Open access peer-reviewed chapter

Vitamin B1 (Thiamine) Metabolism and Regulation in Archaea

Written By

Julie A. Maupin-Furlow

Submitted: 12 February 2018 Reviewed: 11 April 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.77170

From the Edited Volume

B Group Vitamins - Current Uses and Perspectives

Edited by Jean Guy LeBlanc and Graciela Savoy de Giori

Chapter metrics overview

1,785 Chapter Downloads

View Full Metrics

Abstract

Thiamine is the water-soluble sulfur containing vitamin B1 that is used to form thiamine diphosphate (ThDP), an enzyme cofactor important in the metabolism of carbohydrates, amino acids and other organic molecules. ThDP is synthesized de novo by certain bacteria, archaea, yeast, fungi, plants, and protozoans. Other organisms, such as humans, rely upon thiamine transport and salvage for metabolism; thus, thiamine is considered an essential vitamin. The focus of this chapter is on the regulation and metabolism of thiamine in archaea. The review will discuss the role ThDP has as an enzyme cofactor and the catalytic and regulatory mechanisms that archaea use to synthesize, salvage and transport thiamine. Future perspectives will be articulated in terms of how archaea have advanced our understanding of thiamine metabolism, regulation and biotechnology applications.

Keywords

  • thiamine
  • vitamin B1
  • archaea
  • thiazole
  • thiazolium
  • pyrimidine
  • sulfur mobilization
  • riboswitch

1. Introduction

Thiamine or vitamin B1 consists of a thiazole/thiazolium ring [5-(2-hydroxyethyl)-4-methylthiazole, THZ] linked by a methylene bridge to an aminopyrimidine ring (2-methyl-4-amino-5-hydroxymethylpyrimidine, HMP) (Figure 1A). Thiamine diphosphate (ThDP) is the best-known form of thiamine, as it is a cofactor. Other natural thiamine phosphate derivatives include: thiamine monophosphate (ThMP), thiamine triphosphate (ThTP), adenosine thiamine triphosphate (AThTP) and adenosine thiamine diphosphate (AThDP) (Figure 1A) [1, 2]. These latter forms have yet to be analyzed in archaea and, thus, will not be a focus of this review.

Figure 1.

Thiamin (vitamin B1) and its natural forms. A) Thiamin and its natural derivatives thiamin monophosphate (ThMP), thiamin diphosphate (ThDP), thiamin triphosphate (ThTP), and adenosine thiamin triphosphate (AdThTP). The aminopyrimidine ring (blue), thiazolium ring (red) and methylene bridge (green) are highlighted with carbon indicated by C or blue balls. B) Thiamin diphosphate and its C2 anion/ylid form (ThDP-). Enzyme bound ThDP is in a V-conformation, which positions the 4′-amino group of the pyrimidine to abstract the C2-H proton of the thiazolium ring when activated by a conserved glutamate residue of the enzyme (in red). The two resonance structures of the anion/ylid are presented.

Advertisement

2. Thiamine diphosphate

ThDP is an enzyme cofactor found in all domains of life. In archaea and bacteria, ThDP is considered one of the eight universal cofactors along with NAD, NADP, FAD, FMN, S-adenosyl-methionine (SAM), pyridoxal-5-phosphate (PLP, vitamin B6), CoA and the C1 carrier tetrahydrofolate or tetrahydromethanopterin [3]. The rare exceptions are the bacteria Borrelia and Rickettsia, which do not use ThDP as a coenzyme for metabolism [4].

ThDP-dependent enzymes catalyze the cleavage and formation of C-C, C-N, C-S and C-O bonds in a wide range of catabolic and anabolic reactions [5]. As a coenzyme, ThDP serves as an electrophilic covalent catalyst in the decarboxylation of 2-oxo acids (e.g., pyruvate and 2-oxoglutarate) and in carboligation and lyase-type reactions [6, 7, 8]. The active species of ThDP is typically the C2 anion/ylid (ThDP) form, generated by dissociation of the C2-H proton from the thiazole ring (Figure 1B). ThDP is the source of the catalytic power of ThDP-dependent enzymes, as it can add to unsaturated systems and serve as a sink for mobile electrons [9, 10]. ThDP typically requires Mg2+ or Ca2+ ions to bind the enzyme in a V conformation in which the 4′-amino group of the pyrimidine ring is positioned to abstract the C2-H proton from the thiazole ring (Figure 1B) [11, 12, 13, 14, 15]. This proton abstraction is often assisted by a conserved glutamate residue (Glu) of the enzyme that provides a carboxylate side chain for hydrogen bonding to the N1’ of the pyrimidine ring and for proton relay to form the ThDP catalytic intermediate (Figure 1B). Thus, ThDP is fundamentally distinct among coenzymes in that both rings contribute to catalysis.

ThDP-dependent enzymes are used in pyruvate metabolism, the TCA cycle, the pentose phosphate pathway and branched chain amino acid biosynthesis (Table 1). Archaea commonly use ThDP-dependent 2-oxoacid: ferredoxin oxidoreductases (OFORs) to catalyze the oxidative decarboxylation of 2-oxoacids (e.g., pyruvate, 2-oxoglutarate and 2-oxoisovalerate) into an energy rich CoA thioester [16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32] or the reverse reaction to fix CO2 into cell carbon [33]. ThDP, Mg2+ and Fe-S cluster(s) are the intrinsic cofactors of OFORs with ferredoxin as the electron acceptor. OFORs (typically 270 kDa) are less complex than the 5-6 MDa 2-oxoacid dehydrogenases (ODHs) of mitochondria and aerobic bacteria; ODHs rely upon NAD+ as the electron acceptor and are composed of E1p (ThDP-dependent 2-oxoacid decarboxylase), E2p (lipoate acetyltransferase) and E3p (dihydrolipoamide dehydrogenase) components [16]. While some archaea express mRNAs specific for all three ODH (E1p, E2p and E3p) homologs, ODH activity has yet to be detected in archaea [30]. Other ThDP-dependent enzymes of archaea include the non-oxidative 3-sulfopyruvate decarboxylase of coenzyme M biosynthesis [34, 35] and the acetohydroxyacid synthase of branch-chain amino acid (isoleucine, leucine and valine) biosynthesis [36, 37]. The transketolase activities of archaea [38] are presumed to be catalyzed by ThDP-dependent enzymes based on comparative genomics [39].

Archaea Bacteria Eukarya EC Enzyme (Abbreviation and Description)
+ + + 1.2.4.1 PDH Pyruvate dehydrogenase (E1p component)
n.d. + + 1.2.4.2 OGDH 2-Oxoglutarate dehydrogenase (E1o component)
+ (rare) + + 1.2.4.4 BCOADH Branched chain 2-oxoacid dehydrogenase (E1b component)
+ + + 2.2.1.1 TK Transketolase (glycolaldehyde transferase)
n.d. + (rare) + 4.1.-.- HACL 2-Hydroxyphytanoyl−/2-hydroxyacyl-CoA lyase
+ + n.d. 1.2.3.3 POX Pyruvate oxidase (phosphate-dependent)
+ + n.d. 1.2.7.1 PFOR Pyruvate: ferredoxin oxidoreductase
+ + n.d. 1.2.7.3 KGOR 2-Oxoglutarate: ferredoxin oxidoreductase
+ + n.d. 1.2.7.7 VOR 2-Oxoisovalerate: ferredoxin oxidoreductase
+ + n.d. 1.2.7.8 IOR Indolepyruvate: ferredoxin oxidoreductase
n.d. + (rare) n.d. 1.2.7.10 Oxalate: ferredoxin oxidoreductase
n.d. n.d. + 2.2.1.3 DHAS Dihydroxyacetone synthase (formaldehyde transketolase)
+ + + 2.2.1.6 AHAS Acetohydroxyacid synthase (acetylacetate synthase)
n.d. + + 2.2.1.7 DXPS 1-Deoxy-D-xylulose 5-phosphate synthase
+ + + 2.2.1.9 MenD 2-Succinyl-5-enolpyruvyl-6-hydroxy-3-cyclohexene-1-carboxylic-acid synthase
n.d. + n.d. 2.5.1.66 CeaS N2-(2-carboxyethyl)arginine synthase
? + ? 3.7.1.11 Cyclohexane-1,2-dione hydrolase
? + + 4.1.1.1 PDC Pyruvate decarboxylase
+ + n.d. 4.1.1.7 BFD Benzoylformate decarboxylase
n.d. + n.d. 4.1.1.8 OXC Oxalyl-CoA decarboxylase
? ? + 4.1.1.43 Phenylpyruvate decarboxylase
n.d. + n.d. 4.1.1.47 GCL Glyoxylate carboligase (tartronate semialdehyde synthase)
n.d. + n.d. 4.1.1.71 KGD 2-Oxoglutarate decarboxylase
+ + n.d. 4.1.1.74 IpdC Indolepyruvate decarboxylase
+ + n.d. 4.1.1.79 ComDE Sulfopyruvate decarboxylase
+ (rare) + + 4.1.1.82 PnPyDC 3-Phosphonopyruvate decarboxylase
n.d. + + 4.1.2.9 PHK Phosphoketolase (D-xylulose-5-phosphate phosphoketolase)
? + ? 4.1.2.38 BAL Benzaldehyde lyase (benzoin aldolase)

Table 1.

