmao Zhang, Yuzhong Xu, Zhen Huang, Ruihan Li, Tianen Zhu, Shangyan Liang, Hao Huang, Siyan Zhong, Hui Yang, Xudong Fan, Xiaotian Tan, Yujuan Chai","doi":"10.1021/acs.analchem.4c05390","DOIUrl":"10.1021/acs.analchem.4c05390","url":null,"abstract":"<p><p>Interferon-γ release assay (IGRA) is one of the most important diagnostic tools for tuberculosis (TB) infection. Despite its high accuracy, conventional IGRA has several drawbacks, including complicated procedures, large blood volume requirements, lengthy incubation times, and difficulties in parallel testing. Efforts have been made to develop miniaturized and highly sensitive biosensors for interferon-γ or to evaluate the specific immune response through microfluidic platforms. However, the need for sophisticated consumables and equipment, as well as the partial experimental design, has limited the application of these advanced techniques in TB diagnosis and disease control. Here, we report the development of a tip optofluidic immunoassay (TOI)-based consolidated microscale IGRA (CM-IGRA) for the dynamic and parallel evaluation of TB infection, refining both the blood incubation and interferon-γ quantification processes. The TOI system comprises 12 microfluidic immuno-reactors and a portable chemiluminescent imaging station, capable of quantifying interferon-γ with high sensitivity (8.00 pg/mL in plasma) and a wide detection range (∼10<sup>4</sup>). The results generated with CM-IGRA achieved 98.39% agreement with the standard IGRA while reducing blood sample consumption to 50 μL per assay (20-fold reduction) and significantly shortening the incubation time from 20 to 10 h. This diagnostic method simplifies operations and improves efficiency for the parallel assays required in IGRA, providing a promising solution for TB screening in patients for whom current methods are inconvenient, such as children and older adults.</p>","PeriodicalId":27,"journal":{"name":"Analytical Chemistry","volume":" ","pages":"2863-2872"},"PeriodicalIF":6.7,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143070727","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DNAzyme-Triggered Equilibrium Transfer with Self-Activated CRISPR-Cas12a Biosensor Enables One-Pot Diagnosis of Nucleic Acids.
IF 6.7 1区 化学 Q1 CHEMISTRY, ANALYTICAL Pub Date : 2025-02-11 Epub Date: 2025-01-31 DOI: 10.1021/acs.analchem.4c06066
Di Huang, Yichen He, Chutian Xu, Peijie Shen, Min Li, Mengjun Fang, Zhinan Xu, Xiangming Fang

Integrating recombinase-polymerase amplification (RPA) with CRISPR-Cas12a holds significant potential to simplify and improve nucleic acid diagnostic procedures. However, current strategies face limitations, such as complexity, reduced efficiency, and potential compromises in Cas12a activity. In response, we developed a DNAzyme-triggered equilibrium transfer with a self-activated CRISPR-Cas12a biosensor (DESCRIBER) for integrated nucleic acid detection. This platform features varying balance points to minimize interference between RPA and Cas12a in one pot and maximize their activity at different stages. Initially, the reaction focused on RPA, while Cas12a was silenced by circular-crRNA (C-crRNA). Then, DNAzyme, the activator, was generated during the RPA process, which linearizes C-crRNA to activate Cas12a and transfer the equilibrium toward signal readout. Meanwhile, activated Cas12a can further linearize C-crRNA to promote self-activation and accelerate equilibrium transfer. According to this principle, highly sensitive detection of the HIV-1 genome, as low as 500 CPs/mL, was achieved within 1 h while maintaining universality in detecting common subtypes and specificity against opportunistic infectious pathogens. Compared with qRT-PCR, it also exhibited good accuracy in detecting 35 spiked samples. Overall, we believe that the proposed strategy will enhance existing CRISPR systems to promote their practical applications in clinical diagnosis.

