1
|
Petrich J, Alvarez CE, Gómez Cano L, Dewberry R, Grotewold E, Casati P, Falcone Ferreyra ML. Functional characterization of a maize UDP-glucosyltransferase involved in the biosynthesis of flavonoid 7-O-glucosides and di-O-glucosides. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2025; 221:109583. [PMID: 39923422 DOI: 10.1016/j.plaphy.2025.109583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/11/2025]
Abstract
Flavonoids are glycosylated in the final steps of their synthesis by UDP-dependent glycosyltransferase enzymes. We present the functional characterization of the first maize flavonoid O-glucosyltransferase enzyme from family 85, which exhibits properties not previously described. ZmUGT85W2 catalyzes the O-glucosylation of flavonols, flavones and flavanones, to form primarily 7-O-glucosides, but also flavonol O-glucoside positional isomers, flavones and flavonol di-O-glucosides. ZmUGT85W2 exhibited a differential kinetic behavior depending on the flavonoid acceptor, showing hyperbolic dependence for flavonols and sigmoidal response for flavanones and flavones. Structural and molecular docking analyses predicted conserved residues interacting with the sugar donor, with close contact with the 7-hydroxyl of the flavonoid acceptors, consistent with enzymatic activity results. In addition, ZmUGT85W2 is induced by UV-B radiation, and its expression is controlled by the B and PL1 transcription factors. Consistently, higher levels of flavone and flavonol O-glycosides are accumulated in leaves of plants exposed to solar UV-B compared to control plants, suggesting that ZmUGT85W2 is involved in the biosynthesis of these metabolites in maize leaves, contributing to UV-B tolerance. The activity of ZmUGT85W2, along with its elevated expression in silks and pericarps expressing the R2R3-MYB transcription factor P1, highlights its critical role in the accumulation of flavonoid O-glucosides in these tissues. Together, our findings reveal a key step in maize flavonoid O-glycosides biosynthesis, with the observed positive cooperative behaviors suggesting that ZmUGT85W2 plays a crucial role in finely regulating metabolic flux towards these compounds in planta.
Collapse
Affiliation(s)
- Julieta Petrich
- Centro de Estudios Fotosintéticos y Bioquímicos, Universidad Nacional de Rosario, Rosario, S2002LRK, Argentina
| | - Clarisa Ester Alvarez
- Centro de Estudios Fotosintéticos y Bioquímicos, Universidad Nacional de Rosario, Rosario, S2002LRK, Argentina
| | - Lina Gómez Cano
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Ronnie Dewberry
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Erich Grotewold
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Paula Casati
- Centro de Estudios Fotosintéticos y Bioquímicos, Universidad Nacional de Rosario, Rosario, S2002LRK, Argentina
| | | |
Collapse
|
2
|
Liao J, Shahul Hameed UF, Hoffmann TD, Kurze E, Sun G, Steinchen W, Nicoli A, Di Pizio A, Kuttler C, Song C, Catici DAM, Assaad-Gerbert F, Hoffmann T, Arold ST, Schwab WG. β-Carotene alleviates substrate inhibition caused by asymmetric cooperativity. Nat Commun 2025; 16:3065. [PMID: 40157902 PMCID: PMC11954892 DOI: 10.1038/s41467-025-58259-7] [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: 05/08/2024] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
Enzymes are essential catalysts in biological systems. Substrate inhibition, once dismissed, is now observed in 20% of enzymes1 and is attributed to the formation of an unproductive enzyme-substrate complex, with no structural evidence of unproductivity provided to date1-6. This study uncovers the molecular mechanism of substrate inhibition in tobacco glucosyltransferase NbUGT72AY1, which transfers glucose to phenols for plant protection. The peculiarity that β-carotene strongly attenuates the substrate inhibition of NbUGT72AY1, despite being a competitive inhibitor, allows to determine the conformational changes that occur during substrate binding in both active and substrate-inhibited complexes. Crystallography reveals structurally different ternary enzyme-substrate complexes that do not conform to classical mechanisms. An alternative pathway suggests substrates bind randomly, but the reaction occurs only if a specific order is followed (asymmetric cooperativity). This unreported paradigm explains substrate inhibition and reactivation by competitive inhibitors, opening new research avenues in metabolic regulation and industrial applications.
Collapse
Affiliation(s)
- Jieren Liao
- Biotechnology of Natural Products, School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
| | - Umar F Shahul Hameed
- KAUST Center of Excellence for Smart Health, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Timothy D Hoffmann
- Biotechnology of Natural Products, School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
| | - Elisabeth Kurze
- Biotechnology of Natural Products, School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
| | - Guangxin Sun
- Biotechnology of Natural Products, School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Wieland Steinchen
- Center for Synthetic Microbiology (SYNMIKRO), Philipps-University Marburg, 35043, Marburg, Germany
- Department of Chemistry, Philipps-University Marburg, 35043, Marburg, Germany
| | - Alessandro Nicoli
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, 85354, Freising, Germany
- Chemoinformatics and Protein Modelling, School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
| | - Antonella Di Pizio
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, 85354, Freising, Germany
- Chemoinformatics and Protein Modelling, School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
| | - Christina Kuttler
- Analysis and Mathematical Biology, Technical University of Munich, School of Computation, Information and Technology, 85748, Garching, Germany
| | - Chuankui Song
- State Key Laboratory of Tea Plant Biology and Utilization, International Joint Laboratory on Tea Chemistry and Health Effects, Anhui Agricultural University, 230036, Hefei, Anhui, China
| | - Dragana A M Catici
- Center for Protein Assemblies (CPA), Technical University of Munich, 85748, Garching, Germany
| | - Farhah Assaad-Gerbert
- Biotechnology of Natural Products, School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
| | - Thomas Hoffmann
- Biotechnology of Natural Products, School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
| | - Stefan T Arold
- KAUST Center of Excellence for Smart Health, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Wilfried G Schwab
- Biotechnology of Natural Products, School of Life Sciences, Technical University of Munich, 85354, Freising, Germany.
| |
Collapse
|
3
|
Pal S, Panigrahy M, Adhikari R, Dua A. Memory, hysteresis, and kinetic cooperativity in stochastic mnemonic networks. J Chem Phys 2025; 162:125102. [PMID: 40135612 DOI: 10.1063/5.0252386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
Mnemonic networks are cyclic catalytic networks of monomeric enzymes that exhibit kinetic cooperativity as departures of the mean velocity from the hyperbolic, Michaelis-Menten-like response. In addition, such networks admit a hysteretic response when conformational fluctuations are slow compared to the catalytic rate. Here, we show how these fluctuation-driven effects emerge from the underlying stochasticity in the network. We use the chemical master equation to study the stochastic kinetics of mnemonic networks, which, in their minimal form, include a pair of conformers and triangular reaction pathways. We introduce statistical measures that are conditional on the turnovers to comprehensively analyze molecular fluctuations in the transient and stationary states of these networks. In the transient state, temporal correlations between enzyme turnovers lead to an inequivalence between number and temporal fluctuations, yielding a hysteretic response of the mean velocity to substrates. The transient relaxes to a stationary state with independent and identically distributed turnovers and equality between number and temporal fluctuations. This state is a non-equilibrium stationary state (NESS) when the Kolmogorov loop criterion is not satisfied, leading to the emergence of kinetic cooperativity. The symmetry of the number correlation functions allows us to distinguish between the absence of cooperativity in equilibrium and the accidental vanishing of cooperativity in a NESS. We conclude that memory and hysteresis are transient effects while kinetic cooperativity emerges as the macroscopic manifestation of the microscopic irreversibility of the NESS in a network with cyclic reaction pathways.
Collapse
Affiliation(s)
- Subham Pal
- Department of Chemistry, Indian Institute of Technology, Madras, Chennai 600036, India
| | - Manmath Panigrahy
- Department of Chemistry, Indian Institute of Technology, Madras, Chennai 600036, India
| | - R Adhikari
- DAMTP, Centre for Mathematical Sciences, University of Cambridge, Wilberforce Road, Cambridge CB3 0WA, United Kingdom
| | - Arti Dua
- Department of Chemistry, Indian Institute of Technology, Madras, Chennai 600036, India
| |
Collapse
|
4
|
Howard JA, Aziz A, Davis LA, Pugh D, Ahamed MS, Ramkissoon R, Corrales J, Nguyen NT, Mandimutsira C, Beyene T, Ha C, Dao C, Nikumbh P, Zacharias AO, Chowdhury SM, Johnson-Winters K. From Negative to No Cooperativity: Effects of Mutations on Intersubunit Communication within F 420H 2:NADP + Oxidoreductase Using Steady-State and Pre-Steady-State Kinetic Methods. Biochemistry 2025; 64:1338-1347. [PMID: 40036042 DOI: 10.1021/acs.biochem.4c00416] [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: 03/06/2025]
Abstract
F420H2:NADP+ oxidoreductase (Fno) catalyzes the reversible production of NADPH by transferring a hydride from the reduced F420 cofactor to NADP+. Previous kinetic studies suggest that wild-type Fno (wtFno) displays half-site reactivity and negative cooperativity, making Fno regulatory within methanogenic and sulfate-reducing archaea. These studies identified four amino acids; R186, T192, S190, and H133, as potential candidates involved in intersubunit communication due to their location either at or within close proximity to the interface of the dimer. Therefore, a library of Fno variants─R186K, R186Q, R186I, T192V, T192A, S190A, H133A, and H133N─was generated and characterized using binding, steady-state, and pre-steady-state kinetic experiments to understand their involvement in communication. The Hill coefficient for wtFno was previously reported as 0.61 ± 0.03, while the R186K, R186Q, R186I, and T192V Fno variant values were close or equal to 1, indicating a switch to no cooperativity behavior. The S190A variant displayed increased Hill coefficients of 0.8 ± 0.1 when compared to wtFno, showing that cooperativity was affected. The steady-state double reciprocal plots of the R186 variants, S190A, and T192V Fno variants were linear, which is indicative of no cooperativity, departing from the negative cooperativity shape displayed by wtFno. Unlike wtFno, the pre-steady-state kinetic experiments did not display half-site reactivity for the variants. Additionally, the hydride transfer step became rate-limiting in catalysis for the R186K Fno variant only. Our data suggest that negative cooperativity can be disrupted and that the amino acids R186, T192, and S190 are involved in intersubunit communication.
Collapse
Affiliation(s)
- Jamariya A Howard
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Alaa Aziz
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Lindsay A Davis
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Denzel Pugh
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Md Sabid Ahamed
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Ravi Ramkissoon
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Juan Corrales
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Nathan T Nguyen
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Charlene Mandimutsira
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Tekleab Beyene
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Co Ha
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Calvin Dao
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Parth Nikumbh
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Adway O Zacharias
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Saiful M Chowdhury
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| | - Kayunta Johnson-Winters
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019-0065, United States
| |
Collapse
|
5
|
Raihan MT, Tanaka Y, Ishikawa T. Characterization of chloroplastic thioredoxin dependent glutathione peroxidase like protein in Euglena gracilis: biochemical and functional perspectives. Biosci Biotechnol Biochem 2024; 88:1034-1046. [PMID: 38925644 DOI: 10.1093/bbb/zbae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Euglena gracilis, a fascinating organism in the scientific realm, exhibits characteristics of both animals and plants. It maintains redox homeostasis through a variety of enzymatic and non-enzymatic antioxidant molecules. In contrast to mammals, Euglena possesses nonselenocysteine glutathione peroxidase homologues that regulate its intracellular pools of reactive oxygen species. In the present study, a full-length cDNA of chloroplastic EgGPXL-1 was isolated and subjected to biochemical and functional characterization. Recombinant EgGPXL-1 scavenged H2O2 and t-BOOH, utilizing thioredoxin as an electron donor rather than glutathione. Despite its monomeric nature, EgGPXL-1 exhibits allosteric behavior with H2O2 as the electron acceptor and follows typical Michaelis-Menten kinetics with t-BOOH. Suppression of EgGPXL-1 gene expression under normal and high-light conditions did not induce critical situations in E. gracilis, suggesting the involvement of compensatory mechanisms in restoring normal conditions.
Collapse
Affiliation(s)
- Md Topu Raihan
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori, Japan
| | - Yasuhiro Tanaka
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori, Japan
| | - Takahiro Ishikawa
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori, Japan
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, Matsue, Shimane, Japan
| |
Collapse
|
6
|
Dijkema FM, Escarpizo‐Lorenzana MI, Nordentoft MK, Rabe HC, Sahin C, Landreh M, Branca RM, Sørensen ES, Christensen B, Prestel A, Teilum K, Winther JR. A suicidal and extensively disordered luciferase with a bright luminescence. Protein Sci 2024; 33:e5115. [PMID: 39023083 PMCID: PMC11255867 DOI: 10.1002/pro.5115] [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: 02/28/2024] [Revised: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/20/2024]
Abstract
Gaussia luciferase (GLuc) is one of the most luminescent luciferases known and is widely used as a reporter in biochemistry and cell biology. During catalysis, GLuc undergoes inactivation by irreversible covalent modification. The mechanism by which GLuc generates luminescence and how it becomes inactivated are however not known. Here, we show that GLuc unlike other enzymes has an extensively disordered structure with a minimal hydrophobic core and no apparent binding pocket for the main substrate, coelenterazine. From an alanine scan, we identified two Arg residues required for light production. These residues separated with an average of about 22 Å and a major structural rearrangement is required if they are to interact with the substrate simultaneously. We furthermore show that in addition to coelenterazine, GLuc also can oxidize furimazine, however, in this case without production of light. Both substrates result in the formation of adducts with the enzyme, which eventually leads to enzyme inactivation. Our results demonstrate that a rigid protein structure and substrate-binding site are no prerequisites for high enzymatic activity and specificity. In addition to the increased understanding of enzymes in general, the findings will facilitate future improvement of GLuc as a reporter luciferase.
