Enterobactin

Last updated
Enterobactin
Enterobactin.svg
Names
Preferred IUPAC name
N,N′,N′′-[(3S,7S,11S)-2,6,10-Trioxo-1,5,9-trioxacyclododecane-3,7,11-triyl]tris(2,3-dihydroxybenzamide)
Identifiers
3D model (JSmol)
ChEBI
ChEMBL
ChemSpider
PubChem CID
UNII
  • InChI=1S/C30H27N3O15/c34-19-7-1-4-13(22(19)37)25(40)31-16-10-46-29(44)18(33-27(42)15-6-3-9-21(36)24(15)39)12-48-30(45)17(11-47-28(16)43)32-26(41)14-5-2-8-20(35)23(14)38/h1-9,16-18,34-39H,10-12H2,(H,31,40)(H,32,41)(H,33,42)/t16-,17-,18-/m0/s1 Yes check.svgY
    Key: SERBHKJMVBATSJ-BZSNNMDCSA-N Yes check.svgY
  • InChI=1/C30H27N3O15/c34-19-7-1-4-13(22(19)37)25(40)31-16-10-46-29(44)18(33-27(42)15-6-3-9-21(36)24(15)39)12-48-30(45)17(11-47-28(16)43)32-26(41)14-5-2-8-20(35)23(14)38/h1-9,16-18,34-39H,10-12H2,(H,31,40)(H,32,41)(H,33,42)/t16-,17-,18-/m0/s1
    Key: SERBHKJMVBATSJ-BZSNNMDCBT
  • C1C(C(=O)OCC(C(=O)OCC(C(=O)O1)NC(=O)C2=C(C(=CC=C2)O)O)NC(=O)C3=C(C(=CC=C3)O)O)NC(=O)C4=C(C(=CC=C4)O)O
  • c1cc(c(c(c1)O)O)C(=O)N[C@H]2COC(=O)[C@H](COC(=O)[C@H](COC2=O)NC(=O)c3cccc(c3O)O)NC(=O)c4cccc(c4O)O
Properties
C30H27N3O15
Molar mass 669.55 g/mol
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).
Yes check.svgY  verify  (what is  Yes check.svgYX mark.svgN ?)

Enterobactin (also known as enterochelin) is a high affinity siderophore that acquires iron for microbial systems. It is primarily found in Gram-negative bacteria, such as Escherichia coli and Salmonella typhimurium . [1]

Contents

Enterobactin is the strongest siderophore known, binding to the ferric ion (Fe3+) with affinity K = 1052 M−1. [2] This value is substantially larger than even some synthetic metal chelators, such as EDTA (Kf,Fe3+ ~ 1025 M−1). [3] Due to its high affinity, enterobactin is capable of chelating even in environments where the concentration of ferric ion is held very low, such as within living organisms. Pathogenic bacteria can steal iron from other living organisms using this mechanism, even though the concentration of iron is kept extremely low due to the toxicity of free iron.

Structure and biosynthesis

Chorismic acid, an aromatic amino acid precursor, is converted to 2,3-dihydroxybenzoic acid (DHB) by a series of enzymes, EntA, EntB and EntC. An amide linkage of DHB to L-serine is then catalyzed by EntD, EntE, EntF and EntB. Three molecules of the DHB-Ser formed undergo intermolecular cyclization, yielding enterobactin. [4] Although a number of stereoisomers are possible due to the chirality of the serine residues, only the Δ-cis isomer is metabolically active. [3] The first three-dimensional structure of a metal enterobactin complex was determined as the vanadium(IV) complex. [5] Although ferric enterobactin long eluded crystallization, its definitive three-dimensional structure was ultimately obtained using racemic crystallography, in which crystals of a 1:1 mixture of ferric enterobactin and its mirror image (ferric enantioenterobactin) were grown and analyzed by X-ray crystallography. [6]

Synthesis of enterobactin startig from chorismate Enterobactin synthesis.svg
Synthesis of enterobactin startig from chorismate

Mechanism

Iron deficiency in bacterial cells triggers secretion of enterobactin into the extracellular environment, causing formation of a coordination complex "FeEnt" wherein ferric ion is chelated to the conjugate base of enterobactin. In Escherichia coli , FepA in the bacterial outer membrane then allows entrance of FeEnt to the bacterial periplasm. FepB,C,D and G all participate in transport of the FeEnt through the inner membrane by means of an ATP-binding cassette transporter. [4]