将重组酶聚合酶扩增(RPA)与 CRISPR-Cas12a 相结合,在简化和改进核酸诊断程序方面具有巨大潜力。然而,目前的策略面临着一些局限性,如复杂性、效率降低以及可能影响 Cas12a 的活性。为此,我们开发了一种 DNA 酶触发平衡转移与自激活 CRISPR-Cas12a 生物传感器(DESCRIBER),用于集成核酸检测。该平台具有不同的平衡点,可最大限度地减少一锅中 RPA 和 Cas12a 之间的干扰,并最大限度地提高它们在不同阶段的活性。起初,反应集中在 RPA 上,而 Cas12a 则被环状-crRNA(C-crRNA)沉默。然后,在 RPA 过程中产生激活剂 DNAzyme,它将 C-crRNA 线性化,从而激活 Cas12a,并将平衡转移到信号读出上。同时,被激活的 Cas12a 还能进一步线性化 C-crRNA,促进自我激活,加速平衡传递。根据这一原理,可在 1 小时内实现对低至 500 CPs/mL 的 HIV-1 基因组的高灵敏度检测,同时保持检测常见亚型的普遍性和针对机会性感染病原体的特异性。与 qRT-PCR 相比,该方法在检测 35 份加标样本时也表现出良好的准确性。总之,我们相信所提出的策略将增强现有的 CRISPR 系统,促进其在临床诊断中的实际应用。
{"title":"DNAzyme-Triggered Equilibrium Transfer with Self-Activated CRISPR-Cas12a Biosensor Enables One-Pot Diagnosis of Nucleic Acids.","authors":"Di Huang, Yichen He, Chutian Xu, Peijie Shen, Min Li, Mengjun Fang, Zhinan Xu, Xiangming Fang","doi":"10.1021/acs.analchem.4c06066","DOIUrl":"10.1021/acs.analchem.4c06066","url":null,"abstract":"<p><p>Integrating recombinase-polymerase amplification (RPA) with CRISPR-Cas12a holds significant potential to simplify and improve nucleic acid diagnostic procedures. However, current strategies face limitations, such as complexity, reduced efficiency, and potential compromises in Cas12a activity. In response, we developed a <u>D</u>NAzyme-triggered <u>e</u>quilibrium transfer with a <u>s</u>elf-activated <u>CRI</u>SPR-Cas12a <u>b</u>ios<u>e</u>nso<u>r</u> (DESCRIBER) for integrated nucleic acid detection. This platform features varying balance points to minimize interference between RPA and Cas12a in one pot and maximize their activity at different stages. Initially, the reaction focused on RPA, while Cas12a was silenced by circular-crRNA (C-crRNA). Then, DNAzyme, the activator, was generated during the RPA process, which linearizes C-crRNA to activate Cas12a and transfer the equilibrium toward signal readout. Meanwhile, activated Cas12a can further linearize C-crRNA to promote self-activation and accelerate equilibrium transfer. According to this principle, highly sensitive detection of the HIV-1 genome, as low as 500 CPs/mL, was achieved within 1 h while maintaining universality in detecting common subtypes and specificity against opportunistic infectious pathogens. Compared with qRT-PCR, it also exhibited good accuracy in detecting 35 spiked samples. Overall, we believe that the proposed strategy will enhance existing CRISPR systems to promote their practical applications in clinical diagnosis.</p>","PeriodicalId":27,"journal":{"name":"Analytical Chemistry","volume":" ","pages":"3026-3035"},"PeriodicalIF":6.7,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143070729","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SIGNIFICANT IMPACT OF CONSUMABLE MATERIAL AND BUFFER COMPOSITION FOR LOW-CELL NUMBER PROTEOMIC SAMPLE PREPARATION
IF 7.4 1区 化学 Q1 CHEMISTRY, ANALYTICAL Pub Date : 2025-02-11 DOI: 10.1021/acs.analchem.4c03709
Christopher Kune, Sylvia Tielens, Dominique Baiwir, Maximilien Fléron, Denis Vandormael, Gauthier Eppe, Laurent Nguyen, Gabriel Mazzucchelli
Proteomics, essential for understanding gene and cell functions, faces challenges with peptide loss due to adsorption onto vial surfaces, especially in samples with low peptide quantities. Using HeLa tryptic digested standard solutions, we demonstrate preferential adsorption of peptides, particularly hydrophobic ones, onto polypropylene (PP) vials, leading to nonuniform signal loss. This phenomenon can alter protein quantification (e.g., Label-Free Quantification, LFQ) if no appropriate data processing is applied. Our study is based on understanding this adsorption phenomenon to establish recommendations for minimizing peptide loss. To address this issue, we evaluated the nature of surface material and buffer additives to reduce peptide-surface noncovalent binding. Here, we report that using vials made from polymer containing polar monomeric units such as poly(methyl methacrylate) (PMMA) or polyethylene terephthalate (PET) drastically reduces the hydrophobic peptide loss, increasing the global proteomics performance (4-fold increase in identified peptides for the single-cell equivalent peptide content range). Additionally, the incorporation of nonionic detergents like poly(ethylene oxide) (PEO) and n-Dodecyl-Beta-Maltoside (DDM) at optimized concentrations (0.0001% and 0.0075%, respectively) improves the overall proteomic performance and consistency, even across different vial materials. Implementing these recommendations on 0.2 ng/μL HeLa tryptic digest results in a 10-fold increase in terms of peptide signal. Application to True Single-Cell sample preparation without specialized instrumentation dramatically improves the performance, allowing for the identification of approximately 650 proteins, a stark contrast to none detected with classical protocols.