Collapse
Affiliation(s)
- Fenne Marjolein Dijkema
- The Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | | | | | - Hanna Christin Rabe
- The Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Cagla Sahin
- The Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Michael Landreh
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Rui Mamede Branca
- Science for Life Laboratory, Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
| | - Esben Skipper Sørensen
- Department of Molecular Biology and Genetics, Section for Cellular Health, Intervention and NutritionAarhus UniversityAarhus CentrumDenmark
| | - Brian Christensen
- Department of Molecular Biology and Genetics, Section for Cellular Health, Intervention and NutritionAarhus UniversityAarhus CentrumDenmark
| | - Andreas Prestel
- The Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Kaare Teilum
- The Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Jakob Rahr Winther
- The Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
7
|
Basu Choudhury G, Datta S. Implication of Molecular Constraints Facilitating the Functional Evolution of Pseudomonas aeruginosa KPR2 into a Versatile α-Keto-Acid Reductase. Biochemistry 2024; 63:1808-1823. [PMID: 38962820 DOI: 10.1021/acs.biochem.4c00087] [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: 07/05/2024]
Abstract
Theoretical concepts linking the structure, function, and evolution of a protein, while often intuitive, necessitate validation through investigations in real-world systems. Our study empirically explores the evolutionary implications of multiple gene copies in an organism by shedding light on the structure-function modulations observed in Pseudomonas aeruginosa's second copy of ketopantoate reductase (PaKPR2). We demonstrated with two apo structures that the typical active site cleft of the protein transforms into a two-sided pocket where a molecular gate made up of two residues controls the substrate entry site, resulting in its inactivity toward the natural substrate ketopantoate. Strikingly, this structural modification made the protein active against several important α-keto-acid substrates with varied efficiency. Structural constraints at the binding site for this altered functional trait were analyzed with two binary complexes that show the conserved residue microenvironment faces restricted movements due to domain closure. Finally, its mechanistic highlights gathered from a ternary complex structure help in delineating the molecular perspectives behind its kinetic cooperativity toward these broad range of substrates. Detailed structural characteristics of the protein presented here also identified four key amino acid residues responsible for its versatile α-keto-acid reductase activity, which can be further modified to improve its functional properties through protein engineering.
Collapse
Affiliation(s)
- Gourab Basu Choudhury
- CSIR-Indian Institute of Chemical Biology, Raja S C Mullick Road, Jadavpur, Kolkata 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Saumen Datta
- CSIR-Indian Institute of Chemical Biology, Raja S C Mullick Road, Jadavpur, Kolkata 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
8
|
Vinces TC, de Souza AS, Carvalho CF, Visnardi AB, Teixeira RD, Llontop EE, Bismara BAP, Vicente EJ, Pereira JO, de Souza RF, Yonamine M, Marana SR, Farah CS, Guzzo CR. Monomeric Esterase: Insights into Cooperative Behavior, Hysteresis/Allokairy. Biochemistry 2024; 63:1178-1193. [PMID: 38669355 DOI: 10.1021/acs.biochem.3c00668] [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: 04/28/2024]
Abstract
Herein, we present a novel esterase enzyme, Ade1, isolated from a metagenomic library of Amazonian dark earths soils, demonstrating its broad substrate promiscuity by hydrolyzing ester bonds linked to aliphatic groups. The three-dimensional structure of the enzyme was solved in the presence and absence of substrate (tributyrin), revealing its classification within the α/β-hydrolase superfamily. Despite being a monomeric enzyme, enzymatic assays reveal a cooperative behavior with a sigmoidal profile (initial velocities vs substrate concentrations). Our investigation brings to light the allokairy/hysteresis behavior of Ade1, as evidenced by a transient burst profile during the hydrolysis of substrates such as p-nitrophenyl butyrate and p-nitrophenyl octanoate. Crystal structures of Ade1, coupled with molecular dynamics simulations, unveil the existence of multiple conformational structures within a single molecular state (E̅1). Notably, substrate binding induces a loop closure that traps the substrate in the catalytic site. Upon product release, the cap domain opens simultaneously with structural changes, transitioning the enzyme to a new molecular state (E̅2). This study advances our understanding of hysteresis/allokairy mechanisms, a temporal regulation that appears more pervasive than previously acknowledged and extends its presence to metabolic enzymes. These findings also hold potential implications for addressing human diseases associated with metabolic dysregulation.
Collapse
Affiliation(s)
- Tania Churasacari Vinces
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo CEP 05508-000, Brazil
| | - Anacleto Silva de Souza
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo CEP 05508-000, Brazil
| | - Cecília F Carvalho
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo CEP 05508-000, Brazil
| | - Aline Biazola Visnardi
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo CEP 05508-000, Brazil
| | - Raphael D Teixeira
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo CEP 05508-000, Brazil
| | - Edgar E Llontop
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo CEP 05508-000, Brazil
| | - Beatriz Aparecida Passos Bismara
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo CEP 05508-000, Brazil
| | - Elisabete J Vicente
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo CEP 05508-000, Brazil
| | - José O Pereira
- Biotechnology Group, Federal University of Amazonas, Amazonas CEP 69077-000, Brazil
| | - Robson Francisco de Souza
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo CEP 05508-000, Brazil
| | - Mauricio Yonamine
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo CEP 05508-000, Brazil
| | - Sandro Roberto Marana
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo CEP 05508-000, Brazil
| | - Chuck Shaker Farah
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo CEP 05508-000, Brazil
| | - Cristiane R Guzzo
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo CEP 05508-000, Brazil
| |
Collapse
|
9
|
Xie Y, Yan X, Li C, Wang S, Jia L. Characterization and insight mechanism of an acid-adapted β-Glucosidase from Lactobacillus paracasei and its application in bioconversion of glycosides. Front Bioeng Biotechnol 2024; 12:1334695. [PMID: 38333082 PMCID: PMC10851751 DOI: 10.3389/fbioe.2024.1334695] [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: 11/07/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Introduction: β-glucosidase is one class of pivotal glycosylhydrolase enzyme that can cleavage glucosidic bonds and transfer glycosyl group between the oxygen nucleophiles. Lactobacillus is the most abundant bacteria in the human gut. Identification and characterization of new β-glucosidases from Lactobacillus are meaningful for food or drug industry. Method: Herein, an acid-adapted β-glucosidase (LpBgla) was cloned and characterized from Lactobacillus paracasei. And the insight acid-adapted mechanism of LpBgla was investigated using molecular dynamics simulations. Results and Discussion: The recombinant LpBgla exhibited maximal activity at temperature of 30°C and pH 5.5, and the enzymatic activity was inhibited by Cu2+, Mn2+, Zn2+, Fe2+, Fe3+ and EDTA. The LpBgla showed a more stable structure, wider substrate-binding pocket and channel aisle, more hydrogen bonds and stronger molecular interaction with the substrate at pH 5.5 than pH 7.5. Five residues including Asp45, Leu60, Arg120, Lys153 and Arg164 might play a critical role in the acid-adapted mechanism of LpBgla. Moreover, LpBgla showed a broad substrate specificity and potential application in the bioconversion of glycosides, especially towards the arbutin. Our study greatly benefits for the development novel β-glucosidases from Lactobacillus, and for the biosynthesis of aglycones.
Collapse
Affiliation(s)
- Yufeng Xie
- College of Food Science and Engineering, Harbin University, Harbin, China
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| | - Xinrui Yan
- College of Food Science and Engineering, Harbin University, Harbin, China
| | - Changzhuo Li
- College of Food Science and Engineering, Harbin University, Harbin, China
| | - Shumei Wang
- College of Food Science and Engineering, Harbin University, Harbin, China
| | - Longgang Jia
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| |
Collapse
|
10
|
Lavrinenko IA, Vashanov GA, Hernández Cáceres JL, Nechipurenko YD. Mathematical models describing oxygen binding by hemoglobin. Biophys Rev 2023; 15:1269-1278. [PMID: 37974982 PMCID: PMC10643423 DOI: 10.1007/s12551-023-01110-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/03/2023] [Indexed: 11/19/2023] Open
Abstract
Despite the fact that the investigation of the structural and functional properties of hemoglobin dates back more than 150 years, the topic has not lost its relevance today. The most important component of these studies is the development of mathematical models that formalize and generalize the mechanisms determining the cooperative binding of ligands based on data on the structural and functional state of the protein. In this work, we review the mathematical relationships describing oxygen binding by hemoglobin, ranging from the classical Hüfner, Hill, and Adair equations to the Szabo-Karplus and tertiary two-state mathematical models based on the Monod-Wyman-Changeux and Koshland-Némethy-Filmer concepts. The generality of the considered equations as mathematical functions, bearing in their basis a power dependence, is demonstrated. The problems and possible solutions related to approximation of experimental data by the oxygenation equations with correlated fitting parameters are noted. Attention is paid to empirical equations, extended versions of the Hill equation, where the coefficient of cooperation is modulated by Gauss and Lorentz distributions as functions of partial oxygen pressure.
Collapse
Affiliation(s)
- Igor A. Lavrinenko
- Department of Human and Animal Physiology, Voronezh State University, Voronezh, 394018 Russia
| | - Gennady A. Vashanov
- Department of Human and Animal Physiology, Voronezh State University, Voronezh, 394018 Russia
| | | | - Yury D. Nechipurenko
- Laboratory of DNA-Protein Interactions, Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, 119991 Russia
- Laboratory of Molecular and Cellular Biophysics, Sevastopol State University, Sevastopol, 299053 Russia
| |
Collapse
|
11
|
Gomez GM, D’Arrigo G, Sanchez CP, Berger F, Wade RC, Lanzer M. PfCRT mutations conferring piperaquine resistance in falciparum malaria shape the kinetics of quinoline drug binding and transport. PLoS Pathog 2023; 19:e1011436. [PMID: 37285379 PMCID: PMC10281575 DOI: 10.1371/journal.ppat.1011436] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/20/2023] [Accepted: 05/21/2023] [Indexed: 06/09/2023] Open
Abstract
The chloroquine resistance transporter (PfCRT) confers resistance to a wide range of quinoline and quinoline-like antimalarial drugs in Plasmodium falciparum, with local drug histories driving its evolution and, hence, the drug transport specificities. For example, the change in prescription practice from chloroquine (CQ) to piperaquine (PPQ) in Southeast Asia has resulted in PfCRT variants that carry an additional mutation, leading to PPQ resistance and, concomitantly, to CQ re-sensitization. How this additional amino acid substitution guides such opposing changes in drug susceptibility is largely unclear. Here, we show by detailed kinetic analyses that both the CQ- and the PPQ-resistance conferring PfCRT variants can bind and transport both drugs. Surprisingly, the kinetic profiles revealed subtle yet significant differences, defining a threshold for in vivo CQ and PPQ resistance. Competition kinetics, together with docking and molecular dynamics simulations, show that the PfCRT variant from the Southeast Asian P. falciparum strain Dd2 can accept simultaneously both CQ and PPQ at distinct but allosterically interacting sites. Furthermore, combining existing mutations associated with PPQ resistance created a PfCRT isoform with unprecedented non-Michaelis-Menten kinetics and superior transport efficiency for both CQ and PPQ. Our study provides additional insights into the organization of the substrate binding cavity of PfCRT and, in addition, reveals perspectives for PfCRT variants with equal transport efficiencies for both PPQ and CQ.
Collapse
Affiliation(s)
- Guillermo M. Gomez
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld, Heidelberg, Germany
| | - Giulia D’Arrigo
- Molecular and Cellular Modelling Group, Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg, Heidelberg, Germany
| | - Cecilia P. Sanchez
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld, Heidelberg, Germany
| | - Fiona Berger
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld, Heidelberg, Germany
| | - Rebecca C. Wade
- Molecular and Cellular Modelling Group, Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg, Heidelberg, Germany
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Im Neuenheimer Feld, Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Im Neuenheimer Feld, Heidelberg, Germany
| | - Michael Lanzer
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld, Heidelberg, Germany
| |
Collapse
|
12
|
Kratky J, Eggerichs D, Heine T, Hofmann S, Sowa P, Weiße RH, Tischler D, Sträter N. Structural and Mechanistic Studies on Substrate and Stereoselectivity of the Indole Monooxygenase VpIndA1: New Avenues for Biocatalytic Epoxidations and Sulfoxidations. Angew Chem Int Ed Engl 2023; 62:e202300657. [PMID: 36762980 DOI: 10.1002/anie.202300657] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/11/2023]
Abstract
Flavoprotein monooxygenases are a versatile group of enzymes for biocatalytic transformations. Among these, group E monooxygenases (GEMs) catalyze enantioselective epoxidation and sulfoxidation reactions. Here, we describe the crystal structure of an indole monooxygenase from the bacterium Variovorax paradoxus EPS, a GEM designated as VpIndA1. Complex structures with substrates reveal productive binding modes that, in conjunction with force-field calculations and rapid mixing kinetics, reveal the structural basis of substrate and stereoselectivity. Structure-based redesign of the substrate cavity yielded variants with new substrate selectivity (for sulfoxidation of benzyl phenyl sulfide) or with greatly enhanced stereoselectivity (from 35.1 % to 99.8 % ee for production of (1S,2R)-indene oxide). This first determination of the substrate binding mode of GEMs combined with structure-function relationships opens the door for structure-based design of these powerful biocatalysts.
Collapse
Affiliation(s)
- Julia Kratky
- Institute of Bioanalytical Chemistry, Leipzig University, Deutscher Platz 5, 04103, Leipzig, Germany
| | - Daniel Eggerichs
- Microbial Biotechnology, Ruhr-Universität Bochum, Universitätsstr. 150, 44780, Bochum, Germany
| | - Thomas Heine
- Environmental Microbiology, TU Bergakademie Freiberg, Leipziger Str. 29, 09599, Freiberg, Germany
| | - Sarah Hofmann
- Environmental Microbiology, TU Bergakademie Freiberg, Leipziger Str. 29, 09599, Freiberg, Germany
| | - Philipp Sowa
- Microbial Biotechnology, Ruhr-Universität Bochum, Universitätsstr. 150, 44780, Bochum, Germany
| | - Renato H Weiße
- Institute of Bioanalytical Chemistry, Leipzig University, Deutscher Platz 5, 04103, Leipzig, Germany
| | - Dirk Tischler
- Microbial Biotechnology, Ruhr-Universität Bochum, Universitätsstr. 150, 44780, Bochum, Germany.,Environmental Microbiology, TU Bergakademie Freiberg, Leipziger Str. 29, 09599, Freiberg, Germany
| | - Norbert Sträter
- Institute of Bioanalytical Chemistry, Leipzig University, Deutscher Platz 5, 04103, Leipzig, Germany
| |
Collapse
|
13
|
Faylo JL, van Eeuwen T, Gupta K, Murakami K, Christianson DW. Transient Prenyltransferase-Cyclase Association in Fusicoccadiene Synthase, an Assembly-Line Terpene Synthase. Biochemistry 2022; 61:2417-2430. [PMID: 36227241 PMCID: PMC9648990 DOI: 10.1021/acs.biochem.2c00509] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fusicoccadiene synthase from the fungus Phomopsis amygdali (PaFS) is an assembly-line terpene synthase that catalyzes the first two steps in the biosynthesis of Fusiccocin A, a diterpene glycoside. The C-terminal prenyltransferase domain of PaFS catalyzes the condensation of one molecule of C5 dimethylallyl diphosphate and three molecules of C5 isopentenyl diphosphate to form C20 geranylgeranyl diphosphate, which then transits to the cyclase domain for cyclization to form fusicoccadiene. Previous structural studies of PaFS using electron microscopy (EM) revealed a central octameric prenyltransferase core with eight cyclase domains tethered in random distal positions through flexible 70-residue linkers. However, proximal prenyltransferase-cyclase configurations could be captured by covalent cross-linking and observed by cryo-EM and mass spectrometry. Here, we use cryo-EM to show that proximally configured prenyltransferase-cyclase complexes are observable even in the absence of covalent cross-linking; moreover, such complexes can involve multiple cyclase domains. A conserved basic patch on the prenyltransferase domain comprises the primary touchpoint with the cyclase domain. These results support a model for transient prenyltransferase-cyclase association in which the cyclase domains of PaFS are in facile equilibrium between proximal associated and random distal positions relative to the central prenyltransferase octamer. The results of biophysical measurements using small-angle X-ray scattering, analytical ultracentrifugation, dynamic light scattering, and size-exclusion chromatography in-line with multi-angle light scattering are consistent with this model. This model accordingly provides a framework for understanding substrate transit between the prenyltransferase and cyclase domains as well as the cooperativity observed for geranylgeranyl diphosphate cyclization.