Thiamin diphosphate (ThDP)-dependent enzymes and their distribution among the three domains of life. Enzyme homolog detected (+), not detected (n.d.), or low homology (?) as indicated.

Advertisement

3. Thiamine biosynthesis de novo

Thiamine is synthesized de novo by generating thiazole and aminopyrimidine rings separately and then joining the rings to form ThMP, the precursor of ThDP. The de novo pathways rely upon energy input (ATP), carbon- and nitrogen-based intermediates and a source of sulfur (the latter incorporated into the thiazole ring).

3.1. Synthesis and phosphorylation of the aminopyrimidine ring of thiamine

ThiC (HMP-P synthase; EC 4.1.99.17) is the major enzyme used by bacteria [40, 41], plant chloroplasts [42] and archaea [43] to synthesize the aminopyrimidine ring of thiamine (Figures 2-4). ThiC converts 5′-phosphoribosyl-5-aminoimidazole (AIR) to 4-amino-5-hydroxymethyl-2-methylpyrimidine phosphate (HMP-P), thus, diverting carbon/nitrogen skeletons of purine metabolism to thiamine biosynthesis. ThiC is a radical SAM enzyme, that initiates this catalytic reaction by use of a [4Fe-4S]+ cluster that reductively cleaves SAM to methionine and an 5′-deoxyadenosyl radical [40], a presumed oxidizing cosubstrate of the reaction [44].

Figure 2.

Thiamin (vitamin B1) biosynthesis in bacteria. Enzymes are discussed in text and colored by phylogenetic distribution (red, restricted to one domain of life; blue, found in all domains of life; green, apparent homologs in all domains of life but no direct evidence). Abbreviations: AIR, 5-aminoimidazole ribotide; SAM, S-adenosyl-methionine; GAP3P, D-glyceraldehyde 3-phosphate; HMP-P, 4-aminohydroxymethyl-2-methylpyrimidine phosphate; HMP-PP, 4aminohydroxymethyl-2-methylpyrimidine diphosphate; ThMP, thiamin monophosphate; ThDP, thiamin diphosphate; DXP, 1-deoxy-D-xylulose 5-phosphate; cTHZ-P, 2-[(2R,5Z)-2-carboxy-4-methylthiazol-5(2H)-ylidene]ethyl phosphate; THZ-P, 4-methyl-5-(β-hydroxyethyl)thiazolium phosphate; X, electron carrier.

Figure 3.

Thiamin (vitamin B1) biosynthesis in eukaryotes. Blue shading indicates restricted to yeast. Abbreviations: ADP-thiazole, ADP-5-ethyl-4methylthiazole-2-carboxylate; PLP, pyridoxal phosphate; R5P, D-ribose 5-phosphate.?, not determined to date. For additional abbreviations and coloring scheme see Figure 2.

Figure 4.

Thiamin (vitamin B1) biosynthesis in archaea. For abbreviations and coloring scheme see Figures 2, 3.

THI5 forms the aminopyrimidine ring of thiamine from the substrates PLP and histidine in yeast [45, 46] (Figure 3). Only a subset of THI5 family (IPR027939) proteins have the conserved histidine residue needed for HMP-P synthesis [45] and appear restricted to yeast, fungi, plants (non-chloroplast) and select γ-proteobacteria. Bacterial ABC-type solute binding proteins for HMP precursor (ThiY) [47] and riboflavin (RibY) [48] transport are structurally related to THI5. Thus, the archaeal THI5 family proteins, which are devoid of the conserved histidine residue, are suggested to serve a similar role in transport.

ThiD domain proteins are used as bifunctional HMP kinase (EC 2.7.1.49)/HMP-P kinase (EC 2.7.4.7) enzymes in thiamine biosynthesis and salvage (Figures 2-4). Bacterial ThiD [49, 50] and yeast THI20 and THI21 (N-terminal ThiD domain proteins) [51] phosphorylate HMP-P to HMP-PP in the de novo pathway and successively phosphorylate HMP to HMP-PP in the salvage pathway. Proteins with an unusual ThiD2 domain (standalone or fused to ThiE) are identified in bacteria to catalyze only HMP-P kinase activity, potentially to avoid misincorporation of damaged and/or toxic analogs of HMP into ThDP-dependent enzymes [52]. ThiD homologs (IPR004399) are widespread in all domains of life, including organisms that only salvage HMP and do not synthesize thiamine de novo. Archaeal ThiD proteins are standalone or fused to a ThiN-type ThMP synthase domain (see later discussion) [43, 53, 54].

3.2. Synthesis of the thiazole ring of thiamine

De novo biosynthesis of the thiazole ring can be classified into two fundamentally distinct pathways based on the type of thiazole synthase (ThiG vs. Thi4) used. While similar in nomenclature, the ThiG- and Thi4-type thiazole synthases differ in terms of structure and function. The ThiG-dependent pathway relies upon at least six steps to form THZ-P and appears limited to bacteria based on the phylogenetic distribution of ThiG (EC 2.8.1.10) (Figure 2). By contrast, the Thi4-type branch for thiazole biosynthesis is simpler in having only two steps (Figures 3-4) and appears more widespread, as Thi4-homologs (KEGG K03146) are represented in all domains of life and are demonstrated to function in thiazole ring biosynthesis in yeast [55] and archaea [56, 57].

3.2.1. Synthesis of the thiazole ring of thiamine by the ThiG-pathway

To form the thiazole ring, ThiG uses three substrates: () dehydroglycine, (ii) 1-deoxy-D-xylulose-5-phosphate (DXP) and (iii) thiocarboxylated ThiS [58, 59, 60, 61] (Figure 2).

(i) Dehydroglycine is synthesized by either oxygen-dependent (ThiO; EC 1.4.3.19) or SAM radical enzymes (ThiH; EC 4.1.99.19), both of which are broadly distributed in bacteria but generally absent in archaea and eukaryotes. The ThiO glycine oxidase catalyzes the oxidative deamination of glycine to form the dehydroglycine required for thiazole ring synthesis [62, 63, 64, 65]. By contrast, the ThiH tyrosine lyase forms a 5′-deoxyadenosyl radical that initiates cleavage of the C alpha-C beta bond of tyrosine to generate the dehydroglycine (needed for thiamine biosynthesis) and p-cresol (the byproduct) [66, 67, 68].

(ii) The 1-deoxy-D-xylulose-5-phosphate synthase (Dxs; EC 2.2.1.7) is a ThDP-dependent enzyme that condenses the (hydroxyethyl)-group derived from pyruvate with the C1 aldehyde group of D-glyceraldehyde 3-phosphate (GAP3P) to generate DXP and CO2 [69, 70]. Dxs homologs (IPR005477) are widespread in bacteria, green algae, higher plants and protists but rare in archaea. Dxs generates the DXP precursor of thiamine, pyridoxol and non-mevalonate isoprenoid biosynthesis pathways [69, 70]. DXP is used for thiamine biosynthesis in bacteria but not in eukaryotes or archaea (Figure 2).

(iii) The ThiG-dependent pathway uses a protein-based relay system to mobilize sulfur to the thiazole ring. Sulfur is transferred from L-cysteine to an active site cysteine residue of a sulfurtransferase (e.g., IscS-SH) [71] to form an enzyme persulfide intermediate (e.g., IscS-S-SH) [72]. In a separate reaction, the E1-like ThiF adenylates the C-terminus of the ubiquitin-fold protein, ThiS, in a mechanism resembling the activation step of ubiquitination [73]. This modification step readies the C-terminus of ThiS for thiocarboxylation. The sulfur is relayed from IcsS-S-SH to ThiS through the ThiI rhodanese (RHD) domain [71, 74, 75, 76]. The resulting thiocarboxylated ThiS serves as the sulfur donor for the ThiG mediated synthesis of the thiazole ring [58, 59, 60, 61].

3.2.2. Synthesis of the thiazole ring of thiamine by the Thi4-pathway

The Thi4-pathway used to form the thiazole ring (Figures 3, 4) is distinct from that of ThiG (Figure 2). Key to the pathway is Thi4-mediated formation of ADP-thiazole, which is then hydrolyzed to THZ-P by a presumed NUDIX hydrolase [55]. Thi4 family (IPR002922) proteins are distributed in all domains of life and generally absent from ThiG-containing bacteria. Although initially annotated as ribose-1,5-bisphosphate isomerases (R15Pi) based on indirect assay [77], archaeal Thi4 homologs are found to be distinct from archaeal R15Pi of the e2b2 family [78, 79] and demonstrated to catalyze thiazole synthase activity [56] that is transcriptionally repressed when thiamine and THZ levels are sufficient [43] and is required for thiazole ring formation [57]. In vitro, yeast Thi4 operates by a suicide mechanism by mobilizing the sulfur of its active site cysteine (C205) to form ADP-thiazole from NAD and glycine [55]. By contrast, the methanogen Thi4, uses an active site histidine residue and iron to catalyze the synthesis of ADP-thiazole from NAD, glycine and sulfide [56]. Thi4 enzymes of archaea, yeast [80] and plant [81] are related based on X-ray crystal structure; in addition, yeast Thi4 modified to use an active site histidine residue can operate by a catalytic mechanism with iron similarly to the methanogen Thi4 [56, 80].