{"title":"SIGNIFICANT IMPACT OF CONSUMABLE MATERIAL AND BUFFER COMPOSITION FOR LOW-CELL NUMBER PROTEOMIC SAMPLE PREPARATION","authors":"Christopher Kune, Sylvia Tielens, Dominique Baiwir, Maximilien Fléron, Denis Vandormael, Gauthier Eppe, Laurent Nguyen, Gabriel Mazzucchelli","doi":"10.1021/acs.analchem.4c03709","DOIUrl":"https://doi.org/10.1021/acs.analchem.4c03709","url":null,"abstract":"Proteomics, essential for understanding gene and cell functions, faces challenges with peptide loss due to adsorption onto vial surfaces, especially in samples with low peptide quantities. Using HeLa tryptic digested standard solutions, we demonstrate preferential adsorption of peptides, particularly hydrophobic ones, onto polypropylene (PP) vials, leading to nonuniform signal loss. This phenomenon can alter protein quantification (e.g., Label-Free Quantification, LFQ) if no appropriate data processing is applied. Our study is based on understanding this adsorption phenomenon to establish recommendations for minimizing peptide loss. To address this issue, we evaluated the nature of surface material and buffer additives to reduce peptide-surface noncovalent binding. Here, we report that using vials made from polymer containing polar monomeric units such as poly(methyl methacrylate) (PMMA) or polyethylene terephthalate (PET) drastically reduces the hydrophobic peptide loss, increasing the global proteomics performance (4-fold increase in identified peptides for the single-cell equivalent peptide content range). Additionally, the incorporation of nonionic detergents like poly(ethylene oxide) (PEO) and n-Dodecyl-Beta-Maltoside (DDM) at optimized concentrations (0.0001% and 0.0075%, respectively) improves the overall proteomic performance and consistency, even across different vial materials. Implementing these recommendations on 0.2 ng/μL HeLa tryptic digest results in a 10-fold increase in terms of peptide signal. Application to True Single-Cell sample preparation without specialized instrumentation dramatically improves the performance, allowing for the identification of approximately 650 proteins, a stark contrast to none detected with classical protocols.","PeriodicalId":27,"journal":{"name":"Analytical Chemistry","volume":"15 1","pages":""},"PeriodicalIF":7.4,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143393676","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
iSODA: A Comprehensive Tool for Integrative Omics Data Analysis in Single- and Multi-Omics Experiments.
IF 6.7 1区 化学 Q1 CHEMISTRY, ANALYTICAL Pub Date : 2025-02-11 Epub Date: 2025-01-31 DOI: 10.1021/acs.analchem.4c04355
Damien Olivier-Jimenez, Rico J E Derks, Oscar Harari, Carlos Cruchaga, Muhammad Ali, Alessandro Ori, Domenico Di Fraia, Birol Cabukusta, Andy Henrie, Martin Giera, Yassene Mohammed

Thanks to the plummeting costs of continuously evolving omics analytical platforms, research centers collect multiomics data more routinely. They are, however, confronted with the lack of a versatile software solution to harmoniously analyze single-omics and interpret multiomics data. We have developed iSODA, a web-based application for the analysis of single- and multiomics data. The tool emphasizes intuitive interactive visualizations designed for user-driven data exploration. Researchers can access a variety of functions ranging from simple visualization like volcano plots and PCA to advanced functional analyses like enrichment analysis and lipid saturation analysis. For integrated multiomics, iSODA incorporates multi-omics factor analysis and similarity network fusion. The ability to adapt the data on-the-fly allows for tasks, such as the removal of outlier samples or failed features, imputation, or normalization. All results are presented through interactive plots, the modular design supports extensions, and tooltips and tutorials provide comprehensive guidance for users. iSODA is accessible under http://isoda.online/.

