Pub Date : 2025-09-19DOI: 10.1021/acscentsci.5c01666
Alexandra T. Barth, and , Felix N. Castellano,
Earth-abundant ligand-to-metal charge transfer (LMCT) chromophores in donor–acceptor dyads unlock an electron transfer pathway for efficient triplet state formation.
{"title":"Chasing Waterfalls: A Cascade Mechanism to Generate Triplets from 2LMCT States","authors":"Alexandra T. Barth, and , Felix N. Castellano, ","doi":"10.1021/acscentsci.5c01666","DOIUrl":"https://doi.org/10.1021/acscentsci.5c01666","url":null,"abstract":"<p >Earth-abundant ligand-to-metal charge transfer (LMCT) chromophores in donor–acceptor dyads unlock an electron transfer pathway for efficient triplet state formation.</p>","PeriodicalId":10,"journal":{"name":"ACS Central Science","volume":"11 10","pages":"1802–1804"},"PeriodicalIF":10.4,"publicationDate":"2025-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acscentsci.5c01666","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145332098","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}
Pub Date : 2025-09-19DOI: 10.1021/acscentsci.5c01713
Kwang S. Kim,
A quantum algorithm navigating the immense design space of multivariate porous materials demonstrates a logical and practical roadmap for the future of chemical synthesis.
在多元多孔材料的巨大设计空间中导航的量子算法为化学合成的未来展示了一个合乎逻辑且实用的路线图。
{"title":"A Quantum Compass for Materials Discovery: Navigating the Combinatorial Explosion","authors":"Kwang S. Kim, ","doi":"10.1021/acscentsci.5c01713","DOIUrl":"https://doi.org/10.1021/acscentsci.5c01713","url":null,"abstract":"<p >A quantum algorithm navigating the immense design space of multivariate porous materials demonstrates a logical and practical roadmap for the future of chemical synthesis.</p>","PeriodicalId":10,"journal":{"name":"ACS Central Science","volume":"11 10","pages":"1808–1811"},"PeriodicalIF":10.4,"publicationDate":"2025-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acscentsci.5c01713","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145332097","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}
Pub Date : 2025-09-19DOI: 10.1021/acscentsci.5c01712
Xin Wan, and , Jianglan Shui*,
An electron-enriched PtNiCo catalyst enabled by TiN support boosts stability in methanol fuel cells by simultaneously overcoming CO poisoning and metal dissolution.
{"title":"Electron Donation Stabilizes Pt Catalysts in Methanol Fuel Cells","authors":"Xin Wan, and , Jianglan Shui*, ","doi":"10.1021/acscentsci.5c01712","DOIUrl":"https://doi.org/10.1021/acscentsci.5c01712","url":null,"abstract":"<p >An electron-enriched PtNiCo catalyst enabled by TiN support boosts stability in methanol fuel cells by simultaneously overcoming CO poisoning and metal dissolution.</p>","PeriodicalId":10,"journal":{"name":"ACS Central Science","volume":"11 10","pages":"1799–1801"},"PeriodicalIF":10.4,"publicationDate":"2025-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acscentsci.5c01712","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145332096","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}
Pub Date : 2025-09-17DOI: 10.1021/acscentsci.5c01046
Seul Ki Yeon, , , Jenna Pellegrino, , , Tushar Raskar, , , Minh L. N. Tran, , , Mohamad Dandan, , , François Guérin, , , Manuel Einsiedler, , , Vincent Cattoir, , , James S. Fraser, , and , Ian B. Seiple*,
Antimicrobial resistance remains a formidable challenge to modern medicine, with bacterial resistance mechanisms increasingly eroding the utility of clinically important antibiotics. While recent efforts have expanded the antibacterial pipeline, the development of resistance in priority pathogens continues to exceed the pace of new drug development. One emerging strategy to overcome resistance is the rational design of hybrid antibiotics that engage multiple binding sites. Here we describe the design, synthesis, and microbiological and structural characterization of hybrid antibiotics of azithromycin, tedizolid, and chloramphenicol that span the peptidyltransferase center (PTC) and nascent peptide exit tunnel (NPET) in the bacterial ribosome. We characterize the binding of four such hybrids by cryo-electron microscopy, granting insight into their molecular mechanisms of action. We identify a hybrid of azithromycin and tedizolid that is active against a diverse panel of multidrug-resistant Gram-positive bacteria and is minimally affected by ribosomal protection (ABC-F) resistance mechanisms. These results extend our understanding of ribosome inhibition and provide a pipeline for the rational design of dual-action antibiotics that target the ribosome. In a broader context, this work offers a framework for developing bifunctional inhibitors that engage adjacent binding sites by means of a rational cycle of synthetic optimization, biological evaluation, and structural characterization.
