5,7,8,10,13,15,17,20,21 Recognition of Substrates by Enzymes

Total Page:16

File Type:pdf, Size:1020Kb

5,7,8,10,13,15,17,20,21 Recognition of Substrates by Enzymes Recommended problems from chapter 15: 5,7,8,10,13,15,17,20,21 Recognition of substrates by enzymes See chemical mechanisms notes on web page, covered in chapter 14 in your textbook. Also see chapter 13 for discussion of the “lock and key” vs. the “induced fit” substrate binding modes. 2 hypotheses on how substrates fit within the active site of enzymes: 1. “Lock and key” •Exact fit between substrate and enzyme 2. “Induced fit” •Enzyme active site is “flexible” or “dynamic” and can assume distinct (but probably related) conformations. A corollary is that it can probably accommodate distinct (but probably somewhat similar) substrates. • “Good” substrates can fit into the active site in such a way that the transition state is approximated. 1 General considerations in the regulation of enzymes 1. Genetic control. At this level of regulation the amount of the enzyme is changed in response to environmental conditions. For example, in response to an abundance of glucose, the production of metabolic enzymes that cleave large polysaccharides stops. In the case of lac repressor, this regulation is on the transcription level. 2. Control of enzymatic activity. At this level the amount of an enzyme is not varied. Rather, the activity of the enzyme is regulated (up or down). A. For S ↔ P, as the amount of product builds up the reverse reaction becomes more pronounced, until equilibrium is reached, where there is no net change in the amounts of product and substrate. B. Enzymatic rates can be modulated by the amount of available substrates and their Km values. C. Covalent modification can alter the affinity of enzyme to substrate (alter the Km). Phosphorylation and de-phosphorylation are such control systems. Nobel prize in 1992 was awarded to Edmond Fischer and Edwin Krebs for this discovery. D. Allosteric effectors are molecules that alter the affinity of an enzyme to a substrate. These are non-covalent modifications. Regulation of enzyme activity by phosphorylation Phosphorylation can occur on serine, threonine, or tyrosine residues. What are the predicted physical consequences of this modification on an enzyme active site? 2 Aspartate transcarbamoylase as a model system of allosteric control The reaction catalyzed by ATCase is diagrammed below. This is one of the initial reactions in pyrimidine synthesis. O O- C NH2 CH2 + C C 2- O OPO3 + - H3N COO H Carbamoyl phosphate Aspartic acid aspartate transcarbamoylase O O- C NH CH - 2 2 + H2PO4 C C O N COO- H H N-Carbamoyl aspartate Feedback inhibition of ATCase by CTP 3 Allosteric models •Allosteric interactions affect the activity of an enzyme by binding at a site distinct from the active site (allos = different; stereo = place or solid). •Allosteric effectors can be the substrate(s); this is a homotropic effect. •The effector(s) can be distinct from the substrate(s) molecules; this is a heterotropic effect. •An allosteric effector can be positive (increases enzymatic activity) or negative (decreases enzymatic activity). Often both effects are found for a given enzyme. For example, in the hemoglobin system: O2 is a homotropic positive effector (increases affinity for oxygen at the oxygen binding site). Bisphosphoglycerate (BPG) and CO2 are negative heterotropic effectors (decrease affinity for oxygen). In general, allosteric effects result from interactions among subunits of oligomeric proteins. It is possible, however, that single-subunit proteins can exhibit allosteric effects if there are multiple binding sites for substrates within the single polypeptide. The Monod, Wyman, and Changeux allosteric model: cooperative binding Consider a protein that may assume two states. The “taut” state binds substrate very poorly, whereas the “relaxed” state has a high affinity for substrate. The subscript 0 denotes an unbound protein, and these two states exist in equilibrium, such that: R0 ↔ T0 Further assume that the equilibrium of R0 and T0 lies substantially to the right, such that in the absence of substrate there is a lot more taut form than relaxed. If L is the association equilibrium constant, L = [T0] / [R0] and L is large. If KR is the equilibrium dissociation constant of substrate from the relaxed form, and KT is the equilibrium dissociation constant of substrate from the taut form, then by the conditions above KR << KT. The extreme case is KR / KT = 0, and in this case there is no affinity of T0 to substrate. 4 The Monod, Wyman, and Changeux allosteric model: cooperative binding R0 ↔ T0 If substrate is added to this system, the equilibrium is perturbed. R0 binds the substrate and no longer exchanged with T0. Now the reaction above goes to the left, more R0 is made, and, in turn, more substrate is bound by R0, and so on. This is a positive homotropic effect. The Monod, Wyman, and Changeux allosteric model: cooperative binding c = KR / KT ; L = [T0] / [R0]; n = number of substrate binding sites; Y = [filled substrate binding sites] / [total number of substrate binding sites]. This substrate binding behavior is cooperative. Filling of a binding site alters the apparent overall affinity for substrate. All binding sites, however, are equivalent. 