Competitive Inhibition That Barrier, As We Shall See

Total Page:16

File Type:pdf, Size:1020Kb

Competitive Inhibition That Barrier, As We Shall See Chapter 4 Catalysis In living systems, speed is everything. Providing the reaction speeds necessary to support life are the catalysts, mostly in the form of enzymes. Catalysis Introduction Introduction Activation Energy General Mechanisms of Action If there is a magical component to life, an argument can surely be made for it Substrate Binding being catalysis. Thanks to catalysis, reactions that could take hundreds of years Enzyme Flexibility to complete in the “real world,” occur in seconds in the presence of a catalyst. Active Site Chymotrypsin Chemical catalysts, like platinum, speed reactions, but enzymes (which are simply Enzyme Parameters super-catalysts with a twist) put chemical catalysts to shame. To understand VMAX & KCAT enzymatic catalysis, we must first understand energy. In Chapter 2, we noted the KM tendency for processes to move in the direction of lower energy. Chemical Perfect Enzymes Lineweaver-Burk Plots reactions follow this universal trend, but they often have a barrier in place that Enzyme Inhibition must be overcome. The secret to catalytic action is reducing the magnitude of Competitive Inhibition that barrier, as we shall see. No Effect On VMAX Increased KM Non-Competitive Inhibition Uncompetitive Inhibition Suicide Inhibition Control of Enzymes Allosterism Covalent Control of Enzymes Other Controls of Enzymes Ribozymes Enzyme Rate Enhancements 81 Activation Energy Another feature to note about catalyzed reactions is Click HERE, HERE, and HERE for The figure below schematically depicts the energy changes that the reduced energy barrier Kevin’s Catalytic Mechanism occur during the progression of a simple reaction. In the figure, lectures on YouTube (also called the activation the energy differences during the reaction are compared for a energy or free energy of catalyzed (plot on the right) and an uncatalyzed reaction (plot on activation) to reach the transition state of the catalyzed reaction. the left). Notice that the reactants start at the same energy level This is the second important point for both conditions and about catalyzed reactions – catalysts that the products end work by lowering activation energies at the same energy for of reactions and thus molecules more both as well. Thus, the easily reach the energy necessary to difference in energy get to the point where the reaction between the energy of occurs. Note that these reactions are the ending compounds reversible. The extent to which they and the starting will proceed is a function of the size compounds is the same of the energy difference between the in both cases. This is product and reactant states. The the first important rule lower the energy of the products to understand any kind compared to the reactants, the larger of catalysis – catalysts the percentage of molecules that will do not change the be present as products at equilibrium. overall energy of a At equilibrium, of course, no change reaction. Given enough in concentration of reactants and time, a non-catalyzed products occurs because at this reaction will get to the point, the forward and reverse same equilibrium as a Energetic Considerations of Catalysis reaction rates are the same. catalyzed one. 82 General Mechanisms of Action Substrate Binding As noted above, enzymes are orders of magnitude more effective Another important difference between the mechanism of action of (faster) than chemical catalysts. The secret of their success lies an enzyme and a chemical catalyst is that an enzyme has binding in a fundamental difference in their mechanisms of action. Every sites that not only ‘grab’ the substrate (molecule involved in the chemistry student has had hammered into their heads the fact reaction being catalyzed), but also place it in a position to be that a catalyst speeds a reaction without being consumed by it. electronically induced to react, either within itself or with another In other words, the catalyst ends up after a reaction just the way it substrate. The enzyme itself may play a role in the electronic started so it can catalyze induction or the induction may occur as a result of substrates other reactions, as well. being placed in very close proximity to each other. Chemical Click HERE, HERE, HERE, and Enzymes share this catalysts have no such ability to bind substrates and are HERE for Kevin’s enzyme property, but in the middle, dependent upon them colliding in the right orientation at or near catalysis lectures on YouTube during the catalytic action, their surfaces. an enzyme is transiently changed. Such changes may be subtle electronic ones or more significant covalent modifications. It is also important to recognize that enzymes are not fixed, rigid structures, but rather are flexible. Flexibility allows movement and movement facilitates alteration of electronic environments necessary for catalysis. Enzymes are, thus, much more efficient than rigid