1
|
Volkov VV, Perry CC, Chelli R. Binding Zinc and Oxo-Vanadium Insulin-Mimetic Complexes to Phosphatase Enzymes: Structure, Electronics and Implications. Molecules 2025; 30:1469. [PMID: 40286074 PMCID: PMC11990500 DOI: 10.3390/molecules30071469] [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: 02/27/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025] Open
Abstract
We explore the structural and electronic properties of representative insulin-mimetic oxovanadium and zinc complexes as computed in vacuum, in water clusters and upon binding to PTEN and PTP1B phosphatases. Albeit diverse, the enzymes' active sites represent evolutionary variant choices of the same type of biochemistry. Though different in respect to covalency and the orbital nature of bonding, theory predicts comparable ionic radii, bond lengths and square pyramidal coordination for the considered vanadyl and zinc systems when in an aqueous environment. Employing docking, DFT and quantum mechanics/molecular mechanics methods, we address possible polar interactions in the protein environments and compute infrared/Raman modes and optical electronic properties, which may be suitable for the structural analysis of the specific chemical moieties in binding studies. Accounting for how protein embedding may alter the electronic states of metal centres, we discuss artificial intelligence-assisted protein field engineering to assist biomedical and quantum information applications.
Collapse
Affiliation(s)
- Victor V. Volkov
- Independent Researcher, Bereozovaya 2a, Konstantinovo 140207, Moscow Region, Russia
| | - Carole C. Perry
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| | - Riccardo Chelli
- Dipartimento di Chimica, Università di Firenze, Via della Lastruccia 3, I-50019 Sesto Fiorentino, Italy
| |
Collapse
|
2
|
Yehorova D, Alansson N, Shen R, Denson JM, Robinson M, Risso VA, Molina NR, Loria JP, Gaucher EA, Sanchez-Ruiz JM, Hengge AC, Johnson SJ, Kamerlin SCL. Conformational Dynamics and Catalytic Backups in a Hyper-Thermostable Engineered Archaeal Protein Tyrosine Phosphatase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645524. [PMID: 40196513 PMCID: PMC11974932 DOI: 10.1101/2025.03.26.645524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Protein tyrosine phosphatases (PTPs) are a family of enzymes that play important roles in regulating cellular signaling pathways. The activity of these enzymes is regulated by the motion of a catalytic loop that places a critical conserved aspartic acid side chain into the active site for acid-base catalysis upon loop closure. These enzymes also have a conserved phosphate binding loop that is typically highly rigid and forms a well-defined anion binding nest. The intimate links between loop dynamics and chemistry in these enzymes make PTPs an excellent model system for understanding the role of loop dynamics in protein function and evolution. In this context, archaeal PTPs, which have evolved in extremophilic organisms, are highly understudied, despite their unusual biophysical properties. We present here an engineered chimeric PTP (ShufPTP) generated by shuffling the amino acid sequence of five extant hyperthermophilic archaeal PTPs. Despite ShufPTP's high sequence similarity to its natural counterparts, ShufPTP presents a suite of unique properties, including high flexibility of the phosphate binding P-loop, facile oxidation of the active site cysteine, mechanistic promiscuity, and most notably, hyperthermostability, with a denaturation temperature likely >130 °C (>8°C higher than the highest recorded growth temperature of any archaeal strain). Our combined structural, biochemical, biophysical and computational analysis provides insight both into how small steps in evolutionary space can radically modulate the biophysical properties of an enzyme, and showcase the tremendous potential of archaeal enzymes for biotechnology, to generate novel enzymes capable of operating under extreme conditions.
Collapse
Affiliation(s)
- Dariia Yehorova
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive NW, Atlanta, GA 30332, USA
| | - Nikolas Alansson
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive NW, Atlanta, GA 30332, USA
| | - Ruidan Shen
- Department of Chemistry & Biochemistry, Utah State University, 0300 Old Main Hill, Logan, UT 84322-0300, USA
| | - Joshua M Denson
- Department of Chemistry & Biochemistry, Utah State University, 0300 Old Main Hill, Logan, UT 84322-0300, USA
| | - Michael Robinson
- Department of Chemistry - BMC, Uppsala University, BMC Box 576, S-751 23 Uppsala, Sweden
| | - Valeria A Risso
- Departamento de Química Física, Facultad de Ciencias, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente (UEQ), Universidad de Granada, Granada, 18071, Spain
| | - Nuria Ramirez Molina
- Department of Biology, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - J Patrick Loria
- Department of Chemistry, Yale University, P. O. Box 208107, New Haven, CT, 06520-8107
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA
| | - Eric A Gaucher
- Department of Biology, Georgia State University, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Jose M Sanchez-Ruiz
- Departamento de Química Física, Facultad de Ciencias, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente (UEQ), Universidad de Granada, Granada, 18071, Spain
| | - Alvan C Hengge
- Department of Chemistry & Biochemistry, Utah State University, 0300 Old Main Hill, Logan, UT 84322-0300, USA
| | - Sean J Johnson
- Department of Chemistry & Biochemistry, Utah State University, 0300 Old Main Hill, Logan, UT 84322-0300, USA
| | - Shina C L Kamerlin
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive NW, Atlanta, GA 30332, USA
- Department of Chemistry, Lund University, Box 124, 22100 Lund, Sweden
| |
Collapse
|
3
|
Dinda R, Garribba E, Sanna D, Crans DC, Costa Pessoa J. Hydrolysis, Ligand Exchange, and Redox Properties of Vanadium Compounds: Implications of Solution Transformation on Biological, Therapeutic, and Environmental Applications. Chem Rev 2025; 125:1468-1603. [PMID: 39818783 DOI: 10.1021/acs.chemrev.4c00475] [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: 01/19/2025]
Abstract
Vanadium is a transition metal with important industrial, technological, biological, and biomedical applications widespread in the environment and in living beings. The different reactions that vanadium compounds (VCs) undergo in the presence of proteins, nucleic acids, lipids and metabolites under mild physiological conditions are reviewed. In the environment vanadium is present naturally or through anthropogenic sources, the latter having an environmental impact caused by the dispersion of VCs in the atmosphere and aquifers. Vanadium has a versatile chemistry with interconvertible oxidation states, variable coordination number and geometry, and ability to form polyoxidovanadates with various nuclearity and structures. If a VC is added to a water-containing environment it can undergo hydrolysis, ligand-exchange, redox, and other types of changes, determined by the conditions and speciation chemistry of vanadium. Importantly, the solution is likely to differ from the VC introduced into the system and varies with concentration. Here, vanadium redox, hydrolytic and ligand-exchange chemical reactions, the influence of pH, concentration, salt, specific solutes, biomolecules, and VCs on the speciation are described. One of our goals with this work is highlight the need for assessment of the VC speciation, so that beneficial or toxic species might be identified and mechanisms of action be elucidated.
Collapse
Affiliation(s)
- Rupam Dinda
- Department of Chemistry, National Institute of Technology, Rourkela, 769008 Odisha, India
| | - Eugenio Garribba
- Dipartimento di Medicina, Chirurgia e Farmacia, Università di Sassari, Viale San Pietro, I-07100 Sassari, Italy
| | - Daniele Sanna
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Trav. La Crucca 3, I-07040 Sassari, Italy
| | - Debbie C Crans
- Department Chemistry and Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523, United States
| | - João Costa Pessoa
- Centro de Química Estrutural and Departamento de Engenharia Química, Institute of Molecular Sciences, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
4
|
Shen R, Brownless ALR, Alansson N, Corbella M, Kamerlin SCL, Hengge AC. SHP-1 Variants Broaden the Understanding of pH-Dependent Activities in Protein Tyrosine Phosphatases. JACS AU 2024; 4:2874-2885. [PMID: 39211599 PMCID: PMC11350601 DOI: 10.1021/jacsau.4c00078] [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: 01/26/2024] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 09/04/2024]
Abstract
The protein tyrosine phosphatase (PTP) SHP-1 plays an important role in both immune regulation and oncogenesis. This enzyme is part of a broader family of PTPs that all play important regulatory roles in vivo. Common to these enzymes is a highly conserved aspartic acid (D421 in SHP-1) that acts as an acid/base catalyst during the PTP-catalyzed reaction. This residue is located on a mobile loop, the WPD-loop, the dynamic behavior of which is intimately connected to the catalytic activity. The SHP-1 WPD-loop variants H422Q, E427A, and S418A have been kinetically characterized and compared to those of the wild-type (WT) enzyme. These variants exhibit limiting magnitudes of k cat ranging from 43 to 77% of the WT enzyme. However, their pH profiles are significantly broadened in the basic pH range. As a result, above pH 6, the E427A and S418A variants have turnover numbers notably higher than those of WT SHP-1. Molecular modeling results indicate that the shifted pH dependencies result primarily from changes in solvation and hydrogen-bonding networks that affect the pK a of the D421 residue, explaining the changes in pH-rate profiles for k cat on the basic side. In contrast, a previous study of a noncatalytic residue variant of the PTP YopH, which also exhibited changes in pH dependency, showed that the catalytic change arose from mutation-induced changes in conformational equilibria of the WPD-loop. This finding and the present study show the existence of distinct strategies for nature to tune the activity of PTPs in particular environments through controlling the pH dependency of catalysis.
Collapse
Affiliation(s)
- Ruidan Shen
- Department
of Chemistry and Biochemistry, Utah State
University, Logan, Utah 84322-0300, United States
| | - Alfie-Louise R. Brownless
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, 901 Atlantic Drive NW, Atlanta, Georgia 30332-0400, United States
| | - Nikolas Alansson
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, 901 Atlantic Drive NW, Atlanta, Georgia 30332-0400, United States
| | - Marina Corbella
- Departament
de Quımica Inorgànica i Orgànica (Secció
de Quımica Orgànica) & Institut de Quımica
Teòrica i Computacional (IQTCUB), Universitat de Barcelona, Martı i Franquès 1, 08028 Barcelona, Spain
- Science
for Life Laboratory, Department of Chemistry—BMC, Uppsala University, BMC, P.O. Box 576, S-751 23 Uppsala, Sweden
| | - Shina C. L. Kamerlin
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, 901 Atlantic Drive NW, Atlanta, Georgia 30332-0400, United States
- Science
for Life Laboratory, Department of Chemistry—BMC, Uppsala University, BMC, P.O. Box 576, S-751 23 Uppsala, Sweden
| | - Alvan C. Hengge
- Department
of Chemistry and Biochemistry, Utah State
University, Logan, Utah 84322-0300, United States
| |
Collapse
|
5
|
Imhoff A, Sweeney NL, Bongard RD, Syrlybaeva R, Gupta A, Del Carpio E, Talipov MR, Garcia-Keller C, Crans DC, Ramchandran R, Sem DS. Structural and kinetic characterization of DUSP5 with a Di-phosphorylated tripeptide substrate from the ERK activation loop. FRONTIERS IN CHEMICAL BIOLOGY 2024; 3:1385560. [PMID: 39749114 PMCID: PMC11694514 DOI: 10.3389/fchbi.2024.1385560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Introduction Dual specific phosphatases (DUSPs) are mitogen-activated protein kinase (MAPK) regulators, which also serve as drug targets for treating various vascular diseases. Previously, we have presented mechanistic characterizations of DUSP5 and its interaction with pERK, proposing a dual active site. Methods Herein, we characterize the interactions between the DUSP5 phosphatase domain and the pT-E-pY activation loop of ERK2, with specific active site assignments. We also report the full NMR chemical shift assignments of DUSP5 that now enable chemical shift perturbation and dynamics studies. Results and Discussion Both phosphates of the pT-E-pY tripeptide are dephosphorylated, based on 31P NMR; but, steady state kinetic studies of the tripeptide both as a substrate and as an inhibitor indicate a preference for binding and dephosphorylation of the phospho-tyrosine before the phospho-threonine. Catalytic efficiency (kcat/Km) is 3.7 M-1S-1 for T-E-pY vs 1.3 M-1S-1 for pT-E-Y, although the diphosphorylated peptide (pT-E-pY) is a better substrate than both, with kcat/Km = 18.2 M-1S-1. Steady state inhibition studies with the pNPP substrate yields Kis values for the peptide inhibitors of: 15.82 mM (pT-E-Y), 4.932 mM (T-E-pY), 1.672 mM (pT-E-pY). Steady state inhibition studies with pNPP substrate and with vanadate or phosphate inhibitors indicated competitive inhibition with Kis values of 0.0006122 mM (sodium vanadate) and 17.32 mM (sodium phosphate), similar to other Protein Tyrosine Phosphatases with an active site cysteine nucleophile that go through a five-coordinate high energy transition state or intermediate. Molecular dynamics (MD) studies confirm preferential binding of the diphosphorylated peptide, but with preference for binding the pY over the pT reside in the catalytic site proximal to the Cys263 nucleophile. Based on MD, the monophosphorylated peptide binds tighter if phosphorylated on the Tyr vs the Thr. And, if the starting pose of the docked diphosphorylated peptide has pT in the catalytic site, it will adjust to have the pY in the catalytic site, suggesting a dynamic shifting of the peptide orientation. 2D 1H-15N HSQC chemical shift perturbation studies confirm that DUSP5 with tripeptide bound is in a dynamic state, with extensive exchange broadening observed-especially of catalytic site residues. The availability of NMR chemical shift assignments enables additional future studies of DUSP5 binding to the ERK2 diphosphorylated activation loop. Summary These studies indicate a preference for pY before pT binding, but with ability to bind and dephosphorylate both residues, and with a dynamic active site pocket that accommodates multiple tripeptide orientations.