3.2.3. Condensation of the aminopyrimidine and thiazole rings to form ThMP

Once formed, the thiamine ring precursors (i.e., THZ-P and HMP-PP) are condensed to ThMP by a ThMP synthase of the ThiE- or ThiN-type (EC 2.5.1.3).

ThiE-type ThMP synthases are widespread in all domains of life (IPR036206) and are found to catalyze the substitution of the diphosphate of HMP-PP with THZ-P to yield ThMP, CO2 and diphosphate (PPi) in bacteria [82, 83], plants [84] and yeast [85]. ThiE homologs are often bifunctional, fused to an additional catalytic domain such as HMP-P kinase (EC 2.7.4.7) [5284, 85]. ThiE serves as a ThMP synthase in certain archaea based on its requirement for growth of haloarchaea in the absence of thiamine, HMP and/or THZ [43].

ThMP synthases of the ThiN-type are also identified in archaea and bacteria, but absent in eukaryotes. ThiN domain (IPR019293) proteins are of three major types: I) fused to an N-terminal DNA binding domain (ThiR type), II) fused to an N- or C-terminal catalytic domain (e.g., ThiD) and III) standalone ThiN domains. The ThiDN proteins are ThMP synthases based on in vitro assay and complementation of ΔthiE mutants for growth in the absence of thiamine [43, 53, 54]. Fusion of the ThiN domain to the HMP/HMP-P kinase domain (ThiD) is suggested to minimize the release of HMP-PP prior to its condensation with THZ-P and, thus, channel substrate to the ThMP product [43]. ThiN domains that lack a conserved α-helix near the active site histidine are not ThMP synthases and instead can serve as apparent ligand binding sites for transcriptional regulation as in ThiR (see later discussion) [43].

3.2.4. Formation of ThDP from ThMP or thiamine

Thiamine diphosphate (ThDP), the biologically active form of thiamine, is produced from ThMP by two routes. ThMP is commonly phosphorylated to ThDP by the ATP-dependent ThiL ThMP kinase (EC 2.7.4.16 of IPR006283) in bacteria [86] and archaea [87]. Alternatively, ThMP is hydrolyzed to thiamine, and thiamine, is converted to ThDP by a Mg2+-dependent thiamine pyrophosphokinase TPK (THI80) that catalyzes thiamine + ATP ⇆ ThDP + AMP (EC 2.7.6.2) in eukaryotes [88, 89, 90, 91]. Consistent with this latter route, TPK is required for the de novo biosynthesis of thiamine in yeast [89, 90] and the ThMP phosphatase TH2 can hydrolyze ThMP to thiamine in plants [92]. TPK is also used to salvage thiamine to ThDP in eukaryotes [91, 93] and certain bacteria (TPK homolog YloS) [93]; by contrast, γ-proteobacteria use a thiamine kinase (ThiK, EC 2.7.1.89) to phosphorylate thiamine to ThMP [93] prior to ThiL-mediated phosphorylation of ThMP to ThDP. While TPK (IPR036759) homologs are conserved in some archaea, ThiK is not. Puzzling then is that certain archaea (e.g., haloarchaea and Pyrobaculum) have ThiBQP thiamine transport and ThiL ThMP kinase homologs but do not have ThiK or TPK homologs or activities (e.g., Pyrobaculum californica) [87]. Furthermore, archaea lacking TPK and ThiK homologs can transport thiamine and generate ThDP as demonstrated by growth of a ThMP synthase mutant, Haloferax volcanii ΔthiE, when supplemented with thiamine but not THZ or HMP [43, 57]. These findings suggest that certain archaea use an alternative pathway to salvage thiamine to ThDP.

Advertisement

4. Thiamine transport

Thiamine is a micronutrient that is actively transported into cells against a concentration gradient. Transport of thiamine and its precursors alleviates the need for de novo biosynthesis of thiamine. Thiamine transporters are predicted in archaea based on homology to bacterial transport systems or identification of putative transporter genes that are either in genomic synteny with thiamine biosynthesis genes or downstream of ThDP-binding riboswitch (THI- box) motifs [57, 94, 95, 96].

Bacterial transporters of thiamine and thiamine precursors, conserved in archaea, can be classified into: (i) ABC-type transporters (e.g., ThiBPQ and ThiYXZ) [47, 97, 98], (ii) a new ABC-type class termed energy coupling factor (ECF) importers [95, 99], (iii) NiaP transporters [100] of the major facilitator superfamily (MSF, IPR036259) that use an ion gradient [101] and (iv) PnuT transporters that mediate the facilitated diffusion of thiamine [102, 103]. ABC and ECF are primary active transporters that hydrolyze ATP in thiamine uptake by use of conserved ATPases (Figure 5). ECF and ABC transporters are distinguished by the type of protein used to bind solute: ECF uses a transmembrane substrate-capture protein (S component, ThiT) while ABC uses an extracytoplasmic solute binding protein (e.g., ThiB or ThiY) [95, 99]. ECF systems are typically modular in that ThiT and other S-components (e.g., the biotin specific BioY) interchangeably bind to the transmembrane (T) component of the system [95, 99, 104]. By comparison, ABC systems are not modular and have solute binding proteins (ThiB/Y) that bind to the extracytoplasmic domain of the transporter [47, 48, 105, 106].

Figure 5.

Comparison of thiamin transport by ABC and ECF importers. The nucleotide-binding domains that hydrolyze ATP and drive transporter are shown in blue. The ABC-type transmembrane domain protein (ThiP) and ECF-type Tcomponent (EcfT) are in shades of green. The soluble binding protein (ThiB, ThiY) of the ABC importer is in dark orange. The ECF importer S-components of thiamin (ThiT) and biotin (BioY), which can be swapped, are in shades of orange.

Advertisement

5. Thiamine salvage

Thiamine and its derivatives are salvaged from the outside and inside of a cell to replenish and repair the ThDP cofactor for metabolism. Thiamine salvage pathways are widespread in all domains of life and overcome the need for de novo biosynthesis of thiamine, minimize energy cost, and reduce the misincorporation of thiamine breakdown products into ThDP-dependent enzyme active sites [107].

Archaea are found to salvage thiamine and its derivatives (HMP and THZ) from the environment [43, 57] and repress the de novo biosynthesis of thiamine when thiamine levels are sufficient [43, 108]. Archaeal salvage pathways are predicted to include enzymes of de novo biosynthesis (i.e., ThiD, ThiE or ThiDN, and ThiL) with enzymes specific for salvage such as ThiM (THZ kinase, EC 2.7.1.50), TenA (aminopyrimidine aminohydrolase, EC 3.5.99.2) and/or YlmB (formylaminopyrimidine deformylase, EC 3.5.1.-) the latter speculative as it clusters to a family of proteins (IPR010182) that includes succinyl-diaminopimelate desuccinylase and YodQ of N-acetyl-beta-lysine synthesis [57] (Figure 6). ThiM is a THZ kinase in bacteria [49109, 110, 111], protists [112], and plants [113] and is predicted in archaea (e.g., UniProtKB D4GV40) based on conserved active site residues [114]. TenA homologs are subclassified into TenA_C and TenA_E [115], based on conserved active site cysteine and glutamate residues, respectively. Both types of TenA proteins are conserved in archaea. TenA_C is demonstrated to be an aminohydrolase that works in concert with the YlmB deformylase to regenerate HMP from thiamine degradation products and to function as a thiaminase II that hydrolyzes thiamine to THZ and HMP in bacteria [94, 116]. Note that thiaminase I (EC 2.5.1.2) which is secreted by certain bacteria to degrade thiamine [117, 118] is distinct from TenA. In plants, TenA_E is bifunctional in catalyzing deformylase and aminohydrolase activities to regenerate HMP from thiamine breakdown products, thus, overcoming the need for YlmB [115]. TenA_C and TenA_E are conserved in archaea and likely to function in thiamine salvage.

Figure 6.

Thiamin (vitamin B1) salvage in archaea. Abbreviations: Formylaminio-HMP, N-formyl-4-amino5-aminomethyl-2-methylpyrimidine; amino-HMP, 4amino-5-aminomethyl-2-methylpyrimidine; HMP, 4amino-5-hydroxymethyl-2-methylpyrimidine; THZ, 4methyl-5-(2-hydroxyethyl)thiazole. For additional abbreviations and coloring scheme see Figures 2-4.