An integrated platform to develop hybrid antibiotics that inhibit the bacterial ribosome.
{"title":"Hybrid Antibiotics Targeting the Bacterial Ribosome","authors":"Seul Ki Yeon, , , Jenna Pellegrino, , , Tushar Raskar, , , Minh L. N. Tran, , , Mohamad Dandan, , , François Guérin, , , Manuel Einsiedler, , , Vincent Cattoir, , , James S. Fraser, , and , Ian B. Seiple*, ","doi":"10.1021/acscentsci.5c01046","DOIUrl":"https://doi.org/10.1021/acscentsci.5c01046","url":null,"abstract":"<p >Antimicrobial resistance remains a formidable challenge to modern medicine, with bacterial resistance mechanisms increasingly eroding the utility of clinically important antibiotics. While recent efforts have expanded the antibacterial pipeline, the development of resistance in priority pathogens continues to exceed the pace of new drug development. One emerging strategy to overcome resistance is the rational design of hybrid antibiotics that engage multiple binding sites. Here we describe the design, synthesis, and microbiological and structural characterization of hybrid antibiotics of azithromycin, tedizolid, and chloramphenicol that span the peptidyltransferase center (PTC) and nascent peptide exit tunnel (NPET) in the bacterial ribosome. We characterize the binding of four such hybrids by cryo-electron microscopy, granting insight into their molecular mechanisms of action. We identify a hybrid of azithromycin and tedizolid that is active against a diverse panel of multidrug-resistant Gram-positive bacteria and is minimally affected by ribosomal protection (ABC-F) resistance mechanisms. These results extend our understanding of ribosome inhibition and provide a pipeline for the rational design of dual-action antibiotics that target the ribosome. In a broader context, this work offers a framework for developing bifunctional inhibitors that engage adjacent binding sites by means of a rational cycle of synthetic optimization, biological evaluation, and structural characterization.</p><p >An integrated platform to develop hybrid antibiotics that inhibit the bacterial ribosome.</p>","PeriodicalId":10,"journal":{"name":"ACS Central Science","volume":"11 11","pages":"2133–2142"},"PeriodicalIF":10.4,"publicationDate":"2025-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acscentsci.5c01046","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145594392","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}
Pub Date : 2025-09-17DOI: 10.1021/acscentsci.5c01285
Kunyang Sun, , , Dorian Bagni, , , Joseph M. Cavanagh, , , Yingze Wang, , , Jacob M. Sawyer, , , Bo Zhou, , , Andrew Gritsevskiy, , , Oufan Zhang, , and , Teresa Head-Gordon*,
Generative machine learning models for exploring chemical space have shown immense promise, but many molecules that they generate are too difficult to synthesize, making them impractical for further investigation or development. In this work, we present a novel approach by fine-tuning Meta’s Llama3 Large Language Models (LLMs) to create SynLlama, which generates full synthetic pathways made of commonly accessible building blocks and robust organic reaction templates. SynLlama explores a large synthesizable space using significantly less data and offers strong performance in both forward and bottom-up synthesis planning compared to other state-of-the-art methods. We find that SynLlama, even without training on external building blocks, can effectively generalize to unseen yet purchasable building blocks, meaning that its reconstruction capabilities extend to a broader synthesizable chemical space than those of the training data. We also demonstrate the use of SynLlama in a pharmaceutical context for synthesis planning of analog molecules and hit expansion leads for proposed inhibitors of target proteins, offering medicinal chemists a valuable tool for discovery.
Fine-tuning on synthetic reactions from commercial building blocks and high-fidelity reactions creates a versatile LLM, SynLlama, for key drug discovery tasks.