Collapse
Affiliation(s)
- Jacque L. Faylo
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, 19104-6323, USA
| | - Trevor van Eeuwen
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6073, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6073, USA
| | - Kushol Gupta
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6073, USA
| | - Kenji Murakami
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6073, USA
| | - David W. Christianson
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, 19104-6323, USA
| |
Collapse
|
14
|
Atypical kinetics of cytochrome P450 enzymes in pharmacology and toxicology. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:131-176. [PMID: 35953154 DOI: 10.1016/bs.apha.2022.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Atypical kinetics are observed in metabolic reactions catalyzed by cytochrome P450 enzymes (P450). Yet, this phenomenon is regarded as experimental artifacts in some instances despite increasing evidence challenging the assumptions of typical Michaelis-Menten kinetics. As P450 play a major role in the metabolism of a wide range of substrates including drugs and endogenous compounds, it becomes critical to consider the impact of atypical kinetics on the accuracy of estimated kinetic and inhibitory parameters which could affect extrapolation of pharmacological and toxicological implications. The first half of this book chapter will focus on atypical non-Michaelis-Menten kinetics (e.g. substrate inhibition, biphasic and sigmoidal kinetics) as well as proposed underlying mechanisms supported by recent insights in mechanistic enzymology. In particular, substrate inhibition kinetics in P450 as well as concurrent drug inhibition of P450 in the presence of substrate inhibition will be further discussed. Moreover, mounting evidence has revealed that despite the high degree of sequence homology between CYP3A isoforms (i.e. CYP3A4 and CYP3A5), they have the propensities to exhibit vastly different susceptibilities and potencies of mechanism-based inactivation (MBI) with a common drug inhibitor. These experimental observations pertaining to the presence of these atypical isoform- and probe substrate-specific complexities in CYP3A isoforms by several clinically-relevant drugs will therefore be expounded and elaborated upon in the second half of this book chapter.
Collapse
|
15
|
Gallegos EM, Reed TD, Mathes FA, Guevara NV, Neau DB, Huang W, Newcomer ME, Gilbert NC. Helical remodeling augments 5-lipoxygenase activity in the synthesis of pro-inflammatory mediators. J Biol Chem 2022; 298:102282. [PMID: 35863431 PMCID: PMC9418500 DOI: 10.1016/j.jbc.2022.102282] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/29/2022] Open
Abstract
The synthesis of pro-inflammatory leukotrienes implicated in asthma, allergic rhinitis, and atherosclerosis is initiated by the enzyme 5-lipoxygenase (5-LOX). The crystal structure of human Stable-5-LOX revealed a conformation where the catalytic iron was inaccessible to bulk solvent as two aromatic residues on a conserved helix-α2 (Hα2) plugged the substrate access portal. Whether 5-LOX can also adopt a more open conformation has not been resolved. Here, we present a new conformation of 5-LOX where Hα2 adopts an elongated conformation equivalent to that described in other animal lipoxygenase structures. Our observation of the sigmoidal kinetic behavior of 5-LOX, which is indicative of positive cooperativity, is consistent with a substrate-induced conformational change that shifts the ensemble of enzyme populations to favor the catalytically competent state. Strategic point mutations along Hα2 designed to unlock the closed conformation and elongate Hα2 resulted in improved kinetic parameters, altered limited-proteolysis data, and a drastic reduction in the length of the lag phase yielding the most active Stable-5-LOX to date. Structural predictions by AlphaFold2 of these variants statistically favor an elongated Hα2 and reinforce a model in which improved kinetic parameters correlate with a more readily adopted, open conformation. Taken together, these data provide valuable insights into the synthesis of leukotrienes.
Collapse
Affiliation(s)
- Eden M Gallegos
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Tanner D Reed
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Forge A Mathes
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Nelson V Guevara
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - David B Neau
- Cornell University, Northeastern Collaborative Access Team, Argonne National Laboratory, Argonne, IL, USA
| | - Wei Huang
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Marcia E Newcomer
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Nathaniel C Gilbert
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
16
|
Structural and biochemical analysis reveals how ferulic acid improves catalytic efficiency of Humicola grisea xylanase. Sci Rep 2022; 12:11409. [PMID: 35794132 PMCID: PMC9259647 DOI: 10.1038/s41598-022-15175-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Humicolagrisea var. thermoidea is an aerobic and thermophilic fungus that secretes the GH11 xylanase HXYN2 in the presence of sugarcane bagasse. In this study, HXYN2 was expressed in Pichiapastoris and characterized biochemically and structurally in the presence of beechwood xylan substrate and ferulic acid (FA). HXYN2 is a thermally stable protein, as indicated by circular dichroism, with greater activity in the range of 40–50 °C and pH 5.0–9.0, with optimal temperature and pH of 50 °C and 6.0, respectively. FA resulted in a 75% increase in enzyme activity and a 2.5-fold increase in catalytic velocity, catalytic efficiency, and catalytic rate constant (kcat), with no alteration in enzyme affinity for the substrate. Fluorescence quenching indicated that FA forms a complex with HXYN2 interacting with solvent-exposed tryptophan residues. The binding constants ranged from moderate (pH 7.0 and 9.0) to strong (pH 4.0) affinity. Isothermal titration calorimetry, structural models and molecular docking suggested that hydrogen bonds and hydrophobic interactions occur in the aglycone region inducing conformational changes in the active site driven by initial and final enthalpy- and entropy processes, respectively. These results indicate a potential for biotechnological application for HXYN2, such as in the bioconversion of plant residues rich in ferulic acid.
Collapse
|
17
|
Moraes BC, Ribeiro-Filho HV, Roldão AP, Toniolo EF, Carretero GPB, Sgro GG, Batista FAH, Berardi DE, Oliveira VRS, Tomasin R, Vieceli FM, Pramio DT, Cardoso AB, Figueira ACM, Farah SC, Devi LA, Dale CS, de Oliveira PSL, Schechtman D. Structural analysis of TrkA mutations in patients with congenital insensitivity to pain reveals PLCγ as an analgesic drug target. Sci Signal 2022; 15:eabm6046. [PMID: 35471943 DOI: 10.1126/scisignal.abm6046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Chronic pain is a major health issue, and the search for new analgesics has become increasingly important because of the addictive properties and unwanted side effects of opioids. To explore potentially new drug targets, we investigated mutations in the NTRK1 gene found in individuals with congenital insensitivity to pain with anhidrosis (CIPA). NTRK1 encodes tropomyosin receptor kinase A (TrkA), the receptor for nerve growth factor (NGF) and that contributes to nociception. Molecular modeling and biochemical analysis identified mutations that decreased the interaction between TrkA and one of its substrates and signaling effectors, phospholipase Cγ (PLCγ). We developed a cell-permeable phosphopeptide derived from TrkA (TAT-pQYP) that bound the Src homology domain 2 (SH2) of PLCγ. In HEK-293T cells, TAT-pQYP inhibited the binding of heterologously expressed TrkA to PLCγ and decreased NGF-induced, TrkA-mediated PLCγ activation and signaling. In mice, intraplantar administration of TAT-pQYP decreased mechanical sensitivity in an inflammatory pain model, suggesting that targeting this interaction may be analgesic. The findings demonstrate a strategy to identify new targets for pain relief by analyzing the signaling pathways that are perturbed in CIPA.
Collapse
Affiliation(s)
- Beatriz C Moraes
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Helder V Ribeiro-Filho
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio) Campinas, SP 13083-100, Brazil
| | - Allan P Roldão
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Elaine F Toniolo
- Laboratory of Neuromodulation of Experimental Pain (LaNed), Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, SP 05508-000, Brazil
| | - Gustavo P B Carretero
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Germán G Sgro
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040903, Brazil
| | - Fernanda A H Batista
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio) Campinas, SP 13083-100, Brazil
| | - Damian E Berardi
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Victoria R S Oliveira
- Laboratory of Neuromodulation of Experimental Pain (LaNed), Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, SP 05508-000, Brazil
| | - Rebeka Tomasin
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Felipe M Vieceli
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Dimitrius T Pramio
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Alexandre B Cardoso
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Ana C M Figueira
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio) Campinas, SP 13083-100, Brazil
| | - Shaker C Farah
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Camila S Dale
- Laboratory of Neuromodulation of Experimental Pain (LaNed), Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, SP 05508-000, Brazil
| | - Paulo S L de Oliveira
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian Biosciences National Laboratory (LNBio) Campinas, SP 13083-100, Brazil
| | - Deborah Schechtman
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| |
Collapse
|
18
|
Bruekers JPJ, Hellinghuizen MA, Swartjes A, Tinnemans P, White PB, Elemans JAAW, Nolte RJM. 113Cd as a Probe in NMR Studies of Allosteric Host-Guest-Ligand Complexes of Porphyrin Cage Compounds. European J Org Chem 2022; 2022:e202200111. [PMID: 35915641 PMCID: PMC9324901 DOI: 10.1002/ejoc.202200111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/22/2022] [Indexed: 11/16/2022]
Abstract
Cadmium porphyrin cage compounds Cd1 and 113 Cd1 have been synthesized from the free base porphyrin cage derivative H21 and Cd(OAc)2 ⋅ 2 H2O or 113Cd(OAc)2 ⋅ 2 H2O, respectively. The compounds form allosteric complexes with the positively charged guests N,N'-dimethylimidazolium hexafluorophosphate (DMI) and N,N'-dimethylviologen dihexafluorophosphate (Me2V), which bind in the cavity of the cage, and tbupy, which coordinates as an axial ligand to the outside of the cage. In the presence of tbupy, the binding of DMI in Cd1 is enhanced by a factor of ∼31, while the presence of DMI or Me2V in the cavity of Cd1 enhances the binding of tbupy by factors of 55 and 85, respectively. The X-ray structures of the coordination complexes of Cd1 with acetone, acetonitrile, and pyridine, the host-guest complex of Cd1 with a bound viologen guest, and the ternary allosteric complex of Cd1 with a bound DMI guest and a coordinated tbupy ligand, were solved. These structures revealed relocations of the cadmium center in and out of the porphyrin plane, depending on whether a guest or a ligand is present. 113Cd NMR could be employed as a tool to quantify the binding of guests and ligands to 113 Cd1. 1D EXSY experiments on the ternary allosteric system Cd1-tbupy-Me2V revealed that the coordination of tbupy significantly slowed down the dissociation of the Me2V guest. Eyring plots of the dissociation process revealed that this kinetic allosteric effect is entropic in nature.
Collapse
Affiliation(s)
- Jeroen P. J. Bruekers
- Radboud UniversityInstitute for Molecules and MaterialsHeyendaalseweg 1356525AJNijmegenThe Netherlands
| | - Matthijs A. Hellinghuizen
- Radboud UniversityInstitute for Molecules and MaterialsHeyendaalseweg 1356525AJNijmegenThe Netherlands
| | - Anne Swartjes
- Radboud UniversityInstitute for Molecules and MaterialsHeyendaalseweg 1356525AJNijmegenThe Netherlands
| | - Paul Tinnemans
- Radboud UniversityInstitute for Molecules and MaterialsHeyendaalseweg 1356525AJNijmegenThe Netherlands
| | - Paul B. White
- Radboud UniversityInstitute for Molecules and MaterialsHeyendaalseweg 1356525AJNijmegenThe Netherlands
| | - Johannes A. A. W. Elemans
- Radboud UniversityInstitute for Molecules and MaterialsHeyendaalseweg 1356525AJNijmegenThe Netherlands
| | - Roeland J. M. Nolte
- Radboud UniversityInstitute for Molecules and MaterialsHeyendaalseweg 1356525AJNijmegenThe Netherlands
| |
Collapse
|
19
|
Versluys M, Porras-Domínguez JR, De Coninck T, Van Damme EJM, Van den Ende W. A novel chicory fructanase can degrade common microbial fructan product profiles and displays positive cooperativity. JOURNAL OF EXPERIMENTAL BOTANY 2022; 73:1602-1622. [PMID: 34750605 DOI: 10.1093/jxb/erab488] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/03/2021] [Indexed: 06/13/2023]
Abstract
Fructan metabolism in bacteria and plants relies on fructosyltransferases and fructanases. Plant fructanases (fructan exohydrolase, FEH) only hydrolyse terminal fructose residues. Levan (β-2,6 linkages) is the most abundant fructan type in bacteria. Dicot fructan accumulators, such as chicory (Cichorium intybus), accumulate inulin (β-2,1 linkages), harbouring several 1-FEH isoforms for their degradation. Here, a novel chicory fructanase with high affinity for levan was characterized, providing evidence that such enzymes widely occur in higher plants. It is adapted to common microbial fructan profiles, but has low affinity towards chicory inulin, in line with a function in trimming of microbial fructans in the extracellular environment. Docking experiments indicate the importance of an N-glycosylation site close to the active site for substrate specificity. Optimal pH and temperature for levan hydrolysis are 5.0 and 43.7 °C, respectively. Docking experiments suggested multiple substrate binding sites and levan-mediated enzyme dimerization, explaining the observed positive cooperativity. Alignments show a single amino acid shift in the position of a conserved DXX(R/K) couple, typical for sucrose binding in cell wall invertases. A possible involvement of plant fructanases in levan trimming is discussed, in line with the emerging 'fructan detour' concepts, suggesting that levan oligosaccharides act as signalling entities during plant-microbial interactions.