Advertisement

6. Thiamine regulation

Thiamine biosynthesis, salvage and/or transport pathways are regulated by THI-box riboswitches in bacteria [119, 120, 121], eukaryotes [122, 123, 124, 125], and a few archaea (based on Rfam RF00059) [43, 96]. The THI-box riboswitch is a regulatory element of an mRNA/pre-mRNA aptamer that binds a thiamine metabolite and an expression platform that transduces the ligand binding to control gene expression [126]. In bacteria, when ThDP levels are sufficient, ThDP binds the 5′ untranslated region (UTR) of the THI-box and triggers the formation of a stem-loop structure that masks the Shine-Dalgarno (SD) sequence of the mRNA and inhibits translation initiation [119, 120, 121]. The major targets of this regulation are the mRNAs of the thiamine metabolic operons (e.g., thiCEFSGH and thiMD in E. coli) [119, 120, 121] and the ABC-type thiamine transporter (thiBPQ), with the latter based on motif analysis (Rfam RF00059). Eukaryotes (plants, fungi, and algae) also use a THI-box riboswitch to regulate expression of thiamine metabolism but do so by modulating the alternative splicing of pre-mRNAs [42, 122, 123, 124, 125, 127, 128, 129, 130]. In these eukaryotic systems, ThDP or HMP-PP binds the THI-box riboswitch of an intron located in the 5′- or 3’-UTR and causes mispairing of the splice donor (GU) and acceptor (AG) of the pre-mRNA (e.g., THIC and THI4). This incorrect pairing promotes alternative mRNA slicing and, thus, reduces thiamine biosynthesis.

Thiamine metabolism is also regulated by transcription factors, as exemplified by organisms that synthesize thiamine de novo but do not have a THI-box riboswitch motif including yeast and many archaea. In yeast, three proteins (Thi2p, Thi3p, and Pdc2p) coordinate the induction of thiamine biosynthetic (THI) gene expression in response to thiamine starvation [131, 132, 133, 134, 135, 136]. Thi3p serves as the thiamine sensor for the two transcription factors (Thi2p and Pdc2p) that bind specific DNA sequences upstream of the THI genes. When thiamine is low, Thi3p forms a ternary complex with Thi2p and Pdc2p that activates transcription of the THI genes. Once the levels of thiamine are sufficient, Thi3p binds ThDP, triggering dissociation of Thi3p from the ternary complex and reduced expression of the THI genes. In archaea from the phyla Euryarchaeota [43] and Crenarchaeota [108], a novel transcription factor, ThiR, is found to repress thiamine metabolic gene (thi4 and thiC) expression when the levels of thiamine are sufficient. ThiR is composed of an N-terminal DNA binding domain and C-terminal ThiN domain. The ThiN domain of ThiR is not catalytic, as it is missing an α-helix extension and conserved Met near the active-site His that are needed for the thiazole synthase activity of ThiDN proteins [43]. Instead the ThiN domain of ThiR serves as an apparent sensor of thiamine metabolites that triggers ThiR-mediated repression of thi4 and thiC transcription during thiamine sufficient conditions. This type of transcriptional regulation appears common in archaea based on the widespread phylogenetic distribution of ThiR homologs vs. THI-box motifs.

Advertisement

7. Future perspectives and conclusions

Thiamine is an important vitamin for improving human health [137], is a strategic nutritional supplement [138, 139], is targeted for production in probiotics [140], is useful in drug discovery including developing antimetabolites to treat cancer or fungal infections [141, 142, 143, 144], has potential for use as antitoxic agent in the food industry [145], may improve crop resistance [146], is a starting point for design of novel riboswitches [147], functions in central metabolism and unusual biocatalytic reactions [6, 7, 8, 148, 149, 150, 151], may modulate global nutrient cycles [152], and holds promise for other applications.

Discovery of the metabolic route for the de novo biosynthesis of thiamine in archaea opens a new window for the use of extremophiles in thiamine-related biotechnology applications. Archaea are designated as GRAS (generally recognized as safe) by the FDA, are amenable to genetic manipulation [153], and can readily express ThDP-dependent enzymes from foreign systems (e.g., bacterial pyruvate decarboxylase) [154]. Thus, archaea provide a useful resource to discover and optimize ThDP-dependent biocatalysts for the generation of renewable fuels and chemicals.

Archaea also provide an evolutionary perspective on the origins of thiamine biosynthesis pathways. The aminopyrimidine biosynthesis branch, composed of the radical SAM enzyme ThiC and the HMP/HMP-P kinase ThiD, appears ancient based on its functional conservation in all three domains of life. By contrast, thiazole biosynthesis can be divided into two major pathways: ThiG- and Thi4-dependent. Of these two divisions, the Thi4-type is suggested to be fairly ancient as Thi4 depends on Fe for catalytic activity, can use sulfide as a source of sulfur for thiazole ring formation, is functionally conserved in archaea and eukaryotes, and is predicted to function in certain bacteria (including anaerobes) based on genome sequencing.

Identification of genes needed to transport, synthesize, and salvage thiamine (from the three domains of life) improves understanding of how vitamin B1 may be trafficked in the environment. Finding that Thi4 is important for thiazole ring formation in eukaryotes and archaea provides new perspective on defining the organisms that synthesize thiamine de novo. Microbes that produce thiamine and thiamine precursors are suggested to be of benefit to other microbial taxa that cannot produce thiamine yet require this vitamin as a cofactor for their metabolic activity [152]. Thus, interspecies vitamin transfer may influence the metabolism of microbial consortia and global/carbon energy cycles.

Finally, thiamine is damaged by extreme conditions such as oxidation. Plant and yeast have a hydrolase (Tnr3, YJR142W) that converts the oxy- and oxo-damaged forms of ThDP into monophosphates to avoid misincorporation of the damaged thiamine molecules into the ThDP-dependent enzymes [155]. Many archaea thrive in conditions of extreme thermal and oxidative stress suggesting these microbes use unique mechanisms to avoid and/or repair damaged ThDP for use as a cofactor.

Advertisement

Acknowledgments

Funds for this project were awarded to JM-F through the Bilateral NSF/BIO-BBSRC program (NSF 1642283), the U.S. Department of Energy, Office of Basic Energy Sciences, Division of Chemical Sciences, Geosciences and Biosciences, Physical Biosciences Program (DOE DE-FG02-05ER15650) and the National Institutes of Health (NIH R01 GM57498).

Advertisement

Conflict of interest

The author has no conflict of interest to declare.

