1
|
Jawed R, Bhatti H, Khan A. Genetic profile of ferroptosis in non-small cell lung carcinoma and pharmaceutical options for ferroptosis induction. Clin Transl Oncol 2025; 27:1867-1886. [PMID: 39460894 DOI: 10.1007/s12094-024-03754-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Lung cancer (LC) is the leading cause of cancer-related deaths and the second most commonly diagnosed malignancy worldwide. Lung adenocarcinoma (LUAD) and lung squamous cell LC (LUSCC) are the most common subtypes of non-small cell LC (NSCLC). Early diagnosis of LC can be challenging due to a lack of biomarkers. The overall survival (OS) of patients with NSCLC is still poor despite the enormous efforts that have been made to develop novel treatments. Understanding fundamental molecular and genetic mechanisms is necessary to develop new therapeutic approaches for NSCLC. A recently identified type of programmed cell death known as ferroptosis is one potential approach. Ferroptosis causes oxidative damage and the death of cancerous cells by peroxidizing unsaturated phospholipids and accumulating reactive oxygen species (ROS) in an iron-dependent manner. Ferroptosis-related gene (FRG) signatures have recently been evaluated for their ability to predict patient OS and prognosis. These analyses show FRGs are involved in cancer progression, and may serve as promising biomarkers for tumor diagnosis and therapy. Moreover, we summarize the current pharmaceutical options of ferroptosis induction and their underlying molecular mechanism in LC. Therefore, this review aims to provide a comprehensive summary of FRG-based prognostic models, their associated metabolic and signaling pathways, and promising therapeutic options for ferroptosis induction in NSCLC.
Collapse
Affiliation(s)
- Rohil Jawed
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210023, China.
| | - Huma Bhatti
- School of Chemistry and Chemical Engineering, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| | - Adnan Khan
- Clinical and Molecular Labs, Karachi Institute of Radiotherapy and Nuclear Medicine (KIRAN), KDA Scheme 33 Near Safoora Chowk, Karachi, Pakistan
| |
Collapse
|
2
|
Cáceres JC, Michellys NG, Greene BL. Nitric Oxide Inhibition of Glycyl Radical Enzymes and Their Activases. J Am Chem Soc 2025; 147:11777-11788. [PMID: 40133071 PMCID: PMC11987019 DOI: 10.1021/jacs.4c14786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/06/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Innate immune response cells produce high concentrations of the free radical nitric oxide (NO) in response to pathogen infection. The antimicrobial properties of NO include nonspecific damage to essential biomolecules and specific inactivation of enzymes central to aerobic metabolism. However, the molecular targets of NO in anaerobic metabolism are less understood. Here, we demonstrate that the Escherichia coli glycyl radical enzyme pyruvate formate lyase (PFL), which catalyzes the anaerobic metabolism of pyruvate, is irreversibly inhibited by NO. Using electron paramagnetic resonance and site-directed mutagenesis we show that NO destroys the glycyl radical of PFL. The activation of PFL by its cognate radical S-adenosyl-l-methionine-dependent activating enzyme (PFL-AE) is also inhibited by NO, resulting in the conversion of the essential iron-sulfur cluster to dinitrosyl iron complexes. Whole-cell EPR and metabolic flux analyses of anaerobically growing E. coli show that PFL and PFL-AE are inhibited by physiologically relevant levels of NO in bacterial cell cultures, resulting in diminished growth and a metabolic shift to lactate fermentation. The class III ribonucleotide reductase (RNR) glycyl radical enzyme and its corresponding RNR-AE are also inhibited by NO in a mechanism analogous to those observed in PFL and PFL-AE, which likely contributes to the bacteriostatic effect of NO. Based on the similarities in reactivity of the PFL/RNR and PFL-AE/RNR-AE enzymes with NO, the mechanism of inactivation by NO appears to be general to the respective enzyme classes. The results implicate an immunological role of NO in inhibiting glycyl radical enzyme chemistry in the gut.
Collapse
Affiliation(s)
- Juan Carlos Cáceres
- Interdisciplinary
Program in Quantitative Biosciences, University
of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Nathan G. Michellys
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, Santa
Barbara, California 93106, United States
| | - Brandon L. Greene
- Interdisciplinary
Program in Quantitative Biosciences, University
of California Santa Barbara, Santa Barbara, California 93106, United States
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, Santa
Barbara, California 93106, United States
| |
Collapse
|
3
|
Graciano A, Liu A. Protein-derived cofactors: chemical innovations expanding enzyme catalysis. Chem Soc Rev 2025. [PMID: 40151987 PMCID: PMC11951088 DOI: 10.1039/d4cs00981a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Indexed: 03/29/2025]
Abstract
Protein-derived cofactors, formed through posttranslational modification of a single amino acid or covalent crosslinking of amino acid side chains, represent a rapidly expanding class of catalytic moieties that redefine enzyme functionality. Once considered rare, these cofactors are recognized across all domains of life, with their repertoire growing from 17 to 38 types in two decades in our survey. Their biosynthesis proceeds via diverse pathways, including oxidation, metal-assisted rearrangements, and enzymatic modifications, yielding intricate motifs that underpin distinctive catalytic strategies. These cofactors span paramagnetic and non-radical states, including both mono-radical and crosslinked radical forms, sometimes accompanied by additional modifications. While their discovery has accelerated, mechanistic understanding lags, as conventional mutagenesis disrupts cofactor assembly. Emerging approaches, such as site-specific incorporation of non-canonical amino acids, now enable precise interrogation of cofactor biogenesis and function, offering a viable and increasingly rigorous means to gain mechanistic insights. Beyond redox chemistry and electron transfer, these cofactors confer enzymes with expanded functionalities. Recent studies have unveiled new paradigms, such as long-range remote catalysis and redox-regulated crosslinks as molecular switches. Advances in structural biology, mass spectrometry, and biophysical spectroscopy continue to elucidate their mechanisms. Moreover, synthetic biology and biomimetic chemistry are increasingly leveraging these natural designs to engineer enzyme-inspired catalysts. This review integrates recent advances in cofactor biogenesis, reactivity, metabolic regulation, and synthetic applications, highlighting the expanding chemical landscape and growing diversity of protein-derived cofactors and their far-reaching implications for enzymology, biocatalysis, and biotechnology.
Collapse
Affiliation(s)
- Angelica Graciano
- Department of Chemistry, The University of Texas at San Antonio, Texas 78249, USA.
| | - Aimin Liu
- Department of Chemistry, The University of Texas at San Antonio, Texas 78249, USA.
| |
Collapse
|
4
|
Wernet ND, Tecle E, Sarmiento MB, Kuo CJ, Chhan CB, Baick I, Batachari LE, Franklin L, Herneisen A, Bhabha G, Ekiert DC, Hanna-Rose W, Troemel ER. Adenosine deaminase and deoxyadenosine regulate intracellular immune response in C. elegans. iScience 2025; 28:111950. [PMID: 40034845 PMCID: PMC11872409 DOI: 10.1016/j.isci.2025.111950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 11/22/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
Adenosine deaminase (ADA) and purine nucleoside phosphorylase (PNP) are enzymes in the purine salvage pathway, which recycles purines to meet cellular demands. Mutations of these enzymes in humans cause inflammatory and immunodeficiency syndromes, but the mechanisms are not well understood. Prior work in the nematode Caenorhabditis elegans demonstrated that loss of PNP ortholog PNP-1 induced an immune response called the intracellular pathogen response (IPR). Here, we show that loss of the enzyme upstream of PNP-1 called ADAH-1 (ADA homolog) also induces the IPR and promotes resistance against intracellular pathogens. Unlike PNP-1, ADAH-1 is essential for organismal development. Importantly, we find that supplementation of deoxyadenosine, a substrate for ADA, induces the IPR and promotes resistance to intracellular pathogens in C. elegans, a finding we extend to human cells. Thus, mutations in ADA and PNP induce innate immunity through increased deoxyadenosine, a phenomenon that is conserved from C. elegans to humans.
Collapse
Affiliation(s)
- Nicole D. Wernet
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Eillen Tecle
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Cheng-Ju Kuo
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Crystal B. Chhan
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Ian Baick
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Lakshmi E. Batachari
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Latisha Franklin
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Alice Herneisen
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Gira Bhabha
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Damian C. Ekiert
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Wendy Hanna-Rose
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Emily R. Troemel
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
5
|
Xu D, Thomas WC, Burnim AA, Ando N. Conformational landscapes of a class I ribonucleotide reductase complex during turnover reveal intrinsic dynamics and asymmetry. Nat Commun 2025; 16:2458. [PMID: 40075098 PMCID: PMC11903788 DOI: 10.1038/s41467-025-57735-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Understanding the structural dynamics associated with enzymatic catalysis has been a long-standing goal of biochemistry. With the advent of modern cryo-electron microscopy (cryo-EM), it has become conceivable to redefine a protein's structure as the continuum of all conformations and their distributions. However, capturing and interpreting this information remains challenging. Here, we use classification and deep-learning-based analyses to characterize the conformational heterogeneity of a class I ribonucleotide reductase (RNR) during turnover. By converting the resulting information into physically interpretable 2D conformational landscapes, we demonstrate that RNR continuously samples a wide range of motions while maintaining surprising asymmetry to regulate the two halves of its turnover cycle. Remarkably, we directly observe the appearance of highly transient conformations needed for catalysis, as well as the interaction of RNR with its endogenous reductant thioredoxin also contributing to the asymmetry and dynamics of the enzyme complex. Overall, this work highlights the role of conformational dynamics in regulating key steps in enzyme mechanisms.
Collapse
Affiliation(s)
- Da Xu
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - William C Thomas
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
- Department of Chemistry, Princeton University, Princeton, NJ, 08544, USA
| | - Audrey A Burnim
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Nozomi Ando
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA.
- Department of Chemistry, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
6
|
Martinie RJ, Livada J, Kothiya N, Bollinger JM, Krebs C, Silakov A. Structural Elucidation of the Reduced Mn(III)/Fe(III) Intermediate of the Radical-Initiating Metallocofactor in Chlamydia trachomatis Ribonucleotide Reductase. Biochemistry 2025; 64:1157-1167. [PMID: 39960811 PMCID: PMC11883743 DOI: 10.1021/acs.biochem.4c00692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 03/05/2025]
Abstract
Ribonucleotide reductases (RNRs) are the sole de novo source of deoxyribonucleotides for DNA synthesis and repair across all organisms and carry out their reaction via a radical mechanism. RNR from Chlamydia trachomatis generates its turnover-initiating cysteinyl radical by long-range reduction of a Mn(IV)/Fe(III) cofactor, producing a Mn(III)/Fe(III) intermediate. Herein, we characterize the protonation states of the inorganic ligands in this reduced state using advanced pulse electron paramagnetic resonance (EPR) spectroscopy and 2H-isotope labeling. A strongly coupled deuteron is observed by hyperfine sublevel correlation (HYSCORE) spectroscopy experiments and indicates the presence of a bridging hydroxo ligand. Isotope-dependent EPR line broadening analysis and the magnitude of the estimated Mn-Fe exchange coupling constant together suggest a μ-oxo/μ-hydroxo core. Two distinct signals detected in electron-nuclear double resonance (ENDOR) spectra are attributable to less strongly coupled hydrons of a terminal water ligand to Mn(III). Together, these experiments imply that the reduced cofactor has a mixed μ-oxo/μ-hydroxo core with a terminal water ligand on Mn(III). This structural assignment sheds light generally on the reactivity of Mn/Fe heterobimetallic sites and, more specifically, on the proton-coupling in the electron transfer that initiates ribonucleotide reduction in this subclass of RNRs.
Collapse
Affiliation(s)
- Ryan J. Martinie
- Department
of Chemistry, Hamilton College, Clinton, New York 13323, United States
| | - Jovan Livada
- Department
of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Nyaari Kothiya
- Department
of Chemistry, Hamilton College, Clinton, New York 13323, United States
| | - J. Martin Bollinger
- Department
of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department
of Biochemistry and Molecular Biology, The
Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Carsten Krebs
- Department
of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department
of Biochemistry and Molecular Biology, The
Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Alexey Silakov
- Department
of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
7
|
Cáceres JC, Michellys NG, Greene BL. Nitric Oxide Inhibition of Glycyl Radical Enzymes and Their Activases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.23.639758. [PMID: 40060521 PMCID: PMC11888291 DOI: 10.1101/2025.02.23.639758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Innate immune response cells produce high concentrations of the free radical nitric oxide (NO) in response to pathogen infection. The antimicrobial properties of NO include non-specific damage to essential biomolecules and specific inactivation of enzymes central to aerobic metabolism. However, the molecular targets of NO in anaerobic metabolism are less understood. Here, we demonstrate that the Escherichia coli glycyl radical enzyme pyruvate formate lyase (PFL), which catalyzes the anaerobic metabolism of pyruvate, is irreversibly inhibited by NO. Using electron paramagnetic resonance and site-directed mutagenesis we show that NO destroys the glycyl radical of PFL. The activation of PFL by its cognate radical S-adenosyl-L-methionine-dependent activating enzyme (PFL-AE) is also inhibited by NO, resulting in the conversion of the essential iron-sulfur cluster to dinitrosyl iron complexes. Whole-cell EPR and metabolic flux analyses of anaerobically growing Escherichia coli show that PFL and PFL-AE are inhibited by physiologically relevant levels of NO in bacterial cell cultures, resulting in diminished growth and a metabolic shift to lactate fermentation. The class III ribonucleotide reductase (RNR) glycyl radical enzyme and its corresponding RNR-AE are also inhibited by NO in a mechanism analogous to those observed in PFL and PFL-AE, which likely contributes to the bacteriostatic effect of NO. Based on the similarities in reactivity of the PFL/RNR and PFL-AE/RNR-AE enzymes with NO, the mechanism of inactivation by NO appears to be general to the respective enzyme classes. The results implicate an immunological role of NO in inhibiting glycyl radical enzyme chemistry in the gut.