{"title":"iSODA: A Comprehensive Tool for Integrative Omics Data Analysis in Single- and Multi-Omics Experiments.","authors":"Damien Olivier-Jimenez, Rico J E Derks, Oscar Harari, Carlos Cruchaga, Muhammad Ali, Alessandro Ori, Domenico Di Fraia, Birol Cabukusta, Andy Henrie, Martin Giera, Yassene Mohammed","doi":"10.1021/acs.analchem.4c04355","DOIUrl":"10.1021/acs.analchem.4c04355","url":null,"abstract":"<p><p>Thanks to the plummeting costs of continuously evolving omics analytical platforms, research centers collect multiomics data more routinely. They are, however, confronted with the lack of a versatile software solution to harmoniously analyze single-omics and interpret multiomics data. We have developed iSODA, a web-based application for the analysis of single- and multiomics data. The tool emphasizes intuitive interactive visualizations designed for user-driven data exploration. Researchers can access a variety of functions ranging from simple visualization like volcano plots and PCA to advanced functional analyses like enrichment analysis and lipid saturation analysis. For integrated multiomics, iSODA incorporates multi-omics factor analysis and similarity network fusion. The ability to adapt the data on-the-fly allows for tasks, such as the removal of outlier samples or failed features, imputation, or normalization. All results are presented through interactive plots, the modular design supports extensions, and tooltips and tutorials provide comprehensive guidance for users. iSODA is accessible under http://isoda.online/.</p>","PeriodicalId":27,"journal":{"name":"Analytical Chemistry","volume":" ","pages":"2689-2697"},"PeriodicalIF":6.7,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11822744/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143062327","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ultrasensitive COFs Functionalized Electrochemical Biosensor for DNA Methyltransferase Activity Detection by DNA Walking and Rolled Circular Strand Displacement Amplification
IF 7.4 1区 化学 Q1 CHEMISTRY, ANALYTICAL Pub Date : 2025-02-11 DOI: 10.1021/acs.analchem.4c06572
Jialin Zhang, Biyao Mao, Shuoshuo Cheng, Kangqiang Lu, Herui Wen, Jiali Ren
Assessing the activity of DNA methyltransferases (MTases) and screening for methyltransferase inhibitors not only allow for a deep exploration of the role of methylation regulation in disease initiation and progression but also provide an important experimental and clinical basis for the diagnosis and treatment of diseases. Herein, a new COFs functionalized electrochemical biosensor has been developed to detect DNA adenine methylation (Dam) MTase activity with high sensitivity and rapidity by taking advantage of the DNA walker and rolled circular strand displacement amplification (RC-SDA) reaction. Specifically, hairpin probe H1 was methylated by Dam MTase, followed by methylation site-specific cleavage of DpnI enzyme to generate the S5 probe. The padlock probes with two nucleic acid endonuclease sites were introduced to trigger the RC-SDA reaction under the action of the primer, releasing a large number of single-stranded S6 probes. The released probe worked synergistically with the Exo III enzyme to trigger DNA walking, which exposed the binding sites to enable the Au-COF-MB signal probes to bind effectively to the electrode surface, and the electrochemical signals were thus generated. The result showed that the designed electrochemical biosensor demonstrated excellent sensitivity and specificity in detecting the activity of Dam MTase, with the detection limit (LOD) of 6.85 × 10–5 U/mL. This method provides support for the development of new treatment strategies for methylation related diseases.