5 The Monod, Wyman, and Changeux allosteric model; positive heterotropic effects Each subunit has an allostery site and a substrate binding site. If an allosteric activator (A) is present, then the R0 ↔ T0 reaction goes to the left, and there are more substrate binding sites available. This equilibrium shift results in an increase of the apparent (measured) affinity for substrate at a fixed activator concentration. Overall, the cooperativity of substrate binding decreases, because there are more substrate binding sites available. The allosteric activator has the same effect as a lower equilibrium constant L. The Monod, Wyman, and Changeux allosteric model; negative heterotropic effects In the presence of inhibitor, the equilibrium R0 ↔ T0 goes to the right. This reduces the number of substrate binding sites available, and lowers the apparent affinity for substrate. In the presence of a fixed concentration of inhibitor, a titration of substrate would show an increase in the cooperativity of binding, but a decrease in the apparent affinity. 6 The Monod, Wyman, and Changeux allosteric model; heterotropic effects Plot of v0 versus [Aspartate] for the ATCase reaction Is ATP an inhibitor or activator? How does it affect cooperativity of aspartate binding? Is UTP an inhibitor or activator? How does it affect cooperativity of aspartate binding? Does this make sense if you consider the role of ATCase in pyrimidine synthesis? 7 Structure of E. coli ATCase •A view along the 3-fold axis of the 300 kDa protein complex. Composition is c6r6. •The 6 catalytic subunits are in red and blue, and consist of 2 trimers stacked atop each other. •The 6 regulatory subunits are in yellow, there are 3 dimers that bind the two trimers. •Each trimer has catalytic activity that is higher than the catalytic activity of the complex. The trimer is neither affected by ATP nor CTP. What does this argue about the role of the regulatory subunits? Where do you expect the regulatory allostery effectors to bind? Structure of E. coli ATCase The T-state conformation The R-state conformation A view perpendicular to the previous slide, along the molecular two-fold symmetry axis. Given the information about ATCase in the previous slides, what reagent would you add to the protein in order to obtain these two distinct structure? 8 Structure of E. coli ATCase The T-state conformation The R-state conformation O O PALA is a compound that C C PO 2- Carbamoyl binds the enzyme but 3 H N C O PO 2- H2 2 3 phosphate does not react. NH + NH3 Predict to which -OOC C C COO- - - H2 H OOC C C COO Asp artate conformation PALA H2 H would bind. N-(Phosphonaetyl)-L-aspartate (PAL A) The Koshland, Nemethy, and Filmer model, AKA the sequential model •The symmetry model assumes a constant, fixed substrate binding site (Emil Fischer’s “lock and key” model). •In contrast, the sequential model allows for changed in the conformation of the other binding sites as a result of substrate binding at a particular site on the oligomeric protein. The binding sites are NOT always equivalent. •This means that substrate binding sites are “coupled,” such that filling one binding site changes the other binding sites on the oligomeric protein. •The degree of “coupling” among the subunits can vary. At the extreme, when the subunits are completely coupled, the sequential model actually resembles the symmetry model. 9 The symmetry model of allostery •A tetramer protein is represented. •All the subunits of a given protein can be either in the T or the R form. •No combinations of R and T subunits are allowed within the protein. •The T and R states are at equilibrium regardless of the number of substrates bound. The sequential model of allostery •Binding of S to one subunit of the enzyme can induce a conformational change in the other subunits of the enzyme. •The greatest change occurs at the subunits that have bound ligand. •The symmetry of the protein subunits is not preserved as was the case with the symmetry model. •At the heart of this model are the distinct conformations that the subunits assume as a consequence of substrate binding. 10 Hemoglobin as a model system for allosteric regulation Hemoglobin and myoglobin serve similar functions. They bind O2 and deliver it as needed. Hemoglobin is found in blood, whereas myoglobin is found in muscle. Each polypeptide binds a single heme group, a porphyrin ring structure that binds Fe2+. The porphyrin ring system also binds O2. Oxygen binding by myoglobin If the heme group contains Fe2+, oxygen binding is efficient. In contrast, if the heme group contains Fe3+, oxygen binding is poor.