Due to the extreme iron binding affinity of enterobactin, it is necessary to cleave FeEnt with ferrienterobactin esterase to remove the iron. This degradation yields three 2,3-dihydroxybenzoyl-L-serine units. Reduction of the iron (Fe3+ to Fe2+) occurs in conjunction with this cleavage, but no FeEnt bacterial reductase enzyme has been identified, and the mechanism for this process is still unclear. [8] The reduction potential for Fe3+/Fe2+enterobactin complex is pH dependent and varies from 0.57 V (vs NHE) at pH 6 to 0.79 V at pH 7.4 to 0.99 at pH values higher than 10.4. [9]

History

Enterobactin was discovered by Gibson and Neilands groups in 1970. [10] [11] These initial studies established the structure and its relationship to 2,3-dihydroxybenzoic acid.

Related Research Articles

Bioleaching is the extraction or liberation of metals from their ores through the use of living organisms. Bioleaching is one of several applications within biohydrometallurgy and several methods are used to treat ores or concentrates containing copper, zinc, lead, arsenic, antimony, nickel, molybdenum, gold, silver, and cobalt.

<span class="mw-page-title-main">Siderophore</span> Iron compounds secreted by microorganisms

Siderophores (Greek: "iron carrier") are small, high-affinity iron-chelating compounds that are secreted by microorganisms such as bacteria and fungi. They help the organism accumulate iron. Although a widening range of siderophore functions is now being appreciated, siderophores are among the strongest (highest affinity) Fe3+ binding agents known. Phytosiderophores are siderophores produced by plants.

Iron–sulfur proteins are proteins characterized by the presence of iron–sulfur clusters containing sulfide-linked di-, tri-, and tetrairon centers in variable oxidation states. Iron–sulfur clusters are found in a variety of metalloproteins, such as the ferredoxins, as well as NADH dehydrogenase, hydrogenases, coenzyme Q – cytochrome c reductase, succinate – coenzyme Q reductase and nitrogenase. Iron–sulfur clusters are best known for their role in the oxidation-reduction reactions of electron transport in mitochondria and chloroplasts. Both Complex I and Complex II of oxidative phosphorylation have multiple Fe–S clusters. They have many other functions including catalysis as illustrated by aconitase, generation of radicals as illustrated by SAM-dependent enzymes, and as sulfur donors in the biosynthesis of lipoic acid and biotin. Additionally, some Fe–S proteins regulate gene expression. Fe–S proteins are vulnerable to attack by biogenic nitric oxide, forming dinitrosyl iron complexes. In most Fe–S proteins, the terminal ligands on Fe are thiolate, but exceptions exist.

<span class="mw-page-title-main">Chorismic acid</span> Chemical compound

Chorismic acid, more commonly known as its anionic form chorismate, is an important biochemical intermediate in plants and microorganisms. It is a precursor for:

In enzymology, a 2,3-dihydro-2,3-dihydroxybenzoate dehydrogenase (EC 1.3.1.28) is an enzyme that catalyzes the chemical reaction

<span class="mw-page-title-main">Aerobactin</span> Chemical compound

Aerobactin is a bacterial iron chelating agent (siderophore) found in E. coliand other Enterobacteriaceae species. It is a virulence factor enabling E. coli to sequester iron in iron-poor environments such as the urinary tract.

2,3-Dihydroxybenzoic acid is a natural phenol found in Phyllanthus acidus and in the aquatic fern Salvinia molesta. It is also abundant in the fruits of Flacourtia inermis. It is a dihydroxybenzoic acid, a type of organic compound.

<span class="mw-page-title-main">Ferrichrome</span> Chemical compound

Ferrichrome is a cyclic hexa-peptide that forms a complex with iron atoms. It is a siderophore composed of three glycine and three modified ornithine residues with hydroxamate groups [-N(OH)C(=O)C-]. The 6 oxygen atoms from the three hydroxamate groups bind Fe(III) in near perfect octahedral coordination.

<span class="mw-page-title-main">Rhodotorulic acid</span> Chemical compound

Rhodotorulic acid is the smallest of the 2,5-diketopiperazine family of hydroxamate siderophores which are high-affinity chelating agents for ferric iron, produced by bacterial and fungal phytopathogens for scavenging iron from the environment. It is a tetradentate ligand, meaning it binds one iron atom in four locations (two hydroxamate and two lactam moieties), and forms Fe2(siderophore)3 complexes to fulfill an octahedral coordination for iron.