{"title":"Ultrasensitive COFs Functionalized Electrochemical Biosensor for DNA Methyltransferase Activity Detection by DNA Walking and Rolled Circular Strand Displacement Amplification","authors":"Jialin Zhang, Biyao Mao, Shuoshuo Cheng, Kangqiang Lu, Herui Wen, Jiali Ren","doi":"10.1021/acs.analchem.4c06572","DOIUrl":"https://doi.org/10.1021/acs.analchem.4c06572","url":null,"abstract":"Assessing the activity of DNA methyltransferases (MTases) and screening for methyltransferase inhibitors not only allow for a deep exploration of the role of methylation regulation in disease initiation and progression but also provide an important experimental and clinical basis for the diagnosis and treatment of diseases. Herein, a new COFs functionalized electrochemical biosensor has been developed to detect DNA adenine methylation (Dam) MTase activity with high sensitivity and rapidity by taking advantage of the DNA walker and rolled circular strand displacement amplification (RC-SDA) reaction. Specifically, hairpin probe H1 was methylated by Dam MTase, followed by methylation site-specific cleavage of DpnI enzyme to generate the S5 probe. The padlock probes with two nucleic acid endonuclease sites were introduced to trigger the RC-SDA reaction under the action of the primer, releasing a large number of single-stranded S6 probes. The released probe worked synergistically with the Exo III enzyme to trigger DNA walking, which exposed the binding sites to enable the Au-COF-MB signal probes to bind effectively to the electrode surface, and the electrochemical signals were thus generated. The result showed that the designed electrochemical biosensor demonstrated excellent sensitivity and specificity in detecting the activity of Dam MTase, with the detection limit (LOD) of 6.85 × 10<sup>–5</sup> U/mL. This method provides support for the development of new treatment strategies for methylation related diseases.","PeriodicalId":27,"journal":{"name":"Analytical Chemistry","volume":"208 1","pages":""},"PeriodicalIF":7.4,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143385674","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Triple-Responsive and Dual-NIR Emissive Fluorescence Probe for Precise Cancer Imaging and Therapy by Activating Pyroptosis Pathway.
IF 6.7 1区 化学 Q1 CHEMISTRY, ANALYTICAL Pub Date : 2025-02-11 Epub Date: 2025-01-31 DOI: 10.1021/acs.analchem.4c06015
Min Deng, Peipei Wang, Zibo Zhai, Ying Liu, Dan Cheng, Longwei He, Songjiao Li

Revealing changes in the tumor microenvironment is crucial for understanding cancer and developing sensitive methods for precise cancer imaging and diagnosis. Intracellular hydrogen peroxide (H2O2) and microenvironmental factors (e.g., viscosity and polarity) are closely linked to various physiological and pathological processes, making them potential biomarkers for cancer. However, a triple-response theranostic probe for precise tumor imaging and therapy has not yet been achieved due to the lack of effective tools. Herein, we present a mitochondria-targeting near-infrared (NIR) fluorescent probe, VPH-5DF, capable of simultaneously monitoring H2O2, viscosity, and polarity through dual NIR channels. The probe specifically detects H2O2 via NIR emission (λem = 650 nm) and shows high sensitivity to microenvironmental viscosity/polarity in the deep NIR channel (λem ≈ 750 nm). Furthermore, the probe not only monitors mitochondrial polarity, viscosity, and fluctuations in endogenous/exogenous H2O2 levels but also distinguishes cancer cells from normal cells through multiple parameters. Additionally, VPH-5DF can be employed to monitor alterations in H2O2 levels, as well as changes in viscosity and polarity, during drug-induced pyroptosis in living cells. After treatment with VPH-5DF, chemotherapy-induced oxidative damage to the mitochondria in tumor cells activated the pyroptosis pathway, leading to a robust antitumor response, as evidenced in xenograft tumor models. Thus, this triple-response theranostic prodrug offers a new platform for precise in vivo cancer diagnosis and anticancer chemotherapy.

{"title":"A Triple-Responsive and Dual-NIR Emissive Fluorescence Probe for Precise Cancer Imaging and Therapy by Activating Pyroptosis Pathway.","authors":"Min Deng, Peipei Wang, Zibo Zhai, Ying Liu, Dan Cheng, Longwei He, Songjiao Li","doi":"10.1021/acs.analchem.4c06015","DOIUrl":"10.1021/acs.analchem.4c06015","url":null,"abstract":"<p><p>Revealing changes in the tumor microenvironment is crucial for understanding cancer and developing sensitive methods for precise cancer imaging and diagnosis. Intracellular hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>) and microenvironmental factors (e.g., viscosity and polarity) are closely linked to various physiological and pathological processes, making them potential biomarkers for cancer. However, a triple-response theranostic probe for precise tumor imaging and therapy has not yet been achieved due to the lack of effective tools. Herein, we present a mitochondria-targeting near-infrared (NIR) fluorescent probe, <b>VPH-5DF</b>, capable of simultaneously monitoring H<sub>2</sub>O<sub>2</sub>, viscosity, and polarity through dual NIR channels. The probe specifically detects H<sub>2</sub>O<sub>2</sub> via NIR emission (λ<sub>em</sub> = 650 nm) and shows high sensitivity to microenvironmental viscosity/polarity in the deep NIR channel (λ<sub>em</sub> ≈ 750 nm). Furthermore, the probe not only monitors mitochondrial polarity, viscosity, and fluctuations in endogenous/exogenous H<sub>2</sub>O<sub>2</sub> levels but also distinguishes cancer cells from normal cells through multiple parameters. Additionally, VPH-5DF can be employed to monitor alterations in H<sub>2</sub>O<sub>2</sub> levels, as well as changes in viscosity and polarity, during drug-induced pyroptosis in living cells. After treatment with VPH-5DF, chemotherapy-induced oxidative damage to the mitochondria in tumor cells activated the pyroptosis pathway, leading to a robust antitumor response, as evidenced in xenograft tumor models. Thus, this triple-response theranostic prodrug offers a new platform for precise <i>in vivo</i> cancer diagnosis and anticancer chemotherapy.</p>","PeriodicalId":27,"journal":{"name":"Analytical Chemistry","volume":" ","pages":"2998-3008"},"PeriodicalIF":6.7,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143062292","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"化学","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 Top-Down Mass Spectrometry Strategies in the Chromatographic Time Scale (LC-TD-MS) for the Extended Characterization of an Anti-EGFR Single-Domain Antibody-Drug Conjugate in Both Reduced and Nonreduced Forms.