Recommended publications
  • Allosteric Regulation in Drug Design
    Mini Review Curr Trends Biomedical Eng & Biosci Volume 4 Issue 1 - May 2017 Copyright © All rights are reserved by Ashfaq Ur Rehman DOI: 10.19080/CTBEB.2017.04.5555630 Allosteric regulation in drug design Ashfaq Ur Rehman1,2*, Shah Saud3, Nasir Ahmad4, Abdul Wadood2 and R Hamid5 1State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, China 2Department of Biochemistry, Abdul Wali Khan University Mardan, Pakistan 3Laboratory of Analytical Biochemistry and Bio separation, Shanghai Jiao Tong University, China 4Department of Chemistry, Islama College University Peshawar, Pakistan 5Department of Bioinformatics, Muhammad Ali Jinnah University Islamabad, Pakistan Submission: May 02, 2017; Published: May 23, 2017 *Corresponding author: Ashfaq Ur Rehman, State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China, Tel: ; Fax: 86-21-34204348; Email: Abstract mechanism, which are initiated through attachment of ligand or inhibitors with the protein or enzymes other than active (orthosteric) sites. ThisProtein mini review and enzymes involved play mechanism, significant types roles and in importancebiological processes of allosteric of all regulations living organisms; in drug theirdesign functions process. are regulated through allosteric Keywords: Allosteric, Activator: Drug design Introduction and ultimately cause disease. While various biological processes expressed the control at different points in life time of protein function is pivotal. As all the cell processes are under carful For the survival of all organisms the significance of protein included regulation of gene expression, translation into protein control and if not properly controls this leads to the abnormality through control of activity and at last degradation of protein [1].
    [Show full text]
  • Nucleosome-Mediated Cooperativity Between Transcription Factors
    Nucleosome-mediated cooperativity between transcription factors Leonid A. Mirny1 Harvard–MIT Division of Health Sciences and Technology, and Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139 Edited* by José N. Onuchic, University of California San Diego, La Jolla, CA, and approved October 12, 2010 (received for review December 3, 2009) Cooperative binding of transcription factors (TFs) to promoters and A B other regulatory regions is essential for precise gene expression. The classical model of cooperativity requires direct interactions between TFs, thus constraining the arrangement of TF sites in reg- ulatory regions. Recent genomic and functional studies, however, demonstrate a great deal of flexibility in such arrangements with variable distances, numbers of sites, and identities of TF sites lo- cated in cis-regulatory regions. Such flexibility is inconsistent with L C N O D T R cooperativity by direct interactions between TFs. Here, we demon- P 0 0 0 0 P O O strate that strong cooperativity among noninteracting TFs can be K K 2 2 NN O O T R achieved by their competition with nucleosomes. We find that the P 1 1 P 1 1 K O O mechanism of nucleosome-mediated cooperativity is analogous to c = O 10 1..10 3 2 2 K cooperativity in another multimolecular complex: hemoglobin. This N N2 O2 T R [P] P 2 2 = 15 P surprising analogy provides deep insights, with parallels between O2 O2 KO … … the heterotropic regulation of hemoglobin (e.g., the Bohr effect) [N ] L = 0 100 1000 N O and the roles of nucleosome-positioning sequences and chromatin [O0] n n T4 R4 modifications in gene expression.