Collapse
Affiliation(s)
- Maxime Versluys
- Laboratory of Molecular Plant Biology, KU Leuven, Kasteelpark Arenberg 31, 3001 Leuven, Belgium
| | | | - Tibo De Coninck
- Laboratory of Biochemistry and Glycobiology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Els J M Van Damme
- Laboratory of Biochemistry and Glycobiology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Wim Van den Ende
- Laboratory of Molecular Plant Biology, KU Leuven, Kasteelpark Arenberg 31, 3001 Leuven, Belgium
| |
Collapse
|
20
|
Kum SL, Ho JCS, Parikh AN, Liedberg B. Amphiphilic Membrane Environments Regulate Enzymatic Behaviors of Salmonella Outer Membrane Protease. ACS BIO & MED CHEM AU 2022; 2:73-83. [PMID: 37102179 PMCID: PMC10114716 DOI: 10.1021/acsbiomedchemau.1c00027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
The role of an amphiphilic environment in the functional regulation of integral membrane proteins is well appreciated but how specific amphiphilic surrounding influences the conformational plasticity and function of a protein is less obvious. We focus on the Salmonella phosphoglycerate transport system (pgt)-encoded outer membrane protease E (PgtE), which plays an important role in tissue infiltration and survival of Salmonella enterica. Despite our understanding of its physiological functions, elucidation of its enzymatic behavior in response to the immediate amphiphilic surrounding is lacking. We monitor the proteolytic activity of PgtE reconstituted in Zwittergent 3-12 detergent micelles or a 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine (POPC) bilayer and examine factors that influence its activity. We find, to our surprise, that PgtE, which is thought to elicit a rapid response toward various substrates, showed hysteretic enzymatic behavior, characterized by a prominent lag phase prior to achieving the exponential steady state in its detergent-stabilized form as well as in the outer membrane embedded native state in live bacteria. The lag phase was abolished under three conditions: preformation of an inactive detergent-stabilized PgtE-substrate complex without lipopolysaccharide (LPS), LPS-bound detergent-stabilized PgtE that had reached steady state velocity, or PgtE reconstituted into a POPC bilayer environment. Interestingly, detergent- and bilayer-stabilized PgtE showed comparable steady-state activity. And strikingly, lipopolysaccharide (LPS) becomes nonessential for the activation of PgtE when the protein is reconstituted in the phospholipid bilayer, contrasting a long-standing notion that LPS is required for proteases belonging to the omptin family to be proteolytically active. These findings suggest intriguing biological nuances for the proteolytic function of PgtE that were not well appreciated previously and offer new perspectives that may generally be applicable for omptins.
Collapse
Affiliation(s)
- Siau Ling Kum
- Centre
for Biomimetic Sensor Science, Nanyang Technological
University, 50 Nanyang Drive, 637553 Singapore
- School
of Materials Science and Engineering, Nanyang
Technological University, 50 Nanyang Ave, 639798 Singapore
| | - James C. S. Ho
- Centre
for Biomimetic Sensor Science, Nanyang Technological
University, 50 Nanyang Drive, 637553 Singapore
- School
of Materials Science and Engineering, Nanyang
Technological University, 50 Nanyang Ave, 639798 Singapore
| | - Atul N. Parikh
- Centre
for Biomimetic Sensor Science, Nanyang Technological
University, 50 Nanyang Drive, 637553 Singapore
- School
of Materials Science and Engineering, Nanyang
Technological University, 50 Nanyang Ave, 639798 Singapore
- Department of Chemistry and Department of
Biomedical Engineering, University of California, Davis, California 95616, United States
| | - Bo Liedberg
- Centre
for Biomimetic Sensor Science, Nanyang Technological
University, 50 Nanyang Drive, 637553 Singapore
- School
of Materials Science and Engineering, Nanyang
Technological University, 50 Nanyang Ave, 639798 Singapore
| |
Collapse
|
21
|
Bazzone A, Tesmer L, Kurt D, Kaback HR, Fendler K, Madej MG. Investigation of sugar binding kinetics of the E. coli sugar/H + symporter XylE using solid supported membrane-based electrophysiology. J Biol Chem 2021; 298:101505. [PMID: 34929170 PMCID: PMC8784342 DOI: 10.1016/j.jbc.2021.101505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 12/19/2022] Open
Abstract
Bacterial transporters are difficult to study using conventional electrophysiology because of their low transport rates and the small size of bacterial cells. Here, we applied solid-supported membrane–based electrophysiology to derive kinetic parameters of sugar translocation by the Escherichia coli xylose permease (XylE), including functionally relevant mutants. Many aspects of the fucose permease (FucP) and lactose permease (LacY) have also been investigated, which allow for more comprehensive conclusions regarding the mechanism of sugar translocation by transporters of the major facilitator superfamily. In all three of these symporters, we observed sugar binding and transport in real time to determine KM, Vmax, KD, and kobs values for different sugar substrates. KD and kobs values were attainable because of a conserved sugar-induced electrogenic conformational transition within these transporters. We also analyzed interactions between the residues in the available X-ray sugar/H+ symporter structures obtained with different bound sugars. We found that different sugars induce different conformational states, possibly correlating with different charge displacements in the electrophysiological assay upon sugar binding. Finally, we found that mutations in XylE altered the kinetics of glucose binding and transport, as Q175 and L297 are necessary for uncoupling H+ and d-glucose translocation. Based on the rates for the electrogenic conformational transition upon sugar binding (>300 s−1) and for sugar translocation (2 s−1 − 30 s−1 for different substrates), we propose a multiple-step mechanism and postulate an energy profile for sugar translocation. We also suggest a mechanism by which d-glucose can act as an inhibitor for XylE.
Collapse
Affiliation(s)
- Andre Bazzone
- Max-Planck-Institute of Biophysics, Department of Biophysical Chemistry in Frankfurt/M, Germany
| | - Laura Tesmer
- Max-Planck-Institute of Biophysics, Department of Biophysical Chemistry in Frankfurt/M, Germany
| | - Derya Kurt
- Max-Planck-Institute of Biophysics, Department of Biophysical Chemistry in Frankfurt/M, Germany
| | - H Ronald Kaback
- University of California, Department of Physiology and Department of Microbiology, Immunology, Molecular Genetics, Molecular Biology Institute in Los Angeles CA, USA
| | - Klaus Fendler
- Max-Planck-Institute of Biophysics, Department of Biophysical Chemistry in Frankfurt/M, Germany
| | - M Gregor Madej
- Institute of Biophysics and Biophysical Chemistry, Department of Structural Biology, University of Regensburg, Universitätsstr. 31, 95053 Regensburg, Germany; Institute of Biophysics, Department of Structural Biology, Saarland University, Center of Human and Molecular Biology, Building 60, 66421 Homburg, Germany
| |
Collapse
|
22
|
Carroll BL, Zahn KE, Hanley JP, Wallace SS, Dragon JA, Doublié S. Caught in motion: human NTHL1 undergoes interdomain rearrangement necessary for catalysis. Nucleic Acids Res 2021; 49:13165-13178. [PMID: 34871433 PMCID: PMC8682792 DOI: 10.1093/nar/gkab1162] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/02/2021] [Accepted: 12/03/2021] [Indexed: 01/08/2023] Open
Abstract
Base excision repair (BER) is the main pathway protecting cells from the continuous damage to DNA inflicted by reactive oxygen species. BER is initiated by DNA glycosylases, each of which repairs a particular class of base damage. NTHL1, a bifunctional DNA glycosylase, possesses both glycolytic and β-lytic activities with a preference for oxidized pyrimidine substrates. Defects in human NTHL1 drive a class of polyposis colorectal cancer. We report the first X-ray crystal structure of hNTHL1, revealing an open conformation not previously observed in the bacterial orthologs. In this conformation, the six-helical barrel domain comprising the helix-hairpin-helix (HhH) DNA binding motif is tipped away from the iron sulphur cluster-containing domain, requiring a conformational change to assemble a catalytic site upon DNA binding. We found that the flexibility of hNTHL1 and its ability to adopt an open configuration can be attributed to an interdomain linker. Swapping the human linker sequence for that of Escherichia coli yielded a protein chimera that crystallized in a closed conformation and had a reduced activity on lesion-containing DNA. This large scale interdomain rearrangement during catalysis is unprecedented for a HhH superfamily DNA glycosylase and provides important insight into the molecular mechanism of hNTHL1.
Collapse
Affiliation(s)
- Brittany L Carroll
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA
| | - Karl E Zahn
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA
| | - John P Hanley
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA
| | - Susan S Wallace
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA
| | - Julie A Dragon
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA
| | - Sylvie Doublié
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405, USA
| |
Collapse
|
23
|
Choudhury A, Khanppnavar B, Datta S. Crystallographic and biophysical analyses of Pseudomonas aeruginosa ketopantoate reductase: Implications of ligand induced conformational changes in cofactor recognition. Biochimie 2021; 193:103-114. [PMID: 34757166 DOI: 10.1016/j.biochi.2021.10.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/15/2021] [Accepted: 10/28/2021] [Indexed: 11/26/2022]
Abstract
Ketopantoate reductases (KPRs) catalyse NADPH-dependent reduction of ketopantoate to pantoate, the rate-limiting step of pantothenate biosynthetic pathway. In our recent study, we showed KPRs are under dynamic evolutionary selection and highlighted the possible role of ordered substrate binding kinetics for cofactor selection. To further delineate this at molecular level, here, we perform X-ray crystallographic and biophysical analyses of KPR in presence of non-canonical cofactor NAD+. In our structure, NAD+ was found to be highly dynamic in catalytic pocket of KPR, which could attain stable conformation only in presence of ketopantoate. Further, isothermal calorimetric (ITC) titrations showed that affinity of KPR for ketopantoate is higher in presence of NADP+ than in presence of NAD+ and lowest in absence of redox cofactors. In sum, our results clearly depict two modes of redox cofactor selections in KPRs, firstly by specific salt bridge interactions with unique phosphate moiety of NADP+ and secondly via ordered sequential heterotrophic cooperative binding of substrate ketopantoate.
Collapse
Affiliation(s)
- Arkaprabha Choudhury
- Department of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology (CSIR-IICB), Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), India
| | - Basavraj Khanppnavar
- Department of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology (CSIR-IICB), Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), India
| | - Saumen Datta
- Department of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology (CSIR-IICB), Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), India.
| |
Collapse
|
24
|
Guerreiro BM, Silva JC, Lima JC, Reis MAM, Freitas F. Antioxidant Potential of the Bio-Based Fucose-Rich Polysaccharide FucoPol Supports Its Use in Oxidative Stress-Inducing Systems. Polymers (Basel) 2021; 13:3020. [PMID: 34577923 PMCID: PMC8470694 DOI: 10.3390/polym13183020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) are dangerous sources of macromolecular damage. While most derive from mitochondrial oxidative phosphorylation, their production can be triggered by exogenous stresses, surpassing the extinction capacity of intrinsic antioxidant defense systems of cells. Here, we report the antioxidant activity of FucoPol, a fucose-rich polyanionic polysaccharide produced by Enterobacter A47, containing ca. 17 wt% of negatively charged residues in its structure. Ferric reducing antioxidant power (FRAP) assays coupled to Hill binding kinetics fitting have shown FucoPol can neutralize ferricyanide and Fe3+-TPTZ species at an EC50 of 896 and 602 µg/mL, respectively, with positive binding cooperativity (2.52 ≤ H ≤ 4.85). This reducing power is greater than most polysaccharides reported. Moreover, an optimal 0.25% w/v FucoPol concentration shown previously to be cryo- and photoprotective was also demonstrated to protect Vero cells against H2O2-induced acute exposure not only by attenuating metabolic viability decay, but also by accentuating post-stress proliferation capacity, whilst preserving cell morphology. These results on antioxidant activity provide evidence for the biopolymer's ability to prevent positive feedback cascades of the radical-producing Fenton reaction. Ultimately, FucoPol provides a biotechnological alternative for implementation in cryopreservation, food supplementation, and photoprotective sunscreen formula design, as all fields benefit from an antioxidant functionality.
Collapse
Affiliation(s)
- Bruno M. Guerreiro
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal; (B.M.G.); (M.A.M.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- CENIMAT/I3N, Department of Physics, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2819-516 Caparica, Portugal;
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2819-516 Caparica, Portugal;
| | - Jorge Carvalho Silva
- CENIMAT/I3N, Department of Physics, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2819-516 Caparica, Portugal;
| | - João Carlos Lima
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2819-516 Caparica, Portugal;
| | - Maria A. M. Reis
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal; (B.M.G.); (M.A.M.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Filomena Freitas
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal; (B.M.G.); (M.A.M.R.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| |
Collapse
|
25
|
A Novel Cooperative Metallo-β-Lactamase Fold Metallohydrolase from Pathogen Vibrio vulnificus Exhibits β-Lactam Antibiotic-Degrading Activities. Antimicrob Agents Chemother 2021; 65:e0032621. [PMID: 34228542 DOI: 10.1128/aac.00326-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vibrio vulnificus is a pathogen that accounts for one of the highest mortality rates and is responsible for most reported seafood-related illnesses and deaths worldwide. Owing to the threats of pathogens with β-lactamase activity, it is important to identify and characterize β-lactamases with clinical significance. In this study, the protein sequence of the metallo-β-lactamase (MBL) fold metallohydrolase from V. vulnificus (designated Vmh) was analyzed, and its oligomeric state, β-lactamase activity, and metal binding ability were determined. BLASTp analysis indicated that the V. vulnificus Vmh protein showed no significant sequence identity with any experimentally identified Ambler class B MBLs or enzymes containing the MBL protein fold; it was also predicted to have a signal peptide of 19 amino acids at its N terminus and an MBL protein fold from amino acid residues 23 to 216. Recombinant V. vulnificus Vmh protein was overexpressed and purified. Analytical ultracentrifugation and electrospray ionization-mass spectrometry (MS) data demonstrated its monomeric state in an aqueous solution. Recombinant V. vulnificus Vmh protein showed broad degrading activities against β-lactam antibiotics, such as penicillins, cephalosporins, and imipenems, with kcat/Km values ranging from 6.23 × 102 to 1.02 × 104 M-1 s-1. The kinetic reactions of this enzyme exhibited sigmoidal behavior, suggesting the possibility of cooperativity. Zinc ions were required for the enzyme activity, which was abolished by adding the metal chelator EDTA. Inductively coupled plasma-MS indicated that this enzyme might bind two zinc ions per molecule as a cofactor.
