View of the Tetrapyrrole Biosynthesis Pathway with Its Substrates, Major Intermediates and Products

Total Page:16

File Type:pdf, Size:1020Kb

View of the Tetrapyrrole Biosynthesis Pathway with Its Substrates, Major Intermediates and Products Florida State University Libraries Electronic Theses, Treatises and Dissertations The Graduate School 2019 Siroheme Biosynthesis and Regulation of SJosierpho Mhasetemrs Peen nDingetoni IcV iency Follow this and additional works at the DigiNole: FSU's Digital Repository. For more information, please contact [email protected] FLORIDA STATE UNIVERSITY COLLEGE OF ARTS AND SCIENCES SIROHEME BIOSYNTHESIS AND REGULATION OF SIROHEME DEFICIENCY By JOSEPH MASTERS PENNINGTON IV A Dissertation submitted to the Institute of Molecular Biophysics in partial fulfillment of the requirements for the degree of Doctor of Philosophy 2019 Joseph Masters Pennington IV defended this dissertation on April 12, 2019. The members of the supervisory committee were: Hong Li Professor Directing Dissertation Kathryn M. Jones University Representative M. Elizabeth Stroupe Committee Member Michael Blaber Committee Member Wei Yang Committee Member The Graduate School has verified and approved the above-named committee members, and certifies that the dissertation has been approved in accordance with university requirements. ii This dissertation is dedicated to my parents, Joe and Patricia; Your love and support have made this possible. iii ACKNOWLEDGMENTS I would like to initially thank my PI, Beth Stroupe, for all her support, patience, and training over the past years. The collaborative atmosphere of the lab and encouragment of undergradaute and high school students to pursue research played a large role in my develpoment as an independent researcher and a future mentor. I would also like to thank my committee, Hong Li, Kathryn Jones, Michael Blaber, and Wei Yang for their input and feedback during each step of my PhD journey. A huge thank you to the FSU core facilites, specifically Cladius Mundoma, Soma Sundaram, Joan Hare, Brian Washburn, Cheryl Pye, and Kristina Poduch. Much of this work would have been impossible, or at best much harder then needed, without your work, guidance, and advice. Next, I want to thank my current and former lab mates, Daniel Murray, Mike Kopylov, Isabel Askenasy, Matt Johnson, Jay Rai, Rachel Andrews, Marisa Cepeda, Lauren McGarry, Angela Tavolieri, Trent Quist, and Ashley Santiago. It has been an honor working and learning alongside of each of you. Lastly, I want to thank my family. All of your love and support as I pursued this crazy degree has meant more than I can ever show. iv TABLE OF CONTENTS LIST OF TABLES .................................................................................................................. viii LIST OF FIGURES .................................................................................................................. ix ABSTRACT ...............................................................................................................................x CHAPTER 1 INTRODUCTION .................................................................................................1 1.1. Preface ..........................................................................................................................1 1.2. Anatomy of a tetrapyrrole .............................................................................................1 1.3. Tetrapyrrole biosynthesis ..............................................................................................2 1.4. The discovery of siroheme and the siroheme biosynthetic pathway ...............................4 1.5. Siroheme biosynthesis in different organisms ................................................................5 1.6. Siroheme in sulfite and nitrite reductases ......................................................................5 CHAPTER 2 THE DEHYDROGENASE AND CHELATASE MECHANISMS OF SALMONELLA TYPHIMURIUM SIROHEME SYNTHASE .......................................................7 2.1. Introduction ..................................................................................................................7 2.2. Materials and methods ..................................................................................................8 2.2.1. Materials and chemicals .........................................................................................8 2.2.2. Cloning of StCysG mutants ....................................................................................8 2.2.3. Protein expression and purification ........................................................................8 2.2.4. Structure determination ..........................................................................................9 2.2.5. Substrate biosynthesis ............................................................................................9 2.2.6. Anaerobic crystal soaking and freezing ................................................................ 10 2.2.7. Data collection and processing ............................................................................. 10 2.2.8. In vivo complementation assays of StCysG mutants ............................................. 10 2.2.9. In vitro specific activity enzyme assays ................................................................ 10 2.3. Results ........................................................................................................................ 11 2.3.1. Structure determination of CysG bound to PC2 + NADH, SHC, and Co-SHC and CysG variants .................................................................................................................... 11 2.3.2. In vivo activity assays of StCysG mutants ............................................................ 