    [Show full text]
  • Tyrosine Kinase – Role and Significance in Cancer
    Int. J. Med. Sci. 2004 1(2): 101-115 101 International Journal of Medical Sciences ISSN 1449-1907 www.medsci.org 2004 1(2):101-115 ©2004 Ivyspring International Publisher. All rights reserved Review Tyrosine kinase – Role and significance in Cancer Received: 2004.3.30 Accepted: 2004.5.15 Manash K. Paul and Anup K. Mukhopadhyay Published:2004.6.01 Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S Nagar, Mohali, Punjab, India-160062 Abstract Tyrosine kinases are important mediators of the signaling cascade, determining key roles in diverse biological processes like growth, differentiation, metabolism and apoptosis in response to external and internal stimuli. Recent advances have implicated the role of tyrosine kinases in the pathophysiology of cancer. Though their activity is tightly regulated in normal cells, they may acquire transforming functions due to mutation(s), overexpression and autocrine paracrine stimulation, leading to malignancy. Constitutive oncogenic activation in cancer cells can be blocked by selective tyrosine kinase inhibitors and thus considered as a promising approach for innovative genome based therapeutics. The modes of oncogenic activation and the different approaches for tyrosine kinase inhibition, like small molecule inhibitors, monoclonal antibodies, heat shock proteins, immunoconjugates, antisense and peptide drugs are reviewed in light of the important molecules. As angiogenesis is a major event in cancer growth and proliferation, tyrosine kinase inhibitors as a target for anti-angiogenesis can be aptly applied as a new mode of cancer therapy. The review concludes with a discussion on the application of modern techniques and knowledge of the kinome as means to gear up the tyrosine kinase drug discovery process.
    [Show full text]
  • DNA Breakpoint Assay Reveals a Majority of Gross Duplications Occur in Tandem Reducing VUS Classifications in Breast Cancer Predisposition Genes
    © American College of Medical Genetics and Genomics ARTICLE Corrected: Correction DNA breakpoint assay reveals a majority of gross duplications occur in tandem reducing VUS classifications in breast cancer predisposition genes Marcy E. Richardson, PhD1, Hansook Chong, PhD1, Wenbo Mu, MS1, Blair R. Conner, MS1, Vickie Hsuan, MS1, Sara Willett, MS1, Stephanie Lam, MS1, Pei Tsai, CGMBS, MB (ASCP)1, Tina Pesaran, MS, CGC1, Adam C. Chamberlin, PhD1, Min-Sun Park, PhD1, Phillip Gray, PhD1, Rachid Karam, MD, PhD1 and Aaron Elliott, PhD1 Purpose: Gross duplications are ambiguous in terms of clinical cohort, while the remainder have unknown tandem status. Among interpretation due to the limitations of the detection methods that the tandem gross duplications that were eligible for reclassification, cannot infer their context, namely, whether they occur in tandem or 95% of them were upgraded to pathogenic. are duplicated and inserted elsewhere in the genome. We Conclusion: DBA is a novel, high-throughput, NGS-based method investigated the proportion of gross duplications occurring in that informs the tandem status, and thereby the classification of, tandem in breast cancer predisposition genes with the intent of gross duplications. This method revealed that most gross duplica- informing their classifications. tions in the investigated genes occurred in tandem and resulted in a Methods: The DNA breakpoint assay (DBA) is a custom, paired- pathogenic classification, which helps to secure the necessary end, next-generation sequencing (NGS) method designed to treatment options for their carriers. capture and detect deep-intronic DNA breakpoints in gross duplications in BRCA1, BRCA2, ATM, CDH1, PALB2, and CHEK2. Genetics in Medicine (2019) 21:683–693; https://doi.org/10.1038/s41436- Results: DBA allowed us to ascertain breakpoints for 44 unique 018-0092-7 gross duplications from 147 probands.
    [Show full text]
  • Pi-Pi Stacking Mediated Cooperative Mechanism for Human Cytochrome P450 3A4
    Molecules 2015, 20, 7558-7573; doi:10.3390/molecules20057558 OPEN ACCESS molecules ISSN 1420-3049 www.mdpi.com/journal/molecules Article Pi-pi Stacking Mediated Cooperative Mechanism for Human Cytochrome P450 3A4 Botao Fa 1, Shan Cong 2 and Jingfang Wang 1,* 1 Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China; E-Mail: [email protected] 2 Department of Bioinformatics and Biostatistics, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; E-Mail: [email protected] * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +86-21-3420-7344 (ext. 123); Fax: +86-21-3420-4348. Academic Editor: Antonio Frontera Received: 13 January 2015 / Accepted: 19 March 2015 / Published: 24 April 2015 Abstract: Human Cytochrome P450 3A4 (CYP3A4) is an important member of the cytochrome P450 superfamily with responsibility for metabolizing ~50% of clinical drugs. Experimental evidence showed that CYP3A4 can adopt multiple substrates in its active site to form a cooperative binding model, accelerating substrate metabolism efficiency. In the current study, we constructed both normal and cooperative binding models of human CYP3A4 with antifungal drug ketoconazoles (KLN). Molecular dynamics simulation and free energy calculation were then carried out to study the cooperative binding mechanism. Our simulation showed that the second KLN in the cooperative binding model had a positive impact on the first one binding in the active site by two significant pi-pi stacking interactions. The first one was formed by Phe215, functioning to position the first KLN in a favorable orientation in the active site for further metabolism reactions.