References

  1. 1. Bettendorff L, Wins P. Thiamine diphosphate in biological chemistry: New aspects of thiamine metabolism, especially triphosphate derivatives acting other than as cofactors. The FEBS Journal. 2009;276(11):2917-2925
  2. 2. Frederich M, Delvaux D, Gigliobianco T, Gangolf M, Dive G, Mazzucchelli G, et al. Thiaminylated adenine nucleotides. Chemical synthesis, structural characterization and natural occurrence. The FEBS Journal. 2009;276(12):3256-3268
  3. 3. Xavier JC, Patil KR, Rocha I. Integration of biomass formulations of genome-scale metabolic models with experimental data reveals universally essential cofactors in prokaryotes. Metabolic Engineering. 2017;39:200-208
  4. 4. Zhang K, Bian J, Deng Y, Smith A, Nunez RE, Li MB, et al. Lyme disease spirochaete Borrelia burgdorferi does not require thiamine. Nature Microbiology. 2016;2:16213
  5. 5. Müller M, Sprenger GA, Pohl M. CC bond formation using ThDP-dependent lyases. Current Opinion in Chemical Biology. 2013;17(2):261-270
  6. 6. Jordan F. Current mechanistic understanding of thiamine diphosphate-dependent enzymatic reactions. Natural Product Reports. 2003;20(2):184-201
  7. 7. Nemeria N, Binshtein E, Patel H, Balakrishnan A, Vered I, Shaanan B, et al. Glyoxylate carboligase: A unique thiamine diphosphate-dependent enzyme that can cycle between the 4′-aminopyrimidinium and 1′,4′-iminopyrimidine tautomeric forms in the absence of the conserved glutamate. Biochemistry. 2012;51(40):7940-7952
  8. 8. Shaanan B, Chipman DM. Reaction mechanisms of thiamine diphosphate enzymes: New insights into the role of a conserved glutamate residue. The FEBS Journal. 2009;276(9):2447-2453
  9. 9. Schellenberger A. Sixty years of thiamine diphosphate biochemistry. Biochimica et Biophysica Acta (BBA) - Protein Structure and Molecular Enzymology. 1998;1385(2):177-186
  10. 10. Stetter H. Catalyzed addition of aldehydes to activated double bonds - a new synthetic approach. Angewandte Chemie International Edition in English. 1976;15(11):639-647
  11. 11. Schellenberger A, Hubner G, Neef H. Cofactor designing in functional analysis of thiamine diphosphate enzymes. Methods in Enzymology. 1997;279:131-146
  12. 12. Frank RA, Titman CM, Pratap JV, Luisi BF, Perham RN. A molecular switch and proton wire synchronize the active sites in thiamine enzymes. Science. 2004;306(5697):872-876
  13. 13. Chabriere E, Charon MH, Volbeda A, Pieulle L, Hatchikian EC, Fontecilla-Camps JC. Crystal structures of the key anaerobic enzyme pyruvate: Ferredoxin oxidoreductase, free and in complex with pyruvate. Nature Structural Biology. 1999;6(2):182-190
  14. 14. Caines ME, Elkins JM, Hewitson KS, Schofield CJ. Crystal structure and mechanistic implications of N2-(2-carboxyethyl)arginine synthase, the first enzyme in the clavulanic acid biosynthesis pathway. The Journal of Biological Chemistry. 2004;279(7):5685-5692
  15. 15. Xiang S, Usunow G, Lange G, Busch M, Tong L. Crystal structure of 1-deoxy-D-xylulose 5-phosphate synthase, a crucial enzyme for isoprenoids biosynthesis. The Journal of Biological Chemistry. 2007;282(4):2676-2682
  16. 16. Yan Z, Maruyama A, Arakawa T, Fushinobu S, Wakagi T. Crystal structures of archaeal 2-oxoacid: Ferredoxin oxidoreductases from Sulfolobus tokodaii. Scientific Reports. 2016;6:33061
  17. 17. Plaga W, Lottspeich F, Oesterhelt D. Improved purification, crystallization and primary structure of pyruvate: Ferredoxin oxidoreductase from Halobacterium halobium. European Journal of Biochemistry. 1992;205(1):391-397
  18. 18. Kerscher L, Oesterhelt D. Purification and properties of two 2-oxoacid: Ferredoxin oxidoreductases from Halobacterium halobium. European Journal of Biochemistry. 1981;116(3):587-594
  19. 19. Kunow J, Linder D, Thauer RK. Pyruvate: Ferredoxin oxidoreductase from the sulfate-reducing Archaeoglobus fulgidus: Molecular composition, catalytic properties, and sequence alignments. Archives of Microbiology. 1995;163(1):21-28
  20. 20. Mai X, Adams MW. Indolepyruvate ferredoxin oxidoreductase from the hyperthermophilic archaeon Pyrococcus furiosus. A new enzyme involved in peptide fermentation. The Journal of Biological Chemistry. 1994;269(24):16726-16732
  21. 21. Smith ET, Blamey JM, Adams MW. Pyruvate ferredoxin oxidoreductases of the hyperthermophilic archaeon, Pyrococcus furiosus, and the hyperthermophilic bacterium, Thermotoga maritima, have different catalytic mechanisms. Biochemistry. 1994;33(4):1008-1016
  22. 22. Blamey JM, Adams MW. Purification and characterization of pyruvate ferredoxin oxidoreductase from the hyperthermophilic archaeon Pyrococcus furiosus. Biochimica et Biophysica Acta. 1993;1161(1):19-27
  23. 23. Kletzin A, Adams MW. Molecular and phylogenetic characterization of pyruvate and 2-ketoisovalerate ferredoxin oxidoreductases from Pyrococcus furiosus and pyruvate ferredoxin oxidoreductase from Thermotoga maritima. Journal of Bacteriology. 1996;178(1):248-257
  24. 24. Heider J, Mai X, Adams MW. Characterization of 2-ketoisovalerate ferredoxin oxidoreductase, a new and reversible coenzyme A-dependent enzyme involved in peptide fermentation by hyperthermophilic archaea. Journal of Bacteriology. 1996;178(3):780-787
  25. 25. Bock AK, Kunow J, Glasemacher J, Schonheit P. Catalytic properties, molecular composition and sequence alignments of pyruvate: Ferredoxin oxidoreductase from the methanogenic archaeon Methanosarcina barkeri (strain Fusaro). European Journal of Biochemistry. 1996;237(1):35-44
  26. 26. Mai X, Adams MW. Characterization of a fourth type of 2-keto acid-oxidizing enzyme from a hyperthermophilic archaeon: 2-ketoglutarate ferredoxin oxidoreductase from Thermococcus litoralis. Journal of Bacteriology. 1996;178(20):5890-5896
  27. 27. Zhang Q, Iwasaki T, Wakagi T, Oshima T. 2-oxoacid:Ferredoxin oxidoreductase from the thermoacidophilic archaeon, Sulfolobus sp. strain 7. Journal of Biochemistry. 1996;120(3):587-599
  28. 28. Ma K, Hutchins A, Sung SJ, Adams MW. Pyruvate ferredoxin oxidoreductase from the hyperthermophilic archaeon, Pyrococcus furiosus, functions as a CoA-dependent pyruvate decarboxylase. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(18):9608-9613
  29. 29. Bock AK, Schonheit P, Teixeira M. The iron-sulfur centers of the pyruvate:Ferredoxin oxidoreductase from Methanosarcina barkeri (Fusaro). FEBS Letters. 1997;414(2):209-212
  30. 30. Jolley KA, Maddocks DG, Gyles SL, Mullan Z, Tang SL. Dyall-smith ML, et al. 2-Oxoacid dehydrogenase multienzyme complexes in the halophilic Archaea? Gene sequences and protein structural predictions. Microbiology. 2000;146(Pt 5):1061-1069
  31. 31. Ozawa Y, Nakamura T, Kamata N, Yasujima D, Urushiyama A, Yamakura F, et al. Thermococcus profundus 2-ketoisovalerate ferredoxin oxidoreductase, a key enzyme in the archaeal energy-producing amino acid metabolic pathway. Journal of Biochemistry. 2005;137(1):101-107
  32. 32. van Ooyen J, Soppa J. Three 2-oxoacid dehydrogenase operons in Haloferax volcanii: Expression, deletion mutants and evolution. Microbiology 2007;153(Pt 10):3303-3313
  33. 33. Jahn U, Huber H, Eisenreich W, Hugler M, Fuchs G. Insights into the autotrophic CO2 fixation pathway of the archaeon Ignicoccus hospitalis: Comprehensive analysis of the central carbon metabolism. Journal of Bacteriology. 2007;189(11):4108-4119
  34. 34. Sarmiento F, Ellison CK, Whitman WB. Genetic confirmation of the role of sulfopyruvate decarboxylase in coenzyme M biosynthesis in Methanococcus maripaludis. Archaea. 2013;2013:185250
  35. 35. Graupner M, Xu H, White RH. Identification of the gene encoding sulfopyruvate decarboxylase, an enzyme involved in biosynthesis of coenzyme M. Journal of Bacteriology. 2000;182(17):4862-4867
  36. 36. Xing RY, Whitman WB. Sulfometuron methyl-sensitive and -resistant acetolactate synthases of the archaebacteria Methanococcus spp. Journal of Bacteriology. 1987;169(10):4486-4492
  37. 37. Xing R, Whitman WB. Purification and characterization of the oxygen-sensitive acetohydroxy acid synthase from the archaebacterium Methanococcus aeolicus. Journal of Bacteriology. 1994;176(5):1207-1213
  38. 38. Yu JP, Ladapo J, Whitman WB. Pathway of glycogen metabolism in Methanococcus maripaludis. Journal of Bacteriology. 1994;176(2):325-332
  39. 39. Soderberg T. Biosynthesis of ribose-5-phosphate and erythrose-4-phosphate in archaea: A phylogenetic analysis of archaeal genomes. Archaea. 2005;1(5):347-352