<span class="mw-page-title-main">Yersiniabactin</span> Chemical compound

Yersiniabactin (Ybt) is a siderophore found in the pathogenic bacteria Yersinia pestis, Yersinia pseudotuberculosis, and Yersinia enterocolitica, as well as several strains of enterobacteria including enteropathogenic Escherichia coli and Salmonella enterica. Siderophores, compounds of low molecular mass with high affinities for ferric iron, are important virulence factors in pathogenic bacteria. Iron—an essential element for life used for such cellular processes as respiration and DNA replication—is extensively chelated by host proteins like lactoferrin and ferritin; thus, the pathogen produces molecules with an even higher affinity for Fe3+ than these proteins in order to acquire sufficient iron for growth. As a part of such an iron-uptake system, yersiniabactin plays an important role in pathogenicity of Y. pestis, Y. pseudotuberculosis, and Y. entercolitica.

Many bacteria secrete small iron-binding molecules called siderophores, which bind strongly to ferric ions. FepA is an integral bacterial outer membrane porin protein that belongs to outer membrane receptor family and provides the active transport of iron bound by the siderophore enterobactin from the extracellular space, into the periplasm of Gram-negative bacteria. FepA has also been shown to transport vitamin B12, and colicins B and D as well. This protein belongs to family of ligand-gated protein channels.

Ferric-chelate reductase (NADPH) (EC 1.16.1.9, ferric chelate reductase, iron chelate reductase, NADPH:Fe3+-EDTA reductase, NADPH-dependent ferric reductase, yqjH (gene)) is an enzyme with systematic name Fe(II):NADP+ oxidoreductase. This enzyme catalyses the following chemical reaction

<span class="mw-page-title-main">Ascorbate ferrireductase (transmembrane)</span> Class of enzymes

Ascorbate ferrireductase (transmembrane) (EC 1.16.5.1, cytochrome b561) is an enzyme with systematic name Fe(III):ascorbate oxidorectuctase (electron-translocating). This enzyme catalyses the following chemical reaction

N2-citryl-N6-acetyl-N6-hydroxylysine synthase (EC 6.3.2.38, N(alpha)-citryl-N(epsilon)-acetyl-N(epsilon)-hydroxylysine synthase, iucA (gene)) is an enzyme with systematic name citrate:N6-acetyl-N6-hydroxy-L-lysine ligase (ADP-forming). This enzyme catalyses the following chemical reaction

<span class="mw-page-title-main">Bacillibactin</span> Chemical compound

Bacillibactin is a catechol-based siderophore secreted by members of the genus Bacillus, including Bacillus anthracis and Bacillus subtilis. It is involved in the chelation of ferric iron (Fe3+) from the surrounding environment and is subsequently transferred into the bacterial cytoplasm via the use of ABC transporters.

Siderocalin(Scn), lipocalin-2, NGAL, 24p3 is a mammalian lipocalin-type protein that can prevent iron acquisition by pathogenic bacteria by binding siderophores, which are iron-binding chelators made by microorganisms. Iron serves as a key nutrient in host-pathogen interactions, and pathogens can acquire iron from the host organism via synthesis and release siderophores such as enterobactin. Siderocalin is a part of the mammalian defence mechanism and acts as an antibacterial agent. Crystallographic studies of Scn demonstrated that it includes a calyx, a ligand-binding domain that is lined with polar cationic groups. Central to the siderophore/siderocalin recognition mechanism are hybrid electrostatic/cation-pi interactions. To evade the host defences, pathogens evolved to produce structurally varied siderophores that would not be recognized by siderocalin, allowing the bacteria to acquire iron.

The iron/lead transporter (ILT) family is a family of transmembrane proteins within the lysine exporter (LysE) superfamily. The ILT family includes two subfamilies, the iron-transporting (OFeT) family and the lead-transporting (PbrT) family. A representative list of the proteins belonging to these subfamilies of the ILT family can be found in the Transporter Classification Database.