{"title":"SynLlama: Generating Synthesizable Molecules and Their Analogs with Large Language Models","authors":"Kunyang Sun, , , Dorian Bagni, , , Joseph M. Cavanagh, , , Yingze Wang, , , Jacob M. Sawyer, , , Bo Zhou, , , Andrew Gritsevskiy, , , Oufan Zhang, , and , Teresa Head-Gordon*, ","doi":"10.1021/acscentsci.5c01285","DOIUrl":"https://doi.org/10.1021/acscentsci.5c01285","url":null,"abstract":"<p >Generative machine learning models for exploring chemical space have shown immense promise, but many molecules that they generate are too difficult to synthesize, making them impractical for further investigation or development. In this work, we present a novel approach by fine-tuning Meta’s Llama3 Large Language Models (LLMs) to create SynLlama, which generates full synthetic pathways made of commonly accessible building blocks and robust organic reaction templates. SynLlama explores a large synthesizable space using significantly less data and offers strong performance in both forward and bottom-up synthesis planning compared to other state-of-the-art methods. We find that SynLlama, even without training on external building blocks, can effectively generalize to unseen yet purchasable building blocks, meaning that its reconstruction capabilities extend to a broader synthesizable chemical space than those of the training data. We also demonstrate the use of SynLlama in a pharmaceutical context for synthesis planning of analog molecules and hit expansion leads for proposed inhibitors of target proteins, offering medicinal chemists a valuable tool for discovery.</p><p >Fine-tuning on synthetic reactions from commercial building blocks and high-fidelity reactions creates a versatile LLM, SynLlama, for key drug discovery tasks.</p>","PeriodicalId":10,"journal":{"name":"ACS Central Science","volume":"11 11","pages":"2108–2120"},"PeriodicalIF":10.4,"publicationDate":"2025-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acscentsci.5c01285","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145594391","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}
Tumor resistance to immune checkpoint blockade (ICB) therapy is frequently driven by adaptive metabolic reprogramming in the hypoxic tumor microenvironment (TME). The key N4-acetylcytidine (ac4C) RNA modification mediator N-acetyltransferase 10 (NAT10) emerges as a promising therapeutic target, despite the lack of potent targeting agents. Here, we engineered NP1192, a PROTAC degrader targeting NAT10. NP1192 achieved nearly 70% NAT10 degradation and a 26.8% lower IC50 than canonical NAT10 inhibitor Remodelin in cervical cancer cells, outperforming Remodelin in antitumor effect in vivo, in vitro, and across three tumor organoids. It abrogated ac4C modifications on HIF1A mRNA and translation, reducing hypoxic lactate production and depleted ATP, and suppressed HIF-1α-mediated PD-L1 upregulation. In xenograft models, NP1192 combined with anti-PD-L1 inhibited subcutaneous xenograft growth and reduced tumor-core lactate gradients by > 80%. Furthermore, scRNA-seq and in vitro coculture experiments identified expansion of IFN-γ+ effector CD8+ T cells (Teff) and decline in exhausted CD8+ T cells (Tex). NP1192 in combination with anti-PD-L1 enhanced proliferation and effector function of CD8+ Teff cells, thereby reversing resistance to anti-PD-L1 blockade therapy and synergizing with immunotherapy. These findings establish PROTAC-mediated NAT10 degradation as a dual metabolic-immune strategy to enhance checkpoint blockade efficacy.
PROTAC NP1192 degrades NAT10 inhibiting HIF-1α via ac4C modification. It reduces hypoxic glycolysis, enhances CD8+ Teff function, and synergizes with anti-PD-L1 to reverse immunotherapy resistance.