Collapse
Affiliation(s)
- Andrea Imhoff
- Center for Structure-based Drug Design and Development, Department of Pharmaceutical Sciences, Concordia University Wisconsin, Mequon, WI, United States
| | - Noreena L. Sweeney
- Center for Structure-based Drug Design and Development, Department of Pharmaceutical Sciences, Concordia University Wisconsin, Mequon, WI, United States
| | - Robert D. Bongard
- Center for Structure-based Drug Design and Development, Department of Pharmaceutical Sciences, Concordia University Wisconsin, Mequon, WI, United States
| | - Raulia Syrlybaeva
- Department of Chemistry and Biochemistry, New Mexico State University, MSC 3C, Las Cruces, NM, United States
| | - Ankan Gupta
- Department of Pediatrics, Division of Neonatology, Children’s Research Institute (CRI) Developmental Vascular Biology Program, Translational and Biomedical Research Center, Milwaukee, WI, United States
| | - Edgar Del Carpio
- Department of Chemistry, Colorado State University, Fort Collins, CO, United States
| | - Marat R. Talipov
- Department of Chemistry and Biochemistry, New Mexico State University, MSC 3C, Las Cruces, NM, United States
| | - Costanza Garcia-Keller
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Debbie C. Crans
- Department of Chemistry, Colorado State University, Fort Collins, CO, United States
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, United States
| | - Ramani Ramchandran
- Department of Pediatrics, Division of Neonatology, Children’s Research Institute (CRI) Developmental Vascular Biology Program, Translational and Biomedical Research Center, Milwaukee, WI, United States
| | - Daniel S. Sem
- Center for Structure-based Drug Design and Development, Department of Pharmaceutical Sciences, Concordia University Wisconsin, Mequon, WI, United States
| |
Collapse
|
6
|
Frando A, Grundner C. More than two components: complexities in bacterial phosphosignaling. mSystems 2024; 9:e0028924. [PMID: 38591891 PMCID: PMC11097640 DOI: 10.1128/msystems.00289-24] [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] [Indexed: 04/10/2024] Open
Abstract
For over 40 years, the two-component systems (TCSs) have taken front and center in our thinking about the signaling mechanisms by which bacteria sense and respond to their environment. In contrast, phosphorylation on Ser/Thr and Tyr (O-phosphorylation) was long thought to be mostly restricted to eukaryotes and a somewhat accessory signaling mechanism in bacteria. Several recent studies exploring systems aspects of bacterial O-phosphorylation, however, now show that it is in fact pervasive, with some bacterial proteomes as highly phosphorylated as those of eukaryotes. Labile, non-canonical protein phosphorylation sites on Asp, Arg, and His are now also being identified in large numbers in bacteria and first cellular functions are discovered. Other phosphomodifications on Cys, Glu, and Lys remain largely unexplored. The surprising breadth and complexity of bacterial phosphosignaling reveals a vast signaling capacity, the full scope of which we may only now be beginning to understand but whose functions are likely to affect all aspects of bacterial physiology and pathogenesis.
Collapse
Affiliation(s)
- Andrew Frando
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Christoph Grundner
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
7
|
Chen L, Qian Z, Zheng Y, Zhang J, Sun J, Zhou C, Xiao H. Structural analysis of PTPN21 reveals a dominant-negative effect of the FERM domain on its phosphatase activity. SCIENCE ADVANCES 2024; 10:eadi7404. [PMID: 38416831 PMCID: PMC10901363 DOI: 10.1126/sciadv.adi7404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 01/24/2024] [Indexed: 03/01/2024]
Abstract
PTPN21 belongs to the four-point-one, ezrin, radixin, moesin (FERM) domain-containing protein tyrosine phosphatases (PTP) and plays important roles in cytoskeleton-associated cellular processes like cell adhesion, motility, and cargo transport. Because of the presence of a WPE loop instead of a WPD loop in the phosphatase domain, it is often considered to lack phosphatase activity. However, many of PTPN21's biological functions require its catalytic activity. To reconcile these findings, we have determined the structures of individual PTPN21 FERM, PTP domains, and a complex between FERM-PTP. Combined with biochemical analysis, we have found that PTPN21 PTP is weakly active and is autoinhibited by association with its FERM domain. Disruption of FERM-PTP interaction results in enhanced ERK activation. The oncogenic HPV18 E7 protein binds to PTP at the same location as PTPN21 FERM, indicating that it may act by displacing the FERM domain from PTP. Our results provide mechanistic insight into PTPN21 and benefit functional studies of PTPN21-mediated processes.
Collapse
Affiliation(s)
- Lu Chen
- Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Zijun Qian
- Department of Hematology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Yuyuan Zheng
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Jie Zhang
- Department of Hematology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Jie Sun
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Chun Zhou
- Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Haowen Xiao
- Department of Hematology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
8
|
Welsh CL, Madan LK. Allostery in Protein Tyrosine Phosphatases is Enabled by Divergent Dynamics. J Chem Inf Model 2024; 64:1331-1346. [PMID: 38346324 PMCID: PMC11144062 DOI: 10.1021/acs.jcim.3c01615] [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: 02/27/2024]
Abstract
Dynamics-driven allostery provides important insights into the working mechanics of proteins, especially enzymes. In this study, we employ this paradigm to answer a basic question: in enzyme superfamilies, where the catalytic mechanism, active sites, and protein fold are conserved, what accounts for the difference in the catalytic prowess of the individual members? We show that when subtle changes in sequence do not translate to changes in structure, they do translate to changes in dynamics. We use sequentially diverse PTP1B, TbPTP1, and YopH as representatives of the conserved protein tyrosine phosphatase (PTP) superfamily. Using amino acid network analysis of group behavior (community analysis) and influential node dominance on networks (eigenvector centrality), we explain the dynamic basis of the catalytic variations seen between the three proteins. Importantly, we explain how a dynamics-based blueprint makes PTP1B amenable to allosteric control and how the same is abstracted in TbPTP1 and YopH.
Collapse
Affiliation(s)
- Colin L. Welsh
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, College of Medicine, Medical University of South Carolina, Charleston, SC-29425, USA
| | - Lalima K. Madan
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, College of Medicine, Medical University of South Carolina, Charleston, SC-29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC-29425, USA
| |
Collapse
|
9
|
Mahdizadeh S, Stier M, Carlesso A, Lamy A, Thomas M, Eriksson LA. Multiscale In Silico Study of the Mechanism of Activation of the RtcB Ligase by the PTP1B Phosphatase. J Chem Inf Model 2024; 64:905-917. [PMID: 38282538 PMCID: PMC10865347 DOI: 10.1021/acs.jcim.3c01600] [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: 10/03/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/30/2024]
Abstract
Inositol-requiring enzyme 1 (IRE1) is a transmembrane sensor that is part of a trio of sensors responsible for controlling the unfolded protein response within the endoplasmic reticulum (ER). Upon the accumulation of unfolded or misfolded proteins in the ER, IRE1 becomes activated and initiates the cleavage of a 26-nucleotide intron from human X-box-containing protein 1 (XBP1). The cleavage is mediated by the RtcB ligase enzyme, which splices together two exons, resulting in the formation of the spliced isoform XBP1s. The XBP1s isoform activates the transcription of genes involved in ER-associated degradation to maintain cellular homeostasis. The catalytic activity of RtcB is controlled by the phosphorylation and dephosphorylation of three tyrosine residues (Y306, Y316, and Y475), which are regulated by the ABL1 tyrosine kinase and PTP1B phosphatase, respectively. This study focuses on investigating the mechanism by which the PTP1B phosphatase activates the RtcB ligase using a range of advanced in silico methods. Protein-protein docking identified key interacting residues between RtcB and PTP1B. Notably, the phosphorylated Tyr306 formed hydrogen bonds and salt bridge interactions with the "gatekeeper" residues Arg47 and Lys120 of the inactive PTP1B. Classical molecular dynamics simulation emphasized the crucial role of Asp181 in the activation of PTP1B, driving the conformational change from an open to a closed state of the WPD-loop. Furthermore, QM/MM-MD simulations provided insights into the free energy landscape of the dephosphorylation reaction mechanism of RtcB, which is mediated by the PTP1B phosphatase.
Collapse
Affiliation(s)
- Sayyed
Jalil Mahdizadeh
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, 405 30 Gothenburg, Sweden
| | - Michael Stier
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, 405 30 Gothenburg, Sweden
| | - Antonio Carlesso
- Department
of Pharmacology, Sahlgrenska Academy, University
of Gothenburg, 413 90 Gothenburg, Sweden
- Università
della Svizzera italiana (USI), Faculty of Biomedical Sciences, Euler
Institute, Via G. Buffi
13, CH-6900 Lugano, Switzerland
| | - Aurore Lamy
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, 405 30 Gothenburg, Sweden
- Department
of Bioinformatics and Chemical Communication, Research Institute in Semiochemistry and Applied Ethology, Quartier Salignan, 84400 Apt, France
| | - Melissa Thomas
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, 405 30 Gothenburg, Sweden
| | - Leif A. Eriksson
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, 405 30 Gothenburg, Sweden
| |
Collapse
|
10
|
Sharma S, Skaist Mehlman T, Sagabala RS, Boivin B, Keedy DA. High-resolution double vision of the allosteric phosphatase PTP1B. Acta Crystallogr F Struct Biol Commun 2024; 80:1-12. [PMID: 38133579 PMCID: PMC10833341 DOI: 10.1107/s2053230x23010749] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B) plays important roles in cellular homeostasis and is a highly validated therapeutic target for multiple human ailments, including diabetes, obesity and breast cancer. However, much remains to be learned about how conformational changes may convey information through the structure of PTP1B to enable allosteric regulation by ligands or functional responses to mutations. High-resolution X-ray crystallography can offer unique windows into protein conformational ensembles, but comparison of even high-resolution structures is often complicated by differences between data sets, including non-isomorphism. Here, the highest resolution crystal structure of apo wild-type (WT) PTP1B to date is presented out of a total of ∼350 PTP1B structures in the PDB. This structure is in a crystal form that is rare for PTP1B, with two unique copies of the protein that exhibit distinct patterns of conformational heterogeneity, allowing a controlled comparison of local disorder across the two chains within the same asymmetric unit. The conformational differences between these chains are interrogated in the apo structure and between several recently reported high-resolution ligand-bound structures. Electron-density maps in a high-resolution structure of a recently reported activating double mutant are also examined, and unmodeled alternate conformations in the mutant structure are discovered that coincide with regions of enhanced conformational heterogeneity in the new WT structure. These results validate the notion that these mutations operate by enhancing local dynamics, and suggest a latent susceptibility to such changes in the WT enzyme. Together, these new data and analysis provide a detailed view of the conformational ensemble of PTP1B and highlight the utility of high-resolution crystallography for elucidating conformational heterogeneity with potential relevance for function.
Collapse
Affiliation(s)
- Shivani Sharma
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY 10031, USA
- PhD Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Tamar Skaist Mehlman
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY 10031, USA
| | - Reddy Sudheer Sagabala
- Department of Nanobioscience, College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA
| | - Benoit Boivin
- Department of Nanobioscience, College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA
| | - Daniel A. Keedy
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY 10031, USA
- Department of Chemistry and Biochemistry, City College of New York, New York, NY 10031, USA
- PhD Programs in Biochemistry, Biology and Chemistry, CUNY Graduate Center, New York, NY 10016, USA
| |
Collapse
|
11
|
Šinko G. Modeling of a near-attack conformation of oxime in phosphorylated acetylcholinesterase via a reactivation product, a phosphorylated oxime. Chem Biol Interact 2023; 383:110656. [PMID: 37579936 DOI: 10.1016/j.cbi.2023.110656] [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: 02/09/2023] [Revised: 07/26/2023] [Accepted: 08/09/2023] [Indexed: 08/16/2023]
Abstract
At the present, only four antidotes are in use in therapy for poisoning by organophosphorus compounds: 2-PAM, HI-6, obidoxime and trimedoxime. Numerous compounds have been designed and synthetized to be more effective reactivators than those currently in use. Many of those new compounds fail at the enzyme level because interactions formed within the AChE active site are not favourable ones that lead to a successful reactivation. The approach in which the modeling of a phosphorylated oxime (POX), a product of successful reactivation in the AChE active site, may be a way to better understand the role of active site residues during the process of formation of the Michaelis type of complex between an enzyme and oxime. After reactivation, a change in phosphorus stereochemistry occurs leading to a different spatial arrangement of attached substituents, now including an oxime. To study interactions between the AChE oxyanion hole and a phosphorylated oxime, an S203G mutant was used to avoid the steric hindrance caused by the catalytic serine. In this way, the POX could be positioned close to the oxyanion hole. In the final step, the oxime without a phosphoester moiety was transferred into the phosphorylated AChE and molecular dynamics was used to test the stability of the near-attack conformation of the oxime near the phosphorylated serine.