Collapse
|
26
|
Freire MCLC, Noske GD, Bitencourt NV, Sanches PRS, Santos-Filho NA, Gawriljuk VO, de Souza EP, Nogueira VHR, de Godoy MO, Nakamura AM, Fernandes RS, Godoy AS, Juliano MA, Peres BM, Barbosa CG, Moraes CB, Freitas-Junior LHG, Cilli EM, Guido RVC, Oliva G. Non-Toxic Dimeric Peptides Derived from the Bothropstoxin-I Are Potent SARS-CoV-2 and Papain-like Protease Inhibitors. Molecules 2021; 26:4896. [PMID: 34443484 PMCID: PMC8401042 DOI: 10.3390/molecules26164896] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 12/24/2022] Open
Abstract
The COVID-19 outbreak has rapidly spread on a global scale, affecting the economy and public health systems throughout the world. In recent years, peptide-based therapeutics have been widely studied and developed to treat infectious diseases, including viral infections. Herein, the antiviral effects of the lysine linked dimer des-Cys11, Lys12,Lys13-(pBthTX-I)2K ((pBthTX-I)2K)) and derivatives against SARS-CoV-2 are reported. The lead peptide (pBthTX-I)2K and derivatives showed attractive inhibitory activities against SARS-CoV-2 (EC50 = 28-65 µM) and mostly low cytotoxic effect (CC50 > 100 µM). To shed light on the mechanism of action underlying the peptides' antiviral activity, the Main Protease (Mpro) and Papain-Like protease (PLpro) inhibitory activities of the peptides were assessed. The synthetic peptides showed PLpro inhibition potencies (IC50s = 1.0-3.5 µM) and binding affinities (Kd = 0.9-7 µM) at the low micromolar range but poor inhibitory activity against Mpro (IC50 > 10 µM). The modeled binding mode of a representative peptide of the series indicated that the compound blocked the entry of the PLpro substrate toward the protease catalytic cleft. Our findings indicated that non-toxic dimeric peptides derived from the Bothropstoxin-I have attractive cellular and enzymatic inhibitory activities, thereby suggesting that they are promising prototypes for the discovery and development of new drugs against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Marjorie C. L. C. Freire
- São Carlos Institute of Physics, University of Sao Paulo, Avenida João Dagnone, 1100, São Carlos 13563-120, SP, Brazil; (M.C.L.C.F.); (G.D.N.); (V.O.G.); (V.H.R.N.); (M.O.d.G.); (A.M.N.); (R.S.F.); (A.S.G.)
| | - Gabriela D. Noske
- São Carlos Institute of Physics, University of Sao Paulo, Avenida João Dagnone, 1100, São Carlos 13563-120, SP, Brazil; (M.C.L.C.F.); (G.D.N.); (V.O.G.); (V.H.R.N.); (M.O.d.G.); (A.M.N.); (R.S.F.); (A.S.G.)
| | - Natália V. Bitencourt
- Department of Biochemistry and Organic Chemistry, Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil; (N.V.B.); (P.R.S.S.); (N.A.S.-F.); (E.M.C.)
| | - Paulo R. S. Sanches
- Department of Biochemistry and Organic Chemistry, Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil; (N.V.B.); (P.R.S.S.); (N.A.S.-F.); (E.M.C.)
| | - Norival A. Santos-Filho
- Department of Biochemistry and Organic Chemistry, Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil; (N.V.B.); (P.R.S.S.); (N.A.S.-F.); (E.M.C.)
| | - Victor O. Gawriljuk
- São Carlos Institute of Physics, University of Sao Paulo, Avenida João Dagnone, 1100, São Carlos 13563-120, SP, Brazil; (M.C.L.C.F.); (G.D.N.); (V.O.G.); (V.H.R.N.); (M.O.d.G.); (A.M.N.); (R.S.F.); (A.S.G.)
| | - Eduardo P. de Souza
- Department of Genetics and Evolution, Federal University of São Carlos, Rodovia Washington Luís km 235, São Carlos 13565-905, SP, Brazil;
| | - Victor H. R. Nogueira
- São Carlos Institute of Physics, University of Sao Paulo, Avenida João Dagnone, 1100, São Carlos 13563-120, SP, Brazil; (M.C.L.C.F.); (G.D.N.); (V.O.G.); (V.H.R.N.); (M.O.d.G.); (A.M.N.); (R.S.F.); (A.S.G.)
| | - Mariana O. de Godoy
- São Carlos Institute of Physics, University of Sao Paulo, Avenida João Dagnone, 1100, São Carlos 13563-120, SP, Brazil; (M.C.L.C.F.); (G.D.N.); (V.O.G.); (V.H.R.N.); (M.O.d.G.); (A.M.N.); (R.S.F.); (A.S.G.)
| | - Aline M. Nakamura
- São Carlos Institute of Physics, University of Sao Paulo, Avenida João Dagnone, 1100, São Carlos 13563-120, SP, Brazil; (M.C.L.C.F.); (G.D.N.); (V.O.G.); (V.H.R.N.); (M.O.d.G.); (A.M.N.); (R.S.F.); (A.S.G.)
| | - Rafaela S. Fernandes
- São Carlos Institute of Physics, University of Sao Paulo, Avenida João Dagnone, 1100, São Carlos 13563-120, SP, Brazil; (M.C.L.C.F.); (G.D.N.); (V.O.G.); (V.H.R.N.); (M.O.d.G.); (A.M.N.); (R.S.F.); (A.S.G.)
| | - Andre S. Godoy
- São Carlos Institute of Physics, University of Sao Paulo, Avenida João Dagnone, 1100, São Carlos 13563-120, SP, Brazil; (M.C.L.C.F.); (G.D.N.); (V.O.G.); (V.H.R.N.); (M.O.d.G.); (A.M.N.); (R.S.F.); (A.S.G.)
| | - Maria A. Juliano
- The Sao Paulo School of Medicine, Federal University of São Paulo, Rua Três de Maio, 100, São Paulo 04044-020, SP, Brazil;
| | - Bianca M. Peres
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo 05508-900, SP, Brazil; (B.M.P.); (C.G.B.); (L.H.G.F.-J.)
| | - Cecília G. Barbosa
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo 05508-900, SP, Brazil; (B.M.P.); (C.G.B.); (L.H.G.F.-J.)
| | - Carolina B. Moraes
- Department of Pharmaceutical Sciences, Federal University of São Paulo, Rua São Nicolau, 210, Diadema 09913-030, SP, Brazil;
| | - Lucio H. G. Freitas-Junior
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo 05508-900, SP, Brazil; (B.M.P.); (C.G.B.); (L.H.G.F.-J.)
| | - Eduardo M. Cilli
- Department of Biochemistry and Organic Chemistry, Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil; (N.V.B.); (P.R.S.S.); (N.A.S.-F.); (E.M.C.)
| | - Rafael V. C. Guido
- São Carlos Institute of Physics, University of Sao Paulo, Avenida João Dagnone, 1100, São Carlos 13563-120, SP, Brazil; (M.C.L.C.F.); (G.D.N.); (V.O.G.); (V.H.R.N.); (M.O.d.G.); (A.M.N.); (R.S.F.); (A.S.G.)
| | - Glaucius Oliva
- São Carlos Institute of Physics, University of Sao Paulo, Avenida João Dagnone, 1100, São Carlos 13563-120, SP, Brazil; (M.C.L.C.F.); (G.D.N.); (V.O.G.); (V.H.R.N.); (M.O.d.G.); (A.M.N.); (R.S.F.); (A.S.G.)
| |
Collapse
|
27
|
Le Coq J, López Navajas P, Rodrigo Martin B, Alfonso C, Lietha D. A new layer of phosphoinositide-mediated allosteric regulation uncovered for SHIP2. FASEB J 2021; 35:e21815. [PMID: 34314064 DOI: 10.1096/fj.202100561r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/26/2021] [Accepted: 07/08/2021] [Indexed: 11/11/2022]
Abstract
The Src homology 2 containing inositol 5-phosphatase 2 (SHIP2) is a large multidomain enzyme that catalyzes the dephosphorylation of the phospholipid phosphatidylinositol 3,4,5-triphosphate (PI(3,4,5)P3 ) to form PI(3,4)P2 . PI(3,4,5)P3 is a key lipid second messenger controlling the recruitment of signaling proteins to the plasma membrane, thereby regulating a plethora of cellular events, including proliferation, growth, apoptosis, and cytoskeletal rearrangements. SHIP2, alongside PI3K and PTEN, regulates PI(3,4,5)P3 levels at the plasma membrane and has been heavily implicated in serious diseases such as cancer and type 2 diabetes; however, many aspects of its regulation mechanism remain elusive. We recently reported an activating effect of the SHIP2 C2 domain and here we describe an additional layer of regulation via the pleckstrin homology-related (PHR) domain. We show a phosphoinositide-induced transition to a high activity state of the enzyme that increases phosphatase activity up to 10-15 fold. We further show that PI(3,4)P2 directly interacts with the PHR domain to trigger this allosteric activation. Modeling of the PHR-phosphatase-C2 region of SHIP2 on the membrane suggests no major inter-domain interactions with the PHR domain, but close contacts between the two linkers offer a possible path of allosteric communication. Together, our data show that the PHR domain acts as an allosteric module regulating the catalytic activity of SHIP2 in response to specific phosphoinositide levels in the cell membrane.
Collapse
Affiliation(s)
- Johanne Le Coq
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Pilar López Navajas
- Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, Spanish National Research Council (CSIC), Madrid, Spain
| | - Bárbara Rodrigo Martin
- Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, Spanish National Research Council (CSIC), Madrid, Spain
| | - Carlos Alfonso
- Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, Spanish National Research Council (CSIC), Madrid, Spain
| | - Daniel Lietha
- Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, Spanish National Research Council (CSIC), Madrid, Spain
| |
Collapse
|
28
|
Mak CA, Weis K, Henao T, Kuchtova A, Chen T, Sharma S, Meekins DA, Thalmann M, Vander Kooi CW, Raththagala M. Cooperative Kinetics of the Glucan Phosphatase Starch Excess4. Biochemistry 2021; 60:2425-2435. [PMID: 34319705 DOI: 10.1021/acs.biochem.1c00307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glucan phosphatases are members of a functionally diverse family of dual-specificity phosphatase (DSP) enzymes. The plant glucan phosphatase Starch Excess4 (SEX4) binds and dephosphorylates glucans, contributing to processive starch degradation in the chloroplast at night. Little is known about the complex kinetics of SEX4 when acting on its complex physiologically relevant glucan substrate. Therefore, we explored the kinetics of SEX4 against both insoluble starch and soluble amylopectin glucan substrates. SEX4 displays robust activity and a unique sigmoidal kinetic response to amylopectin, characterized by a Hill coefficient of 2.77 ± 0.63, a signature feature of cooperativity. We investigated the basis for this positive kinetic cooperativity and determined that the SEX4 carbohydrate-binding module (CBM) dramatically influences the binding cooperativity and substrate transformation rates. These findings provide insights into a previously unknown but important regulatory role for SEX4 in reversible starch phosphorylation and further advances our understanding of atypical kinetic mechanisms.
Collapse
Affiliation(s)
- Claudia A Mak
- Department of Chemistry, Skidmore College, Saratoga Springs, New York 12866, United States
| | - Kenyon Weis
- Department of Chemistry, Skidmore College, Saratoga Springs, New York 12866, United States
| | - Tiffany Henao
- Department of Chemistry, Skidmore College, Saratoga Springs, New York 12866, United States
| | - Andrea Kuchtova
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky 40506, United States
| | - Tiantian Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky 40506, United States
| | - Savita Sharma
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky 40506, United States
| | - David A Meekins
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky 40506, United States
| | - Matthias Thalmann
- The John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom
| | - Craig W Vander Kooi
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky 40506, United States
| | - Madushi Raththagala
- Department of Chemistry, Skidmore College, Saratoga Springs, New York 12866, United States
| |
Collapse
|
29
|
Faylo JL, van Eeuwen T, Kim HJ, Gorbea Colón JJ, Garcia BA, Murakami K, Christianson DW. Structural insight on assembly-line catalysis in terpene biosynthesis. Nat Commun 2021; 12:3487. [PMID: 34108468 PMCID: PMC8190136 DOI: 10.1038/s41467-021-23589-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 05/05/2021] [Indexed: 12/20/2022] Open
Abstract
Fusicoccadiene synthase from Phomopsis amygdali (PaFS) is a unique bifunctional terpenoid synthase that catalyzes the first two steps in the biosynthesis of the diterpene glycoside Fusicoccin A, a mediator of 14-3-3 protein interactions. The prenyltransferase domain of PaFS generates geranylgeranyl diphosphate, which the cyclase domain then utilizes to generate fusicoccadiene, the tricyclic hydrocarbon skeleton of Fusicoccin A. Here, we use cryo-electron microscopy to show that the structure of full-length PaFS consists of a central octameric core of prenyltransferase domains, with the eight cyclase domains radiating outward via flexible linker segments in variable splayed-out positions. Cryo-electron microscopy and chemical crosslinking experiments additionally show that compact conformations can be achieved in which cyclase domains are more closely associated with the prenyltransferase core. This structural analysis provides a framework for understanding substrate channeling, since most of the geranylgeranyl diphosphate generated by the prenyltransferase domains remains on the enzyme for cyclization to form fusicoccadiene.
Collapse
Affiliation(s)
- Jacque L Faylo
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, PA, United States
| | - Trevor van Eeuwen
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hee Jong Kim
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jose J Gorbea Colón
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Benjamin A Garcia
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kenji Murakami
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David W Christianson
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
30
|
Zitare UA, Habib MH, Rozeboom H, Mascotti ML, Todorovic S, Fraaije MW. Mutational and structural analysis of an ancestral fungal dye-decolorizing peroxidase. FEBS J 2021; 288:3602-3618. [PMID: 33369202 PMCID: PMC8248431 DOI: 10.1111/febs.15687] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 12/31/2022]
Abstract
Dye-decolorizing peroxidases (DyPs) constitute a superfamily of heme-containing peroxidases that are related neither to animal nor to plant peroxidase families. These are divided into four classes (types A, B, C, and D) based on sequence features. The active site of DyPs contains two highly conserved distal ligands, an aspartate and an arginine, the roles of which are still controversial. These ligands have mainly been studied in class A-C bacterial DyPs, largely because no effective recombinant expression systems have been developed for the fungal (D-type) DyPs. In this work, we employ ancestral sequence reconstruction (ASR) to resurrect a D-type DyP ancestor, AncDyPD-b1. Expression of AncDyPD-b1 in Escherichia coli results in large amounts of a heme-containing soluble protein and allows for the first mutagenesis study on the two distal ligands of a fungal DyP. UV-Vis and resonance Raman (RR) spectroscopic analyses, in combination with steady-state kinetics and the crystal structure, reveal fine pH-dependent details about the heme active site structure and show that both the aspartate (D222) and the arginine (R390) are crucial for hydrogen peroxide reduction. Moreover, the data indicate that these two residues play important but mechanistically different roles on the intraprotein long-range electron transfer process. DATABASE: Structural data are available in the PDB database under the accession number 7ANV.