    [Show full text]
  • Allosteric Effects and Cooperative Binding
    Biochemistry I, Fall Term Lecture 13 Sept 28, 2005 Lecture 13: Allosteric Effects and Cooperative Binding Assigned reading in Campbell: Chapter 4.7, 7.2 Key Terms: • Homotropic Allosteric effects • Heterotropic Allosteric effects • T and R states of cooperative systems • Role of proximal His residue in cooperativity of O2 binding by Hb • Regulation of O2 binding to Hb by bis-phosphoglycerate (BPG) Oxygen Binding to Myoglobin and Hemolobin: Myoglobin binds one O2: Hemoglobin binds four O2: Allosteric Effects and Cooperativity: Allosteric effects occur when the binding properties of a macromolecule change as a consequence of a second ligand binding to the macromolecule and altering its affinity towards the first, or primary, ligand. There need not be a direct connection between the two ligands (i.e. they may bind to opposite sides of the protein, or even to different subunits) • If the two ligands are the same (e.g. oxygen) then this is called a homo-tropic allosteric effect. • If the two ligands are different (e.g. oxygen and BPG), then this is called a hetero-tropic allosteric effect. In the case of macromolecules that have multiple ligand binding sites (e.g. Hb), allosteric effects can generate cooperative behavior. Allosteric effects are important in the regulation of enzymatic reactions. Both allosteric activators (which enhance activity) and allosteric inhibitors (which reduce activity) are utilized to control enzyme reactions. Allosteric effects require the presence of two forms of the macromolecule. One form, usually called the T or tense state, binds the primary ligand (e.g. oxygen) with low affinity. The other form, usually called the R or relaxed state, binds ligand with high affinity.
    [Show full text]
  • Integrated Computational Approaches and Tools for Allosteric Drug Discovery
    International Journal of Molecular Sciences Review Integrated Computational Approaches and Tools for Allosteric Drug Discovery Olivier Sheik Amamuddy 1 , Wayde Veldman 1 , Colleen Manyumwa 1 , Afrah Khairallah 1 , Steve Agajanian 2, Odeyemi Oluyemi 2, Gennady M. Verkhivker 2,3,* and Özlem Tastan Bishop 1,* 1 Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, Grahamstown 6140, South Africa; [email protected] (O.S.A.); [email protected] (W.V.); [email protected] (C.M.); [email protected] (A.K.) 2 Graduate Program in Computational and Data Sciences, Keck Center for Science and Engineering, Schmid College of Science and Technology, Chapman University, One University Drive, Orange, CA 92866, USA; [email protected] (S.A.); [email protected] (O.O.) 3 Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA * Correspondence: [email protected] (G.M.V.); [email protected] (Ö.T.B.); Tel.: +714-516-4586 (G.M.V.); +27-46-603-8072 (Ö.T.B.) Received: 25 December 2019; Accepted: 21 January 2020; Published: 28 January 2020 Abstract: Understanding molecular mechanisms underlying the complexity of allosteric regulation in proteins has attracted considerable attention in drug discovery due to the benefits and versatility of allosteric modulators in providing desirable selectivity against protein targets while minimizing toxicity and other side effects. The proliferation of novel computational approaches for predicting ligand–protein interactions and binding using dynamic and network-centric perspectives has led to new insights into allosteric mechanisms and facilitated computer-based discovery of allosteric drugs. Although no absolute method of experimental and in silico allosteric drug/site discovery exists, current methods are still being improved.