Collapse
Affiliation(s)
- Juan Carlos Cáceres
- Interdisciplinary Program in Quantitative Biosciences, University of California Santa Barbara, Santa Barbara, CA, 93106 United States
| | - Nathan G. Michellys
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA, 93106 United States
| | - Brandon L. Greene
- Interdisciplinary Program in Quantitative Biosciences, University of California Santa Barbara, Santa Barbara, CA, 93106 United States
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA, 93106 United States
| |
Collapse
|
8
|
Ziener J, Henao-Restrepo JA, Leonhardi J, Sturm MJ, Becker S, Morales-Prieto DM, Milde T, Beck JF, Sonnemann J. Combined inhibition of ribonucleotide reductase and WEE1 induces synergistic anticancer activity in Ewing's sarcoma cells. BMC Cancer 2025; 25:277. [PMID: 39962391 PMCID: PMC11831844 DOI: 10.1186/s12885-025-13691-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Ewing's sarcoma is a childhood bone and soft tissue cancer with poor prognosis. Treatment outcomes for Ewing's sarcoma patients have improved only modestly over the past decades, making the development of new treatment strategies paramount. In this study, the combined targeting of ribonucleotide reductase (RNR) and WEE1 was explored for its effectiveness against Ewing's sarcoma cells. METHODS The RNR inhibitor triapine and the WEE1 inhibitors adavosertib and ZN-c3 were tested in p53 wild-type and p53 mutant Ewing's sarcoma cells. The combination of adavosertib with the PARP inhibitors olaparib and veliparib was tested for comparison. Combinatorial effects were determined by flow cytometric analyses of cell death, loss of mitochondrial membrane potential and DNA fragmentation as well as by caspase 3/7 activity assay, immunoblotting and real-time RT-PCR. The drug interactions were assessed using combination index analysis. RESULTS RNR and WEE1 inhibitors were weakly to moderately effective on their own, but highly effective in combination. The combination treatments were similarly effective in p53 wild-type and p53 mutant cells. They synergistically induced cell death and cooperated to elicit mitochondrial membrane potential decay, to activate caspase 3/7 and to trigger DNA fragmentation, evidencing the induction of the apoptotic cell death cascade. They also cooperated to boost CHK1 phosphorylation, indicating augmented replication stress after combination treatment. In comparison, the combination of adavosertib with PARP inhibitors produced weaker synergistic effects. CONCLUSION Our findings show that combined inhibition of RNR and WEE1 was effective against Ewing's sarcoma in vitro. They thus provide a rationale for the evaluation of the potential of combined targeting of RNR and WEE1 in Ewing's sarcoma in vivo.
Collapse
Affiliation(s)
- Judy Ziener
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | | | - Johanna Leonhardi
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Max-Johann Sturm
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Sabine Becker
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Comprehensive Cancer Centre Central Germany (CCCG), Jena, Germany
| | | | - Till Milde
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Comprehensive Cancer Centre Central Germany (CCCG), Jena, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
| | - James F Beck
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Jürgen Sonnemann
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.
- Comprehensive Cancer Centre Central Germany (CCCG), Jena, Germany.
- Klinik für Kinder- und Jugendmedizin, Friedrich-Schiller-Universität Jena, Am Klinikum 1, D-07747, Jena, Germany.
| |
Collapse
|
9
|
Zhong J, Zhu Q, Soudackov AV, Hammes-Schiffer S. Hydrogen Tunneling and Conformational Motions in Nonadiabatic Proton-Coupled Electron Transfer between Interfacial Tyrosines in Ribonucleotide Reductase. J Am Chem Soc 2025; 147:4459-4468. [PMID: 39841588 PMCID: PMC11829447 DOI: 10.1021/jacs.4c15756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Ribonucleotide reductase (RNR) is essential for DNA synthesis and repair in all living organisms. The mechanism of E. coli RNR requires long-range radical transport through a proton-coupled electron transfer (PCET) pathway spanning two different protein subunits. Herein, the direct PCET reaction between the interfacial tyrosine residues, Y356 and Y731, is investigated with a vibronically nonadiabatic theory that treats the transferring proton and all electrons quantum mechanically. The input quantities to the PCET rate constant expression are computed with a combination of density functional theory and molecular dynamics simulations. The calculations highlight the importance of hydrogen tunneling in this PCET reaction. Compression of the distance between the proton donor and acceptor oxygen atoms of the interfacial tyrosine residues is essential to facilitate hydrogen tunneling by increasing the overlap between the reactant and product proton vibrational wave functions. This compression occurs by thermal conformational fluctuations of these interfacial tyrosine residues. N733 and R411 are identified as key residues that can hydrogen bond to Y731 and Y356, respectively, and thereby compete with the hydrogen-bonding interaction between Y731 and Y356 required for direct PCET. Understanding the roles of hydrogen tunneling and conformational motions in this interfacial PCET reaction, as well as identifying other residues that may impact the kinetics, is important for targeted protein engineering efforts to modulate RNR activity.
Collapse
Affiliation(s)
- Jiayun Zhong
- Department of Chemistry, Yale University, New Haven, Connecticut 06520, United States
| | - Qiwen Zhu
- Department of Chemistry, Yale University, New Haven, Connecticut 06520, United States
| | | | - Sharon Hammes-Schiffer
- Department of Chemistry, Yale University, New Haven, Connecticut 06520, United States
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
10
|
Li RN, Chen SL. Recent Insights into the Reaction Mechanisms of Non-Heme Diiron Enzymes Containing Oxoiron(IV) Complexes. Chembiochem 2025; 26:e202400788. [PMID: 39508533 DOI: 10.1002/cbic.202400788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/15/2024]
Abstract
Oxoiron(IV) complexes are key intermediates in the catalytic reactions of some non-heme diiron enzymes. These enzymes, across various subfamilies, activate dioxygen to generate high-valent diiron-oxo species, which, in turn, drive the activation of substrates and mediate a variety of challenging oxidative transformations. In this review, we summarize the structures, formation mechanisms, and functions of high-valent diiron-oxo intermediates in eight representative diiron enzymes (sMMO, RNR, ToMO, MIOX, PhnZ, SCD1, AlkB, and SznF) spanning five subfamilies. We also categorize and analyze the structural and mechanistic differences among these enzymes.
Collapse
Affiliation(s)
- Rui-Ning Li
- Key Laboratory of Cluster Science of Ministry of Education, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Shi-Lu Chen
- Key Laboratory of Cluster Science of Ministry of Education, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
11
|
Guo L, Zhao Y, Bai X, Wang X, Tuoheti K, Cao Y, Zuo Y, Zhang X, Liu T. RRM2 Is a Putative Biomarker and Promotes Bladder Cancer Progression via PI3K/AKT/mTOR Pathway. J Cell Physiol 2025; 240:e31501. [PMID: 39676643 DOI: 10.1002/jcp.31501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/24/2024] [Accepted: 11/25/2024] [Indexed: 12/17/2024]
Abstract
Bladder cancer (BLCA) is the tenth most common cancer worldwide, characterized by its high recurrence and progression rates. Thus, identifying prognostic biomarkers and understanding its underlying mechanisms are imperative to enhance patient outcomes. In this study, we aimed to investigate the prognostic significance, expression, functional activity, and underlying mechanisms of RRM2 in BLCA. RRM2 expression and its association with pathological grading and survival were investigated in samples from TCGA dataset and BLCA tissue microarray. CCK8 assays, colony formation assays, wound healing, and transwell assays were performed to assess the role of RRM2 in BLCA cell proliferation and migration. Western blot was employed to investigate alterations in markers associated with epithelial-to-mesenchymal transition (EMT), apoptosis, and cell cycle regulation. Gene set enrichment analysis was performed to investigate the biological processes associated with RRM2, which were subsequently validated. The expression of RRM2 was significantly elevated in both BLCA tissues and cells. Our results also indicated a positive correlation between RRM2 expression and high tumor stage, high tumor grade, and poor survival. Knockdown of RRM2 inhibited cell proliferation and migration of BLCA. RRM2 knockdown significantly induced apoptosis and arrested the cell cycle at the G0/G1 phase. Opposite results were observed in the RRM2 overexpression cells. Additionally, our study demonstrates that RRM2 promotes BLCA progression by activating the PI3K/AKT/mTOR pathway. RRM2 is remarkably correlated with poor prognosis in BLCA and facilitate its progression via PI3K/AKT/mTOR pathway. It is suggested that RRM2 might become an effective prognostic biomarker and potential therapeutic target for BLCA.
Collapse
Affiliation(s)
- Linfa Guo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yiqiao Zhao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaojie Bai
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaolong Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kuerban Tuoheti
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuanfei Cao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yingtong Zuo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xinhua Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tongzu Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
12
|
Rodríguez I, Acosta C, Nieves-Escobar C, Strangmark E, Claudio-Ares O, Vargas Figueroa AI, Soto-Millán AM, M. Orta-Rivera A, Astashkin AV, Tinoco AD. DefNEtTrp: An Iron Dual Chelator Approach for Anticancer Application. JACS AU 2024; 4:4799-4808. [PMID: 39735911 PMCID: PMC11672142 DOI: 10.1021/jacsau.4c00774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024]
Abstract
Targeting iron metabolism has emerged as a novel therapeutic strategy for the treatment of cancer. As such, iron chelator drugs are repurposed or specifically designed as anticancer agents. Two important chelators, deferasirox (Def) and triapine (Trp), attack the intracellular supply of iron (Fe) and inhibit Fe-dependent pathways responsible for cellular proliferation and metastasis. Trp, in particular, forms a redox active ferrous complex that inactivates the Fe-dependent ribonucleotide reductase (RNR), responsible for DNA replication. Building on recent efforts to employ intracellular Fe chelation for anticancer therapy, this work aimed to develop the Fe dual chelator ligand DefNEtTrp, consisting of the Def and Trp moieties, to exploit their high affinity Fe(II/III) binding and redox modulation. Using UV-vis spectroscopy, EPR spectroscopy, ESI and MALDI-TOF mass spectrometry, and cyclic voltammetry analyses, DefNEtTrp is shown to retain its Fe binding at both chelator moieties and generate a redox active Fe(III) complex Fe3(DefNEtTrp)2 featuring a reduction potential (E 1/2 = +0.103 V vs normal hydrogen electrode) within the biological window. Screened against different cancer cell line types, DefNEtTrp exhibits potent and broad-spectrum antiproliferative and cell death behavior. Its cytotoxicity (IC50 0.77 ± 0.06 μM) is superior to that of unconjugated Def and Trp ligands (IC50 2.6 ± 0.15 μM and 1.1 ± 0.04 μM, respectively) in single-compound and combination treatments and is selective toward cancer cells. The cell death mechanism of the dual chelator is assessed in the context of intracellular labile Fe binding and was found to induce both apoptosis and ferroptosis.
Collapse
Affiliation(s)
- Israel Rodríguez
- Department
of Chemistry, University of Puerto Rico,
Río Piedras Campus, Río
Piedras, Puerto Rico 00931, United States
| | - Carmen Acosta
- Department
of Chemistry, University of Puerto Rico,
Río Piedras Campus, Río
Piedras, Puerto Rico 00931, United States
| | - Christopher Nieves-Escobar
- Department
of Chemistry, University of Puerto Rico,
Río Piedras Campus, Río
Piedras, Puerto Rico 00931, United States
| | - Estelle Strangmark
- Department
of Chemistry, University of Puerto Rico,
Río Piedras Campus, Río
Piedras, Puerto Rico 00931, United States
| | - Oscar Claudio-Ares
- Department
of Chemistry, University of Puerto Rico,
Río Piedras Campus, Río
Piedras, Puerto Rico 00931, United States
| | - Adriana I. Vargas Figueroa
- Department
of Chemistry, University of Puerto Rico,
Río Piedras Campus, Río
Piedras, Puerto Rico 00931, United States
| | - Alexandra M. Soto-Millán
- Department
of Chemistry, University of Puerto Rico,
Río Piedras Campus, Río
Piedras, Puerto Rico 00931, United States
| | - Aixa M. Orta-Rivera
- Department
of Chemistry, University of Puerto Rico,
Río Piedras Campus, Río
Piedras, Puerto Rico 00931, United States
| | - Andrei V. Astashkin
- Department
of Chemistry and Biochemistry, The University
of Arizona, Tucson, Arizona 85721-0041, United States
| | - Arthur D. Tinoco
- Department
of Chemistry, University of Puerto Rico,
Río Piedras Campus, Río
Piedras, Puerto Rico 00931, United States
| |
Collapse
|
13
|
Yang S, Wang R, Liu L, Xu F, Zhao X, Yao Z, Zhang J, Cheng X, Xu A, Wu L, Zhao G. RRM1 promotes homologous recombination and radio/chemo-sensitivity via enhancing USP11 and E2F1-mediated RAD51AP1 transcription. Cell Death Discov 2024; 10:496. [PMID: 39695160 DOI: 10.1038/s41420-024-02267-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Abstract
Ribonucleotide reductase M1 (RRM1), the catalytic subunit of ribonucleotide reductase, plays a pivotal role in converting ribonucleotides (NTP) into deoxyribonucleotides (dNTP), essential for DNA replication and repair. Elevated RRM1 expression is associated with various human cancers, correlating with poorer prognosis and reduced overall survival rates. Our previous study found that RRM1 will enter the nucleus to promote DNA damage repair. However, the underlying mechanism remains elusive. Here, we unveil a novel role of RRM1 in promoting homologous recombination (HR) by upregulating the expression of RAD51AP1, a critical HR factor, in an E2F1-dependent manner. We demonstrate that RRM1 interacts with USP11 in the cytoplasm, and the recruitment of RRM1 to LaminB1 induced by ionizing radiation (IR) facilitates the binding of USP11 to the nuclear pore complex (NPC), promoting USP11 entry into the nucleus. Upon nuclear translocation, USP11 binds to E2F1 and inhibits the ubiquitin-mediated degradation of E2F1, thereby enhancing the transcriptional expression of RAD51AP1. Moreover, a specific RRM1 mutant lacking amino acids 731-793, crucial for its interaction with USP11 and recruitment to LaminB1, exhibits a dominant-negative effect on RAD51AP1 expression and HR. Truncations of RRM1 fail to inhibit the ubiquitin-mediated degradation of E2F1 and cannot promote the E2F1-mediated transactivation of RAD51AP1. Lastly, the full length of RRM1, not truncations, enhances tumor cells' sensitivity to IR, underscoring its importance in radiotherapy resistance. Collectively, our results suggest a novel function of RRM1 in promoting HR-mediated DSB repair through positive regulation of RAD51AP1 transcription by direct interaction with USP11 and promoting subsequent USP11-mediated deubiquitination of E2F1. Our findings elucidate a previously unknown mechanism whereby RRM1 promotes HR-mediated DNA repair, presenting a potential therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Shuai Yang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Ruru Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Lingling Liu
- University of Science and Technology of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Feng Xu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- University of Science and Technology of China, Hefei, Anhui, China
| | - Xipeng Zhao
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- University of Science and Technology of China, Hefei, Anhui, China
| | - Zhicheng Yao
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- University of Science and Technology of China, Hefei, Anhui, China
| | - Jie Zhang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- University of Science and Technology of China, Hefei, Anhui, China
| | - Xu Cheng
- University of Science and Technology of China, Hefei, Anhui, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - An Xu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Lijun Wu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China
| | - Guoping Zhao
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.