chemical catalysts as a result of their abilities to facilitate the changes necessary to optimize the catalytic process. Mechanism of Induced Fit 83 Enzyme Flexibility The Way They Work To the tune of “The Way We Were” As mentioned earlier, a difference between an enzyme and a chemical catalyst is that an enzyme is flexible. Its slight Enzymes Mighty powerhouse peptides changes in shape (often arising from the binding of the Cause reactions to go faster In the cell’s insides substrate itself) help to position substrates for reaction Tiny substrates after they bind. These changes in shape are explained, in Bring about an induced fit part, by Koshland’s Induced Fit Model of Catalysis, Enzyme structure is affect-ed By what binds to it which illustrates that not only do enzymes change Can it be that it’s just simple zen? substrates, but that substrates also transiently change How the enzymes activate enzymes. At the end of the catalysis, the enzyme is If they bind effector, will they go To an R-State, T-State? returned to its original state. Enzyme flexibility also is Folding important for control of enzyme activity. Two distinct Gives the mechanistic might structures are typically described– the T (tight) state, which To three-D arrangement Of the active site is a lower activity state and the R (relaxed) state, which has Enzymes greater activity. Have a bias they can’t hide Hydrophobic side chains are Mostly found inside Active Site So it’s the structure Reactions in enzymes are catalyzed at a specific location For celebrating Whenever there’s debating known as the ‘active site’. Substrate binding sites are The way they work located in close physical proximity to the active site and oriented to provide access for the relevant portion of the T molecule to the electronic environment of the enzyme where catalysis is initiated. Recorded by Liz Bacon and David Simmons Lyrics by Kevin Ahern 84 Chymotrypsin two phases – a fast phase that occurs first and a slower phase Consider the mechanism that follows. The enzyme has a of catalysis of the enzyme substrate binding site that known as chymotrypsin. includes a region of the enzyme Found in our digestive known as the S1 pocket. Let us system, chymotrypsin’s step through the mechanism by catalytic action is cleaving which chymotrypsin cuts peptide bonds in proteins adjacent to phenylalanine. and it uses the side chain of a serine in its The process starts with the mechanism of catalysis. binding of the substrate in the Many other protein- S1 pocket. The S1 pocket in cutting enzymes employ a chymotrypsin has a hydrophobic very similar mechanism hole in which the substrate is and they are known bound. Preferred substrates will collectively as serine include amino acid side chains proteases. As a that are hydrophobic, like protease, it acts fairly phenylalanine. If an ionized side specifically, cutting not all chain, like that of glutamic acid peptide bonds, but only binds in the S1 pocket, it will those that are adjacent to quickly exit, much like water specific amino acids in the would avoid an oily interior. protein. One of the amino When the proper substrate acids it cuts adjacent to is binds, it stays and its presence phenylalanine. The induces an ever so slight shift in enzyme’s action occurs in Serine Protease Mechanism the shape of the enzyme. This 85 subtle shape change on the binding of the proper substrate starts The Serine Protease Song the steps of the catalysis and is the reason that the enzyme To the tune of “Blackbird” shows specificity for cutting at specific enzyme positions in the Substrate floating in the cell’s insides Enzyme snags it with its binding site target protein. Only amino acids with the side chains that interact It supplies well with the S1 pocket start the catalytic wheels turning. Shuffling of electrons in the act to catalyze The slight changes in shape of the enzyme upon binding of the Proteases of the serine kind Break up peptide bonds in rapid time proper substrate cause changes in the positioning of three amino Fast and slow acids (aspartic acid, histidine, and serine) in the active site known Steps in breaking bonds are mechanisms you should know as the catalytic triad, during the second step of the catalytic Asp – his - ser action. The shift of the negatively charged aspartic acid towards Bonds beware the electron rich histidine ring favors the abstraction of a proton Inside the S1 pocket substrate sits by the histidine from the hydroxyl group on the side chain of Alkoxides serine, resulting in production of a very reactive alkoxide ion in Break peptides Nucleophiles give bonds the fits the active site. Since the active site at this point also contains the polypeptide chain positioned with the phenylalanine side chain Peptide one exits easily But water has to let the other flee embedded in the S1 pocket, the alkoxide ion performs a Bound not free nucleophilic attack on the peptide bond on the carboxyl side of ‘Cuz the enzyme’s linked to it in mechanism three phenylalanine sitting in the active site.