  40. 40. Palmer LD, Downs DM. The thiamine biosynthetic enzyme ThiC catalyzes multiple turnovers and is inhibited by S-adenosylmethionine (AdoMet) metabolites. The Journal of Biological Chemistry. 2013;288(42):30693-30699
  41. 41. Martinez-Gomez NC, Downs DM. ThiC is an [Fe-S] cluster protein that requires AdoMet to generate the 4-amino-5-hydroxymethyl-2-methylpyrimidine moiety in thiamine synthesis. Biochemistry. 2008;47(35):9054-9056
  42. 42. Raschke M, Burkle L, Muller N, Nunes-Nesi A, Fernie AR, Arigoni D, et al. Vitamin B1 biosynthesis in plants requires the essential iron sulfur cluster protein, THIC. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(49):19637-19642
  43. 43. Hwang S, Cordova B, Abdo M, Pfeiffer F, Maupin-Furlow JA. ThiN as a versatile domain of transcriptional repressors and catalytic enzymes of thiamine biosynthesis. Journal of Bacteriology. 2017;199(7):e00810-16
  44. 44. Chatterjee A, Hazra AB, Abdelwahed S, Hilmey DG, Begley TP. A “radical dance” in thiamine biosynthesis: Mechanistic analysis of the bacterial hydroxymethylpyrimidine phosphate synthase. Angewandte Chemie (International Ed. in English). 2010;49(46):8653-8656
  45. 45. Coquille S, Roux C, Fitzpatrick TB, Thore S. The last piece in the vitamin B1 biosynthesis puzzle: Structural and functional insight into yeast 4-amino-5-hydroxymethyl-2-methylpyrimidine phosphate (HMP-P) synthase. The Journal of Biological Chemistry. 2012;287(50):42333-42343
  46. 46. Wightman R, Meacock PA. The THI5 gene family of Saccharomyces cerevisiae: Distribution of homologues among the hemiascomycetes and functional redundancy in the aerobic biosynthesis of thiamine from pyridoxine. Microbiology. 2003;149(Pt 6):1447-1460
  47. 47. Bale S, Rajashankar KR, Perry K, Begley TP, Ealick SE. HMP binding protein ThiY and HMP-P synthase THI5 are structural homologues. Biochemistry. 2010;49(41):8929-8936
  48. 48. Rodionova IA, Li X, Plymale AE, Motamedchaboki K, Konopka AE, Romine MF, et al. Genomic distribution of B-vitamin auxotrophy and uptake transporters in environmental bacteria from the Chloroflexi phylum. Environmental Microbiology Reports. 2015;7(2):204-210
  49. 49. Mizote T, Tsuda M, Smith DD, Nakayama H, Nakazawa T. Cloning and characterization of the thiD/J gene of Escherichia coli encoding a thiamine-synthesizing bifunctional enzyme, hydroxymethylpyrimidine kinase/phosphomethylpyrimidine kinase. Microbiology. 1999;145(Pt 2):495-501
  50. 50. Reddick JJ, Kinsland C, Nicewonger R, Christian T, Downs DM, Winkler ME, et al. Overexpression, purification and characterization of two pyrimidine kinases involved in the biosynthesis of thiamine: 4-amino-5-hydroxymethyl-2-methylpyrimidine kinase and 4-amino-5-hydroxymethyl-2-methylpyrimidine phosphate kinase. Tetrahedron. 1998;54:15983-15991
  51. 51. Kawasaki Y, Onozuka M, Mizote T, Nosaka K. Biosynthesis of hydroxymethylpyrimidine pyrophosphate in Saccharomyces cerevisiae. Current Genetics. 2005;47(3):156-162
  52. 52. Thamm AM, Li G, Taja-Moreno M, Gerdes SY, de Crecy-Lagard V, Bruner SD, et al. A strictly monofunctional bacterial hydroxymethylpyrimidine phosphate kinase precludes damaging errors in thiamine biosynthesis. Biochemical Journal. 2017;474:2887-2895
  53. 53. Hayashi M, Kobayashi K, Esaki H, Konno H, Akaji K, Tazuya K, et al. Enzymatic and structural characterization of an archaeal thiamine phosphate synthase. Biochimica et Biophysica Acta. 2014;1844(4):803-809
  54. 54. Morett E, Korbel JO, Rajan E, Saab-Rincon G, Olvera L, Olvera M, et al. Systematic discovery of analogous enzymes in thiamine biosynthesis. Nature Biotechnology. 2003;21(7):790-795
  55. 55. Chatterjee A, Abeydeera ND, Bale S, Pai PJ, Dorrestein PC, Russell DH, et al. Saccharomyces cerevisiae THI4p is a suicide thiamine thiazole synthase. Nature. 2011;478(7370):542-546
  56. 56. Eser BE, Zhang X, Chanani PK, Begley TP, Ealick SE. From suicide enzyme to catalyst: The iron-dependent sulfide transfer in Methanococcus jannaschii thiamine thiazole biosynthesis. Journal of the American Chemical Society. 2016;138(11):3639-3642
  57. 57. Hwang S, Cordova B, Chavarria N, Elbanna D, McHugh S, Rojas J, et al. Conserved active site cysteine residue of archaeal THI4 homolog is essential for thiamine biosynthesis in Haloferax volcanii. BMC Microbiology. 2014;14:260
  58. 58. Park JH, Dorrestein PC, Zhai H, Kinsland C, McLafferty FW, Begley TP. Biosynthesis of the thiazole moiety of thiamine pyrophosphate (vitamin B1). Biochemistry. 2003;42(42):12430-12438
  59. 59. Dorrestein PC, Zhai H, Taylor SV, McLafferty FW, Begley TP. The biosynthesis of the thiazole phosphate moiety of thiamine (vitamin B1): The early steps catalyzed by thiazole synthase. Journal of the American Chemical Society. 2004;126(10):3091-3096
  60. 60. Dorrestein PC, Zhai H, McLafferty FW, Begley TP. The biosynthesis of the thiazole phosphate moiety of thiamine: The sulfur transfer mediated by the sulfur carrier protein ThiS. Chemistry & Biology. 2004;11(10):1373-1381
  61. 61. Zhang J, Zhang B, Zhao Y, Yang X, Huang M, Cui P, et al. Snapshots of catalysis: Structure of covalently bound substrate trapped in Mycobacterium tuberculosis thiazole synthase (ThiG). Biochemical and Biophysical Research Communications. 2018;497(1):214-219
  62. 62. Settembre EC, Dorrestein PC, Park JH, Augustine AM, Begley TP, Ealick SE. Structural and mechanistic studies on ThiO, a glycine oxidase essential for thiamine biosynthesis in Bacillus subtilis. Biochemistry. 2003;42(10):2971-2981
  63. 63. Nishiya Y, Imanaka T. Purification and characterization of a novel glycine oxidase from Bacillus subtilis. FEBS Letters. 1998;438(3):263-266
  64. 64. Job V, Marcone GL, Pilone MS, Pollegioni L. Glycine oxidase from Bacillus subtilis. Characterization of a new flavoprotein. The Journal of Biological Chemistry. 2002;277(9):6985-6993
  65. 65. Pedotti M, Rosini E, Molla G, Moschetti T, Savino C, Vallone B, et al. Glyphosate resistance by engineering the flavoenzyme glycine oxidase. The Journal of Biological Chemistry. 2009;284(52):36415-36423
  66. 66. Kriek M, Martins F, Leonardi R, Fairhurst SA, Lowe DJ, Roach PL. Thiazole synthase from Escherichia coli: An investigation of the substrates and purified proteins required for activity in vitro. The Journal of Biological Chemistry. 2007;282(24):17413-17423
  67. 67. Challand MR, Martins FT, Roach PL. Catalytic activity of the anaerobic tyrosine lyase required for thiamine biosynthesis in Escherichia coli. The Journal of Biological Chemistry. 2010;285(8):5240-5248
  68. 68. Kriek M, Martins F, Challand MR, Croft A, Roach PL. Thiamine biosynthesis in Escherichia coli: Identification of the intermediate and by-product derived from tyrosine. Angewandte Chemie (International Ed. in English). 2007;46(48):9223-9226
  69. 69. Sprenger GA, Schorken U, Wiegert T, Grolle S, de Graaf AA, Taylor SV, et al. Identification of a thiamine-dependent synthase in Escherichia coli required for the formation of the 1-deoxy-D-xylulose 5-phosphate precursor to isoprenoids, thiamine, and pyridoxol. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(24):12857-12862
  70. 70. Lois LM, Campos N, Putra SR, Danielsen K, Rohmer M, Boronat A. Cloning and characterization of a gene from Escherichia coli encoding a transketolase-like enzyme that catalyzes the synthesis of D-1-deoxyxylulose 5-phosphate, a common precursor for isoprenoid, thiamine, and pyridoxol biosynthesis. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(5):2105-2110
  71. 71. Lauhon CT, Kambampati R. The iscS gene in Escherichia coli is required for the biosynthesis of 4-thiouridine, thiamine, and NAD. The Journal of Biological Chemistry. 2000;275(26):20096-20103
  72. 72. Flint DH. Escherichia coli contains a protein that is homologous in function and N-terminal sequence to the protein encoded by the nifS gene of Azotobacter vinelandii and that can participate in the synthesis of the Fe-S cluster of dihydroxy-acid dehydratase. The Journal of Biological Chemistry. 1996;271(27):16068-16074
  73. 73. Xi J, Ge Y, Kinsland C, McLafferty FW, Begley TP. Biosynthesis of the thiazole moiety of thiamine in Escherichia coli: Identification of an acyldisulfide-linked protein–protein conjugate that is functionally analogous to the ubiquitin/E1 complex. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(15):8513-8518