| |
Collapse
|
14
|
Giang LH, Wu KS, Lee WC, Chu SS, Do AD, Huang MH, Lin YL, Hsieh CL, Sung SY, Yen Y, Wong TT, Chang CC. RRM2 inhibition alters cell cycle through ATM/Rb/E2F1 pathway in atypical teratoid rhabdoid tumor. Neoplasia 2024; 58:101075. [PMID: 39437704 PMCID: PMC11536058 DOI: 10.1016/j.neo.2024.101075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/28/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Atypical teratoid rhabdoid tumor (ATRT) is an aggressive brain tumor that mainly affects young children. Our recent study reported a promising therapeutic strategy to trigger DNA damage, impede homologous recombination repair, and induce apoptosis in ATRT cells by targeting ribonucleotide reductase regulatory subunit M2 (RRM2). COH29, an inhibitor of RRM2, effectively reduced tumor growth and prolonged survival in vivo. Herein, we explored the underlying mechanisms controlling these functions to improve the clinical applicability of COH29 in ATRT. METHODS Molecular profiling of ATRT patients and COH29-treated cells was analyzed to identify the specific signaling pathways, followed by validation using a knockdown system, flow cytometry, q-PCR, and western blot. RESULTS Elevated E2F1 and its signaling pathway were correlated with poor prognosis. RRM2 inhibition induced DNA damage and activated ATM, which reduced Rb phosphorylation to promote Rb-E2F1 interaction and hindered E2F1 functions. E2F1 activity suppression led to decreased E2F1-dependent target expressions, causing cell cycle arrest in the G1 phase, decreased S phase cells, and blocked DNA damage repair. CONCLUSION Our study highlights the role of ATM/Rb/E2F1 pathway in controlling cell cycle arrest and apoptosis in response to RRM2 inhibition-induced DNA damage. This provides insight into the therapeutic benefits of COH29 and suggests targeting this pathway as a potential treatment for ATRT.
Collapse
Affiliation(s)
- Le Hien Giang
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Department of Biology and Genetics, Hai Phong University of Medicine and Pharmacy, Hai Phong 180000, Vietnam
| | - Kuo-Sheng Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Wei-Chung Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Shing-Shung Chu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Anh Duy Do
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Department of Physiology, Pathophysiology and Immunology, Pham Ngoc Thach University of Medicine, Ho Chi Minh City 700000, Vietnam
| | - Man-Hsu Huang
- Department of Pathology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
| | - Yu-Ling Lin
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chia-Ling Hsieh
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; General Clinical Research Center, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Shian-Ying Sung
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Yun Yen
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Tai-Tong Wong
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Pediatric Brain Tumor Program, Taipei Cancer Center, Taipei Medical University, Taipei 110, Taiwan; Division of Pediatric Neurosurgery, Department of Neurosurgery, Taipei Medical University Hospital and Taipei Neuroscience Institute, Taipei Medical University, Taipei 110, Taiwan; Neuroscience Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; TMU Research Center for Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Che-Chang Chang
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Neuroscience Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan; TMU Research Center for Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan; Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan; Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei 11031, Taiwan.
| |
Collapse
|
15
|
López-Gil CI, Téllez-Jurado A, Velasco-Velázquez MA, Anducho-Reyes MA. Identification and Analysis of Anticancer Therapeutic Targets from the Polysaccharide Krestin (PSK) and Polysaccharopeptide (PSP) Using Inverse Docking. Molecules 2024; 29:5390. [PMID: 39598781 PMCID: PMC11596896 DOI: 10.3390/molecules29225390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
The natural compounds PSK and PSP have antitumor and immunostimulant properties. These pharmacological benefits have been documented in vitro and in vivo, although there is no information in silico which describes the action mechanisms at the molecular level. In this study, the inverse docking method was used to identify the interactions of PSK and PSP with two local databases: BPAT with 66 antitumor proteins, and BPSIC with 138 surfaces and intracellular proteins. This led to the identification interactions and similarities of PSK and the AB680 inhibitor in the active site of CD73. It was also found that PSK binds to CD59, interacting with the amino acids APS22 and PHE23, which coincide with the rlLYd4 internalization inhibitor. With the isoform of the K-RAS protein, PSK bonded to the TYR32 amino acid at switch 1, while with BAK it bonded to the region of the α1 helix, while PSP bonded to the activation site and the C-terminal and N-terminal ends of that helix. In Bcl-2, PSK interacted at the binding site of the Venetoclax inhibitor, showing similarities with the amino acids ASP111, VAL133, LEU137, MET115, PHE112, and TYR108, while PSP had similarities with THR132, VAL133, LEU137, GLN118, MET115, APS111, PHE112, and PHE104.
Collapse
Affiliation(s)
- Carlos Iván López-Gil
- Department of Biotechnology, Universidad Politécnica de Pachuca, Zempoala 43830, Mexico; (C.I.L.-G.); (A.T.-J.)
| | - Alejandro Téllez-Jurado
- Department of Biotechnology, Universidad Politécnica de Pachuca, Zempoala 43830, Mexico; (C.I.L.-G.); (A.T.-J.)
| | | | - Miguel Angel Anducho-Reyes
- Department of Biotechnology, Universidad Politécnica de Pachuca, Zempoala 43830, Mexico; (C.I.L.-G.); (A.T.-J.)
| |
Collapse
|
16
|
Westmoreland DE, Feliciano PR, Kang G, Cui C, Kim A, Stubbe J, Nocera DG, Drennan CL. 2.6-Å resolution cryo-EM structure of a class Ia ribonucleotide reductase trapped with mechanism-based inhibitor N 3CDP. Proc Natl Acad Sci U S A 2024; 121:e2417157121. [PMID: 39475643 PMCID: PMC11551348 DOI: 10.1073/pnas.2417157121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/03/2024] [Indexed: 11/06/2024] Open
Abstract
Ribonucleotide reductases (RNRs) reduce ribonucleotides to deoxyribonucleotides using radical-based chemistry. For class Ia RNRs, the radical species is stored in a separate subunit (β2) from the subunit housing the active site (α2), requiring the formation of a short-lived α2β2 complex and long-range radical transfer (RT). RT occurs via proton-coupled electron transfer (PCET) over a long distance (~32-Å) and involves the formation and decay of multiple amino acid radical species. Here, we use cryogenic electron microscopy and a mechanism-based inhibitor 2'-azido-2'-deoxycytidine-5'-diphosphate (N3CDP) to trap a wild-type α2β2 complex of Escherichia coli class Ia RNR. We find that one α subunit has turned over and that the other is trapped, bound to β in a midturnover state. Instead of N3CDP in the active site, forward RT has resulted in N2 loss, migration of the third nitrogen from the ribose C2' to C3' positions, and attachment of this nitrogen to the sulfur of cysteine-225. In this study, an inhibitor has been visualized as an adduct to an RNR. Additionally, this structure reveals the positions of PCET residues following forward RT, complementing the previous structure that depicted a preturnover PCET pathway and suggesting how PCET is gated at the α-β interface. This N3CDP-trapped structure is also of sufficient resolution (2.6 Å) to visualize water molecules, allowing us to evaluate the proposal that water molecules are proton acceptors and donors as part of the PCET process.
Collapse
Affiliation(s)
- Dana E Westmoreland
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Patricia R Feliciano
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- HHMI, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Gyunghoon Kang
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- HHMI, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Chang Cui
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Albert Kim
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - JoAnne Stubbe
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Daniel G Nocera
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Catherine L Drennan
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- HHMI, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
17
|
Chemla Y, Kaufman F, Amiram M, Alfonta L. Expanding the Genetic Code of Bioelectrocatalysis and Biomaterials. Chem Rev 2024; 124:11187-11241. [PMID: 39377473 DOI: 10.1021/acs.chemrev.4c00077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Genetic code expansion is a promising genetic engineering technology that incorporates noncanonical amino acids into proteins alongside the natural set of 20 amino acids. This enables the precise encoding of non-natural chemical groups in proteins. This review focuses on the applications of genetic code expansion in bioelectrocatalysis and biomaterials. In bioelectrocatalysis, this technique enhances the efficiency and selectivity of bioelectrocatalysts for use in sensors, biofuel cells, and enzymatic electrodes. In biomaterials, incorporating non-natural chemical groups into protein-based polymers facilitates the modification, fine-tuning, or the engineering of new biomaterial properties. The review provides an overview of relevant technologies, discusses applications, and highlights achievements, challenges, and prospects in these fields.
Collapse
|
18
|
Saura P, Kim H, Beghiah A, Young L, Moore AL, Kaila VRI. Proton-coupled electron transfer dynamics in the alternative oxidase. Chem Sci 2024:d4sc05060f. [PMID: 39444558 PMCID: PMC11492382 DOI: 10.1039/d4sc05060f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024] Open
Abstract
The alternative oxidase (AOX) is a membrane-bound di-iron enzyme that catalyzes O2-driven quinol oxidation in the respiratory chains of plants, fungi, and several pathogenic protists of biomedical and industrial interest. Yet, despite significant biochemical and structural efforts over the last decades, the catalytic principles of AOX remain poorly understood. We develop here multi-scale quantum and classical molecular simulations in combination with biochemical experiments to address the proton-coupled electron transfer (PCET) reactions responsible for catalysis in AOX from Trypanosoma brucei, the causative agent of sleeping sickness. We show that AOX activates and splits dioxygen via a water-mediated PCET reaction, resulting in a high-valent ferryl/ferric species and tyrosyl radical (Tyr220˙) that drives the oxidation of the quinol via electric field effects. We identify conserved carboxylates (Glu215, Asp100) within a buried cluster of ion-pairs that act as a transient proton-loading site in the quinol oxidation process, and validate their function experimentally with point mutations that result in drastic activity reduction and pK a-shifts. Our findings provide a key mechanistic understanding of the catalytic machinery of AOX, as well as a molecular basis for rational drug design against energy transduction chains of parasites. On a general level, our findings illustrate how redox-triggered conformational changes in ion-paired networks control the catalysis via electric field effects.
Collapse
Affiliation(s)
- Patricia Saura
- Department of Biochemistry and Biophysics, Stockholm University Stockholm 10691 Sweden
| | - Hyunho Kim
- Department of Biochemistry and Biophysics, Stockholm University Stockholm 10691 Sweden
| | - Adel Beghiah
- Department of Biochemistry and Biophysics, Stockholm University Stockholm 10691 Sweden
| | - Luke Young
- Biochemistry and Biomedicine, School of Life Sciences, University of Sussex Falmer Brighton BN1 9QG UK
| | - Anthony L Moore
- Biochemistry and Biomedicine, School of Life Sciences, University of Sussex Falmer Brighton BN1 9QG UK
| | - Ville R I Kaila
- Department of Biochemistry and Biophysics, Stockholm University Stockholm 10691 Sweden
| |
Collapse
|
19
|
Westmoreland DE, Feliciano PR, Kang G, Cui C, Kim A, Stubbe J, Nocera DG, Drennan CL. 2.6-Å resolution cryo-EM structure of a class Ia ribonucleotide reductase trapped with mechanism-based inhibitor N 3CDP. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.09.617422. [PMID: 39416103 PMCID: PMC11482829 DOI: 10.1101/2024.10.09.617422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Ribonucleotide reductases (RNRs) reduce ribonucleotides to deoxyribonucleotides using radical-based chemistry. For class Ia RNRs, the radical species is stored in a separate subunit (β2) from the subunit housing the active site (α2), requiring the formation of a short-lived α2β2 complex and long-range radical transfer (RT). RT occurs via proton-coupled electron transfer (PCET) over a long distance (~32-Å) and involves the formation and decay of multiple amino acid radical species. Here, we use cryogenic-electron microscopy and a mechanism-based inhibitor 2'-azido-2'-deoxycytidine-5'-diphosphate (N3CDP) to trap a wild-type α2β2 complex of E. coli class Ia RNR. We find that one α subunit has turned over and that the other is trapped, bound to β in a mid-turnover state. Instead of N3CDP in the active site, forward RT has resulted in N2 loss, migration of the third nitrogen from the ribose C2' to C3' positions, and attachment of this nitrogen to the sulfur of cysteine-225. To the best of our knowledge, this is the first time an inhibitor has been visualized as an adduct to an RNR. Additionally, this structure reveals the positions of PCET residues following forward RT, complementing the previous structure that depicted a pre-turnover PCET pathway and suggesting how PCET is gated at the α-β interface. This N3CDP-trapped structure is also of sufficient resolution (2.6 Å) to visualize water molecules, allowing us to evaluate the proposal that water molecules are proton acceptors and donors as part of the PCET process.
Collapse
Affiliation(s)
- Dana E. Westmoreland
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Patricia R. Feliciano
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Gyunghoon Kang
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Chang Cui
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Albert Kim
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - JoAnne Stubbe
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Daniel G. Nocera
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Catherine L. Drennan
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
20
|
Alabugin IV, Eckhardt P, Christopher KM, Opatz T. The Photoredox Paradox: Electron and Hole Upconversion as the Hidden Secrets of Photoredox Catalysis. J Am Chem Soc 2024; 146:27233-27254. [PMID: 39316772 DOI: 10.1021/jacs.4c10422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Although photoredox catalysis is complex from a mechanistic point of view, it is also often surprisingly efficient. In fact, the quantum efficiency of a puzzlingly large portion of photoredox reactions exceeds 100% (i.e., the measured quantum yields (QYs) are >1). Hence, these photoredox reactions can be more than perfect with respect to photon utilization. In several documented cases, a single absorbed photon can lead to the formation of >100 molecules of the product, behavior known to originate from chain processes. In this Perspective, we explore the underlying reasons for this efficiency, identify the nature of common catalytic chains, and highlight the differences between HAT and SET chains. Our goal is to show why chains are especially important in photoredox catalysis and where the thermodynamic driving force that sustains the SET catalytic cycles comes from. We demonstrate how the interplay of polar and radical processes can activate hidden catalytic pathways mediated by electron and hole transfer (i.e., electron and hole catalysis). Furthermore, we illustrate how the phenomenon of redox upconversion serves as a thermodynamic precondition for electron and hole catalysis. After discussing representative mechanistic puzzles, we analyze the most common bond forming steps, where redox upconversion frequently occurs (and issometimes unavoidable). In particular, we highlight the importance of 2-center-3-electron bonds as a recurring motif that allows a rational chemical approach to the design of redox upconversion processes.