Recommended publications
  • The Serpintine Solution
    & Experim l e ca n i t in a l l C C f a Journal of Clinical & Experimental o r d l i a o n l o r g u Lucas et al., J Clin Exp Cardiolog 2017, 8:1 y o J Cardiology ISSN: 2155-9880 DOI: 10.4172/2155-9880.1000e150 Editorial Open Access The Serpintine Solution Alexandra Lucas, MD, FRCP(C)1,2,*, Sriram Ambadapadi, PhD1, Brian Mahon, PhD3, Kasinath Viswanathan, PhD4, Hao Chen, MD, PhD5, Liying Liu, MD6, Erbin Dai, MD6, Ganesh Munuswami-Ramanujam, PhD7, Jacek M. Kwiecien, DVM, MSc, PhD8, Jordan R Yaron, PhD1, Purushottam Shivaji Narute, BVSc & AH, MVSc, PhD1,9, Robert McKenna, PhD10, Shahar Keinan, PhD11, Westley Reeves, MD, PhD12, Mark Brantly, MD, PhD13, Carl Pepine, MD, FACC14 and Grant McFadden, PhD1 1Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe AZ, USA 2Saint Josephs Hospital, Dignity Health Phoenix, Phoenix, AZ, USA 3NIH/ NIDDK, Bethesda MD, USA 4Zydus Research Centre, Ahmedabad, India 5Department of tumor surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu, P.R.China 6Beth Israel Deaconess Medical Center, Harvard, Boston, MA, USA 7Interdisciplinary Institute of Indian system of Medicine (IIISM), SRM University, Chennai, Tamil Nadu, India 8MacMaster University, Hamilton, ON, Canada 9Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda MD, USA 10Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA 11Cloud Pharmaceuticals, Durham, North Carolina, USA 12Division of Rheumatology, University of Florida,
    [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]
  • Insights Into the Role of Tick Salivary Protease Inhibitors During Ectoparasite–Host Crosstalk
    International Journal of Molecular Sciences Review Insights into the Role of Tick Salivary Protease Inhibitors during Ectoparasite–Host Crosstalk Mohamed Amine Jmel 1,† , Hajer Aounallah 2,3,† , Chaima Bensaoud 1, Imen Mekki 1,4, JindˇrichChmelaˇr 4, Fernanda Faria 3 , Youmna M’ghirbi 2 and Michalis Kotsyfakis 1,* 1 Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, Branišovská 1160/31, 37005 Ceskˇ é Budˇejovice,Czech Republic; [email protected] (M.A.J.); [email protected] (C.B.); [email protected] (I.M.) 2 Institut Pasteur de Tunis, Université de Tunis El Manar, LR19IPTX, Service d’Entomologie Médicale, Tunis 1002, Tunisia; [email protected] (H.A.); [email protected] (Y.M.) 3 Innovation and Development Laboratory, Innovation and Development Center, Instituto Butantan, São Paulo 05503-900, Brazil; [email protected] 4 Faculty of Science, University of South Bohemia in Ceskˇ é Budˇejovice, 37005 Ceskˇ é Budˇejovice, Czech Republic; [email protected] * Correspondence: [email protected] † These authors contributed equally. Abstract: Protease inhibitors (PIs) are ubiquitous regulatory proteins present in all kingdoms. They play crucial tasks in controlling biological processes directed by proteases which, if not tightly regulated, can damage the host organism. PIs can be classified according to their targeted proteases or their mechanism of action. The functions of many PIs have now been characterized and are showing clinical relevance for the treatment of human diseases such as arthritis, hepatitis, cancer, AIDS, and cardiovascular diseases, amongst others. Other PIs have potential use in agriculture as insecticides, anti-fungal, and antibacterial agents.