  74. 74. Mueller EG, Palenchar PM, Buck CJ. The role of the cysteine residues of ThiI in the generation of 4-thiouridine in tRNA. The Journal of Biological Chemistry. 2001;276(36):33588-33595
  75. 75. Martinez-Gomez NC, Palmer LD, Vivas E, Roach PL, Downs DM. The rhodanese domain of ThiI is both necessary and sufficient for synthesis of the thiazole moiety of thiamine in Salmonella enterica. Journal of Bacteriology. 2011;193(18):4582-4587
  76. 76. Kambampati R, Lauhon CT. Evidence for the transfer of sulfane sulfur from IscS to ThiI during the in vitro biosynthesis of 4-thiouridine in Escherichia coli tRNA. The Journal of Biological Chemistry. 2000;275(15):10727-10730
  77. 77. Finn MW, Tabita FR. Modified pathway to synthesize ribulose 1,5-bisphosphate in methanogenic archaea. Journal of Bacteriology. 2004;186(19):6360-6366
  78. 78. Sato T, Atomi H, Imanaka T. Archaeal type III RuBisCOs function in a pathway for AMP metabolism. Science. 2007;315(5814):1003-1006
  79. 79. Gogoi P, Kanaujia SP. A presumed homologue of the regulatory subunits of eIF2B functions as ribose-1,5-bisphosphate isomerase in Pyrococcus horikoshii OT3. Scientific Reports. 2018;8(1):1891
  80. 80. Zhang X, Eser BE, Chanani PK, Begley TP, Ealick SE. Structural basis for iron-mediated sulfur transfer in archael and yeast thiazole synthases. Biochemistry. 2016;55(12):1826-1838
  81. 81. Godoi PH, Galhardo RS, Luche DD, Van Sluys MA, Menck CF, Oliva G. Structure of the thiazole biosynthetic enzyme THI1 from Arabidopsis thaliana. The Journal of Biological Chemistry 2006;281(41):30957-30966
  82. 82. Backstrom AD, McMordie RAS, Begley TP. Biosynthesis of thiamine I: The function of the thiE gene product. Journal of the American Chemical Society. 1995;117(8):2351-2352
  83. 83. Chiu HJ, Reddick JJ, Begley TP, Ealick SE. Crystal structure of thiamine phosphate synthase from Bacillus subtilis at 1.25 Å resolution. Biochemistry. 1999;38(20):6460-6470
  84. 84. Suk Kim Y, Nosaka K, Downs DM, Myoung Kwak J, Park D, Kyung Chung I, et al. A Brassica cDNA clone encoding a bifunctional hydroxymethylpyrimidine kinase/thiamine-phosphate pyrophosphorylase involved in thiamine biosynthesis. Plant Molecular Biology. 1998;37(6):955-966
  85. 85. Paul D, Chatterjee A, Begley TP, Ealick SE. Domain organization in Candida glabrata THI6, a bifunctional enzyme required for thiamine biosynthesis in eukaryotes. Biochemistry. 2010;49(45):9922-9934
  86. 86. Webb E, Downs D. Characterization of thiL, encoding thiamine-monophosphate kinase, in Salmonella typhimurium. The Journal of Biological Chemistry. 1997;272(25):15702-15707
  87. 87. Hayashi M, Nosaka K. Characterization of thiamine phosphate kinase in the hyperthermophilic archaeon Pyrobaculum calidifontis. Journal of Nutritional Science and Vitaminology (Tokyo). 2015;61(5):369-374
  88. 88. Voskoboyev AI, Ostrovsky YM. Thiamine pyrophosphokinase: Structure, properties, and role in thiamine metabolism. Annals of the New York Academy of Sciences. 1982;378:161-176
  89. 89. Nosaka K, Kaneko Y, Nishimura H, Iwashima A. Isolation and characterization of a thiamine pyrophosphokinase gene, THI80, from Saccharomyces cerevisiae. The Journal of Biological Chemistry. 1993;268(23):17440-17447
  90. 90. Fankhauser H, Zurlinden A, Schweingruber AM, Edenharter E, Schweingruber ME. Schizosaccharomyces pombe thiamine pyrophosphokinase is encoded by gene tnr3 and is a regulator of thiamine metabolism, phosphate metabolism, mating, and growth. The Journal of Biological Chemistry. 1995;270(47):28457-28462
  91. 91. Nosaka K, Onozuka M, Nishino H, Nishimura H, Kawasaki Y, Ueyama H. Molecular cloning and expression of a mouse thiamine pyrophosphokinase cDNA. The Journal of Biological Chemistry. 1999;274(48):34129-34133
  92. 92. Mimura M, Zallot R, Niehaus TD, Hasnain G, Gidda SK, Nguyen TN, et al. Arabidopsis TH2 encodes the orphan enzyme thiamine monophosphate phosphatase. The Plant Cell. 2016;28(10):2683-2696
  93. 93. Melnick J, Lis E, Park JH, Kinsland C, Mori H, Baba T, et al. Identification of the two missing bacterial genes involved in thiamine salvage: Thiamine pyrophosphokinase and thiamine kinase. Journal of Bacteriology. 2004;186(11):3660-3662
  94. 94. Jenkins AH, Schyns G, Potot S, Sun G, Begley TP. A new thiamine salvage pathway. Nature Chemical Biology. 2007;3(8):492-497
  95. 95. Majsnerowska M, Ter Beek J, Stanek WK, Duurkens RH, Slotboom DJ. Competition between different S-components for the shared energy coupling factor module in energy coupling factor transporters. Biochemistry 2015;54(31):4763-4766
  96. 96. Rodionov DA, Vitreschak AG, Mironov AA, Gelfand MS. Comparative genomics of thiamine biosynthesis in procaryotes. New genes and regulatory mechanisms. The Journal of Biological Chemistry. 2002;277(50):48949-48959
  97. 97. Webb E, Claas K, Downs D. thiBPQ encodes an ABC transporter required for transport of thiamine and thiamine pyrophosphate in Salmonella typhimurium. The Journal of Biological Chemistry. 1998;273(15):8946-8950
  98. 98. Dermoun Z, Foulon A, Miller MD, Harrington DJ, Deacon AM, Sebban-Kreuzer C, et al. TM0486 from the hyperthermophilic anaerobe Thermotoga maritima is a thiamine-binding protein involved in response of the cell to oxidative conditions. Journal of Molecular Biology. 2010;400(3):463-476
  99. 99. Rodionov DA, Hebbeln P, Eudes A, ter Beek J, Rodionova IA, Erkens GB, et al. A novel class of modular transporters for vitamins in prokaryotes. Journal of Bacteriology. 2009;191(1):42-51
  100. 100. Jeanguenin L, Lara-Nunez A, Rodionov DA, Osterman AL, Komarova NY, Rentsch D, et al. Comparative genomics and functional analysis of the NiaP family uncover nicotinate transporters from bacteria, plants, and mammals. Functional & Integrative Genomics. 2012;12(1):25-34
  101. 101. Zhang XC, Zhao Y, Heng J, Jiang D. Energy coupling mechanisms of MFS transporters. Protein Science. 2015;24(10):1560-1579
  102. 102. Genee HJ, Bali AP, Petersen SD, Siedler S, Bonde MT, Gronenberg LS, et al. Functional mining of transporters using synthetic selections. Nature Chemical Biology. 2016;12(12):1015-1022
  103. 103. Jaehme M, Singh R, Garaeva AA, Duurkens RH, Slotboom DJ. PnuT uses a facilitated diffusion mechanism for thiamine uptake. The Journal of General Physiology. 2018;150(1):41-50
  104. 104. ter Beek J, Duurkens RH, Erkens GB, Slotboom DJ. Quaternary structure and functional unit of energy coupling factor (ECF)-type transporters. The Journal of Biological Chemistry 2011;286(7):5471-5475
  105. 105. Hollenbach AD, Dickson KA, Washabaugh MW. Overexpression, purification, and characterization of the periplasmic space thiamine-binding protein of the thiamine traffic ATPase in Escherichia coli. Protein Expression and Purification. 2002;25(3):508-518
  106. 106. Soriano EV, Rajashankar KR, Hanes JW, Bale S, Begley TP, Ealick SE. Structural similarities between thiamine-binding protein and thiaminase-I suggest a common ancestor. Biochemistry. 2008;47(5):1346-1357
  107. 107. Jurgenson CT, Begley TP, Ealick SE. The structural and biochemical foundations of thiamine biosynthesis. Annual Review of Biochemistry. 2009;78:569-603
  108. 108. Rodionov DA, Leyn SA, Li X, Rodionova IA. A novel transcriptional regulator related to thiamine phosphate synthase controls thiamine metabolism genes in Archaea. Journal of Bacteriology. 2017;199(4)
  109. 109. Tani Y, Kimura K, Mihara H. Purification and properties of 4-methyl-5-hydroxyethylthiazole kinase from Escherichia coli. Bioscience, Biotechnology, and Biochemistry. 2016;80(3):514-517
  110. 110. Zhang Y, Taylor SV, Chiu HJ, Begley TP. Characterization of the Bacillus subtilis thiC operon involved in thiamine biosynthesis. Journal of Bacteriology. 1997;179(9):3030-3035
  111. 111. Mizote T, Nakayama H. The thiM locus and its relation to phosphorylation of hydroxyethylthiazole in Escherichia coli. Journal of Bacteriology. 1989;171(6):3228-3232
  112. 112. Wrenger C, Eschbach ML, Muller IB, Laun NP, Begley TP, Walter RD. Vitamin B1 de novo synthesis in the human malaria parasite Plasmodium falciparum depends on external provision of 4-amino-5-hydroxymethyl-2-methylpyrimidine. Biological Chemistry. 2006;387(1):41-51
  113. 113. Yazdani M, Zallot R, Tunc-Ozdemir M, de Crécy-Lagard V, Shintani DK, Hanson AD. Identification of the thiamine salvage enzyme thiazole kinase in Arabidopsis and maize. Phytochemistry. 2013;94:68-73