IF 6.7 1区 化学 Q1 CHEMISTRY, ANALYTICAL Pub Date : 2025-02-11 Epub Date: 2025-01-31 DOI: 10.1021/acs.analchem.4c03323
Rania Benazza, Léa Letissier, Greg Papadakos, Jen Thom, Helene Diemer, Graham Cotton, Sarah Cianférani, Oscar Hernandez-Alba

Even though mAbs have attracted the biggest interest in the development of therapeutic proteins, next-generation therapeutics such as single-domain antibodies (sdAb) are propelling increasing attention as new alternatives with appealing applications in different clinical areas. These constructs are small therapeutic proteins formed by a variable domain of the heavy chain of an antibody with multiple therapeutic and production benefits compared with their mAb counterparts. These proteins can be subjected to different bioconjugation processes to form single-domain antibody-drug conjugates (sdADCs) and hence increase their therapeutic potency, and akin to other therapeutic proteins, nanobodies and related products require dedicated analytical strategies to fully characterize their primary structure prior to their release to the market. In this study, we report for the first time the extensive sequence characterization of a conjugated anti-EGFR 14 kDa sdADC by using state-of-the-art top-down mass spectrometry strategies in combination with liquid chromatography (LC-TD-MS). Mass analysis revealed a highly homogeneous sample with one conjugated molecule. Subsequently, the reduced sdADC was submitted to different fragmentation techniques, namely, higher-energy collisional dissociation, electron-transfer dissociation, and electron-transfer higher-energy collision dissociation, allowing to unambiguously assess the conjugation site with 24 diagnostic fragment ions and 85% of global sequence coverage. The sequence coverage of the nonreduced protein was significantly lower (around 16%); however, the analysis of the fragmentation spectra corroborated the presence of the intramolecular disulfide bridge along with the localization of the conjugation site. Altogether, our results pinpoint the difficulties and challenges associated with the fragmentation of sdAb-derived formats in the LC time scale due to their remarkable stability as a consequence of the intramolecular disulfide bridge. However, the use of complementary activation techniques along with the identification of specific ion fragments allows an improved sequence coverage, the characterization of the intramolecular disulfide bond, and the unambiguous localization of the conjugation site.