{"title":"Targeted NAT10 Degradation by PROTAC NP1192 Suppresses Hypoxia-Adaptive Glycolysis and Reinvigorates CD8+ Effector T-Cell Function for Synergistic Cancer Immunotherapy","authors":"Keyi Ao, , , Zhiqiang Sun, , , Yi Hao, , , Jiaqi Qin, , , Chenglong Xu, , , Xiuli Wen, , , Zichao Yang, , , Li Li, , , Shaoyan Gan, , , Xiaona Chen, , , Xin Li*, , , Jian Zhang*, , , Jianjun Chen*, , and , Xia Guo*, ","doi":"10.1021/acscentsci.5c00812","DOIUrl":"https://doi.org/10.1021/acscentsci.5c00812","url":null,"abstract":"<p >Tumor resistance to immune checkpoint blockade (ICB) therapy is frequently driven by adaptive metabolic reprogramming in the hypoxic tumor microenvironment (TME). The key N4-acetylcytidine (ac4C) RNA modification mediator N-acetyltransferase 10 (NAT10) emerges as a promising therapeutic target, despite the lack of potent targeting agents. Here, we engineered NP1192, a PROTAC degrader targeting NAT10. NP1192 achieved nearly 70% NAT10 degradation and a 26.8% lower IC<sub>50</sub> than canonical NAT10 inhibitor Remodelin in cervical cancer cells, outperforming Remodelin in antitumor effect <i>in vivo</i>, <i>in vitro</i>, and across three tumor organoids. It abrogated ac4C modifications on <i>HIF1A</i> mRNA and translation, reducing hypoxic lactate production and depleted ATP, and suppressed HIF-1α-mediated PD-L1 upregulation. In xenograft models, NP1192 combined with anti-PD-L1 inhibited subcutaneous xenograft growth and reduced tumor-core lactate gradients by > 80%. Furthermore, scRNA-seq and <i>in vitro</i> coculture experiments identified expansion of IFN-γ<sup>+</sup> effector CD8<sup>+</sup> T cells (T<sub>eff</sub>) and decline in exhausted CD8<sup>+</sup> T cells (T<sub>ex</sub>). NP1192 in combination with anti-PD-L1 enhanced proliferation and effector function of CD8<sup>+</sup> T<sub>eff</sub> cells, thereby reversing resistance to anti-PD-L1 blockade therapy and synergizing with immunotherapy. These findings establish PROTAC-mediated NAT10 degradation as a dual metabolic-immune strategy to enhance checkpoint blockade efficacy.</p><p >PROTAC NP1192 degrades NAT10 inhibiting HIF-1α via ac4C modification. It reduces hypoxic glycolysis, enhances CD8<sup>+</sup> T<sub>eff</sub> function, and synergizes with anti-PD-L1 to reverse immunotherapy resistance.</p>","PeriodicalId":10,"journal":{"name":"ACS Central Science","volume":"11 11","pages":"2087–2107"},"PeriodicalIF":10.4,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acscentsci.5c00812","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145594426","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}
Pub Date : 2025-09-16DOI: 10.1021/acscentsci.5c00509
Su Yeon Lim, , , Pin Liu, , , Ju Hwa Shin, , , Bum Soo Lee, , , Sun Ju Kim, , , Dahwun Kim, , , Siyan Lyu, , , Byung Deok Kim, , , Chaeeun Park, , , Junku Jung, , , Jihyun Lee, , , Jinbeom Seo, , , Taegwan Yun, , , Hyo Jin Park, , , Min Sang Lee, , , Ki Hyun Kim*, , , Wonsik Lee*, , and , Ji Hoon Jeong*,
The efficacy of cancer immunotherapy is often limited by the immunosuppressive tumor microenvironment (TME) and insufficient immune activation in tumor-draining lymph nodes (TDLN). Since the TME and TDLN form a dynamic axis crucial for tumor metastasis and resistance to immune checkpoint blockade, strategies that effectively modulate both sites are critical. Here, we present a dissolving microneedle (MN) system that generates nanomicelles (NMCs) for localized delivery of a newly identified dual-functional macrocyclic trichothecene, Roridin E (R.E). R.E induces cancer cell-autonomous secretion of IFN-β and immunogenic cancer cell death (ICD). Direct delivery of R.E to the TDLN via the MN platform reshapes the local immune landscape to suppress cancer while minimizing off-target toxicity. In a B16F10 melanoma model, MN-guided R.E. delivery significantly improved tumor control, reduced lung metastases, and extended overall survival. This approach provides a minimally invasive and effective strategy for integrating natural-product-based therapies with advanced drug delivery systems to target the TME–TDLN axis, thereby improving outcomes in metastatic cancer.
Microneedle-directed delivery of a dual-functional immune modulator reprograms tumor−lymph node immunity to enhance cancer immunotherapy.