    [Show full text]
  • Optimizing Defence, Counter-Defence and Counter-Counter Defence in Parasitic and Trophic Interactions – a Modelling Study
    1 Optimizing defence, counter-defence and counter-counter defence in parasitic and trophic interactions – A modelling study Stefan Schuster1*, Jan Ewald1,2, Thomas Dandekar2, Sybille Dühring1 1Dept. of Bioinformatics, Friedrich Schiller University of Jena Ernst-Abbe-Platz 2, 07743 Jena, Germany 2Dept. of Bioinformatics, Biocenter Julius Maximilian University of Würzburg Am Hubland, 97074 Würzburg *Corresponding author, e-mail: [email protected] Abstract In host-pathogen interactions, often the host (attacked organism) defends itself by some toxic compound and the parasite, in turn, responds by producing an enzyme that inactivates that compound. In some cases, the host can respond by producing an inhibitor of that enzyme, which can be considered as a counter-counter defence. An example is provided by cephalosporins, β-lactamases and clavulanic acid (an inhibitor of β-lactamases). Here, we tackle the question under which conditions it pays, during evolution, to establish a counter- counter defence rather than to intensify or widen the defence mechanisms. We establish a mathematical model describing this phenomenon, based on enzyme kinetics for competitive inhibition. We use an objective function based on Haber’s rule, which says that the toxic effect is proportional to the time integral of toxin concentration. The optimal allocation of defence and counter-counter defence can be calculated in an analytical way despite the nonlinearity in the underlying differential equation. The calculation provides a threshold value for the dissociation constant of the inhibitor. Only if the inhibition constant is below that threshold, that is, in the case of strong binding of the inhibitor, it pays to have a counter-counter defence.
    [Show full text]
  • Bsc Chemistry
    Subject Chemistry Paper No and Title 16, Bio-organic and Biophysical Chemistry Module No and Title 30, Enzymes as targets or drug design Module Tag CHE_P16_M30 Chemistry PAPER No. : 16, Bioorganic and biophysical chemistry MODULE No. : 30, Enzymes as targets of drug design TABLE OF CONTENTS 1. Learning outcomes 2. Why Enzymes as Drug Targets? 3. Enzyme structure and Catalysis 4. Concept of Enzyme Inhibition 5. Types of Enzyme Inhibition 5.1. Nonspecific Inhibitors 5.2. Specific Inhibitors 6. Summary Chemistry PAPER No. : 16, Bioorganic and biophysical chemistry MODULE No. : 30, Enzymes as targets of drug design 1. Learning outcomes After studying this module you shall be able to: Know about enzymes as drug targets Learn about enzyme structure and catalysis Know the concept of enzyme inhibition Understand the different types of enzyme inhibition 2. Introduction 2.1 Why Enzymes as Drug Targets? Medicine in twenty first century has become a science in which drug molecules are directed against the macromolecules. Enzymes hold a prominent position among the biological macromolecules that can be used as drug targets. Some features of enzyme structures and reaction pathways which make them attractive and primary target for drug action are as follows They play an essential role in biological life processes and pathophysiology The structures of active sites of enzymes and the ligand binding pockets are highly amenable for high affinity interactions with small drug like molecules Presence of potential allosteric sites Conformational variations in the binding sites Can be directly used as a therapeutic agent 3. Enzyme structure and catalysis Enzymes are biological catalysts as well as can act as receptors by acting with the substrates.