Collapse
Affiliation(s)
- Igor V Alabugin
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306-4390, United States
| | - Paul Eckhardt
- Department of Chemistry, Johannes Gutenberg University, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Kimberley M Christopher
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306-4390, United States
| | - Till Opatz
- Department of Chemistry, Johannes Gutenberg University, Duesbergweg 10-14, 55128 Mainz, Germany
| |
Collapse
|
21
|
Fatima S, Olshansky L. Conformational control over proton-coupled electron transfer in metalloenzymes. Nat Rev Chem 2024; 8:762-775. [PMID: 39223400 PMCID: PMC11531298 DOI: 10.1038/s41570-024-00646-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
From the reduction of dinitrogen to the oxidation of water, the chemical transformations catalysed by metalloenzymes underlie global geochemical and biochemical cycles. These reactions represent some of the most kinetically and thermodynamically challenging processes known and require the complex choreography of the fundamental building blocks of nature, electrons and protons, to be carried out with utmost precision and accuracy. The rate-determining step of catalysis in many metalloenzymes consists of a protein structural rearrangement, suggesting that nature has evolved to leverage macroscopic changes in protein molecular structure to control subatomic changes in metallocofactor electronic structure. The proton-coupled electron transfer mechanisms operative in nitrogenase, photosystem II and ribonucleotide reductase exemplify this interplay between molecular and electronic structural control. We present the culmination of decades of study on each of these systems and clarify what is known regarding the interplay between structural changes and functional outcomes in these metalloenzyme linchpins.
Collapse
Affiliation(s)
- Saman Fatima
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Lisa Olshansky
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Materials Research Laboratory, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- The Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
22
|
Funk M, Zimanyi CM, Andree GA, Hamilos AE, Drennan CL. How ATP and dATP Act as Molecular Switches to Regulate Enzymatic Activity in the Prototypical Bacterial Class Ia Ribonucleotide Reductase. Biochemistry 2024; 63:2517-2531. [PMID: 39164005 PMCID: PMC11447812 DOI: 10.1021/acs.biochem.4c00329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/22/2024]
Abstract
Class Ia ribonucleotide reductases (RNRs) are allosterically regulated by ATP and dATP to maintain the appropriate deoxyribonucleotide levels inside the cell for DNA biosynthesis and repair. RNR activity requires precise positioning of the β2 and α2 subunits for the transfer of a catalytically essential radical species. Excess dATP inhibits RNR through the creation of an α-β interface that restricts the ability of β2 to obtain a position that is capable of radical transfer. ATP breaks the α-β interface, freeing β2 and restoring enzyme activity. Here, we investigate the molecular basis for allosteric activity regulation in the well-studied Escherichia coli class Ia RNR through the determination of six crystal structures and accompanying biochemical and mutagenesis studies. We find that when dATP is bound to the N-terminal regulatory cone domain in α, a helix unwinds, creating a binding surface for β. When ATP displaces dATP, the helix rewinds, dismantling the α-β interface. This reversal of enzyme inhibition requires that two ATP molecules are bound in the cone domain: one in the canonical nucleotide-binding site (site 1) and one in a site (site 2) that is blocked by phenylalanine-87 and tryptophan-28 unless ATP is bound in site 1. When ATP binds to site 1, histidine-59 rearranges, prompting the movement of phenylalanine-87 and trytophan-28, and creating site 2. dATP hydrogen bonds to histidine-59, preventing its movement. The importance of site 2 in the restoration of RNR activity by ATP is confirmed by mutagenesis. These findings have implications for the design of bacterial RNR inhibitors.
Collapse
Affiliation(s)
- Michael
A. Funk
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Christina M. Zimanyi
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Gisele A. Andree
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Allison E. Hamilos
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
| | - Catherine L. Drennan
- Department
of Chemistry, Massachusetts Institute of
Technology, Cambridge, Massachusetts 02139, United States
- Department
of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Howard
Hughes Medical Institute, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Center
for Environmental Health Sciences, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
23
|
Jones BJ, Greene BL. Singlet oxygen-mediated photochemical cross-linking of an engineered fluorescent flavoprotein iLOV. J Biol Chem 2024; 300:107845. [PMID: 39357827 PMCID: PMC11541845 DOI: 10.1016/j.jbc.2024.107845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
Genetically encoded photoactive proteins are integral tools in modern biochemical and molecular biological research. Within this tool box, truncated variants of the phototropin two light-oxygen-voltage flavoprotein have been developed to photochemically generate singlet oxygen (1O2) in vitro and in vivo, yet the effect of 1O2 on these genetically encoded photosensitizers remains underexplored. In this study, we demonstrate that the "improved" light-oxygen-voltage flavoprotein is capable of photochemical 1O2 generation. Once generated, 1O2 induces protein oligomerization via covalent cross-linking. The molecular targets of protein oligomerization by cross-linking are not endogenous tryptophans or tyrosines, but rather primarily histidines. Substitution of surface-exposed histidines for serine or glycine residues effectively eliminates protein cross-linking. When used in biochemical applications, such protein-protein cross-links may interfere with native biological responses to 1O2, which can be ameliorated by substitution of the surface exposed histidines of improved" light-oxygen-voltage or other 1O2-generating flavoproteins.
Collapse
Affiliation(s)
- Benjamin J Jones
- Department of Chemistry and Biochemistry the University of California Santa Barbara, Santa Barbara, California, USA
| | - Brandon L Greene
- Department of Chemistry and Biochemistry the University of California Santa Barbara, Santa Barbara, California, USA; Biomolecular Science and Engineering Program, The University of California Santa Barbara, Santa Barbara, California, USA.
| |
Collapse
|
24
|
Owusu SB, Zaher A, Ahenkorah S, Pandya DN, Wadas TJ, Petronek MS. Gallium Uncouples Iron Metabolism to Enhance Glioblastoma Radiosensitivity. Int J Mol Sci 2024; 25:10047. [PMID: 39337531 PMCID: PMC11432413 DOI: 10.3390/ijms251810047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Gallium-based therapy has been considered a potentially effective cancer therapy for decades and has recently re-emerged as a novel therapeutic strategy for the management of glioblastoma tumors. Gallium targets the iron-dependent phenotype associated with aggressive tumors by mimicking iron in circulation and gaining intracellular access through transferrin-receptor-mediated endocytosis. Mechanistically, it is believed that gallium inhibits critical iron-dependent enzymes like ribonucleotide reductase and NADH dehydrogenase (electron transport chain complex I) by replacing iron and removing the ability to transfer electrons through the protein secondary structure. However, information regarding the effects of gallium on cellular iron metabolism is limited. As mitochondrial iron metabolism serves as a central hub of the iron metabolic network, the goal of this study was to investigate the effects of gallium on mitochondrial iron metabolism in glioblastoma cells. Here, it has been discovered that gallium nitrate can induce mitochondrial iron depletion, which is associated with the induction of DNA damage. Moreover, the generation of gallium-resistant cell lines reveals a highly unstable phenotype characterized by impaired colony formation associated with a significant decrease in mitochondrial iron content and loss of the mitochondrial iron uptake transporter, mitoferrin-1. Moreover, gallium-resistant cell lines are significantly more sensitive to radiation and have an impaired ability to repair any sublethal damage and to survive potentially lethal radiation damage when left for 24 h following radiation. These results support the hypothesis that gallium can disrupt mitochondrial iron metabolism and serve as a potential radiosensitizer.
Collapse
Affiliation(s)
- Stephenson B. Owusu
- Department of Radiation Oncology, Division of Free Radical and Radiation Biology, The University of Iowa, Iowa City, IA 52242, USA; (S.B.O.); (A.Z.)
| | - Amira Zaher
- Department of Radiation Oncology, Division of Free Radical and Radiation Biology, The University of Iowa, Iowa City, IA 52242, USA; (S.B.O.); (A.Z.)
| | - Stephen Ahenkorah
- Department of Radiology, The University of Iowa, Iowa City, IA 52242, USA; (S.A.)
| | - Darpah N. Pandya
- Department of Radiology, The University of Iowa, Iowa City, IA 52242, USA; (S.A.)
| | - Thaddeus J. Wadas
- Department of Radiology, The University of Iowa, Iowa City, IA 52242, USA; (S.A.)
| | - Michael S. Petronek
- Department of Radiation Oncology, Division of Free Radical and Radiation Biology, The University of Iowa, Iowa City, IA 52242, USA; (S.B.O.); (A.Z.)
| |
Collapse
|
25
|
Zhao Y, Xue S, Wei D, Zhang J, Zhang N, Mao L, Liu N, Zhao L, Yan J, Wang Y, Cai X, Zhu S, Roessler S, Ji J. Membrane RRM2-positive cells represent a malignant population with cancer stem cell features in intrahepatic cholangiocarcinoma. J Exp Clin Cancer Res 2024; 43:255. [PMID: 39243109 PMCID: PMC11378576 DOI: 10.1186/s13046-024-03174-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (iCCA) is one of the most lethal malignancies and highly heterogeneous. We thus aimed to identify and characterize iCCA cell subpopulations with severe malignant features. METHODS Transcriptomic datasets from three independent iCCA cohorts (iCCA cohorts 1-3, n = 382) and formalin-fixed and paraffin-embedded tissues from iCCA cohort 4 (n = 31) were used. An unbiased global screening strategy was established, including the transcriptome analysis with the activated malignancy/stemness (MS) signature in iCCA cohorts 1-3 and the mass spectrometry analysis of the sorted stemness reporter-positive iCCA cells. A group of cellular assays and subcutaneous tumor xenograft assay were performed to investigate functional roles of the candidate. Immunohistochemistry was performed in iCCA cohort 4 to examine the expression and localization of the candidate. Molecular and biochemical assays were used to evaluate the membrane localization and functional protein domains of the candidate. Cell sorting was performed and the corresponding cellular molecular assays were utilized to examine cancer stem cell features of the sorted cells. RESULTS The unbiased global screening identified RRM2 as the top candidate, with a significantly higher level in iCCA patients with the MS signature activation and in iCCA cells positive for the stemness reporter. Consistently, silencing RRM2 significantly suppressed iCCA malignancy phenotypes both in vitro and in vivo. Moreover, immunohistochemistry in tumor tissues of iCCA patients revealed an unreported cell membrane localization of RRM2, in contrast to its usual cytoplasmic localization. RRM2 cell membrane localization was then confirmed in iCCA cells via immunofluorescence with or without cell membrane permeabilization, cell fractionation assay and cell surface biotinylation assay. Meanwhile, an unclassical signal peptide and a transmembrane domain of RRM2 were revealed experimentally. They were essential for RRM2 trafficking to cell membrane via the conventional endoplasmic reticulum (ER)-Golgi secretory pathway. Furthermore, the membrane RRM2-positive iCCA cells were successfully sorted. These cells possessed significant cancer stem cell malignant features including cell differentiation ability, self-renewal ability, tumor initiation ability, and stemness/malignancy gene signatures. Patients with membrane RRM2-positive iCCA cells had poor prognosis. CONCLUSIONS RRM2 had an alternative cell membrane localization. The membrane RRM2-positive iCCA cells represented a malignant subpopulation with cancer stem cell features.
Collapse
Affiliation(s)
- Yongzhi Zhao
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Department of General Surgery in Sir Run Run Shaw Hospital Affiliated to School of Medicine, Cancer Center, Center for Life Sciences in Shaoxing Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Shuting Xue
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Department of General Surgery in Sir Run Run Shaw Hospital Affiliated to School of Medicine, Cancer Center, Center for Life Sciences in Shaoxing Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Danduo Wei
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Department of General Surgery in Sir Run Run Shaw Hospital Affiliated to School of Medicine, Cancer Center, Center for Life Sciences in Shaoxing Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jianjuan Zhang
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Department of General Surgery in Sir Run Run Shaw Hospital Affiliated to School of Medicine, Cancer Center, Center for Life Sciences in Shaoxing Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Nachuan Zhang
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Department of General Surgery in Sir Run Run Shaw Hospital Affiliated to School of Medicine, Cancer Center, Center for Life Sciences in Shaoxing Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Liping Mao
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Department of General Surgery in Sir Run Run Shaw Hospital Affiliated to School of Medicine, Cancer Center, Center for Life Sciences in Shaoxing Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Niya Liu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Department of General Surgery in Sir Run Run Shaw Hospital Affiliated to School of Medicine, Cancer Center, Center for Life Sciences in Shaoxing Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Lei Zhao
- Shandong Cancer Hospital and Institute, Shandong Cancer Hospital of Shandong First Medical University, Jinan, Shandong Province, China
| | - Jianing Yan
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Department of General Surgery in Sir Run Run Shaw Hospital Affiliated to School of Medicine, Cancer Center, Center for Life Sciences in Shaoxing Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yifan Wang
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Department of General Surgery in Sir Run Run Shaw Hospital Affiliated to School of Medicine, Cancer Center, Center for Life Sciences in Shaoxing Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Xiujun Cai
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Department of General Surgery in Sir Run Run Shaw Hospital Affiliated to School of Medicine, Cancer Center, Center for Life Sciences in Shaoxing Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Saiyong Zhu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Department of General Surgery in Sir Run Run Shaw Hospital Affiliated to School of Medicine, Cancer Center, Center for Life Sciences in Shaoxing Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Stephanie Roessler
- Institute of Pathology, Heidelberg University, University Hospital Heidelberg, Heidelberg, Germany
| | - Junfang Ji
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Department of General Surgery in Sir Run Run Shaw Hospital Affiliated to School of Medicine, Cancer Center, Center for Life Sciences in Shaoxing Institute, Zhejiang University, Hangzhou, Zhejiang Province, China.
- Life Sciences Institute, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
26
|
Birch-Price Z, Hardy FJ, Lister TM, Kohn AR, Green AP. Noncanonical Amino Acids in Biocatalysis. Chem Rev 2024; 124:8740-8786. [PMID: 38959423 PMCID: PMC11273360 DOI: 10.1021/acs.chemrev.4c00120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
In recent years, powerful genetic code reprogramming methods have emerged that allow new functional components to be embedded into proteins as noncanonical amino acid (ncAA) side chains. In this review, we will illustrate how the availability of an expanded set of amino acid building blocks has opened a wealth of new opportunities in enzymology and biocatalysis research. Genetic code reprogramming has provided new insights into enzyme mechanisms by allowing introduction of new spectroscopic probes and the targeted replacement of individual atoms or functional groups. NcAAs have also been used to develop engineered biocatalysts with improved activity, selectivity, and stability, as well as enzymes with artificial regulatory elements that are responsive to external stimuli. Perhaps most ambitiously, the combination of genetic code reprogramming and laboratory evolution has given rise to new classes of enzymes that use ncAAs as key catalytic elements. With the framework for developing ncAA-containing biocatalysts now firmly established, we are optimistic that genetic code reprogramming will become a progressively more powerful tool in the armory of enzyme designers and engineers in the coming years.