Collapse
Affiliation(s)
- Ulises A. Zitare
- Molecular Enzymology GroupUniversity of GroningenThe Netherlands
- Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE)Departamento de Química Inorgánica, Analítica y Química FísicaFacultad de Ciencias Exactas y NaturalesUniversidad de Buenos Aires and CONICETArgentina
| | - Mohamed H. Habib
- Molecular Enzymology GroupUniversity of GroningenThe Netherlands
- Department of Microbiology and ImmunologyFaculty of PharmacyCairo UniversityEgypt
| | | | - Maria L. Mascotti
- Molecular Enzymology GroupUniversity of GroningenThe Netherlands
- IMIBIO‐SL CONICETFacultad de Química Bioquímica y FarmaciaUniversidad Nacional de San LuisArgentina
| | - Smilja Todorovic
- Instituto de Tecnologia Química e BiológicaUniversidade Nova de LisboaOeirasPortugal
| | - Marco W. Fraaije
- Molecular Enzymology GroupUniversity of GroningenThe Netherlands
| |
Collapse
|
31
|
Olmeda I, Casino P, Collins RE, Sendra R, Callejón S, Huesa J, Soares AS, Ferrer S, Pardo I. Structural analysis and biochemical properties of laccase enzymes from two Pediococcus species. Microb Biotechnol 2021; 14:1026-1043. [PMID: 33635570 PMCID: PMC8085982 DOI: 10.1111/1751-7915.13751] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/22/2020] [Accepted: 12/31/2020] [Indexed: 11/29/2022] Open
Abstract
Prokaryotic laccases are emergent biocatalysts. However, they have not been broadly found and characterized in bacterial organisms, especially in lactic acid bacteria. Recently, a prokaryotic laccase from the lactic acid bacterium Pediococcus acidilactici 5930, which can degrade biogenic amines, was discovered. Thus, our study aimed to shed light on laccases from lactic acid bacteria focusing on two Pediococcus laccases, P. acidilactici 5930 and Pediococcus pentosaceus 4816, which have provided valuable information on their biochemical activities on redox mediators and biogenic amines. Both laccases are able to oxidize canonical substrates as ABTS, ferrocyanide and 2,6-DMP, and non-conventional substrates as biogenic amines. With ABTS as a substrate, they prefer an acidic environment and show sigmoidal kinetic activity, and are rather thermostable. Moreover, this study has provided the first structural view of two lactic acid bacteria laccases, revealing new structural features not seen before in other well-studied laccases, but which seem characteristic for this group of bacteria. We believe that understanding the role of laccases in lactic acid bacteria will have an impact on their biotechnological applications and provide a framework for the development of engineered lactic acid bacteria with enhanced properties.
Collapse
Affiliation(s)
- Isidoro Olmeda
- ENOLABInstitut de Biotecnologia i Biomedicina (BioTecMed)Universitat de ValènciaValenciaSpain
| | - Patricia Casino
- Departament de Bioquímica i Biologia MolecularUniversitat de ValènciaValenciaSpain
- Institut de Biotecnologia i Biomedicina (BioTecMed)Universitat de ValènciaValenciaSpain
- Group 739 of the Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBERER) del Instituto de Salud Carlos IIIValenciaSpain
| | - Robert E. Collins
- Office of Educational ProgramsBrookhaven National LaboratoryUptonNY11973USA
- Department of Chemistry and Physical SciencesQuinnipiac UniversityHamdenCT06518USA
| | - Ramón Sendra
- Departament de Bioquímica i Biologia MolecularUniversitat de ValènciaValenciaSpain
| | - Sara Callejón
- ENOLABInstitut de Biotecnologia i Biomedicina (BioTecMed)Universitat de ValènciaValenciaSpain
- Present address:
ENARTIS Wine TechENARTIS SEPSA S.A.U. PolIndustrial AlcesAvda de los vinos, 18Alcázar de San JuanCiudad Real13600Spain
| | - Juanjo Huesa
- Departament de Bioquímica i Biologia MolecularUniversitat de ValènciaValenciaSpain
| | - Alexei S. Soares
- Photon Sciences DirectorateBrookhaven National LaboratoryUptonNY11973USA
| | - Sergi Ferrer
- ENOLABInstitut de Biotecnologia i Biomedicina (BioTecMed)Universitat de ValènciaValenciaSpain
| | - Isabel Pardo
- ENOLABInstitut de Biotecnologia i Biomedicina (BioTecMed)Universitat de ValènciaValenciaSpain
- Departament de Microbiologia i EcologiaUniversitat de ValènciaValenciaSpain
| |
Collapse
|
32
|
Abstract
The sodium pump (Na+, K+-ATPase, NKA) is vital for animal cells, as it actively maintains Na+ and K+ electrochemical gradients across the cell membrane. It is a target of cardiotonic steroids (CTSs) such as ouabain and digoxin. As CTSs are almost unique strong inhibitors specific to NKA, a wide range of derivatives has been developed for potential therapeutic use. Several crystal structures have been published for NKA-CTS complexes, but they fail to explain the largely different inhibitory properties of the various CTSs. For instance, although CTSs are thought to inhibit ATPase activity by binding to NKA in the E2P state, we do not know if large conformational changes accompany binding, as no crystal structure is available for the E2P state free of CTS. Here, we describe crystal structures of the BeF3 - complex of NKA representing the E2P ground state and then eight crystal structures of seven CTSs, including rostafuroxin and istaroxime, two new members under clinical trials, in complex with NKA in the E2P state. The conformations of NKA are virtually identical in all complexes with and without CTSs, showing that CTSs bind to a preformed cavity in NKA. By comparing the inhibitory potency of the CTSs measured under four different conditions, we elucidate how different structural features of the CTSs result in different inhibitory properties. The crystal structures also explain K+-antagonism and suggest a route to isoform specific CTSs.
Collapse
|
33
|
Panda A, Karhadkar S, Acharya B, Banerjee A, De S, Dasgupta S. Enhancement of angiogenin inhibition by polyphenol-capped gold nanoparticles. Biopolymers 2021; 112:e23429. [PMID: 33851721 DOI: 10.1002/bip.23429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/27/2021] [Accepted: 03/22/2021] [Indexed: 11/10/2022]
Abstract
Angiogenin (Ang), is a ribonucleolytic protein that is associated with angiogenesis, the formation of blood vessels. The involvement of Ang in vascularisation makes it a potential target for the identification of compounds that have the potential to inhibit the process. The compounds may be assessed for their ability to inhibit the ribonucleolytic activity of the protein and subsequently blood vessel formation, a crucial requirement for tumor formation. We report an inhibition of the ribonucleolytic activity of Ang with the gallate containing green tea polyphenols, ECG and EGCG that exhibits an increased efficacy upon forming polyphenol-capped gold nanoparticles (ECG-AuNPs and EGCG-AuNPs). The extent of inhibition was confirmed using an agarose gel-based assay followed by fluorescence titration studies that indicated a hundred fold stronger binding of polyphenol-capped gold nanoparticles (GTP-AuNPs) compared to the bare polyphenols. Interestingly, we found a change in the mode of inhibition from a noncompetitive type to a competitive mode of inhibition in case of the GTP-AuNPs, which is in agreement with the 'n' values obtained from the fluorescence quenching studies. The effect on angiogenesis has also been assessed by the chorioallantoic membrane (CAM) assay. We find an increase in the inhibition potency of GTP-AuNPs that could find applications in the development of anti-angiogenic compounds.
Collapse
Affiliation(s)
- Atashi Panda
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Siddhant Karhadkar
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Bidisha Acharya
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Anwesha Banerjee
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Soumya De
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Swagata Dasgupta
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
34
|
Mu W, Kong J, Cao J. Understanding the Optimal Cooperativity of Human Glucokinase: Kinetic Resonance in Nonequilibrium Conformational Fluctuations. J Phys Chem Lett 2021; 12:2900-2904. [PMID: 33724849 DOI: 10.1021/acs.jpclett.1c00438] [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: 06/12/2023]
Abstract
The cooperativity of a monomeric enzyme arises from dynamic correlation instead of spatial correlation and is a consequence of nonequilibrium conformation fluctuations. We investigate the conformation-modulated kinetics of human glucokinase, a monomeric enzyme with important physiological functions, using a five-state kinetic model. We derive the non-Michealis-Menten (MM) correction term of the activity (i.e., turnover rate), predict its relationship to cooperativity, and reveal the violation of conformational detailed balance. Most importantly, we reproduce and explain the observed resonance effect in human glucokinase (i.e., maximal cooperativity when the conformational fluctuation rate is comparable to the catalytic rate). With the realistic parameters, our theoretical results are in quantitative agreement with the reported measurement by Miller and co-workers. The analysis can be extended to a general chemical network beyond the five-state model, suggesting the generality of kinetic cooperativity and resonance.
Collapse
Affiliation(s)
- Weihua Mu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge 02139, U.K
| | - Jing Kong
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge 02139, U.K
| | - Jianshu Cao
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge 02139, U.K
| |
Collapse
|
35
|
Perrin-Cocon L, Vidalain PO, Jacquemin C, Aublin-Gex A, Olmstead K, Panthu B, Rautureau GJP, André P, Nyczka P, Hütt MT, Amoedo N, Rossignol R, Filipp FV, Lotteau V, Diaz O. A hexokinase isoenzyme switch in human liver cancer cells promotes lipogenesis and enhances innate immunity. Commun Biol 2021; 4:217. [PMID: 33594203 PMCID: PMC7886870 DOI: 10.1038/s42003-021-01749-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
During the cancerous transformation of normal hepatocytes into hepatocellular carcinoma (HCC), the enzyme catalyzing the first rate-limiting step of glycolysis, namely the glucokinase (GCK), is replaced by the higher affinity isoenzyme, hexokinase 2 (HK2). Here, we show that in HCC tumors the highest expression level of HK2 is inversely correlated to GCK expression, and is associated to poor prognosis for patient survival. To further explore functional consequences of the GCK-to-HK2 isoenzyme switch occurring during carcinogenesis, HK2 was knocked-out in the HCC cell line Huh7 and replaced by GCK, to generate the Huh7-GCK+/HK2− cell line. HK2 knockdown and GCK expression rewired central carbon metabolism, stimulated mitochondrial respiration and restored essential metabolic functions of normal hepatocytes such as lipogenesis, VLDL secretion, glycogen storage. It also reactivated innate immune responses and sensitivity to natural killer cells, showing that consequences of the HK switch extend beyond metabolic reprogramming. Many cancers fuel their rapid growth by replacing glucokinase with its higher affinity isoenzyme, hexokinase 2 (HK2), making HK2 an attractive drug target. In this study, Perrin-Cocon and Vidalain et al. use CRISPR/Cas-9 gene editing to reverse this enzymatic switch in human liver cancer cells, and find this restores innate immune function as well as reversing cancer-associated metabolic reprogramming.
Collapse
Affiliation(s)
- Laure Perrin-Cocon
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, Lyon, F-69007, France
| | - Pierre-Olivier Vidalain
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, Lyon, F-69007, France
| | - Clémence Jacquemin
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, Lyon, F-69007, France
| | - Anne Aublin-Gex
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, Lyon, F-69007, France
| | - Keedrian Olmstead
- Cancer Systems Biology, Institute for Diabetes and Cancer, Helmholtz Zentrum München, Ingolstädter Landstraße 1, München, D-85764, Germany
| | - Baptiste Panthu
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, Lyon, F-69007, France.,Univ Lyon, CarMeN Laboratory, Inserm, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Hôpital Lyon Sud, Bâtiment CENS ELI-2D, 165 Chemin du grand Revoyet, Pierre-Bénite, F-69310, France
| | - Gilles Jeans Philippe Rautureau
- Université de Lyon, CNRS, Université Claude Bernard Lyon 1, ENS de Lyon, Centre de RMN à Très Hauts Champs (CRMN), FRE 2034, 5 rue de la Doua, Villeurbanne, F-69100, France
| | - Patrice André
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, Lyon, F-69007, France
| | - Piotr Nyczka
- Department of Life Sciences and Chemistry, Jacobs University, Campus Ring 1, Bremen, D-28759, Germany
| | - Marc-Thorsten Hütt
- Department of Life Sciences and Chemistry, Jacobs University, Campus Ring 1, Bremen, D-28759, Germany
| | - Nivea Amoedo
- CELLOMET, Centre de Génomique Fonctionnelle de Bordeaux, 146 Rue Léo Saignat, Bordeaux, F-33000, France
| | - Rodrigue Rossignol
- CELLOMET, Centre de Génomique Fonctionnelle de Bordeaux, 146 Rue Léo Saignat, Bordeaux, F-33000, France.,Univ. Bordeaux, Inserm U1211, MRGM, Centre hospitalier universitaire Pellegrin, place Amélie Raba Léon, Bordeaux, F-33076, France
| | - Fabian Volker Filipp
- Cancer Systems Biology, Institute for Diabetes and Cancer, Helmholtz Zentrum München, Ingolstädter Landstraße 1, München, D-85764, Germany.,School of Life Sciences Weihenstephan, Technical University München, Maximus-von-Imhof-Forum 3, Freising, D-85354, Germany
| | - Vincent Lotteau
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, Lyon, F-69007, France.
| | - Olivier Diaz
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, Lyon, F-69007, France.
| |
Collapse
|
36
|
Srinivasan B. Explicit Treatment of Non-Michaelis-Menten and Atypical Kinetics in Early Drug Discovery*. ChemMedChem 2020; 16:899-918. [PMID: 33231926 DOI: 10.1002/cmdc.202000791] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Indexed: 12/27/2022]
Abstract
Biological systems are highly regulated. They are also highly resistant to sudden perturbations enabling them to maintain the dynamic equilibrium essential to sustain life. This robustness is conferred by regulatory mechanisms that influence the activity of enzymes/proteins within their cellular context to adapt to changing environmental conditions. However, the initial rules governing the study of enzyme kinetics were mostly tested and implemented for cytosolic enzyme systems that were easy to isolate and/or recombinantly express. Moreover, these enzymes lacked complex regulatory modalities. Now, with academic labs and pharmaceutical companies turning their attention to more-complex systems (for instance, multiprotein complexes, oligomeric assemblies, membrane proteins and post-translationally modified proteins), the initial axioms defined by Michaelis-Menten (MM) kinetics are rendered inadequate, and the development of a new kind of kinetic analysis to study these systems is required. This review strives to present an overview of enzyme kinetic mechanisms that are atypical and, oftentimes, do not conform to the classical MM kinetics. Further, it presents initial ideas on the design and analysis of experiments in early drug-discovery for such systems, to enable effective screening and characterisation of small-molecule inhibitors with desirable physiological outcomes.