{"title":"Reshaping Tumor-Lymph Node Immune Axis via Targeted Lymphatic Delivery of Dual-Functional Immune Modulator for Enhanced Cancer Immunotherapy","authors":"Su Yeon Lim, , , Pin Liu, , , Ju Hwa Shin, , , Bum Soo Lee, , , Sun Ju Kim, , , Dahwun Kim, , , Siyan Lyu, , , Byung Deok Kim, , , Chaeeun Park, , , Junku Jung, , , Jihyun Lee, , , Jinbeom Seo, , , Taegwan Yun, , , Hyo Jin Park, , , Min Sang Lee, , , Ki Hyun Kim*, , , Wonsik Lee*, , and , Ji Hoon Jeong*, ","doi":"10.1021/acscentsci.5c00509","DOIUrl":"https://doi.org/10.1021/acscentsci.5c00509","url":null,"abstract":"<p >The efficacy of cancer immunotherapy is often limited by the immunosuppressive tumor microenvironment (TME) and insufficient immune activation in tumor-draining lymph nodes (TDLN). Since the TME and TDLN form a dynamic axis crucial for tumor metastasis and resistance to immune checkpoint blockade, strategies that effectively modulate both sites are critical. Here, we present a dissolving microneedle (MN) system that generates nanomicelles (NMCs) for localized delivery of a newly identified dual-functional macrocyclic trichothecene, Roridin E (R.E). R.E induces cancer cell-autonomous secretion of IFN-β and immunogenic cancer cell death (ICD). Direct delivery of R.E to the TDLN via the MN platform reshapes the local immune landscape to suppress cancer while minimizing off-target toxicity. In a B16F10 melanoma model, MN-guided R.E. delivery significantly improved tumor control, reduced lung metastases, and extended overall survival. This approach provides a minimally invasive and effective strategy for integrating natural-product-based therapies with advanced drug delivery systems to target the TME–TDLN axis, thereby improving outcomes in metastatic cancer.</p><p >Microneedle-directed delivery of a dual-functional immune modulator reprograms tumor−lymph node immunity to enhance cancer immunotherapy.</p>","PeriodicalId":10,"journal":{"name":"ACS Central Science","volume":"11 11","pages":"2074–2086"},"PeriodicalIF":10.4,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acscentsci.5c00509","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145594388","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}
Pub Date : 2025-09-15DOI: 10.1021/acscentsci.5c00949
Tobias Vornholt*, , , Peter Stockinger, , , Mojmír Mutný, , , Markus Jeschek, , , Bettina Nestl, , , Gustav Oberdorfer, , , Silvia Osuna, , , Jürgen Pleiss, , , Ditte Hededam Welner, , , Andreas Krause, , , Rebecca Buller, , and , Thomas R. Ward*,
Machine learning (ML) is rapidly turning into a key technology for biocatalysis. By learning patterns in amino acid sequences, protein structures, and functional data, ML models can help navigate complex fitness landscapes, uncover new enzymes in databases, and even design biocatalysts de novo. Along with advances in DNA synthesis and sequencing, laboratory automation, and high-throughput screening, ML is increasing the speed and efficiency of enzyme development. In this Outlook, we highlight recent applications of ML in the fields of enzyme discovery, design, and engineering, with a focus on current challenges and emerging solutions. Furthermore, we discuss barriers that impede a broader and faster adoption of ML-based workflows in the biocatalysis community. We conclude by suggesting best practices for fostering effective collaborations in this interdisciplinary field.
We highlight how machine learning accelerates enzyme discovery, design, and engineering, outlining recent advances, key challenges, and emerging opportunities in biocatalysis.