Collapse
Affiliation(s)
| | | | | | | | - Anthony P. Green
- Manchester Institute of Biotechnology,
School of Chemistry, University of Manchester, Manchester M1 7DN, U.K.
| |
Collapse
|
27
|
An L, Said M, Tran L, Majumder S, Goreshnik I, Lee GR, Juergens D, Dauparas J, Anishchenko I, Coventry B, Bera AK, Kang A, Levine PM, Alvarez V, Pillai A, Norn C, Feldman D, Zorine D, Hicks DR, Li X, Sanchez MG, Vafeados DK, Salveson PJ, Vorobieva AA, Baker D. Binding and sensing diverse small molecules using shape-complementary pseudocycles. Science 2024; 385:276-282. [PMID: 39024436 PMCID: PMC11542606 DOI: 10.1126/science.adn3780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 06/03/2024] [Indexed: 07/20/2024]
Abstract
We describe an approach for designing high-affinity small molecule-binding proteins poised for downstream sensing. We use deep learning-generated pseudocycles with repeating structural units surrounding central binding pockets with widely varying shapes that depend on the geometry and number of the repeat units. We dock small molecules of interest into the most shape complementary of these pseudocycles, design the interaction surfaces for high binding affinity, and experimentally screen to identify designs with the highest affinity. We obtain binders to four diverse molecules, including the polar and flexible methotrexate and thyroxine. Taking advantage of the modular repeat structure and central binding pockets, we construct chemically induced dimerization systems and low-noise nanopore sensors by splitting designs into domains that reassemble upon ligand addition.
Collapse
Affiliation(s)
- Linna An
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Meerit Said
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Long Tran
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Chemistry, University of Washington, Seattle, WA, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Sagardip Majumder
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Inna Goreshnik
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Gyu Rie Lee
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - David Juergens
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Graduate Program in Molecular Engineering, University of Washington, Seattle, WA, USA
| | - Justas Dauparas
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Ivan Anishchenko
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Brian Coventry
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Asim K. Bera
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alex Kang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Paul M. Levine
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Valentina Alvarez
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Arvind Pillai
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | | | - Dmitri Zorine
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Derrick R. Hicks
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Xinting Li
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - Dionne K. Vafeados
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Patrick J. Salveson
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Anastassia A. Vorobieva
- VIB-VUB Center for Structural Biology, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Chemistry, University of Washington, Seattle, WA, USA
| |
Collapse
|
28
|
Milunovic MM, Ohui K, Besleaga I, Petrasheuskaya TV, Dömötör O, Enyedy ÉA, Darvasiova D, Rapta P, Barbieriková Z, Vegh D, Tóth S, Tóth J, Kucsma N, Szakács G, Popović-Bijelić A, Zafar A, Reynisson J, Shutalev AD, Bai R, Hamel E, Arion VB. Copper(II) Complexes with Isomeric Morpholine-Substituted 2-Formylpyridine Thiosemicarbazone Hybrids as Potential Anticancer Drugs Inhibiting Both Ribonucleotide Reductase and Tubulin Polymerization: The Morpholine Position Matters. J Med Chem 2024; 67:9069-9090. [PMID: 38771959 PMCID: PMC11181322 DOI: 10.1021/acs.jmedchem.4c00259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/19/2024] [Accepted: 05/09/2024] [Indexed: 05/23/2024]
Abstract
The development of copper(II) thiosemicarbazone complexes as potential anticancer agents, possessing dual functionality as inhibitors of R2 ribonucleotide reductase (RNR) and tubulin polymerization by binding at the colchicine site, presents a promising avenue for enhancing therapeutic effectiveness. Herein, we describe the syntheses and physicochemical characterization of four isomeric proligands H2L3-H2L6, with the methylmorpholine substituent at pertinent positions of the pyridine ring, along with their corresponding Cu(II) complexes 3-6. Evidently, the position of the morpholine moiety and the copper(II) complex formation have marked effects on the in vitro antiproliferative activity in human uterine sarcoma MES-SA cells and the multidrug-resistant derivative MES-SA/Dx5 cells. Activity correlated strongly with quenching of the tyrosyl radical (Y•) of mouse R2 RNR protein, inhibition of RNR activity in the cancer cells, and inhibition of tubulin polymerization. Insights into the mechanism of antiproliferative activity, supported by experimental results and molecular modeling calculations, are presented.
Collapse
Affiliation(s)
| | - Katerina Ohui
- Institute
of Inorganic Chemistry, University of Vienna, Vienna A-1090, Austria
| | - Iuliana Besleaga
- Institute
of Inorganic Chemistry, University of Vienna, Vienna A-1090, Austria
| | - Tatsiana V. Petrasheuskaya
- Department
of Molecular and Analytical Chemistry, Interdisciplinary Excellence
Centre, University of Szeged, Dóm tér 7-8, Szeged H-6720, Hungary
- MTA-SZTE
Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Orsolya Dömötör
- Department
of Molecular and Analytical Chemistry, Interdisciplinary Excellence
Centre, University of Szeged, Dóm tér 7-8, Szeged H-6720, Hungary
- MTA-SZTE
Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Éva A. Enyedy
- Department
of Molecular and Analytical Chemistry, Interdisciplinary Excellence
Centre, University of Szeged, Dóm tér 7-8, Szeged H-6720, Hungary
- MTA-SZTE
Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Denisa Darvasiova
- Institute
of Physical Chemistry and Chemical Physics, Faculty of Chemical and
Food Technology, Slovak University of Technology
in Bratislava, Bratislava SK-81237, Slovakia
| | - Peter Rapta
- Institute
of Physical Chemistry and Chemical Physics, Faculty of Chemical and
Food Technology, Slovak University of Technology
in Bratislava, Bratislava SK-81237, Slovakia
| | - Zuzana Barbieriková
- Institute
of Physical Chemistry and Chemical Physics, Faculty of Chemical and
Food Technology, Slovak University of Technology
in Bratislava, Bratislava SK-81237, Slovakia
| | - Daniel Vegh
- Institute
of Organic Chemistry, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Bratislava SK-81237, Slovakia
| | - Szilárd Tóth
- Institute
of Molecular Life Sciences, HUN-REN Research
Centre for Natural Sciences, Hungarian Research Network, Magyar Tudósok körútja
2, Budapest H-1117, Hungary
| | - Judit Tóth
- Institute
of Molecular Life Sciences, HUN-REN Research
Centre for Natural Sciences, Hungarian Research Network, Magyar Tudósok körútja
2, Budapest H-1117, Hungary
| | - Nóra Kucsma
- Institute
of Molecular Life Sciences, HUN-REN Research
Centre for Natural Sciences, Hungarian Research Network, Magyar Tudósok körútja
2, Budapest H-1117, Hungary
| | - Gergely Szakács
- Institute
of Molecular Life Sciences, HUN-REN Research
Centre for Natural Sciences, Hungarian Research Network, Magyar Tudósok körútja
2, Budapest H-1117, Hungary
- Center
for Cancer Research, Medical University
of Vienna, Vienna A-1090, Austria
| | - Ana Popović-Bijelić
- Faculty
of Physical Chemistry, University of Belgrade, Belgrade 11158, Serbia
| | - Ayesha Zafar
- School
of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jóhannes Reynisson
- School
of Pharmacy and Bioengineering, Keele University, Newcastle-under-Lyme, Staffordshire ST5 5BG, United
Kingdom
| | - Anatoly D. Shutalev
- N.
D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow 119991, Russian Federation
| | - Ruoli Bai
- Molecular
Pharmacology Branch, Developmental Therapeutics Program, Division
of Cancer Diagnosis and Treatment, National Cancer Institute, Frederick
National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Ernest Hamel
- Molecular
Pharmacology Branch, Developmental Therapeutics Program, Division
of Cancer Diagnosis and Treatment, National Cancer Institute, Frederick
National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Vladimir B. Arion
- Institute
of Inorganic Chemistry, University of Vienna, Vienna A-1090, Austria
- Inorganic
Polymers Department, “Petru Poni”
Institute of Macromolecular Chemistry, Aleea Gr. Ghica Voda 41 A, Iasi 700487, Romania
| |
Collapse
|
29
|
Haight JA, Koppenhafer SL, Geary EL, Gordon DJ. Auranofin and reactive oxygen species inhibit protein synthesis and regulate the level of the PLK1 protein in Ewing sarcoma cells. Front Oncol 2024; 14:1394653. [PMID: 38933441 PMCID: PMC11199525 DOI: 10.3389/fonc.2024.1394653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Novel therapeutic approaches are needed for the treatment of Ewing sarcoma tumors. We previously identified that Ewing sarcoma cell lines are sensitive to drugs that inhibit protein translation. However, translational and therapeutic approaches to inhibit protein synthesis in tumors are limited. In this work, we identified that reactive oxygen species, which are generated by a wide range of chemotherapy and other drugs, inhibit protein synthesis and reduce the level of critical proteins that support tumorigenesis in Ewing sarcoma cells. In particular, we identified that both hydrogen peroxide and auranofin, an inhibitor of thioredoxin reductase and regulator of oxidative stress and reactive oxygen species, activate the repressor of protein translation 4E-BP1 and reduce the levels of the oncogenic proteins RRM2 and PLK1 in Ewing and other sarcoma cell lines. These results provide novel insight into the mechanism of how ROS-inducing drugs target cancer cells via inhibition of protein translation and identify a mechanistic link between ROS and the DNA replication (RRM2) and cell cycle regulatory (PLK1) pathways.
Collapse
Affiliation(s)
- Joseph A. Haight
- Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Stacia L. Koppenhafer
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, United States
| | - Elizabeth L. Geary
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, United States
| | - David J. Gordon
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
30
|
Tong Z, Zhao Y, Bai S, Ebner B, Lienhard L, Zhao Y, Wang Z, Pan Q, Guo P, Bracht T, Sitek B, Gschwend JE, Xu W, Nawroth R. The mechanism of resistance to CDK4/6 inhibition and novel combination therapy with RNR inhibition for chemo-resistant bladder cancer. Cancer Commun (Lond) 2024; 44:700-704. [PMID: 38468431 PMCID: PMC11194448 DOI: 10.1002/cac2.12532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/13/2024] Open
Affiliation(s)
- Zhichao Tong
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangP. R. China
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted TheranosticsHarbin Medical UniversityHarbinHeilongjiangP. R. China
- Department of UrologyKlinikum rechts der Isar, Technical university of MunichMunichGermany
- Department of UrologyThe Fourth Hospital of Harbin Medical UniversityHarbinHeilongjiangP. R. China
| | - Yubo Zhao
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangP. R. China
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted TheranosticsHarbin Medical UniversityHarbinHeilongjiangP. R. China
| | - Shiyu Bai
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted TheranosticsHarbin Medical UniversityHarbinHeilongjiangP. R. China
- Department of UrologyThe Fourth Hospital of Harbin Medical UniversityHarbinHeilongjiangP. R. China
| | - Benedikt Ebner
- Department of UrologyKlinikum rechts der Isar, Technical university of MunichMunichGermany
- Department of UrologyLudwig‐Maximilians‐University of MunichMunichGermany
| | - Lou Lienhard
- Department of UrologyKlinikum rechts der Isar, Technical university of MunichMunichGermany
| | - Yuling Zhao
- Department of UrologyKlinikum rechts der Isar, Technical university of MunichMunichGermany
| | - Ziqi Wang
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangP. R. China
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted TheranosticsHarbin Medical UniversityHarbinHeilongjiangP. R. China
| | - Qi Pan
- Department of UrologyKlinikum rechts der Isar, Technical university of MunichMunichGermany
- Department of UrologyShanghai General Hospital, Shanghai Jiao Tong UniversityShanghaiP. R. China
| | - Pengyu Guo
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangP. R. China
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted TheranosticsHarbin Medical UniversityHarbinHeilongjiangP. R. China
| | - Thilo Bracht
- Medizinisches Proteom‐Center, Ruhr‐Universität BochumBochumGermany
- Clinic for Anesthesiology, Intensive Care and Pain Therapy, University Medical Center Knappschaftskrankenhaus BochumBochumGermany
| | - Barbara Sitek
- Medizinisches Proteom‐Center, Ruhr‐Universität BochumBochumGermany
- Clinic for Anesthesiology, Intensive Care and Pain Therapy, University Medical Center Knappschaftskrankenhaus BochumBochumGermany
| | - Jürgen E. Gschwend
- Department of UrologyKlinikum rechts der Isar, Technical university of MunichMunichGermany
| | - Wanhai Xu
- Department of UrologyHarbin Medical University Cancer HospitalHarbinHeilongjiangP. R. China
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted TheranosticsHarbin Medical UniversityHarbinHeilongjiangP. R. China
| | - Roman Nawroth
- Department of UrologyKlinikum rechts der Isar, Technical university of MunichMunichGermany
| |
Collapse
|
31
|
Zhang X, Joseph S, Wu D, Bowser JL, Vaziri C. The DNA Damage Response (DDR) landscape of endometrial cancer defines discrete disease subtypes and reveals therapeutic opportunities. NAR Cancer 2024; 6:zcae015. [PMID: 38596432 PMCID: PMC11000323 DOI: 10.1093/narcan/zcae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/12/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024] Open
Abstract
Genome maintenance is an enabling characteristic that allows neoplastic cells to tolerate the inherent stresses of tumorigenesis and evade therapy-induced genotoxicity. Neoplastic cells also deploy many mis-expressed germ cell proteins termed Cancer Testes Antigens (CTAs) to promote genome maintenance and survival. Here, we present the first comprehensive characterization of the DNA Damage Response (DDR) and CTA transcriptional landscapes of endometrial cancer in relation to conventional histological and molecular subtypes. We show endometrial serous carcinoma (ESC), an aggressive endometrial cancer subtype, is defined by gene expression signatures comprising members of the Replication Fork Protection Complex (RFPC) and Fanconi Anemia (FA) pathway and CTAs with mitotic functions. DDR and CTA-based profiling also defines a subset of highly aggressive endometrioid endometrial carcinomas (EEC) with poor clinical outcomes that share similar profiles to ESC yet have distinct characteristics based on conventional histological and genomic features. Using an unbiased CRISPR-based genetic screen and a candidate gene approach, we confirm that DDR and CTA genes that constitute the ESC and related EEC gene signatures are required for proliferation and therapy-resistance of cultured endometrial cancer cells. Our study validates the use of DDR and CTA-based tumor classifiers and reveals new vulnerabilities of aggressive endometrial cancer where none currently exist.