Collapse
Affiliation(s)
- Bharath Srinivasan
- Mechanistic Biology and Profiling Discovery Sciences, R&D, AstraZeneca, 310, Milton Rd, Milton CB4 0WG, Cambridge, UK
| |
Collapse
|
37
|
Adams NBP, Bisson C, Brindley AA, Farmer DA, Davison PA, Reid JD, Hunter CN. The active site of magnesium chelatase. NATURE PLANTS 2020; 6:1491-1502. [PMID: 33257858 DOI: 10.1038/s41477-020-00806-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/14/2020] [Indexed: 06/12/2023]
Abstract
The insertion of magnesium into protoporphyrin initiates the biosynthesis of chlorophyll, the pigment that underpins photosynthesis. This reaction, catalysed by the magnesium chelatase complex, couples ATP hydrolysis by a ChlID motor complex to chelation within the ChlH subunit. We probed the structure and catalytic function of ChlH using a combination of X-ray crystallography, computational modelling, mutagenesis and enzymology. Two linked domains of ChlH in an initially open conformation of ChlH bind protoporphyrin IX, and the rearrangement of several loops envelops this substrate, forming an active site cavity. This induced fit brings an essential glutamate (E660), proposed to be the key catalytic residue for magnesium insertion, into proximity with the porphyrin. A buried solvent channel adjacent to E660 connects the exterior bulk solvent to the active site, forming a possible conduit for the delivery of magnesium or abstraction of protons.
Collapse
Affiliation(s)
- Nathan B P Adams
- Department of Molecular Biology and Biotechnology, The University of Sheffield, Sheffield, UK.
| | - Claudine Bisson
- Department of Molecular Biology and Biotechnology, The University of Sheffield, Sheffield, UK
- Centre for Ultrastructural Imaging, New Hunt's House, Guy's Campus, King's College London, London, UK
| | - Amanda A Brindley
- Department of Molecular Biology and Biotechnology, The University of Sheffield, Sheffield, UK
| | - David A Farmer
- Department of Molecular Biology and Biotechnology, The University of Sheffield, Sheffield, UK
| | - Paul A Davison
- Department of Molecular Biology and Biotechnology, The University of Sheffield, Sheffield, UK
| | - James D Reid
- Department of Chemistry, The University of Sheffield, Sheffield, UK
| | - C Neil Hunter
- Department of Molecular Biology and Biotechnology, The University of Sheffield, Sheffield, UK.
| |
Collapse
|
38
|
Liu S, Refaei M, Liu S, Decker A, Hinerman JM, Herr AB, Howell M, Musier-Forsyth K, Tsang P. Hairpin RNA-induced conformational change of a eukaryotic-specific lysyl-tRNA synthetase extension and role of adjacent anticodon-binding domain. J Biol Chem 2020; 295:12071-12085. [PMID: 32611767 PMCID: PMC7443506 DOI: 10.1074/jbc.ra120.013852] [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: 04/11/2020] [Revised: 06/26/2020] [Indexed: 11/06/2022] Open
Abstract
Human lysyl-tRNA synthetase (hLysRS) is essential for aminoacylation of tRNALys Higher eukaryotic LysRSs possess an N-terminal extension (Nterm) previously shown to facilitate high-affinity tRNA binding and aminoacylation. This eukaryote-specific appended domain also plays a critical role in hLysRS nuclear localization, thus facilitating noncanonical functions of hLysRS. The structure is intrinsically disordered and therefore remains poorly characterized. Findings of previous studies are consistent with the Nterm domain undergoing a conformational transition to an ordered structure upon nucleic acid binding. In this study, we used NMR to investigate how the type of RNA, as well as the presence of the adjacent anticodon-binding domain (ACB), influences the Nterm conformation. To explore the latter, we used sortase A ligation to produce a segmentally labeled tandem-domain protein, Nterm-ACB. In the absence of RNA, Nterm remained disordered regardless of ACB attachment. Both alone and when attached to ACB, Nterm structure remained unaffected by titration with single-stranded RNAs. The central region of the Nterm domain adopted α-helical structure upon titration of Nterm and Nterm-ACB with RNA hairpins containing double-stranded regions. Nterm binding to the RNA hairpins resulted in CD spectral shifts consistent with an induced helical structure. NMR and fluorescence anisotropy revealed that Nterm binding to hairpin RNAs is weak but that the binding affinity increases significantly upon covalent attachment to ACB. We conclude that the ACB domain facilitates induced-fit conformational changes and confers high-affinity RNA hairpin binding, which may be advantageous for functional interactions of LysRS with a variety of different binding partners.
Collapse
Affiliation(s)
- Sheng Liu
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| | - Maryanne Refaei
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| | - Shuohui Liu
- Department of Chemistry and Biochemistry, Center for RNA Biology, Ohio State University, Columbus, Ohio, USA
| | - Aaron Decker
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jennifer M. Hinerman
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Andrew B. Herr
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Mike Howell
- Protein Express, Inc., Cincinnati, Ohio, USA
| | - Karin Musier-Forsyth
- Department of Chemistry and Biochemistry, Center for RNA Biology, Ohio State University, Columbus, Ohio, USA
| | - Pearl Tsang
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
39
|
Bharmoria P, Correia SFH, Martins M, Hernández-Rodríguez MA, Ventura SPM, Ferreira RAS, Carlos LD, Coutinho JAP. Protein Cohabitation: Improving the Photochemical Stability of R-Phycoerythrin in the Solid State. J Phys Chem Lett 2020; 11:6249-6255. [PMID: 32643938 DOI: 10.1021/acs.jpclett.0c01491] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The poor photochemical stability of R-phycoerythrin (R-PE) has been a bottleneck for its broad-spectrum applications. Inspired by nature, we studied a sustainable strategy of protein cohabitation to enhance R-PE stability by embedding it in a solid matrix of gelatin. Both pure R-PE and fresh phycobiliprotein (PBP) extracts recovered from Gracilaria gracilis were studied. The incorporation of R-PE in the gelatin-based films (gelatin-RPE and gelatin-PBPs) has improved its photochemical stability for at least 8 months, the longest time period reported so far. These results were evidenced by not only absorption but also emission quantum yield measurements (Φ). Moreover, the photostability of gelatin-RPE films upon continuous excitation with an AM1.5G solar simulator was tested and found to remain stable for 23 h after initial decreasing up to 250 min. In the end, another approach was established to allow 100% photostability for a 3 h exposure to an AM1.5G solar simulator by doping the gelatin-based film including R-Phycoerythrin with n-propyl gallate stabilized with Tween 80, allowing their use as naturally based optically active centers in photovoltaic applications.
Collapse
Affiliation(s)
- Pankaj Bharmoria
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Sandra F H Correia
- Phantom-g, CICECO-Aveiro Institute of Materials, Department of Physics, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Margarida Martins
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Miguel A Hernández-Rodríguez
- Phantom-g, CICECO-Aveiro Institute of Materials, Department of Physics, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Sónia P M Ventura
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rute A S Ferreira
- Phantom-g, CICECO-Aveiro Institute of Materials, Department of Physics, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Luís D Carlos
- Phantom-g, CICECO-Aveiro Institute of Materials, Department of Physics, University of Aveiro, 3810-193 Aveiro, Portugal
| | - João A P Coutinho
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
40
|
Viñambres M, Espada M, Martínez AT, Serrano A. Screening and Evaluation of New Hydroxymethylfurfural Oxidases for Furandicarboxylic Acid Production. Appl Environ Microbiol 2020; 86:e00842-20. [PMID: 32503910 PMCID: PMC7414962 DOI: 10.1128/aem.00842-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/31/2020] [Indexed: 11/20/2022] Open
Abstract
The enzymatic production of 2,5-furandicarboxylic acid (FDCA) from 5-hydroxymethylfurfural (HMF) has gained interest in recent years, as FDCA is a renewable precursor of poly(ethylene-2,5-furandicarboxylate) (PEF). 5-Hydroxymethylfurfural oxidases (HMFOs) form a flavoenzyme family with genes annotated in a dozen bacterial species but only one enzyme purified and characterized to date (after heterologous expression of a Methylovorus sp. HMFO gene). This oxidase acts on both furfuryl alcohols and aldehydes and, therefore, is able to catalyze the conversion of HMF into FDCA through 2,5-diformylfuran (DFF) and 2,5-formylfurancarboxylic acid (FFCA), with only the need of oxygen as a cosubstrate. To enlarge the repertoire of HMFO enzymes available, genetic databases were screened for putative HMFO genes, followed by heterologous expression in Escherichia coli After unsuccessful trials with other bacterial HMFO genes, HMFOs from two Pseudomonas species were produced as active soluble enzymes, purified, and characterized. The Methylovorus sp. enzyme was also produced and purified in parallel for comparison. Enzyme stability against temperature, pH, and hydrogen peroxide, three key aspects for application, were evaluated (together with optimal conditions for activity), revealing differences between the three HMFOs. Also, the kinetic parameters for HMF, DFF, and FFCA oxidation were determined, the new HMFOs having higher efficiencies for the oxidation of FFCA, which constitutes the bottleneck in the enzymatic route for FDCA production. These results were used to set up the best conditions for FDCA production by each enzyme, attaining a compromise between optimal activity and half-life under different conditions of operation.IMPORTANCE HMFO is the only enzyme described to date that can catalyze by itself the three consecutive oxidation steps to produce FDCA from HMF. Unfortunately, only one HMFO enzyme is currently available for biotechnological application. This availability is enlarged here by the identification, heterologous production, purification, and characterization of two new HMFOs, one from Pseudomonas nitroreducens and one from an unidentified Pseudomonas species. Compared to the previously known Methylovorus HMFO, the new enzyme from P. nitroreducens exhibits better performance for FDCA production in wider pH and temperature ranges, with higher tolerance for the hydrogen peroxide formed, longer half-life during oxidation, and higher yield and total turnover numbers in long-term conversions under optimized conditions. All these features are relevant properties for the industrial production of FDCA. In summary, gene screening and heterologous expression can facilitate the selection and improvement of HMFO enzymes as biocatalysts for the enzymatic synthesis of renewable building blocks in the production of bioplastics.
Collapse
Affiliation(s)
- Mario Viñambres
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | - Marta Espada
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | - Angel T Martínez
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | - Ana Serrano
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| |
Collapse
|
41
|
Fricke J, Kargbo R, Regestein L, Lenz C, Peschel G, Rosenbaum MA, Sherwood A, Hoffmeister D. Scalable Hybrid Synthetic/Biocatalytic Route to Psilocybin. Chemistry 2020; 26:8281-8285. [PMID: 32101345 PMCID: PMC7383583 DOI: 10.1002/chem.202000134] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Indexed: 01/24/2023]
Abstract
Psilocybin, the principal indole alkaloid of Psilocybe mushrooms, is currently undergoing clinical trials as a medication against treatment-resistant depression and major depressive disorder. The psilocybin supply for pharmaceutical purposes is met by synthetic chemistry. We replaced the problematic phosphorylation step during synthesis with the mushroom kinase PsiK. This enzyme was biochemically characterized and used to produce one gram of psilocybin from psilocin within 20 minutes. We also describe a pilot-scale protocol for recombinant PsiK that yielded 150 mg enzyme in active and soluble form. Our work consolidates the simplicity of tryptamine chemistry with the specificity and selectivity of enzymatic catalysis and helps provide access to an important drug at potentially reasonable cost.
Collapse
Affiliation(s)
- Janis Fricke
- Department Pharmaceutical Microbiology at the Hans-Knöll-InstituteFriedrich-Schiller-UniversitätBeutenbergstrasse 11a07745JenaGermany
| | - Robert Kargbo
- Usona Institute2780 Woods Hollow RoadMadisonWI53711USA
| | - Lars Regestein
- Bio Pilot PlantLeibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-InstituteBeutenbergstrasse 11a07745JenaGermany
| | - Claudius Lenz
- Department Pharmaceutical Microbiology at the Hans-Knöll-InstituteFriedrich-Schiller-UniversitätBeutenbergstrasse 11a07745JenaGermany
| | - Gundela Peschel
- Bio Pilot PlantLeibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-InstituteBeutenbergstrasse 11a07745JenaGermany
| | - Miriam A. Rosenbaum
- Bio Pilot PlantLeibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-InstituteBeutenbergstrasse 11a07745JenaGermany
| | | | - Dirk Hoffmeister
- Department Pharmaceutical Microbiology at the Hans-Knöll-InstituteFriedrich-Schiller-UniversitätBeutenbergstrasse 11a07745JenaGermany
| |
Collapse
|
42
|
2-Aminopyridine Analogs Inhibit Both Enzymes of the Glyoxylate Shunt in Pseudomonas aeruginosa. Int J Mol Sci 2020; 21:ijms21072490. [PMID: 32260167 PMCID: PMC7177833 DOI: 10.3390/ijms21072490] [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: 02/28/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/16/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen responsible for many hospital-acquired infections. P. aeruginosa can thrive in diverse infection scenarios by rewiring its central metabolism. An example of this is the production of biomass from C2 nutrient sources such as acetate via the glyoxylate shunt when glucose is not available. The glyoxylate shunt is comprised of two enzymes, isocitrate lyase (ICL) and malate synthase G (MS), and flux through the shunt is essential for the survival of the organism in mammalian systems. In this study, we characterized the mode of action and cytotoxicity of structural analogs of 2-aminopyridines, which have been identified by earlier work as being inhibitory to both shunt enzymes. Two of these analogs were able to inhibit ICL and MS in vitro and prevented growth of P. aeruginosa on acetate (indicating cell permeability). Moreover, the compounds exerted negligible cytotoxicity against three human cell lines and showed promising in vitro drug metabolism and safety profiles. Isothermal titration calorimetry was used to confirm binding of one of the analogs to ICL and MS, and the mode of enzyme inhibition was determined. Our data suggest that these 2-aminopyridine analogs have potential as anti-pseudomonal agents.