    [Show full text]
  • Mathematical Toolkit for Quantitative Analysis of Cooperative Binding of Two Or More Ligands to a Substrate Jacob Peacock and James B
    Mathematical toolkit for quantitative analysis of cooperative binding of two or more ligands to a substrate Jacob Peacock and James B. Jaynes Dept. of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia PA 19107 United States of America [email protected] Abstract We derive mathematical expressions for quantitative analysis of cooperative binding covering the following cases: 1) a single ligand binds to either two non-equivalent sites, or an arbitrary number of equivalent sites, on a substrate (Fig. 1), and 2) two different ligands bind distinct sites on a substrate (Figs. 2, 3). We show how to analyze "competition experiments" using non-linear regression, where a ligand binds to a single site on a labeled substrate in the presence of increasing amounts of identical but unlabeled competitor substrate, to simultaneously determine the Kd and active ligand concentration (Fig. 4A). We compare the performance of this competition method with the commonly used saturation binding method (Fig. 4B). We also provide methods to analyze such experiments that include a second competitor substrate with non-specific binding sites. We show how to build on results from single-ligand competition experiments to fully characterize cooperative binding in systems with two distinct ligands and binding sites (Fig. 4C and Section 5). We generalize the methodology to more than two cooperating ligands, such as an array of DNA binding proteins (Fig. 6). See Peacock and Jaynes [1] for discussion of the various ways these tools can be used, and results using them. • Visualize characteristics and limitations of Hill plots applied to more realistic binding models than those described by the Hill equation, which implies multiple simultaneous ligand binding.
    [Show full text]
  • Tyrosine Kinases
    KEVANM SHOKAT MINIREVIEW Tyrosine kinases: modular signaling enzymes with tunable specificities Cytoplasmic tyrosine kinases are composed of modular domains; one (SHl) has catalytic activity, the other two (SH2 and SH3) do not. Kinase specificity is largely determined by the binding preferences of the SH2 domain. Attaching the SHl domain to a new SH2 domain, via protein-protein association or mutation, can thus dramatically change kinase function. Chemistry & Biology August 1995, 2:509-514 Protein kinases are one of the largest protein families identified, This is a result of the overlapping substrate identified to date; over 45 new members are identified specificities of many tyrosine kinases, which makes it each year. It is estimated that up to 4 % of vertebrate pro- difficult to dissect the individual signaling pathways by teins are protein kinases [l].The protein kinases are cate- scanning for unique target motifs [2]. gorized by their specificity for serineithreonine, tyrosine, or histidine residues. Protein tyrosine kinases account for The apparent promiscuity of individual tyrosine kinases roughly half of all kinases. They occur as membrane- is a result of their unique structural organization. bound receptors or cytoplasmic proteins and are involved Enzyme specificity is typically programmed by one in a wide variety of cellular functions, including cytokine binding site, which recognizes the substrate and also con- responses, antigen-dependent immune responses, cellular tains exquisitely oriented active-site functional groups transformation by RNA viruses, oncogenesis, regulation that help to lower the energy of the transition state for of the cell cycle, and modification of cell morphology the conversion of specific substrates to products.Tyrosine (Fig.
    [Show full text]
  • Structure and Evolution of Protein Allosteric Sites
    Structure and evolution of protein allosteric sites by Alejandro Panjkovich Thesis submitted to Universitat Aut`onoma de Barcelona in partial fulfillment of the requirements for the degree of Doctor of Philosophy Director - Prof. Xavier Daura Tesi Doctoral UAB/ANY 2013 Ph.D. Program - Protein Structure and Function Institut de Biotecnologia i Biomedicina caminante, no hay camino, se hace camino al andar Antonio Machado,1912 Acknowledgements First of all I would like to thank my supervisor and mentor Prof. Xavier Daura for his consistent support and trust in my work throughout these years. Xavi, I deeply appreciate the freedom you gave me to develop this project while you were still carefully aware of the small details. Working under your supervision has been a rich and fulfilling experience. Of course, thanks go as well to current and past members of our institute, especially Rita Rocha, Pau Marc Mu˜noz,Oscar Conchillo, Dr. Mart´ınIndarte, Dr. Mario Ferrer, Prof. Isidre Gibert, Dr. Roman Affentranger and Dr. Juan Cedano for their technical and sometimes philo- sophical assistance. Help from the administrative staff was also significant, I would like to thank in particular Eva, Alicia and Miguel who where always ready to help me in sorting out unexpected bureaucratic affairs. I would also like to thank Dr. Mallur Srivatasan Madhusudhan and his group (especially Kuan Pern Tan, Dr. Minh Nguyen and Binh Nguyen), and also Dr. Gloria Fuentes, Cassio Fernandes, Youssef Zaki, Thijs Kooi, Rama Iyer, Christine Low and many others at the Bioinformatics Institute BII - A∗STAR in Singapore for the many interesting discussions and support during my stage over there.