{"title":"Of Revolutions and Roadblocks: The Emerging Role of Machine Learning in Biocatalysis","authors":"Tobias Vornholt*, , , Peter Stockinger, , , Mojmír Mutný, , , Markus Jeschek, , , Bettina Nestl, , , Gustav Oberdorfer, , , Silvia Osuna, , , Jürgen Pleiss, , , Ditte Hededam Welner, , , Andreas Krause, , , Rebecca Buller, , and , Thomas R. Ward*, ","doi":"10.1021/acscentsci.5c00949","DOIUrl":"https://doi.org/10.1021/acscentsci.5c00949","url":null,"abstract":"<p >Machine learning (ML) is rapidly turning into a key technology for biocatalysis. By learning patterns in amino acid sequences, protein structures, and functional data, ML models can help navigate complex fitness landscapes, uncover new enzymes in databases, and even design biocatalysts <i>de novo</i>. Along with advances in DNA synthesis and sequencing, laboratory automation, and high-throughput screening, ML is increasing the speed and efficiency of enzyme development. In this Outlook, we highlight recent applications of ML in the fields of enzyme discovery, design, and engineering, with a focus on current challenges and emerging solutions. Furthermore, we discuss barriers that impede a broader and faster adoption of ML-based workflows in the biocatalysis community. We conclude by suggesting best practices for fostering effective collaborations in this interdisciplinary field.</p><p >We highlight how machine learning accelerates enzyme discovery, design, and engineering, outlining recent advances, key challenges, and emerging opportunities in biocatalysis.</p>","PeriodicalId":10,"journal":{"name":"ACS Central Science","volume":"11 10","pages":"1828–1838"},"PeriodicalIF":10.4,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acscentsci.5c00949","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145332095","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}
Pub Date : 2025-09-15DOI: 10.1021/acscentsci.5c00851
Justin A. Hopkins, , , Benjamin J. Page, , , Shengguang Wang, , , Jesse R. Canavan, , , Jason A. Chalmers, , , Susannah L. Scott, , , Lars C. Grabow, , , James R. McKone, , , Paul J. Dauenhauer, , and , Omar A. Abdelrahman*,
The extent of charge transfer between an adsorbate and thermocatalytic surface plays a key role in determining catalytic activity, but direct and quantitative measures have remained elusive. Here, we report the method of isopotential electron titration (IET), an approach that directly measures charge transfer between adsorbates and catalytic surfaces. Charge transfer between Pt and adsorbed hydrogen adatoms was investigated using a catalytic condenser, where the Pt surface was separated from a p-type silicon layer by a hafnia dielectric film. By forcing the Pt and Si layers into isopotential conditions, charge transfer between the adsorbate and Pt surface was titrated through an external circuit. Hydrogen atoms donated electrons to Pt upon adsorption, which was quantitatively reversed upon desorption. Across a temperature range of 125–200 °C (surface hydrogen fractional coverages of 80–100%), the charge transferred to Pt by an adsorbed hydrogen atom was measured to be 0.19 ± 0.01% |e|/H. Bader charge analysis of the extent of charge transfer was in agreement with experimental measurements, with a calculated net donation of 0.4% |e|/H. The ability to experimentally quantify surface charge transfer provides an electronic-based approach to characterize catalytic surfaces, the adsorbed moieties residing on them, and the chemical reactions they accelerate.
The electrostatic generosity of hydrogen to metals: Isopotential electron titration measures the nonfaradaic charge transfer between adsorbates and catalytic surfaces
{"title":"Isopotential Electron Titration: Hydrogen Adsorbate-Metal Charge Transfer","authors":"Justin A. Hopkins, , , Benjamin J. Page, , , Shengguang Wang, , , Jesse R. Canavan, , , Jason A. Chalmers, , , Susannah L. Scott, , , Lars C. Grabow, , , James R. McKone, , , Paul J. Dauenhauer, , and , Omar A. Abdelrahman*, ","doi":"10.1021/acscentsci.5c00851","DOIUrl":"https://doi.org/10.1021/acscentsci.5c00851","url":null,"abstract":"<p >The extent of charge transfer between an adsorbate and thermocatalytic surface plays a key role in determining catalytic activity, but direct and quantitative measures have remained elusive. Here, we report the method of isopotential electron titration (IET), an approach that directly measures charge transfer between adsorbates and catalytic surfaces. Charge transfer between Pt and adsorbed hydrogen adatoms was investigated using a catalytic condenser, where the Pt surface was separated from a p-type silicon layer by a hafnia dielectric film. By forcing the Pt and Si layers into isopotential conditions, charge transfer between the adsorbate and Pt surface was titrated through an external circuit. Hydrogen atoms donated electrons to Pt upon adsorption, which was quantitatively reversed upon desorption. Across a temperature range of 125–200 °C (surface hydrogen fractional coverages of 80–100%), the charge transferred to Pt by an adsorbed hydrogen atom was measured to be 0.19 ± 0.01% |e|/H. Bader charge analysis of the extent of charge transfer was in agreement with experimental measurements, with a calculated net donation of 0.4% |e|/H. The ability to experimentally quantify surface charge transfer provides an electronic-based approach to characterize catalytic surfaces, the adsorbed moieties residing on them, and the chemical reactions they accelerate.