Collapse
|
43
|
Goblirsch BR, Wiener MC. Ste24: An Integral Membrane Protein Zinc Metalloprotease with Provocative Structure and Emergent Biology. J Mol Biol 2020; 432:5079-5090. [PMID: 32199981 PMCID: PMC7172729 DOI: 10.1016/j.jmb.2020.03.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 03/06/2020] [Accepted: 03/12/2020] [Indexed: 01/30/2023]
Abstract
Ste24, an integral membrane protein zinc metalloprotease, is found in every kingdom of eukaryotes. It was discovered approximately 20 years ago by yeast genetic screens identifying it as a factor responsible for processing the yeast mating a-factor pheromone. In animals, Ste24 processes prelamin A, a component of the nuclear lamina; mutations in the human ortholog of Ste24 diminish its activity, giving rise to genetic diseases of accelerated aging (progerias). Additionally, lipodystrophy, acquired from the standard highly active antiretroviral therapy used to treat AIDS patients, likely results from off-target interactions of HIV (aspartyl) protease inhibitor drugs with Ste24. Ste24 possesses a novel “α-barrel” structure, consisting of a ring of seven transmembrane α-helices enclosing a large (> 12,000 Å3) interior volume that contains the active-site and substrate-binding region; this “membrane-interior reaction chamber” is unprecedented in integral membrane protein structures. Additionally, the surface of the membrane-interior reaction chamber possesses a strikingly large negative electrostatic surface potential, adding additional “functional mystery.” Recent publications implicate Ste24 as a key factor in several endoplasmic reticulum processes, including the unfolded protein response, a cellular stress response of the endoplasmic reticulum, and removal of misfolded proteins from the translocon. Ste24, with its provocative structure, enigmatic mechanism, and recently emergent new biological roles including “translocon unclogger” and (non-enyzmatic) broad-spectrum viral restriction factor, presents far differently than before 2016, when it was viewed as a “CAAX protease” responsible for cleavage of prenylated (farnesylated or geranylgeranylated) substrates. The emphasis of this review is on Ste24 of the “Post-CAAX-Protease Era.” Ste24 is a eukaryotic integral membrane protein of novel structure. Ste24 is a gluzincin ZMP whose structure/function relationships are poorly explored. ZMP core, ZMP accessory, and “ɑ-barrel modules form the Ste24 tripartite architecture. Emergent biology of Ste24 includes roles as a translocon unclogger and a viral restriction factor.
Collapse
Affiliation(s)
- Brandon R Goblirsch
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908, USA
| | - Michael C Wiener
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
44
|
Yadav PK, Vitvitsky V, Carballal S, Seravalli J, Banerjee R. Thioredoxin regulates human mercaptopyruvate sulfurtransferase at physiologically-relevant concentrations. J Biol Chem 2020; 295:6299-6311. [PMID: 32179647 DOI: 10.1074/jbc.ra120.012616] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/13/2020] [Indexed: 11/06/2022] Open
Abstract
3-Mercaptopyruvate sulfur transferase (MPST) catalyzes the desulfuration of 3-mercaptopyruvate (3-MP) and transfers sulfane sulfur from an enzyme-bound persulfide intermediate to thiophilic acceptors such as thioredoxin and cysteine. Hydrogen sulfide (H2S), a signaling molecule implicated in many physiological processes, can be released from the persulfide product of the MPST reaction. Two splice variants of MPST, differing by 20 amino acids at the N terminus, give rise to the cytosolic MPST1 and mitochondrial MPST2 isoforms. Here, we characterized the poorly-studied MPST1 variant and demonstrated that substitutions in its Ser-His-Asp triad, proposed to serve a general acid-base role, minimally affect catalytic activity. We estimated the 3-MP concentration in murine liver, kidney, and brain tissues, finding that it ranges from 0.4 μmol·kg-1 in brain to 1.4 μmol·kg-1 in kidney. We also show that N-acetylcysteine, a widely-used antioxidant, is a poor substrate for MPST and is unlikely to function as a thiophilic acceptor. Thioredoxin exhibits substrate inhibition, increasing the KM for 3-MP ∼15-fold compared with other sulfur acceptors. Kinetic simulations at physiologically-relevant substrate concentrations predicted that the proportion of sulfur transfer to thioredoxin increases ∼3.5-fold as its concentration decreases from 10 to 1 μm, whereas the total MPST reaction rate increases ∼7-fold. The simulations also predicted that cysteine is a quantitatively-significant sulfane sulfur acceptor, revealing MPST's potential to generate low-molecular-weight persulfides. We conclude that the MPST1 and MPST2 isoforms are kinetically indistinguishable and that thioredoxin modulates the MPST-catalyzed reaction in a physiologically-relevant concentration range.
Collapse
Affiliation(s)
- Pramod Kumar Yadav
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109-0600
| | - Victor Vitvitsky
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109-0600
| | - Sebastián Carballal
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109-0600.,Departamento de Bioquímica, Facultad de Medicina and Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Javier Seravalli
- Department of Biochemistry and the Redox Biology Center, University of Nebraska, Lincoln, Nebraska 68588
| | - Ruma Banerjee
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109-0600
| |
Collapse
|
45
|
Expression and purification of the 5'-nucleotidase YitU from Bacillus species: its enzymatic properties and possible applications in biotechnology. Appl Microbiol Biotechnol 2020; 104:2957-2972. [PMID: 32040605 PMCID: PMC7062661 DOI: 10.1007/s00253-020-10428-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/17/2020] [Accepted: 02/03/2020] [Indexed: 11/05/2022]
Abstract
5’-Nucleotidases (EC 3.1.3.5) are enzymes that catalyze the hydrolytic dephosphorylation of 5′-ribonucleotides and 5′-deoxyribonucleotides to their corresponding nucleosides plus phosphate. In the present study, to search for new genes encoding 5′-nucleotidases specific for purine nucleotides in industrially important Bacillus species, “shotgun” cloning and the direct selection of recombinant clones grown in purine nucleosides at inhibitory concentrations were performed in the Escherichia coli GS72 strain, which is sensitive to these compounds. As a result, orthologous yitU genes from Bacillus subtilis and Bacillus amyloliquefaciens, whose products belong to the ubiquitous haloacid dehalogenase superfamily (HADSF), were selected and found to have a high sequence similarity of 87%. B. subtilis YitU was produced in E. coli as an N-terminal hexahistidine-tagged protein, purified and biochemically characterized as a soluble 5′-nucleotidase with broad substrate specificity with respect to various deoxyribo- and ribonucleoside monophosphates: dAMP, GMP, dGMP, CMP, AMP, XMP, IMP and 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranosyl 5′-monophosphate (AICAR-P). However, the preferred substrate for recombinant YitU was shown to be flavin mononucleotide (FMN). B. subtilis and B. amyloliquefaciens yitU overexpression increased riboflavin (RF) and 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) accumulation and can be applied to breed highly performing RF- and AICAR-producing strains.
Collapse
|
46
|
Sternisha SM, Whittington AC, Martinez Fiesco JA, Porter C, McCray MM, Logan T, Olivieri C, Veglia G, Steinbach PJ, Miller BG. Nanosecond-Timescale Dynamics and Conformational Heterogeneity in Human GCK Regulation and Disease. Biophys J 2020; 118:1109-1118. [PMID: 32023434 DOI: 10.1016/j.bpj.2019.12.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/11/2019] [Accepted: 12/19/2019] [Indexed: 11/25/2022] Open
Abstract
Human glucokinase (GCK) is the prototypic example of an emerging class of proteins with allosteric-like behavior that originates from intrinsic polypeptide dynamics. High-resolution NMR investigations of GCK have elucidated millisecond-timescale dynamics underlying allostery. In contrast, faster motions have remained underexplored, hindering the development of a comprehensive model of cooperativity. Here, we map nanosecond-timescale dynamics and structural heterogeneity in GCK using a combination of unnatural amino acid incorporation, time-resolved fluorescence, and 19F nuclear magnetic resonance spectroscopy. We find that a probe inserted within the enzyme's intrinsically disordered loop samples multiple conformations in the unliganded state. Glucose binding and disease-associated mutations that suppress cooperativity alter the number and/or relative population of these states. Together, the nanosecond kinetics characterized here and the millisecond motions known to be essential for cooperativity provide a dynamical framework with which we address the origins of cooperativity and the mechanism of activated, hyperinsulinemia-associated, noncooperative variants.
Collapse
Affiliation(s)
- Shawn M Sternisha
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida
| | - A Carl Whittington
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida; Department of Biological Science, Florida State University, Tallahassee, Florida
| | | | - Carol Porter
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida
| | - Malcolm M McCray
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida
| | - Timothy Logan
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida; Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida
| | - Cristina Olivieri
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Gianluigi Veglia
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota; Department of Chemistry, University of Minnesota, Minneapolis, Minnesota
| | - Peter J Steinbach
- Center for Molecular Modeling, Center for Information Technology, National Institutes of Health, Bethesda, Maryland.
| | - Brian G Miller
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida.
| |
Collapse
|
47
|
Bocedi A, Cattani G, Gambardella G, Ticconi S, Cozzolino F, Di Fusco O, Pucci P, Ricci G. Ultra-Rapid Glutathionylation of Ribonuclease: Is this the Real Incipit of its Oxidative Folding? Int J Mol Sci 2019; 20:ijms20215440. [PMID: 31683668 PMCID: PMC6862303 DOI: 10.3390/ijms20215440] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 11/16/2022] Open
Abstract
Many details of oxidative folding of proteins remain obscure, in particular, the role of oxidized glutathione (GSSG). This study reveals some unknown aspects. When a reduced ribonuclease A refolds in the presence of GSSG, most of its eight cysteines accomplish a very fast glutathionylation. In particular, one single cysteine, identified as Cys95 by mass spectrometry, displays 3600 times higher reactivity when compared with an unperturbed protein cysteine. Furthermore, the other five cysteines show 40-50 times higher reactivity toward GSSG. This phenomenon is partially due to a low pKa value of most of these cysteines (average pKa = 7.9), but the occurrence of a reversible GSSG-ribonuclease complex (KD = 0.12 mM) is reasonably responsible for the extraordinary hyper-reactivity of Cys95. Neither hyper-reactivity nor some protein-disulfide complexes have been found by reacting a reduced ribonuclease with other natural disulfides i.e., cystine, cystamine, and homocystine. Hyper-reactivity of all cysteines was observed toward 5,5'-dithiobis-(2-nitrobenzoic acid). Given that GSSG is present in high concentrations in the endoplasmic reticulum, this property may shed light on the early step of its oxidative folding. The ultra-rapid glutathionylation of cysteines, only devoted to form disulfides, is a novel property of the molten globule status of the ribonuclease.
Collapse
Affiliation(s)
- Alessio Bocedi
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Giada Cattani
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Giorgia Gambardella
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Silvia Ticconi
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Flora Cozzolino
- CEINGE Biotecnologie Avanzate and Department of Chemical Science, University of Naples "Federico II", 80126 Naples, Italy.
| | - Ornella Di Fusco
- CEINGE Biotecnologie Avanzate and Department of Chemical Science, University of Naples "Federico II", 80126 Naples, Italy.
| | - Piero Pucci
- CEINGE Biotecnologie Avanzate and Department of Chemical Science, University of Naples "Federico II", 80126 Naples, Italy.
| | - Giorgio Ricci
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133 Rome, Italy.
| |
Collapse
|
48
|
Khanppnavar B, Chatterjee R, Choudhury GB, Datta S. Genome-wide survey and crystallographic analysis suggests a role for both horizontal gene transfer and duplication in pantothenate biosynthesis pathways. Biochim Biophys Acta Gen Subj 2019; 1863:1547-1559. [DOI: 10.1016/j.bbagen.2019.05.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 05/21/2019] [Accepted: 05/24/2019] [Indexed: 01/13/2023]
|
49
|
Munshi S, Gopi S, Subramanian S, Campos LA, Naganathan AN. Protein plasticity driven by disorder and collapse governs the heterogeneous binding of CytR to DNA. Nucleic Acids Res 2019. [PMID: 29538715 PMCID: PMC5934615 DOI: 10.1093/nar/gky176] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The amplitude of thermodynamic fluctuations in biological macromolecules determines their conformational behavior, dimensions, nature of phase transitions and effectively their specificity and affinity, thus contributing to fine-tuned molecular recognition. Unique among large-scale conformational changes in proteins are temperature-induced collapse transitions in intrinsically disordered proteins (IDPs). Here, we show that CytR DNA-binding domain, an IDP that folds on binding DNA, undergoes a coil-to-globule transition with temperature in the absence of DNA while exhibiting energetically decoupled local and global structural rearrangements, and maximal thermodynamic fluctuations at the optimal bacterial growth temperature. The collapse is shown to be a continuous transition through a combination of statistical-mechanical modeling and all-atom implicit solvent simulations. Surprisingly, CytR binds single-site cognate DNA with negative cooperativity, described by Hill coefficients less than one, resulting in a graded binding response. We show that heterogeneity arising from varying binding-competent CytR conformations or orientations at the single-molecular level contributes to negative binding cooperativity at the level of bulk measurements due to the conflicting requirements of collapse transition, large fluctuations and folding-upon-binding. Our work reports strong evidence for functionally driven thermodynamic fluctuations in determining the extent of collapse and disorder with implications in protein search efficiency of target DNA sites and regulation.
Collapse
Affiliation(s)
- Sneha Munshi
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Soundhararajan Gopi
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Sandhyaa Subramanian
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Luis A Campos
- National Biotechnology Center, Consejo Superior de Investigaciones Científicas, Darwin 3, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Athi N Naganathan
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| |
Collapse
|
50
|
Pectin hydrolysis in cashew apple juice by Aspergillus aculeatus URM4953 polygalacturonase covalently-immobilized on calcium alginate beads: A kinetic and thermodynamic study. Int J Biol Macromol 2019; 126:820-827. [DOI: 10.1016/j.ijbiomac.2018.12.236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/28/2018] [Accepted: 12/25/2018] [Indexed: 12/17/2022]
|