Collapse
Affiliation(s)
- Goran Šinko
- Institute for Medical Research and Occupational Health, Ksaverska cesta 2, HR-10 000, Zagreb, Croatia.
| |
Collapse
|
12
|
Corbella M, Pinto GP, Kamerlin SCL. Loop dynamics and the evolution of enzyme activity. Nat Rev Chem 2023; 7:536-547. [PMID: 37225920 DOI: 10.1038/s41570-023-00495-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2023] [Indexed: 05/26/2023]
Abstract
In the early 2000s, Tawfik presented his 'New View' on enzyme evolution, highlighting the role of conformational plasticity in expanding the functional diversity of limited repertoires of sequences. This view is gaining increasing traction with increasing evidence of the importance of conformational dynamics in both natural and laboratory evolution of enzymes. The past years have seen several elegant examples of harnessing conformational (particularly loop) dynamics to successfully manipulate protein function. This Review revisits flexible loops as critical participants in regulating enzyme activity. We showcase several systems of particular interest: triosephosphate isomerase barrel proteins, protein tyrosine phosphatases and β-lactamases, while briefly discussing other systems in which loop dynamics are important for selectivity and turnover. We then discuss the implications for engineering, presenting examples of successful loop manipulation in either improving catalytic efficiency, or changing selectivity completely. Overall, it is becoming clearer that mimicking nature by manipulating the conformational dynamics of key protein loops is a powerful method of tailoring enzyme activity, without needing to target active-site residues.
Collapse
Affiliation(s)
- Marina Corbella
- Department of Chemistry, Uppsala University, Uppsala, Sweden
| | - Gaspar P Pinto
- Department of Chemistry, Uppsala University, Uppsala, Sweden
- Cortex Discovery GmbH, Regensburg, Germany
| | - Shina C L Kamerlin
- Department of Chemistry, Uppsala University, Uppsala, Sweden.
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
13
|
Liang S, Tran E, Du X, Dong J, Sudholz H, Chen H, Qu Z, Huntington ND, Babon JJ, Kershaw NJ, Zhang ZY, Baell JB, Wiede F, Tiganis T. A small molecule inhibitor of PTP1B and PTPN2 enhances T cell anti-tumor immunity. Nat Commun 2023; 14:4524. [PMID: 37500611 PMCID: PMC10374545 DOI: 10.1038/s41467-023-40170-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/15/2023] [Indexed: 07/29/2023] Open
Abstract
The inhibition of protein tyrosine phosphatases 1B (PTP1B) and N2 (PTPN2) has emerged as an exciting approach for bolstering T cell anti-tumor immunity. ABBV-CLS-484 is a PTP1B/PTPN2 inhibitor in clinical trials for solid tumors. Here we have explored the therapeutic potential of a related small-molecule-inhibitor, Compound-182. We demonstrate that Compound-182 is a highly potent and selective active site competitive inhibitor of PTP1B and PTPN2 that enhances T cell recruitment and activation and represses the growth of tumors in mice, without promoting overt immune-related toxicities. The enhanced anti-tumor immunity in immunogenic tumors can be ascribed to the inhibition of PTP1B/PTPN2 in T cells, whereas in cold tumors, Compound-182 elicited direct effects on both tumor cells and T cells. Importantly, treatment with Compound-182 rendered otherwise resistant tumors sensitive to α-PD-1 therapy. Our findings establish the potential for small molecule inhibitors of PTP1B and PTPN2 to enhance anti-tumor immunity and combat cancer.
Collapse
Affiliation(s)
- Shuwei Liang
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Eric Tran
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Xin Du
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Jiajun Dong
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
| | - Harrison Sudholz
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Hao Chen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, 3052, Australia
| | - Zihan Qu
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Nicholas D Huntington
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Jeffrey J Babon
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, 3052, Australia
| | - Nadia J Kershaw
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, 3052, Australia
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Jonathan B Baell
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Lyterian Therapeutics, South San Francisco, San Francisco, CA, 94080, USA
| | - Florian Wiede
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
| | - Tony Tiganis
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
14
|
Welsh CL, Madan LK. Allostery in Protein Tyrosine Phosphatases is Enabled by Divergent Dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.23.550226. [PMID: 37547015 PMCID: PMC10402003 DOI: 10.1101/2023.07.23.550226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Dynamics-driven allostery provides important insights into the working mechanics of proteins, especially enzymes. In this study we employ this paradigm to answer a basic question: in enzyme superfamilies where the catalytic mechanism, active sites and protein fold are conserved, what accounts for the difference in the catalytic prowess of the individual members? We show that when subtle changes in sequence do not translate to changes in structure, they do translate to changes in dynamics. We use sequentially diverse PTP1B, TbPTP1, and YopH as the representatives of the conserved Protein Tyrosine Phosphatase (PTP) superfamily. Using amino acid network analysis of group behavior (community analysis) and influential node dominance on networks (eigenvector centrality), we explain the dynamic basis of catalytic variations seen between the three proteins. Importantly, we explain how a dynamics-based blueprint makes PTP1B amenable to allosteric control and how the same is abstracted in TbPTP1 and YopH.
Collapse
|
15
|
Liang S, Tran E, Du X, Dong J, Sudholz H, Chen H, Qu Z, Huntington N, Babon J, Kershaw N, Zhang ZY, Baell J, Wiede F, Tiganis T. A small molecule inhibitor of PTP1B and PTPN2 enhances T cell anti-tumor immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.545220. [PMID: 37397992 PMCID: PMC10312756 DOI: 10.1101/2023.06.16.545220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The inhibition of protein tyrosine phosphatases (PTPs), such as PTP1B and PTPN2 that function as intracellular checkpoints, has emerged as an exciting new approach for bolstering T cell anti-tumor immunity to combat cancer. ABBV-CLS-484 is a dual PTP1B and PTPN2 inhibitor currently in clinical trials for solid tumors. Here we have explored the therapeutic potential of targeting PTP1B and PTPN2 with a related small molecule inhibitor, Compound 182. We demonstrate that Compound 182 is a highly potent and selective active site competitive inhibitor of PTP1B and PTPN2 that enhances antigen-induced T cell activation and expansion ex vivo and represses the growth of syngeneic tumors in C57BL/6 mice without promoting overt immune-related toxicities. Compound 182 repressed the growth of immunogenic MC38 colorectal and AT3-OVA mammary tumors as well as immunologically cold AT3 mammary tumors that are largely devoid of T cells. Treatment with Compound 182 increased both the infiltration and activation of T cells, as well as the recruitment of NK cells and B cells that promote anti-tumor immunity. The enhanced anti-tumor immunity in immunogenic AT3-OVA tumors could be ascribed largely to the inhibition of PTP1B/PTPN2 in T cells, whereas in cold AT3 tumors, Compound 182 elicited both direct effects on tumor cells and T cells to facilitate T cell recruitment and thereon activation. Importantly, treatment with Compound 182 rendered otherwise resistant AT3 tumors sensitive to anti-PD1 therapy. Our findings establish the potential for small molecule active site inhibitors of PTP1B and PTPN2 to enhance anti-tumor immunity and combat cancer.
Collapse
|
16
|
Hashmi K, Gupta S, Siddique A, Khan T, Joshi S. Medicinal applications of vanadium complexes with Schiff bases. J Trace Elem Med Biol 2023; 79:127245. [PMID: 37406475 DOI: 10.1016/j.jtemb.2023.127245] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/31/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
Many transition metal complexes have been explored for their therapeutic properties after the discovery of cisplatin. Schiff bases have an efficient complexation tendency with the transition metals and several medicinal properties have been reported. However, fewer studies have reported the medicinal utility of vanadium and its Schiff base complexes. This paper provides a comprehensive overview of vanadium complexes with Schiff bases along with their mechanistic insight. Vanadium complexes in + 4 and + 5 oxidation states have exhibited well-defined geometry and found to be thermodynamically stable. The studies have reported the G0/G1 phase cell cycle arrest and decreased delta psi m, inducing mitochondrial membrane depolarization in cancer cell lines along with the alterations in the metabolism of the cancer cells upon dosing with the vanadium complexes. Cancer cell invasion and growth are also found to be markedly reduced by peroxo complexes of vanadium. The studies included in the review paper have been taken from leading indexing databases and focus was laid on recent reports in literature. The biological potential of vanadium complexes of Schiff bases opens new horizons for future interdisciplinary studies and investigation focussed on understanding the biochemistry of these complexes, along with designing new complexes which have better bioavailability, solubility and low or non-toxicity.
Collapse
Affiliation(s)
- Kulsum Hashmi
- Department of Chemistry, Isabella Thoburn College, Lucknow, UP 226007, India
| | - Sakshi Gupta
- Department of Chemistry, Isabella Thoburn College, Lucknow, UP 226007, India
| | - Armeen Siddique
- Department of Chemistry, Isabella Thoburn College, Lucknow, UP 226007, India
| | - Tahmeena Khan
- Department of Chemistry, Integral University, Lucknow, UP 226026, India
| | - Seema Joshi
- Department of Chemistry, Isabella Thoburn College, Lucknow, UP 226007, India.
| |
Collapse
|
17
|
Hardie A, Cossins BP, Lovera S, Michel J. Deconstructing allostery by computational assessment of the binding determinants of allosteric PTP1B modulators. Commun Chem 2023; 6:125. [PMID: 37322137 PMCID: PMC10272186 DOI: 10.1038/s42004-023-00926-1] [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: 02/13/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023] Open
Abstract
Fragment-based drug discovery is an established methodology for finding hit molecules that can be elaborated into lead compounds. However it is currently challenging to predict whether fragment hits that do not bind to an orthosteric site could be elaborated into allosteric modulators, as in these cases binding does not necessarily translate into a functional effect. We propose a workflow using Markov State Models (MSMs) with steered molecular dynamics (sMD) to assess the allosteric potential of known binders. sMD simulations are employed to sample protein conformational space inaccessible to routine equilibrium MD timescales. Protein conformations sampled by sMD provide starting points for seeded MD simulations, which are combined into MSMs. The methodology is demonstrated on a dataset of protein tyrosine phosphatase 1B ligands. Experimentally confirmed allosteric inhibitors are correctly classified as inhibitors, whereas the deconstructed analogues show reduced inhibitory activity. Analysis of the MSMs provide insights into preferred protein-ligand arrangements that correlate with functional outcomes. The present methodology may find applications for progressing fragments towards lead molecules in FBDD campaigns.
Collapse
Affiliation(s)
- Adele Hardie
- EaStChem School of Chemistry, Joseph Black Building, University of Edinburgh, Edinburgh, EH9 3FJ, UK
| | - Benjamin P Cossins
- UCB Pharma, 216 Bath Road, Slough, UK
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford, UK
| | - Silvia Lovera
- UCB Pharma, Chemin du Foriest 1, 1420, Braine-l'Alleud, Belgium
| | - Julien Michel
- EaStChem School of Chemistry, Joseph Black Building, University of Edinburgh, Edinburgh, EH9 3FJ, UK.
| |
Collapse
|
18
|
Morris R, Keating N, Tan C, Chen H, Laktyushin A, Saiyed T, Liau NPD, Nicola NA, Tiganis T, Kershaw NJ, Babon JJ. Structure guided studies of the interaction between PTP1B and JAK. Commun Biol 2023; 6:641. [PMID: 37316570 DOI: 10.1038/s42003-023-05020-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/06/2023] [Indexed: 06/16/2023] Open
Abstract
Protein Tyrosine Phosphatase 1B (PTP1B) is the prototypical protein tyrosine phosphatase and plays an essential role in the regulation of several kinase-driven signalling pathways. PTP1B displays a preference for bisphosphorylated substrates. Here we identify PTP1B as an inhibitor of IL-6 and show that, in vitro, it can dephosphorylate all four members of the JAK family. In order to gain a detailed understanding of the molecular mechanism of JAK dephosphorylation, we undertook a structural and biochemical analysis of the dephosphorylation reaction. We identified a product-trapping PTP1B mutant that allowed visualisation of the tyrosine and phosphate products of the reaction and a substrate-trapping mutant with a vastly decreased off-rate compared to those previously described. The latter mutant was used to determine the structure of bisphosphorylated JAK peptides bound to the enzyme active site. These structures revealed that the downstream phosphotyrosine preferentially engaged the active site, in contrast to the analogous region of IRK. Biochemical analysis confirmed this preference. In this binding mode, the previously identified second aryl binding site remains unoccupied and the non-substrate phosphotyrosine engages Arg47. Mutation of this arginine disrupts the preference for the downstream phosphotyrosine. This study reveals a previously unappreciated plasticity in how PTP1B interacts with different substrates.