  114. 114. Drebes J, Kunz M, Windshugel B, Kikhney AG, Muller IB, Eberle RJ, et al. Structure of ThiM from vitamin B1 biosynthetic pathway of Staphylococcus aureus - insights into a novel pro-drug approach addressing MRSA infections. Scientific Reports. 2016;6:22871
  115. 115. Zallot R, Yazdani M, Goyer A, Ziemak MJ, Guan JC, McCarty DR, et al. Salvage of the thiamine pyrimidine moiety by plant TenA proteins lacking an active-site cysteine. The Biochemical Journal. 2014;463(1):145-155
  116. 116. Müller IB, Bergmann B, Groves MR, Couto I, Amaral L, Begley TP, et al. The vitamin B1 metabolism of Staphylococcus aureus is controlled at enzymatic and transcriptional levels. PLoS One. 2009;4(11):e7656
  117. 117. Costello CA, Kelleher NL, Abe M, McLafferty FW, Begley TP. Mechanistic studies on thiaminase I. Overexpression and identification of the active site nucleophile. The Journal of Biological Chemistry. 1996;271(7):3445-3452
  118. 118. Cooper LE, O'Leary SE, Begley TP. Biosynthesis of a thiamine antivitamin in Clostridium botulinum. Biochemistry. 2014;53(14):2215-2217
  119. 119. Miranda-Ríos J, Navarro M, Soberón M. A conserved RNA structure (thi box) is involved in regulation of thiamine biosynthetic gene expression in bacteria. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(17):9736-9741
  120. 120. Winkler W, Nahvi A, Breaker RR. Thiamine derivatives bind messenger RNAs directly to regulate bacterial gene expression. Nature. 2002;419(6910):952-956
  121. 121. Serganov A, Polonskaia A, Phan AT, Breaker RR, Patel DJ. Structural basis for gene regulation by a thiamine pyrophosphate-sensing riboswitch. Nature. 2006;441(7097):1167-1171
  122. 122. Thore S, Leibundgut M, Ban N. Structure of the eukaryotic thiamine pyrophosphate riboswitch with its regulatory ligand. Science. 2006;312(5777):1208-1211
  123. 123. Cheah MT, Wachter A, Sudarsan N, Breaker RR. Control of alternative RNA splicing and gene expression by eukaryotic riboswitches. Nature. 2007;447(7143):497-500
  124. 124. Wachter A, Tunc-Ozdemir M, Grove BC, Green PJ, Shintani DK, Breaker RR. Riboswitch control of gene expression in plants by splicing and alternative 3′ end processing of mRNAs. The Plant Cell. 2007;19(11):3437-3450
  125. 125. Croft MT, Moulin M, Webb ME, Smith AG. Thiamine biosynthesis in algae is regulated by riboswitches. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(52):20770-20775
  126. 126. Garst AD, Batey RT. A switch in time: Detailing the life of a riboswitch. Biochimica et Biophysica Acta. 2009;1789(9-10):584-591
  127. 127. Bocobza SE, Aharoni A. Small molecules that interact with RNA: Riboswitch-based gene control and its involvement in metabolic regulation in plants and algae. The Plant Journal. 2014;79(4):693-703
  128. 128. Bocobza S, Adato A, Mandel T, Shapira M, Nudler E, Aharoni A. Riboswitch-dependent gene regulation and its evolution in the plant kingdom. Genes & Development. 2007;21(22):2874-2879
  129. 129. Kubodera T, Watanabe M, Yoshiuchi K, Yamashita N, Nishimura A, Nakai S, et al. Thiamine-regulated gene expression of Aspergillus oryzae thiA requires splicing of the intron containing a riboswitch-like domain in the 5'-UTR. FEBS Letters. 2003;555(3):516-520
  130. 130. Moulin M, Nguyen GT, Scaife MA, Smith AG, Fitzpatrick TB. Analysis of Chlamydomonas thiamine metabolism in vivo reveals riboswitch plasticity. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(36):14622-14627
  131. 131. Hohmann S, Meacock PA. Thiamine metabolism and thiamine diphosphate-dependent enzymes in the yeast Saccharomyces cerevisiae: Genetic regulation. Biochimica et Biophysica Acta. 1998;1385(2):201-219
  132. 132. Harbison CT, Gordon DB, Lee TI, Rinaldi NJ, Macisaac KD, Danford TW, et al. Transcriptional regulatory code of a eukaryotic genome. Nature. 2004;431(7004):99-104
  133. 133. Nosaka K, Esaki H, Onozuka M, Konno H, Hattori Y, Akaji K. Facilitated recruitment of Pdc2p, a yeast transcriptional activator, in response to thiamine starvation. FEMS Microbiology Letters. 2012;330(2):140-147
  134. 134. Nosaka K, Onozuka M, Konno H, Kawasaki Y, Nishimura H, Sano M, et al. Genetic regulation mediated by thiamine pyrophosphate-binding motif in Saccharomyces cerevisiae. Molecular Microbiology. 2005;58(2):467-479
  135. 135. Nosaka K, Onozuka M, Konno H, Akaji K. Thiamine-dependent transactivation activity of PDC2 in Saccharomyces cerevisiae. FEBS Letters. 2008;582(29):3991-3996
  136. 136. Nosaka K. Recent progress in understanding thiamine biosynthesis and its genetic regulation in Saccharomyces cerevisiae. Applied Microbiology and Biotechnology. 2006;72(1):30-40
  137. 137. Liu D, Ke Z, Luo J. Thiamine deficiency and neurodegeneration: The interplay among oxidative stress, endoplasmic reticulum stress, and autophagy. Molecular Neurobiology. 2017;54(7):5440-5448
  138. 138. Revuelta JL, Buey RM, Ledesma-Amaro R, Vandamme EJ. Microbial biotechnology for the synthesis of (pro)vitamins, biopigments and antioxidants: Challenges and opportunities. Microbial Biotechnology. 2016;9(5):564-567
  139. 139. Wolak N, Zawrotniak M, Gogol M, Kozik A, Rapala-Kozik M. Vitamins B1, B2, B3 and B9–Occurrence, biosynthesis pathways and functions in human nutrition. Mini Reviews in Medicinal Chemistry. 2017;17(12):1075-1111
  140. 140. LeBlanc JG, Chain F, Martin R, Bermudez-Humaran LG, Courau S, Langella P. Beneficial effects on host energy metabolism of short-chain fatty acids and vitamins produced by commensal and probiotic bacteria. Microbial Cell Factories. 2017;16(1):79
  141. 141. Tylicki A, Lotowski Z, Siemieniuk M, Ratkiewicz A. Thiamine and selected thiamine antivitamins–biological activity and methods of synthesis. Bioscience Reports. 2018;38(1):BSR20171148
  142. 142. Lu'o'ng KV, Nguyen LT. The role of thiamine in cancer: Possible genetic and cellular signaling mechanisms. Cancer Genomics & Proteomics. 2013;10(4):169-185
  143. 143. Chhabria MT, Patel S, Modi P, Brahmkshatriya PS. Thiazole: A review on chemistry, synthesis and therapeutic importance of its derivatives. Current Topics in Medicinal Chemistry. 2016;16(26):2841-2862
  144. 144. Rouf A, Tanyeli C. Bioactive thiazole and benzothiazole derivatives. European Journal of Medicinal Chemistry. 2015;97:911-927
  145. 145. Nazemi L, Kordbacheh P, Daei Ghazvini R, Moazeni M, Akbari Dana M, Rezaie S. Effects of thiamine on growth, aflatoxin production, and aflr gene expression in A. parasiticus. Current Medical Mycology. 2015;1(1):26-34
  146. 146. Boubakri H, Gargouri M, Mliki A, Brini F, Chong J, Jbara M. Vitamins for enhancing plant resistance. Planta. 2016;244(3):529-543
  147. 147. Lunse CE, Scott FJ, Suckling CJ, Mayer G. Novel TPP-riboswitch activators bypass metabolic enzyme dependency. Frontiers in Chemistry. 2014;2:53
  148. 148. Lu T, Li X, Gu L, Zhang Y. Vitamin B1-catalyzed acetoin formation from acetaldehyde: A key step for upgrading bioethanol to bulk C(4) chemicals. ChemSusChem. 2014;7(9):2423-2426
  149. 149. Resch V, Schrittwieser JH, Siirola E, Kroutil W. Novel carbon-carbon bond formations for biocatalysis. Current Opinion in Biotechnology. 2011;22(6):793-799
  150. 150. Muller M, Gocke D, Pohl M. Thiamin diphosphate in biological chemistry: Exploitation of diverse thiamin diphosphate-dependent enzymes for asymmetric chemoenzymatic synthesis. The FEBS Journal. 2009;276(11):2894-2904
  151. 151. Pohl M, Lingen B, Muller M. Thiamine-diphosphate-dependent enzymes: New aspects of asymmetric C-C bond formation. Chemistry. 2002;8(23):5288-5295
  152. 152. Carini P, Campbell EO, Morre J, Sanudo-Wilhelmy SA, Thrash JC, Bennett SE, et al. Discovery of a SAR11 growth requirement for thiamin's pyrimidine precursor and its distribution in the Sargasso Sea. The ISME Journal. 2014;8(8):1727-1738
  153. 153. Leigh JA, Albers SV, Atomi H, Allers T. Model organisms for genetics in the domain Archaea: Methanogens, halophiles, Thermococcales and Sulfolobales. FEMS Microbiology Reviews. 2011;35(4):577-608
  154. 154. Kaczowka SJ, Reuter CJ, Talarico LA, Maupin-Furlow JA. Recombinant production of Zymomonas mobilis pyruvate decarboxylase in the haloarchaeon Haloferax volcanii. Archaea. 2005;1(5):327-334
  155. 155. Goyer A, Hasnain G, Frelin O, Ralat MA, Gregory JF 3rd, Hanson AD. A cross-kingdom Nudix enzyme that pre-empts damage in thiamine metabolism. The Biochemical Journal. 2013;454(3):533-542

Written By

Julie A. Maupin-Furlow

Submitted: 12 February 2018 Reviewed: 11 April 2018 Published: 05 November 2018