<span class="mw-page-title-main">Rebecca Abergel</span> French inorganic chemist

Rebecca Abergel is a professor of nuclear engineering and of chemistry at University of California, Berkeley. Abergel is also a senior faculty scientist in the chemical sciences division of Lawrence Berkeley National Laboratory, where she directs the Glenn T. Seaborg Center and leads the Heavy Element Chemistry research group. She is the recipient of several awards for her research in nuclear and inorganic chemistry.

<span class="mw-page-title-main">Vibriobactin</span> Chemical compound

Vibriobactin is a catechol siderophore that helps the microbial system to acquire iron. It was first isolated from Vibrio cholerae.

<span class="mw-page-title-main">Mirubactin</span> Chemical compound

Mirubactin is a siderophore produced by the bacterium Actinosynnema mirum.A.mirum was first isolated from the Raritan River in New Jersey in 1976, and its full genome sequence was published in 2009. In 2012, mirubactin was isolated and characterized, and the biosynthesis was connected with the gene cluster Amir_2714-Amir_2728, since renamed mrbA-mrbO.

References

  1. Dertz EA, Xu J, Stintzi A, Raymond KN (January 2006). "Bacillibactin-mediated iron transport in Bacillus subtilis". Journal of the American Chemical Society. 128 (1): 22–3. doi:10.1021/ja055898c. PMID   16390102.
  2. Carrano CJ, Raymond KN (1979). "Ferric Ion Sequestering Agents. 2. Kinetics and Mechanism of Iron Removal From Transferrin by Enterobactin and Synthetic Tricatechols". J. Am. Chem. Soc. 101 (18): 5401–5404. doi:10.1021/ja00512a047.
  3. 1 2 Walsh CT, Liu J, Rusnak F, Sakaitani M (1990). "Molecular Studies on Enzymes in Chorismate Metabolism and the Enterobactin Biosynthetic Pathway". Chemical Reviews . 90 (7): 1105–1129. doi:10.1021/cr00105a003.
  4. 1 2 Raymond KN, Dertz EA, Kim SS (April 2003). "Enterobactin: an archetype for microbial iron transport". Proceedings of the National Academy of Sciences of the United States of America. 100 (7): 3584–8. doi: 10.1073/pnas.0630018100 . PMC   152965 . PMID   12655062.
  5. Karpishin TB, Raymond KN (1992). "The First Structural Characterization of A Metal-Enterobactin Complex: [V(enterobactin)]2-". Angewandte Chemie International Edition in English . 31 (4): 466–468. doi:10.1002/anie.199204661.
  6. Johnstone TC, Nolan EM (October 2017). "Determination of the Molecular Structures of Ferric Enterobactin and Ferric Enantioenterobactin Using Racemic Crystallography". Journal of the American Chemical Society. 139 (42): 15245–15250. doi:10.1021/jacs.7b09375. PMC   5748154 . PMID   28956921.
  7. Raines, D. J.; Sanderson, T. J.; Wilde, E. J.; Duhme-Klair, A. -K. (2015-01-01), "Siderophores", Reference Module in Chemistry, Molecular Sciences and Chemical Engineering, Elsevier, ISBN   978-0-12-409547-2 , retrieved 2024-07-06
  8. Ward TR, Lutz A, Parel SP, Ensling J, Gütlich P, Buglyó P, Orvig C (November 1999). "An Iron-Based Molecular Redox Switch as a Model for Iron Release from Enterobactin via the Salicylate Binding Mode". Inorganic Chemistry. 38 (22): 5007–5017. doi:10.1021/ic990225e. PMID   11671244.
  9. Lee CW, Ecker DJ, Raymond KN (1985). "Coordination chemistry of microbial iron transport compounds. 34. The pH-dependent reduction of ferric enterobactin probed by electrochemical methods and its implications for microbial iron transport". J. Am. Chem. Soc. 107 (24): 6920–6923. doi:10.1021/ja00310a030.
  10. Pollack JR, Neilands JB (March 1970). "Enterobactin, an iron transport compound from Salmonella typhimurium". Biochemical and Biophysical Research Communications. 38 (5): 989–92. doi:10.1016/0006-291X(70)90819-3. PMID   4908541.
  11. O'Brien IG, Cox GB, Gibson F (March 1970). "Biologically active compounds containing 2,3-dihydroxybenzoic acid and serine formed by Escherichia coli". Biochimica et Biophysica Acta (BBA) - General Subjects. 201 (3): 453–60. doi:10.1016/0304-4165(70)90165-0. PMID   4908639.