    [Show full text]
  • Imexon and Gemcitabine: Mechanisms of Synergy Against Human Pancreatic Cancer
    Imexon and Gemcitabine: Mechanisms of Synergy against Human Pancreatic Cancer Item Type text; Electronic Dissertation Authors Roman, Nicholas Publisher The University of Arizona. Rights Copyright © is held by the author. Digital access to this material is made possible by the University Libraries, University of Arizona. Further transmission, reproduction or presentation (such as public display or performance) of protected items is prohibited except with permission of the author. Download date 02/10/2021 18:39:06 Link to Item http://hdl.handle.net/10150/194494 IMEXON AND GEMCITABINE: MECHANISMS OF SYNERGY AGAINST HUMAN PANCREATIC CANCER by NICHOLAS ORDUÑO ROMÁN _________________________ A Dissertation Submitted to the Faculty of the COMMITTEE ON PHARMACOLOGY AND TOXICOLOGY (GRADUATE) In Partial Fulfillment of the Requirements For the Degree of DOCTOR OF PHILOSOPHY In the Graduate College THE UNIVERSITY OF ARIZONA 2 0 0 5 2 THE UNIVERSITY OF ARIZONA GRADUATE COLLEGE As members of the Dissertation Committee, we certify that we have read the disser tation prepared by Nicholas Ordu ño Rom án entitled Imexon and Gemcitabine: Mechanisms of Synergy against Human Pancreatic Cancer and recommend that it be accepted as fulfilling the dissertation requirement for the Degree of Doctor of Philosophy _____________________________________ ___________________________ Date:_______________ Robert T. Dorr, Ph.D. 4/12/05 _________ _______________________________________________________ Date:_______________ Terrence J. Monks. Ph.D. 4/12/05 _____ ___________________________________________________________ Date:_______________ Nathan J. Cherrington, Ph.D. 4/12/05 __________________________________________ ______________________ Date:_______________ Garth Powis, D. Phil. 4/12/05 ________ __ ______________________________________________________ Date:_______________ Terry H. Landowski, Ph.D. 4/12/05 Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertatio n to the Graduate College.
    [Show full text]
  • Identification of Regulatory Mechanisms of the Hepatic
    Available online at www.sciencedirect.com Computers and Chemical Engineering 32 (2008) 356–369 Identification of regulatory mechanisms of the hepatic response to thermal injury E. Yang a, T.J. Maguire a, M.L. Yarmush a, F. Berthiaume b, I.P. Androulakis a,∗ a Biomedical Engineering Department, Rutgers University, Piscataway, NJ, USA b Center for Engineering in Medicine/Surgical Services, Massachusetts General Hospital, Harvard Medical School and the Shriners Hospitals for Children, Boston, MA, USA Received 30 October 2006; received in revised form 11 February 2007; accepted 13 February 2007 Available online 25 February 2007 Abstract The purpose of this paper is to evaluate the hypothesis that a systems biology approach can be developed such that a set of transcription factors, relevant to burn-induced inflammatory response can be identified and modulated to control the host response. We explore a novel method for identifying coherent and informative expression motifs and we subsequently determine conserved transcription factor binding sites for the sub-sets of co-expressed genes. The responses are rationalized in the context of burn-induced inflammation and the putative transcription factors are rationalized in the context of intervention targets for controlling gene expression. © 2007 Published by Elsevier Ltd. Keywords: Gene expression; Burn; Inflammation; Transcritpion factors 1. Introduction to thermal injury predispose to the later hypermetabolic state would make it possible to define points of intervention by which Deep thermal injury over greater than 20 percent of the total such outcomes can be avoided. body surface area is one of the most severe forms of trauma. The response to burn injury and trauma results from a com- Following the early acute phase response dealing with the initial plex interplay between inflammation caused by the initial injury injury and shock (Carter, Tompkins, Yarmush, Walker, & Burke, and hypermetabolism.
    [Show full text]
  • Reversible and Mechanism-Based Irreversible Inhibitor Studies on Human Steroid Sulfatase and Protein Tyrosine Phosphatase 1B
    Reversible and Mechanism-Based Irreversible Inhibitor Studies on Human Steroid Sulfatase and Protein Tyrosine Phosphatase 1B by F. Vanessa Ahmed A thesis presented to the University of Waterloo in fulfilment of the thesis requirement for the degree of Doctor of Philosophy in Chemistry Waterloo, Ontario, Canada, 2009 © F. Vanessa Ahmed 2009 Author’s Declaration I hereby declare that I am the sole author of this thesis. This is a true copy of the thesis, including any required final revisions, as accepted by my examiners. I understand that my thesis may be made electronically available to the public. ii Abstract The development of reversible and irreversible inhibitors of steroid sulfatase (STS) and protein tyrosine phosphatase 1B (PTP1B) is reported herein. STS belongs to to the aryl sulfatase family of enzymes that have roles in diverse processes such as hormone regulation, cellular degradation, bone and cartilage development, intracellular communication, and signalling pathways. STS catalyzes the desulfation of sulfated steroids which are the storage forms of many steroids such as the female hormone estrone. Its crucial role in the regulation of estrogen levels has made it a therapeutic target for the treatment of estrogen-dependent cancers. Estrone sulfate derivatives bearing 2- and 4- mono- and difluoromethyl substitutions were examined as quinone methide-generating suicide inhibitors of STS with the goal of developing these small molecules as activity- based probes for proteomic profiling of sulfatases. Kinetic studies suggest that inhibition by the monofluoro derivatives is a result of a quinone methide intermediate that reacts with active-site nucleophiles. However, the main inhibition pathway of the 4- difluoromethyl derivative involved an unexpected process in which initially formed quinone methide diffuses from the active site and decomposes to an aldehyde in solution which then acts as a potent, almost irreversible STS inhibitor.