Collapse
Affiliation(s)
- Rhiannon Morris
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, 3052, VIC, Australia
| | - Narelle Keating
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, 3052, VIC, Australia
| | - Cyrus Tan
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, 3052, VIC, Australia
| | - Hao Chen
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, 3052, VIC, Australia
| | - Artem Laktyushin
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, VIC, Australia
| | - Tamanna Saiyed
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, VIC, Australia
| | - Nicholas P D Liau
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, 3052, VIC, Australia
| | - Nicos A Nicola
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, 3052, VIC, Australia
| | - Tony Tiganis
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Nadia J Kershaw
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, 3052, VIC, Australia
| | - Jeffrey J Babon
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, Royal Parade, Parkville, 3052, VIC, Australia.
| |
Collapse
|
19
|
Gao X, Wu Y, Chick JM, Abbott A, Jiang B, Wang DJ, Comte-Walters S, Johnson RH, Oberholtzer N, Nishimura MI, Gygi SP, Mehta A, Guttridge DC, Ball L, Mehrotra S, Sicinski P, Yu XZ, Wang H. Targeting protein tyrosine phosphatases for CDK6-induced immunotherapy resistance. Cell Rep 2023; 42:112314. [PMID: 37000627 PMCID: PMC10544673 DOI: 10.1016/j.celrep.2023.112314] [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: 01/19/2022] [Revised: 12/20/2022] [Accepted: 03/14/2023] [Indexed: 04/01/2023] Open
Abstract
Elucidating the mechanisms of resistance to immunotherapy and developing strategies to improve its efficacy are challenging goals. Bioinformatics analysis demonstrates that high CDK6 expression in melanoma is associated with poor progression-free survival of patients receiving single-agent immunotherapy. Depletion of CDK6 or cyclin D3 (but not of CDK4, cyclin D1, or D2) in cells of the tumor microenvironment inhibits tumor growth. CDK6 depletion reshapes the tumor immune microenvironment, and the host anti-tumor effect depends on cyclin D3/CDK6-expressing CD8+ and CD4+ T cells. This occurs by CDK6 phosphorylating and increasing the activities of PTP1B and T cell protein tyrosine phosphatase (TCPTP), which, in turn, decreases tyrosine phosphorylation of CD3ζ, reducing the signal transduction for T cell activation. Administration of a PTP1B and TCPTP inhibitor prove more efficacious than using a CDK6 degrader in enhancing T cell-mediated immunotherapy. Targeting protein tyrosine phosphatases (PTPs) might be an effective strategy for cancer patients who resist immunotherapy treatment.
Collapse
Affiliation(s)
- Xueliang Gao
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Yongxia Wu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Joel M Chick
- Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| | - Andrea Abbott
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Baishan Jiang
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA 02215, USA
| | - David J Wang
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Susana Comte-Walters
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Roger H Johnson
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Nathaniel Oberholtzer
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| | - Anand Mehta
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Denis C Guttridge
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Lauren Ball
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Shikhar Mehrotra
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Xue-Zhong Yu
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Haizhen Wang
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
20
|
Brickel S, Demkiv AO, Crean RM, Pinto GP, Kamerlin SCL. Q-RepEx: A Python pipeline to increase the sampling of empirical valence bond simulations. J Mol Graph Model 2023; 119:108402. [PMID: 36610324 DOI: 10.1016/j.jmgm.2022.108402] [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: 11/17/2022] [Revised: 12/17/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
The exploration of chemical systems occurs on complex energy landscapes. Comprehensively sampling rugged energy landscapes with many local minima is a common problem for molecular dynamics simulations. These multiple local minima trap the dynamic system, preventing efficient sampling. This is a particular challenge for large biochemical systems with many degrees of freedom. Replica exchange molecular dynamics (REMD) is an approach that accelerates the exploration of the conformational space of a system, and thus can be used to enhance the sampling of complex biomolecular processes. In parallel, the empirical valence bond (EVB) approach is a powerful approach for modeling chemical reactivity in biomolecular systems. Here, we present an open-source Python-based tool that interfaces with the Q simulation package, and increases the sampling efficiency of the EVB free energy perturbation/umbrella sampling approach by means of REMD. This approach, Q-RepEx, both decreases the computational cost of the associated REMD-EVB simulations, and opens the door to more efficient studies of biochemical reactivity in systems with significant conformational fluctuations along the chemical reaction coordinate.
Collapse
Affiliation(s)
- Sebastian Brickel
- Department of Chemistry - BMC, Uppsala University, BMC Box 576, S-751 23, Uppsala, Sweden
| | - Andrey O Demkiv
- Department of Chemistry - BMC, Uppsala University, BMC Box 576, S-751 23, Uppsala, Sweden
| | - Rory M Crean
- Department of Chemistry - BMC, Uppsala University, BMC Box 576, S-751 23, Uppsala, Sweden
| | - Gaspar P Pinto
- Department of Chemistry - BMC, Uppsala University, BMC Box 576, S-751 23, Uppsala, Sweden
| | - Shina Caroline Lynn Kamerlin
- Department of Chemistry - BMC, Uppsala University, BMC Box 576, S-751 23, Uppsala, Sweden; School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive NW, Atlanta, GA, 30332-0400, USA.
| |
Collapse
|
21
|
Molecular and Biological Investigation of Isolated Marine Fungal Metabolites as Anticancer Agents: A Multi-Target Approach. Metabolites 2023; 13:metabo13020162. [PMID: 36837781 PMCID: PMC9964656 DOI: 10.3390/metabo13020162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Cancer is the leading cause of death globally, with an increasing number of cases being annually reported. Nature-derived metabolites have been widely studied for their potential programmed necrosis, cytotoxicity, and anti-proliferation leading to enrichment for the modern medicine, particularly within the last couple of decades. At a more rapid pace, the concept of multi-target agents has evolved from being an innovative approach into a regular drug development procedure for hampering the multi-fashioned pathophysiology and high-resistance nature of cancer cells. With the advent of the Red Sea Penicillium chrysogenum strain S003-isolated indole-based alkaloids, we thoroughly investigated the molecular aspects for three major metabolites: meleagrin (MEL), roquefortine C (ROC), and isoroquefortine C (ISO) against three cancer-associated biological targets Cdc-25A, PTP-1B, and c-Met kinase. The study presented, for the first time, the detailed molecular insights and near-physiological affinity for these marine indole alkaloids against the assign targets through molecular docking-coupled all-atom dynamic simulation analysis. Findings highlighted the superiority of MEL's binding affinity/stability being quite in concordance with the in vitro anticancer activity profile conducted via sulforhodamine B bioassay on different cancerous cell lines reaching down to low micromolar or even nanomolar potencies. The advent of lengthy structural topologies via the metabolites' extended tetracyclic cores and aromatic imidazole arm permitted multi-pocket accommodation addressing the selectivity concerns. Additionally, the presence decorating polar functionalities on the core hydrophobic tetracyclic ring contributed compound's pharmacodynamic preferentiality. Introducing ionizable functionality with more lipophilic characters was highlighted to improve binding affinities which was also in concordance with the conducted drug-likeness/pharmacokinetic profiling for obtaining a balanced pharmacokinetic/dynamic profile. Our study adds to the knowledge regarding drug development and optimization of marine-isolated indole-based alkaloids for future iterative synthesis and pre-clinical investigations as multi-target anticancer agents.
Collapse
|
22
|
Chirgadze YN, Battaile KP, Likhachev IV, Balabaev NK, Gordon RD, Romanov V, Lin A, Karisch R, Lam R, Ruzanov M, Brazhnikov EV, Pai EF, Neel BG, Chirgadze NY. Signal transfer in human protein tyrosine phosphatase PTP1B from allosteric inhibitor P00058. J Biomol Struct Dyn 2022; 40:13823-13832. [PMID: 34705594 DOI: 10.1080/07391102.2021.1994879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Protein tyrosine phosphatases constitute a family of cytosolic and receptor-like signal transducing enzymes that catalyze the hydrolysis of phospho-tyrosine residues of phosphorylated proteins. PTP1B, encoded by PTPN1, is a key negative regulator of insulin and leptin receptor signaling, linking it to two widespread diseases: type 2 diabetes mellitus and obesity. Here, we present crystal structures of the PTP1B apo-enzyme and a complex with a newly identified allosteric inhibitor, 2-(2,5-dimethyl-pyrrol-1-yl)-5-hydroxy-benzoic acid, designated as P00058. The inhibitor binding site is located about 18 Å away from the active center. However, the inhibitor causes significant re-arrangements in the active center of enzyme: residues 45-50 of catalytic Tyr-loop are shifted at their Cα-atom positions by 2.6 to 5.8 Å. We have identified an event of allosteric signal transfer from the inhibitor to the catalytic area using molecular dynamic simulation. Analyzing change of complex structure along the fluctuation trajectory we have found the large Cα-atom shifts in external strand, residues 25-40, which occur at the same time with the shifts in adjacent catalytic p-Tyr-loop. Coming of the signal to this loop arises due to dynamic fluctuation of protein structure at about 4.0 nanoseconds after the inhibitor takes up its space. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yuri N Chirgadze
- Institute of Protein Research, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | | | - Ilya V Likhachev
- Institute of Mathematical Problems of Biology, Branch of Keldysh Institute of Applied Mathematics, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Nikolay K Balabaev
- Institute of Mathematical Problems of Biology, Branch of Keldysh Institute of Applied Mathematics, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Roni D Gordon
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | - Vladimir Romanov
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | - Andres Lin
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | - Robert Karisch
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | - Robert Lam
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada
| | - Max Ruzanov
- Molecular Structure and Design, Molecular Discovery Technologies, Bristol-Myers Squibb Research & Development, Princeton, NJ, USA
| | - Evgeniy V Brazhnikov
- Institute of Protein Research, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Emil F Pai
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada.,Department of Biochemistry and Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Benjamin G Neel
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada.,Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, NY, USA
| | - Nickolay Y Chirgadze
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, ON, Canada.,X-CHIP Technologies Inc., Toronto, ON, Canada
| |
Collapse
|
23
|
Shen R, Crean RM, Olsen KJ, Corbella M, Calixto AR, Richan T, Brandão TAS, Berry RD, Tolman A, Loria JP, Johnson SJ, Kamerlin SCL, Hengge AC. Insights into the importance of WPD-loop sequence for activity and structure in protein tyrosine phosphatases. Chem Sci 2022; 13:13524-13540. [PMID: 36507179 PMCID: PMC9682893 DOI: 10.1039/d2sc04135a] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/25/2022] [Indexed: 12/15/2022] Open
Abstract
Protein tyrosine phosphatases (PTPs) possess a conserved mobile catalytic loop, the WPD-loop, which brings an aspartic acid into the active site where it acts as an acid/base catalyst. Prior experimental and computational studies, focused on the human enzyme PTP1B and the PTP from Yersinia pestis, YopH, suggested that loop conformational dynamics are important in regulating both catalysis and evolvability. We have generated a chimeric protein in which the WPD-loop of YopH is transposed into PTP1B, and eight chimeras that systematically restored the loop sequence back to native PTP1B. Of these, four chimeras were soluble and were subjected to detailed biochemical and structural characterization, and a computational analysis of their WPD-loop dynamics. The chimeras maintain backbone structural integrity, with somewhat slower rates than either wild-type parent, and show differences in the pH dependency of catalysis, and changes in the effect of Mg2+. The chimeric proteins' WPD-loops differ significantly in their relative stability and rigidity. The time required for interconversion, coupled with electrostatic effects revealed by simulations, likely accounts for the activity differences between chimeras, and relative to the native enzymes. Our results further the understanding of connections between enzyme activity and the dynamics of catalytically important groups, particularly the effects of non-catalytic residues on key conformational equilibria.
Collapse
Affiliation(s)
- Ruidan Shen
- Department of Chemistry and Biochemistry, Utah State University Logan Utah 84322-0300 USA
| | - Rory M Crean
- Science for Life Laboratory, Department of Chemistry - BMC, Uppsala University, BMC Box 576 S-751 23 Uppsala Sweden
| | - Keith J Olsen
- Department of Chemistry and Biochemistry, Utah State University Logan Utah 84322-0300 USA
| | - Marina Corbella
- Science for Life Laboratory, Department of Chemistry - BMC, Uppsala University, BMC Box 576 S-751 23 Uppsala Sweden
| | - Ana R Calixto
- Science for Life Laboratory, Department of Chemistry - BMC, Uppsala University, BMC Box 576 S-751 23 Uppsala Sweden
| | - Teisha Richan
- Department of Chemistry and Biochemistry, Utah State University Logan Utah 84322-0300 USA
| | - Tiago A S Brandão
- Departamento de Química, ICEX, Universidade Federal de Minas Gerais Belo Horizonte Minas Gerais 31270-901 Brazil
| | - Ryan D Berry
- Department of Chemistry and Biochemistry, Utah State University Logan Utah 84322-0300 USA
| | - Alex Tolman
- Department of Chemistry and Biochemistry, Utah State University Logan Utah 84322-0300 USA
| | - J Patrick Loria
- Department of Chemistry, Yale University 225 Prospect Street New Haven CT 06520 USA
- Department of Molecular Biophysics and Biochemistry, Yale University 266 Whitney Avenue New Haven CT 06520 USA
| | - Sean J Johnson
- Department of Chemistry and Biochemistry, Utah State University Logan Utah 84322-0300 USA
| | - Shina C L Kamerlin
- Science for Life Laboratory, Department of Chemistry - BMC, Uppsala University, BMC Box 576 S-751 23 Uppsala Sweden
- School of Chemistry and Biochemistry, Georgia Institute of Technology 901 Atlantic Drive NW Atlanta, GA 30332-0400 USA
| | - Alvan C Hengge
- Department of Chemistry and Biochemistry, Utah State University Logan Utah 84322-0300 USA
| |
Collapse
|
24
|
Friedman AJ, Liechty ET, Kramer L, Sarkar A, Fox JM, Shirts MR. Allosteric Inhibition of PTP1B by a Nonpolar Terpenoid. J Phys Chem B 2022; 126:8427-8438. [PMID: 36223525 PMCID: PMC10040085 DOI: 10.1021/acs.jpcb.2c05423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Protein tyrosine phosphatases (PTPs) are promising drug targets for treating a wide range of diseases such as diabetes, cancer, and neurological disorders, but their conserved active sites have complicated the design of selective therapeutics. This study examines the allosteric inhibition of PTP1B by amorphadiene (AD), a terpenoid hydrocarbon that is an unusually selective inhibitor. Molecular dynamics (MD) simulations carried out in this study suggest that AD can stably sample multiple neighboring sites on the allosterically influential C-terminus of the catalytic domain. Binding to these sites requires a disordered α7 helix, which stabilizes the PTP1B-AD complex and may contribute to the selectivity of AD for PTP1B over TCPTP. Intriguingly, the binding mode of AD differs from that of the most well-studied allosteric inhibitor of PTP1B. Indeed, biophysical measurements and MD simulations indicate that the two molecules can bind simultaneously. Upon binding, both inhibitors destabilize the α7 helix by disrupting interactions at the α3-α7 interface and prevent the formation of hydrogen bonds that facilitate closure of the catalytically essential WPD loop. These findings indicate that AD is a promising scaffold for building allosteric inhibitors of PTP1B and illustrate, more broadly, how unfunctionalized terpenoids can engage in specific interactions with protein surfaces.