11 2.3.3. In vitro activity assays of StCysG mutants ........................................................... 11 2.4. Discussion................................................................................................................... 12 2.4.1. PC2 is puckered in the CysGB active site .............................................................. 12 2.4.2. Coordination of SHC in CysGB ............................................................................ 12 2.4.3. Co-SHC is weakly bound in the second CysGB active site .................................... 13 2.4.4. Fluorescence of CysG substrates in vivo .............................................................. 13 2.4.5. Conservation with BmSirC .................................................................................. 13 v CHAPTER 3 IDENTIFICATION AND CHARACTERIZATION OF MTCYSG AND CHE1, TWO ENZYMES RESPONSIBLE FOR SIROHEME PRODUCTION IN MYCOBACTERIUM TUBERCULOSIS ...................................................................................................................... 25 Introduction ................................................................................................................ 25 Materials and methods ................................................................................................ 26 3.2.1. Materials and chemicals ....................................................................................... 26 3.2.2. Identification of tuberculosis CysG candidates ..................................................... 27 3.2.3. Cloning of CysG Candidates ................................................................................ 27 3.2.4. In vivo CysG complementation assays ................................................................. 27 3.2.5. Protein expression and purification of MtCysG and Che1 .................................... 27 3.2.6. Substrate biosynthesis .......................................................................................... 28 3.2.7. In vitro activity assays of MtCysG and Che1 ........................................................ 28 3.2.8. Crystallization of MtCysG and diffraction ............................................................ 29 Results and discussion................................................................................................. 29 3.3.1. MtCysG is located in an operon associated with vitamin B12 synthesis................ 29 3.3.2. MtCysG is missing the alpha-helical bundle domain ............................................ 30 3.3.3. MtcysG does not complement cysG-deficient E. coli ........................................... 30 3.3.4. MtCysG is an NAD+-dependent PC2 dehydrogenase but not a ferrochelatase....... 30 3.3.5. Diffraction of MtCysG crystals ............................................................................ 31 CHAPTER 4 THE N-TERMINAL DOMAIN OF ESCHERICHIA COLI ASSIMILATORY NADPH-SULFITE REDUCTASE HEMOPROTEIN IS AN OLIGOMERIZATION DOMAIN THAT MEDIATES HOLOENZYME ASSEMBLY .................................................................. 37 4.1. Introduction .................................................................................................................... 37 4.2. Materials and methods .................................................................................................... 39 4.2.1. Recombinant protein production: SiRHP ................................................................. 39 4.2.2. Recombinant protein production: SiRFP .................................................................. 39 4.2.3. Recombinant protein production: SiRFPFMN............................................................. 40 4.2.4. Recombinant protein production: SiRFP43................................................................ 40 4.2.5. Recombinant protein production: SiR ...................................................................... 41 4.2.6. SiR, SiRFP43,
Recommended publications
  • Light-Independent Nitrogen Assimilation in Plant Leaves: Nitrate Incorporation Into Glutamine, Glutamate, Aspartate, and Asparagine Traced by 15N
    plants Review Light-Independent Nitrogen Assimilation in Plant Leaves: Nitrate Incorporation into Glutamine, Glutamate, Aspartate, and Asparagine Traced by 15N Tadakatsu Yoneyama 1,* and Akira Suzuki 2,* 1 Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan 2 Institut Jean-Pierre Bourgin, Institut national de recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), UMR1318, RD10, F-78026 Versailles, France * Correspondence: [email protected] (T.Y.); [email protected] (A.S.) Received: 3 September 2020; Accepted: 29 September 2020; Published: 2 October 2020 Abstract: Although the nitrate assimilation into amino acids in photosynthetic leaf tissues is active under the light, the studies during 1950s and 1970s in the dark nitrate assimilation provided fragmental and variable activities, and the mechanism of reductant supply to nitrate assimilation in darkness remained unclear. 15N tracing experiments unraveled the assimilatory mechanism of nitrogen from nitrate into amino acids in the light and in darkness by the reactions of nitrate and nitrite reductases, glutamine synthetase, glutamate synthase, aspartate aminotransferase, and asparagine synthetase. Nitrogen assimilation in illuminated leaves and non-photosynthetic roots occurs either in the redundant way or in the specific manner regarding the isoforms of nitrogen assimilatory enzymes in their cellular compartments. The electron supplying systems necessary to the enzymatic reactions share in part a similar electron donor system at the expense of carbohydrates in both leaves and roots, but also distinct reducing systems regarding the reactions of Fd-nitrite reductase and Fd-glutamate synthase in the photosynthetic and non-photosynthetic organs.