    [Show full text]
  • Cefoxitin, N-Formimidoyl Thienamycin, Clavulanic
    1594 THE JOURNAL OF ANTIBIOTICS NOV. 1982 CEFOXITIN, N-FORMIMIDOYL THIENAMYCIN, CLAVULANIC ACID, AND PENICILLANIC ACID SULFONE AS SUICIDE INHIBITORS FOR DIFFERENT TYPES OF ~-LACTAMASES PRODUCED BY GRAM-NEGATIVE BACTERIA TETSUO SAWAI and KIKUO TSUKAMOTO Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (Received for publication July 30, 1982) Cefoxitin, N-formimidoyl thienamycin (MK0787), clavulanic acid, and penicillanic acid sulfone (CP45,899) were studied to determine their potency as suicide inhibitors of three very different kinds of Vii-lactamases produced by Gram-negative bacteria: type Ib penicillinase (TEM-2 type), a typical cephalosporinase (the enzyme of Proteus morganii), and a cephalo- sporinase with broad substrate specificity (the enzyme of Proteus vulgaris). All these ~-lactams were confirmed to be quite stable to the three 19-lactamases. The absolute values of the turn- over numbers (kcat) for these enzyme-catalyzed hydrolyses were determined, the values ranged from 0.25 minute-1 to 660 minute-1. All the j3-lactams studied, except cefoxitin, acted as suicide inhibitors of the typical cephalosporinase. Although cefoxitin did not exhibit such an effect, it was a powerful competitive inhibitor of this enzyme. Although the four j3-lactams acted as suicide inhibitors of the P. vulgaris cephalosporinase, the inactivated enzyme partially regained its activity after removing the effect of the free inhibitor. Type lb penicillinase was also inactivated by the four p-lactams, and the activity of the inactivated enzyme, except in the case of cefoxitin, was partially restored. The rate constants for enzyme inactivation or reacti- vation were calculated and are presented. The information obtained from this study suggests that the catalytic center of the P.
    [Show full text]
  • Chapter 8. Enzymes: Basic Concepts and Kinetics
    CHAPTER 8. ENZYMES: BASIC CONCEPTS AND KINETICS coelenterazine The activity of an enzyme is responsible for the glow of the luminescent jellyfish at left. The enzyme aequorin catalyzes the oxidation of a compound by oxygen in the presence of calcium to release CO2 and light CHAPTER 8. ENZYMES: BASIC CONCEPTS AND KINETICS Introduction . Enzymes are large biological molecules that catalyze chemical transformations . The molecule also mediates the transformation of one form of energy into another . 25% of the genes in the human genome encode enzymes . Catalytic power and specificity – important characteristics . Active site – the place where enzymatic reactions take place CHAPTER 8. ENZYMES: BASIC CONCEPTS AND KINETICS Introduction . Nearly all known enzymes are proteins • RNA can function as a catalyst . Proximity effect – enzymes bring substrates closer together in an optimal orientation . They catalyze reactions by stabilizing transition states 8.1 CATALYTIC POWER AND SPECIFICITY Rate enhancement by enzymes . Enzymes accelerate reactions by factors of as much as a million or more 8.1 CATALYTIC POWER AND SPECIFICITY Substrate specificity . A substrate is a molecule upon which an enzyme acts . Some proteases cleaves specific amide bonds in a peptide . The specificity of an enzyme is due to the precise interaction of the substrate with the enzyme . This precision is a result of the intricate 3D structure of the enzyme protein Fig 8.1 Enzyme specificity. (A) Trypsin cleaves on the carboxyl side of arginine and lysine residues, whereas (B) thrombin cleaves Arg-Gly bonds in particular sequences only 8.1 CATALYTIC POWER AND SPECIFICITY Cofactors . The catalytic activity of enzymes depends on the presence of small molecules termed cofactors .