Collapse
Affiliation(s)
- Anika J Friedman
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado80309, United States
| | - Evan T Liechty
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado80309, United States
| | - Levi Kramer
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado80309, United States
| | - Ankur Sarkar
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado80309, United States
| | - Jerome M Fox
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado80309, United States
| | - Michael R Shirts
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado80309, United States
| |
Collapse
|
25
|
Netto LES, Machado LESF. Preferential redox regulation of cysteine‐based protein tyrosine phosphatases: structural and biochemical diversity. FEBS J 2022; 289:5480-5504. [DOI: 10.1111/febs.16466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/20/2022] [Accepted: 04/28/2022] [Indexed: 12/30/2022]
Affiliation(s)
- Luís Eduardo S. Netto
- Departamento de Genética e Biologia Evolutiva Instituto de Biociências Universidade de São Paulo Brazil
| | | |
Collapse
|
26
|
Kostrzewa T, Jończyk J, Drzeżdżon J, Jacewicz D, Górska-Ponikowska M, Kołaczkowski M, Kuban-Jankowska A. Synthesis, In Vitro, and Computational Studies of PTP1B Phosphatase Inhibitors Based on Oxovanadium(IV) and Dioxovanadium(V) Complexes. Int J Mol Sci 2022; 23:7034. [PMID: 35806035 PMCID: PMC9267097 DOI: 10.3390/ijms23137034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 02/06/2023] Open
Abstract
One of the main goals of recent bioinorganic chemistry studies has been to design and synthesize novel substances to treat human diseases. The promising compounds are metal-based and metal ion binding components such as vanadium-based compounds. The potential anticancer action of vanadium-based compounds is one of area of investigation in this field. In this study, we present five oxovanadium(IV) and dioxovanadium(V) complexes as potential PTP1B inhibitors with anticancer activity against the MCF-7 breast cancer cell line, the triple negative MDA-MB-231 breast cancer cell line, and the human keratinocyte HaCaT cell line. We observed that all tested compounds were effective inhibitors of PTP1B, which correlates with anticancer activity. [VO(dipic)(dmbipy)]·2 H2O (Compound 4) and [VOO(dipic)](2-phepyH)·H2O (Compound 5) possessed the greatest inhibitory effect, with IC50 185.4 ± 9.8 and 167.2 ± 8.0 nM, respectively. To obtain a better understanding of the relationship between the structure of the examined compounds and their activity, we performed a computer simulation of their binding inside the active site of PTP1B. We observed a stronger binding of complexes containing dipicolinic acid with PTP1B. Based on our simulations, we suggested that the studied complexes exert their activity by stabilizing the WPD-loop in an open position and limiting access to the P-loop.
Collapse
Affiliation(s)
- Tomasz Kostrzewa
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Jakub Jończyk
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Krakow, Poland; (J.J.); (M.K.)
| | - Joanna Drzeżdżon
- Department of Environmental Technology, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland; (J.D.); (D.J.)
| | - Dagmara Jacewicz
- Department of Environmental Technology, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland; (J.D.); (D.J.)
| | - Magdalena Górska-Ponikowska
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland;
- IEMEST Istituto Euro-Mediterraneo di Scienza e Tecnologia, 90127 Palermo, Italy
- Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70174 Stuttgart, Germany
| | - Marcin Kołaczkowski
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Krakow, Poland; (J.J.); (M.K.)
| | - Alicja Kuban-Jankowska
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland;
| |
Collapse
|
27
|
Liu R, Mathieu C, Berthelet J, Zhang W, Dupret JM, Rodrigues Lima F. Human Protein Tyrosine Phosphatase 1B (PTP1B): From Structure to Clinical Inhibitor Perspectives. Int J Mol Sci 2022; 23:ijms23137027. [PMID: 35806030 PMCID: PMC9266911 DOI: 10.3390/ijms23137027] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 01/27/2023] Open
Abstract
Phosphorylation is an essential process in biological events and is considered critical for biological functions. In tissues, protein phosphorylation mainly occurs on tyrosine (Tyr), serine (Ser) and threonine (Thr) residues. The balance between phosphorylation and dephosphorylation is under the control of two super enzyme families, protein kinases (PKs) and protein phosphatases (PPs), respectively. Although there are many selective and effective drugs targeting phosphokinases, developing drugs targeting phosphatases is challenging. PTP1B, one of the most central protein tyrosine phosphatases (PTPs), is a key player in several human diseases and disorders, such as diabetes, obesity, and hematopoietic malignancies, through modulation of different signaling pathways. However, due to high conservation among PTPs, most PTP1B inhibitors lack specificity, raising the need to develop new strategies targeting this enzyme. In this mini-review, we summarize three classes of PTP1B inhibitors with different mechanisms: (1) targeting multiple aryl-phosphorylation sites including the catalytic site of PTP1B; (2) targeting allosteric sites of PTP1B; (3) targeting specific mRNA sequence of PTP1B. All three types of PTP1B inhibitors present good specificity over other PTPs and are promising for the development of efficient small molecules targeting this enzyme.
Collapse
Affiliation(s)
- Rongxing Liu
- Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France; (R.L.); (J.B.); (W.Z.); (J.-M.D.)
| | | | - Jérémy Berthelet
- Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France; (R.L.); (J.B.); (W.Z.); (J.-M.D.)
- Centre Epigénétique et Destin Cellulaire, Université Paris Cité, CNRS, F-75013 Paris, France
| | - Wenchao Zhang
- Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France; (R.L.); (J.B.); (W.Z.); (J.-M.D.)
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jean-Marie Dupret
- Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France; (R.L.); (J.B.); (W.Z.); (J.-M.D.)
| | - Fernando Rodrigues Lima
- Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France; (R.L.); (J.B.); (W.Z.); (J.-M.D.)
- Correspondence:
| |
Collapse
|
28
|
Wang H, Perera L, Jork N, Zong G, Riley AM, Potter BVL, Jessen HJ, Shears SB. A structural exposé of noncanonical molecular reactivity within the protein tyrosine phosphatase WPD loop. Nat Commun 2022; 13:2231. [PMID: 35468885 PMCID: PMC9038691 DOI: 10.1038/s41467-022-29673-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 03/25/2022] [Indexed: 01/06/2023] Open
Abstract
Structural snapshots of protein/ligand complexes are a prerequisite for gaining atomic level insight into enzymatic reaction mechanisms. An important group of enzymes has been deprived of this analytical privilege: members of the protein tyrosine phosphatase (PTP) superfamily with catalytic WPD-loops lacking the indispensable general-acid/base within a tryptophan-proline-aspartate/glutamate context. Here, we provide the ligand/enzyme crystal complexes for one such PTP outlier: Arabidopsis thaliana Plant and Fungi Atypical Dual Specificity Phosphatase 1 (AtPFA-DSP1), herein unveiled as a regioselective and efficient phosphatase towards inositol pyrophosphate (PP-InsP) signaling molecules. Although the WPD loop is missing its canonical tripeptide motif, this structural element contributes to catalysis by assisting PP-InsP delivery into the catalytic pocket, for a choreographed exchange with phosphate reaction product. Subsequently, an intramolecular proton donation by PP-InsP substrate is posited to substitute functionally for the absent aspartate/glutamate general-acid. Overall, we expand mechanistic insight into adaptability of the conserved PTP structural elements.
Collapse
Affiliation(s)
- Huanchen Wang
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA.
| | - Lalith Perera
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Nikolaus Jork
- Institute of Organic Chemistry, and CIBSS - the Center for Integrative Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Guangning Zong
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Andrew M Riley
- Drug Discovery and Medicinal Chemistry, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Barry V L Potter
- Drug Discovery and Medicinal Chemistry, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Henning J Jessen
- Institute of Organic Chemistry, and CIBSS - the Center for Integrative Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Stephen B Shears
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
29
|
Pessoa JC, Santos MF, Correia I, Sanna D, Sciortino G, Garribba E. Binding of vanadium ions and complexes to proteins and enzymes in aqueous solution. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214192] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
30
|
Phong NV, Oanh VT, Yang SY, Choi JS, Min BS, Kim JA. PTP1B inhibition studies of biological active phloroglucinols from the rhizomes of Dryopteris crassirhizoma: Kinetic properties and molecular docking simulation. Int J Biol Macromol 2021; 188:719-728. [PMID: 34416263 DOI: 10.1016/j.ijbiomac.2021.08.091] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 10/20/2022]
Abstract
By various chromatographic methods, 30 phloroglucinols (1-30) were isolated from a methanol extract of Dryopteris crassirhizoma, including two new dimeric phloroglucinols (13 and 25). The structures of the isolates were confirmed by HR-MS, 1D, and 2D NMR as well as by comparison with the literature. The protein tyrosine phosphatase 1B (PTP1B) effects of the isolated compounds (1-30) were evaluated using sodium orthovanadate and ursolic acid as a positive control. Among them, trimeric phloroglucinols 26-28 significantly exhibited the PTP1B inhibitory effects with the IC50 values of 1.19 ± 0.13, 1.00 ± 0.04, 1.23 ± 0.05 μM, respectively. In addition, the kinetic analysis revealed compounds 26-28 acted as competitive inhibitors against PTP1B enzyme with Ki values of 0.63, 0.61, 1.57 μM, respectively. Molecular docking simulations were performed to demonstrate that these active compounds can bind with the catalytic sites of PTP1B with negative binding energies and the results are in accordance with that of the kinetic studies. In vitro and in silico results suggest that D. crassirhizoma rhizomes together with compounds 26-28 are potential candidates for treating type 2 diabetes.
Collapse
Affiliation(s)
- Nguyen Viet Phong
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea; Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Vu Thi Oanh
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea; Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Seo Young Yang
- Department of Pharmaceutical Engineering, Sangji University, Wonju 26339, Republic of Korea
| | - Jae Sue Choi
- Department of Food and Life Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Byung Sun Min
- College of Pharmacy, Drug Research and Development Center, Catholic University of Daegu, Gyeongbuk, Republic of Korea
| | - Jeong Ah Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea; Vessel-Organ Interaction Research Center, VOICE (MRC), College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
31
|
Pinkston J, Jo J, Olsen KJ, Comer D, Glaittli CA, Loria JP, Johnson SJ, Hengge AC. Significant Loop Motions in the SsoPTP Protein Tyrosine Phosphatase Allow for Dual General Acid Functionality. Biochemistry 2021; 60:2888-2901. [PMID: 34496202 DOI: 10.1021/acs.biochem.1c00365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Conformational dynamics are important factors in the function of enzymes, including protein tyrosine phosphatases (PTPs). Crystal structures of PTPs first revealed the motion of a protein loop bearing a conserved catalytic aspartic acid, and subsequent nuclear magnetic resonance and computational analyses have shown the presence of motions, involved in catalysis and allostery, within and beyond the active site. The tyrosine phosphatase from the thermophilic and acidophilic Sulfolobus solfataricus (SsoPTP) displays motions of its acid loop together with dynamics of its phosphoryl-binding P-loop and the Q-loop, the first instance of such motions in a PTP. All three loops share the same exchange rate, implying their motions are coupled. Further evidence of conformational flexibility comes from mutagenesis, kinetics, and isotope effect data showing that E40 can function as an alternate general acid to protonate the leaving group when the conserved acid, D69, is mutated to asparagine. SsoPTP is not the first PTP to exhibit an alternate general acid (after VHZ and TkPTP), but E40 does not correspond to the sequence or structural location of the alternate general acids in those precedents. A high-resolution X-ray structure with the transition state analogue vanadate clarifies the role of the active site arginine R102, which varied in structures of substrates bound to a catalytically inactive mutant. The coordinated motions of all three functional loops in SsoPTP, together with the function of an alternate general acid, suggest that catalytically competent conformations are present in solution that have not yet been observed in crystal structures.