    [Show full text]
  • Bacterial Heme Biosynthesis and Its Biotechnological Application
    Appl Microbiol Biotechnol (2003) 63: 115–127 DOI 10.1007/s00253-003-1432-2 MINI-REVIEW N. Frankenberg . J. Moser . D. Jahn Bacterial heme biosynthesis and its biotechnological application Received: 23 June 2003 / Revised: 22 August 2003 / Accepted: 26 July 2003 / Published online: 16 September 2003 # Springer-Verlag 2003 Abstract Proteins carrying a prosthetic heme group are The nickel-containing yellow coenzyme F430 is the vital parts of bacterial energy conserving and stress prosthetic group of methyl-coenzyme M reductase that response systems. They also mediate complex enzymatic catalyzes the final step of methane formation in methano- reactions and regulatory processes. Here, we review the genenic archaea (Thauer and Bonacker 1994). The pink multistep biosynthetic pathway of heme formation includ- cobalt-containing vitamin B12 derivatives are the most ing the enzymes involved and reaction mechanisms. complex known tetrapyrroles (Martens et al. 2002). They Potential biotechnological implications are discussed. are involved in multiple enzymatic reactions, e.g., radical- dependent nucleotide reduction and methyl transfer. The iron-chelating yellow-greenish siroheme is required for the Introduction six electron transfer reactions during assimilatory nitrite or sulfite reduction (Raux et al. 2003). The green pigment Structure and function of tetrapyrroles heme d1, is part of the dissimilatory nitrite reductase in Pseudomonads and is a typical porphinoid that differs Compounds of the tetrapyrrole class are characterized by significantly in structure and color from the other their four five-membered pyrrole rings, usually linked porphyrin-based hemes (Chang 1994). together via single atom bridges (Fig. 1). The four rings of The other commonly found tetrapyrrole structures are the macrocycle are labelled clockwise A–D starting with the open-chain molecules that are all derived from cleaved the first of the three symmetric rings with regard to the macrocycles.
    [Show full text]
  • Ferredoxin: the Central Hub Connecting Photosystem I to Cellular Metabolism
    DOI: 10.1007/s11099-018-0793-9 PHOTOSYNTHETICA 56 (1): 279-293, 2018 REVIEW Ferredoxin: the central hub connecting photosystem I to cellular metabolism J. MONDAL* and B.D. BRUCE*,**,+ Department of Biochemistry, Cellular and Molecular Biology*, Graduate School of Genome Science and Technology**, University of Tennessee at Knoxville, Knoxville, Tennessee, USA Abstract Ferredoxin (Fd) is a small soluble iron-sulfur protein essential in almost all oxygenic photosynthetic organisms. It contains a single [2Fe-2S] cluster coordinated by four cysteine ligands. It accepts electrons from the stromal surface of PSI and facilitates transfer to a myriad of acceptors involved in diverse metabolic processes, including generation of NADPH via Fd-NADP-reductase, cyclic electron transport for ATP synthesis, nitrate reduction, nitrite reductase, sulfite reduction, hydrogenase and other reductive reactions. Fd serves as the central hub for these diverse cellular reactions and is integral to complex cellular metabolic networks. We describe advances on the central role of Fd and its evolutionary role from cyanobacteria to algae/plants. We compare structural diversity of Fd partners to understand this orchestrating role and shed light on how Fd dynamically partitions between competing partner proteins to enable the optimum transfer of PSI-derived electrons to support cell growth and metabolism. Additional key words: cellular metabolism; electron transfer; ferredoxin; global interaction; oxidation-reduction. Introduction The discovery of Fd is itself an interesting achievement (Fd). Dan Arnon and collaborators were the first to investi- in the history of biochemistry. Its role in the cellular gate the role of Fd in photosynthesis as described over 50 oxidation-reduction processes is essential in organisms years ago (Tagawa and Arnon 1962).