    [Show full text]
  • Pdf (734.95 K)
    Biochemistry Letters, 11(20) 2016, pages 259-274 Scientific Research & Studies Center-Faculty of Science- Zagazig University- Egypt Biochemistry Letters Journal home page: Interactions of Monoamine Oxidase from Human And Rat Liver Mitochondria with (S)-2-[4-(3-Flouro-Benzyloxy)-Benzylamino]-Propionamide Zakia Ben Ramadan*1 , Keith Tipton2 , Safaa khater3 1-Department of Biochemistry, Faculty of Medicine University of Tripoli-Libya. 2-Department of Biochemistry, Trinity College, Dublin 2, Ireland . 3- Department of Biochemistry, Zagazig University –Egypt. ARTICLE INFO ABSTRACT Article history: Background:The interactions of the anticonvulsant (S)-2-[4- Received : (3-Flouro-Benzyloxy) Benzylamino]- Propionamide (FCE Accepted : 26743) with MAO-A and MAO -B from human and rat liver Available online : mitochondria has been studied. Objective: This compound is an alanine derivative that differs from the parent anticonvulsant Keywords: Monoamine compound, 2-n-pentylamino acetamide in that alaninamide oxidase-A and-B (MAO-A& residue replaces the glycinamide moiety, and in the presence of MAO -B), kinetics of inhibition, a phenyl ring substituted in the para-position by a 3-flouro- (S)-2-[4-(3-flourobenzyloxy)- benzyloxy group. Results: The results indicated that, The Ki benzylamino] propionamide, values for rat and human MAO-A were > 1000 times higher (FCE 26743), Inhibition than their corresponding values for rat and human MAO- Studies. Consistent with the IC50values that showed this compound to be > 1000 times more potent as an inhibitor of MAO-B than of MAO-A of the same species. There was an increase in the strength of inhibition when MAO-B was incubated with this compound. This was confirmed by the extended time-courses studies which showed a rather rapid increase in the degree of inhibition of MAO-B, despite incubation being continued for a total of 4h.
    [Show full text]
  • Crossroads of Antibiotic Resistance and Biosynthesis
    Review Crossroads of Antibiotic Resistance and Biosynthesis Timothy A. Wencewicz Department of Chemistry, Washington University in St. Louis, One Brookings Drive, St. Louis, MO 63130, USA Correspondence to : [email protected] https://doi.org/10.1016/j.jmb.2019.06.033 Abstract The biosynthesis of antibiotics and self-protection mechanisms employed by antibiotic producers are an integral part of the growing antibiotic resistance threat. The origins of clinically relevant antibiotic resistance genes found in human pathogens have been traced to ancient microbial producers of antibiotics in natural environments. Widespread and frequent antibiotic use amplifies environmental pools of antibiotic resistance genes and increases the likelihood for the selection of a resistance event in human pathogens. This perspective will provide an overview of the origins of antibiotic resistance to highlight the crossroads of antibiotic biosynthesis and producer self-protection that result in clinically relevant resistance mechanisms. Some case studies of synergistic antibiotic combinations, adjuvants, and hybrid antibiotics will also be presented to show how native antibiotic producers manage the emergence of antibiotic resistance. © 2019 Elsevier Ltd. All rights reserved. Introduction panel of isogenic Escherichia coli strains. Plazomicin is a semisynthetic aminoglycoside derived from Antibiotics continue to be the most important sisomicin and is designed to chemically block the resource in the global management of infectious sites of chemical modification by aminoglycoside diseases [1,2]. The increased occurrence of antibi- inactivating enzymes, including N-acetyltransferases otic resistance in human pathogens has raised (AACs), O-nucleotidylyltransferases, and O- global concern as antibiotics steadily lose efficacy in phosphotransferases (APHs) [9,10]. Indeed, plazomi- clinical and community settings.
    [Show full text]