Collapse
Affiliation(s)
- Xingyuan Zhang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC - 27599, USA
| | - Sayali Joseph
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC - 27599, USA
| | - Di Wu
- Department of Biostatistics, University of North Carolina at Chapel Hill, School of Dentistry, Chapel Hill, NC - 27599, USA
| | - Jessica L Bowser
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC - 27599, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC - 27599, USA
| | - Cyrus Vaziri
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC - 27599, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC - 27599, USA
| |
Collapse
|
32
|
Wei X, Zhou Y, Shen X, Fan L, Liu D, Gao X, Zhou J, Wu Y, Li Y, Feng W, Zhang Z. Ciclopirox inhibits SARS-CoV-2 replication by promoting the degradation of the nucleocapsid protein. Acta Pharm Sin B 2024; 14:2505-2519. [PMID: 38828154 PMCID: PMC11143514 DOI: 10.1016/j.apsb.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/04/2024] [Accepted: 02/28/2024] [Indexed: 06/05/2024] Open
Abstract
The nucleocapsid protein (NP) plays a crucial role in SARS-CoV-2 replication and is the most abundant structural protein with a long half-life. Despite its vital role in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) assembly and host inflammatory response, it remains an unexplored target for drug development. In this study, we identified a small-molecule compound (ciclopirox) that promotes NP degradation using an FDA-approved library and a drug-screening cell model. Ciclopirox significantly inhibited SARS-CoV-2 replication both in vitro and in vivo by inducing NP degradation. Ciclopirox induced abnormal NP aggregation through indirect interaction, leading to the formation of condensates with higher viscosity and lower mobility. These condensates were subsequently degraded via the autophagy-lysosomal pathway, ultimately resulting in a shortened NP half-life and reduced NP expression. Our results suggest that NP is a potential drug target, and that ciclopirox holds substantial promise for further development to combat SARS-CoV-2 replication.
Collapse
Affiliation(s)
- Xiafei Wei
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen 518112, China
| | - Yuzheng Zhou
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen 518112, China
| | - Xiaotong Shen
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen 518112, China
| | - Lujie Fan
- Guangzhou Laboratory, Guangzhou Medical University, Guangzhou 511495, China
| | - Donglan Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Xiang Gao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen 518112, China
| | - Jian Zhou
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen 518112, China
| | - Yezi Wu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen 518112, China
| | - Yunfei Li
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen 518112, China
| | - Wei Feng
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen 518112, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen 518112, China
| |
Collapse
|
33
|
Sharma V, Fedoseyenko D, Joshi S, Abdelwahed S, Begley TP. Phosphomethylpyrimidine Synthase (ThiC): Trapping of Five Intermediates Provides Mechanistic Insights on a Complex Radical Cascade Reaction in Thiamin Biosynthesis. ACS CENTRAL SCIENCE 2024; 10:988-1000. [PMID: 38799670 PMCID: PMC11117688 DOI: 10.1021/acscentsci.4c00125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 05/29/2024]
Abstract
Phosphomethylpyrimidine synthase (ThiC) catalyzes the conversion of AIR to the thiamin pyrimidine HMP-P. This reaction is the most complex enzyme-catalyzed radical cascade identified to date, and the detailed mechanism has remained elusive. In this paper, we describe the trapping of five new intermediates that provide snapshots of the ThiC reaction coordinate and enable the formulation of a revised mechanism for the ThiC-catalyzed reaction.
Collapse
Affiliation(s)
- Vishav Sharma
- Department of Chemistry, Texas A&M University, College Station, Texas 77842, United States
| | - Dmytro Fedoseyenko
- Department of Chemistry, Texas A&M University, College Station, Texas 77842, United States
| | - Sumedh Joshi
- Department of Chemistry, Texas A&M University, College Station, Texas 77842, United States
| | - Sameh Abdelwahed
- Department of Chemistry, Texas A&M University, College Station, Texas 77842, United States
| | - Tadhg P. Begley
- Department of Chemistry, Texas A&M University, College Station, Texas 77842, United States
| |
Collapse
|
34
|
Haight JA, Koppenhafer SL, Geary EL, Gordon DJ. Auranofin and reactive oxygen species inhibit protein synthesis and regulate the level of the PLK1 protein in Ewing sarcoma cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593567. [PMID: 38798568 PMCID: PMC11118274 DOI: 10.1101/2024.05.13.593567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Novel therapeutic approaches are needed for the treatment of Ewing sarcoma tumors. We previously identified that Ewing sarcoma cell lines are sensitive to drugs that inhibit protein translation. However, translational and therapeutic approaches to inhibit protein synthesis in tumors are limited. In this work, we identified that reactive oxygen species, which are generated by a wide range of chemotherapy and other drugs, inhibit protein synthesis and reduce the level of critical proteins that support tumorigenesis in Ewing sarcoma cells. In particular, we identified that both hydrogen peroxide and auranofin, an inhibitor of thioredoxin reductase and regulator of oxidative stress and reactive oxygen species, activate the repressor of protein translation 4E-BP1 and reduce the levels of the oncogenic proteins RRM2 and PLK1 in Ewing and other sarcoma cell lines. These results provide novel insight into the mechanism of how ROS-inducing drugs target cancer cells via inhibition of protein translation and identify a mechanistic link between ROS and the DNA replication (RRM2) and cell cycle regulatory (PLK1) pathways.
Collapse
|
35
|
Song DY, Stubbe J, Nocera DG. Protein engineering a PhotoRNR chimera based on a unifying evolutionary apparatus among the natural classes of ribonucleotide reductases. Proc Natl Acad Sci U S A 2024; 121:e2317291121. [PMID: 38648489 PMCID: PMC11067019 DOI: 10.1073/pnas.2317291121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 03/19/2024] [Indexed: 04/25/2024] Open
Abstract
Ribonucleotide reductases (RNRs) are essential enzymes that catalyze the de novo transformation of nucleoside 5'-di(tri)phosphates [ND(T)Ps, where N is A, U, C, or G] to their corresponding deoxynucleotides. Despite the diversity of factors required for function and the low sequence conservation across RNRs, a unifying apparatus consolidating RNR activity is explored. We combine aspects of the protein subunit simplicity of class II RNR with a modified version of Escherichia coli class la photoRNRs that initiate radical chemistry with light to engineer a mimic of a class II enzyme. The design of this RNR involves fusing a truncated form of the active site containing α subunit with the functionally important C-terminal tail of the radical-generating β subunit to render a chimeric RNR. Inspired by a recent cryo-EM structure, a [Re] photooxidant is located adjacent to Y356[β], which is an essential component of the radical transport pathway in class I RNRs. Combination of this RNR photochimera with cytidine diphosphate (CDP), adenosine triphosphate (ATP), and light resulted in the generation of Y356• along with production of deoxycytidine diphosphate (dCDP) and cytosine. The photoproducts reflect an active site chemistry consistent with both the consensus mechanism of RNR and chemistry observed when RNR is inactivated by mechanism-based inhibitors in the active site. The enzymatic activity of the RNR photochimera in the absence of any β metallocofactor highlights the adaptability of the 10-stranded αβ barrel finger loop to support deoxynucleotide formation and accommodate the design of engineered RNRs.
Collapse
Affiliation(s)
- David Y. Song
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA02138
| | - JoAnne Stubbe
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA02138
| | - Daniel G. Nocera
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA02138
| |
Collapse
|
36
|
Hou Y, Wang S, Zhang Y, Huang X, Zhang X, He F, Tian C, Sun A. Proteomics Identifies LUC7L3 as a Prognostic Biomarker for Hepatocellular Carcinoma. Curr Issues Mol Biol 2024; 46:4004-4020. [PMID: 38785515 PMCID: PMC11120364 DOI: 10.3390/cimb46050247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Alternative splicing has been shown to participate in tumor progression, including hepatocellular carcinoma. The poor prognosis of patients with HCC calls for molecular classification and biomarker identification to facilitate precision medicine. We performed ssGSEA analysis to quantify the pathway activity of RNA splicing in three HCC cohorts. Kaplan-Meier and Cox methods were used for survival analysis. GO and GSEA were performed to analyze pathway enrichment. We confirmed that RNA splicing is significantly correlated with prognosis, and identified an alternative splicing-associated protein LUC7L3 as a potential HCC prognostic biomarker. Further bioinformatics analysis revealed that high LUC7L3 expression indicated a more progressive HCC subtype and worse clinical features. Cell proliferation-related pathways were enriched in HCC patients with high LUC7L3 expression. Consistently, we proved that LUC7L3 knockdown could significantly inhibit cell proliferation and suppress the activation of associated signaling pathways in vitro. In this research, the relevance between RNA splicing and HCC patient prognosis was outlined. Our newly identified biomarker LUC7L3 could provide stratification for patient survival and recurrence risk, facilitating early medical intervention before recurrence or disease progression.
Collapse
Affiliation(s)
- Yushan Hou
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Siqi Wang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yiming Zhang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiaofen Huang
- College of Life Sciences, Hebei University, Baoding 071002, China
| | - Xiuyuan Zhang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Fuchu He
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
- Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing 102206, China
| | - Chunyan Tian
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
- College of Life Sciences, Hebei University, Baoding 071002, China
| | - Aihua Sun
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
- College of Life Sciences, Hebei University, Baoding 071002, China
- Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing 102206, China
| |
Collapse
|
37
|
Fontecilla-Camps JC. Reflections on the Origin of Coded Protein Biosynthesis. Biomolecules 2024; 14:518. [PMID: 38785925 PMCID: PMC11117964 DOI: 10.3390/biom14050518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
The principle of continuity posits that some central features of primordial biocatalytic mechanisms should still be present in the genetically dependent pathway of protein synthesis, a crucial step in the emergence of life. Key bimolecular reactions of this process are catalyzed by DNA-dependent RNA polymerases, aminoacyl-tRNA synthetases, and ribosomes. Remarkably, none of these biocatalysts contribute chemically active groups to their respective reactions. Instead, structural and functional studies have demonstrated that nucleotidic α-phosphate and β-d-ribosyl 2' OH and 3' OH groups can help their own catalysis, a process which, consequently, has been called "substrate-assisted". Furthermore, upon binding, the substrates significantly lower the entropy of activation, exclude water from these catalysts' active sites, and are readily positioned for a reaction. This binding mode has been described as an "entropy trap". The combination of this effect with substrate-assisted catalysis results in reactions that are stereochemically and mechanistically simpler than the ones found in most modern enzymes. This observation is consistent with the way in which primordial catalysts could have operated; it may also explain why, thanks to their complementary reactivities, β-d-ribose and phosphate were naturally selected to be the central components of early coding polymers.
Collapse
|
38
|
Kulagin KA, Starodubova ES, Osipova PJ, Lipatova AV, Cherdantsev IA, Poddubko SV, Karpov VL, Karpov DS. Synergistic Effect of a Combination of Proteasome and Ribonucleotide Reductase Inhibitors in a Biochemical Model of the Yeast Saccharomyces cerevisiae and a Glioblastoma Cell Line. Int J Mol Sci 2024; 25:3977. [PMID: 38612788 PMCID: PMC11011839 DOI: 10.3390/ijms25073977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Proteasome inhibitors are used in the therapy of several cancers, and clinical trials are underway for their use in the treatment of glioblastoma (GBM). However, GBM becomes resistant to chemotherapy relatively rapidly. Recently, the overexpression of ribonucleotide reductase (RNR) genes was found to mediate therapy resistance in GBM. The use of combinations of chemotherapeutic agents is considered a promising direction in cancer therapy. The present work aimed to evaluate the efficacy of the combination of proteasome and RNR inhibitors in yeast and GBM cell models. We have shown that impaired proteasome function results in increased levels of RNR subunits and increased enzyme activity in yeast. Co-administration of the proteasome inhibitor bortezomib and the RNR inhibitor hydroxyurea was found to significantly reduce the growth rate of S. cerevisiae yeast. Accordingly, the combination of bortezomib and another RNR inhibitor gemcitabine reduced the survival of DBTRG-05MG compared to the HEK293 cell line. Thus, yeast can be used as a simple model to evaluate the efficacy of combinations of proteasome and RNR inhibitors.
Collapse
Affiliation(s)
- Kirill A. Kulagin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (K.A.K.); (E.S.S.); (P.J.O.); (A.V.L.); (I.A.C.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Elizaveta S. Starodubova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (K.A.K.); (E.S.S.); (P.J.O.); (A.V.L.); (I.A.C.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Pamila J. Osipova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (K.A.K.); (E.S.S.); (P.J.O.); (A.V.L.); (I.A.C.)
- Institute of Biomedical Problems of Russian Academy of Sciences, 123007 Moscow, Russia;
| | - Anastasia V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (K.A.K.); (E.S.S.); (P.J.O.); (A.V.L.); (I.A.C.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Igor A. Cherdantsev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (K.A.K.); (E.S.S.); (P.J.O.); (A.V.L.); (I.A.C.)
| | - Svetlana V. Poddubko
- Institute of Biomedical Problems of Russian Academy of Sciences, 123007 Moscow, Russia;
| | - Vadim L. Karpov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Dmitry S. Karpov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (K.A.K.); (E.S.S.); (P.J.O.); (A.V.L.); (I.A.C.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| |
Collapse
|
39
|
Kass D, Larson VA, Corona T, Kuhlmann U, Hildebrandt P, Lohmiller T, Bill E, Lehnert N, Ray K. Trapping of a phenoxyl radical at a non-haem high-spin iron(II) centre. Nat Chem 2024; 16:658-665. [PMID: 38216752 DOI: 10.1038/s41557-023-01405-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 11/17/2023] [Indexed: 01/14/2024]
Abstract
The activation of dioxygen at haem and non-haem metal centres, and subsequent functionalization of unactivated C‒H bonds, has been a focal point of much research. In iron-mediated oxidation reactions, O2 binding at an iron(II) centre is often accompanied by an oxidation of the iron centre. Here we demonstrate dioxygen activation by sodium tetraphenylborate and protons in the presence of an iron(II) complex to form a reactive radical species, whereby the iron oxidation state remains unaltered in the presence of a highly oxidizing phenoxyl radical and O2. This complex, containing an unusual iron(II)-phenoxyl radical motif, represents an elusive example of a spectroscopically characterized oxygen-derived iron(II)-reactive intermediate during chemical and biological dioxygen activation at haem and non-haem iron active centres. The present report opens up strategies for the stabilization of a phenoxyl radical cofactor, with its full oxidizing capabilities, to act as an independent redox centre next to an iron(II) site during substrate oxidation reactions.