    [Show full text]
  • Characterization of the Cobalamin (Vitamin B12) Biosynthetic Genes of Salmonella Typhimurium JOHN R
    JOURNAL OF BACTERIOLOGY, June 1993, p. 3303-3316 Vol. 175, No. 11 0021-9193/93/113303-14$02.00/0 Copyright © 1993, American Society for Microbiology Characterization of the Cobalamin (Vitamin B12) Biosynthetic Genes of Salmonella typhimurium JOHN R. ROTH,`* JEFFREY G. LAWRENCE,1 MARC RUBENFIELD 2t STEPHEN KIEFFER-HIGGINS,2 AND GEORGE M. CHURCH2 Department ofBiology, University of Utah, Salt Lake City, Utah 84112,1 and Department of Genetics, Harvard Medical School, Howard Hughes Medical Institute, Boston, Massachusetts 021152 Received 20 November 1992/Accepted 16 March 1993 Salmonella typhimurium synthesizes cobalamin (vitamin B12) de novo under anaerobic conditions. Of the 30 cobalamin synthetic genes, 25 are clustered in one operon, cob, and are arranged in three groups, each group encoding enzymes for a biochemically distinct portion of the biosynthetic pathway. We have determined the DNA sequence for the promoter region and the proximal 17.1 kb of the cob operon. This sequence includes 20 translationally coupled genes that encode the enzymes involved in parts I and III of the cobalamin biosynthetic pathway. A comparison of these genes with the cobalamin synthetic genes from Pseudomonas denitrificans allows assignment of likely functions to 12 of the 20 sequenced Salmonella genes. Three additional Salmonela genes encode proteins likely to be involved in the transport of cobalt, a component of vitamin B12. However, not all Salmonella and Pseudomonas cobalamin synthetic genes have apparent homologs in the other species. These differences suggest that the cobalamin biosynthetic pathways differ between the two organisms. The evolution of these genes and their chromosomal positions is discussed. Cobalamin (vitamin B12) is an evolutionarily ancient co- a known cofactor for numerous enzymes mediating methyl- factor (9, 44, 46) and one of the largest, most structurally ation, reduction, and intramolecular rearrangements (91, complex, nonpolymeric biomolecules described.
    [Show full text]
  • Downloaded from the GTDB Server29
    bioRxiv preprint doi: https://doi.org/10.1101/2021.01.11.426183; this version posted January 11, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 2 Heme auxotrophy in abundant aquatic microbial lineages 3 4 5 Suhyun Kim1, Ilnam Kang2*, Jin-Won Lee3, Che-Ok Jeon4, Stephen J. Giovannoni5, and 6 Jang-Cheon Cho1,6* 7 8 1Department of Biological Sciences, Inha University, Incheon 22212, Republic of Korea 9 2Center for Molecular and Cell Biology, Inha University, Incheon 22212, Republic of Korea 10 3Department of Life Science, Hanyang University, Seoul 04763, Republic of Korea 11 4Department of Life Science, Chung-Ang University, Seoul 06974, Republic of Korea 12 5Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA 13 6Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea 14 15 *E-mail: [email protected] (Ilnam Kang); [email protected] (Jang-Cheon Cho) 16 17 Corresponding authors’ information: 18 *Ilnam Kang: Center for Molecular and Cell Biology, Inha University, Inharo 100, Incheon 22212, Republic of 19 Korea. Tel: +82-32-876-5541 20 *Jang-Cheon Cho: Department of Biological Sciences, Inha University, Inharo 100, Incheon 22212, Republic of 21 Korea. Tel: +82-32-860-7711 22 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.01.11.426183; this version posted January 11, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • Molecular Cloning, Sequencing, and Regulation of the Rhodobacter Capsulatushemb Gene
    Louisiana State University LSU Digital Commons LSU Historical Dissertations and Theses Graduate School 1995 Molecular Cloning, Sequencing, and Regulation of the Rhodobacter capsulatushemB Gene. Karl Joseph Indest Louisiana State University and Agricultural & Mechanical College Follow this and additional works at: https://digitalcommons.lsu.edu/gradschool_disstheses Recommended Citation Indest, Karl Joseph, "Molecular Cloning, Sequencing, and Regulation of the Rhodobacter capsulatushemB Gene." (1995). LSU Historical Dissertations and Theses. 