    [Show full text]
  • Understanding Drug-Drug Interactions Due to Mechanism-Based Inhibition in Clinical Practice
    pharmaceutics Review Mechanisms of CYP450 Inhibition: Understanding Drug-Drug Interactions Due to Mechanism-Based Inhibition in Clinical Practice Malavika Deodhar 1, Sweilem B Al Rihani 1 , Meghan J. Arwood 1, Lucy Darakjian 1, Pamela Dow 1 , Jacques Turgeon 1,2 and Veronique Michaud 1,2,* 1 Tabula Rasa HealthCare Precision Pharmacotherapy Research and Development Institute, Orlando, FL 32827, USA; [email protected] (M.D.); [email protected] (S.B.A.R.); [email protected] (M.J.A.); [email protected] (L.D.); [email protected] (P.D.); [email protected] (J.T.) 2 Faculty of Pharmacy, Université de Montréal, Montreal, QC H3C 3J7, Canada * Correspondence: [email protected]; Tel.: +1-856-938-8697 Received: 5 August 2020; Accepted: 31 August 2020; Published: 4 September 2020 Abstract: In an ageing society, polypharmacy has become a major public health and economic issue. Overuse of medications, especially in patients with chronic diseases, carries major health risks. One common consequence of polypharmacy is the increased emergence of adverse drug events, mainly from drug–drug interactions. The majority of currently available drugs are metabolized by CYP450 enzymes. Interactions due to shared CYP450-mediated metabolic pathways for two or more drugs are frequent, especially through reversible or irreversible CYP450 inhibition. The magnitude of these interactions depends on several factors, including varying affinity and concentration of substrates, time delay between the administration of the drugs, and mechanisms of CYP450 inhibition. Various types of CYP450 inhibition (competitive, non-competitive, mechanism-based) have been observed clinically, and interactions of these types require a distinct clinical management strategy. This review focuses on mechanism-based inhibition, which occurs when a substrate forms a reactive intermediate, creating a stable enzyme–intermediate complex that irreversibly reduces enzyme activity.
    [Show full text]
  • Spring 2013 Lecture 13-14
    CHM333 LECTURE 13 – 14: 2/13 – 15/13 SPRING 2013 Professor Christine Hrycyna INTRODUCTION TO ENZYMES • Enzymes are usually proteins (some RNA) • In general, names end with suffix “ase” • Enzymes are catalysts – increase the rate of a reaction – not consumed by the reaction – act repeatedly to increase the rate of reactions – Enzymes are often very “specific” – promote only 1 particular reaction – Reactants also called “substrates” of enzyme catalyst rate enhancement non-enzymatic (Pd) 102-104 fold enzymatic up to 1020 fold • How much is 1020 fold? catalyst time for reaction yes 1 second no 3 x 1012 years • 3 x 1012 years is 500 times the age of the earth! Carbonic Anhydrase Tissues ! + CO2 +H2O HCO3− +H "Lungs and Kidney 107 rate enhancement Facilitates the transfer of carbon dioxide from tissues to blood and from blood to alveolar air Many enzyme names end in –ase 89 CHM333 LECTURE 13 – 14: 2/13 – 15/13 SPRING 2013 Professor Christine Hrycyna Why Enzymes? • Accelerate and control the rates of vitally important biochemical reactions • Greater reaction specificity • Milder reaction conditions • Capacity for regulation • Enzymes are the agents of metabolic function. • Metabolites have many potential pathways • Enzymes make the desired one most favorable • Enzymes are necessary for life to exist – otherwise reactions would occur too slowly for a metabolizing organis • Enzymes DO NOT change the equilibrium constant of a reaction (accelerates the rates of the forward and reverse reactions equally) • Enzymes DO NOT alter the standard free energy change, (ΔG°) of a reaction 1. ΔG° = amount of energy consumed or liberated in the reaction 2.
    [Show full text]