Collapse
Affiliation(s)
- Dustin Kass
- Department of Chemistry, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Virginia A Larson
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, MI, USA
| | - Teresa Corona
- Department of Chemistry, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Uwe Kuhlmann
- Department of Chemistry, Technische Universität Berlin, Berlin, Germany
| | - Peter Hildebrandt
- Department of Chemistry, Technische Universität Berlin, Berlin, Germany
| | - Thomas Lohmiller
- Department of Chemistry, Humboldt-Universität zu Berlin, Berlin, Germany
- EPR4Energy Joint Lab, Department Spins in Energy Conversion and Quantum Information Science, Helmholtz Zentrum Berlin für Materialien und Energie GmbH, Berlin, Germany
| | - Eckhard Bill
- Max-Planck-Institut für Chemische Energiekonversion, Mülheim an der Ruhr, Germany
| | - Nicolai Lehnert
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, MI, USA.
| | - Kallol Ray
- Department of Chemistry, Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
40
|
Kienbeck K, Malfertheiner L, Zelger-Paulus S, Johannsen S, von Mering C, Sigel RKO. Identification of HDV-like theta ribozymes involved in tRNA-based recoding of gut bacteriophages. Nat Commun 2024; 15:1559. [PMID: 38378708 PMCID: PMC10879173 DOI: 10.1038/s41467-024-45653-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/29/2024] [Indexed: 02/22/2024] Open
Abstract
Trillions of microorganisms, collectively known as the microbiome, inhabit our bodies with the gut microbiome being of particular interest in biomedical research. Bacteriophages, the dominant virome constituents, can utilize suppressor tRNAs to switch to alternative genetic codes (e.g., the UAG stop-codon is reassigned to glutamine) while infecting hosts with the standard bacterial code. However, what triggers this switch and how the bacteriophage manipulates its host is poorly understood. Here, we report the discovery of a subgroup of minimal hepatitis delta virus (HDV)-like ribozymes - theta ribozymes - potentially involved in the code switch leading to the expression of recoded lysis and structural phage genes. We demonstrate their HDV-like self-scission behavior in vitro and find them in an unreported context often located with their cleavage site adjacent to tRNAs, indicating a role in viral tRNA maturation and/or regulation. Every fifth associated tRNA is a suppressor tRNA, further strengthening our hypothesis. The vast abundance of tRNA-associated theta ribozymes - we provide 1753 unique examples - highlights the importance of small ribozymes as an alternative to large enzymes that usually process tRNA 3'-ends. Our discovery expands the short list of biological functions of small HDV-like ribozymes and introduces a previously unknown player likely involved in the code switch of certain recoded gut bacteriophages.
Collapse
Affiliation(s)
- Kasimir Kienbeck
- Department of Chemistry, University of Zurich, Zurich, CH-8057, Switzerland
| | - Lukas Malfertheiner
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, CH-8057, Switzerland
| | | | - Silke Johannsen
- Department of Chemistry, University of Zurich, Zurich, CH-8057, Switzerland
| | - Christian von Mering
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, CH-8057, Switzerland.
| | - Roland K O Sigel
- Department of Chemistry, University of Zurich, Zurich, CH-8057, Switzerland.
| |
Collapse
|
41
|
Zhong J, Soudackov AV, Hammes-Schiffer S. Probing Nonadiabaticity of Proton-Coupled Electron Transfer in Ribonucleotide Reductase. J Phys Chem Lett 2024; 15:1686-1693. [PMID: 38315651 PMCID: PMC11586673 DOI: 10.1021/acs.jpclett.3c03552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
The enzyme ribonucleotide reductase, which is essential for DNA synthesis, initiates the conversion of ribonucleotides to deoxyribonucleotides via radical transfer over a 32 Å pathway composed of proton-coupled electron transfer (PCET) reactions. Previously, the first three PCET reactions in the α subunit were investigated with hybrid quantum mechanical/molecular mechanical (QM/MM) free energy simulations. Herein, the fourth PCET reaction in this subunit between C439 and guanosine diphosphate (GDP) is simulated and found to be slightly exoergic with a relatively high free energy barrier. To further elucidate the mechanisms of all four PCET reactions, we analyzed the vibronic and electron-proton nonadiabaticities. This analysis suggests that interfacial PCET between Y356 and Y731 is vibronically and electronically nonadiabatic, whereas PCET between Y731 and Y730 and between C439 and GDP is fully adiabatic and PCET between Y730 and C439 is in the intermediate regime. These insights provide guidance for selecting suitable rate constant expressions for these PCET reactions.
Collapse
Affiliation(s)
- Jiayun Zhong
- Department of Chemistry, Yale University, New Haven, CT 06520
| | | | - Sharon Hammes-Schiffer
- Department of Chemistry, Yale University, New Haven, CT 06520
- Department of Chemistry, Princeton University, Princeton, NJ 08544
| |
Collapse
|
42
|
Wu WY, Zheng WY, Chen WT, Tsai FT, Tsai ML, Pao CW, Chen JL, Liaw WF. Electronic Structure and Transformation of Dinitrosyl Iron Complexes (DNICs) Regulated by Redox Non-Innocent Imino-Substituted Phenoxide Ligand. Inorg Chem 2024; 63:2431-2442. [PMID: 38258796 PMCID: PMC10848267 DOI: 10.1021/acs.inorgchem.3c03367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024]
Abstract
The coupled NO-vibrational peaks [IR νNO 1775 s, 1716 vs, 1668 vs cm-1 (THF)] between two adjacent [Fe(NO)2] groups implicate the electron delocalization nature of the singly O-phenoxide-bridged dinuclear dinitrosyliron complex (DNIC) [Fe(NO)2(μ-ON2Me)Fe(NO)2] (1). Electronic interplay between [Fe(NO)2] units and [ON2Me]- ligand in DNIC 1 rationalizes that "hard" O-phenoxide moiety polarizes iron center(s) of [Fe(NO)2] unit(s) to enforce a "constrained" π-conjugation system acting as an electron reservoir to bestow the spin-frustrated {Fe(NO)2}9-{Fe(NO)2}9-[·ON2Me]2- electron configuration (Stotal = 1/2). This system plays a crucial role in facilitating the ligand-based redox interconversion, working in harmony to control the storage and redox-triggered transport of the [Fe(NO)2]10 unit, while preserving the {Fe(NO)2}9 core in DNICs {Fe(NO)2}9-[·ON2Me]2- [K-18-crown-6-ether)][(ON2Me)Fe(NO)2] (2) and {Fe(NO)2}9-[·ON2Me] [(ON2Me)Fe(NO)2][PF6] (3). Electrochemical studies suggest that the redox interconversion among [{Fe(NO)2}9-[·ON2Me]2-] DNIC 3 ↔ [{Fe(NO)2}9-[ON2Me]-] ↔ [{Fe(NO)2}9-[·ON2Me]] DNIC 2 are kinetically feasible, corroborated by the redox shuttle between O-bridged dimerized [(μ-ONMe)2Fe2(NO)4] (4) and [K-18-crown-6-ether)][(ONMe)Fe(NO)2] (5). In parallel with this finding, the electronic structures of [{Fe(NO)2}9-{Fe(NO)2}9-[·ON2Me]2-] DNIC 1, [{Fe(NO)2}9-[·ON2Me]2-] DNIC 2, [{Fe(NO)2}9-[·ON2Me]] DNIC 3, [{Fe(NO)2}9-[ONMe]-]2 DNIC 4, and [{Fe(NO)2}9-[·ONMe]2-] DNIC 5 are evidenced by EPR, SQUID, and Fe K-edge pre-edge analyses, respectively.
Collapse
Affiliation(s)
- Wun-Yan Wu
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Wei-Yuan Zheng
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Wei-Ting Chen
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Fu-Te Tsai
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ming-Li Tsai
- Department of Chemistry, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Chih-Wen Pao
- National Synchrotron Radiation
Research Center, Hsinchu 30013, Taiwan
| | - Jeng-Lung Chen
- National Synchrotron Radiation
Research Center, Hsinchu 30013, Taiwan
| | - Wen-Feng Liaw
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
43
|
Kagan VE, Straub AC, Tyurina YY, Kapralov AA, Hall R, Wenzel SE, Mallampalli RK, Bayir H. Vitamin E/Coenzyme Q-Dependent "Free Radical Reductases": Redox Regulators in Ferroptosis. Antioxid Redox Signal 2024; 40:317-328. [PMID: 37154783 PMCID: PMC10890965 DOI: 10.1089/ars.2022.0154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 03/10/2023] [Accepted: 04/08/2023] [Indexed: 05/10/2023]
Abstract
Significance: Lipid peroxidation and its products, oxygenated polyunsaturated lipids, act as essential signals coordinating metabolism and physiology and can be deleterious to membranes when they accumulate in excessive amounts. Recent Advances: There is an emerging understanding that regulation of polyunsaturated fatty acid (PUFA) phospholipid peroxidation, particularly of PUFA-phosphatidylethanolamine, is important in a newly discovered type of regulated cell death, ferroptosis. Among the most recently described regulatory mechanisms is the ferroptosis suppressor protein, which controls the peroxidation process due to its ability to reduce coenzyme Q (CoQ). Critical Issues: In this study, we reviewed the most recent data in the context of the concept of free radical reductases formulated in the 1980-1990s and focused on enzymatic mechanisms of CoQ reduction in different membranes (e.g., mitochondrial, endoplasmic reticulum, and plasma membrane electron transporters) as well as TCA cycle components and cytosolic reductases capable of recycling the high antioxidant efficiency of the CoQ/vitamin E system. Future Directions: We highlight the importance of individual components of the free radical reductase network in regulating the ferroptotic program and defining the sensitivity/tolerance of cells to ferroptotic death. Complete deciphering of the interactive complexity of this system may be important for designing effective antiferroptotic modalities. Antioxid. Redox Signal. 40, 317-328.
Collapse
Affiliation(s)
- Valerian E. Kagan
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Environmental Health and Pharmacology and Chemical Biology and University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Radiation Oncology and Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adam C. Straub
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yulia Y. Tyurina
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Environmental Health and Pharmacology and Chemical Biology and University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alexandr A. Kapralov
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Environmental Health and Pharmacology and Chemical Biology and University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert Hall
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sally E. Wenzel
- Department of Environmental Health and Pharmacology and Chemical Biology and University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rama K. Mallampalli
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Hülya Bayir
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, Children's Hospital Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, Columbia University, New York, New York, USA
| |
Collapse
|
44
|
Pan N, Xu H, Chen W, Liu Z, Liu Y, Huang T, Du S, Xu S, Zheng T, Zuo Z. Cyanobacterial VOCs β-ionone and β-cyclocitral poisoning Lemna turionifera by triggering programmed cell death. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 342:123059. [PMID: 38042469 DOI: 10.1016/j.envpol.2023.123059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/07/2023] [Accepted: 11/26/2023] [Indexed: 12/04/2023]
Abstract
β-Ionone and β-cyclocitral are two typical components in cyanobacterial volatiles, which can poison aquatic plants and even cause death. To reveal the toxic mechanisms of the two compounds on aquatic plants through programmed cell death (PCD), the photosynthetic capacities, caspase-3-like activity, DNA fragmentation and ladders, as well as expression of the genes associated with PCD in Lemna turionifera were investigated in exposure to β-ionone (0.2 mM) and β-cyclocitral (0.4 mM) at lethal concentration. With prolonging the treatment time, L. turionifera fronds gradually died, and photosynthetic capacities gradually reduced and even disappeared at the 96th h. This demonstrated that the death process might be a PCD rather than a necrosis, due to the gradual loss of physiological activities. When L. turionifera underwent the death, caspase-3-like was activated after 3 h, and reached to the strongest activity at the 24th h. TUNEL-positive nuclei were detected after 12 h, and appeared in large numbers at the 48th h. The DNA was cleaved by Ca2+-dependent endonucleases and showed obviously ladders. In addition, the expression of 5 genes (TSPO, ERN1, CTSB, CYC, and ATR) positively related with PCD initiation was up-regulated, while the expression of 2 genes (RRM2 and TUBA) negatively related with PCD initiation was down-regulated. Therefore, β-ionone and β-cyclocitral can poison L. turionifera by adjusting related gene expression to trigger PCD.
Collapse
Affiliation(s)
- Ning Pan
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou, 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou, 311300, China
| | - Haozhe Xu
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou, 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou, 311300, China
| | - Wangbo Chen
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou, 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou, 311300, China
| | - Zijian Liu
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou, 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou, 311300, China
| | - Yichi Liu
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou, 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou, 311300, China
| | - Tianyu Huang
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou, 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou, 311300, China
| | - Siyi Du
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou, 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou, 311300, China
| | - Sun Xu
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou, 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou, 311300, China
| | - Tiefeng Zheng
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou, 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou, 311300, China
| | - Zhaojiang Zuo
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou, 311300, China; Zhejiang Provincial Key Laboratory of Forest Aromatic Plants-based Healthcare Functions, Zhejiang A&F University, Hangzhou, 311300, China.
| |
Collapse
|
45
|
Jafari M, Li Z, Song LF, Sagresti L, Brancato G, Merz KM. Thermodynamics of Metal-Acetate Interactions. J Phys Chem B 2024; 128:684-697. [PMID: 38226860 DOI: 10.1021/acs.jpcb.3c06567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Metal ions play crucial roles in protein- and ligand-mediated interactions. They not only act as catalysts to facilitate biological processes but are also important as protein structural elements. Accurately predicting metal ion interactions in computational studies has always been a challenge, and various methods have been suggested to improve these interactions. One such method is the 12-6-4 Lennard-Jones (LJ)-type nonbonded model. Using this model, it has been possible to successfully reproduce the experimental properties of metal ions in aqueous solution. The model includes induced dipole interactions typically ignored in the standard 12-6 LJ nonbonded model. In this we expand the applicability of this model to metal ion-carboxylate interactions. Using 12-6-4 parameters that reproduce the solvation free energies of the metal ions leads to an overestimation of metal ion-acetate interactions, thus, prompting us to fine-tune the model to specifically handle the latter. We also show that the standard 12-6 LJ model significantly falls short in reproducing the experimental binding free energy between acetate and 11 metal ions (Ni(II), Mg(II), Cu(II), Zn(II), Co(II), Cu(I), Fe(II), Mn(II), Cd(II), Ca(II), and Ag(I)). In this study, we describe optimized C4 parameters for the 12-6-4 LJ nonbonded model to be used with three widely employed water models (Transferable Intermolecular Potential with 3 Points (TIP3P), Simple Point Charge Extended (SPC/E), and Optimal Point Charge (OPC) water models). These parameters can accurately match the experimental binding free energy between 11 metal ions and acetate. These parameters can be applied to the study of metalloproteins and transition metal ion channels and transporters, as acetate serves as a representative of the negatively charged amino acid side chains from aspartate and glutamate.