6021. https://digitalcommons.lsu.edu/gradschool_disstheses/6021 This Dissertation is brought to you for free and open access by the Graduate School at LSU Digital Commons. It has been accepted for inclusion in LSU Historical Dissertations and Theses by an authorized administrator of LSU Digital Commons. For more information, please contact [email protected]. INFORMATION TO USERS This manuscript has been reproduced from the microfilm master. UMI films the text directly from the original or copy submitted. Thus, some thesis and dissertation copies are in typewriter face, while others may be from any type of computer printer. The quality of this reproduction is dependent upon the quality of the copy submitted. Broken or indistinct print, colored or poor quality illustrations and photographs, print bleedthrough, substandard margins, and improper alignment can adversely afreet reproduction. In the unlikely event that the author did not send UMI a complete manuscript and there are missing pages, these will be noted. Also, if unauthorized copyright material had to be removed, a note will indicate the deletion. Oversize materials (e.g., maps, drawings, charts) are reproduced by sectioning the original, beginning at the upper left-hand comer and continuing from left to right in equal sections with small overlaps.
    [Show full text]
  • The Marvels of Biosynthesis: Tracking Nature's Pathways
    Pergamon Bioorganic & Medicinal Chemistry, Vol. 4, No. 7, pp 937-964, 1996 Copyright © 1996 Elsevier Science Ltd Printed in Great Britain. All rights reserved PIh S0968-0896(96)00102-2 0968-0896/96 $15.00+0.00 The Marvels of Biosynthesis: Tracking Nature's Pathways Alan R. Battersby University of Cambridge, University Chemical Laboratory, Lensfield Road, Cambridge CB2 1EW, U.K. Introduction and nitric acids, zinc, sulphur, copper sulphate and many more materials. Those days are gone and there How ever did it come about that a substantial part of are pluses and minuses to the change. At any rate, I my research has been aimed at understanding the was able to assemble a good set of equipment to run marvellous chemistry used by living systems to lots of simple experiments which I enjoyed enormously. ,:onstruct the substances they produce? I must admit ".hat I had not in the past thought much about that I believe the next important influence on me came at 9articular 'pathway' but was encouraged to do so by school where I had the great good fortune to be taught Derek Barton, Chairman of the Executive Board of more about chemistry by a superb teacher, Mr Evans. Editors for Tetrahedron Publications. He suggested The seed of my love for chemistry which had been that this article, invited by Professor Chi-Huey Wong, planted earlier by my father's books was strongly fed by should be a personal one giving some background on his teaching. Then I read my first books about organic how my interests evolved.
    [Show full text]
  • Metabolic Engineering of Escherichia Coli for De Novo Biosynthesis Of
    bioRxiv preprint doi: https://doi.org/10.1101/394338; this version posted August 17, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Metabolic engineering of Escherichia coli for de novo biosynthesis of vitamin B12 Huan Fang1, 2, Dong Li1, Jie Kang1, Pingtao Jiang1, Jibin Sun1,2, and Dawei Zhang1, 2, 3* 1Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China. 2Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China. 3National Engineering Laboratory for Industrial Enzymes, Tianjin 300308, China *Correspondence: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/394338; this version posted August 17, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. ABSTRACT The only known source of vitamin B12 (adenosylcobalamin) is from bacteria and archaea, and the only unknown step in its biosynthesis is the production of the intermediate adenosylcobinamide phosphate. Here, using genetic and metabolic engineering, we generated an Escherichia coli strain that produces vitamin B12 via an engineered de novo aerobic biosynthetic pathway. Excitingly, the BluE and CobC enzymes from Rhodobacter capsulatus transform L-threonine into (R)-1-Amino-2- propanol O-2-Phosphate, which is then condensed with adenosylcobyric acid to yield adenosylcobinamide phosphate by either CobD from the aeroic R. capsulatus or CbiB from the anerobic Salmonella typhimurium. These findings suggest that the biosynthetic steps from co(II)byrinic acid a,c-diamide to adocobalamin are the same in both the aerobic and anaerobic pathways.