Collapse
Affiliation(s)
- Justin Pinkston
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300, United States
| | - Jihye Jo
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520, United States
| | - Keith J Olsen
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300, United States
| | - Drake Comer
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300, United States
| | - Charsti A Glaittli
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300, United States
| | - J Patrick Loria
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520, United States.,Department of Molecular Biophysics and Biochemistry, Yale University, 266 Whitney Avenue, New Haven, Connecticut 06520, United States
| | - Sean J Johnson
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300, United States
| | - Alvan C Hengge
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322-0300, United States
| |
Collapse
|
32
|
García‐Marín J, Griera M, Alajarín R, Rodríguez‐Puyol M, Rodríguez‐Puyol D, Vaquero JJ. A Computer-Driven Scaffold-Hopping Approach Generating New PTP1B Inhibitors from the Pyrrolo[1,2-a]quinoxaline Core. ChemMedChem 2021; 16:2895-2906. [PMID: 34137509 PMCID: PMC8518816 DOI: 10.1002/cmdc.202100338] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/13/2021] [Indexed: 11/06/2022]
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is a very promising target for the treatment of metabolic disorders such as type II diabetes mellitus. Although it was validated as a promising target for this disease more than 30 years ago, as yet there is no drug in advanced clinical trials, and its biochemical mechanism and functions are still being studied. In the present study, based on our experience generating PTP1B inhibitors, we have developed and implemented a scaffold-hopping approach to vary the pyrrole ring of the pyrrolo[1,2-a]quinoxaline core, supported by extensive computational techniques aimed to explain the molecular interaction with PTP1B. Using a combination of docking, molecular dynamics and end-point free-energy calculations, we have rationally designed a hypothesis for new PTP1B inhibitors, supporting their recognition mechanism at a molecular level. After the design phase, we were able to easily synthesize proposed candidates and their evaluation against PTP1B was found to be in good concordance with our predictions. Moreover, the best candidates exhibited glucose uptake increments in cellulo model, thus confirming their utility for PTP1B inhibition and validating this approach for inhibitors design and molecules thus obtained.
Collapse
Affiliation(s)
- Javier García‐Marín
- Departamento de Química Orgánica y Química InorgánicaUniversidad de Alcalá28805Alcalá de HenaresSpain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)Ctra. Colmenar Viejo, km. 910028034MadridSpain
- Instituto de Investigación Química Andrés Manuel del Río (IQAR)Universidad de AlcaláAlcalá de HenaresSpain
- Departamento de Química Biológica y EstructuralCentro de Investigaciones Biológicas Margarita Salas (CIB-CSIC)Calle Ramiro de Maeztu 928040MadridSpain
| | - Mercedes Griera
- Graphenano Medical Care, S.L.C/Pablo Casals, no. 13YeclaMurciaSpain
- Departamento de Biología de SistemasUniversidad de Alcalá28805Alcalá de HenaresSpain
| | - Ramón Alajarín
- Departamento de Química Orgánica y Química InorgánicaUniversidad de Alcalá28805Alcalá de HenaresSpain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)Ctra. Colmenar Viejo, km. 910028034MadridSpain
- Instituto de Investigación Química Andrés Manuel del Río (IQAR)Universidad de AlcaláAlcalá de HenaresSpain
| | - Manuel Rodríguez‐Puyol
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)Ctra. Colmenar Viejo, km. 910028034MadridSpain
- Departamento de Biología de SistemasUniversidad de Alcalá28805Alcalá de HenaresSpain
| | - Diego Rodríguez‐Puyol
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)Ctra. Colmenar Viejo, km. 910028034MadridSpain
- Fundación de Investigación BiomédicaUnidad de Nefrología del Hospital Príncipe de Asturias yDepartamento de Medicina y Especialidades MédicasUniversidad de Alcalá28805Alcalá de HenaresSpain
| | - Juan J. Vaquero
- Departamento de Química Orgánica y Química InorgánicaUniversidad de Alcalá28805Alcalá de HenaresSpain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)Ctra. Colmenar Viejo, km. 910028034MadridSpain
- Instituto de Investigación Química Andrés Manuel del Río (IQAR)Universidad de AlcaláAlcalá de HenaresSpain
| |
Collapse
|
33
|
Untargeted Metabolomics Approach for the Discovery of Environment-Related Pyran-2-ones Chemodiversity in a Marine-Sourced Penicillium restrictum. Mar Drugs 2021; 19:md19070378. [PMID: 34210084 PMCID: PMC8305465 DOI: 10.3390/md19070378] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 11/17/2022] Open
Abstract
Very little is known about chemical interactions between fungi and their mollusc host within marine environments. Here, we investigated the metabolome of a Penicillium restrictum MMS417 strain isolated from the blue mussel Mytilus edulis collected on the Loire estuary, France. Following the OSMAC approach with the use of 14 culture media, the effect of salinity and of a mussel-derived medium on the metabolic expression were analysed using HPLC-UV/DAD-HRMS/MS. An untargeted metabolomics study was performed using principal component analysis (PCA), orthogonal projection to latent structure discriminant analysis (O-PLSDA) and molecular networking (MN). It highlighted some compounds belonging to sterols, macrolides and pyran-2-ones, which were specifically induced in marine conditions. In particular, a high chemical diversity of pyran-2-ones was found to be related to the presence of mussel extract in the culture medium. Mass spectrometry (MS)- and UV-guided purification resulted in the isolation of five new natural fungal pyran-2-one derivatives—5,6-dihydro-6S-hydroxymethyl-4-methoxy-2H-pyran-2-one (1), (6S, 1’R, 2’S)-LL-P880β (3), 5,6-dihydro-4-methoxy-6S-(1’S, 2’S-dihydroxy pent-3’(E)-enyl)-2H-pyran-2-one (4), 4-methoxy-6-(1’R, 2’S-dihydroxy pent-3’(E)-enyl)-2H-pyran-2-one (6) and 4-methoxy-2H-pyran-2-one (7)—together with the known (6S, 1’S, 2’S)-LL-P880β (2), (1’R, 2’S)-LL-P880γ (5), 5,6-dihydro-4-methoxy-2H-pyran-2-one (8), (6S, 1’S, 2’R)-LL-P880β (9), (6S, 1’S)-pestalotin (10), 1’R-dehydropestalotin (11) and 6-pentyl-4-methoxy-2H-pyran-2-one (12) from the mussel-derived culture medium extract. The structures of 1-12 were determined by 1D- and 2D-MMR experiments as well as high-resolution tandem MS, ECD and DP4 calculations. Some of these compounds were evaluated for their cytotoxic, antibacterial, antileishmanial and in-silico PTP1B inhibitory activities. These results illustrate the utility in using host-derived media for the discovery of new natural products.
Collapse
|
34
|
Shen R, Crean RM, Johnson SJ, Kamerlin SCL, Hengge AC. Single Residue on the WPD-Loop Affects the pH Dependency of Catalysis in Protein Tyrosine Phosphatases. JACS AU 2021; 1:646-659. [PMID: 34308419 PMCID: PMC8297725 DOI: 10.1021/jacsau.1c00054] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Indexed: 05/08/2023]
Abstract
Catalysis by protein tyrosine phosphatases (PTPs) relies on the motion of a flexible protein loop (the WPD-loop) that carries a residue acting as a general acid/base catalyst during the PTP-catalyzed reaction. The orthogonal substitutions of a noncatalytic residue in the WPD-loops of YopH and PTP1B result in shifted pH-rate profiles from an altered kinetic pK a of the nucleophilic cysteine. Compared to wild type, the G352T YopH variant has a broadened pH-rate profile, similar activity at optimal pH, but significantly higher activity at low pH. Changes in the corresponding PTP1B T177G variant are more modest and in the opposite direction, with a narrowed pH profile and less activity in the most acidic range. Crystal structures of the variants show no structural perturbations but suggest an increased preference for the WPD-loop-closed conformation. Computational analysis confirms a shift in loop conformational equilibrium in favor of the closed conformation, arising from a combination of increased stability of the closed state and destabilization of the loop-open state. Simulations identify the origins of this population shift, revealing differences in the flexibility of the WPD-loop and neighboring regions. Our results demonstrate that changes to the pH dependency of catalysis by PTPs can result from small changes in amino acid composition in their WPD-loops affecting only loop dynamics and conformational equilibrium. The perturbation of kinetic pK a values of catalytic residues by nonchemical processes affords a means for nature to alter an enzyme's pH dependency by a less disruptive path than altering electrostatic networks around catalytic residues themselves.
Collapse
Affiliation(s)
- Ruidan Shen
- Department
of Chemistry and Biochemistry, Utah State
University, Logan, Utah 84322-0300, United States
| | - Rory M. Crean
- Science
for Life Laboratory, Department of Chemistry − BMC, Uppsala University, Box 576, S-751 23 Uppsala, Sweden
| | - Sean J. Johnson
- Department
of Chemistry and Biochemistry, Utah State
University, Logan, Utah 84322-0300, United States
| | - Shina C. L. Kamerlin
- Science
for Life Laboratory, Department of Chemistry − BMC, Uppsala University, Box 576, S-751 23 Uppsala, Sweden
| | - Alvan C. Hengge
- Department
of Chemistry and Biochemistry, Utah State
University, Logan, Utah 84322-0300, United States
| |
Collapse
|
35
|
Crean RM, Biler M, van der Kamp MW, Hengge AC, Kamerlin SCL. Loop Dynamics and Enzyme Catalysis in Protein Tyrosine Phosphatases. J Am Chem Soc 2021; 143:3830-3845. [PMID: 33661624 PMCID: PMC8031367 DOI: 10.1021/jacs.0c11806] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Indexed: 12/16/2022]
Abstract
Protein tyrosine phosphatases (PTPs) play an important role in cellular signaling and have been implicated in human cancers, diabetes, and obesity. Despite shared catalytic mechanisms and transition states for the chemical steps of catalysis, catalytic rates within the PTP family vary over several orders of magnitude. These rate differences have been implied to arise from differing conformational dynamics of the closure of a protein loop, the WPD-loop, which carries a catalytically critical residue. The present work reports computational studies of the human protein tyrosine phosphatase 1B (PTP1B) and YopH from Yersinia pestis, for which NMR has demonstrated a link between their respective rates of WPD-loop motion and catalysis rates, which differ by an order of magnitude. We have performed detailed structural analysis, both conventional and enhanced sampling simulations of their loop dynamics, as well as empirical valence bond simulations of the chemical step of catalysis. These analyses revealed the key residues and structural features responsible for these differences, as well as the residues and pathways that facilitate allosteric communication in these enzymes. Curiously, our wild-type YopH simulations also identify a catalytically incompetent hyper-open conformation of its WPD-loop, sampled as a rare event, previously only experimentally observed in YopH-based chimeras. The effect of differences within the WPD-loop and its neighboring loops on the modulation of loop dynamics, as revealed in this work, may provide a facile means for the family of PTP enzymes to respond to environmental changes and regulate their catalytic activities.
Collapse
Affiliation(s)
- Rory M. Crean
- Science
for Life Laboratory, Department of Chemistry − BMC, Uppsala University, Box 576, S-751 23 Uppsala, Sweden
| | - Michal Biler
- Science
for Life Laboratory, Department of Chemistry − BMC, Uppsala University, Box 576, S-751 23 Uppsala, Sweden
| | - Marc W. van der Kamp
- School
of Biochemistry, University of Bristol, Biomedical Sciences Building, University
Walk, Bristol BS8 1TD, United Kingdom
| | - Alvan C. Hengge
- Department
of Chemistry and Biochemistry, Utah State
University, Logan, Utah 84322-0300, United States
| | - Shina C. L. Kamerlin
- Science
for Life Laboratory, Department of Chemistry − BMC, Uppsala University, Box 576, S-751 23 Uppsala, Sweden
| |
Collapse
|
36
|
Tao Y, Xie J, Zhong Q, Wang Y, Zhang S, Luo F, Wen F, Xie J, Zhao J, Sun X, Long H, Ma J, Zhang Q, Long J, Fang X, Lu Y, Li D, Li M, Zhu J, Sun B, Li G, Diao J, Liu C. A novel partially open state of SHP2 points to a "multiple gear" regulation mechanism. J Biol Chem 2021; 296:100538. [PMID: 33722610 PMCID: PMC8054191 DOI: 10.1016/j.jbc.2021.100538] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 03/03/2021] [Accepted: 03/10/2021] [Indexed: 11/14/2022] Open
Abstract
The protein tyrosine phosphatase SHP2 mediates multiple signal transductions in various cellular pathways, controlled by a variety of upstream inputs. SHP2 dysregulation is causative of different types of cancers and developmental disorders, making it a promising drug target. However, how SHP2 is modulated by its different regulators remains largely unknown. Here, we use single-molecule fluorescence resonance energy transfer and molecular dynamics simulations to investigate this question. We identify a partially open, semiactive conformation of SHP2 that is intermediate between the known open and closed states. We further demonstrate a “multiple gear” regulatory mechanism, in which different activators (e.g., insulin receptor substrate-1 and CagA), oncogenic mutations (e.g., E76A), and allosteric inhibitors (e.g., SHP099) can shift the equilibrium of the three conformational states and regulate SHP2 activity to different levels. Our work reveals the essential role of the intermediate state in fine-tuning the activity of SHP2, which may provide new opportunities for drug development for relevant cancers.