Collapse
Affiliation(s)
- Majid Jafari
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Zhen Li
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Lin Frank Song
- Biochemical and Biophysical Systems Group, Lawrence Livermore National Laboratory, Livermore, California 94550, United States
| | - Luca Sagresti
- Scuola Normale Superiore and CSGI, Piazza dei Cavalieri 7, I-56126 Pisa, Italy
- Istituto Nazionale di Fisica Nucleare (INFN) sezione di Pisa, Largo Bruno Pontecorvo 3, 56127 Pisa, Italy
| | - Giuseppe Brancato
- Scuola Normale Superiore and CSGI, Piazza dei Cavalieri 7, I-56126 Pisa, Italy
- Istituto Nazionale di Fisica Nucleare (INFN) sezione di Pisa, Largo Bruno Pontecorvo 3, 56127 Pisa, Italy
| | - Kenneth M Merz
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
46
|
Claudio-Ares O, Luciano-Rodríguez J, Del Valle-González YL, Schiavone-Chamorro SL, Pastor AJ, Rivera-Reyes JO, Metzler CL, Domínguez-Orona LM, Vargas-Pérez BL, Skouta R, Tinoco AD. Exploring the Use of Intracellular Chelation and Non-Iron Metals to Program Ferroptosis for Anticancer Application. INORGANICS 2024; 12:26. [PMID: 39380574 PMCID: PMC11460773 DOI: 10.3390/inorganics12010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024] Open
Abstract
The discovery of regulated cell death (RCD) revolutionized chemotherapy. With caspase-dependent apoptosis initially being thought to be the only form of RCD, many drug development strategies aimed to synthesize compounds that turn on this kind of cell death. While yielding a variety of drugs, this approach is limited, given the acquired resistance of cancers to these drugs and the lack of specificity of the drugs for targeting cancer cells alone. The discovery of non-apoptotic forms of RCD is leading to new avenues for drug design. Evidence shows that ferroptosis, a relatively recently discovered iron-based cell death pathway, has therapeutic potential for anticancer application. Recent studies point to the interrelationship between iron and other essential metals, copper and zinc, and the disturbance of their respective homeostasis as critical to the onset of ferroptosis. Other studies reveal that several coordination complexes of non-iron metals have the capacity to induce ferroptosis. This collective knowledge will be assessed to determine how chelation approaches and coordination chemistry can be engineered to program ferroptosis in chemotherapy.
Collapse
Affiliation(s)
- Oscar Claudio-Ares
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00925, USA
| | | | | | | | - Alex J. Pastor
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00925, USA
| | - Javier O. Rivera-Reyes
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00925, USA
| | - Carmen L. Metzler
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00925, USA
| | | | - Brenda Lee Vargas-Pérez
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00925, USA
| | - Rachid Skouta
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003-9248, USA
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003-9248, USA
| | - Arthur D. Tinoco
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00925, USA
| |
Collapse
|
47
|
An L, Said M, Tran L, Majumder S, Goreshnik I, Lee GR, Juergens D, Dauparas J, Anishchenko I, Coventry B, Bera AK, Kang A, Levine PM, Alvarez V, Pillai A, Norn C, Feldman D, Zorine D, Hicks DR, Li X, Sanchez MG, Vafeados DK, Salveson PJ, Vorobieva AA, Baker D. De novo design of diverse small molecule binders and sensors using Shape Complementary Pseudocycles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572602. [PMID: 38187589 PMCID: PMC10769206 DOI: 10.1101/2023.12.20.572602] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
A general method for designing proteins to bind and sense any small molecule of interest would be widely useful. Due to the small number of atoms to interact with, binding to small molecules with high affinity requires highly shape complementary pockets, and transducing binding events into signals is challenging. Here we describe an integrated deep learning and energy based approach for designing high shape complementarity binders to small molecules that are poised for downstream sensing applications. We employ deep learning generated psuedocycles with repeating structural units surrounding central pockets; depending on the geometry of the structural unit and repeat number, these pockets span wide ranges of sizes and shapes. For a small molecule target of interest, we extensively sample high shape complementarity pseudocycles to generate large numbers of customized potential binding pockets; the ligand binding poses and the interacting interfaces are then optimized for high affinity binding. We computationally design binders to four diverse molecules, including for the first time polar flexible molecules such as methotrexate and thyroxine, which are expressed at high levels and have nanomolar affinities straight out of the computer. Co-crystal structures are nearly identical to the design models. Taking advantage of the modular repeating structure of pseudocycles and central location of the binding pockets, we constructed low noise nanopore sensors and chemically induced dimerization systems by splitting the binders into domains which assemble into the original pseudocycle pocket upon target molecule addition.
Collapse
Affiliation(s)
- Linna An
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Meerit Said
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Long Tran
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Sagardip Majumder
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Inna Goreshnik
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Gyu Rie Lee
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - David Juergens
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Justas Dauparas
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Ivan Anishchenko
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Brian Coventry
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Asim K. Bera
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alex Kang
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Paul M. Levine
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Valentina Alvarez
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Arvind Pillai
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - David Feldman
- BioInnovation Institute, DK2200 Copenhagen N, Denmark
| | - Dmitri Zorine
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Derrick R. Hicks
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Xinting Li
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - Dionne K. Vafeados
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Patrick J. Salveson
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - David Baker
- Department of Biochemistry, The University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
48
|
Giang LH, Wu KS, Lee WC, Chu SS, Do AD, Changou CA, Tran HM, Hsieh TH, Chen HH, Hsieh CL, Sung SY, Yu AL, Yen Y, Wong TT, Chang CC. Targeting of RRM2 suppresses DNA damage response and activates apoptosis in atypical teratoid rhabdoid tumor. J Exp Clin Cancer Res 2023; 42:346. [PMID: 38124207 PMCID: PMC10731702 DOI: 10.1186/s13046-023-02911-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/19/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Atypical teratoid rhabdoid tumors (ATRT) is a rare but aggressive malignancy in the central nervous system, predominantly occurring in early childhood. Despite aggressive treatment, the prognosis of ATRT patients remains poor. RRM2, a subunit of ribonucleotide reductase, has been reported as a biomarker for aggressiveness and poor prognostic conditions in several cancers. However, little is known about the role of RRM2 in ATRT. Uncovering the role of RRM2 in ATRT will further promote the development of feasible strategies and effective drugs to treat ATRT. METHODS Expression of RRM2 was evaluated by molecular profiling analysis and was confirmed by IHC in both ATRT patients and PDX tissues. Follow-up in vitro studies used shRNA knockdown RRM2 in three different ATRT cells to elucidate the oncogenic role of RRM2. The efficacy of COH29, an RRM2 inhibitor, was assessed in vitro and in vivo. Western blot and RNA-sequencing were used to determine the mechanisms of RRM2 transcriptional activation in ATRT. RESULTS RRM2 was found to be significantly overexpressed in multiple independent ATRT clinical cohorts through comprehensive bioinformatics and clinical data analysis in this study. The expression level of RRM2 was strongly correlated with poor survival rates in patients. In addition, we employed shRNAs to silence RRM2, which led to significantly decrease in ATRT colony formation, cell proliferation, and migration. In vitro experiments showed that treatment with COH29 resulted in similar but more pronounced inhibitory effect. Therefore, ATRT orthotopic mouse model was utilized to validate this finding, and COH29 treatment showed significant tumor growth suppression and prolong overall survival. Moreover, we provide evidence that COH29 treatment led to genomic instability, suppressed homologous recombinant DNA damage repair, and subsequently induced ATRT cell death through apoptosis in ATRT cells. CONCLUSIONS Collectively, our study uncovers the oncogenic functions of RRM2 in ATRT cell lines, and highlights the therapeutic potential of targeting RRM2 in ATRT. The promising effect of COH29 on ATRT suggests its potential suitability for clinical trials as a novel therapeutic approach for ATRT.
Collapse
Affiliation(s)
- Le Hien Giang
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Biology and Genetics, Hai Phong University of Medicine and Pharmacy, Hai Phong, 180000, Vietnam
| | - Kuo-Sheng Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Wei-Chung Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan
| | - Shing-Shung Chu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Anh Duy Do
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Physiology, Pathophysiology and Immunology, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, 700000, Vietnam
| | - Chun A Changou
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Huy Minh Tran
- Department of Neurosurgery, Faculty of Medicine, University of Medicine and Pharmacy, Ho Chi Minh City, 700000, Vietnam
| | - Tsung-Han Hsieh
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, 110, Taiwan
| | - Hsin-Hung Chen
- Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Chia-Ling Hsieh
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan
- Laboratory of Translational Medicine, Development Center for Biotechnology, Taipei, 115, Taiwan
| | - Shian-Ying Sung
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, 333, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Yun Yen
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Tai-Tong Wong
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Pediatric Brain Tumor Program, Taipei Cancer Center, Taipei Medical University, Taipei, 110, Taiwan
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Taipei Medical University Hospital and Taipei Neuroscience Institute, Taipei Medical University, Taipei, 110, Taiwan
- Neuroscience Research Center, Taipei Medical University Hospital, Taipei, 110, Taiwan
- TMU Research Center for Cancer Translational Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Che-Chang Chang
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan.
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan.
- Neuroscience Research Center, Taipei Medical University Hospital, Taipei, 110, Taiwan.
- TMU Research Center for Cancer Translational Medicine, Taipei Medical University, Taipei, 110, Taiwan.
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, 6F., Education & Research Building, Shuang-Ho Campus, No. 301, Yuantong Rd., Zhonghe Dist., New Taipei City, 23564, Taiwan.
| |
Collapse
|
49
|
Were E, Viljoen A, Rasche F. Iron necessity for chlamydospore germination in Fusarium oxysporum f. sp. cubense TR4. Biometals 2023; 36:1295-1306. [PMID: 37380939 PMCID: PMC10684721 DOI: 10.1007/s10534-023-00519-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
Fusarium wilt disease of banana, caused by the notorious soil-borne pathogen Fusarium oxysporum f. sp. cubense Tropical Race 4 (Foc TR4), is extremely difficult to manage. Manipulation of soil pH or application of synthetic iron chelators can suppress the disease through iron starvation, which inhibits the germination of pathogen propagules called chlamydospores. However, the effect of iron starvation on chlamydospore germination is largely unknown. In this study, scanning electron microscopy was used to assemble the developmental sequence of chlamydospore germination and to assess the effect of iron starvation and pH in vitro. Germination occurs in three distinct phenotypic transitions (swelling, polarized growth, outgrowth). Outgrowth, characterized by formation of a single protrusion (germ tube), occurred at 2 to 3 h, and a maximum value of 69.3% to 76.7% outgrowth was observed at 8 to 10 h after germination induction. Germination exhibited plasticity with pH as over 60% of the chlamydospores formed a germ tube between pH 3 and pH 11. Iron-starved chlamydospores exhibited polarized-growth arrest, characterized by the inability to form a germ tube. Gene expression analysis of rnr1 and rnr2, which encode the iron-dependent enzyme ribonucleotide reductase, showed that rnr2 was upregulated (p < 0.0001) in iron-starved chlamydospores compared to the control. Collectively, these findings suggest that iron and extracellular pH are crucial for chlamydospore germination in Foc TR4. Moreover, inhibition of germination by iron starvation may be linked to a different mechanism, rather than repression of the function of ribonucleotide reductase, the enzyme that controls growth by regulation of DNA synthesis.
Collapse
Affiliation(s)
- Evans Were
- Institute of Agricultural Sciences in the Tropics (Hans-Ruthenberg-Institute), University of Hohenheim, 70599, Stuttgart, Germany
| | - Altus Viljoen
- Department of Plant Pathology, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Frank Rasche
- Institute of Agricultural Sciences in the Tropics (Hans-Ruthenberg-Institute), University of Hohenheim, 70599, Stuttgart, Germany.
| |
Collapse
|
50
|
Zhang X, Joseph S, Wu D, Bowser JL, Vaziri C. The DNA Damage Response (DDR) landscape of endometrial cancer defines discrete disease subtypes and reveals therapeutic opportunities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567919. [PMID: 38045328 PMCID: PMC10690150 DOI: 10.1101/2023.11.20.567919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Genome maintenance is an enabling characteristic that allows neoplastic cells to tolerate the inherent stresses of tumorigenesis and evade therapy-induced genotoxicity. Neoplastic cells also deploy mis-expressed germ cell proteins termed Cancer Testes Antigens (CTAs) to promote genome maintenance and survival. Here, we present the first comprehensive characterization of the DNA Damage Response (DDR) and CTA transcriptional landscapes of endometrial cancer in relation to conventional histological and molecular subtypes. We show endometrial serous carcinoma (ESC), an aggressive endometrial cancer subtype, is defined by gene expression signatures comprising members of the Replication Fork Protection Complex (RFPC) and Fanconi Anemia (FA) pathway and CTAs with mitotic functions. DDR and CTA- based profiling also defines a subset of highly aggressive endometrioid endometrial carcinomas (EEC) with poor clinical outcomes that share similar profiles to ESC yet have distinct characteristics based on conventional histological and genomic features. Using an unbiased CRISPR-based genetic screen and a candidate gene approach, we confirm that DDR and CTA genes that constitute the ESC and related EEC gene signatures are required for proliferation and therapy-resistance of cultured endometrial cancer cells. Our study validates the use of DDR and CTA-based tumor classifiers and reveals new vulnerabilities of aggressive endometrial cancer where none currently exist.
Collapse
|