    [Show full text]
  • Kent Academic Repository Full Text Document (Pdf)
    Kent Academic Repository Full text document (pdf) Citation for published version Dailey, Harry A. and Dailey, Tamara A. and Gerdes, Svetlana and Jahn, Dieter and Jahn, Martina and O'Brian, Mark R. and Warren, Martin J. (2017) Prokaryotic Heme Biosynthesis: Multiple Pathways to a Common Essential Product. Review of: Prokaryotic Heme Biosynthesis: Multiple Pathways to a Common Essential Product by UNSPECIFIED. Microbiology and Molecular DOI https://doi.org/10.1128/MMBR.00048-16 Link to record in KAR http://kar.kent.ac.uk/60615/ Document Version Publisher pdf Copyright & reuse Content in the Kent Academic Repository is made available for research purposes. Unless otherwise stated all content is protected by copyright and in the absence of an open licence (eg Creative Commons), permissions for further reuse of content should be sought from the publisher, author or other copyright holder. Versions of research The version in the Kent Academic Repository may differ from the final published version. Users are advised to check http://kar.kent.ac.uk for the status of the paper. Users should always cite the published version of record. Enquiries For any further enquiries regarding the licence status of this document, please contact: [email protected] If you believe this document infringes copyright then please contact the KAR admin team with the take-down information provided at http://kar.kent.ac.uk/contact.html REVIEW crossm Prokaryotic Heme Biosynthesis: Multiple Pathways to a Common Essential Product Downloaded from Harry A. Dailey,a Tamara A. Dailey,a Svetlana Gerdes,b Dieter Jahn,c Martina Jahn,d Mark R.
    [Show full text]
  • Structure and Mechanistic Implications of a Uroporphyrinogen III Synthase-Product Complex†,‡ Heidi L
    8648 Biochemistry 2008, 47, 8648–8655 Structure and Mechanistic Implications of a Uroporphyrinogen III Synthase-Product Complex†,‡ Heidi L. Schubert,*,§ John D. Phillips,| Annie Heroux,⊥ and Christopher P. Hill§ Departments of Biochemistry and Internal Medicine, School of Medicine, UniVersity of Utah, Salt Lake City, Utah 84112, and Department of Biology, BrookhaVen National Laboratory, Upton, New York 11973-5000 ReceiVed April 9, 2008; ReVised Manuscript ReceiVed June 17, 2008 ABSTRACT: Uroporphyrinogen III synthase (U3S) catalyzes the asymmetrical cyclization of a linear tetrapyrrole to form the physiologically relevant uroporphyrinogen III (uro’gen III) isomer during heme biosynthesis. Here, we report four apoenzyme and one product complex crystal structures of the Thermus thermophilus (HB27) U3S protein. The overlay of eight crystallographically unique U3S molecules reveals a huge range of conformational flexibility, including a “closed” product complex. The product, uro’gen III, binds between the two domains and is held in place by a network of hydrogen bonds between the product’s side chain carboxylates and the protein’s main chain amides. Interactions of the product A and B ring carboxylate side chains with both structural domains of U3S appear to dictate the relative orientation of the domains in the closed enzyme conformation and likely remain intact during catalysis. The product C and D rings are less constrained in the structure, consistent with the conformational changes required for the catalytic cyclization with inversion
    [Show full text]
  • Porphyrin, Heme, and Siroheme Biosynthesis Svetlana Gerdes, Fellowship for Interpretation of Genomes Introduction