Collapse
Affiliation(s)
- Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Jingfei Xie
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Qinglu Zhong
- University of the Chinese Academy of Sciences, Beijing, China; Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yongyao Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Shengnan Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Feng Luo
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Fengcai Wen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jingjing Xie
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Jiawei Zhao
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiaoou Sun
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Houfang Long
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Junfeng Ma
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qian Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jiangang Long
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xianyang Fang
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ying Lu
- University of the Chinese Academy of Sciences, Beijing, China; Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Li
- University of the Chinese Academy of Sciences, Beijing, China; Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Jidong Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Bo Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Guohui Li
- University of the Chinese Academy of Sciences, Beijing, China; Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
37
|
Sciortino G, Maréchal JD, Garribba E. Integrated experimental/computational approaches to characterize the systems formed by vanadium with proteins and enzymes. Inorg Chem Front 2021. [DOI: 10.1039/d0qi01507e] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
An integrated instrumental/computational approach to characterize metallodrug–protein adducts at the molecular level is reviewed. A series of applications are described, focusing on potential vanadium drugs with a generalization to other metals.
Collapse
Affiliation(s)
- Giuseppe Sciortino
- Departament de Química
- Universitat Autònoma de Barcelona
- Cerdanyola del Vallès
- Barcelona 08193
- Spain
| | - Jean-Didier Maréchal
- Departament de Química
- Universitat Autònoma de Barcelona
- Cerdanyola del Vallès
- Barcelona 08193
- Spain
| | - Eugenio Garribba
- Dipartimento di Chimica e Farmacia
- Università di Sassari
- 07100 Sassari
- Italy
| |
Collapse
|
38
|
Yang R, Dong Q, Xu H, Gao X, Zhao Z, Qin J, Chen C, Luo D. Identification of Phomoxanthone A and B as Protein Tyrosine Phosphatase Inhibitors. ACS OMEGA 2020; 5:25927-25935. [PMID: 33073119 PMCID: PMC7557999 DOI: 10.1021/acsomega.0c03315] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 08/04/2020] [Indexed: 05/08/2023]
Abstract
Phomoxanthone A and B (PXA and PXB) are xanthone dimers and isolated from the endophytic fungus Phomopsis sp. By254. The results demonstrated that PXB and PXA are noncompetitive inhibitors of SHP2 and PTP1B and competitive inhibitors of SHP1. Molecular docking studies showed that PXB and PXA interact with conserved domains of protein tyrosine phosphatases such as the β5-β6 loop, WPD loop, P loop, and Q loop. PXA and PXB could significantly inhibit the cell proliferation in MCF7 cells. Our results indicated that these two compounds do not efficiently inhibit PTP1B and SHP2 activity. RNA sequencing showed that PXA and PXB may inhibit SHP1 activity in MCF7 cells leading to the upregulation of inflammatory factors. In addition to PTP inhibition, PXA and PXB are multitarget compounds to inhibit the proliferation of tumor cells. In conclusion, both compounds show inhibition of cancer cells and a certain degree of inflammatory stimulation, which make them promising for tumor immunotherapy.
Collapse
Affiliation(s)
- Runlei Yang
- College
of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei 071002, China
| | - Qian Dong
- College
of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei 071002, China
| | - Huibin Xu
- College
of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei 071002, China
| | - XueHui Gao
- College
of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei 071002, China
| | - Ziyue Zhao
- College
of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei 071002, China
| | - Jianchun Qin
- College
of Plant Science, Jilin University, Changchun, Jilin 130062, China
| | - Chuan Chen
- College
of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei 071002, China
| | - Duqiang Luo
- College
of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, Hebei 071002, China
| |
Collapse
|
39
|
Structural Insights into the Active Site Formation of DUSP22 in N-loop-containing Protein Tyrosine Phosphatases. Int J Mol Sci 2020; 21:ijms21207515. [PMID: 33053837 PMCID: PMC7589817 DOI: 10.3390/ijms21207515] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Cysteine-based protein tyrosine phosphatases (Cys-based PTPs) perform dephosphorylation to regulate signaling pathways in cellular responses. The hydrogen bonding network in their active site plays an important conformational role and supports the phosphatase activity. Nearly half of dual-specificity phosphatases (DUSPs) use three conserved residues, including aspartate in the D-loop, serine in the P-loop, and asparagine in the N-loop, to form the hydrogen bonding network, the D-, P-, N-triloop interaction (DPN-triloop interaction). In this study, DUSP22 is used to investigate the importance of the DPN-triloop interaction in active site formation. Alanine mutations and somatic mutations of the conserved residues, D57, S93, and N128 substantially decrease catalytic efficiency (kcat/KM) by more than 102-fold. Structural studies by NMR and crystallography reveal that each residue can perturb the three loops and induce conformational changes, indicating that the hydrogen bonding network aligns the residues in the correct positions for substrate interaction and catalysis. Studying the DPN-triloop interaction reveals the mechanism maintaining phosphatase activity in N-loop-containing PTPs and provides a foundation for further investigation of active site formation in different members of this protein class.
Collapse
|
40
|
Irving E, Tagalakis AD, Maeshima R, Hart SL, Eaton S, Lehtonen A, Stoker AW. The liposomal delivery of hydrophobic oxidovanadium complexes imparts highly effective cytotoxicity and differentiating capacity in neuroblastoma tumour cells. Sci Rep 2020; 10:16660. [PMID: 33028860 PMCID: PMC7542164 DOI: 10.1038/s41598-020-73539-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 09/15/2020] [Indexed: 12/31/2022] Open
Abstract
Oxidovanadium complexes with organic ligands are well known to have cytotoxic or differentiating capabilities against a range of cancer cell types. Their limited use in clinical testing though has resulted largely from uncertainties about the long-term toxicities of such complexes, due in part to the speciation to vanadate ions in the circulation. We hypothesised that more highly stable complexes, delivered using liposomes, may provide improved opportunities for oxidovanadium applications against cancer. In this study we sourced specifically hydrophobic forms of oxidovanadium complexes with the explicit aim of demonstrating liposomal encapsulation, bioavailability in cultured neuroblastoma cells, and effective cytotoxic or differentiating activity. Our data show that four ethanol-solubilised complexes with amine bisphenol, aminoalcohol bisphenol or salan ligands are equally or more effective than a previously used complex bis(maltolato)oxovanadium(V) in neuroblastoma cell lines. Moreover, we show that one of these complexes can be stably incorporated into cationic liposomes where it retains very good bioavailability, apparently low speciation and enhanced efficacy compared to ethanol delivery. This study provides the first proof-of-concept that stable, hydrophobic oxidovanadium complexes retain excellent cellular activity when delivered effectively to cancer cells with nanotechnology. This offers the improved prospect of applying oxidovanadium-based drugs in vivo with increased stability and reduced off-target toxicity.
Collapse
Affiliation(s)
- Elsa Irving
- Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Aristides D Tagalakis
- Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- Department of Biology, Edge Hill University, Ormskirk, L39 4QP, UK
| | - Ruhina Maeshima
- Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Stephen L Hart
- Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Simon Eaton
- Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Ari Lehtonen
- Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- Department of Chemistry, University of Turku, 20014, Turun yliopisto, Finland
| | - Andrew W Stoker
- Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
41
|
Benjamín-Rivera JA, Cardona-Rivera AE, Vázquez-Maldonado ÁL, Dones-Lassalle CY, Pabón-Colon HL, Rodríguez-Rivera HM, Rodríguez I, González-Espiet JC, Pazol J, Pérez-Ríos JD, Catala-Torres JF, Carrasquillo Rivera M, De Jesus-Soto MG, Cordero-Virella NA, Cruz-Maldonado PM, González-Pagan P, Hernández-Ríos R, Gaur K, Loza-Rosas SA, Tinoco AD. Exploring Serum Transferrin Regulation of Nonferric Metal Therapeutic Function and Toxicity. INORGANICS 2020; 8:48. [PMID: 36844373 PMCID: PMC9957567 DOI: 10.3390/inorganics8090048] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Serum transferrin (sTf) plays a pivotal role in regulating iron biodistribution and homeostasis within the body. The molecular details of sTf Fe(III) binding blood transport, and cellular delivery through transferrin receptor-mediated endocytosis are generally well-understood. Emerging interest exists in exploring sTf complexation of nonferric metals as it facilitates the therapeutic potential and toxicity of several of them. This review explores recent X-ray structural and physiologically relevant metal speciation studies to understand how sTf partakes in the bioactivity of key non-redox active hard Lewis acidic metals. It challenges preconceived notions of sTf structure function correlations that were based exclusively on the Fe(III) model by revealing distinct coordination modalities that nonferric metal ions can adopt and different modes of binding to metal-free and Fe(III)-bound sTf that can directly influence how they enter into cells and, ultimately, how they may impact human health. This knowledge informs on biomedical strategies to engineer sTf as a delivery vehicle for metal-based diagnostic and therapeutic agents in the cancer field. It is the intention of this work to open new avenues for characterizing the functionality and medical utility of nonferric-bound sTf and to expand the significance of this protein in the context of bioinorganic chemistry.
Collapse
Affiliation(s)
- Josué A. Benjamín-Rivera
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Andrés E. Cardona-Rivera
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | | | | | - Héctor L. Pabón-Colon
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | | | - Israel Rodríguez
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Jean C. González-Espiet
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Jessika Pazol
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Jobaniel D. Pérez-Ríos
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - José F. Catala-Torres
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | | | - Michael G. De Jesus-Soto
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | | | - Paola M. Cruz-Maldonado
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Patricia González-Pagan
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Raul Hernández-Ríos
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Kavita Gaur
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
| | - Sergio A. Loza-Rosas
- Departamento de Química y Bioquímica, Facultad de Ciencias e Ingeniería, Universidad de Boyacá, Tunja 150003, Colombia
| | - Arthur D. Tinoco
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA)
- Correspondence: ; Tel.: +1-939-319-9701
| |
Collapse
|
42
|
Thingholm TE, Rönnstrand L, Rosenberg PA. Why and how to investigate the role of protein phosphorylation in ZIP and ZnT zinc transporter activity and regulation. Cell Mol Life Sci 2020; 77:3085-3102. [PMID: 32076742 PMCID: PMC7391401 DOI: 10.1007/s00018-020-03473-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 01/13/2020] [Accepted: 01/28/2020] [Indexed: 12/20/2022]
Abstract
Zinc is required for the regulation of proliferation, metabolism, and cell signaling. It is an intracellular second messenger, and the cellular level of ionic, mobile zinc is strictly controlled by zinc transporters. In mammals, zinc homeostasis is primarily regulated by ZIP and ZnT zinc transporters. The importance of these transporters is underscored by the list of diseases resulting from changes in transporter expression and activity. However, despite numerous structural studies of the transporters revealing both zinc binding sites and motifs important for transporter function, the exact molecular mechanisms regulating ZIP and ZnT activities are still not clear. For example, protein phosphorylation was found to regulate ZIP7 activity resulting in the release of Zn2+ from intracellular stores leading to phosphorylation of tyrosine kinases and activation of signaling pathways. In addition, sequence analyses predict all 24 human zinc transporters to be phosphorylated suggesting that protein phosphorylation is important for regulation of transporter function. This review describes how zinc transporters are implicated in a number of important human diseases. It summarizes the current knowledge regarding ZIP and ZnT transporter structures and points to how protein phosphorylation seems to be important for the regulation of zinc transporter activity. The review addresses the need to investigate the role of protein phosphorylation in zinc transporter function and regulation, and argues for a pressing need to introduce quantitative phosphoproteomics to specifically target zinc transporters and proteins involved in zinc signaling. Finally, different quantitative phosphoproteomic strategies are suggested.
Collapse
Affiliation(s)
- T E Thingholm
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, J.B. Winsløws Vej 25, 3, 5000, Odense C, Denmark.
| | - L Rönnstrand
- Division of Translational Cancer Research, Lund University, Medicon Village, Building 404, Scheelevägen 2, Lund, Sweden
- Lund Stem Cell Center, Lund University, Medicon Village, Building 404, Scheelevägen 2, Lund, Sweden
- Division of Oncology, Skåne University Hospital, Lund, Sweden
| | - P A Rosenberg
- Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
- Department of Neurology and Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
43
|
Samart N, Althumairy D, Zhang D, Roess DA, Crans DC. Initiation of a novel mode of membrane signaling: Vanadium facilitated signal transduction. Coord Chem Rev 2020. [DOI: 10.1016/j.ccr.2020.213286] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
44
|
Sharma B, Xie L, Yang F, Wang W, Zhou Q, Xiang M, Zhou S, Lv W, Jia Y, Pokhrel L, Shen J, Xiao Q, Gao L, Deng W. Recent advance on PTP1B inhibitors and their biomedical applications. Eur J Med Chem 2020; 199:112376. [PMID: 32416458 DOI: 10.1016/j.ejmech.2020.112376] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 12/17/2022]
Abstract
Protein Tyrosine Phosphatase 1B (PTP1B), as one of the most important members in PTP superfamily, plays a vital role in conducting various cellular functions. So far, PTP1B has been reported to be involved in the development of many diseases including obesity, diabetes, cancers and cardiovascular diseases. Development of potent and specific PTP1B inhibitors and studies on the structure-activity relationship (SAR) between their chemical structures and their biological activity have drawn increasing attention as they could not only modulate the PTP1B functions inside the cells but also provide useful lead compounds for the treatment of various PTP1B-associated diseases. To this end, we herein summarized the recent developments of PTP1B inhibitors, and different kinds of high-throughput screening strategies for the identification of potential PTP1B inhibitors as well as their potential biomedical applications, and we also provided some perspectives in the concluding remarks in this work.