</p><p >The electrostatic generosity of hydrogen to metals: Isopotential electron titration measures the nonfaradaic charge transfer between adsorbates and catalytic surfaces</p>","PeriodicalId":10,"journal":{"name":"ACS Central Science","volume":"11 11","pages":"2063–2073"},"PeriodicalIF":10.4,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acscentsci.5c00851","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145594424","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}
The site-selective C–H functionalization to install meta-C–S bonds on aniline derivatives is highly desirable, due to the preponderance of resulting compounds in numerous medicinally relevant compounds. However, the execution of the same is far from being trivial, due to the intrinsic electronic bias of anilines and concerns associated with the ready availability of an appropriate and odorless sulfur source. Accordingly, we demonstrate a metal- and additive-free, one-pot, multicomponent reaction between p-anisidines/anilines, carbon disulfide, and aliphatic amines to install an otherwise difficult meta-C–S bond on anilines with exclusive regioselectivity, while furnishing an array of biologically relevant anisidine-derived S-aryl dithiocarbamates. The method exhibits broad scope with appreciable functional group tolerance, as demonstrated through late-stage modification of a variety of amino acids, pharmaceuticals, and natural products. Importantly, final S-aryl dithiocarbamates are amenable to further synthetic manipulations, furnishing highly valuable and medicinally relevant sulfur-containing functional moieties, such as thiols, thioethers, and sulfones. Furthermore, in vitro evaluations demonstrate that many of the synthesized dithiocarbamates exhibit promising drug-like properties, demonstrating antiproliferative activity on a nanomolar level for breast cancer cell lines by affecting microtubule dynamics.
We report a metal-free multicomponent reaction that constructs meta-C−S bonds on anilines to afford S-aryl dithiocarbamates, featuring broad scope, facile diversification, and potent anticancer activity.
{"title":"Metal and Additive-Free Nondirected Meta-C–S Bond Formation on Anilines: Toward Biologically Relevant S-Aryl Dithiocarbamates","authors":"Sushanta Kumar Parida, , , Srishti Sanghi, , , Ardhendu Mondal, , , Nameeta Choudhary, , , Prahallad Meher, , , Priyanka Singh*, , and , Sandip Murarka*, ","doi":"10.1021/acscentsci.5c01231","DOIUrl":"https://doi.org/10.1021/acscentsci.5c01231","url":null,"abstract":"<p >The site-selective C–H functionalization to install meta-C–S bonds on aniline derivatives is highly desirable, due to the preponderance of resulting compounds in numerous medicinally relevant compounds. However, the execution of the same is far from being trivial, due to the intrinsic electronic bias of anilines and concerns associated with the ready availability of an appropriate and odorless sulfur source. Accordingly, we demonstrate a metal- and additive-free, one-pot, multicomponent reaction between <i>p</i>-anisidines/anilines, carbon disulfide, and aliphatic amines to install an otherwise difficult meta-C–S bond on anilines with exclusive regioselectivity, while furnishing an array of biologically relevant anisidine-derived <i>S</i>-aryl dithiocarbamates. The method exhibits broad scope with appreciable functional group tolerance, as demonstrated through late-stage modification of a variety of amino acids, pharmaceuticals, and natural products. Importantly, final <i>S</i>-aryl dithiocarbamates are amenable to further synthetic manipulations, furnishing highly valuable and medicinally relevant sulfur-containing functional moieties, such as thiols, thioethers, and sulfones. Furthermore, in vitro evaluations demonstrate that many of the synthesized dithiocarbamates exhibit promising drug-like properties, demonstrating antiproliferative activity on a nanomolar level for breast cancer cell lines by affecting microtubule dynamics.</p><p >We report a metal-free multicomponent reaction that constructs meta-C−S bonds on anilines to afford <i>S</i>-aryl dithiocarbamates, featuring broad scope, facile diversification, and potent anticancer activity.</p>","PeriodicalId":10,"journal":{"name":"ACS Central Science","volume":"11 11","pages":"2121–2132"},"PeriodicalIF":10.4,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acscentsci.5c01231","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145594425","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}