{"title":"Development of Top-Down Mass Spectrometry Strategies in the Chromatographic Time Scale (LC-TD-MS) for the Extended Characterization of an Anti-EGFR Single-Domain Antibody-Drug Conjugate in Both Reduced and Nonreduced Forms.","authors":"Rania Benazza, Léa Letissier, Greg Papadakos, Jen Thom, Helene Diemer, Graham Cotton, Sarah Cianférani, Oscar Hernandez-Alba","doi":"10.1021/acs.analchem.4c03323","DOIUrl":"10.1021/acs.analchem.4c03323","url":null,"abstract":"<p><p>Even though mAbs have attracted the biggest interest in the development of therapeutic proteins, next-generation therapeutics such as single-domain antibodies (sdAb) are propelling increasing attention as new alternatives with appealing applications in different clinical areas. These constructs are small therapeutic proteins formed by a variable domain of the heavy chain of an antibody with multiple therapeutic and production benefits compared with their mAb counterparts. These proteins can be subjected to different bioconjugation processes to form single-domain antibody-drug conjugates (sdADCs) and hence increase their therapeutic potency, and akin to other therapeutic proteins, nanobodies and related products require dedicated analytical strategies to fully characterize their primary structure prior to their release to the market. In this study, we report for the first time the extensive sequence characterization of a conjugated anti-EGFR 14 kDa sdADC by using state-of-the-art top-down mass spectrometry strategies in combination with liquid chromatography (LC-TD-MS). Mass analysis revealed a highly homogeneous sample with one conjugated molecule. Subsequently, the reduced sdADC was submitted to different fragmentation techniques, namely, higher-energy collisional dissociation, electron-transfer dissociation, and electron-transfer higher-energy collision dissociation, allowing to unambiguously assess the conjugation site with 24 diagnostic fragment ions and 85% of global sequence coverage. The sequence coverage of the nonreduced protein was significantly lower (around 16%); however, the analysis of the fragmentation spectra corroborated the presence of the intramolecular disulfide bridge along with the localization of the conjugation site. Altogether, our results pinpoint the difficulties and challenges associated with the fragmentation of sdAb-derived formats in the LC time scale due to their remarkable stability as a consequence of the intramolecular disulfide bridge. However, the use of complementary activation techniques along with the identification of specific ion fragments allows an improved sequence coverage, the characterization of the intramolecular disulfide bond, and the unambiguous localization of the conjugation site.</p>","PeriodicalId":27,"journal":{"name":"Analytical Chemistry","volume":" ","pages":"2639-2647"},"PeriodicalIF":6.7,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143070728","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Staining Tissues with Basic Blue 7: A New Dual-Polarity Matrix for MALDI Mass Spectrometry Imaging.
IF 6.7 1区 化学 Q1 CHEMISTRY, ANALYTICAL Pub Date : 2025-02-11 Epub Date: 2025-01-30 DOI: 10.1021/acs.analchem.4c05244
Michal Javorek, Michal Hendrych, Kateřina Ondráková, Jan Preisler, Antonín Bednařík

Obtaining high-quality matrix-assisted laser desorption/ionization mass spectrometry (MALDI MS) images and the reproducibility of the technique depend strongly on the sample preparation protocol. The most crucial part is the application of the MALDI matrix, which often relies on expensive spraying or sublimation coaters. In this work, we present a new dual-polarity matrix for MALDI mass spectrometry imaging (MSI): Basic Blue 7 (BB7), which belongs to the group of triarylmethane dyes. Thanks to its good solubility in water, this matrix allows a quick and simple sample preparation protocol without the need for sophisticated spraying or sublimation instrumentation: dipping the glass with tissue into the dye solution. This technique closely resembles the staining methods employed in classical histopathology. The technique is demonstrated on MSI of lipids in mouse brain sections in positive and negative ion modes using a subatmospheric pressure MALDI source coupled with an orbital trap mass spectrometer. The results are compared with traditional matrices, such as 2,5-dihydroxybenzoic acid (DHB) and 1,5-diaminonaphthalene (DAN). BB7 excels, especially in negative ion mode, offering low background signals and high signal intensities of many lipid classes. Furthermore, the stained tissue can simply be inspected visually and allows basic histopathology annotation prior to MSI. Here, we demonstrate that staining offers excellent image quality, reproducible sample preparation, and the potential for automation and utilization for high spatial resolution MSI.