Collapse
Affiliation(s)
- Bigyan Sharma
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, PR China
| | - Liuxing Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, PR China
| | - Fen Yang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, PR China
| | - Wei Wang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, PR China
| | - Quanming Zhou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, PR China
| | - Menghua Xiang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, PR China
| | - Shizhe Zhou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, PR China
| | - Wanting Lv
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, PR China
| | - Yan Jia
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, PR China
| | - Laxman Pokhrel
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Jun Shen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Qicai Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, PR China.
| | - Liqian Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, PR China.
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, PR China.
| |
Collapse
|
45
|
Bheri M, Mahiwal S, Sanyal SK, Pandey GK. Plant protein phosphatases: What do we know about their mechanism of action? FEBS J 2020; 288:756-785. [PMID: 32542989 DOI: 10.1111/febs.15454] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/27/2020] [Accepted: 06/09/2020] [Indexed: 12/30/2022]
Abstract
Protein phosphorylation is a major reversible post-translational modification. Protein phosphatases function as 'critical regulators' in signaling networks through dephosphorylation of proteins, which have been phosphorylated by protein kinases. A large understanding of their working has been sourced from animal systems rather than the plant or the prokaryotic systems. The eukaryotic protein phosphatases include phosphoprotein phosphatases (PPP), metallo-dependent protein phosphatases (PPM), protein tyrosine (Tyr) phosphatases (PTP), and aspartate (Asp)-dependent phosphatases. The PPP and PPM families are serine(Ser)/threonine(Thr)-specific phosphatases (STPs), while PTP family is Tyr specific. Dual-specificity phosphatases (DsPTPs/DSPs) dephosphorylate Ser, Thr, and Tyr residues. PTPs lack sequence homology with STPs, indicating a difference in catalytic mechanisms, while the PPP and PPM families share a similar structural fold indicating a common catalytic mechanism. The catalytic cysteine (Cys) residue in the conserved HCX5 R active site motif of the PTPs acts as a nucleophile during hydrolysis. The PPP members require metal ions, which coordinate the phosphate group of the substrate, followed by a nucleophilic attack by a water molecule and hydrolysis. The variable holoenzyme assembly of protein phosphatase(s) and the overlap with other post-translational modifications like acetylation and ubiquitination add to their complexity. Though their functional characterization is extensively reported in plants, the mechanistic nature of their action is still being explored by researchers. In this review, we exclusively overview the plant protein phosphatases with an emphasis on their mechanistic action as well as structural characteristics.
Collapse
Affiliation(s)
- Malathi Bheri
- Department of Plant Molecular Biology, University of Delhi South Campus, New Delhi, India
| | - Swati Mahiwal
- Department of Plant Molecular Biology, University of Delhi South Campus, New Delhi, India
| | - Sibaji K Sanyal
- Department of Plant Molecular Biology, University of Delhi South Campus, New Delhi, India
| | - Girdhar K Pandey
- Department of Plant Molecular Biology, University of Delhi South Campus, New Delhi, India
| |
Collapse
|
46
|
Treviño S, Diaz A. Vanadium and insulin: Partners in metabolic regulation. J Inorg Biochem 2020; 208:111094. [PMID: 32438270 DOI: 10.1016/j.jinorgbio.2020.111094] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022]
Abstract
Since the 1970s, the biological role of vanadium compounds has been discussed as insulin-mimetic or insulin-enhancer agents. The action of vanadium compounds has been investigated to determine how they influence the insulin signaling pathway. Khan and coworkers proposed key proteins for the insulin pathway study, introducing the concept "critical nodes". In this review, we also considered critical kinases and phosphatases that participate in this pathway, which will permit a better comprehension of a critical node, where vanadium can act: a) insulin receptor, insulin receptor substrates, and protein tyrosine phosphatases; b) phosphatidylinositol 3'-kinase, 3-phosphoinositide-dependent protein kinase and mammalian target of rapamycin complex, protein kinase B, and phosphatase and tensin homolog; and c) insulin receptor substrates and mitogen-activated protein kinases, each node having specific negative modulators. Additionally, leptin signaling was considered because together with insulin, it modulates glucose and lipid homeostasis. Even in recent literature, the possibility of vanadium acting against metabolic diseases or cancer is confirmed although the mechanisms of action are not well understood because these critical nodes have not been systematically investigated. Through this review, we establish that vanadium compounds mainly act as phosphatase inhibitors and hypothesize on their capacity to affect kinases, which are critical to other hormones that also act on common parts of the insulin pathway.
Collapse
Affiliation(s)
- Samuel Treviño
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, University Autonomous of Puebla, 14 South. FCQ1, University City, Puebla, C.P. 72560, Mexico.
| | - Alfonso Diaz
- Department of Pharmacy, Faculty of Chemistry Science, University Autonomous of Puebla, 22 South, FCQ9, University City, Puebla, C.P. 72560, Mexico.
| |
Collapse
|
47
|
Crean RM, Gardner JM, Kamerlin SCL. Harnessing Conformational Plasticity to Generate Designer Enzymes. J Am Chem Soc 2020; 142:11324-11342. [PMID: 32496764 PMCID: PMC7467679 DOI: 10.1021/jacs.0c04924] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Indexed: 02/08/2023]
Abstract
Recent years have witnessed an explosion of interest in understanding the role of conformational dynamics both in the evolution of new enzymatic activities from existing enzymes and in facilitating the emergence of enzymatic activity de novo on scaffolds that were previously non-catalytic. There are also an increasing number of examples in the literature of targeted engineering of conformational dynamics being successfully used to alter enzyme selectivity and activity. Despite the obvious importance of conformational dynamics to both enzyme function and evolvability, many (although not all) computational design approaches still focus either on pure sequence-based approaches or on using structures with limited flexibility to guide the design. However, there exist a wide variety of computational approaches that can be (re)purposed to introduce conformational dynamics as a key consideration in the design process. Coupled with laboratory evolution and more conventional existing sequence- and structure-based approaches, these techniques provide powerful tools for greatly expanding the protein engineering toolkit. This Perspective provides an overview of evolutionary studies that have dissected the role of conformational dynamics in facilitating the emergence of novel enzymes, as well as advances in computational approaches that allow one to target conformational dynamics as part of enzyme design. Harnessing conformational dynamics in engineering studies is a powerful paradigm with which to engineer the next generation of designer biocatalysts.
Collapse
Affiliation(s)
- Rory M. Crean
- Department of Chemistry -
BMC, Uppsala University, Box 576, 751 23 Uppsala, Sweden
| | - Jasmine M. Gardner
- Department of Chemistry -
BMC, Uppsala University, Box 576, 751 23 Uppsala, Sweden
| | - Shina C. L. Kamerlin
- Department of Chemistry -
BMC, Uppsala University, Box 576, 751 23 Uppsala, Sweden
| |
Collapse
|
48
|
Beaumont VA, Reiss K, Qu Z, Allen B, Batista VS, Loria JP. Allosteric Impact of the Variable Insert Loop in Vaccinia H1-Related (VHR) Phosphatase. Biochemistry 2020; 59:1896-1908. [PMID: 32348128 PMCID: PMC7364816 DOI: 10.1021/acs.biochem.0c00245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dynamics and conformational motions are important to the activity of enzymes, including protein tyrosine phosphatases. These motions often extend to regions outside the active site, called allosteric regions. In the tyrosine phosphatase Vaccinia H1-related (VHR) enzyme, we demonstrate the importance of the allosteric interaction between the variable insert region and the active-site loops in VHR. These studies include solution nuclear magnetic resonance, computation, steady-state, and rapid kinetic measurements. Overall, the data indicate concerted millisecond motions exist between the variable insert and the catalytic acid loop in wild-type (WT) VHR. The 150 ns computation studies show a flexible acid loop in WT VHR that opens during the simulation from its initial closed structure. Mutation of the variable insert residue, asparagine 74, to alanine results in a rigidification of the acid loop as observed by molecular dynamics simulations and a disruption of crucial active-site hydrogen bonds. Moreover, enzyme kinetic analysis shows a weakening of substrate affinity in the N74A mutant and a >2-fold decrease in substrate cleavage and hydrolysis rates. These data show that despite being nearly 20 Å from the active site, the variable insert region is linked to the acid loop by coupled millisecond motions, and that disruption of the communication between the variable insert and active site alters the normal catalytic function of VHR and perturbs the active-site environment.
Collapse
Affiliation(s)
- Victor A Beaumont
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520, United States
| | - Krystle Reiss
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520, United States
| | - Zexing Qu
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520, United States
- Institute for Theoretical Chemistry, Jilin University, Changchun 130023, China
| | - Brandon Allen
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520, United States
| | - Victor S Batista
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520, United States
| | - J Patrick Loria
- Department of Chemistry, Yale University, 225 Prospect Street, New Haven, Connecticut 06520, United States
- Department of Molecular Biophysics and Biochemistry, Yale University, 266 Whitney Avenue, New Haven, Connecticut 06520, United States
| |
Collapse
|
49
|
Rocha RF, Rodrigues T, Menegatti ACO, Bernardes GJL, Terenzi H. The antidiabetic drug lobeglitazone has the potential to inhibit PTP1B activity. Bioorg Chem 2020; 100:103927. [PMID: 32422389 DOI: 10.1016/j.bioorg.2020.103927] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/03/2020] [Accepted: 05/08/2020] [Indexed: 12/13/2022]
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is considered a potential therapeutic target for the treatment of type 2 diabetes mellitus (T2DM), since this enzyme plays a significant role to down-regulate insulin and leptin signalling and its over expression has been implicated in the development of insulin resistance, T2DM and obesity. Some thiazolidinediones (TZD) derivatives have been reported as promising PTP1B inhibitors with anti hyperglycemic effects. Recently, lobeglitazone, a new TZD, was described as an antidiabetic drug that targets the PPAR-γ (peroxisome γ proliferator-activated receptor) pathway, but no information on its effects on PTP1B have been reported to date. We investigated the effects of lobeglitazone on PTP1B activity in vitro. Surprisingly, lobeglitazone led to moderate inhibition on PTP1B (IC50 42.8 ± 3.8 µM) activity and to a non-competitive reversible mechanism of action. As lobeglitazone inhibits PTP1B activity in vitro, we speculate that it could also target PTP1B signalling pathway in vivo and thus contribute to potentiate its antidiabetic effects.
Collapse
Affiliation(s)
- Ruth F Rocha
- Centro de Biologia Molecular Estrutural, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Campus Trindade, 88040-900 Florianópolis, SC, Brazil
| | - Tiago Rodrigues
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| | - Angela C O Menegatti
- Centro de Biologia Molecular Estrutural, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Campus Trindade, 88040-900 Florianópolis, SC, Brazil; Universidade Federal do Piauí, CPCE, 64900-000 Bom Jesus, PI, Brazil.
| | - Gonçalo J L Bernardes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal; Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, UK
| | - Hernán Terenzi
- Centro de Biologia Molecular Estrutural, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Campus Trindade, 88040-900 Florianópolis, SC, Brazil
| |
Collapse
|
50
|
Alkoxide ligand controlled self-assembling of (imido)vanadium(V) compounds having a tetrahedral VO 3N geometry. J Inorg Biochem 2019; 203:110880. [PMID: 31726333 DOI: 10.1016/j.jinorgbio.2019.110880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 11/21/2022]
Abstract
The reaction of (1S,2S)-(-)-1,2-diphenylethylenediamine with VO(OiPr)3 in the presence of NaH was found to afford the binuclear (imido)vanadium(V) triisopropoxide, [(OiPr)3V(N-meso-1,2-DPE-N)V(OiPr)3] (DPE = diphenylethylene), (1aRS/SS). Using (1R,2R)-(+)-1,2-diphenylethylenediamine as a starting material, one-step reaction also proceeded to form the binuclear (imido)vanadium(V) triisopropoxide, [(OiPr)3V(N-meso-1,2-DPE-N)V(OiPr)3], (1aRS/RR). The single-crystal X-ray structure determination of 1aRS/SS revealed the hydrogen-bonded self-assembled structure utilizing the advantage of anti-conformation through the intermolecular hydrogen bonds of C-H···O pattern between phenyl and isopropoxide moieties, wherein each vanadium atom is coordinated in a nearly tetrahedral VO3N geometry (τ4 = 0.017 and 0.057). On the contrary, a discrete (imido)vanadium(V) tris(triphenylsiloxide) unit, which possesses a nearly tetrahedral VO3N arrangement around the vanadium metal center (τ4 = 0.060), was observed in the crystal structure of the (4-methoxyphenylimido)vanadium(V) tris(triphenylsiloxide), [(p-MeOC6H4N)V(OSiPh3)3], (1b) with bulky triphenylsiloxide ligands.
Collapse
|