    Subsystem: Porphyrin, Heme, and Siroheme Biosynthesis Svetlana Gerdes, Fellowship for Interpretation of Genomes Introduction Tetrapyrroles and their derivatives play an essential role in all living organisms. They are involved in many metabolic processes, such as energy transfer, catalysis, and signal transduction. In eukaryotes, the synthesis of tetrapyrroles is restricted to heme, siroheme, chlorophyll and bilins. Prokaryotes additionally form most complicated tetrapyrroles, such as corrinoids, heme d1 and coenzyme F430. An abundant and ubiquitous representative of this group of compounds is heme, a cyclic tetrapyrrole that contains a centrally chelated Fe. The biosynthetic pathway of heme and siroheme can be arbitrary divided into 4 fragments: A: Biosynthesis of 5-aminolevulinic acid (ALA), the common precursor of all marcocyclic and linear tetrapyrrolesis, can occur via two alternative unrelated routes: the C5-pathway, or the Shemin pathway. The C5 pathway, found in most bacteria, archaea and plants, starts from the C5-skeleton of glutamate, ligated to tRNAGlu. Some alpha-proteobacteria, fungi, and animals synthesize 5-aminolevulinate via Shemin pathway by condensation of succinyl-CoA with glycine. B. Universal steps in biosynthesis of tetrapyrroles: condensation of 8 molecules of 5-aminolevulinic acid to form Uroporphyrinogen III (Uro-III) - the first cyclic tetrapyrrole intermediate in the pathway. Universally present, very conserved, variations are extremely rare. The corresponding genes form conserved chromosomal clusters in a large number of genomes. Located at the branchpoint of tetrapyrrole biosynthesis, Uro-III can be converted to both Siroheme (via Uro-III methyltransferase, UroM) and protoporphyrin IX (via Uro-III decarboxylase, UroD). Regulation of Uro-III partitioning of into the two main branches, currently poorly understood, can be a fascinating research topic (see below).
    [Show full text]
  • Biochemical and Mutational Analysis of Coenzyme B12 Biosynthesis Chenguang Fan Iowa State University
    Iowa State University Capstones, Theses and Graduate Theses and Dissertations Dissertations 2009 Biochemical and mutational analysis of coenzyme B12 biosynthesis Chenguang Fan Iowa State University Follow this and additional works at: https://lib.dr.iastate.edu/etd Part of the Biochemistry, Biophysics, and Structural Biology Commons Recommended Citation Fan, Chenguang, "Biochemical and mutational analysis of coenzyme B12 biosynthesis" (2009). Graduate Theses and Dissertations. 11247. https://lib.dr.iastate.edu/etd/11247 This Dissertation is brought to you for free and open access by the Iowa State University Capstones, Theses and Dissertations at Iowa State University Digital Repository. It has been accepted for inclusion in Graduate Theses and Dissertations by an authorized administrator of Iowa State University Digital Repository. For more information, please contact [email protected]. Biochemical and mutational analysis of coenzyme B 12 biosynthesis by Chenguang Fan A dissertation submitted to the graduate faculty in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Major: Biochemistry Program of Study Committee: Thomas Bobik, Major Professor Alan DiSpirito Alan Myers Basil Nikolau Anthony Pometto III Iowa State University Ames, Iowa 2009 Copyright © Chenguang Fan, 2009. All rights reserved. ii TABLE OF CONTENTS LIST OF FIGURES iv LIST OF TABLES vi ABSTRACT vii CHAPTER 1. GENERAL INTRODUCTION 1 Introduction 1 Literature Review 1 Thesis Organization 23 References 23 CHAPTER 2. FUNCTIONAL CHARACTERIZATION AND MUTATION ANALYSIS OF HUMAN ATP:COB(I)ALAMIN ADENOSYLTRANSFERASE 50 Abstract 50 Introduction 50 Materials and Methods 53 Results 57 Discussion 63 Acknowledgments 66 References 66 CHAPTER 3. THE PDUX ENZYME OF SALMONELLA ENTERICA IS AN L- THREONINE KINASE USED FOR COENZYME B 12 SYNTHESIS 80 iii Abstract 80 Introduction 81 Experimental Procedures 82 Results 87 Discussion 93 Acknowledgments 96 References 96 CHAPTER 4.
    [Show full text]