{"title":"Staining Tissues with Basic Blue 7: A New Dual-Polarity Matrix for MALDI Mass Spectrometry Imaging.","authors":"Michal Javorek, Michal Hendrych, Kateřina Ondráková, Jan Preisler, Antonín Bednařík","doi":"10.1021/acs.analchem.4c05244","DOIUrl":"10.1021/acs.analchem.4c05244","url":null,"abstract":"<p><p>Obtaining high-quality matrix-assisted laser desorption/ionization mass spectrometry (MALDI MS) images and the reproducibility of the technique depend strongly on the sample preparation protocol. The most crucial part is the application of the MALDI matrix, which often relies on expensive spraying or sublimation coaters. In this work, we present a new dual-polarity matrix for MALDI mass spectrometry imaging (MSI): Basic Blue 7 (BB7), which belongs to the group of triarylmethane dyes. Thanks to its good solubility in water, this matrix allows a quick and simple sample preparation protocol without the need for sophisticated spraying or sublimation instrumentation: dipping the glass with tissue into the dye solution. This technique closely resembles the staining methods employed in classical histopathology. The technique is demonstrated on MSI of lipids in mouse brain sections in positive and negative ion modes using a subatmospheric pressure MALDI source coupled with an orbital trap mass spectrometer. The results are compared with traditional matrices, such as 2,5-dihydroxybenzoic acid (DHB) and 1,5-diaminonaphthalene (DAN). BB7 excels, especially in negative ion mode, offering low background signals and high signal intensities of many lipid classes. Furthermore, the stained tissue can simply be inspected visually and allows basic histopathology annotation prior to MSI. Here, we demonstrate that staining offers excellent image quality, reproducible sample preparation, and the potential for automation and utilization for high spatial resolution MSI.</p>","PeriodicalId":27,"journal":{"name":"Analytical Chemistry","volume":" ","pages":"2828-2836"},"PeriodicalIF":6.7,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11822741/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143062133","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hyphenation of Affinity Capillary Electrophoresis with Mass Spectrometry for the Study of Ligand–Protein Interactions: n-Methylmorpholine Acetate Buffer and Polydopamine-Based Coating as Key Assets 研究配体与蛋白质相互作用的亲和毛细管电泳与质谱联用:作为关键资产的醋酸正甲基吗啉缓冲液和聚多巴胺涂层
IF 7.4 1区 化学 Q1 CHEMISTRY, ANALYTICAL Pub Date : 2025-02-11 DOI: 10.1021/acs.analchem.4c05559
Clara Davoine, Marianne Fillet
The direct and precise assessment of ligand–protein interactions under nearly physiological conditions is the core of drug discovery. In this context, affinity capillary electrophoresis (ACE) has become an emerging and reliable approach. The hyphenation of ACE with mass spectrometry (MS) is even more powerful than the classical ACE-UV methodology. It reduces compound identification errors and increases throughput by facilitating the analysis of the mixtures. However, buffers and capillary coatings compatible with mass spectrometry and operating under physiological conditions are very limited. In this paper, n-methylmorpholine acetate buffer and polydopamine-based coating were highlighted as major assets for CE-MS studies involving native proteins. Thanks to its protein desorption property, n-methylmorpholine improved the peak shape of proteins during CE analysis at physiological pH. The polydopamine-based neutral coating developed in this study is simple to prepare and demonstrated high stability at pH 7.4, enabling its use with an MS detector. The combination of these two key elements enabled us to successfully convert our ACE-UV method for coagulation factor XIIa into an ACE-MS approach operating at physiological pH. This study extends the scope of ACE for medicinal chemistry projects.
{"title":"Hyphenation of Affinity Capillary Electrophoresis with Mass Spectrometry for the Study of Ligand–Protein Interactions: n-Methylmorpholine Acetate Buffer and Polydopamine-Based Coating as Key Assets","authors":"Clara Davoine, Marianne Fillet","doi":"10.1021/acs.analchem.4c05559","DOIUrl":"https://doi.org/10.1021/acs.analchem.4c05559","url":null,"abstract":"The direct and precise assessment of ligand–protein interactions under nearly physiological conditions is the core of drug discovery. In this context, affinity capillary electrophoresis (ACE) has become an emerging and reliable approach. The hyphenation of ACE with mass spectrometry (MS) is even more powerful than the classical ACE-UV methodology. It reduces compound identification errors and increases throughput by facilitating the analysis of the mixtures. However, buffers and capillary coatings compatible with mass spectrometry and operating under physiological conditions are very limited. In this paper, <i>n</i>-methylmorpholine acetate buffer and polydopamine-based coating were highlighted as major assets for CE-MS studies involving native proteins. Thanks to its protein desorption property, <i>n</i>-methylmorpholine improved the peak shape of proteins during CE analysis at physiological pH. The polydopamine-based neutral coating developed in this study is simple to prepare and demonstrated high stability at pH 7.4, enabling its use with an MS detector. The combination of these two key elements enabled us to successfully convert our ACE-UV method for coagulation factor XIIa into an ACE-MS approach operating at physiological pH. This study extends the scope of ACE for medicinal chemistry projects.","PeriodicalId":27,"journal":{"name":"Analytical Chemistry","volume":"63 1","pages":""},"PeriodicalIF":7.4,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143393877","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"化学","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
Analytical Chemistry
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